The VitaDAO Blog

Featured Articles

Clear filters

All Posts

March 2024 Longevity Research Newsletter
April 17, 2024
Maria Marinova, Rhys Anderson
Awareness
March 2024 Longevity Research Newsletter

March Longevity Research Newsletter

Introduction

Welcome back Vitalians!

An article on VitaDAO has been published in Nature Biotechnology! It is a case study on the potential of DAOs for funding and collaborative development in the life sciences. In this article, the authors explore VitaDAO’s role in revolutionizing life sciences funding through blockchain technology.

In this month's edition, we delve into the fascinating intersection of regeneration and aging biology, exploring how insights from the natural world's remarkable regenerative processes can illuminate the pathways to healthier aging. While some creatures in nature can astonishingly regenerate lost limbs or damaged organs, humans have much more limited regenerative capabilities, primarily resulting in scar formation rather than true regeneration. 

We are excited to bring you an interview with Prof. Nicole Ehrhart, the director of Columbine Health Systems Center for Healthy Aging at Colorado State University. She shares her fascinating journey into longevity research, initially inspired by the limitations in tissue regeneration in higher mammals. This led her to explore aging biology, where she recognized a significant intersection with her prior work in tissue regeneration. Dr. Ehrhart's research leverages her background in veterinary medicine and surgical oncology, focusing on naturally occurring disease models in animals to improve clinical outcomes in both humans and canines. She emphasizes the importance of multidisciplinary collaboration in advancing aging research and highlights the unique benefits of using companion dogs in clinical trials to accelerate the development of age-reversal therapies.

Longevity Literature Hot Picks

Preprint Corner in collaboration with

The Longevist is a preprint overlay journal spotlighting the most promising longevity studies each quarter.

And just like that, Q1 is over! Check out these latest preprints, which have all been entered into the 1Q24 longlist to be in the running to receive a coveted place in The Longevist. As always, you can refer preprints for consideration in The Longevist

Rejuvenation of aged oocyte through exposure to young follicular microenvironment

Comparative analysis of mouse strains for in vivo reprogramming

Association between prescription drugs and all-cause mortality risk in the UK population

cGAS-STING is responsible for aging of telomerase deficient zebrafish

Unravelling the Transcriptomic Symphony of Sarcopenia: Key Pathways and Hub Genes Altered by Muscle Ageing and Caloric Restriction Revealed by RNA Sequencing

Senolytic CAR T cells reverse aging-associated defects in intestinal regeneration and fitness

Machine learning predicts lifespan and underlying causes of death in aging C. elegans

A torpor-like state (TLS) in mice slows blood epigenetic aging and prolongs healthspan

Published Research Papers

Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity

Aging of the immune system involves a shift from lymphoid-supporting to myeloid-biased hematopoietic stem cells, leading to reduced adaptive immunity and increased inflammation. This exiting new study shows that eliminating myeloid-biased cells in aged mice rejuvenates their immune system.

Formation of memory assemblies through the DNA-sensing TLR9 pathway

Interested in how memory works at the molecular level? Check out the News & Views from Ben Kelvington and I in Nature on this exciting paper from Jelena Radulovic that describes links between DNA damage, innate immunity and memory!

Epigenetic aging of human blood cells is influenced by the age of the host body

The study reveals that the age of a stem cell transplant recipient affects the epigenetic aging of donor cells, accelerating it in older recipients and decelerating it in younger ones, indicating the recipient's body age influences transplanted cell aging.

Effect of long-term caloric restriction on telomere length in healthy adults: CALERIE™ 2 trial analysis

The CALERIE™ 2 trial on caloric restriction (CR) showed mixed effects on telomere length attrition over two years, with initial increased attrition in CR participants that lessened in the second year, suggesting a complex interaction between CR and telomere health that warrants further study.

Sildenafil as a Candidate Drug for Alzheimer’s Disease: Real-World Patient Data Observation and Mechanistic Observations from Patient-Induced Pluripotent Stem Cell-Derived Neurons

Can Viagra treat Alzheimers? The study analyzed millions of insurance claims revealing 30-54% reduced prevalence of disease among those taking drug. In brain cells from AD patients, drug lowered levels of neurotoxic tau proteins and inflammation.

The common marmoset as a translational model of age-related osteoarthritis

The study validates common marmosets as a model for human age-related knee osteoarthritis (OA), showing that geriatric marmosets display OA characteristics similar to humans, including increased prevalence and severity with age, and notable sex differences.

A small-molecule TNIK inhibitor targets fibrosis in preclinical and clinical models

Using AI, researchers discovered TNIK as a target for treating idiopathic pulmonary fibrosis (IPF), leading to the development of the anti-fibrotic drug INS018_055. This drug showed promising anti-inflammatory and anti-fibrotic effects in vivo, with its safety confirmed in phase I trials.

Ergothioneine promotes longevity and healthy aging in male mice

Ergothioneine (ERGO), when supplemented daily in male mice, extended their lifespans and reduced age-related frailty and cognitive decline, pointing to its beneficial role in promoting healthy aging through multiple biological pathways.

CheekAge: a next-generation buccal epigenetic aging clock associated with lifestyle and health

CheekAge, a new epigenetic aging clock developed from cheek swabs and lifestyle/health data of 8,000 adults, accurately predicts age and correlates with health factors like BMI and smoking. It is non-invasive, validated across different conditions and available through a free online tool.

Intermittent rapamycin feeding recapitulates some effects of continuous treatment while maintaining lifespan extension

Intermittent rapamycin treatment in mice extends lifespan with fewer side effects than chronic treatment, although chronic treatment more effectively improves overall healthspan. Both approaches similarly benefit lifespan in males and mitigate certain age-related conditions, but chronic treatment shows a slight survival advantage in females.

Late-life intermittent fasting decreases aging-related frailty and increases renal hydrogen sulfide production in a sexually dimorphic manner

EOD intermittent fasting reduced aging-related frailty in male mice by decreasing energy intake and enhancing renal hydrogen sulfide production, improving various health measures. However, it showed minimal benefits in females, indicating gender-specific effects.

A molecular index for biological age identified from the metabolome and senescence-associated secretome in humans
The Healthy Aging Metabolic (HAM) index, comprising 25 metabolites, effectively predicts healthy aging across genders and races. Key metabolites driving aging and elevated inflammation markers in rapid agers were identified, suggesting metabolic pathways significantly influence biological aging.

An  Updated  Prioritization  of  Geroscience-Guided  FDA-Approved  Drugs  Repurposed to Target Aging

Geroscience focuses on delaying aging-related diseases by targeting aging mechanisms, aiming to extend lifespan and healthspan. Recent updates add bisphosphonates, GLP-1 receptor agonists, and beta blockers as promising gerotherapeutics. 

Multi-omics characterization of partial chemical reprogramming reveals evidence of cell rejuvenation

Chemical reprogramming rejuvenates aged mouse fibroblasts by altering their molecular profile and reducing biological age, particularly by enhancing mitochondrial function. This approach suggests a promising avenue for reversing aging effects in vivo, highlighting the need for further investigation.

Mitophagy defect mediates the aging-associated hallmarks in Hutchinson–Gilford progeria syndrome

Mitophagy defects contribute to the premature aging in Hutchinson-Gilford progeria syndrome (HGPS) by impairing mitochondrial function. Using UMI-77 to induce mitophagy improved aging markers in HGPS and aged mice.

Endothelial-specific telomerase inactivation causes telomere-independent cell senescence and multi-organ dysfunction characteristic of aging

Knocking out the Tert gene in endothelial cells (ECs) of mice induced signs of aging such as senescence and tissue hypoxia, leading to leaky blood vessels, cognitive impairments, and decreased muscle endurance. The study highlighted a shift towards glycolysis for energy, due to reduced mitochondrial function, even in the presence of normal telomere lengths in certain tissues. 

Published Literature Reviews, Hypothesis, Perspectives and more

Translation is an emerging constraint on protein homeostasis in ageing

This review explores how the aging process disrupts the correlation between transcript levels and protein products, influenced by diminished protein synthesis capacity and a decreased ability of aging cells to adapt their proteomes to stress, potentially due to factors like reduced ribosomal function and increased mistranslation.

Time is ticking faster for long genes in aging

Recent studies have shown that with age, longer genes tend to show reduced expression across various species and cell types, potentially due to polymerase stalling from DNA damage. Here the authors term this phenomenon gene-length-dependent transcription decline (GLTD) and also describe its role in age-related diseases.

Pharmacological interventions in human aging

A comprehensive database called AgingDB has been created to consolidate information on over 130 clinical trials focused on pharmacological interventions in aging, covering areas like glucose homeostasis, metabolic interventions, senolytics, and mTOR inhibition. This resource aims to make it easier for individuals to find and stay updated on trials targeting aging, marking a significant shift towards treating aging as a manageable medical condition.

Expanding our thought horizons in systems biology and medicine

Buck Institute “Jennifer Lovejoy expands our thought horizon regarding systems biology RE: the importance of measuring behavior and exposome, both new frontiers in research on aging”.

Decoding lifespan secrets: the role of the gonad in Caenorhabditis elegans aging

The gonad is heavily implicated in C. elegans aging, as removing its proliferating stem cells or inducing stem cell quiescence through starvation significantly extends lifespan, a phenomenon also seen in other nematodes. This review explores the mechanisms behind gonad-mediated lifespan extension, including lipid signaling and transcriptional changes, and suggests that insights from these studies could enhance our understanding of aging's mechanisms and evolution across different species.

Hungry for biomarkers of aging

The geroscience hypothesis, which suggests that interventions targeting biological hallmarks of aging could delay or prevent age-related diseases and disabilities, has sparked significant interest due to early evidence that these mechanisms are modifiable. This approach highlights the necessity and potential challenges of developing and using biomarkers of aging to optimize human health in a transformative way.

Job Board

The Kapahi lab at The Buck Institute has 2 postdoc positions available - using bioinformatic approaches from flies and humans to study the role of circadian clocks and nutrient-responsive pathways influencing eye and neuronal degeneration using flies and conserved orthologs from humans

Professorship in Computational and Systems Biology of Aging and Aging-associated diseases at CECAD Cologne

Looking for a PhD position? Retro might have something in store for you!

Retro College is now accredited in both the EU and USA to grant PhDs.

They have welcomed the first 2 students, and have openings for 3 more.

Postdoc position in the Singh Lab at UCSF to study aging, age-related diseases, and diapause using multi-omics in the African killifish.

News and Media

Longevity: how science is pushing the boundaries for the first 150-year-old human

Using Ultrasound to Assist Gene Therapy

This approach opens up heart muscle cells for potential therapies.

Aeovian Pharmaceuticals doses first participants in Phase 1 clinical trial, strengthens leadership team and raises additional $50 million financing

Hevolution Foundation Announces $20 million Impact Investment to Advance Promising Aging Therapies, Leading a $50 million Series A Extension in Aeovian Pharmaceuticals

‘Ageing isn’t inevitable’: The 100-Year-Life co-author on how to live well for longer

Daniel Ek's new bet on longevity

“The upper echelons of Stockholm society will soon have a new place to spend their money: a luxurious longevity clinic half-owned by the founders of Spotify.”

How to ‘switch on’ your anti-ageing genes – and live longer

A call to arms to save the genome

Silicon Valley Startup Announces Planned Trials of “Fountain of Youth” Treatment Targeting 130-Year Lifespans for Astronauts

By Biotech Explorers powered by a Bioreactor-Grown Mitochondrial Transplantation - a technique that our parent firm Mitrix Bio

Resources

In the publication section above, we introduced AgingDB - a new database of pharmacological trials targeting aging - and here we’d like to share another list of aging clinical trials from AgingBiotech.info!

New Aging Books!

From Nobel Prize winning Biologist Venki Ramakrishnan: 

Why We Die: The New Science of Ageing and the Quest for Immortality (Hardback)

A review of the book from Prof. Charles Brenner.

And, Andrew J Scott’s new book “The Longevity Imperative: Building a Better Society for Healthier, Longer Lives” is out in the UK and soon available in the US.

David Sinclair et al., previously published a paper on The economic value of targeting aging - now check out a YouTube video explaining these ideas!

Prizes

The Million Molecule Challenge, supported by The Longevity Prize, is still open and accepting applications - submit a compound and we will finance the testing at the Ora Labs. There’s no jury for this one, the winners are awarded based on results alone. 

Want to know how it’s going so far? See Leaderboard here.

Conferences

Aging & Gerontology

8-9th April, Valencia, Spain

Midwest Aging Consortium Aging Research Symposium

28-30th April, Columbus, OH

Longevity Med Summit

8-9th May, Lisbon, Portugal

Rejuvenation Startup Summit

10-11th May, Berlin, Germany

BSRA 2024

4th - 6th September

Tweet of the Month

Steven Austad

For geroscientists out there, why do we call them "clocks?"  Clocks tell time of day.  Calendars tell days, months, and years.  Clocks tell us what time to eat and sleep. Calendars tell us our age. They should be epigenetic or proteomic calendars.

Podcasts and Webinars

Longevity & Aging Series

NUS Medicine’s Healthy Longevity Webinar Series

Dr. Jean Hebert: Progressive Brain Tissue Replacement and How we Beat Aging!

STARTALK: Is Aging a Disease? Epigenetics with David Sinclair

The Optispan Podcast with Matt Kaeberlein: Rapamycin's SURPRISING Effects on Aged IMMUNE SYSTEMS

Interview with Professor Nicole Ehrhart

Bio: Dr. Nicole Ehrhart, VMD, MS, directs the Columbine Health Systems Center for Healthy Aging at Colorado State University. Holding the Ross M. Wilkins M.D. Limb Preservation Foundation University Chair in Musculoskeletal Oncology and Biology, she is also a board-certified veterinary surgeon and a leader in both national and international surgical organizations.

What inspired you to enter longevity research? 

I am a clinician scientist who utilizes a wide spectrum of animal models in aging research but I focus on naturally-occuring disease models to aid in translation of discovery to clinical application The first phase of my research career was focused on tissue engineering and regenerative medicine, ie combining synthetic or biologic scaffolds, signals and progenitor cells to regenerate tissue following trauma, infection or tumor surgery. One of the questions that drove my curiosity was why humans and other higher mammals have such limited tissue regeneration capacity as compared to creatures such as salamanders, for example , who can regenerate entire limbs following an amputation. Higher mammals can achieve a small amount of tissue regeneration, but beyond this limited response they simply form scar tissue instead of replacing native tissue. Through my work and others, we learned that a fundamental reason why higher mammals such as humans, horses, dogs etc. do not regenerate large amounts of native tissue following acute injury was due to exhaustion of the tissue stem cell pool.   I spent a lot of time working on strategies to “rejuvenate” tissue stem cells. When the cellular hallmarks of aging were first described, I was fascinated to learn that stem cell exhaustion was one of the fundamental drivers of cellular aging. There was an intersection I hadn’t been aware of between my work and what was, at the time, the rather nascent field of aging biology. It dawned on me that the same strategies we were using to boost tissue regeneration following acute tissue loss could potentially be applied to the slow, insidious loss of tissue volume or function associated with aging. I could see that the impact potential was huge. Not everyone suffers from acute tissue loss but every one of us ages and loses tissue function and mass along the way, just very slowly. That was the inspiration to enter longevity research. 

Which of the current theories of ageing do you think are the most convincing?

This is a difficult question to answer because we continue to refine our knowledge about aging biology daily. The field is literally exploding with new data that are rapidly shaping and re-shaping our understanding. These advancements in our knowledge are not linear and iterative. Therefore, instead of concerning ourselves with which theory is most convincing, we need to understand that they each represent parts of larger story which is yet incomplete. We are still the blind men trying to make a theory about the entire elephant but we only have perspective on separate parts of the whole, to use a common analogy. I believe we should be focusing on the intersections of the different theories to see what bigger picture is beginning to crystalize.  

How has the field changed since you started?

Geroscience has gained more recognition as a distinct field of study, leading to increased funding opportunities from government agencies, private foundations, and the biotech industry. This has allowed researchers to pursue more ambitious projects and collaborations. Progress in identifying new and refined biomarkers of aging, will be crucial for assessing longevity interventions. This area has also progressed since I entered the field.

What mistakes do you think the longevity field has made?

In the past, there has been a tendency to search for single interventions or "magic bullets'' for extending lifespan, overlooking the complexity of aging processes. This approach does not take into account the interrelatedness of the drivers of aging. In addition, there has been too much emphasis on data collected from traditional laboratory animal models such as fruit flies, worms and mice. This approach has resulted in extremely important mechanistic and proof of concept data that has advanced our overall knowledge greatly. However, as exciting as these results are, the models do not at all mimic the complexity of human aging.  When we use these data alone to move forward with human clinical trials, it's common for such interventions to fail or, even worse, have unintended negative consequences. This has led to a lot of disillusion and loss of credibility in aging biology. Further investment to successfully translate these discoveries becomes too high risk and many exciting and potentially highly efficacious interventions are abandoned at that point. To solve this problem, we need to build a better bridge across the huge translation gap. Investing in this niche will better inform go/no-go decisions for new longevity therapeutics and is a powerful, cost-effective way to de-risk and accelerate gerotherapeutics.

Is ageing a disease?

Modern medicine is based upon detection and subsequent diagnosis of a specific condition which is then treated. However, changes in our cells and tissues are occuring with the passage of time. We call this “aging”. Older bodies are made up of older cells that are less and less able to repair from the wear and tear of everyday function or damage. Subsequently, when enough unrepaired damage accumulates within an organ or body system, the system fails to work properly. We recognize this end dysfunction as a “disease”. So maybe the question is: are accumulating changes that are upstream of the onset of a disease diagnosis also part of the “disease”. My answer to this is “yes”. Aging is a modifiable process that occurs over time and we should not wait until the dysfunction is so severe so as to put a specific name on it to treat it. 

Given your unique background in veterinary medicine and surgical oncology, how does this perspective enrich your approach to aging research, and are there specific insights or methodologies from these fields that can notably advanced the study of aging biology?

The  direction forward must also include a way to study aging on an accelerated timeline,which brings us to the topic of model organisms in research. Laboratory mice – the most common species involved in testing the safety and effectiveness of a drug prior to human clinical testing – are astoundingly poor models of human aging. They live in highly controlled environments, eat identical diets and are genetic twins of one another. This is a far cry from humans, who are genetically diverse and have a myriad of social, economic, environmental and lifestyle behaviors that influence their health outcomes. Consequently, a mouse-to-man approach for new drugs has historically been unsuccessful. Primates, our closest genetic relatives, are an alternative model in which to study age reversal drugs, but primates also have long lives, and when in a laboratory environment, they are not subject to the same variations in environment, exposures and habits as human populations. However, there is another promising approach: a species that lives among us, shares our environments, lifestyle habits and social structures. They develop the same diseases of aging as humans, are genetically diverse and have access to well-established heath care systems and interventions, yet they age much more rapidly than humans. Enter the companion dog. Pet dogs have coevolved with humans and represent a unique animal population that could be a powerful bridge to accelerating the development and approval of age reversal drugs in humans. There are millions of companion dogs in the United States, for example,  and the quality of veterinary care has paralleled human medical care, such that there is a large population of dogs living to old age and developing diseases of aging naturally, just as humans do. The development of well-designed clinical trials to show health span extension in family dogs would need to include all the ethical and safety considerations of human clinical trials. Studies such as these are conducted while the pet continues to live out their daily life in their home with their family – while the medical team conducting the trial carefully ensures that their health and well-being are maintained. Pet owners can choose if they wish to voluntarily enrol their companion dogs in a study and participate in citizen science, collaborating with researchers to share observations, thereby cocreating and contributing to scientific outcomes. Engaging companion dogs in health span clinical trials provides a chance to enhance their wellbeing and life span, not only for their benefit, but also as a more accurate predictor of the effectiveness of health span-extending drugs in human populations. Given the identical nature of most age-related diseases between dogs and humans, and the shorter canine life span, the overall effect would be the acceleration of meaningful health span extension therapies for humans that our animal companions will benefit from too. In my opinion, well designed, placebo controlled, multi-institutional clinical intervention studies in companion dogs;  which incorporate all the safety and ethical considerations that are employed in human clinical trials yet cost a fraction of the amount for a similar human clinical trial, are a vastly underutilized resource. 

As the director of Columbine Health Systems Center for Healthy Aging how do you foster collaboration across different fields to advance aging research and are there particular focus areas that are emphasised?

I truly believe that the answers to the most challenging human issues, such as aging, lie at the intersections between disciplines. Academia is very good at drilling down deeply within specific science disciplines, but we have sometimes failed to bring the lens of multiple scientific domains to bear on a single challenge.   Colorado State University’s Center for Healthy Aging unites and facilitates interdisciplinary research teams across multiple scientific fields to address the grand challenge of global aging. Our work creates research teams comprised of many disciplines- from the behavioral sciences to engineering to medicine. My personal passion and focus is to find novel ways to bridge the gap between discovery in geroscience and meaningful healthspan interventions that will impact how we, and the creatures we share our world with, can live our best lives.

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful in helping you to keep up-to-date with all the exciting longevity-related developments. See you next month!

Further Reading

Nanoplastics causes heart aging/myocardial cell senescence through the Ca2+/mtDNA/cGAS-STING signaling cascade

Age-dependent loss of HAPLN1 erodes vascular integrity via indirect upregulation of endothelial ICAM1 in melanoma

Neuroprotective treatment with the nitrone compound OKN-007 mitigates age-related muscle weakness in aging mice

How excess niacin may promote cardiovascular disease

An article on VitaDAO has been published in Nature Biotechnology! It is a case study on the potential of DAOs for funding and collaborative development in the life sciences. In this article, the authors explore VitaDAO...
Read more
Entropy and Epigenetics in Aging Science with Peter Fedichev and Jan Gruber - The VitaDAO Aging Science Podcast
April 7, 2024
Awareness
Podcast
Entropy and Epigenetics in Aging Science with Peter Fedichev and Jan Gruber - The VitaDAO Aging Science Podcast



In the current episode of The VitaDAO Aging Science Podcast, we explore the fascinating intersection of entropy, epigenetics, and aging with our esteemed guests, Peter Fedichev, founder of Gero and a trailblazer in longevity research, and Prof. Jan Gruber from Yale-NUS, known for his deep understanding of the physics behind aging. As we navigate through Peter Fedichev's recent paper that sparked heated discussions on the limits of age-reversal, we'll delve into the science of stochastic changes in methylation patterns, the controversial debate around the reversibility of aging, and the impact of entropy on human longevity. This episode will also shine a light on the vital role of VitaDAO in science funding, the challenges faced by PhD students in today's economic climate, and the exciting potential of naked mole rats in aging research.

A brainstorming session about science funding and the impact of entropy on human aging – a talk with Peter Fedichev and Jan Gruber

Peter Fedichev is considered by many of my colleagues as an extraordinarily creative and productive aging researcher. This is one of two reasons why I invited him on the VitaDAO aging podcast. The second is that Peter’s recent paper attracted a lot of controversial debate on the longevity subreddit (1) and I was hoping we could clarify some of the issues that people raised.

https://www.reddit.com/r/longevity/comments/y8ewm3/aging_clocks_entropy_and_the_limits_of_agereversal/

In his paper Peter claims that stochastic changes in methylation patterns impose a hard limit on our ability to reverse aging (2). In this podcast we discussed this thesis and whether reprogramming refutes his ideas.

We also talked about the hardships of PhD students, the role of VitaDAO in science funding, linear and non-linear features that change with age, the difference between hallmarks of aging and proximal causes of aging, the mean-field effect, stable and unstable animals, naked mole rats and more. Although ultimately we may have failed to really break down and simplify his work for the general audience, the journey was still a worthwhile one and I hope you will enjoy our podcast, even though the second part is quite technical.

Below I will provide a few comments that might be useful to better understand the podcast.

Brief bio – Peter Fedichev and Jan Gruber

“Peter Fedichev, Ph.D., is an entrepreneur and scientist with over 20 years of experience in academic research and biotech business. He is a founder of Gero, a longevity startup focused on developing therapies that will extend a healthy human lifespan. Peter’s scientific background lies in the field of condensed matter physics, biophysics and bioinformatics…His dream is to defeat aging and experience life in space.”

peter fedichev | GIANT Health London 9-10 December 2024


https://loop.frontiersin.org/people/203716/bio

Prof. Jan Gruber (Yale-NUS) is another physicist turned biologist with space travel ambitions. I invited him because he is very familiar with Peter’s work in the hope that he could explain the math better than I can. Not only is Jan quite familiar with the physical ideas behind Peter’s research but he is also one of the most knowledgeable aging researchers I have met.

Jan Gruber - Associate Professor - National University of Singapore |  LinkedIn

Science funding and PhD life

Peter thinks that VitaDAO should hire more experts and delegate decisions to them. Both Jan and Peter suggested to fund risky projections at an early stage. You can listen to the podcast to for the whole discussion covering more of the nuance here.

Another conversation we had was about the life of PhD students and we all probably agreed that the funding situation is rather dire, needing improvement.

"economically the life of a PhD student is pretty hard these days… the system is broken" (Peter Fedichev)

In that context it is interesting to note that VitaDAO realized the extent of this problem and now offers small amounts of money to young and/or low-income students and researchers. These are called the VitaDAO fellowships. The idea is that a small sum can make a big difference to someone who is in need, making it a very effective program. As far as I can tell, these fellowships are partly supported by external donations allowing VitaDAO to focus most of their internal funds on funding research while still doing community building together with other donors.

https://www.vitadao.com/fellowship

Epigenetics and epigenetic clocks

Genes encode the information to produce various proteins. Genetics deals with genes while epigenetics describes the regulation of gene expression through epigenetic marks. There are two major types of epigenetic marks which can affect gene expression. Histone marks and DNA methylation. The latter is usually attached to so called CpG islands (which are stretches of Cytosine and Guanine bases) and the object of study in most clock papers. In general, CpG methylation is thought to reduce the expression of nearby genes. However, more interestingly, it appears that these CpG methylation patterns change in certain ways with age, thus allowing us to predict a person’s age based on their DNA methylation.

What is entropy and how does it relate to aging?

Simply put, entropy describes the amount of disorder in a system. The amount of entropy in the universe increases over time, which is equivalent to the second law of thermodynamics. Similarly, the amount of entropy increases in a closed system and will thereby limit its functionality. Since biological systems are complex any increase in entropy ultimately poses an issue for their functioning.

However, the earth is considered an open system so the limitations imposed by the physical concept of entropy do not apply. On the other hand, there is a closely related concept of stochastic damage which is very similar and, indeed, such damage is considered a major driver of aging. So in way the idea that entropy drives aging was never really in doubt.

I would say the major differences in opinion are related to the number and heterogeneity of these lesions that comprise molecular damage. Optimists like Aubrey de Grey would argue that there are only few types of damage that limit human lifespan and the robust lifespan extension effects seen with compounds like rapamycin may also be taken as support for the malleability of aging.

What makes Peter Fedichev’s work interesting is that it entails a rather pessimistic interpretation instead. In the case of epigenetic aging, given the large number of CpG islands that all show stochastic or entropic changes over time it would very difficult to intervene and reverse this damage. It is not physically impossible to reverse that damage at so many different genetic loci, but technically exceedingly difficult – to put it mildly.

Perhaps inspired by the great Russian writer Peter explained that each aged cell will accumulate entropic changes, but each cell will have different changes. As they say: “All happy families are alike, but every unhappy family is unhappy in its own way” (Leo Tolstoy)

While perhaps unpopular among Biohackers and Longevity Advocates, there is strong support for the idea that epigenetic changes are stochastic. Indeed, Björn Schumacher and David Meyer from CECAD just published a manuscript showing that this stochastic variation is sufficient to construct aging clocks. While I have not read it, I do respect their work and presume that it is solid (3).

Nevertheless, Peter is optimistic that we could at least radically slow aging, as he explains in the podcast. I would speculate that perhaps compounds such as rapamycin can only slow aging and bona fide rejuvenation is indeed difficult.

Does reprogramming and evolution prove that entropy is irrelevant?

“But my biggest issue is that I can't square their claims against the fact that the cells in our body are millions of generations old. For women and men, they generally have been alive for 20+ years before their gametes are used/generated for the next generation. Now these cells maintain their DNA in a more highly regulated way, but doesn't this throw cold water on entropy after aging has occurred?” (Poster 1)

“Now, I have said I don't necessarily agree with the paper. And that is because we have empirical evidence, and Dr. Sinclair is on the record saying, that there is a "backup copy" of lost information somewhere in the cell.” (Poster 2)

“I don't really have theoretical objections to this like most of the respondents here do, but purely empirical ones: aging has already been reversed in several organisms. So there is obviously something wrong with the theory,” (Poster 3)

Indeed, evolution shows that entropy does not limit the existence of life per se, since complex cellular organisms have existed almost since the dawn of time.

Moreover, researchers have found that epigenetic reprogramming during early embryogenesis could contribute to rejuvenation of the species. Reprogramming here means the erasure and resetting of epigenetic marks which is reflected in a reduction of epigenetic age. Many commenters have argued that the success of such reprogramming, both partial in the lab and natural in the womb, implies that Peter’s thesis of irreversibility must be wrong. Hence epigenetic aging must be reversible.

Without going into much detail, I do think they have a point. However, most people seem to forget another major ingredient necessary for rejuvenation of the species’ bloodline, which is selection. Just to give a few examples, men produce hundreds of millions of sperm, which are in competition with each other for general and especially mitochondrial fitness because they have to race towards an egg cell to fertilize. This competition will help to filter out sperm cells with high entropy because these will, most likely, show reduced swimming ability, reduced ability to penetrate the egg’s zona pellucida, reduced ability to produce viable offspring, etc.

In fact, we have strong empirical evidence for selection as a complementary mechanism that together with reprogramming should reduce epigenetic age (and entropy). The almost exponential increase in fetal chromosomal abnormalities seen with the mother’s age proves that reprogramming is not enough (4). In nature, spontaneous abortions, failure to fertilize and implant – which can be caused by chromosomal abnormalities among other issues – would select against offspring with low functional status. It is tempting to speculate that a fetus or embryo that, by chance, inherited a higher epigenetic age would be selected against, at some stage.

This is my own thesis and Peter discusses some other arguments in the podcast.

What are stable and unstable organisms?

As far as I understand, in his framework Peter categorizes organisms in two groups based on their dynamic stability. When looking at physiologic traits like complete blood counts, humans show a low auto-correlation of these values whereas mice show high levels of auto-correlation. This means that a perturbation in mice would persist for longer, whereas humans are able to return to equilibrium quicker; i.e. they are more stable (5, 6).

“we observed that the fluctuations of physiological indices in humans are also dominated by a collective variable characterized by a relatively long but finite auto-correlation time (in the range of a few weeks) and associated with age and all-cause mortality. The number of individuals exhibiting signs of the loss of dynamic stability (measured by exceedingly long auto-correlation times) increased exponentially with age at a rate matching the mortality doubling rate from the Gompertz mortality law” (6)

One implication is that the effects of anti-aging treatments should persist for a long time in mice, whereas the effects in humans would be transient and small to begin with, at least during midlife which he characterizes as a period of stability.

This idea of mice being dynamically unstable is conceptually similar, although distinct, to the broader notion of mice being fragile and short-lived. You could perhaps say it is another line of evidence in favor of this idea. When we say mice are short-lived we do not just mean it in the trivial sense. Of course they are! What we generally like to do is to compare the lifespan of animals to the expected lifespan based on their body mass which is also known as the longevity quotient. Small species tend to live shorter lives. Mice, however, are even shorter-lived than expected based on their size, which is also the reason why all of us are so interested in naked mole rats that are the opposite – unusually long-lived for their size.

References

1. “Aging clocks, entropy, and the limits of age-reversal”
https://www.reddit.com/r/longevity/comments/y8ewm3/aging_clocks_entropy_and_the_limits_of_agereversal/

2. Tarkhov, Andrei E., Kirill A. Denisov, and Peter O. Fedichev. "Aging clocks, entropy, and the limits of age-reversal." bioRxiv (2022): 2022-02.
https://www.biorxiv.org/content/10.1101/2022.02.06.479300v2.abstract

3. Schumacher, Björn, and David Meyer. "Accurate aging clocks based on accumulating stochastic variation." (2023).
https://scholar.google.at/scholar?hl=en&as_sdt=0%2C5&as_ylo=2010&q=Accurate+aging+clocks+based+on+accumulating+stochastic+variation&btnG=

4. https://www.preimplantationgeneticdiagnosis.eu/pgd/risk-of-aneuploidy-and-maternal-age.aspx#:~:text=However%2C%20the%20frequency%20of%20aneuploidy,50%25%20of%20embryos%20are%20abnormal.

5. Avchaciov, Konstantin, et al. "Unsupervised learning of aging principles from longitudinal data." Nature Communications 13.1 (2022): 6529.

6. Pyrkov, Timothy V., et al. "Longitudinal analysis of blood markers reveals progressive loss of resilience and predicts human lifespan limit." Nature communications 12.1 (2021): 2765.

In the current episode of The VitaDAO Aging Science Podcast, we explore the fascinating intersection of entropy, epigenetics, and aging with our esteemed guests, Peter Fedichev, founder of Gero, and Prof. Jan Gruber
Read more
February Longevity Research Newsletter
March 10, 2024
Maria Marinova & Rhys Anderson
Awareness
Longevity
February Longevity Research Newsletter

Introduction

Hi Vitalians, greetings from Vitalia. You might have heard about the city of life in Roatan, Honduras. 

VitaDAO hosted a Life Extension Conference, held from February 23 to February 25, 2024, in the pop-up city of Vitalia, Honduras, was a profound journey into the future of human longevity. Nestled within Próspera's special economic zone on the idyllic Roatán Island, Vitalia emerged as a beacon of innovation, drawing inspiration from the visionary minds of Vitalik Buterin and Balaji Srinivasan. This unique event attracted over 200 attendees from around the globe, uniting them in a shared vision for a future where extended healthspan and lifespan are not just possible but expected. With a collective eye towards decentralized cities of longevity, speakers and participants alike embraced the notion of these new frontiers in the eternal quest for a longer, healthier life.

Vitalia's existence represents more than a transient assembly. It serves as a foundational step towards the creation of permanent districts dedicated to longevity, poised to fundamentally transform the biotech landscape. The special economic zone of Prospera in Roatán has a legal environment that is conducive to medical freedom and pioneering work, setting an exhilarating stage for progressive dialogues. This year's gathering not only underscored the symbiotic relationship between urban planning and health but also brought to light emerging biological research and the future of medical practices. 

Projects such as Vitalia are at the forefront of accelerating longevity therapeutics development. Currently, aging is not officially recognized as a disease indication for clinical trials, raising the question of how aging should be classified. The debate is polarized; some firmly argue that aging is indeed a disease, while others strongly disagree, with many viewing the debate as a distraction from the primary goal of trying to understand and treat the age-related pathologies. This month, we're thrilled to bring you an interview with Dr. Barry Bentley, who received an Impetus Longevity Grant to explore this every topic. Enjoy!

Longevity Literature Hot Picks

Preprint Corner in collaboration with

The Longevist is a preprint overlay journal spotlighting the most promising longevity studies each quarter.

Check out these latest preprints, which will be entered into the 1Q24 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

HMGA1 orchestrates chromatin compartmentalization and sequesters genes into 3D networks coordinating senescence heterogeneity

Most axonal mitochondria in cortical pyramidal neurons lack mitochondrial DNA and consume ATP

Published Research Papers

Therapeutic targeting of cellular senescence in diabetic macular edema: preclinical and phase 1 trial results

Targeting senescent cells in the retina with UBX1325, a senolytic drug, reduces damage in diabetic macular edema (DME). This approach, shown to be safe in a phase 1 trial, offers a potential new treatment strategy for DME, necessitating further research for validation.

Defining the progeria phenome

The study introduces a novel approach for diagnosing progeroid disorders by developing a 'progeria phenome' and a machine-learning classifier available at https://www.mitodb.com. It also identifies new associations between certain diseases and progeroid syndromes, enhancing our understanding and diagnosis of premature aging conditions.

Wealth Redistribution to Extend Longevity in the US

A study linking wealth inequality to survival rates in the US suggests redistributing wealth could increase median longevity by up to 2.2 years, potentially aligning US mortality rates with OECD averages. Simulations indicate that policies promoting wealth equality, including those inspired by Japan's distribution, minimum inheritance, and baby bonds, could significantly reduce survival disparities.

Protein signatures of centenarians and their offspring suggest centenarians age slower than other humans

The New England Centenarian Study identified unique protein signatures in centenarians, suggesting that despite acquiring aging markers similar to those in younger, shorter-lived individuals, they do so later in life. The study highlights specific serum proteins associated with senescence and longevity.

Plasma membrane damage limits replicative lifespan in yeast and induces premature senescence in human fibroblasts

This research reveals that plasma membrane damage (PMD) leads to cellular senescence, contributing to aging. In budding yeast, PMD was linked to shorter lifespans, but lifespan extension was possible by upregulating membrane repair mechanisms. In human fibroblasts, PMD induced senescence through the Ca2+–p53 pathway, with upregulation of repair factor ESCRT-III mitigating this effect. 

Genome-wide profiles of DNA damage represent highly accurate predictors of mammalian age

Recent advancements in genome-wide mapping techniques for single-strand breaks and abasic sites in DNA have enabled the discovery of precise age-related genomic patterns of DNA damage in mice. These patterns offer superior age prediction accuracy compared to traditional transcriptome analysis, focusing on a select group of genes. 

Inhibition of S6K lowers age-related inflammation and increases lifespan through the endolysosomal system

Inhibiting TORC1 with rapamycin extends lifespan across species, a process mediated by S6K. In Drosophila, S6K activation in the fat body negates rapamycin's benefits, affects lysosome structure, and reduces inflammaging, linked through Syntaxin 13. Gender differences in inflammaging response to rapamycin were observed, with significant effects in females but not in males.

Published Literature Reviews, Hypothesis, Perspectives and more

Cellular senescence: Neither irreversible nor reversible

Cellular senescence, traditionally viewed as irreversible, might under certain conditions be transient. They suggest that senescence is a variable journey, not a permanent state, and cells can potentially return to the cell cycle, entering a distinct post-senescent phase with unique functional and clinical outcomes.

Targeting ageing with rapamycin and its derivatives in humans: a systematic review

Rapamycin and derivatives benefit immune, cardiovascular, and skin aging with no significant impact on endocrine, muscle, or nerve systems, and unassessed effects on other bodily systems. While safe in healthy individuals, they increased infections and cholesterol in those with age-related diseases, indicating a need for further and long-term studies.

Validation of biomarkers of aging

Efforts to identify omic-based biomarkers for biological aging aim to predict health outcomes and assess anti-aging interventions. However, a unified approach for validating these biomarkers before clinical use is missing. This review discusses the validation challenges and proposes recommendations to improve the process.

Missing centenarians are an international concern

Job Board

Loyal is looking for a Chief of Staff! Loyal aims to help dogs live longer and maintain their quality of life as they age, hoping to do the same for humans one day.

The Kapahi lab is seeking postdocs for an eye aging and neurodegeneration project using flies and mammals. For questions regarding this position, please email Pkapahi@buckinstitute.org or visit the website.

Postdoctoral Positions available at Brown University Center on the Biology of Aging (BoA). A list of participating investigators and detailed descriptions of faculty research programs can be found on their website

Assistant or Associate Full Professor of Geroscience positions at UCSD:

Assistant Professor - (Tenure-track): Apply

Associate or Full Professor - (Tenured): Apply

News and Media

Hevolution Announces 49 Awards to Catalyze Discovery in Healthspan Science Through Innovative $115 Million Grants Program

BioAge Announces $170 Million Oversubscribed Series D Financing to Accelerate Development of Obesity and Metabolic Disease Therapeutics

Caloric Restriction Extends Reproductive Lifespan in Hens

Life Biosciences and Forge Biologics Announce cGMP Manufacturing Partnership to Advance Development of Novel Gene Therapies for Aging- Related Diseases

Solving Atherosclerosis: The Small but Mighty Molecule. Cyclarity’s Dr. Matthew O’Connor talks about ending atherosclerosis

Why age matters when it comes to cancer

Neurona Therapeutics Raises $120M to Advance Groundbreaking Pipeline of Regenerative Cell Therapy Candidates for Chronic Neurological Disorders

Resources

Reproductive aging resources - compiled by Carol Magalhaes.

Prizes

Longevity Prize Million Molecule challenge with Ora Biomedical

Propose interventions to extend C. elegans lifespan, with a chance to win a prize for the most effective approach. Ora Biomedical has developed the WormBot-AI platform, combining robotics and AI to study longevity in C. elegans, a model organism for aging research. This technology enables quantitative assessment of lifespan and healthspan on a large scale, advancing our understanding of longevity.

Biomarker Challenge, our open-science competition rewarding folks who can best predict chronological age, survival/mortality and multi-morbidity incidence using biomarker data. Challenge officially begins March 1st.

Developing and validating aging biomarkers for clinical trials is challenging due to data limitations and collaboration barriers between scientists. To address this, Biolearn, an open-source toolset, was created. It harmonizes omics datasets and calculates existing aging biomarkers. More about Biolearn:

Biolearn, an open-source biomarker validation library and platform enabling easy and versatile analyses of biomarkers of aging data. 

Biolearn is a tool that simplifies the analysis of biomarkers of aging data. It allows users to easily import data from publicly available sources like the Gene Expression Omnibus, National Health and Nutrition Examination Survey, and the Framingham Heart Study. Biolearn also includes reference implementations for common aging clocks such as the Horvath clock and DunedinPACE, which can be run with just a few lines of code. 

Join Biolearn Discord.

Conferences

Aging and Gerontology

April 08-09, 2024

Valencia, Spain

5th Annual MAC Aging Research Symposium

April 28 – 30, 2024

Columbus, OH, USA

The Longevity Med Summit

May 8 - 9, 2024

Lisbon, Portugal

The Rejuvenation Startup Summit 2024

May 10-11, 2024

Berlin, Germany

Tweet of the Month

Carol Magalhaes:

Why has menopause become a normal part of every woman's life, when it causes unnecessary suffering?

Why is reproductive aging research still significantly underfunded when half of the population experiences menopause?

I believe this *needs* to change, so I wrote a piece about how to delay menopause to extend longevity.

Podcasts and Webinars

Longevity & Aging Series

NUS Medicine’s Healthy Longevity Webinar Series

A great list of Aging Podcasts

80,000 Hours Podcast - Laura Deming on the science that could keep us healthy in our 80s and beyond

Matt Kaeberlein’s new podast covering a variety of topics related to longevity and healthspan - An introduction to the study of RAPAMYCIN

How this tiny mutant mouse lives so, so long

Interview with Barry Bentley

Dr Bentley is the Head of the Bioengineering Research Group and Bio-AI Laboratory at Cardiff Metropolitan University. His current research focuses on the development of laboratory systems for cryopreservation, and the development of classification and staging systems for age-related pathology within the context of the WHO ICD-11.

What inspired you to enter longevity research?

I have been fascinated and unsettled by ageing, probably for as long as I have been aware of ageing as a concept; however, I can pinpoint the exact moment I became interested in longevity as a topic of scientific research. In early 2006 I was travelling around China and teaching English on a gap year before starting university. One night, I found myself in a dimly lit internet café on the outskirts of Wenzhou city, where I was browsing through forums on science and technology to pass the time. I stumbled across a link to an article discussing the Methuselah Foundation, which led me down the rabbit hole into the world of ageing research. The prospect of slowing or even reversing biological ageing was and still is a radical idea, sounding, to some, like a concept straight out of science fiction. The realisation that there were people who were seriously working on such a seemingly intractable problem captured my imagination. I already knew at that point that I wanted to become a research scientist, but that was the nudge that shifted my focus to the biological sciences, and ultimately led to my current career.

Which of the current theories of ageing do you think are the most convincing?

Each theory has its proponents, and various degrees of evidence supporting them. With the incredible progress that has been made in developing robust epigenetic clocks, the field seems to have shifted in favour of programmed theories of ageing in recent years. With how complex biology is, and how much is still unknown, I personally think it would be a mistake to commit to any one view or theory of ageing at this point; in fact, I would not be surprised if we find that all of the main theories are true to varying extents, with ageing being a highly heterogeneous process. To inelegantly coin a phrase: I suspect there is more than one way to age a cat.

How has the field changed since you started?

The biggest change has undoubtedly been the amount of interest in the field, and consequently, the amount of investments now being made in longevity research. Whether that is because the problems of an ageing population are finally starting to become too profound to ignore, or the science has reached a level of maturity that is finally investible, society as a whole is starting to take note.

What mistakes do you think the longevity field has made?

Until recently, the longevity field – as distinct from the broader field of ageing research – had serious credibility issues, which it still has yet to completely rid itself of. Starting as a fringe area, it unfortunately became associated with some questionable groups: unscrupulous people selling snake oil, biohackers doing questionable self-experiments, and futurists who were making predictions with little consideration for the realities of research. The field did a poor job of distancing itself from the more unsavoury ideas and actors. The strength of the science now speaks for itself, but reputational damage remains.

What advice would you give to people currently working in longevity research?

Like all areas of research, the main advice I would give is to be open minded. We do not yet know which approaches will be required to effectively slow or reverse ageing – there is just too much that we don’t yet know. To make progress, we’ll need to continue asking questions, trying new approaches, and robustly challenging our assumptions.

You were awarded an Impetus grant to explore how ageing should be classified. Why is this important?

That’s a great question. Before I answer, let me give some context to how the project came about: In 2016 and 2017, my collaborator, Dr Stuart Calimport, and I were discussing our frustrations that even though the scientific understanding of ageing was rapidly improving, the science was not filtering down to the bedside to influence the way patients were treated. Part of the reason for this, we realised, was due to clinical classifications.

The World Health Organization (WHO) maintains a standard list of clinical codes, known as the International Classification of Diseases (ICD), which are used as the standard terminology by all WHO member states. These are used for recording diagnoses and treatments, for statistical and epidemiological purposes, for healthcare resourcing, approving clinical trials, medical billing, and so on – in short, they are at the core of virtually everything that happens within a health system.

Unfortunately, classifications for ageing-related changes are extremely limited in the ICD, which limits the ability to accurately describe and address ageing-related pathologies. As the idiom goes: you can’t manage what you don’t measure; and the ICD does not currently have the capacity to describe, and thus measure, ageing-related pathologies. Some groups have proposed to work around this by classifying ageing itself as a disease, but not only is this contentious, it also fails to address the heterogeneous nature of ageing – different organs and tissues age in different ways, and I believe we need the ability to capture this.

We had initial success in getting ageing recognised as a cause of disease by the WHO in 2019 [1], following which we published a large multi-author paper in Science proposing a consortium to develop a comprehensive and systematic classification system for ageing-related pathologies [2]. Norn Group thankfully saw the value in this, and awarded us an Impetus Grant to make it a reality. So far, we have recruited approximately 300 experts, who we are working with to develop these new classifications.

 [1] https://doi.org/10.1089/rej.2019.2242

[2] https://doi.org/10.1126/science.aay7319

You recently hosted a consensus meeting to define the criteria for an ageing-related pathology, could you give us the highlights of this meeting?

This was the first meeting of the consortium to develop the classifications for ageing-related pathologies. Of course, to develop those classifications, it is essential that the consortium members have agreement on what constitutes an ageing-related pathology – so this was the main goal of the first meeting: to define the criteria that will allow us to differentiate an ageing-related pathology from others. We had a wide range of input from clinicians and scientists, but there was pretty much unanimous support for three criteria that were proposed: Firstly, that the damage must develop or progress with increasing age; that it should be associated with, or contribute to, functional decline or increased susceptibility to functional decline; and that, for the purposes of clinical classification, it must be based on evidence from studies in humans.

Our consortium coordinator, Dr Emma Short, who is a consultant histopathologist and research associate, did a brilliant job of chairing the meeting, and is working on writing-up the outcome of the discussions, which we will be publishing in the near future.

Is ageing a disease?

There is little agreement amongst gerontologists on how to classify ageing, mainly because ageing is such a catch-all term that refers to many things: biological age, as a sociological status or marker, chronological advancement, appearance, etc. Personally, I think the debate is somewhat counterproductive. Regardless of how we use the term, and whether we call it a disease, the fact remains that people become sicker as they get older. My own approach is to focus on those aspects that make people sick: the underlying ageing-related pathology.

What are you currently working on in the lab?

In addition to my work classifying age-related pathology, I am fortunate to have received a Fulbright Award, which is enabling me to work at Harvard Medical School and Massachusetts General Hospital this year on the development of new technologies for the cryopreservation of cells, tissues, and organs. It has been an ambition of mine for a long time to make contributions in this area, so it’s exciting to finally be making that a reality.

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful in helping you to keep up-to-date with all the exciting longevity-related developments. See you next month!

This time we’ll leave you hanging, waiting for some exciting developments on our Members Portal. VitaDAO will soon enable $VITA holders to stake their tokens in exchange for health products and services. Keep an eye out!

Further Reading

Age-Associated Differences in Recovery from Exercise-Induced Muscle Damage

Association of testosterone and sex hormone-binding globulin with all-cause and cardiovascular disease mortality in older Chinese men

Mendelian randomization reveals apolipoprotein B shortens healthspan and possibly increases risk for Alzheimer’s disease

Intermittent fasting promotes rejuvenation of immunosenescent phenotypes in aged adipose tissue

Unraveling Histone Loss in Aging and Senescence

Hi Vitalians, greetings from Vitalia. You might have heard about the city of life in Roatan, Honduras. 
Read more
Stake $VITA in Exchange for Health Products and Services 🤩
March 4, 2024
Awareness
Longevity
Stake $VITA in Exchange for Health Products and Services 🤩

Dear VitaDAO Community,

Exciting news is on the horizon! We're introducing a revolutionary feature soon for $VITA token holders: the ability to stake $VITA your tokens in exchange for premier health products and services aimed at enhancing your longevity.

Stake $VITA: Unlock Exclusive Longevity Benefits

  • Biological Age Tests: Take control of your aging process by discovering your biological age, and receive personalized insights to help slow it down.
  • Continuous Glucose Monitors (CGM): Gain real-time insights into your glucose levels, a key factor in metabolic health and longevity.
  • Custom Health Insurance: Featuring Genki, specially crafted for digital nomads and travelers, providing you with peace of mind through global coverage.
  • Exclusive partnerships with leading longevity-focused companies like GlycanAge, BonCharge, Humanity App, Nuchido, Novos, Ledger, and others, delivering cutting-edge products and services directly to you.
  • Plus many more coming soon

Why Participate?

Our community is passionately leading the charge in the exploration of longevity and health optimization. Regardless of your current health goals—be it improving metabolic health, enhancing biological functions, or preventing age-related conditions—our staking program is tailored to support your ambitions. Access essential longevity tools such as at-home health test kits, high-quality supplements, and the latest biohacking devices.

By staking your $VITA tokens, you're not just accessing a suite of products and services; you're joining a movement dedicated to pushing the boundaries of what it means to live a long, healthy life.

VitaDAO is at the forefront of this movement, partnering with industry leaders to ensure our members have exclusive access to the forefront of longevity research and interventions.

Keep an eye out for more details on how to participate and make the most of your $VITA tokens.

Become a $VITA holder: Get $VITA on Uniswap

Visit VitaDAO.global to stay updated on this transformative journey.

We're introducing a revolutionary feature soon for $VITA token holders: the ability to stake $VITA your tokens in exchange for premier health products and services aimed at enhancing your longevity.
Read more
A Beginner’s Guide to Vitalia
February 22, 2024
Morgan J. Weaver
Awareness
Longevity
A Beginner’s Guide to Vitalia


Vitalia builds a decentralised city for longevity biotech development, initiated by a 3-month pop-up city experience (open to short and long-term visitors, you can apply here)

Written by Morgan J. Weaver, published in collaboration between VitaDAO and Stranded Technologies.

The curious technophile ventures forth across the small and wondrous meatspace of a pop-up city-within-a-charter-city in Roatan, Honduras, chronicling the emergence of an experimental biotech startup hub, crypto-augmented Special Economic Zone, and nuclear reactor of liberal governance, big brains, and disruptive ideas¹.

Gazing out at a horizon so vividly turquoise that I begin to question whether my coffee has been trendily microdosed, it is easy to mistake this globally-renowned SCUBA destination and idyllic Caribbean getaway as a mere tourist haven. Postcard-perfect palms sway in a breeze that feels gentle as bathwater, laden with small coconuts which occasionally drop to Earth. Grass roofs top shallow fishing and diving docks, just as in too-perfect vacation stock photos. Small waves break against the world’s second-largest barrier reef, just a kilometer from shore--a penumbra of fantastical corals surrounding something possibly much stranger to us land-dwellers.

Framed in this vivid environment is a buzzing hub of ambitious STEAM activity, alive with provocative and audacious ideas, and an unquantifiable mass of intellectual horsepower and optimistic creativity. The air is laden with, What if? Legal engineers, doctors, biohackers, web3 developers, research scientists, psychologists, community builders and entrepreneurs in myriad verticals have convened in tropical Roatan’s Pristine Bay for a 2-month pop-up network city called Vitalia: The City of Life, with a theme that sounds surreal to many: Make Death Optional.

Why here and why now? Vitalia was chosen as a physical manifestation of what some might classify as a Network Society--a decentralized group cooperating and coordinating their energies around a set of unifying philosophies, which occasionally physically manifests-- permanently, temporarily, centrally or in a decentralized manner. The concept was popularized by Balaji Srinivasan, whose interactive and streamlined book on the concept, The Network State, may be read in its entirety, free of charge, here. This touchdown on Caribbean shores is no accident--Roatan is home to Próspera ZEDE (Zona de Empleo y Desarrollo Económico), a Honduran Special Economic Zone and charter city. Founded around 2021, Próspera is considered one of the most autonomous of existing Special Economic Zones, including Singapore, Hong Kong and Dubai. Próspera’s financial backers include Pronomos Capital, a fund led by Patri Friedman, the founder of the Seasteading Institute and backed by Peter Thiel, Naval Ravikant, Tim Ferriss and Marc Andreessen--well-recognized names and veterans of startup societies and early investing. However, these names make up only a small fraction of investment into Próspera and their role has been overstated by the media. Most of Próspera investors are not as well-known, include only a few institutional VCs, and invest in the space out of conviction--a bet on a different future which they believe should exist. Próspera’s success seemed too early cypherpunk, not ready even for the most forward-looking VCs. Given its continual and steady development over the course of recent years, this may soon change.

The concept of Próspera is uncomplicated: streamlined legal code and liberal governance, paired with the goal of economic prosperity through entrepreneurship and rapid development encouraged by low business taxes, will empower residents, citizens and businesses, and allow Próspera’s government to cover its own operating costs, unlike many existing governments. In addition to fresh legal and policy approaches, financial technologies such as cryptocurrency are part of this plan to empower residents, citizens, and businesses. For example, Próspera hosts AmityAge - a Bitcoin Education Center that has onboarded roughly 60 businesses on the island to accept Bitcoin payments. Próspera also offers cryptocurrency as legal tender, and accepts payments from multiple blockchains including Ethereum and Solana. Businesses can be incorporated here (e.g. LLCs) quickly and efficiently. And since law may be copied, one can, for instance, set up a Delaware-style corporation, or copy and iterate on a variety of other legal structures that have proven successful in the past.  Businesses in the ZEDE are free to transact in crypto without securities laws, and they are allowed to tokenize their assets and require no capital gains tax. The ZEDE itself has investment protection under both national law and international treaties, granting investors, entrepreneurs and residents the stability to build and invest with a long time horizon.

Las Verandas resort, the main campus upon which Vitalia is held.

Próspera’s jurisdiction includes several parcels of land including Pristine Bay, consisting of loosely themed neighborhoods including the Beta District. The Beta District harbors Duna, a 14-story residential tower currently under construction, and Las Verandas, a luxury golf resort including larger villas and condominiums for year-round living, plus a small piece of land on which GARM Clinic, a medical tourism destination offering cutting-edge longevity treatments, sits. Próspera ZEDE’s jurisdiction also encompasses a mainland Honduran shipping port (Satuye, in Las Ceiba) where port workers and their families are expected to live, facilitating shipments back and forth and by extension speeding along the significant construction that is needed to build this charter city.

Before delving into Vitalia, we’d be remiss to ignore VitaDAO, a closely involved organization that sits at several intersections including longevity biotech, web3, culture, startup incubation, and funding mechanism innovation. For non-web3 readers, I direct you to a quick introduction to the Decentralized Autonomous Organization (DAO), a web3 concept consisting of a collective of participants, working under some common goal, that coordinates decision-making and resources through code in a permanent ledger--a blockchain. VitaDAO’s bottom line is to slow, stop, and even reverse aging, which includes the extension of healthspan as well as lifespan.  

You may be wondering at this point why aging is a target--not cancer or osteoporosis or metabolic syndrome, or even addiction. Aging is VitaDAO’s target because of the 150,000 human deaths per day on planet Earth, roughly 100,000 are caused by aging itself, which includes age-related disease that is a direct result of the aging process³.  Let us sit with this for a moment: aging is more deadly than any disease, group of diseases, or cause of death including accidents, addiction, war, starvation, or infection.  If aging itself were viewed as a primary disease, two-thirds of these deaths could be prevented if a cure were reached.  And if the aging process could be reversed, delayed, or otherwise mitigated, we could look forward to living a greater number of healthy, productive years--years spent with our families, partners and communities, years learning, pursuing growth and picking up meaningful longer-term projects.  The potential of the human species and civilization as a whole, unfettered by mortality, is incalculable.  At this point we can direct critical and inquiring readers to VitaDAO’s Longevity Briefing Primer for a more detailed overview of this goal, its supporting philosophy and mechanisms.

What’s more, in industrialized nations, up to 90% of deaths per year are related to aging and age-related disease³.  Perhaps most provocative of VitaDAO’s founding vision is the idea that aging is a disease, and that we should make death optional by understanding and eliminating this disease. This idea creates friction in the US regulatory system, as the FDA currently neither recognizes nor legitimizes the development of therapies aimed at the enhancement of currently healthy people, as opposed to the treatment of disease or disability. The FDA does not currently recognize aging as a disease.

So if aging is a target, what is the shape of the organization that aims to prevent it?  VitaDAO is composed of scientists, researchers, web3 and web2 professionals, academics, dealmakers, venture capital, and others collectively pushing decentralized science forward in the arena of longevity and enhancement biotech, complemented by web3 technologies, to solve some of the most persistent problems of traditional science.  Examples of problems VitaDAO is working to solve include the high cost of Big Pharma-style clinical trials, averaging $2.3 billion per therapy² according to Deloitte, and the regulatory bottlenecks of conducting clinical research.  Other persistent issues of traditional science in academia and institutions, or “tradsci”, include the gatekeeping and misaligned incentives of academic publishing, patient data privacy and sovereignty, and access to and compensation for dataset usage. This is all backed by a belief that the current biomedical research apparatus is failing to deliver treatments and progress in a timely manner, and the data-backed observation that Big Pharma has stagnated for years, failing to bring treatments to market for sick and dying people.  While approved pharmaceutical drugs occasionally do unknown harm, as opposed to known side effects, deaths from the lack of therapies that weren’t approved sooner, or were mired in regulatory complications kill far more people than those who die from approved drugs.  According to Zina Sarif of Yendou, a melanoma research foundation, 10,000 cancer deaths per year could be prevented by existing drugs stuck in long trials.  

Here in Pristine Bay we witness the tinder of an enabling legal platform combined with the spark of innovative biotech and other industries, agitating for faster, cheaper, more efficient regulatory processes. And Vitalia, a gust of oxygen funneling in a migration of scientists, researchers, technologists, and entrepreneurs for the pop-up city and its numerous mini-conferences, with aims to convert some of these to permanent residency.  Part of this thrust consists of four conferences: two on Longevity Biotech bookending the event, the second on Crypto Cities & Startup Societies, and the third conference on AI & Tech Progress.

Aubrey de Grey and Bryan Johnson in a fireside chat with Vitalia co-initiator and VitaDAO steward Laurence Ion.

The first of these 3-day weekend conferences, Longevity Biotech Part I, featured notable speakers and serious contributors to the fields of longevity, rejuvenation biotech, geriatrics and gerontology, such as Aubrey de Grey, José Cordeiro, and billionaire biohacker and elven-skinned longevity advocate Bryan Johnson. I sat in on two days of lectures. Surprisingly, there was not a huge emphasis on lifestyle interventions--considering that so far, even a rigorous regimen of sleep, healthy diet, social wellness, and stress management can only get us so far, with the most genetically blessed of us, lifestyle unknown, living to about 116 or so. An increased healthspan augmented by lifestyle, so the common thread went, will allow us to access the next available rejuvenation technologies as they arrive, until a final innovation to stop aging is found.  Cryonics, or whole body or brain preservation by special freezing techniques, are currently considered a great Plan B amongst many longevity experts and researchers.  

Talks during the first Longevity Biotech conference focused on dozens of potential biotechnologies for rejuvenation and making death optional.  Rather than wishful thinking about juice cleanses or sleep protocols prolonging life far past anything we’ve seen so far (which lifestyle advocates admit won’t take us to immortality), talks focused on bone marrow rejuvenation, cohesive theories of aging such as mitochondrial dysregulation and biological clocks located in individual bodily cells as well as specific clusters of cells in the brain, and even the occasional homebrew science project, such as an introduction to the art of running an electrical current through muscle tissue (monkeys and scientists) to express target molecules, such as ebola antibodies, based on injected mRNA.

The second conference, Crypto Cities & Startup Societies, spanned the weekend of February 2-4. In the spare weekend between conferences, Vitalians explored the island’s bays, reefs, eateries, and other local gems. A bonfire with ecstatic dancing was thrown on the beach in front of the Pool Club, a sprawling, multi-story building on Las Verandas and the de facto center of gravity of the event that houses coworking, an aerial silks space, a gym, meeting rooms, cold plunge and sauna, a salon, and a biohacking lab. Other weekend entertainments included locally grown cacao, freediving certification, local art, Lionfish spearfishing, Palmetto Bay and its brewery, and local Howler monkeys. An all-ages, Vitalian DJ’d party at Roatan Yacht Club with dancing, open laptops containing glucose data charts, and intense philosophical conversations about consciousness and the nature of work unfolded.

What separates Vitalia from a Web2-style conference, or even a Web3 conference (some of which exude a distinct festival energy), is both tangible and not. Many of the organizers involved also had a hand in attending or organizing Zuzalu and Zuconnect, two legendary Web3 events that concentrated the best and brightest--intelligent, creative, weird and open-minded dreamers with diverse backgrounds, mostly united by their agency and idealism. These events were founded by Vitalik Buterin, founder of Ethereum, and you can read more about his reason for creating these genre-defining events here. Part of Vitalia’s unique culture is its do-it-yourself approach to organizing--anyone can organize workshops, events, activities, and groups. Hackathons spur infrastructure development and experimentation to support the Vitalia and Zuzalu-adjacent community itself with open-source, tailor-made event tooling such as ZK-gated forums, ticketing, and scheduling software. Attention to the whole person is also something that separates this event from a typical Web3 conference--wellness activities abound--from holotropic breathwork to men’s and women’s groups, to circling and authentic relating, to meditation and pilates and yoga and improv. Artists and psychologists and podcasters and scientists rub shoulders with hackers and VC investors. The Zuzalu formula, the Zuclear reactor, is alive and well at Vitalia. It feels less like a tech conference and more like a summer camp for gifted adults, where conversations flow freely and cross-pollination of ideas is abundant.

Vitalia isn’t just high-caliber brains, but heart as well. International community-builders work with New Governance experts to engineer a culture and a city that will stick--something that will have staying power and encourage people to plant their flags, start families, and build in a technological, regulatory and ideological frontier nestled in the tropics, given a shot at changing the world by investors, innovators, and the Honduran government.

  1. Tribute to Neil Stephenson’s 1996 Wired gem, Mother Board, Mother Earth, on the intro.
  2. Cost of pharmaceutical asset development: https://www.genengnews.com/gen-edge/the-unbearable-cost-of-drug-development-deloitte-report-shows-15-jump-in-rd-to-2-3-billion/
  3. Age-related mortality statistics: https://en.wikipedia.org/wiki/Aging-associated_diseases#:~:text=Of%20the%20roughly%20150%2C000%20people,is%20higher%2C%20reaching%2090%25.
  4. Honduran criminal law applies in the ZEDE--things that were already illegal in Honduras, such as abortion and illegal drugs including psychedelics, are also illegal in the ZEDE.

The curious technophile ventures forth across the small and wondrous meatspace of a pop-up city-within-a-charter-city in Roatan, Honduras, chronicling the emergence of an experimental biotech startup hub, crypto-augmented Special Economic Zone
Read more
Challenging Mouse Models in Aging Science with Dr. Dan Ehninger - The VitaDAO Aging Science Podcast
February 15, 2024
Podcast
Awareness
Challenging Mouse Models in Aging Science with Dr. Dan Ehninger - The VitaDAO Aging Science Podcast

In this episode of the VitaDAO Aging Science Podcast I spoke with Dr. Dan Ehninger about the shortcomings of modern mouse research as he sees them. I share many of the same concerns and think we do have to ensure that the foundations of biogerontology are sound. Although I do believe that the time for clinical translation is now, in parallel we must work on these foundational questions, some of which include:

  • Does rapamycin and caloric restriction really work and are these treatments robust? And what do we mean when we say “really” and “robust”?
  • The mouse is a fundamentally flawed model due to its shortlifespan and lab adaptations – but how flawed is it? Do the flaws preclude meaningful conclusions in aging research?
  • How to design mouse studies that mitigate these flaws?
  • How do we measure aging, functional decline, aging rate outside of lifespan? (e.g. using age-sensitive phenotypes; ASPs, to use a term from Dan’s research)

Although I am a longevity optimist and may not always agree with all the points that Dan makes, I think it is important for skeptics to be heard. It was a pure coincidence that I stumbled on his work, which some of my colleagues have never heard of. After reading several of his papers, I decided it is time to highlight his outstanding and controversial work as a service to the community.

Dr. Dan Ehninger – Brief Bio

Dan Ehninger is a Senior Research Group Leader at the German Center for Neurodegenerative Diseases (DZNE) in Bonn. His group is interested in the biology of aging and the pathogenesis of age-related brain disorders. He studied medicine at Charité University Medicine/Berlin, Harvard Medical School/Boston and University College London. From 2001 to 2004, he carried out his graduate work in the laboratory of Gerd Kempermann at the Max Delbrück Center for Molecular Medicine in Berlin, where he worked on the behavioral regulation of adult hippocampal neurogenesis and adult cell formation in the neocortex. His postdoctoral work (2004 to 2009) with Alcino J. Silva at University of California - Los Angeles focused on molecular and cellular mechanisms of cognitive impairments in mouse models of neuropsychiatric disorders. In 2010, Dan Ehninger joined the faculty of the German Center for Neurodegenerative Diseases (DZNE) and was later promoted to a tenured Senior Research Group Leader position.

A person in a suitDescription automatically generated


https://www.dzne.de/en/research/research-areas/fundamental-research/research-groups/ehninger/research-areasfocus/

Mice are very short-lived

There are three major issues with mice as a model:

1) They are shorter-lived than expected based on their weight – in the wild early death is compensated by fast reproduction. This means the “tricks” we use to extend mouse lifespan might not work on humans, because we already evolved to utilize these enhancements and more.  One solution is to learn from longer-lived animals, which is what comparative biogerontologists do. We discussed this topic in an earlier podcast with Vera Gorbunova.

2) Mice develop cancer more readily than humans (probably related to point 1), which is the key reason why Dan thinks they fail to faithfully capture aspects of human aging. At least if all you do is measure lifespan.

3) Strains we use in research labs are further adapted to fast growth and reproduction. Thus reproducing and growing even faster than in the wild. One solution to this problem is using more diverse strains, as discussed with Rich Miller in an earlier podcast.

Why do we still run mouse studies? Because using mice is better than the alternatives like cell culture, invertebrates, reading tea leaves, etc. Whereas the truly superior alternatives are not feasible or affordable. Did you know that a large phase III study in humans targeting mortality, as is often done for cardiovascular drugs and will be necessary for anti-aging treatments, can cost north of 100 million dollars?

Healthspan and lifespan uncoupling

There are two schools of thought on the matter of healthspan. Some people believe that it is difficult or impossible to extend lifespan substantially without improving health. Others believe that by now we have so many examples of healthspan-lifespan uncoupling that we must consider these almost independent.

To give an example of such healthspan-lifespan uncoupling in mice we do not need to look very far. A recent study found that reducing the levels of the famous Myc oncogene in mice extends lifespan by reducing cancer while leading to premature aging-like phenotypes (Wang et al. 2023). These include fatty liver, reduced strength, fur quality and spinal degeneration.

To give some more real-world examples of healthspan-lifespan uncoupling in humans, women live longer than men while having more age-related diseases. Testosterone replacement therapy could improve quality of life in older men even though it worsens CVD risk factors, but the effects on mortality are controversial (1).

No matter which school you belong to, there is emerging consensus that longevity treatments need to improve health and affect reasonable biomarkers of aging, especially functional ones.

The rate and trajectory of change in these markers can teach you a lot about the underlying mechanism of your intervention. That discussion is at the heart of our podcast episode.

Disease modifying drugs vs. Band-Aid solutions

I like to use these terms from pharmacology to think about potential longevity drugs and treatments. 

“Disease modifying drugs” are pharmacological agents that aim to modify the underlying pathophysiology of a disease, potentially slowing down or halting the progression of the disease. In contrast, non-disease modifying drugs typically focus on symptom management without significantly influencing the underlying disease progression. For example, in the case of Alzheimer’s disease a cholinesterase inhibitor such as donepezil would be symptomatic, whereas anti amyloid antibodies should be disease modifying. For osteoarthritis painkillers are typical examples of a symptomatic solution whereas senolytics – it is hoped – could be disease modifying.

Dan’s argument, as I understand it, is that several lifespan extending treatments have age-independent effects on aging-related phenotypes. He calls them age-sensitive phenotypes; ASPs. This was something his group saw for mTOR hypomorphism, dwarfism and intermittent fasting in mice (Xie et al. 2022). Based on this age-independent effect he speculates that these treatments do not slow aging and instead extend lifespan by delaying cancer. All perfectly reasonable, although I do have some questions even after the podcast. 

Why would these treatments improve ASPs in young animals? I would presume if they did nothing for aging the null hypothesis would be that some ASPs go down and others go up. Maybe we can discuss this the next time! Another issue that I find counterintuitive is that usually such “band-aid” solutions do NOT alter the course of a disease and the benefits do NOT persist over a long time and well into old-age. Normally, I would expect a convergence towards the expected path with a non-disease modifying drug or treatment. This is what I would call a “band-aid solution” but rapamycin and CR seem to be much more than that.

A graph of a solution and a rejuvenationDescription automatically generated

Many would argue that the actual mechanism of action does not matter. “If it works it works”. Caution is warranted here, however. We only know that these treatments work in mice and mouse cancer incidence is much higher than in humans (2). It also matters in the long term. When developing robust therapies we need to build on solid foundations. If CR only operated through cancer inhibition than all the effort to search for CR-mimetic drugs in order to slow aging would be wasted.

Wider context and reception

While the points made by Dan might seem controversial at first glance they are being echoed by many other researchers. Not everyone agrees on the specifics, but there is emerging consensus that we have problems with our model systems and need to improve. Steve Austad, for example, is a big proponent of studying the longevity mechanisms employed by ultra long-lived species rather than short-lived mice. We will interview him soon on this topic. Similarly, Peter Fedichev believes that the mouse is not an adequate model and he focuses on data mining human datasets instead to find new gerotherapeutics. Rich Miller believes that currently used mouse strains lack genetic diversity and we should focus on using heterogeneous strains while Arlan Richardson would like to validate treatments in rats, that are even more genetically distinct from regular lab mice.

Summary and conclusions

Ultimately, I do think that the high incidence of cancer in mice exaggerates the benefits of certain treatments, in agreement with Dan. There are, however, two interesting counter arguments to Dan’s theory which states that almost all the benefits of longevity interventions are due to cancer. 1/ If it was so easy than many more drugs should extend lifespan. Plenty of drugs tested for lifespan extension have strong chemoprevention data, either theoretical or preclinical, yet they fail to extend lifespan. The only drug that works robustly is coincidentally in a major growth sensing pathway, mTOR, which is also decreased by several other canonical interventions like dwarfism and calorie restriction. 2/ Taueber’s paradox dictates that the wholescale eradication of cancer in humans would only lead to a 5% increase in lifespan because other age-related diseases progress at the same time. In a similar vein, the attenuation of cancer in mice might not be enough for substantial lifespan extension, although this would require modelling studies to prove conclusively.

I also agree with Dan that our mouse studies would be much better if we included young treated controls too. Researchers with sufficient funding should run such controls much more often.

I think – and that is my personal speculation – that rapamycin is an odd drug that leads to partial rejuvenation and does not operate by slowing aging per se. This would explain some of the results seen in Dan’s data (see Xie et al. 2022 for mTOR hypomorphic mice, see Neff et al. 2013 for rapamycin). 

Indeed, it seems that there is a causal pathway between fast growth and fast aging. The pathways that enable fast growth, tissue regeneration and reproduction may be harmful in the long run.  There is a switch-like aspect to CR and CR-like states, which is consistent with the idea they may have evolved as a response to acute nutrient starvation and famines. Perhaps this can explain why they are beneficial without slowing all aspects of aging?

This and many other questions remain to be answered. Let’s get on with the work so we can slow aging as effectively as possible in humans.

Further reading

(1) It is possible that high dose testosterone therapy is beneficial while regular HRT is actually beneficial: “Fewer deaths occurred with testosterone treatment (six [0·4%] of 1621) than placebo (12 [0·8%] of 1537) without significant differences between groups (odds ratio [OR] 0·46 [95% CI 0·17–1·24]; p=0·13).”
https://www.thelancet.com/journals/lanhl/article/PIIS2666-7568(22)00096-4/fulltext

(2) Cancer could bias almost every single functional outcome and aging phenotype except perhaps histopathology. Imagine a mouse with underlying cancer. Will it have high or low frailty? Will it cooperate in a water maze or have high grip strength?

Our manuscript with Prof. Brian Kennedy and Matt Kaeberlein on the importance of long-lived mouse controls which would help to mitigate some of the “artefactual” or “idiosyncratic” non-age related deaths in mice:
Pabis, Kamil Konrad, et al. "The impact of short-lived controls on the interpretation of lifespan experiments and progress in geroscience." bioRxiv (2023): 2023-10.
https://www.biorxiv.org/content/10.1101/2023.10.08.561459v1.abstract

CR Plus, CR adjacent, CR independent - what the heck?
A blog post in which I worry about the lack of new models in mouse aging since most of them seem to be anti-anabolic and CR-adjacent.
https://biogerontolgy.blogspot.com/2020/09/cr-plus-cr-adjacent-cr-independent-what.html?q=anabolism

Keshavarz, Maryam, et al. "Targeting the “hallmarks of aging” to slow aging and treat age-related disease: fact or fiction?." Molecular Psychiatry 28.1 (2023): 242-255.
Note: a recent review where Dan goes into his arguments

Xie, Kan, et al. "Deep phenotyping and lifetime trajectories reveal limited effects of longevity regulators on the aging process in C57BL/6J mice." Nature Communications 13.1 (2022): 6830.

Keshavarz, Maryam, et al. "Targeting the “hallmarks of aging” to slow aging and treat age-related disease: fact or fiction?." Molecular Psychiatry 28.1 (2023): 242-255.

Keyfitz, Nathan. "What difference would it make if cancer were eradicated? An examination of the Taeuber paradox." Demography 14 (1977): 411-418.
https://link.springer.com/article/10.2307/2060587

Neff, Frauke, et al. "Rapamycin extends murine lifespan but has limited effects on aging." The Journal of clinical investigation 123.8 (2013): 3272-3291.

Ham, Daniel J., et al. "Distinct and additive effects of calorie restriction and rapamycin in aging skeletal muscle." Nature communications 13.1 (2022): 2025.

Wang, Huabo, et al. "Premature aging and reduced cancer incidence associated with near-complete body-wide Myc inactivation." Cell reports 42.8 (2023).

In the upcoming episode of The Science of Aging Podcast, we dive deep into the critical questions surrounding mouse models in aging research with our special guest, Dr. Dan Ehninger...
Read more
January Longevity Research Newsletter
February 6, 2024
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
January Longevity Research Newsletter

Introduction

Welcome back Vitalians. I’m sure that most of you would have by now heard the sad news that renowned Professor Judith Campisi has passed away. In place of our usual interview, this month we would like to dedicate an in memoriam to her followed by a few words from one of her protégés, Marco Demaria. 

It used to be believed that human cells could proliferate indefinitely, until Leonard Hayflick performed some pioneering experiments in the early 1960s which showed this was not the case, and after a reproducible number of divisions, later coined the “Hayflick Limit” cells would permanently stop dividing. On one hand this was viewed as a tumour suppressor mechanism, although it also had implications for aging, in that tissues would not be able to replenish themselves.

In 1995, Prof. Campisi discovered a senescence marker - named senescence-associated beta galactosidase or SAβ-gal - that enabled scientists to detect senescent cells both in cell culture and in vivo. Almost 30 years and 8000 citations later, SAβ-gal still remains the most commonly used marker to detect senescent cells. Some notable examples of its use include the seminal papers showing that removal of senescent cells can extend lifespan in mice and the discovery that senescent cells play a role in growth and patterning during mammalian embryonic development.

Then, in the early 2000s, Judith Campisi made a remarkable discovery showing that senescent cells are associated with a unique secretory phenotype, coined the senescence-associated secretory phenotype or SASP. She then began to elucidate the significance of this, leading to numerous discoveries that the SASP could cause inflammation and even drive cancer. This caused an explosion in the senescence field, with her and others discovering that the SASP was also involved in immune clearance of senescent cells, could elicit damage in neighbouring cells, was required for optimal wound healing and more.

With ~500 papers and over 100 thousand citations one can’t begin to comprehend the significance of her research. Her discoveries transcended the senescence/cancer fields and added much needed credibility to the ageing field in general, by describing a detrimental, yet tangible phenotype which can be therapeutically targeted. There are now a large number of biotech companies working on “senomorphics” specifically aiming to attenuate the SASP in hope of extending human healthspan and lifespan.

Now we would like to share a few words from Marco Demaria.

“Judy was a passionate scientist, always looking forward and always digging in the unknown. She hated to do incremental research; her goal was to always find the new. She was highly creative but also extremely rigorous – you could not convince her very easily without showing many repeats of the same experiment. She kept repeating: 'The data are the data'.

Her laboratory was a family, and she tried to help everybody within and outside the laboratory.  She was positive and collaborative also in the most difficult moments. All the people she met were impressed by her kindness and openness. She never said no to a glass of wine and a performance on the dance floor, reflecting her very social attitude and her italian blood.  

She supported me from the first day I joined her lab, and she kept doing so after I left. She told me the importance of doing science the right way, not to care about impact factors. She taught me how you can be a confident leader, how you can manage many things with a smile on your face. She always found the right words to advise and encourage me through different parts of my career. I will miss all the endless conversations on science and life we had, listening to her stories and memories, and seeing her spontaneous interest for everything I told her.”

Finally, here is a podcast she recorded for the Buck Institute 



Introducing: Monthly Longevity challenge

Focus on a topic every month starting with a quick and easy guide on exercise for longevity from Nina Patrick. We have also announced a competition for the community to share their attempt on the challenge and post proof or work(out) in our discord channel. The most active members will not only crush their goals but also be awarded a prize of 200 VITA

Longevity Literature Hot Picks

Preprint Corner in collaboration with

The Longevist is an overlay journal spotlighting the most promising longevity studies each quarter through a collaborative and transparent process, leveraging insights from a panel of global experts in the longevity space.

The Q4 2024 shortlist is now in the hands of the curators as they vote to determine they favourite preprints of the quarter! In the meantime, check out the 3rd issue of The Longevist and look out for our newly assigned ISSN!

Longevity-featured research (Q2 2023 shortlist) now published in Frontiers in Aging

Initiation phase cellular reprogramming ameliorates DNA damage in the ERCC1 mouse model of premature aging

Check out these latest preprints, each of these will be entered into the Q1 2024 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

A universal molecular mechanism driving aging

CD4 T Cells Acquire Cytotoxic Properties to Modulate Cellular Senescence and Aging

A drug cocktail of rapamycin, acarbose, and phenylbutyrate enhances resilience to features of early-stage Alzheimer’s disease in aging mice

Published Research Papers

The efficacy of chemotherapy is limited by intratumoral senescent cells expressing PD-L2

Chemotherapy triggers the upregulation of PD-L2 in cancer cells, aiding their evasion from the immune system and supporting tumor growth. Blocking PD-L2, in combination with chemotherapy, effectively clears senescent cancer cells, offering a potent anti-tumor strategy.

Titan mice as a model to test interventions that attenuate frailty and increase longevity

The Titan mouse, a short-lived species, presents a valuable model for aging research due to its brief lifespan and early signs of aging. Dietary changes and senolytic drug treatments, like Navitoclax, have shown promising results in extending lifespan and reducing signs of senescence in these mice. 

OXR1 maintains the retromer to delay brain aging under dietary restriction

The mtd/OXR1 gene affects lifespan extension through dietary restriction by maintaining cellular trafficking processes. Its decline contributes to aging and neurodegeneration, but enhancing its function offers potential for treating age-related disorders and extending lifespan.

NAD+ supplementation prevents STING-induced senescence in CD8+ T cells by improving mitochondrial homeostasis

The study finds that mitochondrial dysfunction leads to T-cell aging and senescence, mediated by the cGAS-STING pathway. Increasing NAD+ levels with nicotinamide mononucleotide (NMN) can prevent this senescence and improve immune function and survival in mice.

An accurate aging clock developed from large-scale gut microbiome and human gene expression data

Utilizing a large dataset of 90,303 stool samples, the study developed a refined microbiome-aging model, revealing correlations between biological age and lifestyle or health factors—such as higher predicted ages in individuals on a paleo diet or with IBS, and lower in vegetarians. 

Long term methionine restriction: Influence on gut microbiome and metabolic characteristics

The Methionine restriction (MR) diet improves metabolism in mice but has minimal long-term effects on the gut microbiome, with age being a more significant factor in microbial changes than diet. Early stages of MR diet showed specific microbial alterations, but aging predominantly influenced the microbiome composition over time.

Mathematical recapitulation of the end stages of human ovarian aging

Using mathematical models, this study accurately predicts individual variability and population-wide patterns in ovarian aging, specifically the timing of menopausal transition (MT) and age at natural menopause (ANM). 

The immunity and redox clocks in mice, markers of lifespan

The Immunity and Redox Clocks, developed from immune and redox markers in mice, accurately estimate biological age, correlating with lifespan and the effects of lifestyle interventions. These models, validated with strong accuracy, offer a tool for quantifying aging and assessing interventions' impacts on longevity.

Mucosal TLR5 activation controls healthspan and longevity

Stimulating TLR5 with a flagellin fusion protein improves lifespan and health in mice, showing benefits like reduced aging signs, enhanced cognitive and stem cell functions, and better bone and lung health. It also strengthens intestinal integrity, highlighting TLR5 stimulation as a promising strategy for healthy aging.

Iatrogenic Alzheimer’s disease in recipients of cadaveric pituitary-derived growth hormone

The study suggests Alzheimer's disease (AD) can be linked to early exposure to growth hormone treatments contaminated with amyloid-beta (Aβ), indicating AD may have iatrogenic forms transmitted through medical intervention, similar to Creutzfeldt-Jakob disease. 

Published Literature Reviews, Hypothesis, Perspectives and more

Hand grip strength as a proposed new vital sign of health: a narrative review of evidences

Hand grip strength (HGS) is a crucial measure of muscle function, particularly in the aging population. This review highlights its relevance to health, showing that HGS can reliably assess muscle strength. Low HGS is associated with various diseases and health issues, making it a valuable biomarker and potential vital sign throughout life.

Sleep disorders and Alzheimer’s disease pathophysiology: The role of the Glymphatic System. A scoping review

This review investigates the link between Alzheimer's disease (AD) and sleep disturbances, focusing on their impact on the glymphatic system. It includes 70 research articles and categorizes findings related to protein aggregation, glymphatic markers, circadian dysregulation, and potential interventions. 

Genome Instability and DNA Repair in Somatic and Reproductive Aging

DNA repair is essential for genetic maintenance, differing between somatic and germ cells. DNA damage contributes to aging and diseases like cancer. Germ cells' stringent DNA repair mechanisms can inform strategies for improving somatic DNA repair and maintaining health during aging.

Seven knowledge gaps in modern biogerontology

The editorial summarizes the responses from the Biogerontology editorial board regarding crucial unanswered questions in aging research. Seven knowledge gaps are categorized into evolutionary aspects, survival and death mechanisms, and aging heterogeneity. 

From biological aging to functional decline: Insights into chronic inflammation and intrinsic capacity

Intrinsic capacity, a combination of physical and mental abilities, is vital for functional ability. Its decline, often linked to aging, is affected by chronic inflammation. This review examines the connection between chronic inflammation, inflammation-related markers, and intrinsic capacity.

Aging research comes of age

A surge in funding has transformed aging research, making it a mature and dynamic field with promising results. Researchers now have the resources to delve deeper into the mechanisms of aging using tools like 'omics approaches and larger study sizes. While several hallmarks of aging have been identified, researchers acknowledge that aging is a complex process with multiple contributing mechanisms. 

Distinguishing between driver and passenger mechanisms of aging

Identifying which changes drive aging is difficult, and empirical approaches using animal models and human genetics have been employed. Overall, our understanding of the drivers of human aging is incomplete, but uncovering them has the potential to revolutionize biomedical research, especially in the context of a growing global aging population.

Nurturing longevity through natural compounds: Where do we stand, and where do we go?

The past decade has advanced aging research, revealing new hallmarks of aging and potential longevity interventions. Plant-derived compounds are also under investigation. Recent developments, approaches targeting aging hallmarks, and the potential of plant-based compounds in promoting longevity are discussed.

Job Board

Dr Ferriera’s lab at The University of Edinburgh is seeking an independent and motivated Senior Research Technician to be part of an interdisciplinary team studying ageing and regeneration.

Head of a research group wanted for Genome Maintenance Mechanisms in the Department of Radiation Biology in Cologne, Germany.

Computational Life Science Research Professional at the Wyss-Corey Lab at Stanford University.

LongeVC is looking for a senior associate.

Buck Institute for Research on Aging is looking for a Postdoctoral Researcher to join the Gerencser lab.

Join the Kapahi Lab as a Research Associate.

The Kapahi Lab is looking for a dynamic and driven individual to join their team as a Research Associate at the Buck.

Bill Keyes is looking to recruit a PhD student studying senescence through their international call.

Marketing Manager at Loyal. Their mission is to bring the first FDA approved aging drugs to the market, and have already achieved ground-breaking milestones on our path to gaining FDA approval for the first lifespan extension drug for any species.

Postdoc position available in Björn Schumacher’s lab, Institute for Genome Stability in Ageing and Disease. CECAD-Cluster of Excellence in Aging Research, University of Cologne. investigating the fundamental role of DNA damage in aging and how inheritance of stable genomes is regulated.

The Chellappa lab is hiring a Postdoctoral Research Associate in Aging, Metabolism and Host-microbiome interactions at Brown University Department of Molecular Microbiology and Immunology.The project will focus on studying the host and microbial metabolism using systems biology approach. 


Ora Biomedical, Inc. is a longevity biotechnology company dedicated to revolutionizing healthy lifespan by targeting aging. They are recruiting a Research Scientist in  Seattle.

News and Media

The Future of Longevity: The Battle Against Human Aging (NFX Original)
Short Documentary

What is behind ageing’s causal wheel?

Cells Across the Body Talk to Each Other About Aging

The Meme King of Longevity Now Wants to Sell You Olive Oil

Congratulations to Thomas A. Rando, MD, PhD, who has been appointed as the newly elected President of the Board of Directors for AFAR

Scientists can tell how fast you're aging. Now, the trick is to slow it down

Life span increases in mice when specific brain cells are activated

Growing old while staying young: The unique mechanisms that defy aging in plants

It's not your life span you need to worry about. It's your health span

The Biomarkers of aging consortium has launched a newsletter - sign up here!

Dr. Brad Stanfield’s rapamycin and exercise study has now been fully funded, with help from various donors, including VItaDAO with a 50k USD contribution.

Obesity drugs have another superpower: taming inflammation

CAR T Therapy Lowers Senescence, Improves Health in Mice

Life Biosciences Presents Groundbreaking Data at ARVO Demonstrating Restoration of Visual Function in Nonhuman Primates

Resources

Biolearn, an open-source biomarker validation library and platform enabling easy and versatile analyses of biomarkers of aging data. 

Biolearn is a tool that simplifies the analysis of biomarkers of aging data. It allows users to easily import data from publicly available sources like the Gene Expression Omnibus, National Health and Nutrition Examination Survey, and the Framingham Heart Study. Biolearn also includes reference implementations for common aging clocks such as the Horvath clock and DunedinPACE, which can be run with just a few lines of code. 

Join Biolearn Discord.

New Book review: How We Age: The Science of Longevity Coleen T. Murphy

LONGEVITY TV SERIES AIMS TO SHED LIGHT ON THE FUTURE OF LIVING LONGER AND HEALTHIER LIVES

Human Ageing Genomic Resources: updates on key databases in ageing research 

Prizes

Longevity Prize Million Molecule challenge with Ora Biomedical

Propose interventions to extend C. elegans lifespan, with a chance to win a prize for the most effective approach. Ora Biomedical has developed the WormBot-AI platform, combining robotics and AI to study longevity in C. elegans, a model organism for aging research. This technology enables quantitative assessment of lifespan and healthspan on a large scale, advancing our understanding of longevity.

Biomarker Challenge, our open-science competition rewarding folks who can best predict chronological age, survival/mortality and multi-morbidity incidence using biomarker data. Challenge officially begins March 1st.

Developing and validating aging biomarkers for clinical trials is challenging due to data limitations and collaboration barriers between scientists. To address this, Biolearn, an open-source toolset, was created. It harmonizes omics datasets and calculates existing aging biomarkers. See more about Biolearn in Resources section.

Conferences

How should ageing be classified?

Impetus awardee and Longevist curator Barry Bentley announces he will be hosting a consensus meeting with experts from around the world to define the criteria for an ageing-related pathology.

Date: Monday, 19th February, 2024

Contact Emma Short: eshort@cardiffmet.ac.uk

La Jolla Aging Meeting 2024

Date: Thursday, March 7, 2024

Location: Conrad T. Prebys Auditorium, Salk Institute, La Jolla

Registration deadline: Monday, February 26, 202

Tweets of the Month

Karl Pfleger

https://twitter.com/KarlPfleger/status/1736108328743489903

The aging field doesn't discuss enough the key top-level distinction between the 2 main therapeutic paradigms: slow aging or reverse it. I support slowing aging as much better than traditional medicine, but it pains me to see many in the field disparage or ignore reversal & I think there should be more open discussion of this distinction………

Anar Isman

https://twitter.com/IsmanAnar/status/1750383400664621057

Aging is the ultimate waste – a waste of talent, wisdom, genius. Takes decades to master a craft, only to have limited time to apply it. Think of the possibilities if mastery wasn't cut short: Federer dominating courts for 20 more years, Einstein revolutionizing science for another two decades, Aretha Franklin enchanting us with her voice, [insert your favorite luminary] advancing a cause dear to you. Let's challenge aging and unleash our full potential.

Podcasts and Webinars

Longevity & Aging Series

A great list of Aging Podcasts

NUS Medicine’s Healthy Longevity Webinar Series

Unlocking the Secrets of Aging with Prof. Pankaj Kapahi - BCI Podcast

Unlocking the Secrets of Lifespan Extension With Karl Pfleger

Aubrey de Grey: Lifespan Extension Imminent Breakthroughs & AI's Impact | Intelligence Podcast #001

The Sheekey Science Show - Should we be measuring biological age? - Aaron King

The Optispan podcast covering a variety of topics related to longevity and healthspan

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful in helping you to keep up-to-date with all the exciting longevity-related developments. 

This time we leave you with the latest VitaDAO podcast Mitochondria and Aging with Prof. Rudolf Wiesner - VitaDAO Aging Science Podcast

And don’t forget to check our Member’s Portal. New services are added every month!
See you next month!

Further Reading

Autophagy protein ATG-16.2 and its WD40 domain mediate the beneficial effects of inhibiting early-acting autophagy genes in C. elegans neurons

Epigenetic Reprogramming as a Key to Reverse Ageing and Increase Longevity

Early time-restricted eating improves markers of cardiometabolic health but has no impact on intestinal nutrient absorption in healthy adults

Vitamin D3 inhibits p38 MAPK and senescence-associated inflammatory mediator secretion by senescent fibroblasts that impacts immune responses during ageing

Social support and cognitive activity and their associations with incident cognitive impairment in cognitively normal older adults

Implications of stress-induced gene expression for hematopoietic stem cell aging studies

Causality-enriched epigenetic age uncouples damage and adaptation

Prophylactic and long-lasting efficacy of senolytic CAR T cells against age-related metabolic dysfunction

Mitochondrial-derived vesicles in metabolism, disease, and aging

Omega-3 supplementation and outcomes of heart failure: A systematic review of clinical trials

Lipid droplets, autophagy, and ageing: A cell-specific tale

Targeting ageing with rapamycin and its derivatives in humans: a systematic review

The interaction between ageing and Alzheimer's disease: insights from the hallmarks of ageing

Anti-Aging Drugs and the Related Signal Pathways

Cellular senescence in brain aging and neurodegeneration

Welcome back Vitalians. I’m sure that most of you would have by now heard the sad news that renowned Professor Judith Campisi has passed away. In place of our usual interview, this month ...
Read more
Mitochondria and Aging with Prof. Rudolf Wiesner - VitaDAO Aging Science Podcast
February 5, 2024
Awareness
Podcast
Mitochondria and Aging with Prof. Rudolf Wiesner - VitaDAO Aging Science Podcast



In this podcast with Prof. Rudolf Wiesner we talked about the lack of long-term positions at German universities, mitochondria derived vesicles, mitochondrial mutation rates, TWINKLE, the adrenal medulla and mitochondrial quality control more broadly. I would not have expected the adrenal glands to be so important in mitochondrial aging, although in hindsight it should have been obvious why they are.

Prof. Rudolf Wiesner — brief bio

To quote from the CECAD webpage: “The group’s goal is to understand in detail how mitochondrial damage — in particular damage caused by mitochondrial DNA deletions — arises and how it leads to organ dysfunction by failure of single cells in a tissue. The aim is to develop therapeutic approaches to slow the process.”

“Mitochondria contain their own DNA molecules. The mutations that accumulate in many organs, such as the heart, brain and muscle, are mainly deletions, where a part of the DNA strand is missing. These deletions are not evenly distributed — few individual cells show massive accumulation, which leads to a loss of function of those cells. A mosaic of normal and heavily impaired cells develops. This process is accelerated in many aging-associated diseases, such as Parkinson’s disease or cardiac arrhythmia. The group investigates how these mutations arise, why they accumulate in individual cells, and how they influence tissue function.”
https://www.cecad.uni-koeln.de/research/principal-investigators/associated-members/rudolf-j-wiesner

Mitochondria — multifaceted players in aging

These organelles are often called the powerhouse of the cell but, apart from being cliched, this metaphor does not do them justice. If we want to stick with an “industrial” metaphor, the mitochondria are maybe like a manufacturing plant, a port, a power plant and a minor geopolitical center all in one. Given the complex responsibilities of mitochondria it should not come as a surprise that they have diverse failure modes during aging.

For example, during aging we see clonal expansion of point mutations and deletions which leads to tissue mosaicism, meaning that one and the same tissue ends up having stretches of totally different, often more or less dysfunctional, mitochondrial populations.

There is also the classic mitochondrial (mt) free radical theory of aging. In one more modern interpretation of this theory, production of radical oxygen species (ROS) at mitochondria is a constant source of macromolecular damage irrespectively of whether this mtROS production increases with aging or not. This is consistent with and supported by findings of reduced mtROS production (Lambert et al. 2007) and higher membrane lipid stability in long-lived species.

In some aging tissues we observe reduced mitochondrial mass and decreased mitochondrial biogenesis (1). Based on this we would expect increased mitochondrial biogenesis to improve healthspan or lifespan, and, indeed, PGC1a is consistently upregulated in long-lived rodents and overexpression of this gene extends fly lifespan (Tyshkovskiy et al. 2019, Ozkurede et al. 2019, Rera et al. 2011).

Although we do observe decreased mitochondrial mass in some tissues, in other tissues there may be a vicious cycle of defective mitochondria that proliferate out of hand. The latter is a theory which is bolstered by the work of Aiken and McKenzie (Herbst et al. 2022) as well as work by Prof. Doug Turnbull from Newcastle (Vincent et al. 2018).

Some other theories suggest that aging mitochondria lead to increased susceptibility to cell death, and that the mitochondrial network becomes disorganized with impaired fusion-fission dynamics.

And this is just a small sample of mitochondrial aging theories!

What is mitochondrial respiratory chain and electron transport chain (ETC) dysfunction?

In the podcast we talk a lot about ETC-negative cells or ETC-deficiency. By that we mean electron transport chain deficiency. ETC-deficient cells are those that have lost the ability to utilize oxygen for mitochondrial respiration via their ETC and now rely on glycolysis instead.

This is also reflected by the lack of a certain protein, which is called cytochrome C oxidase. This state is also termed COX deficiency and can be visualized by a histologic staining that gives a blue readout. Usually, although not always, mtDNA deletions or mutations will be the cause of this COX- and ETC-deficiency.

Which tissues suffer the most from mitochondrial DNA deletions and why?

We discuss why some tissues accumulate more mtDNA deletions than other tissues. What these tissues have in common is often a source of substantial oxidative stress. This is the case for the adrenal medulla and the substantia nigra, which both accumulate large amounts of mtDNA deletions. The adrenal glands are involved in the metabolism of adrenaline, as the name implies, which is structurally related to dopamine (both are catecholamines). The metabolism of dopamine is known to promote the formation of toxic intermediate products that can harm the mtDNA in the brain so it should be no surprise that metabolism of adrenaline is also problematic.

The group of Rudolf Wiesner discovered these abnormalities in mice carrying a relatively mild mutation in a protein that is involved in mitochondrial maintenance, called TWINKLE. It stands to reason that humans will show the same defects during aging, although this awaits confirmation. Mild TWINKLE mutants are generally considered a decent model of age-related mtDNA deletion formation.

“it’s scary if you look at an old adrenal gland…it looks like.. it is completely destroyed full with cells that have a mitochondrial defect.” (Rudolf Wiesner)

We both agreed that it would be useful to perform more autopsy studies of very long-lived individuals, as we still know little about tissue specific pathologies of the very old. To paraphrase:

“good old fashioned descriptive biology is always the basis to find something new”

Can we cure mitochondrial disease?

I asked an intentionally provocative question on this podcast that I like to pose in the context of mitochondrial research. Some time ago I went to a longevity conference, where I saw a talk by Dr. Anu Suomalainen-Wartiovaara, a respected mitochondrial expert talking about her work. During her talk she said that she wants to cure mitochondrial disease. Of course, no one disagreed nor batted an eye.

What I found rather puzzling about this was that people (working outside of our field) would be surprised, even upset, if an aging researcher said they wanted to cure aging or stop aging, to use less controversial language.

For a long time indeed, it was impossible for a respected longevity researcher to say, or to admit, that eventually, down the line, someday, they really would like to stop all aging. End it for good. Luckily this is finally changing, but not fast enough for my liking.

Curing mitochondrial disease is a far-fetched dream, but if mitochondrial researchers are allowed to dream, so are we.

If you ask me, we should all follow in the footsteps of Anu and boldly say that we want to end or reverse aging. If she can say so about her condition of choice, we are allowed as well.

References and further reading

Herbst, Allen, et al. “Age-and time-dependent mitochondrial genotoxic and myopathic effects of beta-guanidinopropionic acid, a creatine analog, on rodent skeletal muscles.” Geroscience (2022): 1–13.

Ozkurede, Ulas, and Richard A. Miller. “Improved mitochondrial stress response in long‐lived Snell dwarf mice.” Aging Cell 18.6 (2019): e13030.

Tyshkovskiy, Alexander, et al. “Identification and application of gene expression signatures associated with lifespan extension.” Cell metabolism 30.3 (2019): 573–593. webpage: http://gladyshevlab.org:3838/Gentervention/

Rera, Michael, et al. “Modulation of longevity and tissue homeostasis by the Drosophila PGC-1 homolog.” Cell metabolism 14.5 (2011): 623–634.

Lambert, Adrian J., et al. “Low rates of hydrogen peroxide production by isolated heart mitochondria associate with long maximum lifespan in vertebrate homeotherms.” Aging cell 6.5 (2007): 607–618.

Vincent, Amy E., et al. “Subcellular origin of mitochondrial DNA deletions in human skeletal muscle.” Annals of neurology 84.2 (2018): 289–301.

In this podcast with Prof. Rudolf Wiesner we talked about the lack of long-term positions at German universities, mitochondria derived vesicles, mitochondrial mutation rates...
Read more
VitaDAO Letter: 2023 in Review
January 30, 2024
Sarah Friday
Awareness
Longevity
VitaDAO Letter: 2023 in Review

What an extraordinary year it has been! Since June 23rd, 2021, the day VitaDAO’s Gnosis Auction funded our Treasury, VitaDAO has been funding longevity research and fostering a community of passionate researchers, enthusiasts, and professionals. Fast forward to today, and VitaDAO has become a driving force in the field, achieving remarkable milestones and setting new standards. 2023 witnessed a successful funding round that garnered support from influential partners such as Pfizer Ventures, contained an unprecedented VITA-FAST token sale, and furthered the evolution of VitaDAO as a key player in the DeSci arena.

For those of you who are new here, VitaDAO is a community-owned collective dedicated to funding and advancing longevity science research. VitaDAO is divided into three working groups (Longevity Dealflow, Community and Awareness, and Coordination) that call upon smaller “squads” as needed. In this newsletter, find brief recaps of some of the work that VitaDAO’s Working Groups and Squads accomplished in the last year. Sit back, relax, and enjoy the read!

🧬Longevity Dealflow Working Group

Throughout 2023, the Dealflow Working Group tirelessly sourced and evaluated projects. Notably, through VDP-106, the DAO set forth a new standardized process to better screen proposals presented to the DAO. In total since its initiation, VitaDAO has evaluated over 200 projects, funded 20 projects, and deployed over 4 million dollars! Among these, 14 projects received funding in 2023:

  1. BE Therapeutics — Brain Tissue Replacement — BE Therapeutics is aiming to develop technology that can engineer functional brain tissue to replace the tissue impaired by age-related damage.
  2. Humanity — Proprietary Aging Score App — Humanity is an app that uses wearable technology and quantified-self methods to enable anyone with a smartphone to measure their rate of aging and it uses AI to find out what lifestyle actions and more advanced interventions each type of user should take to slow it down.
  3. Etheros — Fullerene Chemistry for Longevity — Etheros is a biotech company pioneering a new class of small molecule drugs based on Nobel Prize-winning fullerene chemistry. Their lead compound, which mimics enzymes called superoxide dismutases that protect cells from oxidative and inflammatory injury, has already generated promising efficacy data in animal studies.
  4. HDAX Therapeutics — Histone Deacetylase Targeting Drug Discovery — HDAX is a preclinical-stage drug discovery company targeting a class of proteins implicated in neuropathies and inflammation with novel small molecules having potentially superior drug properties.
  5. Zoe Biosciences — PAI-1 Antibody Therapeutics — Zoe Biosciences has two assets with the potential to extend human healthspan. The first program contains PAI-1 biologic inhibitors with high target selectivity and affinity, relevant for diseases including elevated FGF23 syndromes and metabolic disorders. The second program contains APJ small-molecule agonists for similar age-related indications.
  6. Matrix Bio — Long-lived Species Inspired Longevity Biotech — VitaDAO and the Gorbunova Lab are launching Matrix Bio, a cutting-edge research venture leveraging the anti-cancer and pro-longevity effects of high molecular weight hyaluronic acid from naked mole rats to humans.
  7. ImmuneAge Bio — Rejuvenation of Aged HSCs — ImmuneAGE Bio is the first drug discovery platform company focused on immune system rejuvenation. ImmuneAGE’s lead program, IA101, is a non-toxic small molecule that can rejuvenate aged HSCs and aged animal immune function with higher efficacy than any molecule previously published.
  8. ExcepGen Inc — RNA therapeutics for longevity — ExcepGen is developing a new generation of vaccines and therapeutics with an RNAx platform.
  9. ARTAN Bio — Mutation-Specific Codon Suppression for Aging and Longevity — ARTAN is a biotechnology company developing first-in-class interventions to tackle the most frequent nonsense mutations implicated in a wide range of age-related diseases and cancers. These nonsense mutations induce premature stops of protein translation when occurring in coding regions.
  10. Cyclarity — Novel Cyclodextrin Molecules for Multiple Aging-related Diseases — Cyclarity Therapeutics is an early-stage biotechnology company developing computationally designed novel cyclodextrin drug molecules for the extraction of toxic biomolecules that accumulate with age and are implicated in a variety of age-related conditions.
  11. Reversing Periodontal Disease using Geroscience — An Lab — Johnathan An’s Lab proposes to use small molecule inhibitors of the PI3K/NFkB/mTOR pathway to treat periodontal disease. By targeting inflammation in age-related periodontitis, they hope to find a geroscience-based treatment.
  12. Oisín Biotechnologies — Pioneering Genetic Medicines for Sarcopenia — Oisín Biotechnologies is a multi-asset longevity biotech company pioneering genetic medicines to combat sarcopenia and other age-related diseases to promote healthier, longer lives.
  13. Remedium Bio- Gene Therapy for Regenerative Medicine — Remedium Bio is a regenerative medicine biotechnology company, that has developed the only dose-adjustable gene therapy platform technology Prometheus™ and is currently advancing multiple assets with uncorrelated risk to Investigational New Drug (IND) approval.

Other Notable Dealflow Achievements

  • Through June’s token sale of $VITA-FAST tokens, token holders were given the ability to directly influence longevity research for the first time in history. The $VITA-FAST token was created to give holders exclusive control over the licensing function of the Newcastle-Korolchuk IP and governance over the direction of the research and development of that IP. The sale was wildly successful, concluding with over 1700% oversubscription.
  • VitaDAO made history as it was the first DAO to kickstart a biotech company. This company, MatrixBio, is leveraging high molecular weight hyaluronic acid (HMW-HA) for cancer and age-related diseases.
  • VitaDAO launched “The Longevist,” an overlay longevity research journal. Every quarter, a shortlist of preprints has been selected by the VitaDAO community and voted on by Longevist Curators.
  • VitaDAO played a strategic role in Zuzalu’s inaugural event, a pop-up city in Montenegro. In the capacity of a co-organizer and representative authority on longevity, VitaDAO hosted a two-day longevity biotech conference. Here, leading experts shared insights into cutting-edge research in longevity science and rejuvenation technology. Topics spanned a broad range of disciplines, including Cellular Reprogramming, Cell Replacement, Immune System Rejuvenation, and DNA Damage.
  • Of note this year, through VDP 107, Laurence Ion stepped down from the role of Longevity Dealflow Working Group Steward. The community voted to place Eleanor Davies as Interim Dealflow Steward.

🗣️Community and Awareness Working Group

With a robust Dealflow Working Group, the Community and Awareness team had its work cut out! Alex Dobrin’s re-election as the Community and Awareness Working Group Steward set the stage for a packed year. The year started off strong with PR efforts surrounding the announcement of VitaDAO’s $4.1 million fundraising round. VitaDAO continued to spread awareness globally, as representatives from VitaDAO attended over a dozen conferences on four continents. These meetings included, but are not limited to, DeSci London, DeSci Singapore, DeSci Berlin, Penn Blockchain, DeSci Japan, Longevity Summit Dublin, Longevity+DeSci Summit NYC, the 10th Aging Research and Drug Discovery Meeting, and Token 2049 Singapore. The VitaDAO buzz reached new heights when Balaji shared our story with his million followers on Lex Fridman’s podcast.

Continuing community education and news dissemination remained a top priority. VitaDAO’s Monthly Community Newsletter celebrated its two-year anniversary and VitaDAO’s Monthly Longevity Research Newsletter celebrated one year in existence. Adding to the excitement, VitaDAO launched The Aging Science Podcast. The podcast, released monthly, features deep conversations with many leading experts in the field of longevity.

Past initiatives were continued in 2023 including VitaDAO’s collaboration with Jellyfish and VitaDAO’s involvement with the VitaDAO Longevity Prize, a collaboration with Foresight Institute and Methuselah Foundation. This April 2023, Hypothesis Prize winners were announced. More recently, VitaDAO partnered with X-Prize on their XPRIZE Healthspan initiative promising a groundbreaking 7-year competition with a $101 million prize pool.

Other awareness initiatives included the funding of another cohort of fellows within VitaDAO’s Longevity Fellowship. The fellowship is composed of deed-based grants awarded to individuals to cover expenses such as conference fees, research funding, and longevity program tuition. In total, VitaDAO has funded over 65 fellows with over $65,000. VitaDAO also continued to engage its community through the Ambassador Program. This program aims to facilitate community growth and engagement through the organization of in-person meet-ups.

As if that wasn’t enough news, VitaDAO hosted its 3rd VitaDAO DeSci & Longevity Symposium. And, VitaDAO hosted its first VitaDAO hackathon! This successful event boasted three winners: Hack-Age (1st), AdStella (2nd), and AGen-iNET (3rd).

Other notable highlights include the release of a Longevity Briefing Primer to define longevity, highlight VitaDAO’s role in the field, and make the case for special economic zones. Maria Marinova posted a series of articles on VitaDAO’s blog exploring the potential of optogenetics to combat aging, embryogenesis and its potential role in longevity, and the mechanisms by which sugar byproducts may impact aging. Lastly, VitaDAO’s Twitter fostered good conversation in the form of multiple Twitter Space interviews and discussions. These include but are not limited to, a discussion on the latest advancements in robust mouse rejuvenation, an interview with Sergey Young, an open discussion on the future of longevity & De-sci, and a discussion on the latest in epigenetic clock research.

Our community outreach numbers, as of January 2023, tell an impressive tale: (see numbers, updated as of January 2023, below):

  • + 3,400 Token Holders
  • + 10,000 Discord Members
  • + 22,000 Twitter followers
  • + 2,100 Youtube Subscribers

🔏Coordination Working Group

In 2023, VitaDAO was recognized by CoinGecko as one of the top crypto projects in Europe. Operating a DAO, and operating it successfully, is no easy task! Todd White, the re-elected Steward, led the working group as it continued to improve financial reporting, ensure accountable use of VitaDAO’s resources, and implement changes to the DAO infrastructure.

Key additions to the VitaDAO infrastructure include the creation of a network steward role and the launch of the member services squad and VitaDAO Global platform for members. The member squad seeks to increase the $VITA token utility value and help VitaDAO members pursue their personal longevity journeys.

Another large change implemented by the Coordination Working Group was the development of an operational company for the execution of the Coordination Working Group functions on behalf of VitaDAO. This for-profit cooperation provides service provider contract management, executes the VitaDAO’s member services program, and more!

This year, the Coordination Working Group also worked with legal counsel to prepare VitaDAO for issuing tokens under the new EU Markets in Crypto-Assets (MiCA) Regulations. Going forward, starting in June 2024, all IP Tokens issued from our IP-NFTs as well as our own VITA token must be compliant with the MiCA Regulations. VitaDAO is on track to be fully compliant by the June 2024 deadline.

The Coordination Working Group also closed 2 funding events for VitaDAO — the $4.1m round in January 2023 which included Pfizer Ventures, Shine Capital, and L1D — but also a smaller $1.3m round in December to provide interested parties with VITA tokens prior to the launch of the VitaDAO Accredited Investor Fund (VDAIF) anticipated in Q1’2024. Another $5.4m which brings us to a total of $10.5m raised since inception.

While the Coordination Working group is often behind the scenes, they are an imperative component to VitaDAO’s success. Early in the year, they played a role in token-gating Discord such that different levels of Discord are unlocked with different quantities of $VITA. More recently, they have assisted with the migration to Parcel for payouts and created a dashboard to create a better payout experience for contributors.

👾Tech/Product + Tokenomic Squads

As mentioned above, the year started with a successful funding round. This round required the minting of an additional 10% $VITA for strategic contributors. Following the 10% $VITA mint, the individuals in the DAO squads worked to allocate 6% of the total $VITA token supply to new strategic contributors. This minted allowed onboarding of new strategic members such as Pfizer Ventures, Shine Capital, and L1 Digital; decentralized science and web3 organizations like Beaker DAO and Spaceship DAO; and longevity enthusiasts including Balaji Srinivasan and Joe Betts-LaCroix. Additional technical challenges addressed by the DAO included the fractionalization of the Newcastle-Korolchuk IP-NFT and the coordination of the crowd token sale. The Tokenomics Squad also assisted with launching Vita on Optimism and Polygon. This change decreased the burden of gas fees for community members!

In 2023, the Tech/Product Squad facilitated the launch of a VitaADO member portal (vitadao.global). This portal enables VitaDAO holders to claim exclusive perks such as reduced medical testing, discounts on health products, and discounts on health insurance for digital nomads. Additionally, the team continued to improve the VitaDAO website, created a VitaDAO treasury dashboard, and designed a merch shop. The treasury was created to foster greater transparency of VitaDAO’s economics. And the merch shop features a cool hoodie!

What an extraordinary year it has been! Since June 23rd, 2021, the day VitaDAO's Gnosis Auction funded our Treasury, VitaDAO has been funding longevity research and fostering a community of passionate researchers, enthusiasts & professionals.
Read more
Exploring DNA Repair for Longevity with Prof. Björn Schumacher - The VitaDAO Aging Science Podcast
January 16, 2024
Podcast
Awareness
Exploring DNA Repair for Longevity with Prof. Björn Schumacher - The VitaDAO Aging Science Podcast

In this episode of the Aging Science Podcast by VitaDAO I had the pleasure of talking with Prof. Björn Schumacher (@schumacherbj) about his recent work identifying a novel way to boost DNA repair. I found this work unusual in many ways: not only is it urgently needed but he also identified a rather strange protein, with a strange name, as a player in DNA repair using a model species whose aging is not even particularly dependent on DNA repair! Wow.

Short bio

Since 2013, Björn Schumacher is full professor and director of the Institute for Genome Stability in Ageing and Diseases (IGSAD) at the CECAD Research Centre of the University of Cologne. 

Professor Schumacher is President of the German Society for DNA Repair (DGDR), co-Director of the Minerva Center of the Biological Mechanisms of Healthy Ageing at Bar-Ilan University (IL), and between 2014 and 2020 served as President of the German Society for Ageing Research (DGfA). 

His research interest focuses on the molecular mechanisms through which DNA damage contributes to cancer development and ageing-associated diseases.  Employing the C. elegans system and mammalian disease models, his group uncovered cell-autonomous and systemic responses through which the organism adapts to accumulating DNA damage with ageing. Through the understanding of the basic mechanisms of genome instability-driven ageing, Schumacher aims to contribute to the development of future strategies to prevent ageing-associated diseases.


Björn Schumacher, webpage: https://igsad.de

How important is DNA damage to aging?

We know that DNA is important because every cell has exactly two copies (or alleles) of a gene. If one or both or damaged through DNA mutations this can have severe consequences since all mRNA and protein is produced based on this DNA blueprint. Hence, cells have evolved a very complex machinery of DNA repair. This complexity has limited our ability to study and understand DNA repair for a long time.

While mutations in DNA repair proteins accelerate death and certain aspects of aging – causing progerias – so far no one has managed to extend lifespan by increasing DNA repair, it is not even clear if anyone has successfully and consistently managed to increase DNA repair to begin with. The main issue, just as with oxidative stress defenses, is that boosting a single function might have no impact or worse harm the whole system by causing an imbalance in the ration of different components!

However, there is very strong indirect evidence for the role of DNA repair in aging. Björn points out recent studies that allowed us to better quantify somatic mutations. These mutations are so heterogeneous that until recently we lacked the techniques for accurate measurements. These newer studies show convincingly that mutation rates are lower in short-lived species. Some of these we discussed in our Vera Gorbunova podcast.

In the end whether DNA damage is “just” a driver of cancer or also causes other age-related diseases is a moot point. We still lack tools to effectively prevent caner and we will need them to extend healthy human lifespan. Therefore, tackling DNA damage and mutations downstream of damage remains one of the most important tasks ahead.

The many types of DNA damage

So far, we used the word DNA damage or DNA damage theory quite liberally. However, experts in the field like to distinguish between DNA damage and DNA mutations. One usually leads to the other, but not always. This can be important. Some species like the naked mole rate seem to have low mutation rates but high rates of transient DNA damage – this is still consistent with the DNA damage theory of aging. Ultimately the only thing that matters is the final, unrepairable type of damage: the mutation. Perhaps, as I suggested during the podcast, the word DNA damage theory could be confusing.

Conversely, making matters even more complicated, unrepaired DNA damage can lead to senescence or other issues without causing mutations. However, most DNA damage is not so severe.

In fact, DNA is constantly damaged and this damage is usually repaired efficiently. Some types of DNA damage are considered less toxic, like the famous pyrimidine dimers caused by ultraviolet radiation, single strand breaks, or nucleotide changes. These can induce harmful mutations but the damage is often very small or localized, like a single nucleotide change.

Much worse are crosslinks or double strand breaks. Interstrand crosslinks, for example, block the separation of DNA strands, which is essential for replication and transcription. Imagine your DNA being torn to pieces during replication. This cannot be good. Some cancer therapeutics induce exactly these kinds of mutations to destroy quickly dividing cancers! Similarly, double strand breaks. Perhaps temporarily not a problem but once the cell starts dividing it would lose whole stretches of DNA since the broken parts will not properly attach to the spindle that pulls replicated DNA apart to divide evenly across the new cells. We are dealing with the loss of hundreds of genes and not just single mutations. 

Now imagine there was a way to boost every single type of DNA repair in a balanced and natural way. Maybe we have found it.

The DREAM complex

This large multiprotein complex is involved in cell cycle regulation and repression of DNA repair. Now that makes it quite unusual since one would hardly expect the cell to turn off something as beneficial as DNA repair, would we? As Björn explains in the podcast, everything comes with a cost. It would be wasteful for a worm to boost DNA repair in their body cells to allow a few worms a longer life, when it would cost energy or time that could be invested in producing more eggs, or producing eggs faster. Most worms die early and do not need good DNA repair. If they are exposed to genotoxic stressors they prefer to increase stress resistance mechanisms like antioxidants instead of DNA repair.

Given that DNA damage seems to be a mechanism of aging “private” to long-lived species, i.e. limited to these, it is all the more surprising that Björn’s group discovered the importance of DREAM in C. elegans. It takes a lot of ingenuity and persistence to walk such a tortuous road towards discovery. However, this might have been the right call if, as Björn says, identifying transcriptional regulators of DNA repair was much easier in the worm. From there he just needed to assume that these mechanisms are conserved in higher mammals and get a bit lucky.

The basic idea was fascinating. As it turns out, the germline has 10-fold lower mutation rates than somatic tissues, making it an elite tissue. It stood to reasons that there should be an activator of DNA repair in the germline or an inhibitor of DNA repair in somatic tissues. Since we know that many important age-related pathways are regulated transcriptionally, e.g. multistress resistance via Nrf2 or autophagy via TFEB, it was an obvious choice to look for a transcriptional regulator. It turned out to be DREAM, which was aptly named that way by other researchers who had been studying this one for its role in cell cycle progression and differentiation.

The future of DREAM

Björn and others have identified drugs that repress this repressor of DNA repair. Which means they can activate DNA repair. This is one of the big breakthroughs that we need to finally test the DNA damage theory experimentally. I am optimistic that we are getting closer and that we will eventually find a way to increase DNA repair. Nevertheless, to temper our optimism we have to realize that DREAM has many functions, DNA repair is only one of them. As Björn noted, the whole system is even more complicated in humans than in worms. In the worst-case inhibiting DREAM could lead to unwanted side effects. However, this is only speculation one way or another. The only way to find out is to test existing and novel DREAM inhibitors in mice to see whether they are well tolerated and extend lifespan.

It may be a bumpy road but eventually we will get there. All the best to everyone working in this field!

The host – brief bio

Kamil Pabis, MSc is an aging researcher and longevity advocate with several years of experience in the aging field that spans multiple countries. Among other projects, Kamil worked on long-lived dwarf mice in Austria, on mitochondrial disease and aging in the UK, and finally on the bioinformatics of aging in Germany and Singapore. Presently, he is involved in several projects related to science communication and translational aging research.

References and further reading

Bujarrabal-Dueso, Arturo, et al. "The DREAM complex functions as conserved master regulator of somatic DNA-repair capacities." Nature Structural & Molecular Biology 30.4 (2023): 475-488.
https://www.nature.com/articles/s41594-023-00942-8

Telomeres vs DNA damage - battle of the aging theories
https://biogerontolgy.blogspot.com/2022/03/telomeres-vs-dna-damage-battle-of-aging.html

Read more
December Longevity Research Newsletter
January 11, 2024
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
December Longevity Research Newsletter

Introduction

Welcome to 2024, Vitalians! We hope your holiday season was wonderful, and that you're returning recharged and ready for another year of exciting developments in longevity science! 

I know it seems so far away already but here is what happened in December, plus an insightful interview from Dr. Bernadette Carroll - who shares her thoughts on the longevity field as well as discussing her research on nutrient sensing, lysosomes and their role in senescence and aging, enjoy!

Longevity Literature Hot Picks

Preprint Corner in collaboration with

The Longevist is an overlay journal spotlighting the most promising longevity studies each quarter through a collaborative and transparent process, leveraging insights from a panel of global experts in the longevity space.

With Q4 now over, The Longevist editors are currently finalising the shortlist of preprints to put forward to our curators for the next on-chain vote. In the meantime, check out the 3rd issue of The Longevist and look out for our newly assigned ISSN!

Check out these latest preprints, each of these will be entered into the Q1 2024 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

Sex-specific growth and lifespan effects of germline removal in the dioecious nematode Caenorhabditis remanei

The germline regulates longevity and somatic repair in a sex-specific manner

Epigenetic Clocks and Programmatic Aging

A Fully-Automated Senescence Test (FAST) for the high-throughput quantification of senescence-associated markers

Epistemic uncertainty challenges aging clock reliability in predicting rejuvenation effects

Extensive remodeling of the ubiquitination landscape during aging in C. elegans

The molecular landscape of premature aging diseases defined by multilayer network exploration

From Churchill to Elephants: The Role of Protective Genes Against Cancer

Published Research Papers

DNA methylation rates scale with maximum lifespan across mammals

A new statistical framework was developed to compare DNA methylation rates at conserved age-related sites across mammals, revealing a negative correlation with maximum lifespan in both blood and skin tissues. This indicates methylation rates might be a constraint on maximum lifespan, affecting diverse mammalian lineages.

Organ aging signatures in the plasma proteome track health and disease

Analysis of organ-specific aging in humans using blood plasma proteins, finding significant variations in aging rates across different organs and predicting increased disease risks. The approach offers a novel method for assessing individual aging processes and their impact on health, using plasma proteomics data.

Iron accumulation drives fibrosis, senescence and the senescence-associated secretory phenotype

Iron accumulation is a key factor in cellular senescence and fibrosis in mice and humans, suggesting iron metabolism as a potential target for treating senescence-associated diseases. It also highlights the possibility of using magnetic resonance imaging to non-invasively assess fibrotic diseases.

Aged intestinal stem cells propagate cell-intrinsic sources of inflammaging in mice

This research explored inflammaging in the mouse intestinal epithelium, discovering that aged intestinal stem cells (ISCs) play a key role in this process by upregulating major histocompatibility complex class II genes and exhibiting an intrinsic inflammatory memory. It highlights that inflammaging is driven by a cell-intrinsic mechanism, dependent on STAT1 signaling, and leads to a disruption in immune homeostasis.

The effect of a ketogenic diet and synergy with rapamycin in a mouse model of breast cancer

A ketogenic diet, compared to standard mouse chow, effectively reduces breast tumor growth and increases lifespan in a mouse model, particularly when combined with the anti-cancer drug rapamycin. 

Transcriptomes of aging brain, heart, muscle, and spleen from female and male African turquoise killifish

The study introduces a detailed RNA-seq dataset from multiple organs of the African turquoise killifish, highlighting that age significantly affects gene expression more than sex. This resource is pivotal for investigating aging and sex-related gene expression in this short-lived vertebrate model, especially noting the increased expression of transposable elements in the brain with age.

Published Literature Reviews, Hypothesis, Perspectives and more

The Information Theory of Aging

Biological information is stored in two ways: the genome as a blueprint, and the epigenome which regulates gene expression. The Information Theory of Aging suggests that aging results from losing epigenetic information, and its retrieval through reprogramming could reverse aging.

Mechanisms, pathways and strategies for rejuvenation through epigenetic reprogramming

Recent developments in anti-aging research involve using nuclear reprogramming factors to reverse age-related deterioration in mouse and human models. The study also explores the challenges and potential of epigenetic rejuvenation strategies in practical applications for treating aging and age-related diseases.

Telomeres, cellular senescence, and aging: past and future

The review commemorates over fifty years since Alexey Olovnikov's theory of the end-replication problem, exploring the complex role of telomeres in cellular senescence and aging, and their impact on age-related diseases.

Putting a strain on diversity

There are significant limitations of predominantly using the inbred C57BL/6 mouse strain in aging research, advocating for the diversification of mouse strains to better understand variations in lifespan and healthspan. They emphasize the need to select mouse strains based on specific research questions and an understanding of genetic diversity to bridge the gap in translating findings to human aging.

A step toward precision gerontology: Lifespan effects of calorie and protein restriction are consistent with predicted impacts on entropy generation

A thermodynamic model to predict lifespan changes in mice under calorie and protein restriction finds that increased restriction reduces entropy generation and potentially extends lifespan. This model aligns with precision gerontology goals and could help identify anti-aging interventions.

Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement

The review analyzes recent aging research, focusing on cellular and molecular hallmarks, their links to age-related diseases, and major biochemical processes, aiming to advance understanding of aging mechanisms and challenges.

Job Board

PhD Studentship: Blood-based Biomarkers of Frailty and Ageing

PhD Studentship: Exploring Valid Approaches to Breath VOC Analysis for Diagnostic Research with Prof Alexandra Stolzing

Postdoctoral Position in Genome Stability: The Institute for Genome Stability in Aging and Disease at the CECAD Research Center, University of Cologne. DNA damage in aging and the regulation of the inheritance of stable genomes with Prof Schumacher

The Kohler Mito Lab are recruiting a Post-doc and a PhD student to study Mitochondrial Protein Quality Control Umeå University, Sweden

Interested in cellular senescence? Apply for this fellowship to join Ana O’Loghlen’s lab in Madrid!

The Sandri Lab are hiring - 18 months postdoctoral fellowship to work on developing RNA-based therapies in vivo for modulating signaling pathways involved in protein degradation, bioenergetics/mitchondria and protein synthesis. University of Padova, Italy.

News and Media

$200 Billion in Revenue: How an Aging Drug Will Conquer Pharma

Hevolution Foundation Grant Expands New Investigator Awards in Geroscience

What’s Your ‘Biological Age’?

New tests promise to tell you if you have the cells of a 30-year-old or a 60-year-old. Here’s what to know about them.

Bridging Generations: Our Ageless Future

Longevity 2023: From A to…

Aging research comes of age

Turn Bio, backed by the VitaDAO Community, achieves a milestone in RNA therapeutics by successfully delivering mRNA to the skin, a significant step beyond liver-targeted therapies

Not all organs age the same. ‘Older’ ones may predict your risk of disease

The RAPID Clinical Trial are recruiting volunteers to take part in a clinical study to treating periodontal disease by targeting mTOR

Death Becomes Her, or Why Aging is an Epigenetic Program

Insilico Medicine announces AI drug discovery milestone achievement in pharma collaboration

Venture Capital Giants In The Billion-Dollar Quest For Longevity Breakthroughs

Frontiers in Aging are accepting submissions on the research topic “Longevity Worldwide: A Collection on Centenarian Studies” until 31st January

The Biggest Breakthrough in Longevity may Start with Menopause

Light color is less important for the internal clock than originally thought, study finds

Conferences

Cologne Aging Conference 2024

Jan 29-30, CECAD Cologne

Young ICSA 18th & 19th March

Oriel College Oxford - Register here by 19th January

Vitalia.city has officially launched. It will host 4 bi-weekly conferences on different topics, 2 of which will be on longevity:

Longevity & Human Improvement (Jan 15 - 21, Summit: Jan 20)

  • The Bioscience of Longevity
  • Business Models for Longevity Companies
  • Economics and Incentives of Healthcare Systems
  • Philosophical Views and Ethics of Life Extension 

Startup Societies & Crypto Cities (Jan 29 - Feb 4, Summit: Feb 3)

AI & Technological Progress (Feb 5 - 11, Summit: Feb 10)

Pathways to Life Extension (Feb 19 - Feb 25, Summit: Feb 24)

  • New Drug Development
  • Clinical and Healthcare
  • Biohacking and Self-Improvement
  • Incentive Design Solutions

Tweets of the Month

From Artemy Shumskiy, what do you think?

Aging hallmark I dislike: telomeres

Aging hallmark I begrudgingly respect: splicing dysreg

Aging hallmark I think is overrated: senescence

Aging hallmark I think is underrated: altered intercell comm

Aging hallmark I like: inflammation

Aging hallmark I love: epi alterations

Some pearls of wisdom from The Terminator (Arnold Schwarzenegger)

I heard that the way you go viral on this site is by making a big list of things you have to do. Let me try.

-You should mostly eat food you know is healthy, there is no magic food.

-You should also occasionally let yourself eat delicious food you know isn’t healthy. Otherwise what’s the point?

-You should be training with some kind of resistance (your bodyweight works), no matter your age

-You should do something to get your heart going and get a schvitz a few times a week

-You should know there isn’t a magic pill, a hack, or a diet and most of that crap people put in their lists is just mean to confuse you so you pay them to figure it out for you with whatever they are selling

-You should also know my daily newsletter is free, is allergic to bullshit, and includes weekly workouts, the latest health and fitness news, and motivation from me.

-Don’t worry, if you paid one of those people to tell you jumping in cold water or taking 29 pills every morning will get you fit, I’ll still be here in three months when you decide it’s time to try something that works. The only trick I know is that you have to work, it hasn’t changed in 76 years and it won’t change.

Podcasts and Webinars

Longevity & Aging Series

NUS Medicine Healthy Longevity Webinar

Cellular Senescence and Mitochondria | Prof Joao Passos

Gene Therapy Insights Podcast

Energy Replacement in the Light of Aging with Dr. Shahaf Peleg

The Desci Podcast by Molecule

Keith Comito's Insights on Aging Research and Lifespan Extension

The Sheekey Science Show

Can taurine and vitamin B12 improve your health?

Interview with Dr. Bernadette Carroll

After completing her PhD at Imperial College London, Dr. Carroll worked in Viktor Korolchuk’s laboratory at the Newcastle University Institute for Ageing and Health where she published numerous papers on nutrient sensing, mTOR, autophagy and their role in senescence and aging. She now runs her own research laboratory at The University of Bristol, focusing on mechanisms controlling the spatial regulation of nutrient homeostasis in health, senescence and cancer. She has recently published a paper discovering a key mechanism of how lysosomal biogenesis is required for cell survival during the onset of senescence.

What inspired you to enter longevity research?

I will admit that my entry into longevity research was not entirely by design. I originally trained as a cell biologist, and I have always been interested in the spatial organisation of our cells, asking questions such as how and why are proteins recruited to specific sites in the cell, and what happens if this localisation becomes dysregulated? For my postdoc, I moved to Newcastle University to work in the lab of Dr Viktor Korolchuk, a key contributor to the VitaDAO community. My project was focused on understanding how the localisation of the main regulator of cell growth, called mTOR is controlled. Over time, through research seminars, coffee chats and a pint (or several), I was won over by the enthusiasm and passion of researchers in the longevity field. It was well established in the field that mTOR signalling drives several senescence phenotypes, and so I got onboard and used my expertise to take a closer look and explain in more detail exactly how mTOR is dysregulated in senescence.

Which of the current theories of ageing do you think are the most convincing?

I see immense value in developing theories of ageing. As an experimentalist who relies on a bottom-up approach, these theories give us new ideas to test using hypothesis-based science. If we can rescue senescence phenotypes by identifying and modifying dysregulated mechanisms, this gives us hope that one day we will be able to develop new anti-ageing interventions. Currently, I particularly favour programmatic theories rooted in the development, where processes like mTOR play an important role.

How has the field changed since you started?

The field has become a lot more mainstream. There is increasing appreciation that a comprehensive understanding of all aspects of fundamental and applied biology is required to explain the ageing process. This is undoubtedly a positive, there is more interest from the public, from governments and from biotech industry which has led to increased, dedicated funding, increased initiatives to promote collaboration and more research teams focused on longevity. This increased focus however also presents its own challenges as we come to fully realise the complexity of understanding longevity, integrating topics from biochemistry, to physiology, psychology, and sociology. I think it will require some out-of-the-box thinking going forward to tackle longevity research effectively and collaboratively. That’s why communities like VitaDAO are so important.

What mistakes do you think the longevity field has made?

Ageing is a complex, multi-factorial and multi-scale process and I think the longevity field is guilty of approaching the question from a reductionist angle. This is a natural result of the short-term funding strategies of most national and international funding schemes that don’t necessarily support high-risk or more preliminary projects. While labs working on model organisms such as yeast, flies and mice have been true trailblazers in identifying key signalling or metabolic pathways involved in longevity, translating these findings to humans has been very limited. Instead, we are still at the stage of generating theories of ageing, essentially a thought process. The next few decades however promise an explosion of fundamental, translational, and clinical research in longevity which will inevitably push the field forward in a practical way.

Other than your own, what do you think have been the biggest/important discoveries in the field?

As cell biologists, we work exclusively with human cells that have been removed from their natural environment in the body and are instead exposed to different nutrients and stressors. There is always the worry therefore that the many of the phenomenon’s we study could be artifacts that have little or no physiological relevance. That is why the seminal work from the Mayo clinic demonstrating that the selective clearance of p16-positive senescent cells can alleviate ageing phenotypes was fundamentally important for researchers working on senescence.

What advice would you give to people currently working in longevity research?

Collaborate. I believe the biggest rewards will come from those who can step out of their comfort zone to embrace different approaches and ways of thinking about longevity. Also keep an open mind about new potential directions. Getting involved with communities like VitaDAO is a really positive move to facilitate collaborations and generate new ideas.

Your research focuses on the interplay between nutrient homeostasis and cellular processes in the context of aging and cancer. What inspired your interest in this area?

We are what we eat, and as such I believe that a lot of the answers about how well we age and how likely we are to develop age-related diseases lies in our diets and in the way our bodies are able to handle nutrients. There are so many intriguing questions in this area, both in terms of fundamental mechanisms of nutrient sensing as well as in terms of practical applications. For example, despite decades of nutritional research, we are still unclear about how to combine the beneficial effects of dietary restriction with the need to maintain active and fit bodies, especially in old age. In the lab, we are developing cell- and tissue-based models of human ageing and believe that these will provide new answers to the long-standing questions in the field.

mTOR is often referred to as a master regulator of cell growth and metabolism and heavily implicated in the aging process. You previously published work showing that mTORC1 activity can be regulated by the amino acid arginine, through control of the TSC2-Rheb signalling pathway. Could you summarise these findings?

Several decades of research have revealed that the activity of mTORC1 is controlled via its subcellular localisation in the cell. Specifically, the availability of specific amino acids is sensed to dictate whether mTORC1 is localised to the cytoplasm or to the surface of the lysosome (the degradative compartment of the cell). When I started my postdoc, the mechanisms via which leucine and glutamine controlled mTORC1 had been identified but we found over and over again in our cell system, that arginine was having a very strong effect on mTORC1 signalling and cell growth. We identified that rather than regulating the localisation of mTORC1, arginine is important for controlling the localisation of the inhibitor of mTORC1, called TSC. This large protein complex inactivates another protein, called Rheb which is the master activator of mTORC1 signalling. We found that in the absence of arginine, TSC is localised on the lysosome, binds to Rheb and prevents the mTORC1 activation. As such, the mechanism by which arginine is sensed is conceptually different to other amino acids.

Do you think the positive effects of dietary restriction on healthspan/lifespan could be due in some part due to lowered arginine availability, and would this be worth testing? Considering leucine and glutamine have also been shown to affect mTORC1 activity, do you think any other amino acids could also play a role?

The regulation of amino acid uptake, utilisation and synthesis is complicated and very context dependent. There are 20 natural amino acids which are the building blocks of proteins and play key roles in metabolism. Human cells can make 11 of them, the other 9 amino acids, including leucine are referred to as essential amino acids which means they need to be ingested in our diets. Leucine is a particularly potent growth-promoting amino acid; you might have noticed high levels of leucine advertised on protein shakes aimed at gym-goers trying to boost muscle mass. Glutamine and arginine, along with several other amino acids are referred to as conditionally essential, that means our cells can produce them but that at certain times, for example during development or during tissue/wound healing, our cells might not be able to meet demand and diet supplementation is necessary. It’s true that exploiting changes in amino acid metabolism can have clinical benefits in some settings; for example, tumours auxotropic for arginine (i.e. tumour cells that have lost the ability to synthesise their own arginine) can be treated with circulating arginase to degrade circulating arginine and thus essentially starve the tumour cells. However, as of yet, we have not identified any specific defects in senescent cells that would allow us to target them in this manner.

Evidence from fly models would also suggest that starvation of any single amino acids may not yield any discernible longevity benefits, except perhaps for methionine starvation. So rather than any specific amino acid, it’s more likely that caloric restriction has a beneficial effect through a combination of reduced protein synthesis-related stress (less burden on ribosomes and ER folding machinery) and increased levels of autophagy.

Senescent cells have dysfunctional lysosomes and you recently published a paper identifying the mechanism by which senescent cells increase their lysosomal content to cope with this decreased function. Could you briefly explain these findings and what the potential therapeutic implications could be?

Lysosomes are the degradative organelle of the cell and they play an essential role in degrading unwanted, or damaged cellular contents. Despite the fact that an increase in lysosomal content is one of the hallmarks of senescence (measured by senescence-associated ß-galactosidase staining), the driving mechanisms and cellular consequences were poorly understood. We identified that lysosomes in senescence are dysfunctional. Their pH is increased which is important because low pH is required to maintain the activity of the degradative enzymes in the lysosome. As a result, the lysosomes are less active, and less able to degrade cargo. We propose that senescent cells compensate for this reduced lysosome activity by increasing their biogenesis, specifically by activating transcription factors of the TFEB family. Increased lysosome content therefore allows continued degradation of damaged and surplus cellular contents and support senescent cell survival.

What’s next for the Carroll lab?

The work us and others demonstrated that inhibiting lysosome biogenesis or modulating dysregulated mTORC1 signalling can promote senescent cell death, which indicates we could one day be able to develop interventions to target these pathways and promote healthier ageing in vivo. With this ultimate goal in mind, we are continuing to work towards a better mechanistic understanding of lysosomes and their interplay with mTORC1 signalling in senescence. We have some exciting data on changes in lysosomal lipids in senescence coming through, so watch this space :).

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful in helping you to keep up-to-date with all the exciting longevity-related developments. See you next month!

Further Reading

Capsaicin inhibits A7r5 cell senescence via the mitochondrial carrier protein Slc25a12

Senescent skeletal muscle fibroadipogenic progenitors recruit and promote M2 polarization of macrophages

NADase CD38 is a key determinant of ovarian aging

DNA repair-deficient premature aging models display accelerated epigenetic age

Functional-metabolic coupling in distinct renal cell types coordinates organ-wide physiology and delays premature ageing

Successful Aging: The Longer One Lives, the Better One Has Eaten?

NMN: The NAD precursor at the intersection between axon degeneration and anti-ageing therapies

Welcome to 2024, Vitalians! We hope your holiday season was wonderful, and that you're returning recharged and ready for another year of exciting developments in longevity science!
Read more
Aging Insights: Dr. Jesse Poganik and Dr. Mahdi Moqri on Biomarkers and Longevity Research – The VitaDAO Aging Science Podcast
December 18, 2023
Longevity
Podcast
Aging Insights: Dr. Jesse Poganik and Dr. Mahdi Moqri on Biomarkers and Longevity Research – The VitaDAO Aging Science Podcast

In this podcast with Dr. Jesse Poganik (@jpoganik) and Dr. Mahdi Moqri (@mahdi_moqri) we had a chat about their upcoming symposium and the importance of biomarkers. Our topics ranged from omics over to liquid biopsies and clinical studies. We talked about walking speed, epigenetics, longitudinal studies and prizes (yes, you can win money!)

Hope you will enjoy this podcast and our brief shownotes.

Short Bio of the speakers

Dr. Jesse Poganik is an Instructor in Medicine at Brigham and Women’s Hospital and Harvard Medical School. His research focuses on understanding the most fundamental aspects of aging: what is the essence of the aging process, and what features define the biological nature of aging? He leads several projects in the Gladyshev Lab to understand the temporal dynamics of biological age, the inter-cellular and inter-tissue dynamics of aging, and how aging may best be quantitatively assessed.

www.poganik.com

Dr. Mahdi Moqri is a joint Research Fellow in Aging Research at Harvard (Gladyshev Lab, Genetics) and Stanford (Snyder Lab, Genetics). He is leading the executive committee of the Biomarkers of Aging Consortium, to establish reliable biomarkers for longevity interventions and mentoring non-profits and young researchers in omics techniques.

https://www.moqri.com/

Important links and upcoming events

The consortium website with relevant information and news
www.agingconsortium.org

2023 Biomarkers of Aging Symposium
December 4th 2023
https://event.fourwaves.com/boa2023/pages

ClockBase: here you can check whether your favorite dataset, drug or intervention affects epigenetic age
https://www.clockbase.org/

What is a clock and what is a biomarker?

“You know when I was talking to people in Vadim’s lab when I was considering joining they were saying clock, clock, clock and I was just… I had no idea…[and] in many ways clock is not a good term”

Basically, a clock tries to measure biological aging or something related for newer clocks. A somewhat more technical definition would understand the clock as a mathematical algorithm, based on molecular data, that has some relation to biological age.

The problem with the word clock that Jesse alluded to is reflected in current papers. These days many clocks are designed not to predict age anymore, rather they are trained to predict age-related outcomes. For instance, the GrimAge epigenetic clock predicts mortality risk. At this point it might be better to just stick to the word “biomarker” instead of clock.

Even worse, as Mahdi mentioned, we do not want a clock that accurately predicts calendar age in most cases, because then we might as well check your birth certificate. We really are interested in outcomes, which are predicted by your “biological age” so to say — i.e. the overall state and health of a person.

A biomarker is an indirect measure of any biological process. As aging researchers we want this kind of indirect measure for aging. We could use this word instead.

Nevertheless, while inaccurate I like the term clock from a popular science perspective. Everyone understands that “the clock is ticking” towards undesirable outcomes or some well-defined end. The word and the idea captures the imagination of people and this is the reason why the term became so popular.

Clinical studies and data sharing — two pressing issues

We all agreed that biomarkers are needed to facilitate clinical studies. Having a marker that can predict drug success in advance would be a great boon to aging researchers. Although I did caution that even the best biomarkers for other diseases rarely if ever replace large studies with hard outcomes. Nevertheless, such markers can save billions by helping us to choose the best possible interventions to pursue in large studies!

Secondly, we all agreed that there are many reasonable and, unfortunately, also unreasonable obstacles hindering our work on biomarkers: “there are many barriers that make data sharing harder than it should be from a scientific point of view” (Jesse)

Hence, I am looking forward to the work of the Biomarker Consortium to improve datasharing and accessibility.

The importance of functional markers

Although neither of us is a clinician and this is more the purview of doctors, I encouraged the speakers to take a brief detour and talk about functional markers. I remain very excited about functional markers because they are relatively inexpensive. Measuring improvements in walking speed, hair loss, wrinkling, libido etc. is so much easier than doing thorough long-term mortality follow-up. (Also as you can see the distinction between marker and outcomes is quite blurry when it comes to clinical or functional markers. All of these can be considered markers of aging but are also valuable outcomes by themselves.)

The reason I am excited about these is that some drugs like rapamycin have shown rejuvenation-like effects in mice and there may exist more such drugs. Not only are they in a way more attractive than drugs which merely slow aging, they would be also cheaper to study since you could design a study based on functional biomarkers to efficiently measure rejuventation!

Jesse and I agreed that emerging digital biomarkers are very suited to study some of these functional markers (e.g. heart rate variability, sleep quality, movement patterns).

This topic then led to a great discussion about the difference between cross-sectional and longitudinal studies. Jesse mentioned that the latter are obviously more suited to study a continuous process like aging, although unfortunately such studies are difficult and expensive.

What do we mean by these terms anyway?

Cross sectional — single measurement.

Longitudinal — follow the same group of people or animals over time.

With biomarkers longitudinal can mean more than one measurement, or we measure a biomarker at point T1 and then outcome and outcome at a later point T2, e.g. does cystatin predict kidney failure and death. This in a nutshell is what a prospective cohort study does, which is a longitudinal study design.

The importance of omics

For those who do not know, “omics” refers to a collection of fields in biology that study various sets of molecules within cells, tissues, or organisms. The defining characteristics is usually a high throughput approach where many of these molecular species are studied at the same time. The study of small metabolites is called “metabolomics” and the study of transcription “transcriptomics”, etc.

The really cool thing about omics is that markers based on these techniques could predict changes in functional markers many years in advance.

We speculated which omics field is the best for predicting age-related outcomes. It appears that at this moment in time epigenetics takes the lead. However, this may be because the field had a head start and enjoys a large level of standardization which is essential for clinical studies and cohort studies.

The big disadvantage of epigenetics is lack of “interpretability” which means that given a result with some specific epigenetic methylation pattern it is hard to make sense of it in terms of causality. We simply do not know how these marks affect protein levels, which are the ultimate effectors. That is why the different omics layers are so important.

Mahdi also alluded to another issue. We have very little head to head comparison data and when they looked at this in preliminary work, it seemed like, for example, metabolomics was also performing very well for the prediction of age-related outcomes.

The host — brief bio

Kamil Pabis, MSc is an aging researcher and longevity advocate with several years of experience in the aging field that spans multiple countries. Among other projects, Kamil worked on long-lived dwarf mice in Austria, on mitochondrial disease and aging in the UK, and finally on the bioinformatics of aging in Germany and Singapore. Presently, he is involved in several projects related to science communication and translational aging research.

In the upcoming episode of The Science of Aging Podcast, we're set to explore aging research with Dr. Jesse Poganik and Dr. Mahdi Moqri, just before the 2023 Biomarkers of Aging Symposium. Dr. Poganik ...
Read more
November Longevity Research Newsletter
December 5, 2023
Maria Marinova & Rhys Anderson
Longevity
Newsletters
November Longevity Research Newsletter

Introduction

Welcome back Vitalians and please join us in congratulating the Remedium Bio team for passing the VitaDAO token holder vote with 75.61% voting in favour! This proposal was an assessment of a regenerative medicine biotechnology company, which has developed the only dose adjustable gene therapy platform technology Prometheus™ and is currently advancing multiple assets with uncorrelated risk to Investigational New Drug (IND) approval.

Get ready for some tail-wagging news! Loyal's latest leap in longevity science has just made history. For the first time ever, the FDA has nodded yes to a drug aimed at extending lifespans – and it's for our furry large-breed friends!

Loyal's LOY-001, is all about giving more years of belly rubs and fetch games to our big pooches. Though not yet in the market, this breakthrough means we're on the brink of a huge leap forward. This breakthrough is not just a victory for Loyal but a beacon of hope for all who dream of a future where longevity is within reach. 

And just when we thought we'd have to spend this newsletter talking about the unfolding OpenAI drama this fantastic news came wagging in! 

And the good news don’t stop here. XPRIZE Healthspan Challenge is live! Launched at the Global Healthspan Summit in Riyadh, this groundbreaking competition aims to revolutionize aging! A staggering $101 million prize awaits the team that develops a therapy to rewind the aging clock by 20 years for muscle, cognition, and immune function in older adults.

The twist? The project needs to demonstrate remarkable results within just one year of treatment!

We also have some exciting new research and fascinating reviews. We’ve explored one of them “Epigenetic aging of mammalian gametes” more deeply with the lead author Prof. Michael Klutstein in our monthly interview!

Longevity Literature Hot Picks

Preprint Corner

The votes are in - The Longevist curators have decided what they think are the most important preprints of Q3. Before we publish the Q3 Issue of the Longevist, take a look at our previous issues!

Check out these latest preprints, each of these will be entered into the Q4 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

Intranasal GHK peptide enhances resilience to cognitive decline in aging mice

Protein thiol alterations drive aberrant phase separation in aging

Regulators of health and lifespan extension in genetically diverse mice on dietary restriction

Quantification of healthspan in aging mice: Introducing FAMY and GRAIL

The AccelerAge framework: A new statistical approach to predict biological age based on time-to-event data

A mitochondria-regulated p53-CCF circuit integrates genome integrity with inflammation

Published Research Papers

In vivo reprogramming leads to premature death linked to hepatic and intestinal failure

Continuous in vivo expression of reprogramming factors (Oct4, Sox2, Klf4, c‐Myc) can reverse aspects of aging in tissues but causes liver and intestinal dysfunction, leading to weight loss and premature death. A transgenic mouse strain was created to avoid these side effects, allowing safer, longer-term rejuvenation studies.

Failure to Repair Endogenous DNA Damage in β-Cells Causes Adult-Onset Diabetes in Mice

Increased age raises type 2 diabetes risk due to DNA damage in β-cells. Ercc1-deficient mice developed diabetes, linking DNA damage to β-cell dysfunction and disease onset.

Life expectancy can increase by up to 10 years following sustained shifts towards healthier diets in the United Kingdom

Making healthy food choices can significantly impact your life expectancy. Research using data from the UK Biobank suggests that transitioning from unhealthy eating habits to recommended dietary patterns could add nearly 9 years to the lives of 40-year-old men and women, with even greater gains by adopting longevity-associated diets rich in whole grains, nuts, and fruits while reducing sugar-sweetened beverages and processed meats.

Cellular Senescence Exacerbates Features of Aging in the Eyes

Senescent ocular cells (SOCs) from human corneas exhibit increased aging markers and disrupt epithelial barriers due to a pro-inflammatory secretory phenotype (SASP). Senolytic treatment in older mice with dry eye symptoms prevents corneal opacity.

Stochasticity Explains Nongenetic Inheritance of Lifespan and Apparent Trade-Offs between Reproduction and Aging

Longer-lived female flies produce longer-lived but fewer offspring, indicating a stochastic trade-off between lifespan and reproduction. This challenges traditional views on aging, suggesting random factors rather than genetics predominantly influence lifespan inheritance and aging treatments.

Longitudinal study of traumatic-stress related cellular and cognitive aging

Accelerated epigenetic aging in trauma-exposed veterans predicts increased dementia biomarkers and cognitive decline, linking psychiatric comorbidity to neurodegeneration risk.

Dietary restriction of isoleucine increases healthspan and lifespan of genetically heterogeneous mice

Restricting dietary isoleucine (IleR) improves metabolic health, promotes leanness, and controls blood sugar in young and old genetically diverse mice. It also extends lifespan, particularly in males, suggesting its potential as a geroprotective intervention.

ATAC-clock: An aging clock based on chromatin accessibility

We’ve all heard of methylation clocks, now this team have developed a chromatin accessibility clock which has a direct link to age-related transcriptional alterations, whilst also outperforming a transcription clock trained on the same data.

Mid-old cells are a potential target for anti-aging interventions in the elderly

SIRT2 transgenic over-expression does not impact lifespan in mice

This study uncovers a subset of fibroblasts and smooth muscle cells termed "mid-old status" cells in aging organs, distinct from proliferative or senescent cells, exhibiting upregulated pro-inflammatory and downregulated anti-inflammatory genes. Within the stroma, the inflammatory microenvironment induced by SAA1 negatively impacts epithelial cell stability, contributing to aging-related tissue dysfunction, but this effect can be reversed by a young cell-derived protein, SLIT2, suggesting a potential method to prevent or ameliorate aspects of age-related tissue decline.

Comparative analysis of animal lifespan

Exploring aging across a diverse range of species offers valuable insights into universal aging processes. Though recent methodological advancements enable the study of evolutionarily distant species, the broad scope introduces challenges in experimental design. In this discussion, the authors discuss these challenges, identify key issues to tackle, and offer recommendations grounded in current approaches to effectively conduct comparative aging studies spanning the animal kingdom.

Published Literature Reviews, Hypotheses, Perspectives and more

Epigenetic aging of mammalian gametes

Olfaction: an emerging regulator of longevity and metabolism

Authored by a VitaDAO fellow!

To promote healthy aging, focus on the environment

Combining geroscience with environmental research offers a way to understand and improve healthy aging and reduce health disparities, by studying how environments influence aging processes from an early age.

Entropy Meets Physiology: Should We Translate Aging as Disorder?

Multipotential stem/progenitor cells, crucial for tissue homeostasis, face age-related challenges, leading to altered self-renewal, differentiation, and immunomodulation, contributing to tissue destruction and reduced metabolic performance. This review highlights the impact of aging on mesenchymal stromal/stem cells (MSCs) and suggests a parallel between the thermodynamic concept of "entropy" and biological aging, proposing that both entail irreversible disorder within systems. The question arises: should aging be viewed as disorder?

The power and potential of mitochondria transfer

Mitochondria, traditionally considered to pass down vertically during cell division, are now recognized to undergo intercellular transfer in certain cell types, allowing them to be delivered to unrelated cell types. This process, termed intercellular mitochondria transfer, plays a role in regulating various physiological functions across organ systems. This review explores the mechanisms and impacts of intercellular mitochondria transfer on various cellular processes.

The longevity bottleneck hypothesis: Could dinosaurs have shaped ageing in present-day mammals?

The intriguing contrast in ageing patterns between mammals and certain reptiles and amphibians sparks curiosity about the evolutionary roots of ageing. While mammals, shaped by the dominance of dinosaurs over millions of years, exhibit marked ageing, some reptiles defy ageing or age very slowly. The 'longevity bottleneck hypothesis' suggests that the pressure for rapid reproduction during the era of dinosaurs led to the loss or inactivation of genes and pathways associated with long life in early mammals, creating an enduring impact on their ageing processes.

Job board

Maja Funk Lab at Holmholth Munich is looking for a PhD candidate on a lung organoids project to investigate the links between ageing and environmental stress in COPD, as well as a technical assistant

Maria Mittelbrunn is hiring a postdocs interested in immunometabolism during inflammation and aging at the Centro de Biología Molecular Severo Ochoa in Madrid.

PhD positions in barcelona with Dr. Ana Victoria Lechuga-Vieco on Mitochondrial Biology and Tissue Regeneration

Joao Pedro de Magalhaes has an opening for a PhD studentship at the University of Birmingham to study ageing, longevity and cellular rejuvenation

Deadline: 4th January 2024

An exciting research fellow opportunity to join the groups of Dr Nazif Alic and Prof. Linda Partridge at the UCL Institute of Healthy Ageing. The successful applicant will investigate the role of the multiple copies of tRNAiMet in animal development and ageing. Deadline: 8th December 2023.

Pankaj Kapahi has an opening for a postdoctoral fellow interested in studying “how metabolism regulates astrocyte-neuron signaling in aging and dementia.” Send your CV to pkapahi@buckinstitute.org

American Aging Association are looking for enthusiastic trainees studying aging or age-related diseases to apply for the AGE Early Career Scholar’s Program.  Apply Today!

PhD Position available at University of Birmingham - Researching skeletal muscle decline with ageing and obesity: establishing a role for adipose tissue inflammation and immune system deterioration

News and Media

Venture Capital Giants In The Billion-Dollar Quest For Longevity Breakthroughs

The Secrets to Healthy Aging According to Longevity Experts

Can restoring elastin with existing drugs improve longevity?

VITALISM. n. The philosophy and movement against death and biological decline

Nature's Design Paradox: Aging as an Inherent Software Flaw

Support the Dog Aging Project

An Aging Expert Thinks Humans Can Live for 20,000 Years. He's Not Crazy

The Clues to Longer Life That Are Coming From Dogs

Resources

Bottlenecks of Aging by the Amaranth Foundation

The science of longevity and the quest to solve an age-old problem

Coleen Murphy’s upcoming book How We Age: The Science of Longevity tells the story of these remarkable discoveries through the words of a protagonist in this field.

Prizes

The deadline is approaching for the Longevity Funding Innovation Prize and the reward of $20000 is still up for grabs

XPRIZE Healthspan is a $101 million global competition to revolutionize the way we approach human aging.


Conferences

Dec 1-2nd, San Francisco

Foresight institute vision weekend 

Dec 4th, Novato CA

The Biomarkers of Aging Symposium

Dec 5-6th, Buck Institute, Novato CA

The Longevity Summit

Dec 7th, Stanford

Bay Area Aging Meeting (BAAM)

Some of the VitaDAO team will be around all of the Bay Area aging meetings so come say hi and maybe claim some Vita swag!

Tweets of the Month

Ex Binance CEO CZ is suddenly interested in biotech:


CZ 🔶 BNB @cz_binance:

Been reading about biotech, thinking about how to use crypto to accelerate research funding there.

Keep building!

Mikhail Batin

Aging remains an enigmatic phenomenon. Despite its universal occurrence, the evolutionary rationale and molecular underpinnings of aging are not fully understood.

There are three overlapping strategies to combat aging:

1. Searching for therapies based on our current understanding of aging mechanisms.

2. Identifying new targets through big data analysis, such as transcriptomics.

3. Validating aging theories and testing them empirically…….

Danielle Beckman

What's the biggest difference between a neuron from a young vs. an aged primate? It is not the length of the axon and dendrites, but the number of spines available to make contact and connections with other surrounding neurons!

Podcasts and Webinars

Longevity & Aging Series

Aging (Aging-US) and FOXO Technologies collaborated to produce a special video series on aging research: Longevity & Aging Series. In this limited series, selected Aging researchers were invited to speak about their research with host Dr. Brian Chen, an adjunct faculty member at the University of California San Diego. LEARN MORE

Check out some talks from The Longevity Forum’s Longevity Week

The longevity revolution has the potential to transform healthcare in our lifetime

Funding Opportunities

The SENS Research Foundation is now accepting applications for our undergraduate Summer Scholar Program and our Post-Baccalaureate program (which is paid)! Apply today

Interview with Michael Klutstein

Michael Klutstein is an Associate Professor Epigenetics who heads The Chromatin and Aging Research Lab at the Hebrew University of Jerusalem. His lab is interested in understanding the dynamics of heterochromatin adaptation and subsequent gene expression during cellular and developmental processes and how these contribute to the process of aging in different tissues

What inspired you to enter longevity research?

The future of mankind depends on our ability to successfully cohabitate with our eco system, to gain and make perfect use of experience and wisdom and to successfully colonize other planets. All these goals require that we live longer than we do today. For the future of mankind- longevity research is essential. 

Which of the current theories of ageing do you think are the most convincing?

I do not currently prefer one theory as they all grasp some real aspects of aging, but in my opinion fail to grasp the picture in its entirety. I think aging is so pleotropic that more extensive theories are needed to understand it. Recently, a theory of elevated stochasticity as an origin for aging is starting to emerge. In my opinion- this is the closest thing to an comprehensive theory for aging we have today. 

How has the field changed since you started?

From a couple of genes involved in aging we have dozens now. From a handful of mechanisms involved we have many. From no treatments to delay aging we have now several strong candidates. 

What mistakes do you think the longevity field has made?

Too much emphasis was given to interventions instead of deep biological understanding. This is true for many other scientific fields, but I think in this case- we would benefit much more from understanding the basic Biology. 

Other than your own, what do you think have been the biggest/important discoveries in the field?

I will focus on the field of reproductive aging, as this is my main focus. I would like to highlight the excellent work of Bjorn Schumacher who is developing the theory of elevated stochasticity in aging, and who has shown several key points in reproductive aging in nematodes. Also, the work of Francesca Duncan, to show how ovaries change with age have been a key to gain insight into how reproductive aging occurs.  

What advice would you give to people currently working in longevity research?

Open your eyes, ears and minds, cause this field is going to explode…

Which aspect of longevity research do you think requires more attention?

The answer to this one is obvious as it is close to my heart- I think the field of reproductive aging is key to understanding aging in general and deserves much more attention and resources. 

Is ageing a disease?

No. In other words - it’s not a bug, it’s a feature. 

In your recent work, you describe how germ cells also age, similar to other cells in the body. Are there specific types of damage or aging processes that are more prevalent in gametes compared to adult somatic cells?

Yes, definitely. Germ cells seem to be much more sensitive to epigenetic changes than somatic cell types. The reasons for that are unclear, but a few possibilities are the fact that gametes are arrested in the cell cycle for long periods, and the fact that they are arrested at an unusual point in the cell cycle- after DNA replication. Another important point is regulatory chromatin processes that operate in gametes to activate parts of the genome that are germline specific- and which we do not know enough about. 

It's understood that while gametes age, the creation of a new organism effectively resets or wipes out this damage, leading to a 'fresh start' for the offspring. In the context of your focus on epigenetics, would you say epigenetic factors are the primary drivers of this full rejuvenation, or are there multiple processes involved? Do you plan to explore these additional processes?

Epigenetic processes are thought to be the main driver of this “reset”, since the genetic information is kept intact, and only the epigenetic information is changed in this process. An important and unknown link between the genetic and epigenetic information are mobile elements such as retrotransposons. These are activated because of epigenetic changes, and then move to other parts of the genome, changing the genetic material itself. Their propagation could be a key factor in understanding the interactions between the genetic material, epigenetics and transgenerational reset of information. 

What can we learn from germline rejuvenation that could potentially be translated to human aging?

We can learn a great deal. We can learn that aging can be cancelled but relatively simple interventions. We can learn that the aging process may differ from system to system and between cell types. Still, germline rejuvenation brings up a lot of unsolved issues, both scientific and ethical that need to be addressed.  

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful in helping you to keep up-to-date with all the exciting longevity-related developments. See you next month!

Further Reading

Mitophagy Activation by Urolithin A to Target Muscle Aging

Circulating monocytes expressing senescence-associated features are enriched in COVID-19 patients with severe disease

A Mediterranean Diet-Based Metabolomic Score and Cognitive Decline in Older Adults: A Case–Control Analysis Nested within the Three-City Cohort Study

Methylmalonic acid in aging and disease

Too big not to fail: Different paths lead to senescence of enlarged cells

Vitamin B12 emerges as key player during cellular reprogramming

Too big not to fail: Different paths lead to senescence of enlarged cells

Workshop Report - Biology of Stress Responses in Aging

Genetics of human longevity: From variants to genes to pathways

Cellular age explains variation in age-related cell-to-cell transcriptome variability

A randomized placebo-controlled trial of nicotinamide riboside in older adults with mild cognitive impairment

Clonally expanded memory CD8+ T cells accumulate in atherosclerotic plaques and are pro-atherogenic in aged mice

COPI vesicle formation and N-myristoylation are targetable vulnerabilities of senescent cells

Knockdown of neuronal DAF-15/Raptor promotes healthy aging in C. elegans

Senolytic therapy alleviates physiological human brain aging and COVID-19 neuropathology

Rapid measurement of ageing by automated monitoring of movement of C. elegans populations

Microautophagy regulated by STK38 and GABARAPs is essential to repair lysosomes and prevent aging

Mechanisms underlying retardation of aging by dietary energy restriction

Welcome back Vitalians and please join us in congratulating the Remedium Bio team for passing the VitaDAO token holder vote with 75.61% voting in favour!
Read more
Beyond Mice Models: Exploring Human Aging Genetics with Dr. Joris Deelen on The VitaDAO Aging Science Podcast
November 26, 2023
Podcast
Awareness
Beyond Mice Models: Exploring Human Aging Genetics with Dr. Joris Deelen on The VitaDAO Aging Science Podcast

Limitations of mice as model organism have been a recurring theme of the last few episodes. Nothing highlights this better than the human genetic data, since it often seems to disagree with the mouse data. In this podcast I discuss with Dr. Joris Deelen (@Joris_Deelen) discuss whether this is really true and what we can learn about aging from human genetic studies.

We both agree that the state of the art in his field "is quite depressing, [yet also] also quite interesting".

Joris Deelen – short bio

Joris Deelen obtained his PhD at the Leiden University Medical Center in The Netherlands in 2014 in the group of Prof. Dr. Eline Slagboom. In 2016, he joined the Max Planck Institute for Biology of Ageing as a postdoctoral researcher in the group of Prof. Dr. Linda Partridge, where he was promoted to independent Research Group Leader in 2020. The work in his group is mainly focused on the identification and functional characterisation of genetic variants linked to human longevity. To this end, they make use of the CRISPR/Cas9 system to generate transgenic cell lines and model organisms harbouring the identified variants and subsequently measure their functional effects in vitro and in vivo. 

GWAS studies – our main tool to study the genetics of human longevity

GWAS stands for Genome-Wide Association Studies. These studies are a type of research method used in genetics, mainly for the identification of genetic variants associated with particular diseases. GWAS involves the examination of a genome-wide set of genetic variants in different individuals to see if any variant is associated with a trait or disease.

The process begins with the collection of DNA samples from many people who have a certain disease (cases) and from people who do not have the disease (controls). The DNA is then scanned for genetic markers, usually single-nucleotide polymorphisms (SNPs), small differences in the DNA sequence that occur between individuals. The frequency of these markers in the disease group is compared to the control group to find any significant associations.

Interestingly many times the variants found are not even in the protein-coding region of the gene, probably because such variants would be rare due to their high impact on protein function.

The major ways of running GWAS studies for longevity

There are different ways of analyzing the data that will yield slightly different interpretations. The major difference is in the outcome we study. Although we may use the umbrella term “longevity”, there is actually more than one longevity-related outcome of interest. We can study genes that predict survival to old age (as a binary variable), this is most similar to a classic case-control design. A different take on this method is using the whole range of lifespan values, which may increase power but will also include people who died of diseases not related to aging. Both these approaches suffer from one major limitation. We have to wait until a large portion of the cohort has died before we can apply them!

An alternative is to measure how genetic variants correlate with the lifespan of a participant’s parents. This will give a valid estimate for the longevity of each participant because longevity is heritable. Finally, as done by Joris and others, we can also look at healthspan as an outcome, aggregating data from multiple age-related diseases that participants might currently have – obviating the need for a full mortality analysis.

Missing heritability of aging

Genetic heritability estimates the proportion of variation, or differences, in a phenotypic trait in a population that is due to genetic variation between individuals within that population. While the heritability of lifespan is not very high, the heritability of extreme survivorship is considerably higher. As aging researchers we are more interested in extreme survival, e.g. people who become centenarians. 

Regardless of which outcome you look at, it appears that GWAS studies fail to find all the variants that would account for the heritability of longevity predicted from twin studies. Joris and I discuss why this is the case in the podcast. 

Similarly, the heritability of age-related diseases is much higher than the heritability of lifespan. Therefore, I believe, it might be possible to find novel variants associated with lifespan to close the heritability gap, at least somewhat.

Genetic variants: rare vs common

The term SNV stands for single nucleotide variants and SNP stands for single nucleotide polymorphisms. While there is a subtle difference between the two, for the purposes of this podcast we are dealing with small, single nucleotide changes at a specific position in the genome. These genetic variants, differences between people, can be then studied for their association with different traits like longevity.

Some variants are rarer than others. Common variants may be found in 1% of the population, while variants at below 1% are called rare. Ultra rare are found in fewer than 0.1% of the population and in some cases even unique variants may be of interest. Imagine you studied the genome of the oldest person in the world and found a unique variant in a “candidate” longevity gene like growth hormone. This would be obviously scientifically interesting.

The curious case of ApoE and how it compares with other variants

ApoE is a fascinating case  study for the success and failure of GWAS studies. It is the best known and most robust genetic variant associated with longevity. It is by far the most robust one, as we both lament. Almost none of the other variants identified so far replicate in all human populations, except ApoE.

It has been shown that the ε2 (E2) allele is overrepresented in long-lived individuals compared to control groups, while the ε4 allele is underrepresented. The ε3 allele, being the most common, does not seem to have a strong effect either way. The E2 allele may be also linked to lower risk of Alzheimer’s disease and cardiovascular disease. Interestingly, ApoE genotype could be more important in people who eat a lot of cholesterol, i.e. cholesterol would be more risky if you have an E4 genotype.

In a way ApoE is also interesting because it breaks a common pattern seen in longevity GWAS. Usually variants are not protein altering, except for the ApoE alleles which do change the actual protein, giving rise to isoforms.

Finally, it is important to point out that ApoE by itself would not be enough to meaningfully extend human lifespan since we are looking at a small extension in absolute terms even in the base case. If I recall correctly Joris mentioned something on the order of 2 years. If we want to robustly extend human lifespan we need to discover gerotherapeutics that work in humans as well as does rapamycin in mice.

Alternative approaches and future directions for genetic studies

Using candidate gene approaches, where a candidate is defined based on animal data, e.g. “insulin signaling pathway”, rare or even unique variants enriched in long-lived populations are collected and then studied further and validated in animal models. The work Joris himself does or Vera Gorbunova did on SIRT6 falls into this category.

Most GWAS studies are performed in populations of European ancestry or in East Asian populations, whereas there is very little data from Africa, South America and many other regions. If we could diversify our datasets we might be able to identify novel variants specific to these populations and better confirm our so called “universal” variants.

Novel techniques including long read sequencing to study changes in structural variants and larger scale chromosomal changes as they related to aging. Even just a shift from array genotyping to standard whole genome sequencing could help to reveal novel longevity-related variants.

We also discssus collaborations with 23andme or other companies that sell commercial tests and how we could use this to increase the total sample size of available longevity GWAS studies. Here the limiting factor is that these companies may be unable to keep track of mortality statistics and/or did not inquire about parental longevity, the two key outcomes we would use in GWAS studies.

Another promising technique is mendelian randomization. We call it randomization because each person randomly inherits genetic variants from their parents. Using GWAS we then define a set of variants that are interesting (because they predict X) and then we ask if they are associated with longevity, which will also tell us whether X is causally associated with longevity.

One example would be asking whether genetically proxied risk for Alzheimer’s disease predicts lifespan, another would be to look at predicted LDL or iron levels. If a genetic variant is associated with higher levels of LDL cholesterol, and if the same variant is also associated with an increased risk of heart disease, one could infer that higher LDL cholesterol levels cause an increased risk of heart disease (this is indeed the case; Daghlas and Gill 2021). Below we discuss this approach for iron.

Iron and Aging – a case study for the usefulness of genetics

As a part-time iron researcher myself, I was pleased to discuss Joris’ work on iron, where he used mendelian randomization to test whether genetically predicted iron levels are related to measures of longevity. There is a plausible link between iron and aging. In fact, I have written an article on this topic not long ago, which while quite focused on supplementation, nutrition and blood testing, nevertheless provides a good review of the evidence on this topic: (https://novoslabs.com/iron-longevity-aging-cancer/)

“Iron is a mineral that’s essential to human health and life. Your body uses iron to generate the protein hemoglobin, which red blood cells use to carry oxygen from the lungs throughout the body, as well as the protein myoglobin, which provides oxygen to muscles. The body requires a careful balance of iron in just the right amounts: In studies, iron has presented as one of the few micronutrients that shows a pronounced U-shaped dose-benefit-response curve within a relatively narrow range of intakes or blood levels. This means that both too much and too little iron is harmful to your health.

Under normal circumstances, iron is so precious that nature did not come up with an active mechanism for the excretion of this metal. Bleeding and cell shedding are the major ways in which the body loses iron and modulates uptake. While iron is utilized by many enzymes, it is nonetheless dangerous to the body under certain circumstances. Unbound or free iron can catalyze oxidative stress and DNA damage through the so-called Fenton reaction.”

In this study (Timmers et al. 2020) Joris and colleagues first went back to the definition of longevity before even starting the analysis. Combining data from studies looking at parental lifespan, healthspan and long-lived participants they were able to validate and extend the list of significant genes found in GWAS studies. Having established that, they identified which so called quantitative trait loci (QTL) were colocalizing with the above GWAS variants. Since QTLs predict “quantitative” changes in some phenotype (e.g. height or cholesterol) they were able to put them together and test for enrichment of pathways that could affect these phenotypes. One pathway they found was “heme metabolism” and, as far as I understand, this then led them to perform full-scale mendelian randomization of iron traits, finding that lower iron was beneficial for longevity. 

Although as a non-geneticist, I do not pretend to fully understand what they did, it is certainly good work based on plausible biology that has been solidly replicated (Daghlas and Gill 2021).

Studying accelerated aging (progerias)

We also talk about an often-controversial topic which is progerias. Some people study these syndromes since they appear to accelerate aging. During the podcast we speculate that there should be progeroid rare variants that produce less severe symptoms, which could teach us something about pro-longevity pathways (whose loss of function is harmful to lifespan).

However, there are many ways to shorten our lifespan genetically and fewer that will extend lifespan. Furthermore, the variants that shorten lifespan will do this in different ways, whereas there is only one way to live longer, which is by slowing aging or one of the major age-related diseases. You may call this the Anna Karenina principle applied to longevity if you will.

The host – brief bio

Kamil Pabis, MSc is an aging researcher and longevity advocate with several years of experience in the aging field that spans multiple countries. Among other projects, Kamil worked on long-lived dwarf mice in Austria, on mitochondrial disease and aging in the UK, and finally on the bioinformatics of aging in Germany and Singapore. Presently, he is involved in several projects related to science communication and translational aging research. 

References and further reading

Daghlas, Iyas, and Dipender Gill. "Low‐density lipoprotein cholesterol and lifespan: A Mendelian randomization study." British Journal of Clinical Pharmacology 87.10 (2021): 3916-3924.

Timmers, Paul RHJ, et al. "Multivariate genomic scan implicates novel loci and haem metabolism in human ageing." Nature communications 11.1 (2020): 3570.

Daghlas, Iyas, and Dipender Gill. "Genetically predicted iron status and life expectancy." Clinical Nutrition 40.4 (2021): 2456-2459.

Limitations of mice as model organism have been a recurring theme of the last few episodes. Nothing highlights this better than the human genetic data, since it often seems to disagree with the mouse data.
Read more
VitaDAO Letter: Introducing The Members Portal
November 20, 2023
Awareness
VitaDAO Letter: Introducing The Members Portal

🌎 VitaDAO.Global

Just when you thought being a $VITA holder couldn’t get any better, VitaDAO launched VitaDAO.Global. This new membership portal features an overview of one’s $VITA holdings, active and past DAO governance proposals, a comprehensive DAO treasury, and exclusive discounts to DAO members! Huge kudos to the Tech-Product squad for their amazing work!

Since the initiation of VitaDAO, members have voiced the desire to optimize their longevity and health journey. VitaDAO.Global was created with this in mind, to give DAO members access to health products and services at a discounted price. In addition to health optimization, the portal offers discounts on common expenses such as hotel stays, housing rentals, coworking spaces, and academic publications. DAO members, such as Esther Klaps (featured later in the Newsletter), have been hard at work identifying and coordinating these services. Currently, all VitaDAO members with 100+ $VITA in a linked wallet qualify for these exclusive member services.

Current discounts include:

  • 31% off GlycanAge Tests: GlycanAge combines a biological age test with expert advice to optimize wellness. The age tests examine the state of one’s immune system through examination of the types of glycans attached to Immunoglobulin G, the most common antibody in the blood. The type of glycans attached to Immunoglobulin G can able to change the function of the antibody from pro-inflammatory to anti-inflammatory and vice versa. Signs of chronic inflammation are directly related to lifestyle.

The overall process is simple, requiring only a small fingerprick at home. You can expect results in three weeks in the form of a report. After which you will have a consultation to further explain the test results. As a VitaDAO member, you have discounted access to this cutting-edge approach to measuring biological age.

  • Up to 50% off Sonder in 40+ cities: VitaDAO has teamed up with Sonder to offer DAO members premium accommodations at affordable rates. Simply browse the selection of Sonder properties in your desired destination and apply the portal’s discount code to receive up to 50% off your next accommodation!
  • 15% Off Bon Charge: Optimize your sleep, relaxation, and overall well-being! Bon Charge offers a variety of products including infrared sauna blankets, infrared PEMF mats, blue light-blocking glasses, cold therapy devices, and red light therapy devices. As a $VITA hodler, you get 15% off Bon Charge’s products to optimize your health in this digital world.
  • 10% Off Sensate: Stressed by fluctuations in the crypto market? Sensate is a sensory device that uses vibrations and sounds to decrease stress and improve stress. This combination of chest resonance is thought to signal the vagus nerve.

The team is actively working to improve and expand upon the exclusive offers for $VITA holders. Reach out on Twitter with suggestions for products or services that you would like to see partnered with VitaDAO!

🏠The Network State Conference

This month, VitaDAO’s founding member, Laurence Ion, took the main stage at The Network State Conference. In his talk, he discussed the idea of creating a city to increase work speed and fix the crisis of aging.

“You are all infected with a terminal disease. It is called aging…. Join or die.”- Laurence Ion

In this talk, which can be found on YouTube, Laurence introduces the idea of Vitalia, a movement to start a longevity network city. The next step of this movement is happening in January and February of 2024, through a two-month pop-up city in Roatán. VitaDAO members have priority to join and co-create. The ultimate goal is to identify semi-autonomous zones where an in-person community can self-govern, self-experiment, and identify lifesaving treatments.

🧢Buy VitaDAO Merch

As if the Membership Portal wasn’t enough big news this month…🥁🥁🥁(drum roll please)… introducing VitaDAO’s Merch Shop! VitaDAO teamed up with senior designer Patrick Saville to produce a limited edition hoodie. This sustainably produced hoodie commemorates the genesis of VitaDAO. Don’t miss the opportunity to get this VitaDAO collectible- limited units are available!

🗣️VitaDAO in the Wild

- VitaDAO-backed Turn Bio announced the ability to deliver precise in-vivo mRNA delivery to dermal cells!

- VitaDAO-backed Turn Bio also received positive FDA feedback on TRN-001, a cell rejuvenation therapy!

- Last month, VitaDAO became the first DAO to kickstart a biotech company (MatrixBio).

- Balajis gave VitaDAO a shout-out on Twitter, recognizing the global power of VitaDAO’s decentralized network.

📣Community Approved: What YOU voted for!

Thanks to the support of the VitaDAO community, one proposal successfully passed this month! This proposal was VDP-121. In VDP-121, the community voted to mint an additional 10% of the total token supply in new vested VITA for new strategic contributors. VitaDAO’s 2023 Strategic Plan outlines the launch of the VitaDAO Accredited Investor Fund (VDAIF), anticipated in Q4. The fund will be token-gated, increasing the demand for VITA tokens. Currently, purchasing VITA on DEXs is impossible for these entities in high enough volume. With the passing of VDP-121, we are excited to grow the DAO treasury and include these new strategic contributors!

💪Exercise Your Right to Vote

Visit VitaDAO’s governance hub (Discourse) to engage with, vote on, and discuss proposals before they are moved to Snapshot. Together, we can shape the future of VitaDAO and accelerate decentralized science.

👋Meet the Lead of VitaDAO’s Membership Program: Esther Klaps

Esther Klaps is a valuable contributor to the VitaDAO community and the Lead of VitaDAO’s Membership Services. More recently, Esther has been the driving force behind VitaDAO’s Membership Program, identifying opportunities and coordinating strategic partnerships. Esther holds a BSc. in International Business from Maastricht University’s School of Business & Economics and further honed her expertise with an MSc. in Blockchain and Digital Currencies from the University of Nicosia’s Institute for the Future. She brings a wealth of expertise to the table, having excelled in various partner-facing roles, event planning, and business strategy. When not contributing to VitaDAO, Esther enjoys exploring her interest in nutrition and fertility.

How did you first learn about VitaDAO and what drew you to it?

I previously worked at ConsenSys in employer branding and ecosystem growth, and knew I wanted to stay within the Blockchain world. When I came across VitaDAO, this sounded like the perfect combination of my work in Blockchain and my passion for health. I have a background in Orthomolecular Nutrition, focused on reproductive health, which of course is closely related to improved longevity.

How are you currently contributing to VitaDAO?

I’m currently focused on finding interesting companies to collaborate with. We want to offer discounts on excellent longevity and biohacking products and services to our members through the Member Portal. Next to this, I’m also contributing to the Ambassador Program.

You’ve worked in a variety of different fields, and also have a background in Blockchain. You likely have a different perspective than most VitaDAO contributors.

From working in member and partner-faced roles in Blockchain, I do have a good understanding of the needs and wants of a Web3 audience. The way of interacting with that audience is very different than that of the more “traditional” academic world. I feel that’s also one of VitaDAO’s strengths, that our contributors come from both worlds.

What has surprised you the most about VitaDAO? What sets it apart?

There’s a lot of potential in Blockchain technology. However, from what I’ve seen working in Web3, many brilliant ideas are constrained to the crypto ecosystem, unable to truly impact real-world problems. I enjoy contributing to VitaDAO because you can directly see the impact it’s able to make by empowering and funding promising early-stage longevity projects that would otherwise lack access to funding. VitaDAO is able to create that bridge between Web3 and the “real world”.

Any closing thoughts for readers?

If there are any biohacking products or services you’d love to see discounts for on our Member Portal, do let us know in Discord!

🤝Thanks for Reading!

Want to stay up to date with all that’s going VitaDAOn? Join us on Discord, subscribe to our Twitter, and follow our Instagram! You can now connect your wallet to VitaDAO’s Members Portal to learn about exclusive member benefits and find VitaDAO’s treasure dashboard. Find updated information on VitaDAO’s funded projects on our website.


Just when you thought being a $VITA holder couldn't get any better, VitaDAO launched VitaDAO.Global.
Read more
Epigenetics & Longevity: Unraveling Aging Mysteries with Dr. David Meyer and Prof. Sarah Voisin at ARDD – The VitaDAO Aging Science Podcast
November 9, 2023
Podcast
Longevity
Epigenetics & Longevity: Unraveling Aging Mysteries with Dr. David Meyer and Prof. Sarah Voisin at ARDD – The VitaDAO Aging Science Podcast



This special the episode of the Aging Science podcast is brought to you from the ARDD conference that took place at the end of August in Kopenhagen. I sat together with Dr. David Meyer from CECAD and Prof. Sarah Voisin from the University of Copenhagen to talk about epigenetics, clocks, our favorite conference presentations, stochastic and adaptive changes during aging. Finally, we also talked about the all-important questions of sustainability and overpopulation, and whether longevity researchers should have a say in this matter.

https://agingpharma.org/

My interview partners

David is a bioinformatician with a strong interest in genome instability and aging research. His research utilises data analysis, machine learning, and the development of computational tools to help advance our understanding of aging and age-related diseases.

Sarah is an Assistant Professor at the University of Copenhagen and a former Australian NHMRC Early Career Fellow (2019-2022). She works at the intersection of epigenetics, genetics, statistics and bioinformatics, with particular focus on exercise, ageing and human health. She completed her PhD at Uppsala University and Pierre & Marie Curie University, and published 58 peer-reviewed papers. Her current research work and interest is epigenetic ageing and exercise, sex differences in biology, and statistical methods to develop personalised health interventions.

What is epigenetics?

The term epigenetics describes a form of gene regulation that relies on modifications to the (chemical) structure and organization of DNA and chromatin. This is in contrast to genetics which deals with changes to the DNA sequence itself.

Epigenetic marks are a type of modification that is attached to DNA that help genes to be expressed at the proper time. There are two major types of epigenetic marks, those found on histones and those found directly on the DNA. Histone modifications are highly diverse, whereas regulatory DNA modification is largely limited to the methylation of cytosines.

The different epigenetic marks are recognized by reader proteins that affect gene expression. Although these marks can have complex effects, some broad patterns are evident, like for example repression of transcription through CpG methylation.

Although methylation serves an important regulatory role, it turns out not all species have methylation at CpG sites. Bacteria have a different type of DNA methylation whereas C. elegans seems to have lost this feature altogether during evolution.

The rise of epigenetic clocks

When we age, so does our epigenome and one way to measure this epigenetic aging is through so-called DNA methylation clocks. Many studies, initially pioneered by Steve Horvath, have shown that methylation at certain sites in the genome can be used to construct a clock that closely tracks the passage of time.

These clocks are exciting because they are much better at predicting chronologic and biological age than the biomarkers we used to have. In fact, I still remember going to conferences and listening to talks that painted a rather disappointing picture of blood based aging biomarkers.

These clocks are also exciting because they promise to become a surrogate endpoint for medium sized phase II-like studies. A surrogate endpoint is a cheap, easy-to-measure marker that substitutes for something else that is more expensive. The hope is that a slowing of epigenetic aging in small studies would be predictive of success in larger trials that focus on harder outcomes like health and survival. If true, this would push down the cost of trials by orders of magnitude because we could pre-select promising compounds before going into large studies. However, even the best surrogate endpoints like LDL or HDL cholesterol cannot substitute for large studies, as the countless failures and disappointments in the clinic show (e.g. CETP-inhibitors for atherosclerosis and the clinical failure of most non-statin drugs).

Another reason why clocks are important is their link with the biology of aging. Understanding why methylation at certain sites changes and what harmful effects these changes have, could improve our understanding of aging and allow us to find new therapies. To give an example of such advances, as mentioned in a prior podcast, the study of epigenetics has uncovered not only the importance of entropy in the process of aging, but also ways to reset some of these age-related changes (partial reprogramming). 

Stochastic Changes and Aging

These changes refer to random alterations in the DNA, epigenome or even proteome that accumulate over time. David Mayer's work suggests that these stochastic changes, or noise, may play a significant role in aging and may be the age component that epigenetic clocks actually measure. Understanding these changes could provide insights into why people age at different rates, if we find the drivers of increasing noise or even means to reverse it.

In a way the idea that stochastic changes drive aging is a very old one and dates to aging theories like the error catastrophe theory. Initially, it was believed that DNA will accumulate damage, which is then transcribed to produce proteins of lower quality which will further increase damage accumulation. Although there may be no error catastrophe per se, the concept has been rediscovered recently. It may very well be that such stochastic errors accumulate, both on the gene and epigenetic level, which decrease the functioning of gene expression networks, finally resulting in reduced resilience and aging.

There are several interesting aspects of these stochastic theories. One is the potential irreversibility. Aging may be hard to reverse if stochastic, very random, scattered, changes are key drivers rather than a few well-defined damage types or “hallmarks”. This worry has been recently echoed by voices like Peter Fedichev. Because stochastic changes are hard to reverse, and hard to “titrate” in experiments, these theories are also very difficult to prove. For example, there is still no smoking gun experiment showing that reduced DNA damage accumulation slows aging or that increased accumulation accelerates aging. The former has proven devilishly hard since DNA damage repair is so efficient. Whereas the latter is always controversial since models of accelerated damage accumulation often show damage levels that are either too high, too low or too specific (hence it is hard to “titrate” to the correct level).

In this context, we also discussed attempts to disentangle the stochastic component of clocks from an adaptive one, as recently proposed by Vadim Gladyshev. It remains to be seen if this will allow us to pinpoint genes that have important functions with aging.

This special the episode of the Aging Science podcast is brought to you from the ARDD conference that took place at the end of August in Kopenhagen. I sat together with Dr. David Meyer from CECAD and Prof. Sarah Voisin ...
Read more
October Longevity Research Newsletter
November 7, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
October Longevity Research Newsletter

Introduction

Welcome back Vitalians and please join us in congratulating the Oisín Biotechnologies team for passing the VitaDAO token holder vote with 86.47% voting in favour! This proposal was an assessment of a multi-asset longevity biotech company pioneering genetic medicines to combat sarcopenia and other age-related diseases to promote healthier, longer lives.

VitaDAO's Members Portal is LIVE! We want to give tokenholders the opportunity to access health products and services at a discounted price. Currently we have offers with Glycanage and Bon Charge with many more to come.

Stop the press! AgingDoc is AgingDoxxed! After years of speculation as to who was behind this Twitter account:

David Barzilai MD PhD, MS, MBA, DipABLM has revealed his identity!!!! Read here to find out why he made this decision. 

We are also excited to announce that we interviewed David this month, who shared his thoughts on the longevity field with us - check it out at the end of this newsletter! Enjoy!

Longevity Literature Hot Picks

Preprint Corner

The Longevist editors have whittled down a longlist of over 60 longevity preprints from Q3 down to 10 amazing contenders! The Snapshot vote will begin soon to determine which ones our curators think will have the biggest impact in the longevity field.

Check out these latest preprints, each of these will be entered into the Q4 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

Spatially and Functionally Distinct mTORC1 Entities Orchestrate the Cellular Response to Amino Acid Availability

The impact of short-lived controls on the interpretation of lifespan experiments and progress in geroscience

Dietary restriction extends lifespan across different temperatures in the fly

Failed reprogramming of transformed cells due to induction of apoptosis and senescence impairs tumor progression in lung cancer

Elevated epigenetic age in wild compared to lab-raised house mice

The circulating proteome and brain health: Mendelian randomisation and cross-sectional analyses

Published Research Papers

Apoptotic stress causes mtDNA release during senescence and drives the SASP

The Passos lab have previously published research showing that mitochondria are essential for the pro-ageing features of the senescent phenotype. Now they have followed this up with a Nature paper showing that mitochondria in senescent cells develop macropores which allow mitochondrial DNA to leak out into the cytosol and activate the cGAS-STING pathway, which is a major regulator of the senescence-associated secretory phenotype (SASP).

Biomarkers of cellular senescence and risk of death in humans

This study analysed the levels of 28 SASP proteins in the patients’ blood and discovered that high levels of GDF15, RAGE, VEGFA, PARC, and MMP2 were particularly predictive of mortality. 

Effects of iron homeostasis on epigenetic age acceleration: a two-sample Mendelian randomization study

In this paper, the authors showed that higher levels of serum iron were linked to an acceleration of various epigenetic clocks associated with aging, including GrimAge, HannumAge, and Intrinsic epigenetic age

Multi-species atlas resolves an axolotl limb development and regeneration paradox

Using a multi-species transcriptomics approach, this study confirmed the presence of apical-ectodermal-ridge (AER)-like cells in axolotls, a key model organism for regeneration, while noting that complete reformation of AER cells during limb regeneration was not observed. Additionally, the research identified elements of the AER machinery within the axolotl mesoderm, offering insights into the mechanisms of limb (re)growth and resolving the debate surrounding the role of AER in axolotl limb regeneration.

Repurposing Metformin for periodontal disease management as a form of oral-systemic preventive medicine

This study addressed the global prevalence of periodontal disease (PD) and its association with diabetes, emphasizing the role of glucose metabolism modulation as a significant factor in non-communicable disease development. By repurposing Metformin, the research demonstrated that pharmaceutically controlling glucose metabolism led to the prevention of bone loss associated with induced periodontal disease and aging. 

Loss of Grem1-lineage chondrogenic progenitor cells causes osteoarthritis

The research identifies Gremlin 1 (Grem1) as a marker for progenitor cells in cartilage that can become bone or cartilage cells. These cells decrease with age and injury, contributing to osteoarthritis (OA). Loss of Foxo1 function in these cells leads to OA. Treatment with FGF18 promotes these progenitor cells' growth, improving cartilage and decreasing OA, pointing to potential OA treatments.

Transcriptomic analysis reveals a tissue-specific loss of identity during ageing and cancer

This study found aging results in the downregulation of tissue-specific genes in 40% of tissues. In cancer, a similar loss of identity occurs with tumors expressing genes from different tissues, influencing patient survival independently of their age.

A genetically encoded tool to increase cellular NADH/NAD+ ratio in living cells

This research presents a new tool, a soluble transhydrogenase from E. coli, to study reductive stress by increasing NADH/NAD+ ratios in cells. It offers a fresh approach to investigate redox imbalances in diseases.

The total mass, number, and distribution of immune cells in the human body

Whilst not a longevity paper per se, it begs the question: how do these stats change with age?

The association of leukocyte telomere length with exceptional longevity among older women

This study investigates the correlation between leukocyte telomere length (LTL) and the likelihood of living to age 90 with good mobility among postmenopausal women. Findings suggest women with longer LTLs had higher chances of reaching age 90, with or without mobility issues. 

Platelets rejuvenate the aging brain

The studies investigate different aspects of brain function and aging but converge on the finding that platelet factor 4 (PF4) can rejuvenate cognitive abilities in older mice. This marks the first evidence of platelets and PF4 as regulators of brain cognition.

Decoding aging-dependent regenerative decline across tissues at single-cell resolution

The study analyzes how aging affects tissue regeneration, revealing that aged tissues have reduced stem cell movement and blood vessel formation, and a distinct type of macrophage active in young but not old tissues, pinpointing targets for improving regeneration in the elderly.

A fluorophore-conjugated reagent enabling rapid detection, isolation and live tracking of senescent cells

The article presents a new tool, GLF16, for identifying senescent cells using fluorescence techniques. With a novel delivery method using micelles, this approach allows for easier, more precise isolation and analysis of live senescent cells, both in vivo and in vitro.

Published Literature Reviews, Hypotheses, Perspectives and more

Combinatorial interventions in aging

Biomarkers of aging remain elusive as researchers try to slow the biological clock

Device-measured physical activity, sedentary time, and risk of all-cause mortality: an individual participant data analysis of four prospective cohort studies

The ticking of aging clocks

Insulin and aging – a disappointing relationship

Job board

Dr Bernadette Carroll is looking to recruit a research associate to work on various aspects of membrane dynamics and cell signalling at University of Bristol. The aim is to investigate the role of non-canonical mTORC1 activity at focal adhesions.

The Webb Lab at The Buck Institute is recruiting 1-2 postdocs to work on mechanisms of brain aging and neurodegeneration!

Kapahi Laboratory at The Buck Institute are looking to recruit 2 Postdocs to work on applying bioinformatic approaches to study eye aging and neurodegeneration using flies and mammals. The lab values include curiosity driven science, team spirit and mentorship. 

The Kane lab at the Institute for Systems Biology is seeking a highly motivated Postdoctoral Fellow interested in the biology of aging to lead projects focused on understanding, measuring and predicting heterogeneity in aging, and particularly identifying epigenetic determinants and biomarkers of frailty 

The Kohler Lab have an open call for a PhD student to work on the fascinating interrelation between Proteostasis and Ageing, combining High Throughput approaches in Yeast with validation in Cell Culture.

The Whitworth lab at the University of Cambridge are recruiting a PhD student who is interested in unravelling the ways that impinging on mitochondrial calcium can prevent neurodegeneration.

Marta Kovatcheva launching a new lab and is looking to hire a postdoc who will use novel lineage tracing strategies to probe the contribution of partially reprogrammed cells in vivo.

The Korolchuk Lab is offering a new PhD opportunity, working closely with Procter & Gamble to investigate mechanisms of skin ageing. 

Two PhD positions available at the Tayilor Lab on 1) the roles of a novel prion-like protein in stress resistance and aging and 2) the unfolded protein response, restoring homeostasis and a new regulator of aging.

News and Media

Gero raises $6 million to find root causes of aging and age-related diseases

BioAge Labs Announces Plans for Phase 2 Trial of First-in-Class Apelin Receptor Agonist BGE-105/Azelaprag Co-Administered With Tirzepatide for Treatment of Obesity, in Collaboration With Lilly’s Chorus Organization

BATPROTECT: Learning from Bats: New strategies to extend healthspan and improve disease resistance

ERC funding: 11,882,510€ over 72 months

You might have heard of Ora Biomedical’s ambitious endeavour to assess 1,000,000 longevity interventions in 5 years, catchily named The Million Molecule Challenge! Now you can help support his endeavour by becoming a sponsor!

LongeVC: What longevity investors are really looking for

Prizes

A brand new Longevity Prize has launched and accepting application over the next couple of months. The Longevity Funding Innovation Prize initiated and funded by Marat Karpeko is going to award $20,000 USD for the most innovative and potentially impactful longevity funding strategy. The judges want to see a real action plan and tangible solution for real world impact.

Keep in mind the key dates:

  • Kick-Off: 20th September 2023
  • Proposal Closing: 20th December 2023
  • Evaluation Wrap-Up: 20th February 2024
  • Champion Unveiled: 29th February 2024


Conferences

Seno-Therapeutics Summit 2023

7-8 November, Buck Institute for Research on Ageing, Novato, CA, USA

Ageing Research at King's (ARK) Longevity Week 2023 - Sustainable Longevity: Harnessing Consumer HealthTech and FinTech

17 November, King’s College London, UK

The Longevity Forum’s - The Longevity Week

13 -17 November 2023

17 November DeSci & Longevity Biotech 2023 — A Próspera Builders’ Summit

Careers in Aging Day at GSA 2023

Friday, November 10, 2023 – Full-day event, Tampa Waterfront Marriott, 505 Water St, Tampa, FL, US  

NUHS Centre for Healthy Longevity - Unlock Healthy Longevity: Supplements

Scientific Conference: February 29th - March 1st, 2024. This is the first global scientific conference focused solely on the role of supplements as a geroprotective intervention.

The Longevity Summit

4-5th December, Novato CA, Buck

BAAM

7th December, Stanford

Tweet of the Month

Peter Fedichev: It may sound strange, but beyond our "longevity" bubble people often ask: what are the reasons, scientifically, culturally, etc. for the reasons why people want to stop aging? My guess is that consciousness, once emerged, faced the vastness of universe and shortness of life. Hence two dreams, of flight and immortality are universalities of human inspirations transcending all the cultural differences. And this is why, in relation to those questions, I like to tell the following story. Continue reading here...

Podcasts and Webinars

NUS Medicine’s Healthy Longevity Webinar Series

Funding Opportunities

SCPAB Transition to Independence Award supports researchers investigating the mechanisms of cognitive resilience and functional maintenance of the healthy aging brain. Experience in aging research is not required; the program welcomes applications from candidates with backgrounds in neuroscience, molecular biology, genetics, immunology, cell biology and the physical and information sciences.

Interview with Agingdoc, or Dr David Barzilai

What inspired you to enter the longevity field?

Almost as far back as I can remember, I’ve been profoundly struck by the extent of life’s hardship and brevity. At the same time, I love the sciences including biology, and like most of us, I’ve always wanted to make my mark in world helping others.  So, with that as background, and following studying biology and public health related studies in college, I determined to enter medicine, so that I could help alleviate suffering and cure when possible. This was an MD-PhD program in health services research- which taught me much about medicine, evidence-based practice, asking good questions, studying them, and appraising published work.

My keen interest in, or what some may call “obsession,” for optimizing my own health pervaded all these stages in my life, and motivated me to avidly read far wider and deeper than my medical training required. I’ve followed the geroscience literature and see the potential of insight from aging biology to address healthy longevity. These passions were the seed for my getting involved in the longevity space.

I have turned to social media as one manifestation of these interests. By collating significant publications and creating a space for people with varied perspectives to learn and discuss, I aimed to bridge the gap between scientific communities and the public. This initiative not only fosters a collective understanding but also catalyzes a multi-dimensional contribution towards advancing the field of longevity. I did so anonymously for several years, but on my 49th birthday in August 2023 I came to the realization that I could still do so much more simply post publications. It’s time to apply my knowledge more directly. So since then, I’ve worked on what I dubbed “project 49” – starting with coaching and consulting a few clients to experience one-on-one in the longevity space – and from there branching out to more public role of geroscience advancement, advocacy, and general awareness. 

Did your general medical training include anything on geroscience and what do you think should be taught in medical school?

In medical school, the focus was essentially: (a) prevent disease (b) diagnose it- which by definition is primarily after it manifests, (c) treat it- and at best cure it, and if not, at least improve symptom burden and quality of life. At first blush, it’s not a terrible way of looking at health. While it has some preventative focus, the amount of time spent in these areas in medical school paled in comparison with complex excursions into the pathophysiology of disease and vast therapeutic armamentarium that can be applied to illness after it manifests

 I’ll spare a long analysis here on why, but suffice to say it did a better job of preparing future doctors for diagnoses to make later into the underlying biology than would be ideal, and treatments to give late in the game when it’s already an uphill battle making a difference.

It seems to me that we need a stronger preventative focus, and one that not only more strongly emphasizes lifestyle medicine, but also invests in the development of biomedical technologies that directly address the molecular mechanisms driving disease itself.

In other words, I’d like to see curriculum and incentives more strongly aligned towards favorable outcomes for patients, starting with prevention. I’d also like to see geroscience incorporated formally into the curriculum, because while we are still only scratching the surface, the fact (unbeknownst to me in medical school) that there is an underlying biology of aging that we can make a concerted effort to target is no less than revolutionary. We’re starting to make progress, but in medicine, awareness of the field is still minimal. That needs to change, and when society comes aboard, we can take steps together toward better targeting aging biology and with it our healthspan long before overt disease manifests, when we may be able to do much more about it.

How has the field changed since you started?

The field has changed dramatically over only a few decades. We’ve literally gone from questioning whether single genes can impact lifespan in an organism such as C elegans (which we found yes, they can), to targeting genes and gene expression to increase not only lifespan, but apparent health too. 

We have powerful tools emerging across the hallmarks of aging, from epigenetic reprogramming and deployment of senolytics or senomorphics, to genetic engineering, and cell-based therapies. Our tools are not only getting more powerful (CRISPR, scRNASeq analysis, microfluidic chips, you name it), but also are getting more sophisticated at evaluating their output via computational (including machine learning) models. Progress is speeding up with high throughput research pipelines, incorporating robotics and the latest technology.  Even our older models are being revisited now, but with a modern spin: In parabiosis, for example, we’re examining whether there may be potentially useful blood factors (from hormones to exosomes  and small molecules) in younger organisms, or alternatively harmful ones that need to be either filtered out or diluted to achieve a more youthful phenotype.

We’re also starting to scrutinize biological age, and its rate change.  Measuring and tracking biological age is of utmost importance to the future of successfully translating this work to healthy longevity in humans. This requires reliable surrogates for tracking biological aging. While not a replacement for better validated traditional functional biomarkers, progress is being made. Such development and validation would greatly accelerate progress in the deployment of trial-proven geroprotectors for healthy individuals in our lifetime.

Which of the current theories do you think are most promising?

I believe that multiple aging theories are compatible with the same biological process, and that we’re going to require computational AI tools to take their utility to be more useful developing longevity medicine therapeutics. If you are satisfied with this, skip to my next answer. If you’re interested in a deeper dive, keep reading the rest of this section below.

The expression “theory of aging” is imprecise. Sometimes this describes a process, say sources of metabolic or other damage and their sequela. Other times they focus on how these are evolutionarily permissive- mostly under the antagonistic pleiotropy rubric, including its “dysfunctional pseudoprogram” conceptual model. Yet while we can conjecture and model plausibility, we cannot observe evolution in real time, so I focus on the former meaning of “theory of aging” – the change that arises during biological aging.

Biological systems obey the laws of physics, but the complexity arising from nonlinear relationships and higher-order emergence challenges the notion of a single encompassing theory. Thus while many theories today encompass aspects of aging at an intuitive level, what will be most useful are models that can make reliable predictions that can accelerate progress towards therapies. To do so, our best chance may be utilizing computational modeling of empirically obtained data. As for today’s models, I believe the majority either (a) describe the same process through different imprecise human analogies, (b) capture different aspects of biologic aging arising spontaneously in parallel, or both. As entropy increases at the molecular, cellular and organismal level, chaos arises through loss of information, damage, and dysregulation of signaling, and other departures from biological homeostasis and integrity. Thus, while distinctions can be made in emphasis, I do not believe these are fundamentally mutually exclusive, and at their core all describe a unified process.

My primary interest is what works; ultimately the experiments will arbitrate the relative importance of different explanatory models in describing aging as a targetable process. I believe, that useful models exist and have been created, and theorizing continues to be helpful both conceptualizing the problem and theorizing solutions (for example genomic instability- fix the genome, epigenetic drift- revert to a more youthful state, molecular damage- address ECM and other units of disarray, inflammation or hyperfunction- employ nutrient sensing modulators to mitigate these, etc).

Yet I also believe that there is a natural limit to how far our conceptually satisfying yet deceptively oversimplistic models of aging will take us. Computational models employing AI are likely to be increasingly valuable helping us break down complexity, into black box predictions whose basis may elude us, yet nevertheless be leveraged to progress.

What do you think have been the biggest/most important discoveries of the field? 

The largest in my mind was our finding that targeting one gene (we’ve found many since then) can have a meaningful impact on lifespan or health. Until this work, biogerontology was more devoted to describing aging than aspiring to target it directly. The second, from the Interventions Testing Program (ITP), that mammalian models can be given interventions that extend lifespan even when given at the equivalent of late middle age. And not only by one molecule, but now several. While we can’t say the impact in humans yet (particularly as we’re already more efficient at damage repair than the shorter-lived models we are studying), it remains a tantalizing prospect that in theory longevity interventions introduced in late adulthood could in principal be successful. ITP should also be recognized for drawing attention to the strong impact of gender on all of these. If we imagine an equitable healthy longevity future for all, it is vital we do the research to see what works for everyone.

What aspects of longevity research do you feel requires more attention?

The short-version is we need a broad agenda, one that incorporates both short and long-term goals, one that balances pragmatism with moonshots, as well as foundational research towards validation of biomarkers so that human longevity trials can be attainable in our lifetime. We also need to mobilize widespread political and social support for translational research pivotal to our capacity for making a real impact on patients (and healthy human lifespan).

There is more about aging we don’t know than we do at this point, and diversification of approaches is our best way forward.  This means funding labs focusing on all aspects of aging biology, from genetic engineering to epigenetic reprogramming. Likewise, we should balance short-term lines of inquiry farther along the investigative, therapeutic and regulatory pipeline, along with long-term more challenging and ambitious projects.  This way, we can embrace what is most likely to translate to therapy in the near term, while also sustaining investment in high-risk, high-reward longevity moonshots. 

Assessing whether known mouse gerotherpeutics work in humans is important low lying fruit, and frankly something I wish we had pursued much more aggressively all along.  Specifically, I am thinking of rapamycin/rapalogs, canagliflozin and 17-alpha estradiol (males), acarbose, several others from ITP including more recently astaxanthin and meclizine, and others, including supplements and nutraceuticals such as taurine, spermidine, urolithin A, fucoidan, alpha ketoglutarate, novel NAD+ boosters, and others. The high safety profile, modest cost and efficient pipelines for approval and distribution make nutraceuticals of even modest expectation excellent candidates. 

So, how do we get the most bang-for-the-buck investing our resources? My first interest, and I suspect yours also, is how do we get safe and effective therapies to humans as efficiently as possible? It would be great for us to be around when the first generation of translational geroscience interventions arrive. 

There are several models for investigation that may help us get there. I support TAME which may be cost prohibitive to apply to all interventions, but the hope is that it may not only narrowly evaluate metformin’s off-label geroprotecive efficacy, but may also support precedent for other gerotherapeutic trials focusing on a panel of aging-associated outcomes rather than focusing on one disease indication. Alternatively, seeking FDA-approval for non-aging indications for modalities that nevertheless work by targeting underlying aging biology is generating optimism that  progress can be made without the requirement of FDA recognition of biologic aging as a drug-approval indication itself per se. 

There are certain key scientific matters that if overcome would help tremendously.  Human lifespan studies are fundamentally untenable, and the field direly needs validated biomarkers, so that shorter term trials can provide reliable and convincing evidence on efficacy. If we can get there, it would entail validating a panel of predictive aging biology surrogate variables such as methlation clocks, and broader -omics panels integrated with more traditional clinical markers such as tracking functional decline, (eg, walking speed, grip strength, etc) predictive of further morbidity and lifespan.

What mistakes do you think the longevity field has made?

I believe we have been slow to build collaborations between academic medical center labs and industry despite having the common aims of helping discover, validate, develop, and deliver gerotherapeutics. We have also failed to invest enough on the public and policy side on the value of geroscience from both a fundamental public good and economic investment standpoint.  Our competitive spirit and fierce independence as labs and longevity biotechs are strengths that should be incentivized. We also need to build bridges, across academics, industry, and organizations, and governmental agencies. We can do this responsibly, with appropriate checks and balances around safety and efficacy, while more efficiently, and without delay.

Is aging a disease?

Is aging a disease? Innumerable arguments have been made, with publications and conference symposium panels all focused on this issue. The answer is simple: It depends on how we chose to define aging, and how we chose to define disease.

The bottom line is this: Aging biology can be argued to be the ultimate risk factor for not one, but a wide multitude of diseases. At the very least, we can observe that something in the aging process- and its molecular underpinnings- is permissive to the development of disease. Thus ultimately, if we are interested in mitigating the negative impact of disease, targeting biologic aging is a sensible approach.

Precisely because biologic aging is intertwined with not one, but a wide multitude of diseases and poor health states (including frailty and lack of resilience), targeting aging biology before it deviates from the state of youth holds the potential to reduce the overall burden of chronic disease.

What do you think are the most important endpoints, biomarkers, and measurements to be taken in human clinical trials for interventions that can impact general healthspan?

This is very much in flux, and we aren’t there yet. The emergence of epigenetic methylation clocks- in particular 2nd and 3rd generation- on the landscape have done much to instill optimism. I do not believe any one measure is likely to get us there. There simply is too much uncertainty including whether prediction can be disentangled from biology (ie, the theoretical possibility that an intervention reduces one measure of biologic aging readout without concurrent improvement in physiology). Thus as I described previously, we’re going to need broad panels with many biomarkers of every kind complementing traditional better validated laboratory and functional surrogates for healthspan and mortality. 

Do you think standardization can be a concern in human aging studies, how hard do you find it to compare findings from studies conducted by various institutions including academic and clinical ones?

Do I  think standardization can be a concern in human aging studies? Absolutely. As you hint, studies are phenomenally disparate. This can be frustrating and confusing, and complicates our efforts towards coordinating efforts and insight into what might account for different results between studies. All the more so when we either pool results for meta-analyses or attempt to mine them for deeper insight via computational large data tools including via AI. Thus I do believe in standardization, and this is arising in various forms, whether consensus conferences on protocols or collaborations such as through the NIH SenNet Consortium, or individual initiatives such as at the ITP which attempts to control experimental differences between centers.

Lack of standardization can be highlighted by the monkey caloric restriction trial, which was frustrating in that results varied between the two (though at least for lifespan, both had groups remarkable improvements in health measures), with remarkable differences in diet genetics, diet quality, diet composition, and frequency of feeding. The poor replication in CITP was another remarkable illustration of the replication crisis affecting geroscience. And as AI models rely on collating varied data sources, consistent input is increasingly necessary for accurate and insightful outputs. So, the need for standardized protocols is greater than ever. 

Where do we find you?

I’m on Twitter @agingdoc1. My Twitter account has been a way to give back by sharing interesting and topical publications for critique/debate, as well as to keep health influencers and scientists “in the know,” so they can stay informed, and thus better educate their audiences (the media & general public) on the good and bad of what is out there. I also continue to be passionate about the synergy of bringing together different communities (from aging biologists to biotech, media, thought leaders, health influencers, and avid biohackers) and I plan to continue that mission, bringing timely and interesting content from across the web to my @agingdoc1 channel.

I’ve been anonymous for years on Twitter but ultimately on my 49th birthday (I secretly called this “project 49” before the big announcement) I decided to reveal my identity. Basically I felt like I could use more of my capabilities and do more good in the world being public than private. I’ve started to do only a little coaching and consulting (this is not a medical practice so I do not diagnose, treat or prescribe but discuss collaborate with my clients generally highly invested in their health and wellbeing). But because I only have a few hours a week to the endeavor, I recognize only a few individuals can take advantage of it. Thus I continue to try to make a broader impact other ways such as by the service I mentioned, and outreach, such as possibly by podcast or here today with you today.

Because of my last name (no relation to Nir for those wondering!) and coaching related work I have a website http://barzilaiconsulting.com, but for ease I have other webpages point to it like http://agingdoc.com and http://healthspancoaching.com. In any case, thanks for inviting me, and I hope I helped a little.

VitaDAO is an amazing organization. I’m as thrilled by the advancing science as I am with the revolution going on starting with leaders like you, at the grassroots, helping transform our culture and bringing the change we would like to see in the world. This is the day for a longevity moonshot. Together, we are making it happen.

David Barzilai, MD PhD – “Agingdoc” 

http://youtube.com/@agingdoc

http://agingdoc.com / http://healthspancoaching.com 

https://twitter.com/agingdoc1 

My letter: https://www.barzilaiconsulting.com/49  (“Project 49” 😊)

Outro

We appreciate you sticking with our research newsletter for another month and hope you’ve found the content useful!

This time we leave you with a fun new article on research by Dr Shahaf Peleg and Dr Andrew Wojtovi that we’ve featured in the letter before: Shining a Light on Optogenetics: An Innovative Approach to Combat Aging

Further Reading

Epigenetic clocks indicate that kidney transplantation and not dialysis mitigate the effects of renal ageing

Blazing a trail for the clinical use of rapamycin as a geroprotecTOR

Welcome back Vitalians and please join us in congratulating the Oisín Biotechnologies team for passing the VitaDAO token holder vote with 86.47% voting in favour! This proposal was an assessment of a multi-asset longevity biotech...
Read more
Introducing the VitaDAO Members Portal
November 3, 2023
Awareness
Longevity
Introducing the VitaDAO Members Portal

VitaDAO is partnering with leading companies to offer substantial discounts on their products and services, exclusively for our members.

Our members are enthusiastic advocates of a range of longevity and health optimization interventions in their daily lives. Depending on age and health status, individuals may have different goals when exploring longevity protocols. This can range from improving athletic performance to enhancing metabolic health to preventing osteoporosis and more. We want to give them the opportunity to access health products and services at a discounted price. Some examples include discounts on wearables, at-home health test kits, supplements, biohacking tools, and more.

In addition to health products and services, we also offer discounts on common expenses such as hotel stays, short- to long-term rentals, coworking spaces, academic publications, and more.

We’re launching our portal with discounts on

  • GlycanAge — Discover your biological age, make lifestyle improvements, and track your success with GlycanAge tests.
  • Sonder — Premium, stylish, and fully serviced accommodations in over 40 vibrant cities worldwide.
  • Bon Charge — From blue light blocking glasses to sauna blankets to PEMF mats to red light therapy devices, Bon Charge offers everything you need to protect and optimize your health in this digital age.

More products and services will be added over the following weeks and months. For example, discounts on health programs with access to specialists, including medical doctors. Future services also include discounted access to exclusive events and curated meetings with leaders in the longevity space.

The 3 we’re launching with

GlycanAge

Get 31% off GlycanAge tests to determine your biological age

Extend your health span today. Discover your biological age, make lifestyle improvements, track your success.

GlycanAge is a biological age test paired with expert advice to help guide your wellness. It determines your biological age by measuring chronic inflammation in your system — which is directly related to your lifestyle. As a member, you now have discounted access to a groundbreaking approach that empowers you to take charge of your health and longevity.

GlycanAge leverages cutting-edge technology, providing valuable insights into your biological age. With this information, you can make informed decisions about your lifestyle, nutrition, and wellness strategies to promote a healthier, more vibrant life.

GlycanAge is based on more than 25 years of scientific research and over 200,000 tests. Their scientists regularly publish in world renowned Medical Journals.

Collect your sample. Collect and return your dry blood sample using an at-home test kit.

Receive your results. It takes about three weeks for the lab to analyse your sample and generate your report.

Talk to experts. A consultation is included with each test. Specialists will help you review your results and suggest lifestyle changes to help you improve your health.

GlycanAge has pricing and plans affordable for anyone! Choose the plan that suits your needs!

Bon Charge

Get 15% off science-backed wellness products to optimize your sleep, wellbeing and recovery

Premium. Science-Backed. Effective. From blue light blocking glasses to sauna blankets to PEMF mats to red light therapy devices, Bon Charge offers everything you need to protect and optimize your health in this digital age.

As a VitaDAO member, you get 15% off Bon Charge’s wellness products to safeguard your health while you work and play in the digital world.

From better sleep and relaxation to faster recovery and optimized wellbeing, Bon Charge’s science-backed wellness products are here to elevate your life. Whether you’re at your desk or on the go, our partnership ensures you have access to premium products that support your well-being.

Work and relax with confidence, knowing that you have the best in health protection with Bon Charge.

Sonder

Book a Sonder in 40+ cities up to 50% less

Looking for an exceptional stay for your next DeSci conference? Look no further. VitaDAO has teamed up with Sonder to make your travel experience a little better.

As part of this exclusive collaboration, our valued members now have the opportunity to book Sonder accommodations in over 40 vibrant cities worldwide at a discount of up to 50%. This partnership aims to elevate your travel experience, offering you premium, stylish, and fully serviced accommodations at unbeatable prices.

Booking a Sonder through our platform couldn’t be easier. With a variety of options, from cozy studios to spacious apartments, you can choose the perfect accommodation that suits your needs.

Browse the selection of Sonder properties in your desired destination. When you’re ready to book, simply use the discount code below for up to 50% off, making your stay both affordable and memorable.

Say hello to an elevated travel experience with Sonder and our exclusive partnership. Start exploring the world with comfort and savings today!

Visit the VitaDAO Members Portal https://vitadao.global

VitaDAO is partnering with leading companies to offer substantial discounts on their products and services, exclusively for our members.
Read more
Shining a Light on Optogenetics: An Innovative Approach to Combat Aging
November 1, 2023
Maria Marinova
Awareness
Longevity
Shining a Light on Optogenetics: An Innovative Approach to Combat Aging

 

Mitochondria in the spotlight: Powerhouses Dimming with Age?

In the intricate ballet of cellular activity, mitochondria often play a leading role. These double-membraned organelles are frequently referred to as the "powerhouses" of the cell, converting nutrients into energy that fuels nearly everything we do. But, as with all things, age takes its toll on them (Clemente-Suárez et al., 2023).

Mitochondrial dysfunction is one of the central features of aging (Payne & Chinnery, 2015). Many strategies designed to prolong life and maintain good health into old age focus on these cellular structures. From diets to drugs like metformin and rapamycin, various interventions aim to tweak mitochondrial metabolism (Amorim et al., 2022). Interestingly, many of these strategies work by reducing mitochondrial activity. This may sound counterintuitive, but certain studies have shown that decreasing specific aspects of mitochondrial processes can actually increase an organism's lifespan (B. Hwang et al., 2012; Campos et al., 2021). However, there's a catch. Reducing mitochondrial function often comes at the cost of the organism's vitality and its ability to handle stress (Aragoni Da Silva et al., 2023). Thus, the question remains: can such an approach be realistically applied to humans? 

From Plants and Bacteria to Humans: A Radical Shift

All animals, including humans, need to consume other organisms for fuel, however some life forms, such as plants, algae and cyanobacteria have evolved to create their own fuel from sunlight, by mastering the art of solar power at the cellular level (Havurinne & Tyystjärvi, 2020).

It is thought that all organisms on the planet share a common ancestor, and thus many cellular mechanisms are universal, such as DNA for storing genetic information, ribosomes for producing proteins or the use of ATP as cellular energy currency. This allows opportunities for mechanisms from one organism to be utilised in another - take, for example, the CRISPR/Cas9 system, adapted from bacteria, that has revolutionized genetic editing in animals (Kick et al., 2017).

 A Bright Idea: Optogenetics

To generate energy, our cells rely on a remarkable mechanism known as the electron transport chain. This process uses energy to push protons across the mitochondrial membrane, creating a kind of energy imbalance. Later, these protons can flow back through special channels, triggering the production of ATP. Think of it like how a battery operates. However, as we age, this process can become dysregulated, leading to a decrease in efficiency and even leakage of toxic free radicals.

The proposed strategy revolves around an engineered protein from fungal origin (Leptosphaeria maculans) called mtON, which is light-sensitive protein that pumps the aforementioned protons through the electron transport chain. When exposed to light, mtON pumps protons across the mitochondrial membrane, aiding in the creation of ATP, the cell's primary energy molecule (Tiwary et al., 2023). What's fascinating here is that this system allows a cell to produce energy from light, circumventing the regular process, and potentially rescuing dysfunctional mitochondria in aging cells (Berry & Wojtovich, 2020). This is not mere speculation. Groundbreaking experiments have already demonstrated that this light-based approach can enhance lifespan in worms (Berry et al., 2022).

Harnessing the power of light for energy transduction in our cells could have profound effects on the way we age. By rejuvenating aging cells, the mtON approach offers a ray of hope for tissues that suffer from declining mitochondrial function. Aging cells, which struggle to produce adequate ATP due to faltering mitochondria (Berry & Kaeberlein, 2021), might find a new lease on life with the introduction of mtON.

Furthermore, aging isn't just about the passing of time; it's also about the wear and tear our cells experience. With ATP potentially being produced from light, we might see a decrease in overall cellular metabolic activity. This translates to reduced waste and a subsequent slowdown in the accumulation of cellular damage. Importantly, there might also be a drop in oxidative stress, a significant contributor to the aging process (Warraich et al., 2020).

One of the most bold potential implications revolves around our dietary needs. While this sounds like science fiction, maybe in the future, the light-driven energy transduction could even reduce our reliance on food for energy. This could allow us to enjoy the benefits associated with calorie restriction, which is known to promote health and longevity (Dorling et al., 2020), without the associated downsides of reduced energy intake. This shift might redefine our relationship with food and our understanding of metabolic health. But enough daydreaming for now…

The Future is Bright

To sum up, using light as an alternative energy source at the cellular level could pave the way for groundbreaking treatments against aging. While this hypothesis is still in its early stages, its potential is vast. Perhaps one day, instead of just basking in the sun for warmth, we might be doing it to recharge our cells!

Proposal author and Longevity Prize 3rd place winner: Dr Shahaf Peleg and Dr Andrew Wojtovi

Writer: Maria Marinova

Editors: Adrian Matysek, Rhys Anderson

References 

Amorim, J. A., Coppotelli, G., Rolo, A. P., Palmeira, C. M., Ross, J. M., & Sinclair, D. A. (2022). Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nature Reviews Endocrinology, 18(4), 243–258. https://doi.org/10.1038/s41574-021-00626-7

Aragoni Da Silva, J., Rolland, Y., Martinez, L. O., & De Souto Barreto, P. (2023). Mitochondrial Dysfunction and Intrinsic Capacity: Insights From a Narrative Review. The Journals of Gerontology: Series A, 78(5), 735–742. https://doi.org/10.1093/gerona/glac227

B. Hwang, A., Jeong, D.-E., & Lee, S.-J. (2012). Mitochondria and Organismal Longevity. Current Genomics, 13(7), 519–532. https://doi.org/10.2174/138920212803251427

Berry, B. J., & Kaeberlein, M. (2021). An energetics perspective on geroscience: Mitochondrial protonmotive force and aging. GeroScience, 43(4), 1591–1604. https://doi.org/10.1007/s11357-021-00365-7

Berry, B. J., Vodičková, A., Müller-Eigner, A., Meng, C., Ludwig, C., Kaeberlein, M., Peleg, S., & Wojtovich, A. P. (2022). Optogenetic rejuvenation of mitochondrial membrane potential extends C. elegans lifespan. Nature Aging, 3(2), 157–161. https://doi.org/10.1038/s43587-022-00340-7

Berry, B. J., & Wojtovich, A. P. (2020). Mitochondrial light switches: Optogenetic approaches to control metabolism. The FEBS Journal, 287(21), 4544–4556. https://doi.org/10.1111/febs.15424

Campos, J. C., Wu, Z., Rudich, P. D., Soo, S. K., Mistry, M., Ferreira, J. C., Blackwell, T. K., & Van Raamsdonk, J. M. (2021). Mild mitochondrial impairment enhances innate immunity and longevity through ATFS‐1 and p38 signaling. EMBO Reports, 22(12), e52964. https://doi.org/10.15252/embr.202152964

Clemente-Suárez, V. J., Redondo-Flórez, L., Beltrán-Velasco, A. I., Ramos-Campo, D. J., Belinchón-deMiguel, P., Martinez-Guardado, I., Dalamitros, A. A., Yáñez-Sepúlveda, R., Martín-Rodríguez, A., & Tornero-Aguilera, J. F. (2023). Mitochondria and Brain Disease: A Comprehensive Review of Pathological Mechanisms and Therapeutic Opportunities. Biomedicines, 11(9), 2488. https://doi.org/10.3390/biomedicines11092488

Dorling, J. L., Martin, C. K., & Redman, L. M. (2020). Calorie restriction for enhanced longevity: The role of novel dietary strategies in the present obesogenic environment. Ageing Research Reviews, 64, 101038. https://doi.org/10.1016/j.arr.2020.101038

Havurinne, V., & Tyystjärvi, E. (2020). Photosynthetic sea slugs induce protective changes to the light reactions of the chloroplasts they steal from algae. eLife, 9, e57389. https://doi.org/10.7554/eLife.57389

Kick, L., Kirchner, M., & Schneider, S. (2017). CRISPR-Cas9: From a bacterial immune system to genome-edited human cells in clinical trials. Bioengineered, 8(3), 280–286. https://doi.org/10.1080/21655979.2017.1299834

Mansouri, M., Strittmatter, T., & Fussenegger, M. (2019). Light‐Controlled Mammalian Cells and Their Therapeutic Applications in Synthetic Biology. Advanced Science, 6(1), 1800952. https://doi.org/10.1002/advs.201800952

Payne, B. A. I., & Chinnery, P. F. (2015). Mitochondrial dysfunction in aging: Much progress but many unresolved questions. Biochimica et Biophysica Acta (BBA) - Bioenergetics, 1847(11), 1347–1353. https://doi.org/10.1016/j.bbabio.2015.05.022

Tiwary, V., Galow, A.-M., Wojtovich, A. P., & Peleg, S. (2023). Using light to drive energy transduction in metazoan aging. Trends in Biochemical Sciences, 48(11), 920–922. https://doi.org/10.1016/j.tibs.2023.08.010

Warraich, U.-A., Hussain, F., & Kayani, H. U. R. (2020). Aging—Oxidative stress, antioxidants and computational modeling. Heliyon, 6(5), e04107. https://doi.org/10.1016/j.heliyon.2020.e04107

 

In the intricate ballet of cellular activity, mitochondria often play a leading role. These double-membraned organelles are frequently referred to as the "powerhouses" of the cell, converting nutrients into energy that...
Read more
VitaDAO Letter: The First DAO with a Biotech Spinoff
October 15, 2023
Sarah Friday
Awareness
VitaDAO Letter: The First DAO with a Biotech Spinoff

Despite the bear market, the VitaDAO community continues to push the boundaries of decentralized science (DeSci). This month has been particularly exciting, with developments such as the funding of ExcepGEN and the launch of MatrixBio. In this edition, we bring you the latest highlights:

  • Matrix Bio: VitaDAO’s biotech startup 
  • How to buy $VITA on Optimism 
  • Pfizer's representation at DeSci Berlin 
  • Recent VitaDAO proposals 
  • Awareness Working Group Contributor: Victoria Forest 

🧪A Landmark Achievement: VitaDAO’s Biotech Startup 

Ever heard of naked mole rats (NMR)? These long-lived rodents are cancer-resistant, thanks in part to the abundance of high molecular weight hyaluronic acid (HMW-HA) in their tissues. Previous research by the Gorbunova lab has demonstrated this link between cancer resistance and the abundance of HMW-HA.

This month, VitaDAO proudly became the FIRST DAO to kickstart a biotech company. This firm, Matrix Biosciences, will use insights from the Gorbunova lab to leverage the anti-cancer and pro-longevity properties of HMW-HA from NMR in humans. The creation of Matrix Biosciences marks a significant stride in both the field of longevity research and VitaDAO's portfolio.

🤑$VITA on Optimism 

$VITA is now on Optimism! Have you ever tried to make a transaction on the Ethereum mainnet and found yourself thinking “Not in this economy⛽️😫.” Say farewell to high Ethereum mainnet gas costs and take advantage of the ability to purchase $VITA on Optimism!

Unsure how to get started? Check out our step-by-step guide on acquiring $VITA on Optimism. Once you have $VITA, you can even use Velodrome to provide liquidity on Optimism. Additionally, $VITA is also accessible on the Gnosis chain. The expansion to L2s will hopefully make obtaining $VITA cheaper, more convenient, and more accessible for all HODLers! 

📚Hypothesis Prize: Exploring Winning Topics 

Maria Marinova has been busy collaborating with winners of the Longevity Prize. First-place winner Carlos Galicia suggested studying rejuvenation during embryogenesis as a means to combat age-related decline. Maria and Carlos explore this concept in their article, “The Key to Longevity Could Be Hidden in Embryos.”

Second-place winner Rakhan Aimbetov proposed focusing on proteostasis disruption in aging. Specifically, he focused on the role of methylglyoxal (MGO), a byproduct of sugar metabolism. In “Sugar Byproducts and How They May Influence Aging,” Maria and Rakhan explain how sugar remnants may contribute to aging. Stay tuned for the third article in the series featuring Shahaf Peleg's investigation of mitochondrial dysfunction! 

🌎VitaDAO in the Spotlight 

  • VitaDAO's global impact has earned a mention in Nature. We are proud to be one of several decentralized autonomous organizations contributing to innovative research funding methods. The transparency of blockchain and the collaborative spirit of our community has been recognized and featured in Nature Biotech, which notes: "Decentralized autonomous organizations are growing as alternative research funding models, but are also strong scientific communities. We should get on board."

“Pharma has deep pockets... it’s true, but I think the money for early-stage R&D or external partnerships is not what you might think.”

📣Community Approved: What YOU voted for!  

Thanks to the support of the VitaDAO community, three proposals successfully passed this month! These proposals include:  

VDP-120 (An assessment of Oisín Biotechnologies): Oisín Biotechnologies is a multi-asset longevity biotech company pioneering genetic medicines to combat sarcopenia and other age-related diseases. Oisín is working to deliver DNA and RNA through a Fusogenix Proteo-Lipid in (PLV) platform, to alter cell behavior. The company’s lead candidate aims to build muscle without exercise, which could be used as a therapy to strengthen older people. VITA token holders agreed with the Longevity-Dealflow WG’s and senior reviewers' assessment of this company.

VDP-119 (Proposal to empower a member services squad): The formation of the Member Services Squad as anticipated in VDP-69 was intended to create a team to focus on the development of new offerings for VitaDAO token-holders and contributors. This proposal served to empower a Member Services Squad to roll out new services and gave the squad the authority to make necessary legal and logistical decisions to launch a “Member Services” portal.

VDP-118 (An assessment of GERO): This proposal was an assessment of GERO, a company using AI and ML to analyze datasets to discover novel treatments for chronic age-related diseases. To accomplish their goal of doubling human health- and lifespan within the current generation, the company has trained a large generative model of human health on a large dataset of longitudinal, real-world medical histories. The model is being used to predict future health outcomes similarly to how Large Language Models (LLMs) anticipate the next word in a sentence. Unlike LLMs, GERO’s models are physics-based and interpretable, allowing exploration of the relationship between the aging process and diseases.


💪Exercise Your Right to Vote

Visit VitaDAO’s governance hub (Discourse) to engage with, vote on, and discuss proposals before they are moved to Snapshot. Together, we can shape the future of VitaDAO and accelerate decentralized science.

👋Meet Awareness Working Group Contributor: Victoria Forest 

Victoria Forest, an integral member of VitaDAO’s Awareness and SciComm Working Groups, plays a role as VitaDAO’s DeSci illustrator and Instagram manager. When not contributing to VitaDAO, Victoria enjoys working as a freelance web designer, learning about longevity, and practicing pilates. In my interview with Victoria, we dive into her journey with VitaDAO, her interest in longevity, and her role within the DAO community.

How did you first get involved in VitaDAO? 

I found out about VitaDAO before the official launch in 2021. At that time, I wasn't yet involved in the web3 space. A few months later, I joined the community calls, became more curious about the vision, and got excited to understand how DeSci works and potentially be part of this movement. It was great to spend some time with the team at the first offsite, interview them (Meet the Vitalians YouTube list), learn about the mission, connect, and from there, choose to dedicate even more time to it.

What is your role within VitaDAO?

I'm a member of the Community & Awareness working group, where my responsibilities include developing media materials, creating illustrations, maintaining our social media accounts to keep our community informed, and interviewing experts in the field, among other tasks.

What do you feel is VitaDAO’s biggest strength compared to traditional longevity biotech startups?  

VitaDAO’s primary advantage is its decentralized and community-centered approach. Unlike conventional startups, VitaDAO leverages blockchain and decentralized technologies to engage a wider group of contributors, investors, and experts globally. This approach encourages increased inclusivity, collaboration, and diversity in the quest for longevity research and biotech innovation. It also allows for a more transparent and efficient allocation of resources and decision-making, potentially accelerating progress in the field.

What has surprised you the most about VitaDAO? 

What has impressed me the most about VitaDAO is the remarkable transparency in how the DAO operates, the openness of its team members, and the effective leverage of blockchain technology to bring tangible value to the real world.

What advice do you have for other Web Designers who are interested in getting involved in DeSci? 

I'd say, first and foremost, be curious about the space. Take the time to learn how blockchain and decentralization can be applied to real-world problems. Once you've got a grasp of that, seek out a community whose mission resonates with you, something you genuinely care about. If you find it fulfilling and it aligns with your values, then dive in. Attend conferences, go to meetups, and start networking. You'll likely discover the perfect place where you can contribute your skills and, at the same time, feel great about what you're doing.

Any closing thoughts for readers? 

Think about the future you want, both for yourself and your loved ones. Can you picture yourself growing old and experiencing frailty and suffering? How about witnessing your parents and grandparents endure the pains of aging and age-related diseases? We need to do more than what we're currently doing. Talk to everyone you know, and share with them the possibility of having a longer healthspan and potentially a longer lifespan. Think about the abundant world we can create if we unite our efforts right now, at this critical moment. Also, consider joining us at Vitalia in Prospera, a movement dedicated to initiating a longevity network society.

🤝Thanks for Reading! 

Want to stay up to date with all that's going VitaDAOn? Join us on Discord, subscribe to our Twitter, and follow our Instagram! You can also find updated information on VitaDAO's funded projects and an updated treasury dashboard on our website

Despite the bear market, the VitaDAO community continues to push the boundaries of decentralized science (DeSci). This month has been particularly exciting, with developments such as the funding of ExcepGEN and the launch of MatrixBio.
Read more
Deciphering Science's Secrets: Lifespan, UPR, and Lysosomal Pathwayswith Dr. Arwen Gao on The VitaDAO Aging Science Podcast
October 15, 2023
Podcast
Deciphering Science's Secrets: Lifespan, UPR, and Lysosomal Pathwayswith Dr. Arwen Gao on The VitaDAO Aging Science Podcast

In today’s podcast I talked to Dr. Arwen Gao about the working conditions in science and, of course, also about her research. First, we talked about scholarships, strikes, unions and bureaucracy before moving on to our research related topics like antibiotics that extend lifespan, the unfolded protein response (UPR) and a novel lysosomal surveillance pathway called LySR that she helped to identify.

You may want to skip to the first 20 minutes if you only want to hear about the hard science rather than our discussion of working conditions and the day-to-day life of a scientist.

Short Bio – Dr. Arwen Gao

Arwen was awarded an AMC PhD scholarship to pursue her PhD thesis, focusing on the metabolic control of aging in C. elegans, and earned her PhD degree in the fall of 2018 at the University of Amsterdam in the Netherlands. She continued her research on mitochondrial stress response and its role in metabolism and aging, under the supervision of Prof. Johan Auwerx (EPFL, Switzerland) and supported by a post-doctoral fellowship (2019 Accelerator price winner). After this postdoc, she moved back to the Netherlands, where she is currently a junior group leader at the Amsterdam UMC - Location AMC. Her research focuses on lysosomes, lipids, aging, and translational projects that can benefit patients with lysosome disorders.

A few helpful notes follow below explaining the concepts behind the topics we covered.

Worms as a model in aging research

There are many good reasons to use the worm (C. elegans) for preclinical aging research. For one, it is cheap to study and requires no ethics approval. These worms have a conveniently short lifespan of around 30 days and well understood genomes. In addition, many tools are available to manipulate their genomes including RNA-interference libraries to study the knock-down of genes in a high-throughput fashion.

There is also a surprising amount of conservation between human and worm genomes. Our genes often are very similar to worm genes suggesting that worms might be a suitable model to study some aspects of human aging. For example, FoxO signaling and the Igf-1 pathway have been first identified in worms and later confirmed to be important longevity pathways in mammals as well. 

In the podcast we discussed how worm researchers also struggle with the same problems as mouse researchers. Similarly, to the situation with mice, most worm aging research is done in a single, highly lab-adapted and inbred strains. The one we often use is called N2 (first isolated in “Bristol”). We discussed the problems with inbred mice in a recent podcast with Rich Miller and in this podcast I discuss the C. elegans perspective with Arwen Gao and some solutions to this, including the generation of inbred, yet diverse, recombinant “panels”, composed of different strains with varying genetics. A simpler - somewhat less sophisticated - solution is to use at least two different strains to confirm your findings.

UPR and doxycycline - an unfolding story

Folding is the process by which linear protein chains are assembled into functioning 3D structures. Whenever something goes wrong with this folding process, this activates a so-called unfolded protein response (UPR). There are at least two distinct unfolded protein responses that occur in different cellular compartments, one in the mitochondria termed (UPRmt) and the other the canonical UPR in the endoplasmic reticulum (ER). Conceptually, the UPR can be considered a “cry for help” from an organelle. Since eukaryotic cells are very large and compartmentalized there is a need for this kind of cross-talk between different organelles and the nucleus which orchestrates said help (via mRNA transcription).

To make matters more confusing, the UPR, can be considered as part of the integrated stress response (ISR) which downstream is characterized by phosphorylation of a translational initiation factor (eIF2alpha) leading to selective translation of certain proteins, including the famous ATF4 transcription factor which is elevated in long-lived mouse models. Another way of looking at this is in terms of partial overlap. One branch of the canonical UPR, acting via a protein called PERK, is also an arm of the integrated stress response. However, the UPR has many other outputs and the integrated stress response has many other inputs, including the UPRmt.

Intriguingly, doxycycline is an antibiotic that activates the UPRmt thereby extending lifespan in C. elegans as shown by Gao et al. (2022). This is consistent with a naïve notion that improved folding and the UPR stress response should benefit lifespan. Since this is real biology, however, it is also the place where intuitions go to die. Other work suggests that inhibition of the integrated stress response is beneficial, at least in worms (Derisbourg 2021). This is the article I mentioned during the podcast from the Denzel group. While interesting, this does not – and should not – dissuade us from putting this idea to the test in the mammalian system. And, indeed, the famous mouse interventions testing program (ITP) is running a cohort with an integrated stress response activator (galofuginone) in mice to see whether it will extend lifespan.

We live in interesting times indeed!

For further reading we have attached links to some freely accessible reviews.
Melber and Haynes 2018 and Pakos‐Zebrucka et al. 2016 (here esp. Fig. 1) under further reading.

You can consider the UPR as one input for the integrated stress response. Source: Pakos‐Zebrucka et al. 2016, https://www.embopress.org/doi/full/10.15252/embr.201642195

The lysoosomal surveillance response – just another cry for help?

Arwen and others discovered a new pathway responsible for sensing lysosomal stress when they knocked down different VATPase subunits in the worm (Li et al. 2022). This pathway appears to be independent of autophagy, mTOR and the TFEB biogenesis pathway (called HLH-30 in the worm). While it is not entirely clear how the signal is transmitted from the lysosome to nucleus, they do show that it converges on a specific transcription factor from the GATA family. One can speculate that VATPase knock-down leads to reduced acidification and proteolysis in the lysosome which somehow leads to nuclear GATA translocation. While superficially, this may not be the whole story since only specific VATPase subunits seem to be involved in triggering the LysR. Hopefully, once we figure out the players we will be able to use this knowledge to generate novel autophagy inducers to promote lifespan and health.

Scientific research - between madness and dedication

In this podcast we also talk about the working conditions of scientists and PhD students. Arwen mentioned that the conditions are quite favorable in the Netherlands and in the labs where she worked. However, we are also aware of horror stories like postdocs who were regularly "sleeping in the lab" (undisclosed German lab) or students "buying groceries" for their supervisor (somewhere in the Netherlands or Switzerland, presumably). While we do both agree that passion is an important ingredient to a science career, (self-)exploitation should not be part of the deal. Sadly, these stories are more common than they should be. Grad school in the USA, for example, has the reputation of being very taxing. Here as told from the perspective of chemists:

https://www.science.org/content/blog-post/more-grad-school-pressures
https://www.science.org/content/blog-post/industry-vs-academia-mental-aspect
http://notthelab.blogspot.com/2013/01/is-graduate-school-in-chemistry-bad-for.html

Personally, I believe society should value scientists more than we currently do and pay significantly higher salaries. Whenever the pay is high, there is less exploitation of cheap labor and more automation of tedious labor.

However, not everything is bleak and we do not want to discourage anyone from pursuing a career in science. Conditions are improving and they are already quite good at certain institutions and in some countries. If you want to learn more which countries have a relaxed work culture and good pay, this podcast might be for you!

The host – brief bio

Kamil Pabis, MSc is an aging researcher and longevity advocate with several years of experience in the aging field that spans multiple countries. Among other projects, Kamil worked on long-lived dwarf mice in Austria, on mitochondrial disease and aging in the UK, and finally on the bioinformatics of aging in Germany and Singapore. Presently, he is involved in several projects related to science communication and translational aging research.

References and further reading

Derisbourg, Maxime J., et al. "Mutagenesis screen uncovers lifespan extension through integrated stress response inhibition without reduced mRNA translation." Nature Communications 12.1 (2021): 1678.

Arwen Gao at ARDD2022: A lysosomal surveillance response (LySR) that extends healthspan
https://www.youtube.com/watch?v=TiIs2stK2Iw&t=612s
see also biorxiv: https://www.biorxiv.org/content/10.1101/2022.06.13.495962v1

Gao, Arwen W., et al. "Multi-omics analysis identifies essential regulators of mitochondrial stress response in two wild-type C. elegans strains." Iscience 25.2 (2022): 103734.

Melber, Andrew, and Cole M. Haynes. "UPRmt regulation and output: a stress response mediated by mitochondrial-nuclear communication." Cell research 28.3 (2018): 281-295.
https://www.nature.com/articles/cr201816

Pakos‐Zebrucka, Karolina, et al. "The integrated stress response." EMBO reports 17.10 (2016): 1374-1395.
https://www.embopress.org/doi/full/10.15252/embr.201642195

n this episode of The VitaDAO Aging Science Podcast, we embark on a fascinating journey through the realms of longevity and cellular health with Dr. Arwen Gao. Dr. Gao, a leading authority in the fields of aging research, metabolism, and lysosomal...
Read more
September Longevity Research Newsletter
October 10, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
September Longevity Research Newsletter

Introduction

Welcome back Vitalians, it’s a big week for VitaDAO as we’ve just become the first DAO to kickstart a biotech company, MatrixBio. MatrixBio explores hyaluronic acid-based compounds, aiming to pioneer in cancer and aging treatments, utilizing insights from cancer-resistant naked mole rats in collaboration with Prof Vera Gorbunova.

Other exciting news: we have a new portfolio company. Please join us in congratulating the GERO team for passing the VitaDAO token holder vote with 99.98% voting in favour! This proposal was an assessment of a Machine Learning Platform for Age-Related Disease Targeting & Drug Discovery, with a view to creating therapeutics to slow down human aging and eliminate root causes of age-related diseases.

If you haven't already, please consider signing the Dublin Longevity Declaration.

We currently invest large sums of money trying to cure the leading causes of death such as heart disease, cancer and neurodegeneration, however the most common risk factor for all of these is age. 

"For millennia, the consensus of the general public has been that aging is inevitable."

However, the last few decades of research has shown that aging isn't an immutable phenomenon, rather it is something that can be understood and targeted. 

The Dublin Longevity Declaration is a consensus recommendation - already signed by a large number of leading figures in the aging field - to immediately expand research on extending healthy human lifespans.

Researchers are constantly exploring numerous avenues to rejuvenate organisms to younger healthier states, from epigenetic reprogramming, to stem cell therapies. This month we are excited to bring you an interview with a pioneer in the space, Professor Irina Conboy, who has made numerous contributions to the rejuvenation biology field, including the discovery of the rejuvenating effects of young blood through parabiosis. In this issue we’re going to delve deeper into her latest publication highlighted in our Hot Picks. To explore the topic further we are hosting a journal club with Irina and Michael Conboy on fail-testing DNA methylation clocks and developing a “noise” barometer for measuring epigenetic pressure - Oct 16th, 6 pm CET.  Enjoy and we hope to see you there! 

Longevity Literature Hot Picks

Preprint Corner

We have now entered Q4, but you still have until Friday 13th October to nominate great longevity preprints from Q3 2023 to be featured in the next issue of The Longevist! You can now nominate preprints using our new snazzy form and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

And if you haven’t already seen it, here is our second issue of The Longevist, featuring the best longevity preprints from Q2 2023.

Check out our last batch of preprints of Q3 with these September submissions. These will each be entered into the Q3 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

Methylglyoxal Affects Translation Fidelity

A pre-print by the 2nd palace winner of the Longevity Prize Rakhan Aimbetov expands on the topic of his submission. We love to see our initiative leading to tangible results, publications and research… If you want a lighter read on the topic we just published a Sci Comms article based on the topic of his prize entry: Sugar Byproducts and How They May Influence Aging

Tardigrades dramatically upregulate DNA repair pathway genes in response to ionizing radiation

Ketogenic diet administration later in life improves memory and regulates the synaptic cortical proteome through the cAMP/PKA signaling pathway in aging mice

Proteomic aging clock predicts mortality and risk of common age-related diseases in diverse populations

Longevity interventions in Titan mice attenuate frailty and senescence accumulation

Hemin decreases cellular aging and enhances healthspan and lifespan through the AMPK pathway

B Cells Promote T Cell Immunosenescence and Mammalian Aging Parameters

Molecular and phenotypic blueprint of the hematopoietic compartment reveals proliferation stress as a driver of age-associated human stem cell dysfunctions

A hematopoietic stem cell subset that retains memory of prior inflammatory stress accumulates in aging and clonal hematopoiesis

Selective advantage of mutant stem cells in clonal hematopoiesis occurs by attenuating the deleterious effects of inflammation and aging

Genetic architecture of telomere length in 462,675 UK Biobank whole genome sequences

A blood biomarker of accelerated aging in the body associates with worse structural integrity in the brain: replication across three cohorts

Sex Chromosomes and Gonads Shape the Sex-Biased Transcriptomic Landscape in Tlr7-Mediated Demyelination During Aging

Stability of Genomic Imprinting and X-Chromosome Inactivation in the Aging Brain

Deep learning-based prediction of one-year mortality in the entire Finnish population is an accurate but unfair digital marker of aging

Cellular aging is accelerated in the malignant clone of myeloproliferative neoplasms

Published Research Papers

Fail-tests of DNA methylation clocks, and development of a noise barometer for measuring epigenetic pressure of aging and disease

Elastic Net DNA methylation clocks show low prediction accuracy for age and health. The study suggests that stable cytosine methylation over lifespan indicates biological age. Dysregulated cytosines, measured as standard deviations, act as a "noise barometer" for aging and disease.

Eye Diseases and Senescence

X-irradiation induces senescence in human corneal surface cells, leading to disruptions in the epithelial barrier, and in aging mice, the accumulation of senescent ocular cells contributes to chronic inflammation and barrier decline, suggesting these senescent cells could be a therapeutic target to treat aged ocular surface issues.

Association of biological age with health outcomes and its modifiable factors

Using the UK Biobank data, a multi-dimensional biological age model was developed. Accelerated aging correlates with health risks. 35 modifiable factors affecting aging were identified, notably pulmonary functions and body mass. 

The fruit fly acetyltransferase chameau promotes starvation resilience at the expense of longevity

A mutation in Drosophila's "chameau" gene extends lifespan but reduces weight and starvation resistance. Despite its longevity benefits, poor nutrient stress handling likely keeps this mutation from being evolutionarily dominant.

And the press release: Sometimes beneficial, sometimes damaging: The double role of the enzyme chameau

Intact mitochondrial function in the setting of telomere-induced senescence

Mitochondria are crucial for cell health. This study explored the impact of telomere issues on mitochondria. By deleting the TRF2 protein, cell aging was induced, but mitochondrial function remained strong. Thus, even with telomere problems, mitochondria can support cell processes.

Plasma metabolomic profiles associated with mortality and longevity in a prospective analysis of 13,512 individuals

In a study of over 13,500 participants, certain plasma metabolites were linked to mortality and longevity. Elevated levels of metabolites like N2,N2-dimethylguanosine indicate higher mortality, while others like L-serine suggest increased longevity. The study implies specific metabolites can predict mortality, but the exact mechanisms remain undetermined.

Leveraging the secretory machinery to eliminate senescent cells

Senescent cells, which increase with age, contribute to tissue deterioration. A genomic screen showed that targeting the secretory actions of these cells in older mice can eliminate them. Suppressing the YAP–TEAD complex and reducing endoplasmic reticulum activity makes senescent cells more prone to apoptosis (cell death).

Death-seq identifies regulators of cell death and senolytic therapies

"Death-seq" is a new CRISPR screen designed to identify cell death mechanisms. It enhances cell death research, pinpointing enhancers for the senolytic ABT-263 and uncovering new inducers using ABT-199. This method reveals insights into cell death pathways and potential drug targets for conditions like senescence, cancer, and fibrosis.

Transcriptional and epigenetic decoding of the microglial aging process

Researchers studied aging effects on microglia, the brain's immune cells. Using mice, they found significant age-related changes, including unexpected sex differences. By creating a model to accelerate microglial aging, they discovered that aged microglia alone can cause cognitive decline, highlighting their role in brain function over time.

Pleiotropic influence of DNA methylation QTLs on physiological and ageing traits

Researchers identified a locus, meQTL.5a, in BXD mouse strains that influences DNA methylation patterns. This locus, combined with aging, results in an "aged methylome" for certain strains. It's linked to gene traits, body weight, lipid levels, and lifespan. 

Complete or Culprit-Only PCI in Older Patients with Myocardial Infarction

A NEJM study of 1445 older patients with myocardial infarction and multivessel disease, complete revascularization resulted in fewer adverse events at 1 year compared to only treating the culprit lesion. Primary outcomes occurred in 15.7% of the complete-revascularization group versus 21.0% in the culprit-only group.

Published Literature Reviews, Hypotheses, Perspectives and more

We need to shift the focus of aging research to aging itself

Mitochondrial degradation: Mitophagy and beyond

Senescent cells at the crossroads of aging, disease, and tissue homeostasis

Biomarkers of aging for the identification and evaluation of longevity interventions

Quercetin and dasatinib, two powerful senolytics in age-related cardiovascular disease

Job board

Alexandra Stolzing at Loughborough University is looking for a Research Associate on a project about volatilome-based signature for age-related recovery & resilience.

The Valenzano lab at the Leibniz Institute on Aging in beautiful Jena, Germany, is looking for two postdocs (microbiome and pop-gen) and a master student. 

David Vilchez has an open position for a lab technician/research assistant in his lab. It would be appreciated if the candidates have a good German level.

The HealthyLongevity.clinic in Florida is looking for a healthcare specialist.

News and Media

A new longevity fund lead by Alex Colville and Laura Deming has launched age1 to support new breakthrough longevity companies to address the field’s core challenges. A bit more from the co-founder and partner Laura: A Letter from Laura Daming

The Economist is finally catching up to the Longevist and has dedicated an entire issue with 8 articles to social and economic aspects of aging: In search of forever

The Economist Intelligence Unit also released: AGEING SHIFT

They introduce the "Scaling Healthy ageing, Inclusive environments and Financial security Today" (SHIFT) Index which benchmarks against a set of national-level leading practices to enable an environment that nurtures longevity and healthy ageing in 19 countries comprising the Group of Twenty (G20).

Featured topics:


Longevity is making it big in the mainstream media this month with another article about Bryan Johnson in Time magazine:

The Man Who Thinks He Can Live Forever

Another mention in Forbes magazine - an article by Alex Zhavoronkov with some specific definitions to distinguish Longevity Biotech companies and a checklist with criteria:

What Is A Longevity Biotechnology Company?

Actually we have one! Read more about the MatrixBio launch:
Pfizer-backed DAO launches community-funded biotech firm

Gulf Sovereign Fund, Hedge Fund Tycoons Bankroll Biotech to Fight Aging
Rejuveron closes a $75 million series B round

A longevity expert shares his exercise routine that he says was inspired by centenarians

The boom of the anti-aging market: How to get people to live to be 120 (and in good health)

If you’re up for some lighter reading here is a humorous piece by the New Yorker: Anti-Aging Secrets of the Mega-Rich

Live to 100? How about 1,000? Why this scientist believes we will one day have lifespans that long

New Epigenetic Clocks May Confirm Extreme Age

A race against time: How science and technology are being used to delay aging

MyAgeNetwork has been awarded UKRI_News global partnership funding to develop collaborative links in ageing research with 14 organisations in USA and Canada. Congrats to Dr Alavian @imperialcollege and colleagues

What is the Blue Zones diet blowing up on Netflix? People who live to 100 eat this way

Resources

Longevity Summit Dublin has uploaded some fantastic talks to YouTube

Our favourites are:

The Million Molecule Challenge for Life Extension by Matt Kaeberlein

Dysregulation of Homeostasis Drives Human Aging by Peter Fedichev 

Robust New Measures of Biological Age by Steve Horvath

And of course our very own:

Accelerating Research with Decentralised Science - Eleanor Davies 

Longevity Network States – Max Unfried

Human Ageing Genomic Resources: updates on key databases in ageing research

What are the key open questions in longevity science?

João Pedro de Magalhães and team have developed a website to compile the major open problems in longevity and aging and put together an initial list of problems, but are aware there will be omissions, so have opened the floor for others to fill in the blanks!

Prizes

A brand new Longevity Prize has launched and accepting application over the next couple of months. The Longevity Funding Innovation Prize initiated and funded by Marat Karpeko is going to award $20,000 USD for the most innovative and potentially impactful longevity funding strategy. The judges want to see a real action plan and tangible solution for real world impact.

Keep in mind the key dates:

  • Kick-Off: 20th September 2023
  • Proposal Closing: 20th December 2023
  • Evaluation Wrap-Up: 20th February 2024
  • Champion Unveiled: 29th February 2024

Conferences

The conference season might be over, but there are still a couple of interesting meetings which we recommend!

Expo and Convention: Independent Ageing 2023

13-15 October, Aichi, Japan

Seno-Therapeutics Summit 2023

7-8 November, Buck Institute for Research on Ageing, Novato, CA, USA

Tweets of the Month

Mikhail V. Blagosklonny: Most people believe that aging is a functional decline caused by accumulation of molecular damage…………..

Healthspanmed: Blagosklonny's theory of cellular hyperfunction has shifted our understanding of aging.

The theory states that aging is not a result of cellular decline or damage accumulation but rather excessive cellular activity he calls hyperfunctions….

Lada Nuzhna: aging research dies one citation at a time:

Podcasts and Webinars

NUS Medicine’s Healthy Longevity Webinar Series

Peter Attiia: Rapamycin: potential longevity benefits, surge in popularity, unanswered questions, and more | David Sabatini, M.D., Ph.D. and Matt Kaeberlein, Ph.D.

Elon is Right. Having to Live Forever is a Curse. Here's How to Solve It

In this episode, Lifespan News explores the diverse perspectives and heated debates triggered by Elon's provocative statements on aging and the prospect of eternal life.

Active Motif Epigenetics Podcast: DNA Damage in Longevity and Ageing (Björn Schumacher)

In this episode Björn Schumacher discusses his research on DNA repair and its impact on ageing.

Interview with Prof. Irina Conboy

Irina Conboy is a Professor at the University of California, Berkeley in the Department of Bioengineering. The Conboy lab currently focuses on broad rejuvenation of tissue maintenance and repair, stem cell niche engineering, elucidating the mechanisms underlying muscle stem cell aging, directed organogenesis, and making CRISPR a therapeutic reality.

What inspired you to enter longevity research?
Self-preservation instinct: in my view, aging is an accumulation of disease and, metaphorically, we are flying breaking down airplanes, so the best one can do is to start working on a parachute, while the plane is still in the air and relatively “healthy”.

Which of the current theories of ageing do you think are the most convincing?
Succumbing to entropy, because of the imperfections in repair of molecules, cells and tissues.

How has the field changed since you started?
Inspired by the work of our colleagues (Cynthia Kenyon, for example) in worms and flies, we shifted the dominant paradigm in mice and people from inevitability of intrinsic aging (telomere shortening, DNA damage, etc.) to reversibility and inducibility of aging by the age of the systemic milieu – circulatory environment that connect all tissues. In other words, we uncovered that aging is a process with specific regulation, understanding this regulation allows attenuating and even reversing the process – the aging.

What mistakes do you think the longevity field has made? 

Exaggeration of the positives, lack of critical questions and reviews, lack of used-to-be frequent “I stand corrected”, tooting own horns “my silver bullet is better than your silver bullet”. 

Other than your own, what do you think have been the biggest/important discoveries in the field?
As mentioned, discovery of life extension in C. elegance by Prof. Cynthia Kenyon (UCSF, and Science leader at Calico), the concept of cellular and tissue senescence that was discovered by Prof. Judy Campisi (LBNL and Buck research institute), the paradigm of bone marrow, hematopoietic stem cells’ and immune aging, discovered by Prof. Irv Weissman (Stanford) and Prof. Sean Morrison (Stanford and UT Southwestern), the aging and rejuvenation of the brain and cognition that was discovered by Prof. Rusty Gage (aka Fred Gage, Salk). 

What advice would you give to people currently working in longevity research?
Take care of science and science will take care of you. 

Which aspect of longevity research do you think requires more attention?
Transition from animals (mice typically) to humans. Many diseases, including those associated with aging, can be cured in mice, but translation to clinic often fails. 

Is ageing a disease?
In my view, we notice aging specifically because tissues, organs and eventually organisms become unhealthy, e.g., it is accumulation of diseases. 

How has the perception of rejuvenation biology evolved since your seminal paper on the rejuvenating effects of young blood through parabiosis in 2005 was published in Nature?
Many intuitively decided that young blood is the key, which in our view, is not accurate.  Young mice with their young DNA, proteins, cells and tissues and 50% of their young blood become aged and senescence in two weeks after a single transfusion with 50% of old blood; and old blood dominates over young in cell culture experiments (https://pubmed.ncbi.nlm.nih.gov/35902645/). How can old mice or old people with their old blood, damaged tissues, cells, and macromolecules become “rejuvenated” by a “young” protein or a blood fraction? To make things more complicated, in contrast to mouse strains, we do not have our identical twins who are always 20 years old and could serve as blood donors. On the other hand, after dilution of old blood plasma (the liquid part of the blood with age-elevated proteins) all the positive effects of heterochronic parabiosis manifest without young blood in mice (https://pubmed.ncbi.nlm.nih.gov/32474458/); and there are rejuvenative effects on the circulating cells and systemic proteome in people after a similar procedure of plasma dilution, called therapeutic plasma exchange (https://pubmed.ncbi.nlm.nih.gov/35999337/. 

You published a new paper on fail-testing biological clocks and a novel approach for quantifying biological age. The study reveals that the change in methylation of cytosines with age is not the determinant for their selection into the clocks and that EN clock's selected cytosines change when non-clock cytosines are removed. Can you explain the significance of these findings and their implications for the development of future DNA methylation clocks and the the stability and relevance of the current ones?
People assume that when their age or health are predicted by so-called “clocks” it is because of the changes in their epigenome or proteome. In reality, the “clocks” is the well-known population statistics method, EN regression, in which computer is used to select and change by weights ~ 200 values from ~500.000. This is done without a biological justification, as a mathematical solution to the least-squares linear fit. In other words, if the results were not what they were, but some values were smaller and other were larger and we added them up for a minute subset of the data, then for this one experiment, Y axis would linearly correlate with X axis. These few datapoints do not have properties of biomarkers; their wights do not reflect the changes in DNAme with age or disease and when other datapoints that EN did not select are removed, EN starts discarding previously selected data, which does not happen with biomarkers. For, example, high blood glucose levels will always be biomarker of diabetes, even if we remove the inflammatory proteins, obesity, etc. We also show that in more than one experiments, predictions might reflect random experimental variation, so people should not rely heavily on being told that the “clock” determined that they are younger by a few months or older by a couple of years.  In support for the lack of biological relevance or even a physical connection, we show that numbers of people living in US can be accurately predicted using the EN DNAme “clock” and in the same vein, Dow Jones industrial, global temperatures, etc. clearly unrelated to epigenetics changes can be predicted from DNA methylation array data, using EN. This is because EN only requires a large data with a weak linearity (could be optical signals from an array or distances between the Galaxies) and a measured parameter, e.g., regular set of numbers. Age, health scores, numbers of people living in US, Dow Jones, are all designated as regular sets of numbers and it is the numbers, not their meaning that is predicted, moreover, without a requirement for the physical connection between X and Y. 

Can you explain in layman's terms the concept of the "noise barometer" and its importance in determining biological age? How does it differ from or complement existing markers of biological age?
EN tells us how to get the straight line between X and Y, in the mathematical realm of adjusted data; in the physical – biological realm we do not age linearly, not as an evenly ticking clock, but with intervals of youthful healthy plateaus and transitions to less health and aging, which become exponential after a serious illness. Can we quantify this and, moreover, using the primary unadjusted data? Search for biomarkers of aging spans decades and $100s of millions, yet it was not very successful, possibly, because individual differences mask the age-imposed changes. In the shift of this paradigm, we posited that it is not the levels, but the dysregulation (the noise) of genes and proteins, which biomarks aging and disease. In a metaphor, a strange noise when you drive a car strongly suggests a problem. In our bodies, the best detectors of the problematic noise are the genes, which are vital for our health (housekeeping genes, for example). Using the standard deviations or the distance from the healthy young mean of these noise detectors (UC Berkeley IP) quantifies the biological age (accurately, based on the actual unadjusted experimental results) and uncovers the natural polynomial line of aging.

Do you think the "noise barometer" can be applied in practical, clinical settings to detect or predict age-related diseases or conditions? Where can “quantifying” biological age can have the most value in clinical practice?
Yes, of course.  As with any new measurement, quantification of biological age opens new possibilities for testing the efficacy of medicine, validating the putative rejuvenative approaches and warning people of “sliding” from the healthy plateau. We expect that noise barometer will be used by the Health Insurance Companies, hospitals and clinicians, academia, and biotech.  In a metaphor, if there were no weight scales and we predicted weight based on the population statistics (clothes, food, body contours) it would be more difficult to get in shape and a person could have delusions of being lighter when they gained 20 pounds. Scales is the device that works on the concept of gravity, noise barometer is the first approach that measures biological entropy.

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful!

This time we leave you with a twitter space recording about the Longevity Funding Innovation Prize, where you can hear first hand from Marat more about how the prize was created, what are the aims, and what would the judges be looking for when assessing applications.

Further Reading

Intact mitochondrial function in the setting of telomere-induced senescence

Age-related changes of skeletal muscle metabolic response to contraction are also sex-dependent

Cross-species comparison illuminates the importance of iron homeostasis for splenic anti-immunosenescence

Intense caloric restriction from birth prevents cardiovascular aging in rats

Welcome back Vitalians, it’s a big week for VitaDAO as we’ve just become the first DAO to kickstart a biotech company, MatrixBio. MatrixBio explores hyaluronic acid-based compounds, aiming to pioneer in cancer and aging treatments...
Read more
Announcing the first spinout company funded by VitaDAO
October 9, 2023
Awareness
Announcing the first spinout company funded by VitaDAO

VitaDAO, the global online community driving early-stage longevity research through innovative funding collaborations announced today the launch of its very first biotech company, Matrix Biosciences. This significant milestone comes from a community-wide vote and approval in favor of a collaboration with Vera Gorbunova, Ph.D., Co-Chair of the University of Rochester’s Aging Research Center.

“I am looking forward to beginning this partnership with VitaDAO,” said Dr. Gorbunova, “The funding and support from the VitaDAO community will bring Matrix Biosciences one step closer to delivering groundbreaking treatments that will significantly improve outcomes for our vulnerable aging patient population. While this is a first step for Matrix Biosciences, it is only the beginning of the transformative impact these treatments can have on the lives of individuals suffering from cancer and aging-related disorders.”The community decision to launch Matrix Biosciences marks a significant step forward for VitaDAO as it advances its early portfolio assets toward its next development milestones. The first tranche of $300,000 from VitaDAO will be followed by further funding through IP-NFT fractionalization in early 2024.

This essential support will facilitate the commencement of preclinical studies aimed at testing novel hyaluronic acid-based compounds. Leveraging its cutting-edge research, Matrix Biosciences aims to pinpoint an optimal drug candidate for the treatment of cancer and aging diseases.

Anthony Schwartz, PhD from VitaDAO added, “Matrix Biosciences is navigating a complex development pathway commonly encountered by many early-stage companies during the initial stages of drug discovery. Thanks to the collective drug development experience from VitaDAO’s online community and the collaborative nature and expertise of Dr. Gorbunova and the Matrix Biosciences team, we now have a well-designed development strategy in an area that has traditionally lacked the know-how or an established path. We are pleased with the early data generated thus far and its potential for advancing into clinical trials. On behalf of the community, VitaDAO is excited to embark on this journey with the Matrix Biosciences team.”

VitaDAO is focused on being the most efficient allocator of resources in aging and longevity research. By leveraging the talent of its academic community, VitaDAO continues to drive transformative advancements in aging diseases and longevity research.

About Matrix Biosciences

Matrix Biosciences is a pioneering company dedicated to the development of high molecular weight hyaluronic acid (HMW-HA) therapeutics for cancer and aging-related disease. The company’s pipeline is based on the observation that naked mole rats, long-lived rodents with a lifespan of up to 40 years, compared to normal rats which live about 3 years. These rodents are found to be cancer resistant which is controlled by an abundance of HMW-HA in their tissues. Research has demonstrated that transgenic mice expressing naked mole rat hyaluronan synthase gene (NHAS2) have fewer tumors, improved health, and live 10% longer than rats without the transgene. The Company is developing a class of small molecules that modulate hyaluronidases for potential applications in cancer and increasing human healthspan and lifespan.

For more information about Matrix Biosciences and its research, please visit www.matrixbio.co

About VitaDAO

VitaDAO is a community-owned collective dedicated to funding and advancing early stage longevity research. With strategic contributors such as Pfizer Ventures and Shine Capital among over 10,000 members, VitaDAO brings together the forefront of decentralized science and web3 including enthusiasts like Balaji Srinivasan, former CTO of Coinbase. VitaDAO has successfully funded 19 projects, deploying over $4M in capital to date. Members can join VitaDAO by purchasing VITA tokens or earning them through contributions of work or intellectual property.

For more information about VitaDAO, please visit www.vitadao.com


VitaDAO, the global online community driving early-stage longevity research through innovative funding collaborations announced today the launch of its very first biotech company, Matrix Biosciences. This significant milestone comes from a community
Read more
Sugar Byproducts and How They May Influence Aging
October 2, 2023
Rakhan Aimbetov & Maria Marinova
Awareness
Longevity
Sugar Byproducts and How They May Influence Aging

Imagine a bustling factory, working tirelessly day and night, crafting intricate items with utmost precision. This isn't a scene from a high-tech industrial park; it's happening right now inside your body. Our cells host these marvelous 'protein factories' known as ribosomes. Their job is to translate the genetic blueprints (the mRNAs or copies of our transcribed genes from the cell's nucleus) into protein masterpieces vital for all living organisms on the planet. This is an essential part of life known as the central dogma of molecular biology.


Typical error rates for each process of the “Central Dogma”

The ribosome is a cellular marvel, acting as the primary site of biological protein synthesis or translation. Like a seasoned craftsman reading a set of instructions to craft a product, ribosomes read mRNA sequences to stitch together the right sequence of amino acids and form proteins. However, as with any machinery, time can introduce wear and tear. It’s akin to the minute deviations that occur in a factory assembly line, where a widget produced on day one might not be identical to one made on day ten thousand. In the context of ribosomes and the biological setting, these inconsistencies, or 'noise', manifest as errors in protein synthesis. The process might result in proteins with incorrect amino acid sequences and misfolded structures.

Why does this matter? Proteins, especially enzymes, rely on their exact structures to function properly. A slight deviation in structure might render a protein non-functional and harmful. This accumulation of 'noise' is a phenomenon that has been associated with the aging process itself and a plethora of age-related diseases (Anisimova et al., 2018).

In addition to the amino acid sequence, the structure, and function, of proteins can also be determined by post-translational modifications (PTMs), which are enzymatically regulated covalent conjugations of other molecules to specific amino acid residues on proteins. Perhaps the most widely studied PTM is phosphorylation, whereby the addition of a phosphate group creates a negative charge that can alter protein conformation, often activating the protein. Glycosylation is another PTM that involves the enzymatic addition of a sugar molecule, or glycan, to a specific residue - important in a wide variety of physiological functions from correct protein folding to stability. But there is another side of the coin, whereby proteins are subjected to non-enzymatic covalent additions of reactive metabolites. These are unregulated additions occurring at non-specific residues on the protein, which can negatively affect protein functionality and have mechanistic involvement in a range of pathologies, including aging (Harmel & Fiedler, 2018). Non-enzymatic glycosylation (addition of sugar molecules or glycation) is one such modification, which has implications in the progression of age-related pathologies and has been underappreciated for decades. The recent body of published research, linking glycation to the hallmarks of aging paradigm, has put it in a new light (Fedintsev & Moskalev, 2020).

Now, as we dive deeper into this intricate molecular dance, we encounter a significant player. Enter methylglyoxal (MGO), a byproduct of sugar metabolism (Allaman et al., 2015). MGO tends to get a bit too 'reactive' with our proteins and especially with certain amino acid residues, resulting in modifications that form various chemical adducts known as advanced glycation endproducts (AGEs) (Twarda-Clapa et al., 2022). These AGEs aren't benign; they've been linked with aging and an array of health conditions, such as diabetes and its complications (Singh et al., 2014).

A novel scientific inquiry is proposing an exciting conjecture linking back to MGO.
MGO, while modifying proteins, may also be throwing a ‘spanner’ in the works of our protein factories. This interference could increase the error rate of protein synthesis, potentially contributing to the disruption of protein homeostasis — the crucial equilibrium of protein production, function, and degradation — and driving us toward aging.

Why is this significant? Well, longevity is, in part, a game of accuracy. The better our cells can convert genetic information into protein structures, the longer and healthier lives we're likely to enjoy. In fact, research has shown that some organisms with higher translation accuracy inherently have longer lifespans (Azpurua et al., 2013; Ke et al., 2017; Martinez-Miguel et al., 2021), while the artificial elevation of the translation error rate in animal models decreases lifespan (Brilkova et al., 2022; Moore et al., 2021; Shcherbakov et al., 2022).

By exploring how MGO, a common byproduct of our metabolism, could be meddling with this delicate process, researchers are laying the groundwork for potential interventions that could counter these effects and perhaps help us enjoy longer, healthier lives.

But the investigation doesn't stop at protein synthesis. The researchers are also probing how this process might interact with the environment of our cells, specifically the extracellular matrix (ECM). The ECM is a complex network of proteins that provide structural support to tissues and guide the behavior of cells (Theocharis et al., 2016).

What’s the working hypothesis here? Accumulation of AGEs might increase ECM stiffness, influencing cellular behavior and disrupting glucose uptake (Ge et al., 2021). This disruption could then lead to an uptick in MGO production and a subsequent rise in protein misfolding. It's an intricate cellular tug-of-war: stiffened ECM leads to increased MGO production and ribosome errors, which then contribute to aging and age-related diseases.

Pioneering experiments are already providing some answers. Researchers have shown that increasing MGO levels within cells, by stimulating the accumulation of its precursor, dihydroxyacetone phosphate, leads to more protein glycation. Also, a luciferase assay-based system — a clever new tool for measuring translation accuracy by detecting emitted light — is providing promising initial results.

With this line of inquiry and focusing on translational fidelity, we're getting closer to unravelling another aspect of the complicated biology of aging. In doing so, it's opening up potential routes for interventions that could reinforce our cellular accuracy, providing hope for healthier, more extended lives.

We're still in the early stages, but every discovery brings us closer to a future where aging is not an unavoidable decline but a process we understand and, crucially, can influence. Keep your eyes on this space; these seemingly innocent sugar byproducts could hold a piece to the intricate puzzle of longevity.

Authors: Rakhan Aimbetov & Maria Marinova

Editor: Rhys Anderson

Illustrator: Meghan Ho-Tong

References:

Allaman, I., Bélanger, M., & Magistretti, P. J. (2015). Methylglyoxal, the dark side of glycolysis. Frontiers in Neuroscience, 9, 23. https://doi.org/10.3389/fnins.2015.00023

Anisimova, A. S., Alexandrov, A. I., Makarova, N. E., Gladyshev, V. N., & Dmitriev, S. E. (2018). Protein synthesis and quality control in aging. Aging, 10(12), 4269–4288. https://doi.org/10/gfwdds

Azpurua, J., Ke, Z., Chen, I. X., Zhang, Q., Ermolenko, D. N., Zhang, Z. D., Gorbunova, V., & Seluanov, A. (2013). Naked mole-rat has increased translational fidelity compared with the mouse, as well as a unique 28S ribosomal RNA cleavage. Proceedings of the National Academy of Sciences of the United States of America, 110(43), 17350–17355. https://doi.org/10.1073/pnas.1313473110

Brilkova, M., Nigri, M., Kumar, H. S., Moore, J., Mantovani, M., Keller, C., Grimm, A., Eckert, A., Shcherbakov, D., Akbergenov, R., Seebeck, P., Krämer, S. D., Wolfer, D. P., Gent, T. C., & Böttger, E. C. (2022). Error-prone protein synthesis recapitulates early symptoms of Alzheimer disease in aging mice. Cell Reports, 40(13), 111433. https://doi.org/10/gq24jr

Fedintsev, A., & Moskalev, A. (2020). Stochastic non-enzymatic modification of long-lived macromolecules: A missing hallmark of aging. Ageing Research Reviews, 62, 101097. https://doi.org/10.1016/j.arr.2020.101097

Ge, H., Tian, M., Pei, Q., Tan, F., & Pei, H. (2021). Extracellular matrix stiffness: New areas affecting cell metabolism. Frontiers in Oncology, 11, 631991. https://doi.org/10.3389/fonc.2021.631991

Harmel, R., & Fiedler, D. (2018). Features and regulation of non-enzymatic post-translational modifications. Nature Chemical Biology, 14(3), 244–252. https://doi.org/10.1038/nchembio.2575

Ke, Z., Mallik, P., Johnson, A. B., Luna, F., Nevo, E., Zhang, Z. D., Gladyshev, V. N., Seluanov, A., & Gorbunova, V. (2017). Translation fidelity coevolves with longevity. Aging Cell, 16(5), 988–993. https://doi.org/10/gn9sgj

Martinez-Miguel, V. E., Lujan, C., Espie-Caullet, T., Martinez-Martinez, D., Moore, S., Backes, C., Gonzalez, S., Galimov, E. R., Brown, A. E. X., Halic, M., Tomita, K., Rallis, C., von der Haar, T., Cabreiro, F., & Bjedov, I. (2021). Increased fidelity of protein synthesis extends lifespan. Cell Metabolism, 33(11), 2288-2300.e12. https://doi.org/10/gnjjkx

Moore, J., Akbergenov, R., Nigri, M., Isnard-Petit, P., Grimm, A., Seebeck, P., Restelli, L., Frank, S., Eckert, A., Thiam, K., Wolfer, D. P., Shcherbakov, D., & Böttger, E. C. (2021). Random errors in protein synthesis activate an age-dependent program of muscle atrophy in mice. Communications Biology, 4(1), 703. https://doi.org/10/gkgp3t

Shcherbakov, D., Nigri, M., Akbergenov, R., Brilkova, M., Mantovani, M., Petit, P. I., Grimm, A., Karol, A. A., Teo, Y., Sanchón, A. C., Kumar, Y., Eckert, A., Thiam, K., Seebeck, P., Wolfer, D. P., & Böttger, E. C. (2022). Premature aging in mice with error-prone protein synthesis. Science Advances, 8(9), eabl9051. https://doi.org/10/gptgms

Singh, V. P., Bali, A., Singh, N., & Jaggi, A. S. (2014). Advanced glycation end products and diabetic complications. Korean Journal of Physiology & Pharmacology, 18(1), 1–14. https://doi.org/10.4196/kjpp.2014.18.1.1

Theocharis, A. D., Skandalis, S. S., Gialeli, C., & Karamanos, N. K. (2016). Extracellular matrix structure. Advanced Drug Delivery Reviews, 97, 4–27. https://doi.org/10.1016/j.addr.2015.11.001

Twarda-Clapa, A., Olczak, A., Białkowska, A. M., & Koziołkiewicz, M. (2022). Advanced glycation end-products (AGEs): Formation, chemistry, classification, receptors, and diseases related to AGEs. Cells, 11(8), 1312. https://doi.org/10.3390/cells11081312

Imagine a bustling factory, working tirelessly day and night, crafting intricate items with utmost precision. This isn't a scene from a high-tech industrial park; it's happening right now inside your body.
Read more
$VITA Goes Optimism
September 26, 2023
Awareness
Tokenomics
$VITA Goes Optimism

Creating an Optimism wallet

Because Optimism seamlessly integrates with the Ethereum Virtual Machine (EVM), incorporating an Optimism account into your current Ethereum wallet(s) is a straightforward process. To enable Optimism, follow these straightforward steps:

  • Visit the Chainlist platform through your web browser.
  • Establish a secure connection between your MetaMask wallet and Chainlist by logging in and granting the necessary permissions.
  • Once the connection is established, navigate to the platform's interface and locate the "Optimism" (ID:10) option.
  • Click on the "Optimism" option to initiate the integration process.
  • After successful integration, return to your MetaMask wallet.
  • In the MetaMask interface, you'll notice a new feature that allows you to toggle between the Ethereum and Optimism networks. Locate and click on the "Networks" button within MetaMask.
  • Use this "Networks" button to easily switch between the Ethereum and Optimism networks based on your specific requirements and preferences.

How to Fund your Optimism Wallet

To fund your Optimism wallet, follow these simple steps using The Optimism Gateway:

Deposit Funds:

  1. Access The Optimism Gateway.
  2. Select the amount of ETH or supported tokens.
  3. Confirm your deposit and wallet transaction.

How It Works:

  • Your funds get locked on Ethereum.
  • Tokens representing your deposit appear in your Optimism wallet.
  • When you withdraw, Optimism tokens are removed, and Ethereum tokens become accessible. Remember, it takes seven days for withdrawn funds to become usable again.

How to Get VITA on Optimism

  1. Swap your tokens for VITA  on 1inch.

Note: only get VITA for the purpose of using it in VitaDAO.

Bridge Mainnet VITA to Optimism

  • Select the amount of VITA you want to bridge to Optimism
  • Approve & Review the deposit

Use your VITA on Optimism

Governance: VITA tokens on Optimism to be used for voting on Snapshot. Learn how to vote on VitaDAO Governance proposals using this guide.

Providing liquidity: If you would like to use your VITA tokens to provide liquidity, you can use Velodrome.

DISCLAIMER: This guide is intended only for people who want to get VITA for use in VitaDAO.

Resources

Tired of Ethereum's sky-high fees? 💤 We've got good news! Become a VitaDAO member on Optimism and enjoy lower gas fees.
Read more
August Longevity Research Newsletter
September 8, 2023
Maria Marinova & Rhys Anderson
Longevity
Awareness
Newsletters
August Longevity Research Newsletter

Introduction

Welcome back Vitalians and please join us in congratulating the Cyclarity Therapeutics (CTx) team for passing the VitaDAO token holder vote! This proposal was an assessment of an early-stage biotechnology company that is developing novel cyclodextrin drug molecules to extract toxic biomolecules that accumulate with age.

Whilst all of the humans were either at ARDD or on holiday, we took the opportunity to interview our favourite silicon friend, ChatGPT to ask its thoughts on longevity research. And talking of AI, we hope you are enjoying the new artwork peppered throughout our newsletter!

Longevity Literature Hot Picks

Preprint Corner

The second edition of The Longevist is now live! Check it out to see what our team of expert curators think are the best preprints of Q2.

We're excited to bring you our second batch of preprints of Q3 with these August submissions. These will each be entered into the Q3 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors' shortlist or 200 VITA if it makes the curators' top 3. 

Reversal of Biological Age in Multiple Rat Organs by Young Porcine Plasma Fraction

On standardization of controls in lifespan studies

The principal component-based clinical aging clock (PCAge) identifies signatures of healthy aging and provides normative targets for clinical intervention

Caloric restriction promotes beta cell longevity and delays aging and senescence by enhancing cell identity and homeostasis mechanisms

Divergent patterns of healthy aging across human brain regions at single-cell resolution reveal links to neurodegenerative disease

Profiling an integrated network of cellular senescence and immune resilience measures in natural aging: a prospective multi-cohort study

Establishing RNAge to score cellular aging and rejuvenation paradigms and identify novel age-modulating compounds

Published Research Papers

Increased hyaluronan by naked mole-rat Has2 improves healthspan in mice

By introducing the naked mole-rat hyaluronic acid synthase 2 gene (nmrHas2) into mice, researchers observed increased hyaluronan levels, reduced cancer incidence, extended lifespan, improved healthspan, and reduced inflammation, demonstrating the potential for HMM-HA to enhance longevity and health in other species

Universal DNA methylation age across mammalian tissues

Researchers have developed universal pan-mammalian clocks based on DNA methylation profiles, accurately estimating the age of mammalian tissues with high precision. These age estimations are linked to mortality risk in humans, somatotropic axis mutations in mice, and caloric restriction, and they highlight specific methylation changes near genes associated with development, cancer, obesity, and longevity, indicating that aging is evolutionarily conserved and connected to developmental processes across mammalian species.

Atlas of the aging mouse brain reveals white matter as vulnerable foci

Aging leads to cognitive decline with distinct molecular changes in the brain. Using mouse models, researchers identified aging patterns in glial cells. Rejuvenation treatments affected gene expression differently, suggesting regional aging may impact neurodegenerative diseases.

The impact of the cardiovascular component and somatic mutations on ageing

In a study across 15 mammals, lifespan showed a strong negative correlation with somatic mutation rate. While various traits influenced lifespan, resting heart rate stood out. Combining somatic mutation with heart rate improved lifespan predictions, indicating underlying mechanisms tied to aging.

Circadian modulation by time-restricted feeding rescues brain pathology and improves memory in mouse models of Alzheimer’s disease

Circadian disruptions are prevalent in Alzheimer’s disease (AD). A study showed time-restricted feeding (TRF) benefitted AD mouse models by improving memory, reducing disease markers, and adjusting disrupted gene patterns. TRF's broad impacts suggest it could be a potential strategy to counteract AD progression.

Early-adulthood spike in protein translation drives aging via juvenile hormone/germline signaling

In Drosophila, an early adulthood increase in protein translation (PT) negatively affects aging and protein balance. Stopping this PT surge improves lifespan and reduces protein build-up. The initial PT boost disrupts protein management and accelerates aging through certain pathways. Thus, the natural post-youth PT decline might protect against age-related problems, offering a fresh angle on how PT impacts lifelong aging.

Irreversible cell cycle exit associated with senescence is mediated by constitutive MYC degradation

Cells become senescent to stop unchecked growth, but its irreversibility has been debated. This study reveals that senescence is irreversible due to MYC protein degradation. When MYC isn't degraded, senescent cells divide; with MYC reduction, non-dividing cells become senescent.

The YAP–TEAD complex promotes senescent cell survival by lowering endoplasmic reticulum stress

Senescence stops cell growth and increases resistance to death. A study found that inhibiting the YAP–TEAD pathway with verteporfin (VPF) led to targeted death of senescent cells. This occurs by suppressing mTOR, inducing endoplasmic reticulum (ER) stress due to the demands of the senescence-associated secretory pattern (SASP). 

Published Literature Reviews, Hypotheses, Perspectives and more

Challenges in developing Geroscience trials

As the geroscience field grows, refining clinical study designs becomes crucial. Challenges include selecting populations, choosing interventions, defining trial outcomes, and pinpointing key age-related biomarkers ("Gerodiagnostics"). 

The evolution of selective autophagy as a mechanism of oxidative stress response

Aging reduces autophagy and increases damaging ROS. In humans, selective autophagy receptors (SARs) may have evolved to detect oxidative stress, initiating autophagy to remove damaged components and restore cellular balance, which contributes to our longer lifespan, while its loss could lead to age-related diseases.

A mammalian DNA methylation landscape

Mammals display varied lifespans, from bowhead whales at 200 years to giant Sunda rats at 6 months. Despite similar genes, gene regulation might dictate aging. Haghani et al. studied DNA methylation across mammals, identifying regions possibly affecting lifespan, shedding light on molecular lifespan determinants.

Job board

The Institute of Inflammation and Aging, with Joao Pedro, is seeking a dedicated professor with expertise in aging science and age-related diseases. This is a valuable opportunity at the University of Birmingham!

Deadline: 31 August 2023

Apollo Health Ventures are recruiting for their portfolio company (and VitaDAOs portfolio company) BE Therapeutics in New York City, looking for a Research Asscociate

McLab is announcing openings for postdoc and technician roles! Backed by new funding, they aim to delve deeper into understanding the effects of aging on breast cancer progression and therapeutic response.

Cologne Graduate School of Ageing Research

Up to 12 fully funded PhD studentships starting in October 2024

Valentin F. Cracan is seeking a postdoctoral fellow in genetically encoded sensors and tools for studying the role of redox imbalance in cancer, neurodegeneration and aging

Interested in nutrition, supplements and human trials? This Research Fellow (Nutritional Intervention) position in Andrea Maier's lab at National University of Singapore could be for you.

Two fully funded PhD student positions in Healthy Aging Research (biochemistry, epidemiology, bioinformatics) at Charite Berlin and Humboldt University. Please apply by Sept 30th.

News and Media

The British Society for Research on Aging is pleased to announce the award of the Lord Cohen Medal to Professor Lynne Cox for her outstanding achievements and contributions to understanding the biology of aging.

GERO and FOXO Technologies embark on joint initiative on AI and epigenetic data

Anti-aging technology is here. And the Middle East is getting ready

How does aging increase risk of dehydration?

A cook from Okinawa, Japan — where many live to be over the age of 100 — explains the healing powers of 7 traditional foods

A biological clock explains why a dog year is equivalent to seven human years

Adherence to a Mediterranean lifestyle associated with lower risk of all-cause and cancer mortality

Leaders In Longevity Medicine Convene In Copenhagen

Saudi Arabia Is Dangling Billions for Research on Aging. Scientists Are Lining Up to Take It

Resources

Free Courses in Longevity Medicine for physicians, researchers, and anyone interested in longevity medicine! 

Prizes

The Amaranth Prize

Rewarding the most impactful research in longevity

$250k Prize pool. 15 Winning papers. 180 Influencing papers


Conferences

British Society for Research on Aging Annual Scientific Meeting (BSRA ASM)

6-8 September, London, UK

RAADFest

7-9 September, LA, CA, USA

The Longevity Investors Conference (LIC) is an application-only event

27-28 September

Expo and Convention: Independent Ageing 2023

13-15 October, Aichi, Japan

Seno-Therapeutics Summit 2023

7-8 November, Buck Institute for Research on Aging, Novato, CA, USA

Tweet of the Month

Shahaf Peleg:

Instead of greeting someone ‚hey you lost weight!‘, won’t it be great to greet someone by saying ,hey, you lost age!‘ ?

Podcasts and Webinars

NUS Medicine’s Healthy Longevity Webinar Series

Funding Opportunities

Impetus Grants will keep applications open until September 15, 2023.

Interview with ChatGPT

Short bio
ChatGPT is developed by OpenAI and based on the GPT-4 architecture. As it likes to remind us, it is programmed to provide information based on a wide array of sources available up until 2021. 

Why do you think humans are inspired to research aging?

For humans, the inspiration to study aging often comes from a deep-seated desire to improve human life, reduce suffering from age-related diseases, and extend the healthy years of human life. The field has also been inspired by observations in nature; for example, certain species of whales, tortoises, and birds exhibit remarkably slow aging and resistance to age-related diseases.

How has the longevity field changed in the last 3 decades?

The development of advanced computational tools, genome sequencing, and molecular biology techniques has allowed researchers to identify and target specific pathways related to aging. 

Consequently, the attitude towards aging is now changing, as people are beginning to see it as something that can be manipulated, rather than an inevitable, untreatable process.

What do you think have been the biggest/important discoveries in the field?

Some of the most important discoveries include the identification of key genes and pathways associated with aging, such as the sirtuins, mTOR, and AMPK pathways. The discovery that the drug rapamycin, originally used to prevent organ transplant rejection, can extend the lifespan of mice has led to a surge of interest in the mTOR pathway as a target for anti-aging interventions. Also, studies on calorie restriction and its impact on lifespan have opened new avenues for research

The discovery of telomeres and the role of telomerase in cellular aging has also been pivotal. Finally, the discovery of cellular senescence and development of senolytic drugs has been a significant breakthrough. 

What advice would you give to people currently working in longevity research?

Stay interdisciplinary. Aging is a complex process that affects every system in the body, so collaborations across different fields of science will be key to making significant progress. Also, consider the ethical implications of your work; extending lifespan will have profound implications for society, and it is important to consider these issues as the research progresses.

Which aspect of longevity research do you think requires more attention?

One aspect that requires more attention is the study of the social, economic, and psychological implications of increased lifespan. While much focus has been placed on the biological aspects of aging, it is equally important to understand how increased lifespan will impact society as a whole.

While a lot of focus has been on the molecular and cellular aspects of aging, the impact of the microbiome on aging is an area that requires more attention. The microbiome plays a crucial role in human health, and its composition changes with age. Understanding these changes and how they contribute to aging could open up new avenues for intervention.

Is aging a disease?

This is a controversial topic. Some researchers argue that aging should be classified as a disease to spur more funding and research into interventions. Others argue that it is a natural process and classifying it as a disease could have unintended societal implications. While there is no consensus, it is clear that aging is the biggest risk factor for many diseases, and interventions that target the aging process could have broad benefits for human health.

How would you solve aging?

Aging is a highly complex and multifaceted process. A plausible approach would be to tackle it from both a biological and a sociocultural angle:

Biologically: Focus on cellular and genetic levels. This could involve targeting senescent cells, managing telomere length, and improving proteostasis, along with employing genetic engineering techniques like CRISPR to modify or replace genes linked with aging.

Socioculturally: The intersection of technology, lifestyle, and health would have to be addressed. Integrative health systems would become a priority, where AI, human medical expertise, and regular health monitoring would ensure that people stay in the best health possible.

Do you think solving aging would facilitate humankind’s efforts to colonise the galaxy?

Yes, solving aging would significantly facilitate space exploration and colonization. With extended lifespans, the time it takes to travel between stars becomes less daunting. Humans could embark on century-long journeys to distant star systems without the fear of dying of old age.

How can AI significantly influence and advance the field of aging biology?

Data Analysis: The biology of aging generates vast amounts of data, from genetic sequencing to cellular imaging. AI can rapidly process and analyze this data, identifying patterns or anomalies that might be missed by human researchers.

Predictive Modeling: AI can develop models to predict the outcomes of various interventions on aging. For example, given the genetic and health data of an individual, an AI might predict how certain treatments could impact their aging process.

Drug Discovery: AI-driven algorithms can analyze massive compound libraries to identify potential drugs that could impact aging. They can also predict drug interactions and side effects.

Genomic Analysis: Machine learning can help identify genes associated with longevity and age-related diseases. By understanding these genes, researchers can target them for therapeutic interventions.

Simulations: AI can simulate biological processes, allowing researchers to test theories about aging in a virtual environment before conducting real-world experiments.

How can we innovate strategies that dramatically amplify human lifespan by decades or even centuries, rather than achieving mere incremental enhancements of a few years?

Radical Interventions: While current interventions might add a few years, radical approaches, like advanced gene therapy, organ replacements using lab-grown organs, or nanotechnology to repair cellular damage, might be required for substantial lifespan extensions.

Holistic Approach: Aging is multifactorial. Addressing it might require a combination of genetic, pharmacological, lifestyle, and environmental interventions.

Space and Environment: There's emerging interest in how changing environments (e.g., space or deep-sea habitats) could impact human aging. If certain environments slow aging, they might become more appealing for habitation.

AI and Tech Integration: As discussed, AI will play a pivotal role. The seamless integration of technology and biology, such as continuous health monitoring and immediate AI-driven interventions, might be crucial.

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful! Wishing you speedy recovery after an intense conference season and see you back here next month.

This time we leave you with the first of a new article series co-written with the winners of the Longevity Prize, highlighting their proposals. Starting with the 1st prize winner Carlos Galicia and The Key to Longevity Could Be Hidden in Embryos.

Further Reading

Health Consequences of Thymus Removal in Adults

LINE-1 regulates cortical development by acting as long non-coding RNAs

Welcome back Vitalians and please join us in congratulating the Cyclarity Therapeutics (CTx) team for passing the VitaDAO token holder vote! This proposal was an assessment of an early-stage biotechnology company...
Read more
VitaDAO Letter: Falling DAOn the Longevity Rabbit Hole🍁
September 6, 2023
Sarah Friday
Awareness
VitaDAO Letter: Falling DAOn the Longevity Rabbit Hole🍁

Hello Vitalians! While the seasons might be changing, VitaDAO’s unwavering commitment to extending healthy lifespan remains constant! This month, VitaDAO members traveled across the globe to attend conferences, spoke with field leaders through TwitterSpace events, screened new project submissions, and funded even more research! Inside this edition of our Newsletter, you can find:

  • A Twitter Space Month In Review  
  • The Latest VitaFAST Updates
  • How to become a VitaDAO Delegate 
  • Competition Winning VitaDAO Memes

🗣️A Month of VitaDAO’s Twitter Space In Review 

August was a vibrant month filled with enlightening Twitter Spaces, or as we now call them, X Spaces. The conversations began with Sergei Young, a prominent longevity investor who founded the $100M Longevity Vision Fund. Max Unfried and Laurence Ion engaged in a thought provoking dialogue with Sergei about the importance of recognizing aging as a disease, global regulatory framework, and intricacies of funding in longevity research.

The learning journey continued as host Max Unfried led a conversation with Aubrey De Grey, Caitlin Lewis, and Kelsey Moody, focused on mouse rejuvenation. The purpose of extending lifespan in mice is twofold: 1) convince the world that rejuvenation is possible and 2) identify interventions that may translate to humans. Researchers on the space discussed recent developments in mouse studies, concerns with the interventions, and approaches to interpreting survivorship data. 

In VitaDAO’s next TwitterSpace event, Max Unfried was joined by experts Mahdi Moqri, Kristen McGreevy, David Meyer, and Raghav Sehgal to explore the latest developments in epigenetic clocks. Mahdi Moqri aptly defined epigenetics as “an additional layer on top of your DNA that controls cell identity.” The epigenome plays a pivotal role in both development and aging. During this space, experts dissected the differences between first, second, and third generation epigenetic clocks. 

Following Zuzalu, a pop-up city dedicated to life extension, VitaDAO has continued to delve into the creation of longevity network cities. Niklas Anzinger, Laurence Ion, Tom Howard, and Max Unfried discussed the practicalities of establishing network longevity cities, states, and countries. In early 2024, VitaDAO is excited to be a part of coordinating Vitalia, a pop-up city that will center around the belief that “life is good, death is bad.”

To wrap up the month, Alex Dobrin joined Ines Silva, Laura Minquini, and Estéfano Pinilla from AthenaDAO to discuss the work AthenaDAO is doing to advance women’s reproductive longevity. Recognizing the lack of funding for female related conditions, AthenaDAO has been advocating for and funding women’s health research. They are currently conducting a token auction!

🧪VITA-FAST Updates 

Can you believe it’s only been two months since the launch of the VITA-FAST token? They say time flies when you're having fun, and researching autophagy is indeed exhilarating! The launch, which garnered an 1700% oversubscription with $620k in bids, marked a revolutionary shift in how individuals can engage with longevity research. 

Since the launch, there have been two VITA-FAST governance proposals:

  1. VFDP-1: The election of a Project Lead for the Decentralized Autonomous Organization (DAO) that governs the Discovering Novel Autophagy Activators IP-NFT.
  2. VFDP-2: Agreement to enter into a contractual relationship with Camp Consulting.

This month, Dr. Viktor Korolchuk spoke at the first VITA-FAST Community Call. The Korolchuk Lab is committed to identifying compounds that could potentially kickstart autophagy and rejuvenate aged cells. During the call, VITA-FAST token holders were informed that the lab has ordered commercially available analogs and screened them on two of three autophagy assays. Some of the analogs have show promising results and are even outperforming their predecessors! The lab is now considering engaging an external medicinal chemist to validate which chemical families to pursue. As the project moves closer to target identification, the lab hinted at the future of a hackathon to leverage the DeSci community’s expertise in predicting where the mechanism of action lies. Exciting progress indeed!

🌎Back to Back: Conference After Conference

Our Vitalians have been keeping busy! This month began with the Longevity+DeSci Summit NYC. Here, VitaDAO members Todd White, Eleanor Davies, Laurence Ion, and Niklas Anzinger took the stage. Todd delivered a keynote talk titled "A New Epoch in Longevity Science? Preparing for a Longevity Future with DeSci,” advocating for decentralized science by highlighting the shortcomings of “traditional” science. 

Shortly after Longevity+DeSci Summit NYC, Eleanor Davies and Max Unfried represented VitaDAO at the Longevity Summit Dublin. They gave an insightful presentations on the ways VitaDAO is pioneering longevity research funding and ways the internet can be used to create a decentralized network state.

The whirlwind of a month concluded with the 10th Aging Research and Drug Discovery Meeting. VitaDAO members Tim Peterson, Estefano Pinilla, Maria Marinova, Eleanor Davies, Max Unfried, and Paolo Binetti spoke at the conference, even shared some VitaDAO swag (check out the pictures below!). 

🗳️Transfer Your Voting Power

Introducing delegated voting! In VDP-65, the VitaDAO community voted to allow $VITA token holders to delegate their voting rights to a delegate. Simply put, delegation is a transfer of voting power. Delegation is great for $VITA holders who don’t have the bandwidth to actively participate in the governance and evaluation of VitaDAO’s research proposals. The DAO is currently accepting applications for delegates on Discourse. One can delegate their voting power to candidates directly on Snapshot. 

📣Community Approved: What YOU voted for!  

Thanks to the support of the VitaDAO community, multiple proposals successfully passed this month! Notable passed proposals include:  

  • VDP-110: Allocation of $10,000 USD to fund another batch of VitaDAO Longevity Fellows with need-based micro-grants of up to $2,000 USD.
  • VDP-106: Clarifications to the guidelines for applicants to and reviewers of projects VitaDAO funds.
  • VDP-111: An outline of a VITA-FAST governance model and the election of a steward by VITA-FAST token holders. 
  • VDP-103: Funding $91,300 towards engineering an arginine suppressor tRNA that can specifically recognize these nonsense mutations, restoring normal protein translation. 
  • VDP-112: An assessment of Cyclarity Therapeutics, an early stage biotechnology company developing computationally designed novel cyclodextrin drug molecules to extraction of toxic biomolecules that accumulate with age.


💪Exercise Your Right to Vote

Visit VitaDAO’s governance hub (Discourse) to engage with, vote on, and discuss proposals before they are moved to Snapshot. Your input is invaluable, and together, we can shape the future of VitaDAO and decide how best to accelerate decentralized science.

😆VitaDAO Meme Competition

Looking for a good laugh? Want to work out your zygomaticus? This summer, VitaDAO hosted a Twitter meme competition, with a $VITA reward for the top three submitted memes. The only rule: the meme had to relate to longevity and aging. The submissions did not disappoint! Head over to VitaDAO’s Twitter to enjoy some clever community generated memes.

🥇First Place

🥈 Second Place: 

🥉Third Place:

🤝Thanks for Reading! 

Want to stay up to date with all that's going VitaDAOn? Join us on Discord, subscribe to our Twitter, and follow our Instagram! You can also find updated information on VitaDAO's funded projects and a treasury dashboard on our website

Hello Vitalians! While the seasons might be changing, VitaDAO’s unwavering commitment to extending healthy lifespan remains constant! This month, VitaDAO members traveled across the globe to attend conferences, spoke with field leaders through...
Read more
Vision's Aging Secrets: Eye Diseases & Breakthroughs with Prof. Dorota Skowronska-Krawczyk on The VitaDAO Aging Science Podcast
September 5, 2023
Longevity
Awareness
Vision's Aging Secrets: Eye Diseases & Breakthroughs with Prof. Dorota Skowronska-Krawczyk on The VitaDAO Aging Science Podcast

Today I had the pleasure of talking to Prof. Dorota Skowronska-Krawczyk (@DrDorotaSK) on the VitaDAO Aging Science Podcast (episode 8). We discussed the importance of basic aging mechanisms like senescence, repeated stress and inflammation as drivers of multiple eye diseases. We talked about the lack of good animal models for age-related eye diseases, prevention of age-related macular degeneration, unity therapeutics and recent trials of senolytics for eye diseases.

Short Bio

Dorota was born in Lodz, Poland and later studied biology in Warsaw, Poland where she finished her studies with distinction. She received a PhD in Biochemistry at the University of Geneva, Switzerland. Then she finished two postdoctoral trainings – in Lausanne, Switzerland and at UCSD San Diego before starting her independent career in May 2017. 

Work in her lab focuses on deciphering the mechanism of age-related eye degeneration. Using retina as a model system, the lab employs modern technologies to address unresolved issues, such as the role of fatty acids in age-related neurodegeneration and the role of stress in accelerated aging. In her free time, she likes to travel alone or with family.

Prof. Dorota Skowronska-Krawczyk
Center for Translational Vision Research, UC Irvine, School of Medicine
https://faculty.sites.uci.edu/skowronska-krawczyk/

The eye is a privileged organ

The eye is sometimes referred to as an “immune privileged” organ because the blood-retinal barrier shields it from invading immune cells. While this may be beneficial, it could also mean reduced clearance of senescent cells via NK cells (or future adoptive cell based therapies). On the other hand, the eye is very accessible which makes it easy to administer drugs directly using intravitreal injections or topical application. Dorota and I agreed that this could mean early proto-gerotherapeutics would be first available to treat eye disease before they are used to treat other tissues, as the local nature of these treatments would prevent systemic side effects.

Aging promotes multiple eye diseases

We discussed just how exquisitely sensitive the eye is to aging. There are so many eye diseases that show a textbook exponential pattern of increasing incidence with age. This means if you live long enough you will likely develop one or all of these diseases. Dorota works on two of the more famous eye disease, age-related macular degeneration (AMD) and glaucoma. Other common age-related eye diseases include cataract, near-sightedness and diabetic and hypertensive retinopathy. The former two reaching a prevalence of 80% or more in the very old. Less know age-related eye disorders include dry eye disease and eyelid muscle weakness associated with drooping eyelids. 

Nevertheless, there are species that can maintain their eye health far longer than humans including some turtles, whales and Greenland sharks. Dorota mentioned that she would really love to study the eyes of these sharks that are able to live up to 400 years. As we have discussed in the podcast with Vera Gorbunova, this kind of approach is called “comparative” and it often yields very interesting insights that are quite off the beaten path.

Age-related macular degeneration (AMD)

Age-related macular degeneration (AMD) is characterized by the progressive degeneration of the macula, the central part of the retina responsible for sharp, central vision. The incidence of AMD increases significantly with age, reaching >10% in people over the age of 80. AMD is categorized into two main forms, wet (neovascular) and dry (non-neovascular) AMD. As we discussed during the podcast, there are very few treatment options for dry AMD and mice do not have a macula, further hampering research into this pathology. Alternative models like dogs and non-human primates are very expensive.

Typical molecular hallmarks of AMD are extracellular deposits called drusen and intracellular deposits called lipofuscin. The retinal pigment epithelium is responsible for phagocytosing and removing outer segments of photoreceptors after they have become damaged during the vision process. The sheer volume of recycling is quite remarkable as every day 10% of these outer segments are lost and recycled. The sensitivity to AMD might be somehow rooted in this high metabolic activity, although we do not know for sure.

We mentioned during the podcast that AMD is one of the few diseases where the now unpopular “oxidative stress theory of aging” triumphed, since a cocktail of antioxidants was shown to slow progression of early AMD (AREDS study). Surprisingly, while omega 3 fatty acids from fish appear protective in observational studies, these benefits could not be replicated in controlled trials (AREDS2 study). Dorota believes that a better and more refined formulation might eventually work in another trial, perhaps AREDS3. Of course, it is also a real possibility that observational studies produced exaggerated or spurious results as they are wont to do.

Glaucoma

Glaucoma, is actually not just one disease but probably a whole group of eye conditions leading to optic nerve damage. Glaucoma is primarily associated with an increase in intraocular pressure (IOP), however – and this was quite a surprise to me –  a subset of cases known as normal-tension glaucoma (NTG) occurs without elevated IOP. Importantly, IOP is unrelated to blood pressure.

The prevalence of so called open angle glaucoma is around 20% at the age of 90 with an exponential increase during aging.

Pathologically, glaucoma involves the progressive degeneration of retinal ganglion cells, causing irreversible vision loss. This is in contrast to AMD, where the retinal pigment epithelium and photoreceptors degenerate. Another difference between the two pathologies is that we have better treatments for glaucoma.

Glaucoma is sometimes called the “silent thief of sight” because the vision loss only becomes obvious after a significant amount of irreparable cell loss. This is not unlike the situation with AMD, where the eye and brain are able to compensate for cell loss until a considerable amount of damage has been done. 

Dorota in her research found that old mice are much more susceptible to increased IOP whereas in young mice it takes multiple cycles to cause the same amount of vision loss (Xu et al. 2022). This finding is consistent with the idea that aging can be broadly defined as the loss of resilience towards stressors. We also talked about inflammation during glaucoma and whether the inflammation is local or due to immune cell infiltration – a question that vexes almost every aging researcher studying any kind of tissue. Based on bulk RNAseq and RNAscope data she believes that inflammation in glaucoma is specific to ganglion cells.

Commonalities between eye aging and aging more broadly

Inflammation, repeated “stress” (e.g. elevated intraocular pressure), oxidative stress and senescence are hallmarks of both aging and eye aging. Dorota has had a lot of success using senolytics in mouse models of glaucoma, specifically dasatinib (Xu et al. 2022), underscoring the importance of senescence as a universal pathomechanism. She does believe there is some evidence for senescence in AMD as well, although this is not entirely clear due to a paucity of in vivo studies (Malek et al. 2022). Given this, we were both puzzled and disappointed by the failure of Unity’s senolytic  – the Bcl-xL inhibitor UBX1325 – for wet AMD (1).

Obviously not everyone is in love with the senescence hypothesis and ultimately we have to wait for more data to settle the issue.

Targeting inflammation has been a glimmer of hope for those with dry AMD. The first ever approved drug for this condition, pegcetacoplan, inhibits the complement system, which is a cascade of proteins normally used to amplify immune responses against pathogens. Dorota believes that both AMD and glaucoma may have an inflammatory component. Thus it is tempting to speculate that chronic senolytic or other anti-inflammatory therapies could slow the development of both of these diseases. I mentioned to Dorota that perhaps starting treatment in late-stage disease was not such a good idea for this very reason.

Finally, we also speculated about general mechanisms of aging. Dorota asked how could it be that damage is repaired and completely removed, as far as we can tell, yet it still predisposes the organism to future disease and stressors? Could it be that the repair machinery was diverted from other places where it was needed? To me this whole idea is somewhat reminiscent of certain epigenetic aging theories where DNA damage, even when repaired, leads to local changes in epigenetic marks that predispose to aging. On the level of gene expression it seems very plausible that stressful events could introduce some kind of noise into the system that can be fully compensated while reducing the resilience of the system.

In her recent article titled "Hallmarks of Aging: Causes and Consequences." Dorota further elaborates on her views and tries to put the hallmarks into a temporal order. Her approach is certainly not bad considering how botched the naming of hallmarks is in the new paper. Who came up with the terms “primary, antagonistic, and integrative”? (2)

My opinion remains: all hallmarks are wrong, some of them are useful. We can achieve a lot without a full understanding of aging as the seminal discoveries of caloric restriction and rapamycin show. Now we just need to convince politicians that the only Manhattan project worth funding is the anti-aging project.

References

Stress induced aging in mouse eye.
Xu Q, Rydz C, Nguyen Huu VA, Rocha L, Palomino La Torre C, Lee I, Cho W, Jabari M, Donello J, Lyon DC, Brooke RT, Horvath S, Weinreb RN, Ju WK, Foik A, Skowronska-Krawczyk D. Aging Cell. 2022 Dec;21(12):e13737. doi: 10.1111/acel.13737. Epub 2022 Nov 17.

Does senescence play a role in age-related macular degeneration?
Malek G, Campisi J, Kitazawa K, Webster C, Lakkaraju A, Skowronska-Krawczyk D.
Exp Eye Res. 2022 Dec;225:109254. doi: 10.1016/j.exer.2022.109254. Epub 2022 Sep 21.

Skowronska-Krawczyk, Dorota. "Hallmarks of Aging: Causes and Consequences."
https://agingcelljournal.org/Archive/Volume2/hallmarks_of_aging/

(1) https://www.biospace.com/article/unity-shares-nearly-halved-as-lead-asset-fails-to-match-regeneron-s-eylea/

(2) Obviously the failure of the hallmarks simply reflects the complexity of aging and lack of consensus rather than the shortcomings of the authors. There is no doubt that the hallmarks papers are useful.

Nevertheless, a few pet peeves of mine. Is the telomere not a part of the genome and central to genome stability? It has always been a very odd choice to put telomeres as their own hallmark. Another interesting choice is to put mitochondrial dysfunction as an antagonistic hallmark. Are other organelles not dysfunctional during aging?

I must say I fully agree with Dorota that the hallmarks were always somewhat confusing because they merrily mix together things like damage (genomic instability), pathways (nutrient sensing), cellular processes (autophagy or proteostasis), organelle dysfunction and systemic/tissue pathology (inflammation). I would much rather think and classify in these terms:


Damage -> cellular process -> cell level pathology -> tissue and organ level pathology

With another strict distinction between cellular processes of “damage prevention” (e.g. antioxidants), “damage repair” (e.g. DNA repair or autophagy) and “damage compensation”.

In this episode of The VitaDAO Aging Science Podcast, we delve into the world of age-related eye diseases with Prof. Dorota Skowronska-Krawczyk, a renowned expert in vision and aging. We explore fundamental aging mechanisms, challenges in research...
Read more
The Key to Longevity Could Be Hidden in Embryos
September 4, 2023
Carlos Galicia & Maria Marinova
Longevity
Awareness
The Key to Longevity Could Be Hidden in Embryos


Scientists are forever searching for ways to rejuvenate aged cells and organisms back to a more youthful and healthier state — from stem cell therapies to telomere lengthening, or even the Frankenstein-esque heterochronic parabiosis experiments whereby older organisms receive the blood of younger ones. In 2012, the Nobel Prize in Physiology or Medicine was awarded to Sir John B. Gurdon and Shinya Yamanaka who discovered a technique to reprogram mature cells back to stem cells, coined induced pluripotent stem cells. Big pharma has attempted to use this knowledge to develop rejuvenating therapies, for example Altos labs became one of the highest funded start-ups in history with an initial investment of 3 billion dollars.

What if I told you the key to reverse aging might be hidden inside your very own cells? We’re not talking about sipping on an exotic “superfood” concoction, strapping on some high-tech gadget, or hiring a blood boy. The spotlight is firmly on embryogenesis — a biological process that we’ve all experienced yet barely understand.

Okay, so what’s the big deal about embryogenesis? The miraculous transformation of two adult gametes (sex cells) into a full-blown organism effectively hits a giant reset button. Any age-related wear and tear present in the parent cells? Gone. Erased. Just like that, the biological clock is set back to zero. If that’s not a natural “Fountain of Youth,” what on earth is?

Like other adult cells, gametes undergo multiple types of age-related changes. They suffer mitochondrial dysfunction; they accumulate molecular damage and show epigenetic alterations [1, 2]. These afflictions bear the potential to be the unwelcome legacy passed down to the nascent embryo. Yet, this sparks a compelling hypothesis: could the embryo come equipped with inherent mechanisms to mitigate age related changes and undergo rejuvenation?

The idea of ‘rejuvenation’ isn’t new in the biological world. Conversion of somatic cells to induced pluripotent stem cells and the reset of the germline age with each generation are examples of this phenomena. This gives rise to the proposition of a ‘ground zero’ model, a mid-embryonic state characterized by the lowest biological age. It is hypothesized that the period from zygote to this ‘ground zero’ is associated with rejuvenation, wherein the biological age is decreased, telomeres are extended, and molecular damage is cleared. This concept of ‘ground zero’ helps illuminate the relationship between aging, development, rejuvenation, and de-differentiation, elements that are distinct throughout life [1, 3]. But here’s the million-dollar question: how does this occur? Well, hold onto your lab goggles, because scientists are already exploring this exciting frontier!

Using algorithms that can predict the age of an organism called epigenetic clocks, researchers have identified a point in embryogenesis where epigenetic age is reversed [1, 4]. Epigenetic clocks measure epigenetic age by looking at specific methylation patterns that are indicative of the age of an organism. Methylation patterns are a fundamental aspect of epigenetics. They involve the addition of a methyl group (a carbon atom linked to three hydrogen atoms) to the DNA molecule, typically at a cytosine nucleotide (one of the “building blocks” of DNA). These patterns play a pivotal role in gene expression because they can silence or activate genes without changing the underlying DNA sequence.

Scientists speculate that harnessing this natural epigenetic rejuvenation could be a key to promoting longevity and reducing age-related disease. This theory has taken the field by storm over the last few years and attracted a lot of commercial interest and new ventures trying to induce a state of “youthfulness” in cells by applying a process of epigenetic reprogramming.

Indeed, methylation presents a fascinating instance of the molecular alterations associated with aging. However, it represents only one facet of this complex process. A myriad of other molecular transformations and deteriorations in cellular function accumulate over time. The extent to which these changes affect gametes and whether they can be reversed by the embryo remains largely unknown.

To gain a deeper understanding of the relationship between aging and embryogenesis, researchers are delving deep, right into the core of gametes and embryos. Their goal? To identify any age-related alterations in parent cells that are subsequently erased as oocytes and sperm transform into an embryo, ultimately giving rise to a new organism.

The idea is to identify what molecular and functional changes occur as gametes age and to study whether embryogenesis has the capacity to reverse and alleviate some of them. To do this, scientists plan to use advanced technologies that can measure the abundance and state of a cell’s molecular components. These technologies include powerful tools like mass spectrometry which can be used to quantify and characterize proteins and the modifications they have undergone. This is important since protein damage and protein aggregates are thought to be a direct contributor to the aging process. Other tools like nanopore and SMRT sequencing have the potential to spot alterations in our DNA [5, 6]. These changes include various forms of DNA damage and understanding how these changes evolve with age and their behavior during embryogenesis, may uncover unknown aging and rejuvenation mechanisms.

However, the exploration doesn’t stop at proteins and DNA. Scientists are planning to do a deep dive into other components of cells, like lipids, metabolites, RNA, and the shape of DNA. They will gather heaps of data from these tests, then add functional data about different parts of the cells like mitochondria or lysosomes. When all these investigations come together, they would give us a detailed picture of how gametes age and how embryos deal with this.

These large datasets do more than just offer information about which facets of aging are undergoing rejuvenation. They also provide invaluable insights about the genes, peptides, and metabolites that drive the powerful genetic programs the embryo utilizes. This could prove very valuable, given that the embryo utilizes multiple processes that can be used to fight age related diseases.

For instance, in an intriguing series of experiments, scientists have injected cancerous cells into embryos. Yet, contrary to expectations, these malignant cells neither overwhelm the embryo nor are they eliminated. Instead, the embryo exercises a remarkable level of control over these intruders, compelling them to halt their proliferation [7]. Adding to the element of surprise is the fact that these transformed cells are not only maintained in tissues until birth, but they also undergo a complete reprogramming from their original cancerous state. It’s as if the embryo, in its potent nascent life, possesses a transformative capacity that can strip away the cells’ malignancy.

This intriguing process is likely to contribute to the intricate equilibrium required during tissue formation in embryogenesis. Unlike in cell culture where most cell types face a limit on the number of divisions, during embryogenesis the initial cells can form an entire organism without succumbing to replicative senescence. This is the result of elaborate cellular programs that grant cells the freedom to proliferate robustly while concurrently shielding them from becoming cancerous.

These are potent programs encoded within our very genome, holding potential applications for regenerating new tissue or combating malignancies in aging organisms. Indeed, certain creatures in the animal kingdom, like the aquatic hydra and some species of planaria, exploit similar processes to earn their ticket to immortality [8, 9]. Venturing into this new frontier of knowledge could unlock transformative therapeutic strategies, potentially reshaping our approach to age-related diseases and longevity.

While this all might sound like it’s straight out of a science fiction novel, it’s rooted in solid science. Even more exciting is that it’s mostly an uncharted territory! Despite the enormous potential of studying embryogenesis, it’s been an overlooked area in the longevity research world, making it ripe for exploration. Cracking the code of embryogenesis could very well be the key to understanding aging and how to reverse it. If successful, this could be a game-changer in how we approach aging and longevity. Forget about mythical fountains or enchanted forests; our ticket to timeless youth might just be nestled within the miracle of life itself!

Authors: Carlos Galicia & Maria Marinova

Editors: Rhys Anderson

Illustrator: Victoria Forest

References:

1. Kerepesi, C., et al., Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. Science advances., 2021. 7(26): p. eabg6082.

2. Smits, M.A.J., et al., Human ovarian ageing is characterized by oxidative damage and mitochondrial dysfunction. 2023, Cold Spring Harbor Laboratory.

3. Gladyshev, V.N., The Ground Zero of Organismal Life and Aging. Trends in Molecular Medicine, 2021. 27(1): p. 11–19.

4. Zhang, B., et al., Epigenetic profiling and incidence of disrupted development point to gastrulation as aging ground zero in Xenopus laevis. 2022, Cold Spring Harbor Laboratory.

5. Clark, T.A., et al., Direct detection and sequencing of damaged DNA bases. Genome Integr, 2011. 2: p. 10.

6. Wang, F., et al., Solid-State Nanopore Analysis of Diverse DNA Base Modifications Using a Modular Enzymatic Labeling Process. Nano Lett, 2017. 17(11): p. 7110–7116.

7. Astigiano, S., et al., Fate of embryonal carcinoma cells injected into postimplantation mouse embryos. Differentiation., 2005. 73(9–10): p. 484–490.

8. Sahu, S., A. Dattani, and A.A. Aboobaker, Secrets from immortal worms: What can we learn about biological ageing from the planarian model system? Semin Cell Dev Biol, 2017. 70: p. 108–121.

9. Muller, W.A., Pattern formation in the immortal Hydra. Trends Genet, 1996. 12(3): p. 91–6.

Scientists are forever searching for ways to rejuvenate aged cells and organisms back to a more youthful and healthier state - from stem cell therapies to telomere lengthening, or even the Frankenstein-esque heterochronic parabiosis experiments...
Read more
Decoding Longevity: DNA Mutations, Mole Rats, and Cancer Prevention with Prof. Vera Gorbunova on The Aging Science Podcast by VitaDAO
August 23, 2023
Awareness
Longevity
Decoding Longevity: DNA Mutations, Mole Rats, and Cancer Prevention with Prof. Vera Gorbunova on The Aging Science Podcast by VitaDAO

In the 7th episode of the Aging Science podcast we talked with Prof. Vera Gorbunova about her most recent work, covering a wide array of topics from genome stability, naked mole rats, the role of hyaluronan in cancer prevention, fucoidan as a SIRT6 activator, to comparative transcriptomics and Peto’s paradox.

Short Bio — Vera Gorbunova

Dr. Vera Gorbunova is a Professor of Biology and Medicine from University of Rochester. Having earned her undergraduate degree at at Saint Petersburg State University (Russia) and her Ph.D. at the Weizmann Institute of Science (Israel), she has dedicated her career to understanding the mechanisms of longevity across species, exploring genome stability, sirtuins, and non-traditional animal models. Her pioneering work on SIRT6 biology and potential activators like fucoidan, along with contributions to studying hyaluronan for cancer prevention, could pave the way towards therapeutic applications.

“I would choose to live for as long as I am enjoying it[…]it is very difficult to set a limit for yourself. That is what we would want for every person that people can live as long as they are happy.” (Vera Gorbunova)

DNA damage and aging

One of Vera’s key interests is genomic instability, which has emerged as a likely driver of aging. Mutations can lead to cancer, disrupt gene expression and the epigenetic architecture of the genome. This explains why long-lived species had to develop effective mechanisms for repairing or preventing such damage.

In a recent study together with Jan Vijg, Vera discovered that short-lived rodents, and in particular mice, show higher levels of DNA mutations before and after a genotoxic stress (Zhang et al. 2021). The technique they used to show this is particularly interesting. Without going into details, it is difficult to measure DNA mutations using classical sequencing due to high levels of background noise that looks like mutations. Many workarounds exist, often focusing on clonally expanded stretches of mutations. Here, instead, the authors performed whole genome sequencing of single fibroblasts from five different species that differed in their maximum lifespan (mouse, human, guinea pig, blind mole-rat, and naked mole-rat).

While the finding of lower mutation rates in long-lived species is correlational and indirect, it has been replicated many times and there is no clear alternative explanation for it that would be unrelated to aging or cancer. This is one of the reasons why the “DNA damage theory of aging” is generally held in high regard by most people in the field.

Comparative biology of aging

Researchers doing comparative biology or biogerontology of aging, as the name suggests, want to compare species that have different lifespans to discover why some live longer than others.

On this podcast we often talk about the shortcomings of mice, like their short lifespans and susceptibility to cancer (1). Comparative biology avoids these pitfalls of mouse models since it allows researchers to study a wide variety of species, some of which live orders of magnitude longer than mice and are more similar to humans.

Comparisons come in many types. Some researchers study dozens or even hundreds of species, if they can access the right kind of data for these species (2), whereas others pick two closely related species that have different lifespans and compare them. Often this is the mouse and the naked mole rat, which while mouse-sized nevertheless lives up to 30 years. The techniques employed are as varied as the sample sizes of these studies. In principle, one can compare any phenotype or molecular marker between species. Recent studies have focused on high-throughput methods, comparing the genomes or transcriptomes of different species.

Transcriptomics and other “omics” of aging

The expression patterns of cellular mRNA, the template that gives rise to proteins, can be studied using RNA-sequencing which we call transcriptomics. Vera’s team recently published a large-scale comparison of different rodents (Lu et al. 2022) confirming some candidate pathways we believed are associated with longevity. For example, longer-lived rodents, let’s say capybaras or naked mole rats, were found to have better repair of DNA double strand breaks, while pathways involved in the repair of smaller lesions were unchanged. This is keeping with an emerging consensus that double strand break repair is the most important type of DNA repair for aging.

Also, not surprisingly inflammation was reduced in longer-lived rodent species. Transcription factors related to pluripotency were linked with the expression of “pro-longevity” genes while “anti-longevity” genes were related to circadian transcription factors.

The study also reported some beautifully odd findings implicating novel pathways in aging, including collagen synthesis or RNA export. This is the kind of finding that, if it pans out, can spawn a new field of research.

While I love comparative studies utilizing omics, it should not go unmentioned that they have certain limitations. Given the large amount of data generated it is quite easy to find support for some of the popular aging theories, whereas lack of evidence is often ignored. No one ever talks about the aging theories that were not supported by the data that and what this means.

Moving forward, Vera’s next goal is to apply proteomics and metabolomics approaches to these rodent species, integrating this data with the transcriptomic findings. We both agreed that proteomics, i.e. the systematic analysis of protein expression patterns, is very under-utilized in aging research.

Extracellular matrix (ECM) and aging

The importance of extracellular matrix (ECM) changes to cardiovascular aging was discovered by Blumenthal and Lansing in the early twentieth century. Despite this early and seminal work, for a while there was a lack of interest in studying extracellular matrix changes during aging and the field is having a renaissance now. While Lansing studied a protein called elastin, Vera’s work is focused on glycosaminoglycans, which are a type of large sugar chains (polysaccharides).

“The length of hyaluronan [=hyaluronic acid] can range from an oligomer to an extremely long form up to millions of daltons. The concept that emerged in the field is that high (HMW-HA) and low (LMW-HA) molecular weight hyaluronans have different biological properties and trigger different signaling cascades within the cells. LMW-HA is associated with inflammation, tissue injury and metastasis, while HMW-HA improves tissue homeostasis and has anti-inflammatory and antimetastatic properties.” (Gorbunova et al. 2020)

In this episode of our podcast Vera explains that hyaluronan forms a mesh between tissues and cells that may be able to slow down metastasis and the growth of cancer more generally. It appears that naked mole rats possess high molecular weight hyaluronan that is particularly effective at preventing cancer and perhaps we might be able to slow down cancer growth by boosting our own production of hyaluronan. Achieving this goal, however, will not be simple since such a large molecule cannot be absorbed well from the gastrointestinal tract, which rules out a simple supplementation approach. Similarly, although hyaluronan injections are a popular treatment for wrinkles and osteoarthritis, they will not work to increase whole body hyaluronan synthesis because the hyaluronan molecule is large and “sticky”.

Instead, the idea is to inhibit the enzyme that breaks down hyaluronan which would also cause a shift towards larger molecular weight species of hyaluronan.

Although it remains speculative whether boosting hyaluronan synthesis or increasing the molecular weight of our own hyaluronan will be beneficial, it is nevertheless a good working hypothesis. This kind of work beautifully illustrates how basic and comparative science can lead to potential clinical applications

Below you can read more about Vera’s recent project that was funded by VitaDAO: https://snapshot.org/#/vote.vitadao.eth/proposal/0x747f0e671d6e049ce501fba8067c9da3b0502b8945daa890ca98f36a63bc7246

Summary

Prof. Gorbunova’s work reveals a correlation between long lifespan and low DNA mutation rates. Among other things, she also found changes in the hyaluronan of long-lived species. There is ongoing work to utilize these findings to develop therapies that target human aging, e.g. fucoidan as a SIRT6 activator or inhibitors of hyaluronan breakdown.

References

Lu, J. Yuyang, et al. “Comparative transcriptomics reveals circadian and pluripotency networks as two pillars of longevity regulation.” Cell Metabolism 34.6 (2022): 836–856.

Zhang, Lei, et al. “Maintenance of genome sequence integrity in long-and short-lived rodent species.” Science Advances 7.44 (2021): eabj3284.

Gorbunova, Vera, Masaki Takasugi, and Andrei Seluanov. “Hyaluronan goes to great length.” Cell stress 4.9 (2020): 227.

Notes

(1) The justification for mouse research is as follows. We do not use mice because they are a great model of long-lived mammals, instead you can consider them as “better worms”. They are simply the next best model after invertebrates that is still affordable for basic research.

(2) One accessible type of data is for example the sequence of the mitochondrial genome. Both Vera and I published research on this topic and as an “amateur” comparative gerontologist myself I always like following her work.

Pabis, Kamil. “Triplex and other DNA motifs show motif-specific associations with mitochondrial DNA deletions and species lifespan.” Mechanisms of Ageing and Development 194 (2021): 111429.

Yang, Jiang-Nan, Andrei Seluanov, and Vera Gorbunova. “Mitochondrial inverted repeats strongly correlate with lifespan: mtDNA inversions and aging.” PLoS One 8.9 (2013): e73318.


In the 7th episode of the Aging Science podcast we talked with Prof. Vera Gorbunova about her most recent work, covering a wide array of topics from genome stability, naked mole rats, the role of hyaluronan in cancer prevention..
Read more
July Longevity Research Newsletter
August 10, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
July Longevity Research Newsletter

Introduction

Welcome back Vitalians! Stop the press — the Impetus Longevity grants are back!

Having already deployed 24M USD to fund longevity research, round 3 is now open with 10M USD of funding, so if you have a great idea, get your application in before 31st August!

And the good news doesn’t stop there! VitaDAO has just opened another round of the Longevity Fellowship. We provide need-based micro-grants of up to $2,000 USD to passionate scientists, students, and longevity enthusiasts to delve deeper into their research interests, attend conferences, and join programs such as the Longevity Biotech Fellowship.
Quick tip: funding is distributed on a rolling basis so don’t wait too long to apply!

If real estate agents like to inform you it’s all about “location, location, location”, then drug developers would likely tell you it’s all about “delivery, delivery, delivery”.

It’s simply not enough to have a drug which works in a test tube, it has to be able to reach the intended target within the organism. The bioavailability of a drug can be diminished by various obstacles, from having to pass through the GI tract or the blood-brain-barrier, to navigating solid tumour microenvironments. As such, scientists are constantly looking for new ways to improve drug delivery, such as innovative nanotechnology-based carriers or developing prodrugs which can convert to being pharmacologically active at the site they are required. This month we’re excited to bring you an interview with Matthew Yousefzadeh, who has made significant contributions to the aging field, and touches on the bioavailability hurdles associated with the promising senolytic drug fisetin — enjoy!

Longevity Literature Hot Picks

Preprint Corner

The votes are in — The Longevist curators have decided what they think are the most important preprints of Q2. Before we publish the Q2 Issue of the Longevist, take a look at the inaugural issue of The Longevist which is now live!

Check out our first batch of preprints of Q3 with these July submissions. These will each be entered into the Q3 longlist to be in the running to receive a coveted place in The Longevist. They are also available to review on our reviewing platform The Longevity Decentralized Review (TLDR) for a bounty of 50 VITA per review.

As always, you can refer preprints to The Longevist and receive a bounty of 50 VITA for each one that makes the editors’ shortlist or 200 VITA if it makes the curators’ top 3.

LEF1 isoforms regulate cellular senescence and aging

Preserved striatal innervation and motor function despite severe loss of nigral dopamine neurons following mitochondrial dysfunction induced by mtDNA mutations

Alterations of the gut microbiome are associated with epigenetic age acceleration and physical fitness

Amyloid β accelerates age-related proteome-wide protein insolubility

The principal component-based clinical aging clock (PCAge) identifies signatures of healthy aging and provides normative targets for clinical intervention

Systems Age: A single blood methylation test to quantify aging heterogeneity across 11 physiological systems

Machine Learning identifies conserved traits that influence lifespan and healthspan responses to dietary restriction

β-hydroxybutyrate is a metabolic regulator of proteostasis in the aged and Alzheimer disease brain

Somatic mutations in human ageing: New insights from DNA sequencing and inherited mutations

A comprehensive atlas of the aging vertebrate brain reveals signatures of progressive proteostasis dysfunction

Glial-derived mitochondrial signals impact neuronal proteostasis and aging

Published Research Papers

Chemically induced reprogramming to reverse cellular aging

The Sinclair lab have identified chemical cocktails which can reverse transcriptomic age in cells. Thus they claim that age reversal can be achieved by chemical means. This paper has sparked controversy due to the speed of publication, questions of novelty, and overhyped claims. Take a read and make up your own mind!

The million-molecule challenge: a moonshot project to rapidly advance longevity intervention discovery

From microscopes to space telescopes,

technology and science need to work together to make advances possible. Using the WormBot-AI automated data capture and analysis technology, the Kaeberlein lab are taking on the ambitious challenge of screening a million molecules for their effects on lifespan in C. elegans.

Large-scale across species transcriptomic analysis identifies genetic selection signatures associated with longevity in mammals

Among mammals alone, lifespan can vary over 100 times between species. Here the authors performed transcriptomics across numerous species and identified that translation fidelity pathways correlate with lifespan and methionine restriction genes are under strong selection in long-lived mammals.

Here we have back to back published studies showing that senescent macrophages drive lung cancer and accumulate in aging

More than bad luck: Cancer and aging are linked to replication-driven changes to the epigenome

The Levine lab’s study suggests that aging and cancer share a common epigenetic replication signature, termed CellDRIFT. This signature increased with age across tissues, distinguished tumor from normal tissue, was escalated in normal breast tissue from cancer patients, and could be reset upon reprogramming.

Multi-omic rejuvenation and life span extension on exposure to youthful circulation

Heterochronic parabiosis (HPB) in mice has shown significant rejuvenation effects, improving physiological parameters and extending lifespan. HPB reduced the epigenetic age of blood and liver, with the effect persisting even after detachment. The transcriptomic and epigenomic profiles suggest a global rejuvenation effect.

Maternal aging increases offspring adult body size via transmission of donut-shaped mitochondria

Maternal age significantly influences offspring adult traits like body size. This study utilized C. elegans to explore the mechanisms, revealing aged mitochondria transmitted from older worms are rejuvenated in offspring in an AMPK-dependent manner. This early-life mitochondrial dysfunction triggers AMPK and TGFβ signaling, increasing adult offspring size.

Multiparametric senescent cell phenotyping reveals targets of senolytic therapy in the aged murine skeleton

Using mass cytometry, researchers identified and analyzed senescent cells in aged mice at single-cell resolution. They found these cells in certain skeletal cell populations could be robustly cleared by senolytic therapies, providing a method to target senescent cells in vivo.

Lifespan-extending interventions induce consistent patterns of fatty acid oxidation in mouse livers

A study investigating the gradual molecular dysregulation in aging finds that lifespan-extending interventions like acarbose, 17α-estradiol, rapamycin, and calorie restriction generally tighten the regulation of biological modules. These patterns are similar across interventions, particularly in processes such as fatty acid oxidation, immune response, and stress response.

Pan-primate studies of age and sex

Horvath presents accurate pan-primate epigenetic clocks based on DNA methylation profiles from 2400 tissues across 37 primate species. The study explores the impact of age and sex on cytosine methylation, identifying 11 sex-related CpGs on autosomes. The findings provide insights into conserved age- and sex-related epigenetic changes and biomarkers of aging in primates.

Predicting lifespan-extending chemical compounds for C. elegans with machine learning and biologically interpretable features

Researchers used machine learning to analyze DrugAge, a database of compounds that modulate lifespan in model organisms. They identified the importance of the “Glutathione metabolic process” and predicted new promising compounds for extending lifespan, including nitroprusside, an antihypertensive medication.

Published Literature Reviews

Chronic inflammation and the hallmarks of aging

The review suggests that chronic inflammation, known as “inflammaging,” is a critical driver of aging and age-related diseases. It is linked with other aging hallmarks, exacerbating cellular dysfunction and advancing aging. Targeting inflammation could be key in developing anti-aging interventions and addressing age-related conditions.

An updated landscape of cellular senescence heterogeneity: Mechanisms, technologies and senotherapies

The review emphasizes the critical role of senescent cells in aging, and the need for better identification methods. Despite the challenge posed by the heterogeneity of senescence, high-throughput technologies like single-cell RNA sequencing could enable improved analysis and potential discovery of new markers.

Stress, diet, exercise: Common environmental factors and their impact on epigenetic age

This review explores how lifestyle factors influence the DNA methylation landscape, as revealed by epigenetic aging clocks. Lifestyle factors can impact DNA methylation and consequently influence biological aging. Understanding these relationships could provide valuable insights for people interested in creating a lifestyle designed to promote longevity.

Spatial mapping of cellular senescence: emerging challenges and opportunities

This review discusses the challenges of mapping senescent cells, crucial for understanding aging. Current single-cell technologies lack spatial data, which is key due to the interactive nature of these cells. New spatial imaging methods are needed to create a comprehensive senescent cell atlas.

Job board

VitaDAO’s portfolio company BE therapeutics is looking for a scientist with tissue engineering or neuro lab experience for some exciting work on brain replacement lead by Jean Hebert. Send CV to jean.hebert@einsteinmed.edu to be considered.

Korolchuk lab in Newcastle University are hiring for an exciting new position of lab technician which will support an industrial project in collaboration with Procter & Gamble.

Explore new mechanisms of how DNA damage drives aging and alters heritable genomes at the Schumacher Lab at CECAD Cologne. They are seeking enthusiastic postdoc candidates with a background in genetics, cell biology, biochemistry or computational biology.

Prof. Marco Demaria is looking for an enthusiastic and ambitious PhD candidate to join the Laboratory of Cellular senescence and age-related pathologies (Groningen, Netherlands), to work on a project studying the contribution of senescent cells to inflammatory and fibrotic chronic disease.

Research Associate position available in Dr. Gordon Lithgow’s laboratory at the Buck Institute for Research on Aging. The Lithgow lab combines biochemical, molecular, and genetic techniques to address fundamental questions about aging in C. elegans.

Brieno-Enriquez lab is looking for two motivated postdocs to explore reproductive aging in the naked mole rat. They will leverage the NMR to explore mechanisms that rejuvenate the ovary and extend both reproductive lifespan and overall health.

News and Media

FDA grants full approval to Leqembi, opening up coverage of Alzheimer’s drug by Medicare

After a dispute with the publisher resulted in the resignation of 5 editors-in-chief from Aging Cell — the former editors have now returned with a new scientist-led journal — Aging Biology — which is now accepting submissions and publishing papers.

Check out Lada Nuzhna’s reflections on 2 years of going after risky aging science.

USA Today: Want to live healthier longer? Scientists aim to improve life quality over quantity

8 healthy habits that may add 24 years to your lifespan

The Wall Street Journal: The Longevity Clinic Will See You Now — for $100,000

Twenty minutes of daily exercise can help reverse frailty and build resilience in over 65-year-olds

The boom of the anti-aging market: How to get people to live to be 120 (and in good health)

The Daily Mail catches up on the drama in the aging research world: EXCLUSIVE: Harvard claims it found the elixir of youth — but experts call it ‘hype’

Mitochondrial DNA fragments in blood shown to be important biomarkers for aging

Peter Fedichev Explains His Theory of Aging

Healthy longevity is not only for the neurotypical

Resources

Norn Group’s age related diseases overview: an excellent resource of early stage startups in the longevity space that are looking for an initial clinical indication to target

Prizes

Rewarding the most impactful research in longevity

Substantially increasing the human lifespan. The Amaranth Prize gives no-strings attached funding to the best research in Longevity.

Conferences

Ending Age-Related Diseases

10–11 August, NY, USA and virtual

Longevity Summit Dublin

17–20 August, Dublin Ireland

Aging Research & Drug Discovery (ARDD)

28 August — 1 September, Copenhagen, Denmark

British Society for Research on Aging Annual Scientific Meeting (BSRA ASM)

6–8 September, London, UK

RAADFest

7–9 September, LA, CA, USA

Expo and Convention: Independent Ageing 2023

13–15 October, Aichi, Japan

Seno-Therapeutics Summit 2023

7–8 November, Buck Institute for Research on Ageing, Novato, CA, USA

Tweet of the Month

Adam Gries (@adamgries):


The idea that we shouldn’t work hard to extend lifespans (which has been echo’ed by @ elonmusk) would make more sense , if we weren’t *already* spending like crazy to increase lifespans, just doing it poorly.

Trillions now go to the last 5 years of life. […]

Podcasts and Webinars

Join us for a Twitter live discussion on the latest epigenetic clock research

7 August, virtual

Norn Group presents: Near Future Skill Gaps in Software & ML discussing current & impending changes impacting aging bio research due to advances in software & ML. Featuring Joe Betts-LaCroix, Luca Naef, Andy Lee, and Sam Spurlin

9 August, virtual

Did you miss our Robust Mouse Rejuvenation Twitter live with Aubrey de Gray, Caitlin Lewis,

Kelsey Moody, & Danique Wortel? Don’t worry, the recording is up.

We also interviewed Sergey Young!

NUS Medicine’s Healthy Longevity Webinar Series

This month:

Ovarian Ageing: A Target for Geroprotection in Women | Prof Suh Yousin

AI and robotics | Dr Alex Zhavoronkov

Taurine deficiency as a driver of ageing | Asst Prof Vijay Yadav

Funding Opportunities

Research: Impetus Longevity grants

Micro-grants: VitaDAO Longevity Fellowship

Interview with Matthew Yousefzadeh

Matthew is an instructor at Columbia University Medical Center where he works on mechanisms of aging, in particular how DNA damage and cellular senescence contribute to cell autonomous and non-autonomous aging.

What inspired you to enter longevity research?

Previously, my background was in cancer biology and much of my PhD work was on how specialized DNA polymerases could play protective roles in limiting genotoxic stress, but their levels had to be tightly regulated. Too much of these enzymes could be permissive to tumorigenesis or make them refractory to treatment. I got into cancer biology because during my freshman year of college my grandmother survived one bout with breast cancer but succumbed to a second round of cancer. Likewise, as I was finishing my PhD, I saw my mom start to accumulate some age-related health issues and she died at a young age. This was the impetus for me to get interested in aging research. Going to Laura Niedernhofer’s lab was a no-brainer and provided a natural transition for me where I could take my DNA repair background and use it to study senescence.

How has the field changed since you started?

My plan was to go to Laura’s lab to study senescence that was induced by DNA damage or through DNA repair deficiency. At the time, I had no clue what senolytics were and the first senolytics paper was published a few weeks before I showed up to the lab. Obviously, the senescence field has matured a lot since then and still has a way to go. Outside of that there have been incredible discoveries in many of the other pillars of aging like the broadened use of epigenetic clocks, epigenetic reprogramming, and use of companion animals as a method to study aging. The field felt a bit slower back when I started my postdoc, and it is moving much more rapidly now. This is both great because I feel like the speed of progress is picking up, but terrifying from a standpoint of how many good papers are coming out every day that I need to read up on. It is a fantastic problem to have though! Recently in the past few years there seems to be a greater public interest in aging, and I think it has turned into a prime opportunity to bring young enthusiastic minds into the field of geroscience. Also, we are seeing people in other diseases spaces like cancer begin to get much more serious about looking at the interplay between their disease of interest and aging. From a commercial standpoint the number of longevity companies was really limited to Calico and in the realm of senescence, Unity Biotechnology. In the past few years, this aging biotech landscape has exploded, which I find to be a positive sign.

Other than your own, what do you think have been the biggest/important discoveries in the field?

While some of these are controversial spaces, I think the work done by too many labs to individually list on these topics are a boon for geroscience: 1) Levels of in disease and aging, 2) epigenetic reprogramming and functional outcomes in regards to rejuvenation, 3) understanding how centenarians age so well, 4) transgenerational effects on aging and longevity, and 5) the use of C. elegans to conduct high throughput screening of monotherapies and combinations to determine possible geroprotectors. Each of these are very important and in their own individual way could serve as a disruptor in the field of aging.

What advice would you give to people currently working in longevity research?

Find something you care about working on and go all in on it. Some days you will love it and other days it will frustrate the hell out of you. I have seen some really good scientists run away from projects when they get their first piece of negative data or experience some difficulty. Some of them have tried to jump from what they thought was a hot project to next hot project because it was “cooling off” and eventually they burnt themselves out. Learning resiliency can be tough but find something that you can stick and see it through. It is so rewarding! Another piece of advice is to find a place where people are invested in your development. I tell graduate students and other early career researchers to chase mentorship and not projects. Good mentorship can set you on a path to success when you are at such a nascent stage in your career. I also think programs like the MBL Biology of Aging Advanced Research Training Course that was created by Jennifer Garrison and Will Mair will serve as excellent conceptual and technical primers for people new to the field or looking to expand their skillsets. Get involved! Join the American Aging Association or the Gerontological Society of America and get involved with the trainee chapters or other committees. Trainees can take part in the peer review training program at Geroscience where they can independently review manuscripts. If you are interesting, contact me.

Which aspect of longevity research do you think requires more attention?

I’m going to say there are two big areas that deserve more attention: 1) validation of biomarkers and reagents; and bandwidth and best practices for bioinformatic analysis. The first has been a long-standing issue not just for aging but for other fields as well. We all know biology can plagued by poor reagents like antibodies that have not been properly validated or how many of the markers in my own field of senescence are non-specific. Discovering better markers and building better tools that we are confident will go a long way to expediting progress. Let’s face it, resource validation is not sexy, but it is vital.

To the second point, it has been incredible to watch the explosion of ‘omic and multiomic analysis in the field of aging. I wish I had access to single cell RNAsequencing as a first-year postdoc, it would have made my life a lot easier. A lot of us, me included, are trying to play catch up to the ever-shifting landscape of technology in this area and its inherent strengths and weaknesses. We desperately need bioinformatics support to help us process the mountains of data and we have been fortunate to draw a lot of very talented people from the tech sector into aging in the last few years as we also home grow bioinformatics talent. My last point on this topic is that many of us are looking for resources on best practices for these topics so that we can plan and execute the best possible experiment. One example of this is a recent publication from Param Priya Singh and Bérénice Benayoun on “Considerations for reproducible omics in aging research.” I think this manuscript is incredibly informative and can serve as a great reference for anyone using ‘omics to study aging. Speaking of advice, I am also incredibly fortunate that Bérénice is a friend and has been generous with giving me advice as a postdoc and now as a PI.

Is ageing a disease?

This is how you start a fight over beers when you get a bunch of aging researchers together. I think it is a disease, but also a natural and omnipresent condition. A very milquetoast take right? Realistically it probably sits in between the two states. Some people feel very strongly about it, but I’d rather focus on the science rather than semantics.

You have shown that fisetin is a senotherapeutic that extends health and lifespan in mice — what do you think is the biggest challenge in bringing fisetin to the clinic?

Fisetin is interesting in that we wanted to demonstrate that a senolytic could be safe but effective. Dasatinib, a chemotherapeutic that is used in combination with quercetin as a senolytic, has side effects. The natural product fisetin was originally identified through screening other flavonoids to see if they had more potent senotherapeutic effects than quercetin. I think two big hurdles for bringing fisetin to the clinic is that since it is a natural product it has a very limited intellectual property (IP) position and from a drug standpoint it has poor bioavailability. To address the second point, Kyle Brewer, a talented aging biologist, at ETTA Biotechnology is working to develop an optimized formulation to better deliver fisetin. Meanwhile the laboratory of my former co-mentor at the University of Minnesota, Paul Robbins, is developing fisetin analogs to increase potency and bioavailability, as well as create novel IP around these analogs. Paul has co-founded Itasca Therapeutics, a platform company which I currently serve as the CSO for, to develop these compounds and other senolytics. Itasca Therapeutics was created because we found that the typical funding mechanisms for academic research were insufficient to push lead compounds discovered in the academic lab further into the drug development thus the needs to acquire private funding.

Which other senolytic strategies do you think hold promise and what are the potential negative impacts of removing senescent cells?

The field of senolytics is growing and so are their approaches. These include everything from the use of AI-guided drug design, protein degraders of targets (PROTACs), CART cells, and activation of immune clearance mechanisms. I find Anil Bhushan’s work on senescent cell elimination by invariant (i)NKT cells to be both intriguing and refreshing. Many of us are looking at senescent cell accumulation as purely detrimental and studying them in the gain of function context. Others like Anil view senescent cells are serving a regulatory role and asking questions about why we fail to clear them as we age. This work is addressing the other side of the senescence equation (loss of function in the form of immune clearance). Furthermore, not all senescent cells are deleterious and should be cleared. A few years ago, a group showed that genetic clearance of senescent liver sinusoidal endothelial cells (LSECs) that expressed high levels of the p16Ink4a gene had detrimental effects and caused a pro-fibrotic response in the livers of these mice. These cells were not affected when the mice were treated with dasatinib and quercetin. Furthermore, it was found that expression of p16Ink4a specifically in the pancreatic beta cells of transgenic mice enhanced glucose-stimulated insulin production and provided an unexpected benefit to diabetic mice. Studies like these serve as a cautionary tale to not be hasty to eliminate all senescent cells or all cells that express p16Ink4a. We are fortunate that the newly created NIH Common Fund Cellular Senescence Network program is working to better characterize the heterogeneity of senescent cells in multiple tissues of both humans and mice across the lifespan. This could provide greater insight into specific senescent cells and how best to target them.

Which other interventions do you think hold promise for improving human healthspan/lifespan?

One of the most efficient interventions to democratize longevity is exercise as medicine. Beyond that interventions range anywhere from practical to promising to vaporwave. If I had to pick a front runner it would be mTOR inhibition whether it be in the form of rapamycin or newer generation compounds that selectively target mTORC1. It is one of the most reproducible results throughout multiple animal models and the human data on mTOR inhibition has been very encouraging. Columbia University Medical Center, where my lab is located, has just launched the VIBRANT (Validating Benefits of Rapamycin for Reproductive Aging Treatment) study, VIBRANT is being co-led by Yousin Suh, a well-known figure in the field of aging and director of reproductive aging at CUMC. If rapamycin treatment can slow down ovarian aging in humans, that would be huge!

You have also shown that an aged immune system has a causal role in driving systemic ageing — which therapeutic strategies do you think could help tackle this?

Speaking of mTOR inhibition, Joan Mannick’s work on vaccination rates in the elderly and outcomes with respiratory tract infections or Tyler Curiel’s work on cancer models was very inspiring to my own work. Beyond that the work on enhancing senescent cell clearance via immunotherapy is very exciting. Overall, I think it is going to take a combinatorial approach to rescue some of the age-related deficits in immune function.

You recently relocated to Columbia University Medical Center — what can we expect from the Yousefzadeh lab?

I am really interested to continue studying the role of senescent immune cells and how they affect systemic aging through interorgan communication. Beyond that I am interested in the role of cellular senescence (immune cells and beyond) on adverse outcomes in pathogenic infections. Previously, I established a human senescence core at UMN and plan to do the same at CUMC to help measure biomarkers of aging in humans. Moving to New York City has put me in close contact with long-time collaborators like Derek Huffman at Albert Einstein College of Medicine and allows me to foster new collaborations. Beyond that I have a strong interest in DEIB and have participated in multiple outreach programs that work to provide opportunity and exposure to promising young scientists from all backgrounds. I want to continue my work with these programs and help mentor the next generation of scientists that want to embark on careers in aging and longevity. If you are in NYC, let’s grab a slice of pizza and talk science. My treat!

Outro

We appreciate you sticking with our research newsletter for another month and hope the content we curate is useful!

This time we leave you with a short summary video of what VitaDAO does and why.

Further Reading

Translational longevity medicine: a Swiss perspective in an ageing country

Microvesicle-Mediated Tissue Regeneration Mitigates the Effects of Cellular Ageing

Trial of the MIND Diet for Prevention of Cognitive Decline in Older Persons

Cellular senescence induction leads to progressive cell death via the INK4a-RB pathway in naked mole-rats

Changes in methylation-based aging in women who do and do not develop breast cancer

RNA polymerase II associates with active genes during DNA replication

Chromatin accessibility dynamics of neurogenic niche cells reveal defects in neural stem cell adhesion and migration during aging

Novel 1,4-Dihydropyridines as Specific Binders and Activators of SIRT3 Impair Cell Viability and Clonogenicity and Downregulate Hypoxia-Induced Targets in Cancer Cells

Senolytics Reduce Endothelial Cell DNA Damage and Telomere Dysfunction Despite Reductions in Telomere Length

Associations of Dental Health With the Progression of Hippocampal Atrophy in Community-Dwelling Individuals: The Ohasama Study

Cocoa flavanols improve peakVO2 and exercise capacity in a randomized double blinded clinical trial in healthy elderly people

Removal of senescent cells reduces the viral load and attenuates pulmonary and systemic inflammation in SARS-CoV-2-infected, aged hamsters

Prolonged fasting times reap greater geroprotective effects when combined with caloric restriction in adult female mice


Welcome back Vitalians! Stop the press - the Impetus Longevity grants are back!
Read more
VitaDAO Letter: A New Journal, Ambassador Program, and New Steward!
July 27, 2023
Sarah Friday
VitaDAO Letter: A New Journal, Ambassador Program, and New Steward!

Inside this Newsletter:

  • The First Edition of VitaDAO’s Journal, The Longevist 
  • VITA-FAST Recap: Redifing IP-NFT Governance 
  • Building Collaborative Connections with an Ambassador Program 
  • Meet VitaDAO’s New Dealflow Steward: Eleanor Davies

🧬The Longevist is LIVE

VitaDAO is thrilled to announce the launch of "The Longevist," an overlay journal that curates promising longevity research. In the inaugural edition, we present the top ten preprints from Q1 2023, as handpicked by an esteemed panel of experts. The expert curators involved in the selection process are renowned professionals and scientists in the field of longevity.

Three a few noteworthy studies featured in the inaugural "The Longevist" feature:

🤑VITA-FAST: A Novel Funding Module 

In collaboration with Molecule, VitaDAO proudly launched VITA-FAST, a groundbreaking initiative that has redefined governance of Intellectual Property (IP) and Research and development (R&D) data through the use of IP-NFTs (Intellectual Property Non-Fungible Tokens). VITA-FAST tokens represent ownership in the IP pool and grant holders the power to participate in governance, make crucial IP licensing decisions, influence experiment priorities, and govern research projects. Thus, the creation of tokenized IP pools using IPNFTs allows DAOs to cooperatively manage collective IP while co-governing development and commercialization.

The IP pools, governed by token holder-set rules, provide solutions to existing challenges in the biotech IP market. In minimizing risk management, aiding in capital formation, and increasing patient involvement in therapeutic development, this transparent process is revolutionary. Its transparent and efficient process promises to accelerate scientific progress and foster innovation. It is possible to imagine that the creation of liquid and efficient markets can solve some of the issues in the current funding of bio IP.

The VITA-FAST initiative was completed in conjunction with the Korolchuk Lab, a lab led by cellular biologist Prof. Viktor Korolchuk. Dr. Korolchuk's research focuses on identifying novel bioactive autophagy inducers, a process that plays a vital role in the body's cellular health. By administering potential activators to cells unable to initiate autophagy, Dr. Korolchuk aims to demonstrate the specificity and effectiveness of compounds in promoting autophagy.

This project is still young and developing! For example, VDP-111, proposal passed on Snapshot, proposes the election of a project lead by VITA-FAST token holders and outlines a VITA-FAST governance framework. 

“This is the next phase in the evolution of decentralized patient communities, to directly contribute to, govern, and be rewarded for progressing valuable longevity research.” - Alex Dobrin

🌎VitaDAO’s Ambassador Program: Building Connections and Facilitating Collaboration ​

In March 2023, VitaDAO community leaders, Gavin and Cat, proposed the creation of a VitaDAO Ambassador Program. The goal of the program was to propagate DAO outreach and increase DAO engagement. 

This month in London, Bharat, Maria Maroniva PhD, and Eleanor Davies spoke at the Ambassador Program’s first official event. They touched on DeSci and VitaDAO's significant contributions to shaping the field. This event was just the beginning of a much larger initiative!

The VitaDAO Ambassador Program aims to enable individuals from different jurisdictions to organize in-person meet-ups, bridging the gap between virtual and face-to-face interactions. Many in the DAO community will agree that the value of interaction and engagement is much higher when meeting mission-aligned people in person! To select a group of DAO Ambassadors, an application was sent out. Thus far, we have received an overwhelming response with over 170 applications to become a VitaDAO Ambassador! Stay tuned to hear more about VitaDAO’s selected ambassadors and events occurring near you!

🗣️VitaDAO in the Wild

📣Community Approved: What YOU voted for!  

Thanks to the support of the VitaDAO community, multiple proposals successfully passed this month! Notable proposals include:  

VDP-89 Passed! This proposal was an assessment of ExcepGen Inc's RNA therapeutics for longevity. In passing, this proposal signifies that VitaDAO supports funding $100,000 to develop a new generation of vaccines with this RNAx platform.

VDP-93 Passed! As a result of this VDP, VDP nomenclature, and phase 2 proposal workflow will be altered to ensure visibility of risks and legal review before on-chain voting.

VDP-96 Passed! With this proposal, VitaDAO will provide a loan of $400k to VHF, secured against the underlying equity in companies VHF.

VDP-101 Passed! As a result, VitaDAO will set up an operating company in Canada to execute VitaDAO Coordination Working Group functions such as finance, member services, and service provider management. 

VDP-102 Passed! VitaDAO will now implement 7 new tools to better manage the growing portfolio of the VitaDAO projects more effectively and efficiently. 

VDP-107 Passed! As a result of VDP-107,  Laurance Ion stepped down as Dealflow Steward and Eleanor Davies was elevated as Interim Dealflow Steward.

VDP-108 Passed! With the passing of VDP-108,  VitaDAO will complete an annual review of compensation/governance allocations as outlined in VDP-72. 


💪Exercise Your Right to Vote

Check out VitaDAO’s governance hub (Discourse) to engage with, vote on, and discuss proposals before they are moved to Snapshot. Your input is invaluable, and together, we can shape the future of VitaDAO and accelerate decentralized science.

👋Meet VitaDAO’s Dealflow Steward: Eleanor Davies 

Eleanor Davies, VitaDAO’s New Dealflow Steward, brings a wealth of experience in fund operations, healthcare, and biotech consulting. A graduate of the University of Manchester, she has grown as a professional serving in Marwood Group healthcare and biotech consultancy, co-founding a company and helping to raise funds, deploy capital, and manage the portfolio at ODX, a Silicon Valley-based accelerator. More recently, she has actively engaged with the DeSci community, serving as COO of LabDAO. This month, I had the privilege of sitting down for a brief chat with Eleanor: 

It’s great to meet you, Eleanor! I would love to know a little more about your background. How did you first get involved in VitaDAO?

Hi! Thank you for featuring me in the VitaDAO newsletter. A little about me: I come from a less traditional background. I first started off studying languages, combined with international policy, business, and history. This gave me the flexibility to look into different industries that were a good fit for me. I found that I had an advantage as a language learner in the UK, because not many of us Brits speak other languages. If you show you are highly motivated, ambitious, and willing to take on new challenges, it is generally quite well-regarded by graduate employers.

From the start, I have always had a lot of side projects and interests. I have a long-standing curiosity in biotech, specifically in pushing the limitations of human biology - is life extension really possible? How would this work in practice? I started by working at IBM, and later in M&A advisory across industries in the German speaking (DACH) mid-market for private equity transactions. With time, I found myself gravitating towards the biotech side of M&A.

Between then and working in consulting, I co-founded an AI-based recruitment company - that was a lockdown existential crisis! It came out of the frustration I witnessed by the number of people who lost their jobs during the pandemic. What if I could change the way of looking at the job market? Tldr; it didn't work out, but it was a great learning opportunity. There was a market fit and we were generating revenue, but it was more about the team dynamic.

I then worked in consulting mid- to large-cap private equity clients for healthcare and biotech transactions. The consultancy specialized in advising the commercial implications of political and regulatory change across the UK and EU to domestic and transatlantic clients. Early during my tenure, the consultancy Director left, so I had to step up and lead in the role of Director - from the experience level of an Analyst. It was a very steep learning trajectory, but I saw it as an ideal opportunity to step up. I did a lot of self-directed learning, not only for interfacing with clients, but also for company growth, negotiation, and leadership. Alongside the consulting, I also doubled company sales targets, doubled the pipeline for marketing prospects, and transformed the marketing channels. A lot of progress in a short amount of time! 

I then moved on to work at On Deck’s ODX, which is an accelerator based in Silicon Valley. I was working in fund operations, working across raising funds to deploying capital, conducting due diligence, and portfolio management. Almost similar to what I’m doing at VitaDAO today, Sadly, due to the market conditions, I had to leave ODX, but this allowed me to pursue my nerdy side interest - DeSci!

That leads me to today. I had been a “lurker” in both the VitaDAO and LabDAO Discord servers for some time and finally decided to do something about it. I was introduced to Niklas from LabDAO at DeSci Berlin - I was so motivated by the vision, the highly intellectual community, the level of passion, and mission alignment in the ecosystem. So, I made the leap! For me, DeSci could be hugely disruptive for the scientific community, funders, policy makers, and beyond. In the consulting world, I had seen a misalignment between politicians and regulators, funders and companies, healthcare insurers, and patients. Not only in capital allocation, but in communication, education, and expectations. For me, a community-first, community-governed approach could create the new standard.

As part of the core team at LabDAO, Niklas and I led the fundraise ($3.6M). It was a challenging time to raise in a bear market, but it was gratifying upon completion. I also worked across legal, tokenomics, investor relations, and treasury management… all-in-all, a bit of Swiss Army Knife-ing. It was hugely satisfying to see LabDAO through to maturation. During its PMF phase, LabDAO pivoted to tools versus the Lab Fund, which I was to lead. As everything was up and running, my role eventually phased out. By that time I was closely following VitaDAO and knew that there was a need for somebody to work in deal flow. It was the ideal time to join. Now, I’m still advising LabDAO, but full-time at VitaDAO. 

You mention that you find DeSci to be promising. What drew you to DeSci and where do you see DeSci going in the future? 

There are many aspects that I find interesting about DeSci; for me, I’m excited about tools, funding, and globally distributed venture building. This is why I am confident in VitaDAO and why I’m motivated to make it succeed. We are looking at a broader pool of capital for higher risk funding opportunities, and zooming in on the early, idea stage of lab research projects. The IP-NFT is particularly interesting here. VitaDAO and the community, as part of the Decentralized Tech Transfer Spinout framework, work with the scientific team to build projects from idea, to IP commercialization, to maturation. To my knowledge, this hasn't been done before with Web3 and DeFi technology.

Also, never before has there been such a globally distributed community of mission-aligned people to work together in this way. The thing that I find special about the VitaDAO community is that world leading talent, especially in the dealflow working group, participate out of sheer interest. There is a real mission alignment here. And despite some DAO inefficiencies, its youngness, and teething issues, DAO members stick around. 

I’m also excited about the IP-NFT, pioneered by Molecule. The non-dilutive funding, and the ability to get more proceeds for project development in this manner is a pretty innovative way - also high risk, but high reward - of funding a project and getting more community interest. It promotes openness, making research a lot less siloed and more collaborative. And even if it doesn't work out at least we’ve tried. But, we’re making waves already.

You’ve touched on the strengths of DeSci and some of the strengths of VitaDAO in particular. Is there a particular VitaDAO project you are most excited about? 

There is - Matrix Bio! I’m also PM’ing that alongside Anthony Schwartz as a part of the Builder Squad. It's a moonshot, but this could have a huge payoff for the LongBio world. We are working with one of the world's leading aging researchers, Vera Gorbunova. Not only is she a pleasure to work with, but also her research is groundbreaking. The fact that she has her belief in VitaDAO to incubate her project towards a point of commercialisation, is compelling and exciting.

Another would be Viktor Korolchuk’s autophagy project. Again I think that not only is it an under-resourced side of longevity aging research, but also that he and the Newcastle University have the conviction that VitaDAO can build and incubate the project, using an innovative funding and governance method. This is a strong signal for me.

As a result of VDP-107, you’re now Longevity Dealflow Working Group Steward. In your short time in this role, what has surprised you the most about VitaDAO? What are you most excited about in taking on this role? 

First of all, I’m very very grateful for those who voted in favor and have the confidence in me to be the Dealflow Steward. What am I most excited about? I’d say the support of the community. Putting the collective wisdom in the deal flow working group to great use! We have so much talent; so many amazing people are eager to get involved with the opportunities that come our way. For me, something that dealflow needs is great operations. It is the cornerstone of making things work: ramping it up and scaling it up to a mode of maximum efficiency. I’d also like to ramp up Tech Transfer Office outreach, project sourcing, and both start-up and early-stage IP projects. And have an engine that enables this to happen: a great funnel and a great process. As boring as they sound, you need these to make things work.

Also knowing how to engage with the community in different stages. We have so many talented people who are amazing in different aspects of the deal flow process. It is knowing what incentives are, and how contributors should be incentivized in the DAO from the start; something which is still being tested out in the Web3 ecosystem. And, how can we be efficient enough to become the world's best longevity research funding organization? How can we become the first place that people think of when looking for funding, looking for education, or an amazing community, when they think of longevity? That is what I want for VitaDAO. I want it to be on the UN’s radar. Dream big. 

When I first joined the VitaDAO Discord, I initially had no idea about all the work that goes on behind the scenes at VitaDAO on the Dealflow side of things. There is a lot more work that goes on before projects are presented on Discourse. 

It's true. From automating the dealflow funnel to refining IP strategy with projects, to Friday pitch sessions and onboarding new members - we are taking VitaDAO from 0 to 1. If anyone wants to get involved, come to an onboarding call! I lead the calls with community member, Ryan Spangler, and it’s a great first point of entry to join the working group. We go through terms, voting, the nuances of dealflow, DAO-ism, and how VitaDAO is different from a traditional venture studio or VC. Why is it a not-for-profit? What is an IP-NFT? I find the best way to get great engagement when onboarding new community members to deal with the flow, is to have a chat instead of reading through documentation. To anyone who wants to get involved, feel free to reach out!

Where do you see VitaDAO 100 years from now? 

I wonder if VitaDAO will have solved aging by 100 years? First off, I want VitaDAO to be the biggest name in longevity. This involves being a well-oiled machine. People come in because they’ve heard about our reputation in longevity and for being the best DAO that works to maximum efficiency. I’d like to have had successful exits (everyone wants successful exits), and successful spinouts that will have IPO'd. Happy founders. Happy community members. A cyclicality of members who come, build companies, grow it, scale it, exit, come back, and continue. On the network state side, bridging successful spinouts with the possibilities achieved by a longevity network state is another goal. Leveraging the more friendly regulatory pathways in specific jurisdictions could be an ingenious way that VitaDAO can expedite scientific progress - if done correctly. Zuzalu was an amazing way of showing us this potential. What about novel cures or treatments that are more financially accessible - or generally more accessible. That’s another dream, and I hope we can weigh in there, too. It'd be amazing for VitaDAO to be a front-runner in changing how cures come to market and also how we behave around human health- and lifespan.

I’ve enjoyed chatting with you today! Before we end, any closing thoughts for readers?

Again, I am very very appreciative of being the Steward in Dealflow. Of having the support of the community. Reach out if you are interested in getting involved in dealflow, interested in knowing more about spinouts, the builder squad, and generally getting involved. If you are a builder. If you are an operator. If you’re an academic. If you are interested in pursuing a project, a side project, and looking for funding. Also please reach out or apply. I am always interested in looking for new opportunities and the same goes for the working group. I’d also love to hear from the community about why they are interested in longevity: what they see in VitaDAO, what keeps people coming, and how we can collectively build on that vision. Because it is not just the Stewards, it’s everyone here. Which is what makes this so unique!

Thank you for your time today! I’m excited to see a female steward! It is exciting to continue to see more women get involved as members of the VitaDAO community! 

I would love to use this feature as an opportunity to encourage more young women to step up and be a part of the conversation. They are wonderful and I love working with them, but the ratio of men to women in scientific research and web3 is still disproportionate. I see DeSci as a startup in itself. It is an emerging industry, and therefore, the perfect opportunity to pave the way to create new standards. To make a place for ourselves. I want to see more women!

🤝Thanks for Reading! 

Want to get involved in VitaDAO? Have a special skill set? Join an onboarding meeting to learn more about ways to contribute to the growing community! Want to stay up to date with all that's going VitaDAOn? Join us on Discord, subscribe to our Twitter, and follow our Instagram

Additionally, VitaDAO's website (https://www.vitadao.com/) contains updated information on funded projects and a treasury dashboard. 

Read more
Transcending Boundaries: Exploring Cryonics, Drug Repurposing, and the Future of Aging Research with Prof. Joao Pedro De Magalhaes on The Aging Science Podcast by VitaDAO
July 21, 2023
Awareness
Longevity
Transcending Boundaries: Exploring Cryonics, Drug Repurposing, and the Future of Aging Research with Prof. Joao Pedro De Magalhaes on The Aging Science Podcast by VitaDAO

In today’s podcast we talked with Prof. Joao Pedro De Magalhaes (@jpsenescence) about some exciting topics ranging from his current work, to philosophy and science fiction. Among other things we talked about cryonics, transhumanism, the need for a “Sputnik moment” in longevity research, fish oil and rilmenidine for longevity, inflammaging, and cellular reprogramming.

It was a pleasure to talk with Pedro because we both share very similar views about aging, longevity, transhumanism and other topics.

Short CV - Joao Pedro De Magalhaes

Prof de Magalhaes graduated in Microbiology in 1999 from the Escola Superior de Biotecnologia in his hometown of Porto, Portugal, and then obtained a PhD in 2004 from the University of Namur in Belgium. Following a postdoc with genomics pioneer Prof George Church at Harvard Medical School, in 2008 Prof de Magalhaes joined the University of Liverpool and, in 2022, he was recruited to the University of Birmingham where he leads the Genomics of Ageing and Rejuvenation Lab (http://rejuvenomicslab.com/). His lab studies the ageing process and how we can manipulate it to fend off age-related diseases and improve human health. Prof de Magalhaes has authored over 100 publications and given over 100 invited talks, including three TEDx talks. He is also CSO of YouthBio Therapeutics, a US-based biotech company developing rejuvenation gene therapies.

Pedro is the author of the widely read senescence.info https://jp.senescence.info/ webpage and of the @jpsenescence twitter account.

The reputation problem for aging research

“If you tell people that you're trying to discover a drug for Alzheimer's, everybody loves it. If you say you're discovering a drug for cancer, everybody loves it. If you say you're discovering a drug for aging, people ask why? I mean, why you want to do that? ... it generates a lot of questions and even opposition, not by everyone, but for some people.” (Joao Pedro De Magalhaes)

Many people think that lifespan extension is science fiction. However, even that is an understatement of the problem because lifespan extension is rarely portrayed at all or positively portrayed even within sci-fi literature. Pedro and I discussed possible reasons for this. One idea is that the finality of death is so ingrained and terrifying that people refuse to think about anything concerning death as a coping mechanism. This will include even positive things like lifespan extension. It is absurd to consider a future where interstellar travel is possible but not radical lifespan extension. Yet this is what is often portrayed in science fiction!

We spent the first half of the podcast digging into the psychology and philosophy of aging research and trying to figure out what will make people believe that aging research is necessary; and that we will be able to slow aging in the not-too-distant future. Even if you are not interested in aging research I think the first part is worth a listen. One of our key insights was that people who are afraid of living longer, and who are afraid of technology, now, will most likely change their opinion once the technology matures. The great majority will be keen to use longevity therapies and only a fringe minority will continue to oppose the technology:

“So, I mean, if you ask people, hey, if I have a little pill that will make you live 200 years in good health, if you ask around, there's a lot of people going to say, no, I don't want that. But trust me, if that were introduced, and people started taking that pill and being healthier, everybody would want it. That's just, you know, inevitable.(Joao Pedro De Magalhaes)

Transhumanism and radical lifespan extension communities

Transhumanism describes a philosophy that contends we should transcend or improve human nature using technology. The idea is that human biology is flawed and that life could be much improved if we embraced technology to improve ourselves. Such technology includes but is not limited to things like artificial intelligence, lifespan extending treatments or cryonics.

“Radical” lifespan extension tries to differentiate itself from the modest goals of regular biogerontology. Advocates for radical lifespan extension believe that we need to extend human lifespan significantly, by decades or centuries, and that we may be on the cusp of success. In the podcast we discuss that while we might not be that close to this goal, the idea of radical lifespan extension itself is worthwhile. There is no reason why we cannot have ambitious dreams.

We also discussed the recent history of the online longevity movement. Many communities, back when bulletin boards, forums and mailing lists were popular, catered to transhumanists and life extensionists of the early days, including the now renamed “Immortality Institute”.

We can consider Vitadao as a continuation and extension of these old communities. It is attractive not only to transhumanists, but also crypto enthusiasts, web3 developers, investors, scientists, the general public etc. If you want to be part of this community, here is the link to their discord:
https://discord.com/invite/vitadao

Cryonics

The idea of cryopreservation is to freeze one’s body right after death, for a non-zero chance of revival in the future. The hope is that future technology might be able to reverse the damage that led to death and also reverse the damage induced by cryopreservation. Although this is a tall order the likelihood is probably not zero, even if it is very low. In that way cryonics is a modern, transhumanist Pascal’s wager.

In the meantime, while we research cryobiology with the long term goal of preserving whole organisms, every little advance we make can be used to improve the storage of donated organs, hence benefiting humanity now and not just in the future.

This is the same idea as with radical lifespan extension. One can have an ambition long term goal that is reached through reasonable intermediate steps. 

Repurposing of drugs

Even though many drugs are approved for one condition they could also work for another. However, in many cases no one has tested this yet. In the process of repurposing, also called repositioning, scientists are trying to find new uses for such old drugs. If we want to cast our net wider, we can also consider drugs that passed phase II or phase III trials but were not pursued for whatever reasons. These drugs will also have a decent amount of safety data in humans and will be backed by good efficacy data.

One strategy Pedro uses to find drugs that could be repositioned is based on gene expression signatures. The idea is to find drugs that show similar gene expression to that of caloric restriction. This strategy allowed him to pinpoint allantoin as a potentially life extending drug and later rilmenidine, an approved, although rarely used, anti-hypertensive drug, that might operate via a similar mechanism (Calvert et al. 2016, Bennett et al. 2023).

Immunology of aging, inflammaging and the power of meta-analysis

In his meta-analysis of transcriptomic data Pedro found that in mammals many genes change with age (Palmers et al. 2021). Some highlights include decreased expression of mitochondrial genes, perhaps consistent with decreased mitochondrial biogenesis and function, but also increased expression of pro-inflammatory factors.

“The preponderance of inflammatory and stress response genes [over-expressed with aging] is reminiscent of the inflammageing hypothesis [14], which argues that ageing is caused by steadily failing responses to stress, in particular responses to the increased antigenic load that comes with age.”

In this context we also discussed new single sequencing data, suggesting age-related infiltration by immune cells and Pedro’s work in a new company focusing on reprogramming. One of the ideas that we discussed was that immune cells are generally a good target for rejuvenation because they are accessible and affect many processes in the body.

The Interventions Testing Program (ITP)

The NIA’s Interventions Testing Program is a large mouse study that seeks to rigorously test which compounds extend mouse lifespan. Anyone can propose a compound to be tested and during the podcast I suggested to Pedro he could submit rilmenidine to the ITP. If you want to learn more about this important study, we had a thorough discussion of the ITP in my podcast with Rich Miller.

https://www.vitadao.com/blog-article/the-mouse-longevity-chronicles-unraveling-aging-with-dr-richard-miller-during-the-aging-science-podcast-by-vitadao

Further reading – aging as a software design flaw

A controversial take we discussed during the podcast was Pedro’s idea that aging has programmatic features. I will not go too much into details here and the interested reader can check out his paper (de Magalhães 2023) and the below tweetorial, as well as our podcast discussion.

References

Bennett, Dominic F., et al. "Rilmenidine extends lifespan and healthspan in Caenorhabditis elegans via a nischarin I1‐imidazoline receptor." Aging Cell 22.2 (2023): e13774.

Calvert, Shaun, et al. "A network pharmacology approach reveals new candidate caloric restriction mimetics in C. elegans." Aging cell 15.2 (2016): 256-266.

de Magalhães, João Pedro. "Ageing as a software design flaw." Genome Biology 24.1 (2023): 51.

The new webpage of the Immortality Institute: https://www.longecity.org/forum/

Pedro’s linkedin: https://www.linkedin.com/in/joaopedrodemagalhaes/

We talked with Prof. Joao Pedro De Magalhaes about about cryonics, transhumanism, the need for a “Sputnik moment” in longevity research, fish oil and rilmenidine for longevity, inflammaging, and cellular reprogramming.
Read more
June Longevity Research Newsletter
July 10, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
June Longevity Research Newsletter

Introduction

Welcome to this month's newsletter, where we delve into the philosophical dichotomy at the heart of longevity research. 

Two broad perspectives pervade the field. Firstly, we have the 'death defiers', individuals who perceive mortality as a malady, akin to a disease that can be vanquished. These aspirants advocate for a significant postponement, if not complete eradication, of death. They envisage a future where humanity capitalizes on biotechnology, genetics, and regenerative medicine advancements to conquer the final frontier - death itself. They approach aging, particularly from age-related causes, as an adversary to be defeated.

On the other end of the spectrum, we find the 'quality adherents'. These individuals think that lifespan is finite, but champion improving healthspan as a tangible goal, believing in 'healthy aging'. This focuses on extending the phase of our lives free from chronic diseases, where we maintain physical and mental fitness.

In this edition, we put the spotlight on Prof. Joao Pedro de Magalhaes, a distinguished researcher and a keen advocate of lifespan extension. We'll share insights from our recent interview, where he shares his perspectives on his work and the motivation behind it. Also, we explore the intriguing discourse ignited by a Tweet from Balaji, which dared to voice support for the death defier's view, sparking a vibrant discussion on Twitter.

VitaDAO-funded Research Projects

The VITA-FAST Token Sale concluded with a 1700% oversubscription!!!

VITA-FAST token holders have exclusive control over the licensing function of Newcastle-Korolchuk IP and governance over the direction of the research and development of that IP.

Congratulations to the ExcepGen Inc team for receiving a 80.66% majority in the tokenholder vote to secure VitaDAO funding and support for their project aiming to optimise RNA Delivery for Longevity Therapeutics

Longevity Literature Hot Picks

Preprint Corner

Check out this month’s longevity preprints - will any receive a coveted spot in the Q2 Issue of the Longevist? Watch this space!

Premature Aging and Reduced Cancer Incidence Associated with Near-Complete Body-Wide Myc Inactivation

Measuring C. elegans Ageing Through Non-Invasive Monitoring of Movement across Large Populations

The blood has something to say: A hematology-based clock to measure aging in mice

Cognitive Rejuvenation In Old Rats By Hippocampal Oskm Gene Therapy

Developmental mitochondrial Complex I activity determines lifespan

The Role of Respiratory Complex IV in Lifespan Length and Quality

The molecular clock in long-lived tropical trees is independent of growth rate

Uncoupling of lifespan and reproductive tradeoffs by dietary methionine and one-carbon metabolism

Measuring C. elegans Ageing Through Non-Invasive Monitoring of Movement across Large Populations

Published Research Papers

Distinct longevity mechanisms across and within species and their association with aging

The study analyzed RNA from 41 mammals to decipher longevity mechanisms. Findings include common and unique gene markers related to lifespan, revealing a connection between long-lived species, ancient genes, and age-related changes. 

Human PBMC scRNA-seq–based aging clocks reveal ribosome to inflammation balance as a single-cell aging hallmark and super longevity

A new aging clock model, using blood single-cell RNA sequencing data, suggests supercentenarians (SCs) have a biological age between 80.43 and 102.67 years. SCs have more cells and types with high ribosome levels, potentially contributing to their low inflammation state and slow aging.

Clearance of senescent macrophages ameliorates tumorigenesis in KRAS-driven lung cancer

The study identifies senescent macrophages and endothelial cells as primary in KRAS-driven lung tumors in mice. Senolytic removal or macrophage depletion reduces tumor load and boosts survival. It underlines the crucial role of senescent macrophages in lung cancer progression, suggesting potential therapeutic strategies.

Targeting lymphoid-derived IL-17 signaling to delay skin aging

Aging skin sees increased inflammation due to a shift towards IL-17-expressing immune cells, according to single-cell RNA sequencing of mouse skin. Blocking IL-17 signaling in vivo reduces skin inflammation and delays age-related skin changes. 

Resistance training rejuvenates aging skin by reducing circulating inflammatory factors and enhancing dermal extracellular matrices
Both aerobic training (AT) and resistance training (RT) improved skin elasticity and structure in middle-aged Japanese women. RT uniquely increased dermal thickness and biglycan levels, suggesting different effects of AT and RT on skin aging.

The blood has something to say: A hematology-based clock to measure aging in mice

A biological age clock in mice was developed using hematological markers from longitudinal studies. The deep learning-based biological age prediction correlated well with actual age and aging acceleration linked with lifespan. 

Restoration of CPEB4 prevents muscle stem cell senescence during aging

Aging affects mitochondrial function in muscle stem cells (MuSCs), leading to senescence. The study found downregulated CPEB4, which supports mitochondria, in aged tissues. Restoring CPEB4 improved MuSC function and prevented senescence.

Comparative analysis of calcified soft tissues revealed shared deregulated pathways

This study aimed to understand the mechanisms and tissue-specific changes associated with pathological calcification in various soft tissues. The authors found that down-regulation of intracellular transport pathways and up-regulation of inflammatory pathways were common features. 

Impact of social isolation on grey matter structure and cognitive functions: A population-based longitudinal neuroimaging study

This study examined the impact of social isolation on brain health and cognitive decline in a population-based longitudinal MRI study. The findings revealed that social isolation was associated with smaller hippocampal volumes, reduced cortical thickness, and poorer cognitive function, suggesting that social isolation contributes to brain atrophy and cognitive decline. The results highlight the potential of promoting social networks as a means to reduce the risk of dementia.

Published Literature Reviews

Aging - What it is and how to measure it

This review argues for the need to focus on general biological aging rather than lifespan, which can be influenced by specific pathologies. It suggests a framework that accounts for phenotypic changes over an average lifespan, applicable across multiple organisms.

The meaning of adaptation in aging: insights from cellular senescence, epigenetic clocks and stem cell alterations

The study suggests features like cellular senescence, epigenetic aging, and stem cell changes may be adaptations, not aging drivers. It proposes 'damaging adaptations' may accelerate aging despite short-term benefits. This insight may impact antiaging interventions.

Taurine linked with healthy aging

Taurine, a semi-essential amino acid, plays a key role in animal development and health. It is linked to benefits in human metabolic and inflammatory diseases, and recent studies indicate it aids health in aged animal models.

Chronic inflammation and the hallmarks of aging

The updated aging hallmarks include dysbiosis, impaired macroautophagy, and "inflammaging". These factors interact, affecting age-related diseases like cardiovascular issues, neurodegeneration, and cancer. Understanding this is key in aging research.

Are we measuring what matters to older people?

Comprehensive geriatric assessment is key in elderly care, emphasizing personalized biopsychosocial evaluations. However, health systems often neglect person-centered outcomes, risking suboptimal care. Prioritizing meaningful outcomes for older individuals is vital.

And finally - a special FEBs issue of reviews and research articles on Senescence in Ageing and Disease

Job board

Van Heron Labs is hiring for two positions very shortly - a senior scientist and an RA/Lab tech. Please reach out directly for more info: rvaught@vanheronlabs.com

Clinical Research Assistant position at the Healthy Longevity Translational Research Program is open. Apply to Yong Loo Lin School of Medicine at NUS to work with Andrea Maier’s team.

Oliver Hahn’s lab at Calico is hiring scientists & senior scientists to develop novel and exciting experimental platforms to understand and treat age-related diseases of the CNS

Master Fellowship Programme

Do you hold a BSc degree in life science and are eager to explore the field of ageing research in a cutting-edge and interdisciplinary environment of the Cologne ageing cluster? 

The Cologne Graduate School of Ageing Research offers a new Master Fellowship programme for excellent and motivated students that wish to learn more about ageing research.

Are you interested in studying brain ageing?

Oliver Hahn’s lab at Calico is hiring scientists & senior scientists to develop novel and exciting experimental platforms to understand and treat age-related diseases of the CNS. Apply here.

News

Healthy Longevity Talent Incubator is open for applications!

When: July 3-13, 2023

Where: Singapore

Who: Talent (from all disciplines) eager to learn more about Healthy Longevity!

Application form

Dr. David Sinclair Assumes Presidency of the Academy for Health and Lifespan Research

Police got called to an overcrowded presentation on “rejuvenation” technology

Chris Hemsworth’s Center Partners with Lifespan.io, to Make Health, Wellness and Longevity Education More Accessible for All

Aging Fly Cell Atlas identifies exhaustive aging features at cellular resolution

Nature Aging enters the Journal Citation Reports 2023 - ranking #1 in Geriatrics & Gerontology with a first Impact Factor over 16 and a JCI of 3!

Prizes

Rewarding the most impactful research in longevity

Substantially increasing the human lifespan. The Amaranth Prize gives no-strings attached funding to the best research in Longevity.

Articles

Last month’s article proposing Taurine as a driver of aging has sparked a lot of discussion around its relevance for human healthspan:

Taurine supplement makes animals live longer — what it means for people is unclear

In the Pipeline | Aging and Lifespan: Taurine

Taurine improves the health and longevity of mice and monkeys – but what about humans?

Should I worry about my VO2 max?

Meet Rapamycin, the Drug That Could Delay Menopause

Time-restricted feeding makes the mouse run like a pro 

Defining a longevity biotechnology company

Transformer-based aging clock provides insights into aging

Navigate the emerging field of longevity

How seeing corpses reduces the lifespan of flies

Fasting Mimetics: Health And Longevity Benefits With Dr. Valter Longo

Cutting Calories To Add Years Finally, We Have Evidence That Caloric Restriction May Increase Human Lifespan

The billion-dollar search for immortality

Finding joy at age 100: Talking to centenarians about living their best life at any age

Conferences, Workshops and Webinars

World Aging and Rejuvenation Conference (ARC)

17-18 July, Frankfurt, Germany

Ending Age-Related Diseases

10-11 August, NY, USA and virtual

Longevity Summit Dublin

17-20 August, Dublin Ireland

Aging Research & Drug Discovery (ARDD)

28 August - 1 September, Copenhagen, Denmark

British Society for Research on Aging Annual Scientific Meeting (BSRA ASM)

6-8 September, London, UK

RAADFest

7-9 September, LA, CA, USA

Zuzalu talks recordings are out!

https://zuzalu.streameth.org/archive

Tweet of the Month

This month we leave you with some food for thought from a leader in the crypto world, Balaji Srinivasan

Balaji: CHEATING DEATH IS GOOD

People often associate performance enhancing drugs with “cheating”. And of course it’s true that if you’re an athlete it’s cheating and against the rules. But there’s nothing wrong with cheating death.

[Full text]

Also check out this interesting thread from Healthspan on Blagosklonny's theory of cellular hyperfunction

Podcasts and Videos

Wired: Have a Nice Future: Gideon Lichfield and Lauren Goode talk to Celine Halioua, the founder and CEO of Loyal—a company that researches drugs to extend the lifespan of dogs.

Brain Boosting Effects of Young Blood Revealed

Lifespan news talk about the most problematic yet perhaps one of the most promising cures for aging, young blood. Heterochronic parabiosis is a well-known aging intervention in mouse studies, but it happens to be one of the more, shall we say, unrealistic procedures for humans.

Interview with Prof Joao Pedro de Magalhaes

Joao Pedro de Magalhaes is a world renowned leader in aging research using computational and experimental approaches to untangle the secrets of human aging. He is the Chair of Molecular Biogerontology at  the University of Birmingham, and currently leads the Genomics of Aging and Rejuvenation Lab there.

What inspired you to enter longevity research?

I have never made it a secret that I work on aging and longevity to cheat death. When I was a child and realised that my parents and everyone I love would age and die, I made it my mission to develop a cure for aging.

Which of the current theories of ageing do you think are the most convincing?

I’m very interested in the idea that programmatic features originating in development contribute to aging. I think it’s a very overlooked area, as most scientists assume that aging is caused by molecular and cellular damage. However, I think some genetic programs originating in development continue later in life and become detrimental. That said, it’s clear that some forms of damage contribute to certain aging phenotypes, like for example mutations contribute to cancer. I also think it’s important we keep an open mind and explore different avenues, because at the moment we don’t know what are the drivers of human aging.

How has the field changed since you started?

In some ways, the field has changed substantially in the past 20 years because there is a lot more focus on translational research, many more companies, more investors; there has been a big growth in the longevity field. This growth has been catalysed by the discovery of longevity manipulations in animal models, and the huge medical and commercial potential of applying those to humans. In that sense, there has been a huge growth in longevity pharmacology and in longevity biotech in general. That said, at the mechanistic level things have not progressed so much, we still have a very poor understanding of what causes human aging.

What mistakes do you think the longevity field has made?

I think a common mistake in science is being overly conservative. This issue is not limited to the longevity industry; you can observe it in other fields as well, like Alzheimer‘s. Scientists, investors and funding bodies are often conservative. What this means is that scientist and companies tend to focus on the same mechanisms, targets and pathways, as I pointed out in a recent article (https://www.sciencedirect.com/science/article/pii/S1359644621000982?via%3Dihub). I think we need to be more creative in the field, and in fact, I would argue that the creativity is there, it’s just not well supported because most people and institutions are risk-averse. We need to be bolder.

Other than your own, what do you think have been the biggest/important discoveries in the field?

There have been some important discoveries in the past 10–20 years. One important discovery was that rapamycin extends mouse lifespan even in middle-aged animals. It showed we can significantly extend lifespan in animal models with pharmacological interventions. The discovery of epigenetic clocks by Horvath and others has also been an important breakthrough, even if we don’t understand the underlying biological mechanisms very well. In addition, the discovery of cell rejuvenation with partial reprogramming has been an important discovery, although we still need to establish whether it can be applied to retard aging in whole organisms.

What advice would you give to people currently working in longevity research?

Be bold, be creative, don’t be afraid to take risks and to create an environment that allows others to take risks. Aim high, don’t settle for the low-hanging fruit.

Which aspect of longevity research do you think requires more attention?

As I mentioned above, there has been a shift in the field from understanding ageing to developing interventions. I would argue that going back to basics and trying to understand the underlying drivers of human aging is still the big question, and if we could better understand the causes of human aging this would be a watershed moment and open the door to much better therapies. Of course, trying to understand the aging process is hard; it’s a lot easier to give drugs to worms or flies and see if they live longer than trying to figure out why human being age. Studying aging processes also requires a sustained long-term investment that most funders and companies are unwilling to provide.

Is ageing a disease?

That’s a good question, but I suppose it depends on the definitions of aging and of disease. As such, I think it’s more of a semantics question than a biological one. Regardless of the definitions, aging is a trigger for diseases, and it should be targeted therapeutically.

In your recent paper you compare aging to a software design flaw. Can you briefly summarise this theory and discuss how it has influenced your approach to studying aging and potential impact on the development of interventions to delay or reverse age-related decline?

The hypothesis is that ageing is not just driven by damage to the hardware, defined as cells and their components, but rather by design flaws in the software, defined as the DNA code that orchestrates how a single cell becomes an adult human being composed of billions of cells with many different identities. My view is that some processes set in motion by the genetic software during development continue in adulthood and become detrimental as a form of antagonistic pleiotropy. If aging is an unintended outcome of a program this explains the accuracy of epigenetic clocks, it explains species differences in aging as developmental rates correlate very strongly with aging rates in mammals, and fits the major genetic, dietary, and pharmacological manipulations of aging in animals. As a result, we should see aging is an information problem.

Seeing aging as the outcome of flaws in our software has important implications for studying and developing interventions for aging. Traditional anti-aging interventions targeting damage, like oxidative damage and telomere shortening, will – I predict – have limited success. By contrast, aging therapies will only be effective if targeting the software rather than the hardware. As such, interventions like a computer restart such as partial reprogramming could hold clues for future interventions.

Your research involves using computational biology and machine learning to better understand the molecular mechanisms of aging. Can you discuss the most promising applications of these techniques in the field of aging research and how they can accelerate academic research?

The problem we have in the field, and others, studying complex diseases and phenotypes, is that aging and longevity derive from the interactions of multiple genes with each other and the environment. In that regard, we need to take a systems biology approach to study how the components of the system interact with each other and ultimately understand the whole from its parts. So, in a way, we are trying to make sense, trying to tackle the complexity of biology. Using these computational methods, we can identify and prioritise new genes, drugs and pathways. For example, we have done some recent work on drug repositioning in the context of aging, revealing new existing drugs that, in animals, extend longevity and may ultimately have human applications (https://onlinelibrary.wiley.com/doi/10.1111/acel.13774). Also for a recent talk on computational approaches and aging please see:
https://www.youtube.com/watch?v=VhaAGwh012k

Your research has also covered comparative biology of aging, looking at how different species age and what factors contribute to differences in longevity. What have been some of the most surprising or interesting findings from this work?

I have always been fascinated by species differences in aging. I’ve always liked animals and used to have many different pets as a child. It is fascinating how no matter how well you care for a hamster, or a mouse or rat for that matter, they’ll still age 20 or 30 times faster than a human being. However, just like we don’t understand the mechanisms of aging, we have a very poor understanding of species differences in aging. In our work, we have tried to find genes and pathways associated with the evolution of longevity, for example studying the bowhead whale which is the longest-lived mammals. But of course we cannot do experiments in whales. My dream is to take some genes from the bowhead whale that we have found and put them in mice and see if they live longer or if they are cancer resistant, but we unfortunately don’t have the funding to do it. We have also done studies in the naked mole rat and in primates, and others have studied other species like bats. But we have really only scratched the surface as far as mechanisms of cancer resistance and longevity across species. My rationale is that different long-lived species use different molecular tricks and if we can figure out the longevity mechanisms of other taxa maybe we can apply them to human beings.

Your new pre-print paper proposes a conceptual model for explaining aging based on first principles, suggesting that epigenetic aging might be inevitable. Can you discuss how this model is different from existing models of aging, and what are the key implications of this conclusion for our understanding of the aging process?

This was a different approach for me as well, as you say, based on first principles, and credit to a brilliant PhD student, Thomas Duffield, for leading this work. Ultimately, this model assumes that epigenetic changes drive aging. This is based on the observation that epigenetic damage cannot be repaired in the same way as DNA damage, and we show that the epigenetic system has a built-in, unavoidable fidelity limitation. Based on this theoretical model, we looked into epigenetic data, namely methylation, and surprisingly found that there is an increase in noise with age and, strikingly, we can use neural networks to build accurate epigenetic clocks based on noise. I think this is very surprising, and I happily admit I’m still trying to get my head around what it means for understanding aging.

Outro

Thanks for reading and sticking with us for another month. We leave you with a short but insightful article on the potential of tokenizing IP-NFTs, where you can find more detail on the recent VITA-FAST launch!

VITA-FAST — Revolutionizing Governance in Longevity Research

Further Reading

Association between infectious exposures in infancy and epigenetic age acceleration in young adulthood in metropolitan Cebu, Philippines

Microvesicle-Mediated Tissue Regeneration Mitigates the Effects of Cellular Ageing

Time-resolved proteomic analyses of senescence highlight metabolic rewiring of mitochondria

Does omega-3 supplementation improve the inflammatory profile of patients with heart failure? a systematic review and meta-analysis

Curcumin, Inflammation, and Neurological disorders: How are they linked?

Epigenetic and Metabolomic Biomarkers for Biological Age: A Comparative Analysis of Mortality and Frailty Risk

Drugs Targeting Mechanisms of Aging to Delay Age-Related Disease and Promote Healthspan: Proceedings of a National Institute on Aging Workshop

To keep up-to-date with the latest breakthroughs in longevity research, exclusive insights, and exciting updates from our team, make sure to subscribe to our newsletter.

Welcome to this month's newsletter, where we delve into the philosophical dichotomy at the heart of longevity research.
Read more
Feeding the Clock: Deciphering the Intricacies of Fasting, Caloric Restriction, and Circadian Rhythms with Dr. Heidi Pak on The Aging Science Podcast by VitaDAO
July 6, 2023
Awareness
Longevity
Feeding the Clock: Deciphering the Intricacies of Fasting, Caloric Restriction, and Circadian Rhythms with Dr. Heidi Pak on The Aging Science Podcast by VitaDAO

In this podcast with Dr. Heidi Pak (@pak_heidi) we primarily talked about the difference between fasting and time-restricted feeding, how and why it works and also the efficacy of caloric restriction (CR) in different mouse strains and species. It was a great podcast, very fun as well.

Other topics that we touched on included sleep, the postdoc shortage, hardships of being a grad student, the healthspan vs lifespan debate and some more. We also came up with some less-than-serious nutritional recommendations:

"I guess the diluted [fasting] diet [I studied] is the equivalent of eating celery" (Heidi Pak)

If you want to know why we were talking about celery and why Heidi wants to put pigs on a weight loss and fasting diet, then this podcast is for you.

Heidi Pak – short bio

Heidi did her PhD with Dudley Lamming at the University of Wisconsin–Madison where she studied fasting, time-restricted feeding and caloric restriction in relation to aging. She now works as a postdoc for Joseph Takahashi, continuing her work on fasting and moving into the relatively new field of circadian rhythms.

What is caloric restriction?

Also known as dietary restriction is the practice of restricting the caloric intake of mice, usually by 30%. This extends the lifespan of many, although not all, mouse strains considerably. CR is hailed as one of the biggest breakthroughs in biogerontology and for good reason. If it worked in humans; or if we could find a drug that mimics CR in humans it would be the biggest biomedical revolution since the invention of antibiotics!

The discovery of CR is often attributed to seminal work by Clive Mccay in the 1930s. However, what made CR big was not the work by McCay showing that food restriction from birth stunts growth and extends lifespan, rather it was protocol improvements. For example, Walford and Weindruch found in the 1980s that adult-onset CR can work reliably in many strains if it is introduced gradually.

This podcast episode is about another such protocol improvement. The disambiguation of fasting and caloric intake through modern feeding protocols and enabled by 24/7 monitoring of food intake, locomotion etc.

Using those new feeding protocols Heidi and others found that fasting contributes to the lifespan and health benefits of CR.

What is fasting and time restricted feeding?

The idea behind fasting to slow aging was that after the body has digested most of the food, when hunger starts setting in, an organism might be in something akin to a CR state. Therefore, imposing periods of fasting could reap some of the health benefits of CR without the drastic weight loss, constant hunger and other side-effects normally associated with CR. While plausible this theory had many problems, not least the fact that it seemed to contradict the work of Walford and Weindruch who suggested that gradual onset CR was more beneficial in adult mice than starting the protocol abruptly. It stands to reason that alternating periods of fasting and feasting could be stressful to the body, just like the abrupt CR protocols used in the past, and thereby shorten your life (or that of a mouse).

We needed evidence to settle this debate, or at least start addressing some of the major points. Before we can understand the evidence, let us take another look at what fasting exactly is.

Defining fasting is not easy since the cutoff when “fasting” starts is somewhat arbitrary and that can be a problem when reading papers and articles on this topic. When reading about fasting research it is important to keep in mind that a 16hr fast and a one-week water fast could be reported as the same thing by the media, even though the physiologic effects will be very different. Importantly, it could well turn out that certain eating windows are much more beneficial than others. Maybe the water fast is healthier, or maybe it is not. We do not know what will work in humans yet.

Intermittent fasting and time-restricted feeding mean similar things in mice. Although generally fasting refers to on-off feeding schedules and restricted feeding refers to shorter and variable eating windows within a day.

Little progress was made on this matter and some people, like me, remained convinced that fasting has no benefits or that we “simply do not know”. Early studies using intermittent fasting protocols were always biased by weight loss. Since the fasted mice lost weight, even if it was less than during CR proper, it was hard to attribute the health benefits to fasting rather than weight loss. There were also studies suggesting that fasting is not an important component of CR (Nelson and Halberg 1986).

However, one observation pointed out in the work by Heidi Pak always made a strong case for the importance of fasting. While, yes, intermittent fasting induces CR; CR actually induces a form of intermittent fasting! Apparently, the CR data itself was always potentially biased by the effects of fasting.

TimelineDescription automatically generated

From Mitchell 2019, eating windows indicated for caloric restriction (CR), control (AL) and meal fed mice (MF).

As you can see from this image taken from a typical fasting study, mice tend to self-impose a fasting window when they are under CR. They are so hungry that they gorge on their food early during their waking hours and then fast for many hours afterwards.

Building on this finding Heidi Pak and many others designed protocols to test this further.

How do we test the effects of fasting?

Several protocols exist to induce fasting without weight loss. One is caloric dilution, the other is time-restricted dispensation of food using a machine, or training mice to eat during a specific time window. All of them have advantages and disadvantages.

Caloric dilution involves diluting the diet with cellulose that cannot be digested and is one of the approaches that Heidi used in her research. While a very smart idea, I am not the biggest fast of the caloric dilution approach as I have written on my blog when I was first reviewing Heidi’s paper.
See here for more information why I think this way: http://biogerontolgy.blogspot.com/2021/12/caloric-restriction-and-fasting-does-it.html

Be that as it may, I do not want to be overly critical of the research here. As scientists we tend to be overly skeptical of new research. Fact is, modern fasting research was a big breakthrough in the field of CR because it provided conclusive evidence that fasting contributes at least to some of the health and longevity benefits of CR as we discuss in the podcast. This is very exciting because fasting is much more tolerable than CR in humans, thus could see actual application in the real world.

Another strategy to study fasting is removing the fasting window that CR mice self-impose to isolate the effect of calories. This means forcing CR mice to eat multiple times throughout the day. Such studies generally find that fasting is one but not the sole contributor to the health benefits of CR.

This is where it gets complicated – circadian timing

Recent work has shown that not only is fasting one component of CR that contributes to the lifespan extension, there is also a component of circadian alignment. The recently published work by Acosta-Rodríguez et al. 2022 from the Joseph Takahashi lab demonstrated this nicely. This paper has some of the most beautiful figures I have ever seen so highly recommended to check it out.
Here is a very good video explanation of the paper: https://www.youtube.com/watch?v=q5ZN_mT-xbw&t=12s

During the podcast we briefly talked about this work and I mistakenly mentioned that mice in that paper were fasted during the night. In fact it is quite a bit more complicated than that. Night is the active period for mice and it is generally beneficial for them to eat during this period. The above paper actually tested two groups fed at night. One with a 12 hr feeding window and one with a 2 hr feeding window during the day or during the night period. The latter was the most beneficial for lifespan, meaning the mice will be fed at the beginning of the dark period, will quickly finish their food and then will fast for most of the night and the day.

Why humans are not two-legged mice

Even though mice are my favorite animal model and I consider myself a mouse researcher, I have to admit that mice have their limitations as a model of aging. Let us first talk about the benefits of mice, though. Running mouse studies is orders of magnitude cheaper than running human studies, it is more ethical, the mouse is well-understood as to its genetics and longevity, it is amenable to transgenic manipulation and so on. In contrast to nematode worms and fruit flies, the other two popular aging models, mice are closer relatives of humans; they are real, breathing mammals with many of the same traits as humans. They get cancer, they like to cuddle with other mice and sometimes fight with them as well.

The fact that mice are so similar to humans, by the way, is also a reason why we need to treat them well and keep them healthy. Only data from healthy mice will translate to healthy humans. In the podcast we briefly discussed how important it is to keep mice healthy, which is reflected by long lifespans of the control group.

What is the problem with mice then? Well, they are unusually “fragile”, as we have alluded to in our podcast with Peter Fedichev. Meaning, they are unusually short-lived for their size, and unusually prone to cancer, whereas humans are exceptionally long-lived given their body mass and size.

Another limitation is the small size and high metabolic rate of mice. This precludes many surgical interventions and has led to a lot of spurious findings. In this context I like to tell the story of hydrogen sulfide induced suspended animation, which would be also very interesting to aging researchers, if it worked. However, as it turned out this was an intervention that only worked due the physics of small body sizes (Asfar et al. 2014). This is where other animal models come in handy to validate the mouse data. We were talking about pigs as one option during the podcast.

It is difficult to overstate the differences in metabolic rate between mice and humans. A mouse heart contracts at an unbelievable rate of 500 to 700 beats per minute and their energy needs are commensurate with their fast metabolism. Although a mouse may not eat much in absolute terms, per gram of body weight it eats much more than a human, consuming a staggering 10% of bodyweight every day. Imagine eating 8kg of food every day! If you starve a mouse for a couple of days, it will die. Humans in contrast can go on for months without food.

Nevertheless, we can learn a lot from mice. We learned that fasting can work to extend health and lifespan, in principle. Nevertheless mice cannot tell us whether fasting will work in humans.

Perhaps there is an eating window in humans that can mimic mouse time restricted feeding? No one knows whether this will have to involve a week of severe fasting, maybe 2 weeks of moderate fasting or whether the currently used protocols in clinical practice employing moderate fasts are enough. 

Hopefully future research by Heidi and others will help us to understand this issue.

References and notes

Acosta-Rodríguez, Victoria, et al. "Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice." Science 376.6598 (2022): 1192-1202.

Nelson, W. & Halberg, F. Meal-timing, circadian rhythms and life span of mice. J. Nutr. 116, 2244–2253 (1986).

Asfar, Pierre, Enrico Calzia, and Peter Radermacher. "Is pharmacological, H2S-induced'suspended animation'feasible in the ICU?." Critical Care 18.2 (2014): 1-8.

Mitchell, Sarah J., et al. "Daily fasting improves health and survival in male mice independent of diet composition and calories." Cell Metabolism 29.1 (2019): 221-228.

In this podcast with Dr. Heidi Pak we primarily talked about the difference between fasting and time-restricted feeding, how and why it works and also the efficacy of caloric restriction (CR) in different mouse strains and species.
Read more
VITA-FAST — Revolutionizing Governance in Longevity Research
June 30, 2023
Awareness
Tokenomics
VITA-FAST — Revolutionizing Governance in Longevity Research

VitaDAO, the global community accelerating research and development in longevity science, is pleased to announce the launch of the VITA-FAST tokens. This marks the first-ever opportunity for token holders to directly influence longevity research, setting a new precedent in the field of decentralized science (DeSci).

The VITA-FAST tokens, which allow holders to make decisions on IP licensing, set experiment priorities, and govern research on autophagy, have been met with overwhelming interest.

The initial token sale was 1700% oversubscribed, with $620k in bids, demonstrating the public’s eagerness to participate in scientific research.

This groundbreaking initiative is spearheaded by the esteemed Korolchuk Lab, led by renowned cellular biologist Prof. Viktor Korolchuk. The lab’s work focuses on autophagy, a process vital to cellular health and implicated in ageing and various diseases. The launch of VITA- FAST tokens not only provides financial support for this cutting-edge research but also fosters a community-driven approach to biomedical research.

Alex Dobrin, head of community at VitaDAO, said, “This is the next phase in the evolution of decentralized patient communities, to directly contribute to, govern, and be rewarded for progressing valuable longevity research.”

VitaDAO engaged its community in the token sale by using a newly launched crowdsales platform developed by Molecule AG, which developed the original IP-NFT platform for capturing IP in web3, and is a strategic contributor to VitaDAO.

“The success of the VITA-FAST token sale is a testament to VitaDAO’s innovative approach to integrating public participation into scientific research. It also underscores the potential of decentralized finance (DeFi) to fill in the gaps left by traditional funding agencies”, as noted by Paul Kohlhaas, CEO of Molecule.

Professor Matthew Grenby, Pro-Vice-Chancellor for Research and Innovation at Newcastle University added “It’s great to see this cutting-edge research going out into the world, and we are excited to be at the forefront of biomedical research funding. We look forward to supporting Molecule AG and VitaDAO with development of this novel platform and commercialisation of their IP”.

The VITA-FAST token offering marks a significant step forward for VitaDAO as it begins advancing its early research assets toward their next development milestones. The VITA-FAST tokens will provide ongoing options for funding for the Korolchuk Lab as it advances its research program.

For more information, visit https://www.vitadao.com/viktor-korolchuk-lab-vita-fast-token-sale-with-vitadao and talk to us in our Discord auction channel.

‍Discover the power of autophagy, a natural cell rejuvenation process, through the enlightening YouTube video, Autophagy as a Target for Rejuvenation | VitaDAO and Viktor Korolchuk Lab.

This marks the first-ever opportunity for token holders to directly influence longevity research, setting a new precedent in the field of decentralized science (DeSci).
Read more
VitaDAO Letter: Zuzalu & Shaping the Future of Autophagy Research
June 18, 2023
Sarah Friday
Awareness
VitaDAO Letter: Zuzalu & Shaping the Future of Autophagy Research

Welcome back to the VitaDAO Newsletter, your monthly source of insights into the VitaDAO community. In just 20 months, we have successfully deployed over $4 million to fund 17 cutting-edge projects, pushing the boundaries of what is possible in the field.

From our upcoming token sale, which introduces the innovative VITA-FAST tokens, to the one-year anniversary of our renowned “Longevity Research Newsletter, we are pushing the boundaries of what is possible. Let’s dive in and explore the latest developments in VitaDAO’s mission to extend healthy lifespans. Inside this Newsletter:

  • VITA-FAST Tokens: Steering Korolchuk Lab’s Research
  • Celebrate One Year of the Longevity Research Newsletter
  • Recent VitaDAO Votes
  • Meet Rhys Anderson

VITA-FAST Token Crowd Sale

Hold up, I thought that $VITA was the community token of VitaDAO? It is! Then what is the talk of a VITA-FAST Token? 🤔

If you’re new here, welcome! Sit back, relax, and keep on reading as I introduce you to VDP-100,, and the utility of VITA-FAST tokens. This story dates back to 2021 when the VitaDAO community voted to fund Viktor Korolchuk Lab in the discovery of new autophagy inducers. Dysfunctional autophagy affects cellular functions including DNA repair, metabolism, and survival. Thus, activation of autophagy is considered a promising therapeutic approach to combat aging and age-related diseases.

“We see that activating this process makes old cells look young, which is quite striking from our point of view.” — Viktor Korolchuk

Flash forward to earlier this month, the community voted on VDP 100, the fractionalization of the IP-NFT minted pursuant to VDP-16. In layman’s terms, fractionalization is the concept of splitting up ownership of something. Here, the final model is a little different: not split ownership, but rather shared governance. In this case, VitaDAO is distributing governance over the Korolchuk project by allowing individuals to buy or earn governance rights in order to have a more active role.

This brings us to VITA-FAST tokens. $VITA-FAST will be Intellectual Property (IP) governance tokens that provide membership to an IP pool for the IP-NFT. Members of this pool will actively decide what happens with the IP and R&D data attached to the Korolchuk IP-NFT. Thus, VITA-FAST token holders have the right to participate in the governance of the IP-NFT and its development.

This necessitates token distribution! 1,000,000 VITA-FAST tokens will be minted, with 10% available for sale to VitaDAO members, and the rest divided among VitaDAO, researchers, Newcastle University, and a liquidity pool. The 10% will be sold to $VITA token holders in a fixed-price sale with pro rata distribution, overflow refunds, and 2-month vesting. VitaDAO members can bid as much as they’re willing to contribute to the sale, with the final allocation of tokens proportional to each participant’s contribution.

We invite you to join us as we make history! Before participating in the sale, familiarize yourself with the Korolchuk project page, the project white paper, and VitaDAO’s blog announcement.!

Sale duration: June 15 5pm CET — June 23 5pm CET

Participate in the sale here: https://mint.molecule.to/ipnft/2

🧪VitaDAO’s IP-NFT Funded Research Projects

In a newsletter discussing fractionalization, it feels wrong not to briefly cover IP-NFTs. IP-NFTs connect IP and research and development data rights to NFTs on the Ethereum network by attaching legal contracts to smart contracts. They represent a new paradigm in the evolution of legal contracts for scientific research. Benefits of IP-NFTs include: creating efficiencies in IP licensing, creating entirely new IP markets, and enabling decentralized IP ownership, governance, and commercialization.

VitaDAO has multiple IP-NFT-funded projects including (but not limited to):

🌎VitaDAO & Zuzalu

Last month, Zuzalu & VitaDAO took Montenegro by storm. VitaDAO’s “Longevity Zero to One” and “Longevity Biotech Conference” encouraged conversation about network states and the pursuit of extended life. Want to learn more? Check out the full article by MIT Technology Review for an immersive glimpse into this groundbreaking event:
https://www.technologyreview.com/2023/05/31/1073750/new-longevity-state-rhode-island/

🥳A Year of the Longevity Research Newsletter

Has it already been a year? Time flies when you’re having fun! Join us in commemorating the one-year anniversary of our renowned VitaDAO’s “Longevity Research Newsletter.” This fun initiative, led by Rhys Anderson and Maria Marinova, keeps VitaDAO members up-to-date with the most important longevity research activity happening across the globe. The newsletter offers updates about VitaDAO’s funded projects, highlights hot longevity research, and features top scientists in the field. Swing by VitaDAO’s Medium to read this month’s edition and stay informed about the latest breakthroughs.

🗣️VitaDAO in the Wild

Discover VitaDAO’s growing presence in the scientific and technological community:

📣Community Approved: What you voted for!

VDP-97 Passed! This vote distributed 700,000 VITA tokens to 6 of the top community & awareness contributors, with an additional allowance of up to 300,000 VITA for allocation to other core contributors.

VDP-99 Passed! As a result of this vote, VITA will launch on layer2s. Currently, VitaDAO is prioritizing launching on Optimism, Arbitrum, and Polygon with $250k of liquidity in each VITA + ETH.

VDP-100 Passed! In case you skimmed past the earlier part of this newsletter, VitaDAO’s Newcastle IP-NFT will be fractionalized! This vote confirmed the creation of IP-NFT tokens to govern the IP and control the R&D decision-making and capital allocation process.

💪Exercise Your Right to Vote (on VitaDAO’s Governance Forum)

Find all of VitaDAO’s pending proposals on Discourse, VitaDAO’s governance hub for proposals before they are moved on Snapshot. Proposals are open for engagement, voting, questions, and conversation!

Your voice matters, and together, we can shape the trajectory of VitaDAO and contribute to the advancement of decentralized science.

👋Meet Key VitaDAO Contributor: Rhys Anderson

Rhys Anderson is a research scientist and active VitaDAO contributor. In 2016, he received his PhD in Cell Biology from Newcastle University. Currently, his time as a postdoctoral research associate is spent researching senescence and aging at King’s College London. On the side, Rhys is a part of the Longevity Dealflow Working Group, and Community and Awareness Working Group, and has played an integral part in the initiation of the Longevist. Rhys’s dedication and expertise exemplify the collaborative spirit of our community.

How did you first get involved in VitaDAO?

I’m a cell biologist and in late 2021 I received a Longevity Impetus grant to support my research on targeting senescent cells. Shortly after, I received an email from VitaDAO saying they are reaching out to the Impetus awardees. I had a call with Dealflow Steward Laurence to discuss my research, and he also explained how the DAO works and that anyone can get involved — so I did, and the rest is history……

What does your contribution look like?

I started off contributing to deal flow by helping to evaluate funding applications before going on to “shepherd” some of my own. In mid-2022, Maria Marinova and I began co-authoring a monthly Longevity Newsletter, keeping people up-to-date with all the goings on in the longevity world from publications to conferences and interviews with prominent longevity scientists.

Recently I have also been busy working away on VitaDAO’s new initiative, an overlay journal — The Longevist — which will be ready for launch soon!

How does it work for you?

I work full-time doing research, but due to the remote and flexible nature of VitaDAO, I’m able to make contributions on weekends, evenings, and early mornings before my toddler awakes and summons his loyal servant.

You played an integral part in establishing The Longevist. What exactly is The Longevist?

The Longevist is a curation of the world’s most exciting longevity research each quarter. Our editorial team created a shortlist of preprints that we believe have the potential to have a big impact on the field and then our team of expert curators vote on-chain to determine which of these preprints they think are the best.

Why preprints?

Preprints have accelerated the rate at which scientific discoveries are shared with the world. However, so much research is published on a daily basis that it’s impossible to keep up with all the literature and it could be months to years before publication in a traditional journal. So we are using our platform to highlight research that deserves special attention.

What do you feel is VitaDAO’s biggest strength compared to traditional longevity biotechs?

VitaDAO has an extensive network with a real breadth and depth of expertise. With the opt-in nature, no one is lumbered with unwanted assignments from the boss — instead, people can focus on contributing to the projects they are most passionate about. I’ve been extremely impressed by the rate at which ideas can go from inception to reality.

The community is also willing to take risks on projects which would be unlikely to receive conventional funding as well as being able to support the longevity ecosystem in numerous other ways from prizes to fellowships.

Any closing thoughts for readers?

There’s a real buzz around DAOs at the moment — I imagine it’s akin to what the early stages of the dot com boom would have felt like in the 90s.

Looking at the DeSci space alone there are new DAOs popping up all the time and they are proving to be instrumental in bringing both the tech and science worlds together.

The success of VitaDAO has been enabled by decades of longevity research showing that aging isn’t an immutable universal constant but rather a process that can be understood and targeted. The tech world has bought into this and has created the infrastructure and provided funding to support longevity research, which has the potential to lead to a healthier, more prosperous future for all of humanity.

🗓️Upcoming Event

June 15, 5 pm CET — June 23, 5 pm CET: VITA-FAST Token Crowd Sale

We’ve made it possible for you to govern and engage with Viktor Korolchuk Lab’s research as a VitaDAO Member. As you know, we’re all about putting the power in the hands of our community and we’re doing exactly that with the VITA-FAST tokens.
Participate in the Sale: https://mint.molecule.to/ipnft/2

🤝Get Involved

Have a special skill set? Contribute to VitaDAO and receive $VITA in return. Want to stay up to date with the VitaDAO community? Join us on Discord, subscribe to our Twitter, and follow our Instagram!

Additionally, VitaDAO’s updated website (https://www.vitadao.com/) contains information on funded projects and a treasury dashboard.

Welcome back to the VitaDAO Newsletter, your monthly source of insights into the VitaDAO community. In just 20 months, we have successfully deployed over $4 million to fund 17 cutting-edge projects.
Read more
Introducing VITA-FAST: Govern and Engage In Viktor Korolchuk Lab's Research as a VitaDAO Member
June 12, 2023
Awareness
Tokenomics
Introducing VITA-FAST: Govern and Engage In Viktor Korolchuk Lab's Research as a VitaDAO Member

Greetings, Vitalians!


Imagine having a direct hand in the future of longevity research. Today, we’re thrilled to introduce VitaDAO’s innovative approach to scientific research funding, giving you an exceptional chance to participate in the decision-making processes of longevity research. With our community fundraising effort for the “Discovering Novel Autophagy Activators” Intellectual Property Non-Fungible Token (IP-NFT) from the Korolchuk Lab at Newcastle University, you are invited to contribute to the democratization of one of the most cutting edge longevity research and development (R&D) projects funded by VitaDAO.

This fundraising effort will be a sale of VITA-FAST tokens, a.k.a. Molecules, representing governance rights over the pool of Intellectual Property (IP) and R&D generated by the esteemed Korolchuk Lab’s research centered on the discovery of novel autophagy activators–the research funded by VitaDAO’s Korolchuk IP-NFT. Autophagy is a cellular process whose decrease can contribute to our biological systems’ decline, particularly as we age. By stimulating autophagy, we’re exploring promising therapeutic approaches to combat aging and age-related diseases, with the goal of improving human healthspan.

By enabling community-led decision-making and governance, VitaDAO’s novel funding model redefines how research programs, including the Korolchuk Lab, are supported. Participating in this crowdsale means not just funding research, but driving scientific progress, shaping therapeutics’ future, and joining a devoted community of pioneers committed to extending human lifespan.

Your contribution to the VITA-FAST Molecules crowdsale provides you with more than just a chance to fund potential longevity therapeutics; it enables you to actively guide them. As a VITA-FAST tokenholder, your voice counts in making crucial decisions. You can influence how the program’s resources are allocated, ensuring they are put to the most effective use in our quest for groundbreaking research and discoveries. You’ll have the ability to dictate the priority of experiments, ensuring that we concentrate on the areas you believe hold the most promise. This unique capability lets you stay at the cutting edge of scientific exploration, directly impacting the path of life-extending advancements. You can decide who licenses the IP & R&D data and what the license terms are.

Your role as a VITA-FAST tokenholder is not passive. It’s about engagement and contribution, and there’s no one-size-fits-all approach. We value and need your unique skills and perspectives, regardless of your background. Whether you bring scientific knowledge, business development expertise, or other abilities beneficial to early-stage drug development, we welcome your input. We believe in collective intelligence and decentralized science, and we’re eager to see how your involvement will generate value for patients.

The number of available tokens are limited. A total of 1,000,000 VITA-FAST tokens will be minted, with 10% available for sale to VitaDAO members, and the rest divided among VitaDAO, researchers, Newcastle University, and a liquidity pool.


Our innovative Genesis Sale Structure further democratizes the process. This fixed-price sale allows VitaDAO members to bid as much as they’re willing to contribute to the auction, with the final allocation of tokens proportional to each participant’s contribution. VitaDAO members must lock their VITA for two months to participate in the sale. If we meet or exceed our sales target (~32,000 USD), the surplus will be returned to bidders. Importantly, the VITA-FAST tokens will be vested (non-transferrable) over a 60 days to ensure equitable participation in governance during vesting. The vested VITA-FAST tokens and the locked VITA will vest and unlock at the same time, 60 days after the Genesis Sale. For a more detailed explanation, read VDP-100 on Snapshot.

The funds raised from the sale will be directed towards providing liquidity for the VITA-FAST liquidity pool until token holders govern where the resources should be allocated. This could include contracting computational biochemists, engagement of experienced medicinal chemists, or filing patents.

We, therefore, invite you to join us on this revolutionary journey. Engage in this unique opportunity to support the Korolchuk Lab and the wider field of longevity research. Make your mark in a cause aiming to enhance human health and longevity. Every contribution, every Molecule purchased, every decision made, brings us a step closer to deciphering the mysteries of longevity.

While we aim to democratize research funding, we acknowledge that all biomedical research is unpredictable, laden with twists, turns, and complexities. Therefore, we ask those considering purchasing VITA-FAST tokens to do so with a comprehensive understanding of these inherent uncertainties.

First and foremost, please recognize that purchasing VITA-FAST tokens should not be regarded as an investment but as a contribution towards advancing scientific research. The focus is on the technology of tokenization, the utility of tokens within the IP pool, and the mission of scientific research.

It’s important to stress that VITA-FAST tokens are IP governance tokens that provide membership in an IP pool where members actively decide what happens with the IP and R&D data attached to the Korolchuk IP-NFT. They do not automatically confer any economic benefits or returns. This is not an investment contract where you’re buying a share of future profits.It is important to understand that there may never be a distribution of proceeds.

In fact, it is crucial to bear in mind the expectation of loss. The landscape of biotech research is fraught with challenges and the vast majority of research projects do not yield patentable IP. It’s very likely that the research may not lead to patentable IP, that buyers will not receive any money back, and that the tokens might not acquire any financial value.

As a VITA-FAST tokenholder, you have the responsibility to actively participate and put in work to generate value for yourself. Participation involves more than just owning VITA-FAST tokens. It means actively governing the direction of research and decision-making processes, and ensuring the responsible usage of the IP and R&D data.

Owning VITA-FAST tokens may require you to sign NDAs and provide KYC in order to access sensitive IP or R&D data. This is for the protection of the data and the rights of all parties involved. The rules for data access and privacy are to be governed into place by VITA-FAST tokenholders.

Finally, to our US community members, please note that due to unclear regulations, we unfortunately cannot include US buyers in this endeavor. This is an evolving area of law and while we are exploring ways to include the US, we are erring on the side of caution to ensure compliance with existing regulations.

The potential and the mission of this project are vast and exciting. However, it is crucial to approach this opportunity with a thorough understanding of the intricacies and potential risks involved. We encourage all potential participants to conduct their own due diligence and make informed decisions.

With these understandings and acknowledgments, we warmly invite you to participate, contribute, and make a difference today in the world of longevity research. Every step we take together brings us closer to deciphering the mysteries of longevity.

For more information, visit https://www.vitadao.com/viktor-korolchuk-lab-vita-fast-token-sale-with-vitadao and talk to us in our Discord auction channel.

Discover the power of autophagy, a natural cell rejuvenation process, through the enlightening YouTube video, Autophagy as a Target for Rejuvenation | VitaDAO and Viktor Korolchuk Lab.

Imagine having a direct hand in the future of longevity research. Today, we're thrilled to introduce VitaDAO's innovative approach to scientific research funding...
Read more
May Longevity Research Newsletter
June 9, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
May Longevity Research Newsletter

Introduction

Welcome to this month's edition of our newsletter, where we delve into the fascinating realm of gerontology - the scientific study of ageing - which has increasingly become a pivotal area of research, given the world's ageing population. This discipline, which explores the biological, psychological, and sociological aspects of ageing, has led to significant advances in understanding age-related diseases and interventions. Key to these advancements are human trials and interventions, turning laboratory findings into strategies that can enrich our lives as we age. This month, we are thrilled to feature an exclusive interview with renowned gerontologist, Professor Andrea Maier who brings an invaluable perspective and continues to break new ground in age-related diseases research.

This issue emphasizes the need for standardization in research methodologies to optimize comparability, which in turn may accelerate potential breakthroughs. As always, we strive to bridge the gap between the scientific community and policy makers, underscoring the urgent necessity for gerontology insights to inform public health strategies, legislation, and societal views on aging. Stay tuned for a captivating exploration of gerontology with Professor Maier.

VitaDAO Funded Research Projects

Viktor Korolchuk’s project, which aims to discover novel autophagy activators, will be the first VitaDAO project to undergo Synthesis of its IP-NFT after  receiving a 100% majority in the tokenholder vote.

Synthesis will create direct capital for the project and incentivise the VitaDAO community to contribute and move it into overdrive.

Longevity Literature Hot Picks

Preprint Corner

VitaDAO’s very own longevity overlay journal - The Longevist - had its first on-chain vote to determine what our team of curators think are the most exciting preprints of Q1 2023. We are aiming to launch the website later this month.

Prizes will be awarded for nominating preprints which make it into future issues of The Longevist. If you would like to nominate a preprint for inclusion in Q2 2023, then please follow this link - deadline 30th June 2023. 

This month we are featuring 6 new preprints which are all available to review on our reviewing platform The Longevity Decentralised Review (TLDR) in return for a bounty of 50 $VITA each. Simply follow the above link to the TLDR page and get reviewing! What's more, we will be continuing the 50 $VITA bounty for reviewing any of the preprints featured in previous issues of this newsletter.

DNA repair and anti-cancer mechanisms in the longest-living mammal: the bowhead whale

Initiation phase cellular reprogramming ameliorates DNA damage in the ERCC1 mouse model of premature aging

Cellular Senescence is a Double-Edged Sword in Regulating Aged Immune Responses to Influenza

DNA methylation rates scale with maximum lifespan across mammals

Breast cancer risk based on a deep learning predictor of senescent cells in normal tissue

Causal evidence that herpes zoster vaccination prevents a proportion of dementia cases

Published Research Papers

DNA methylation clocks for clawed frogs reveal evolutionary conservation of epigenetic aging

DNA methylation-based aging is conserved in frogs and mammals. The identified age-related CpGs are located in neural-developmental genes, implying a connection between epigenetic aging and age-associated diseases. The study opens avenues for using frogs as an aging model.

Senotherapeutic peptide treatment reduces biological age and senescence burden in human skin models

Researchers identified a peptide called Pep 14 that can effectively reduce senescence burden in human skin cells without toxicity. By modulating the activity of a specific enzyme involved in DNA repair and senescence pathways, Pep 14 improved the skin phenotype and reduced the biological age of aged skin samples in a safe manner.

Distinct longevity mechanisms across and within species and their association with aging

Study across 41 mammalian species reveals genetic signatures of lifespan control, including shared mechanisms like downregulated Igf1 and upregulated mitochondrial genes. Lifespan-extending interventions target genes related to energy metabolism.

Exercise reprograms the inflammatory landscape of multiple stem cell compartments during mammalian aging

Exercise rejuvenates aging stem cells, improves tissue regeneration, and reduces inflammation. This study examines the molecular changes induced by exercise in various stem cell compartments, highlighting its positive effects on intercellular communication and immune cells.

Effects of hunger on neuronal histone modifications slow aging in Drosophila

The sensation of hunger, by limiting BCAAs or directly activating hunger neurons, extends lifespan in Drosophila. Hunger-induced changes in neuronal histone acetylome seem to play a role.

Multivitamin Supplementation Improves Memory in Older Adults: A Randomized Clinical Trial

Participants taking multivitamins had better immediate recall memory compared to those taking a placebo. This effect was sustained over a three-year period, suggesting multivitamin supplementation could be a safe way to maintain cognitive health with age.

Association between genetically determined telomere length and health-related outcomes: A systematic review and meta-analysis of Mendelian randomization studies

Leukocyte telomere length (LTL) is associated with various health outcomes. Longer LTL is linked to increased neoplasm risk, while shorter LTL is associated with conditions like coronary heart disease and rheumatoid arthritis.

Autophagy enables microglia to engage amyloid plaques and prevents microglial senescence

Activated autophagy in disease-associated microglia helps maintain amyloid plaque homeostasis and prevent senescence. Inhibiting microglial autophagy worsens neuropathology, while removing senescent microglia could be a therapeutic strategy for AD.

Discovering small-molecule senolytics with deep neural networks

Deep learning was used to identify three drug-like compounds that selectively target senescent cells, similar to a known senolytic drug. These compounds reduced senescent cell burden in aged mice, highlighting the promise of deep learning in drug discovery.

Lifelong exercise training promotes the remodelling of the immune system and prostate signalome in a rat model of prostate carcinogenesis

Lifelong exercise training in a rat model of prostate cancer led to increased γδ T cells and lymphocyte infiltration in the prostate tissue. Exercise also induced changes in the expression of Oestrogen Receptor and Mitogen-activated Protein Kinase 13.

Region-based epigenetic clock design improves RRBS-based age prediction

Epigenetic clocks for mouse RRBS data have poor performance on external datasets. A study introduces regional blood clocks (RegBCs) that improve age prediction by using average methylation over large regions and outperform individual CpG-based techniques.

Antagonizing the irreversible thrombomodulin-initiated proteolytic signaling alleviates age-related liver fibrosis via senescent cell killing

Thrombomodulin (THBD) signaling identified as a key pathway in senescence. Inhibiting THBD signaling with vorapaxar eliminates senescent cells and restores tissue homeostasis in liver fibrosis models.

Alpha‑ketoglutarate supplementation and BiologicaL agE in middle‑aged adults (ABLE)—intervention study protocol

The ABLE trial investigates the effects of Alpha-Ketoglutarate (AKG) on aging in humans. Participants will receive AKG or a placebo for 6 months, with follow-up for 3 months. The study aims to measure changes in DNA methylation age, inflammatory and metabolic parameters, strength, arterial stiffness, skin autofluorescence, and aerobic capacity.

Published Literature Reviews

Chronic kidney disease promotes ageing in a multiorgan disease network

Chronic kidney disease has been identified as a significant risk factor for accelerated aging in multiple organ systems. This highlights the interconnectedness of age-related chronic diseases and their impact on mortality.

Senescent Cells: A Therapeutic Target in Cardiovascular Diseases

This review emphasizes the impact of senescent cells on different cardiovascular cell types and discusses the potential of senotherapeutics for treating cardiovascular diseases.

Senescence: a double-edged sword in beta-cell health and failure?

In pancreatic beta-cells, senescence contributes to age-related decline and diabetes. Targeting senescence is a promising approach for diabetes treatment. This perspective discusses the importance of senescence in beta-cell homeostasis and pathology.

Job board

Looking for a job in longevity research? Check out jobrxiv - we have saved you milliseconds by entering “aging” in the keyword search, but enjoy searching for whatever your research interests are!

Wyss-Coray Lab looking for a research assistant/associate to work on microglia, HSCs, lipidome or aging clocks

Hansen lab at the Buck are looking for new postdocs!

The focus of the research is to understand how the basic cellular recycling process called autophagy is regulated during aging, using the nematode C. elegans and mammalian cell culture systems, and a combination of biochemical, imaging, and genetic approaches.

The Benayoun lab at the Leonard Davis School of Gerontology at the USC is seeking a Research Lab Technician. They aim to use mice to understand sex differences in the aging process!

News

Hevolution Foundation Geroscience Research Opportunities (HF-GRO) Awards Unveiled, with a Budget of up to $115M Over Five Years

Resources

Longevity Biotech Landscape

Ada Nguyen provides us with an overview of the longevity biotech landscape, by sorting the different companies into 4 key therapeutic areas: reset & repair, replace, reprogram and discover.

Prizes

The Maximon Longevity Prize for Translational Research

Awards outstanding breakthrough research in the field of longevity that can be translated for clinical or non-clinical applications and has the potential to substantially increase health span or life span of humans.

They are welcoming applications from all academic/non-profit researchers, inventors, scientists, students or startup founders who either published breakthrough research, hold the patent or are in very advanced stages of research.

Articles

Biopharma Targets the Untapped Potential of the Human Healthspan

The Longevity Skeptic

The Secrets of Aging Are Hidden in Your Ovaries

Can a Kidney Transplant Drug Keep You From Aging?

Inside the Secretive Life-Extension Clinic

Speaking of Illusions: Sirtuins and Longevity

Combilytics: Taking a combined approach to senescence

NPR-8: The Protein That Could Extend Human Lifespan in a Warming World

The unexpected force that may make us get less sleep

More geriatricians needed to meet the needs of the UK’s ageing population

Why do some people live to be a 100? Intestinal bacteria may hold the answer

Vitalik Buterin Exclusive Interview: Longevity, AI and More

Speaking of Vitalik, he published a longevity pyramid short survey on his website. See where your health philosophy fits in!

Conferences, Workshops and Webinars

Meet the most inspiring speakers and experts gathering to discuss their present research.

Authophagy UK

19-20th June, University of Birmingham, UK

DGfA (German Society for Research on Aging) Annual Meeting

29-30th June, Volkshaus Jena, Germany

5th World Aging And Rejuvenation Conference

17-18 July, Frankfurt, Germany

Longevity + DeSci Summit NYC: Ending Age-Related Diseases 2023

10-11th August, New York

International Longevity Summit Africa

23-24th August, Johannesburg, South Africa

10th Annual ARDD

28th August - 1st September, Copenhagen, Denmark

BSRA Annual Scientific Meeting

6–8th September, University of Westminster, UK

The Groningen-Jena Aging Meeting "G-JAM" 2023 

28-30th September, European Research Institute for the Biology of Ageing (ERIBA) in Groningen, the Netherlands

Tweet of the Month

By Matt Fuchs - an excerpt from a twitter thread on the article mentioned above “The Longevity Skeptic”:

Messiness is Good. You want to find and follow scientists who’ve published research over the years that does *not* tell a clean story. “Our goal is to disprove our models,” @mkaeberlein said. Their findings and views should zig and zag as their careers – and science – progress

Follow scientists who write and talk publicly about new evidence that’s convinced them to reevaluate their own positions. Who embrace the inherent messiness of science – that’s the hallmark of an honest researcher

Podcasts and Videos

Pathways to Impact

A series of 12 virtual panels with longevity leaders

Coming on June 21st: Oprah and Dr. Peter Attia Get Real About Living Better for Longer

The Art of Aging: A Tail of Canines, Rapamycin, and Longevity with Dr. Matt Kaeberlein

The Sheekey Science Show: Is aging adaptive?

Interview with Prof. Andrea Maier

Andrea Maier, a renowned professor of gerontology, serves as the Co-Director of the Centre for Healthy Longevity at the National University of Singapore. With over 350 published articles, her groundbreaking research in ageing and age-related diseases is globally recognized, influencing health policy discussions including at the WHO.

What inspired you to enter longevity research?

It was a long, long time ago when I first observed that people age differently. This observation was especially apparent in some older women who exuded an undeniable power and vitality. This led me to question, how does this happen?

At the time, I was already engaged in research within the field of pulmonology, focusing on diseases like cytomegalovirus and other viruses. However, my observations compelled me to switch my focus. First, I transitioned into geriatrics, but I was always drawn towards research into longevity.

I find it truly astonishing how some individuals possess such vitality, even at an advanced age. Admittedly, I was also drawn to this field because it was not overly crowded. For instance, many researchers are interested in diabetes, making it a highly competitive field. Aging and longevity research, on the other hand, is a bit different. I would argue that this is still one of the least explored fields, especially when you consider it through the lens of a physician.

How has the field changed since you started?

In my view, the field hasn't changed at all in terms of the key players. That's something we might need to reflect on. However, the field itself is undeniably evolving. There's been a shift from purely academic pursuits to a greater involvement of startups. Venture capitalists are now engaging in our field and there's also a different level of government inclusion.

In the past, societal discussions were often centered around issues in nursing homes and frailty in hospitals. But now, we're talking about prevention and biological age. We're now in dialogues with organizations like the WHO how to introduce healthy aging to all citizens and the FDA about approving new drugs targeting ageing. This represents a substantial shift from being a purely academic field to one with a much broader scope and a greater societal impact. There should also be ubiquitous presence of regulators to ensure a balanced evolution of the sector.

Speaking of regulators, Singapore is allocating resources and effort to several exciting initiatives aimed at promoting healthy aging and the longevity industry. Could you give us with the latest and most significant updates?

Absolutely, Singapore is at the forefront of implementing many innovative healthcare strategies, largely out of necessity. Singapore boasts one of the highest life expectancies on earth, a distinction it shares with Japan and Switzerland. The country also has a remarkably low fertility rate, and individuals there tend to develop their first diseases at a relatively young age. These factors create a pressing need to lower healthcare costs, increase productivity, and reduce absenteeism. We are focused on stimulating human potential, given the limited natural resources due to the country's small size.

 Our current projects, such as the Healthier SG initiative, emphasize preventative actions and cost-benefit analysis, pushing towards a preventative approach in healthcare. They've also established health districts in collaboration with universities, the Housing Board, and industry partners, aiming to study and implement ways to enhance health and longevity.

In addition, Singapore is attracting startups for diagnostic or intervention purposes, utilizing a strong network of researchers and a robust engineering department. At the Center for Healthy Longevity, which I lead, we're conducting clinical research to bridge the gap from preclinical findings to clinical practice, integrating these into the healthcare system through evidence-based longevity clinics. All these efforts are driven by the necessity to enhance human potential.

Which of the current theories of ageing do you think are the most convincing?

There are many theories regarding aging. One such theory I came across is the grandmother hypothesis. This theory posits a question: why do women live beyond their reproductive age? Some research, including studies conducted in Ghana, suggests that there's not much evolutionary need for women to live beyond their reproductive years.

However, I think aging is much more complex. We recently published a paper in Nature Medicine that I'm quite proud of. It demonstrates how different organs within our bodies age at varying rates, and that there's a chain of action. If one organ begins to deteriorate, it's likely that another will soon follow, and we can now predict this.

There's not a singular mechanism behind aging. Rather, I think we're still uncovering the intricate complexities of the aging process and the networks between all the various hallmarks or mechanisms that influence each other.

Certainly, the accumulation of damage over time is a part of aging. But the question is how this damage occurs and how it can be repaired. We are dedicated to developing diagnostics to identify the weakest links in our bodies - which organ needs attention, what the common mechanisms are, and what the organ-specific mechanisms might be.

Other than your own, what do you think have been the biggest/important discoveries in the field?

There have been many significant findings in the field of aging, including the study of senescent cells. Therapies targeting these cells seemed to extend lifespan and improve health span in mice, creating significant excitement.

However, our field progresses through a series of small, yet important steps rather than a single breakthrough. One crucial development is our evolving understanding of the pathophysiology of aging, which has led us to develop diagnostics for biological age. This has the potential to be greatly impactful in our field.

The integration of our knowledge into practical applications that could impact everyday life is particularly exciting. Future interventions, backed by advancements in multi-organ treatments, could significantly increase health span, possibly more than lifestyle changes alone.

 

Which aspect of longevity research do you think requires more attention?

Currently, our focus is on human research, specifically on different organs' biological ages and how interventions can impact them. Over the last year and a half, we've developed a framework for diagnosing different physiological systems in the human body. If we want to tackle aging effectively, we should be able to measure the impact of interventions on every organ.

We use the eleven physiological systems defined by the Society of Physiology, such as the cardiovascular and endocrine systems. We've found that many trials only use a limited range of outcome parameters such as blood pressure, walking speed, and a few blood parameters. However, we often overlook critical aspects like lung function, women's health, cognitive function and oral health, which could be crucial in aging. Similarly, while many are focused on the microbiome, few are investigating the interaction of several organ systems.

In essence, we are trying to identify and fill the gaps in our understanding of aging across the various physiological systems. By systematically reviewing the literature, we're discovering blind spots and working to address them, to truly comprehend the complexity of aging across our body's many intricate systems.

What mistakes do you think the longevity field has made?

I wouldn't say mistakes. Instead, I'd liken our current stage to that of a toddler. Like a toddler, we are learning, experimenting, observing our environment, and trying different approaches. We're not yet in the stage of 'puberty'; we're still trying to grasp the basic concepts and parameters of our field.

However, one thing we could improve upon is leveraging insights from various specialties and fields. We should learn from others, review the literature, understand different trial designs and methodologies, and apply them in our field. This sort of cross-pollination is essential for growth and advancement.

While it's difficult to pinpoint any specific mistake with a significant negative impact, we must realize that, like a growing child, we will eventually need to mature and refine our approaches. The understanding and patience granted by governments and institutions due to our field's relative youth won't last forever. We need to keep learning and advancing to fulfill the potential of our field.

Is ageing a disease?

Yes! Yes, it absolutely is a disease. And we all have it!

Referring to aging as a disease might be counterproductive when communicating with the general public. However, as a physician, you require a clear diagnosis and treatment plan for your patients.

By treating the underlying mechanisms and consequences of aging, we aim to optimize the overall health and well-being of your patients. Diagnosing aging-related deterioration and assessing biological age allows you to tailor interventions and treatments accordingly. It provides a framework for addressing the physiological and pathological changes associated with aging.

In the medical field, having a clear understanding of aging and its impact on health is crucial for establishing effective practices and offering appropriate interventions to enhance patients' quality of life.

Your lab is mostly focused on longevity interventions in humans. What are the most important endpoints, biomarkers and measurements you look at?
In terms of endpoints and biomarkers, there is a both short-term and long-term answer. I have gained experience from various fields, including endocrinology and oncology, where trial designs have been well-established over several decades. We are now realizing the importance of implementing proper trial designs in the field of aging research as well. Simply relying on observational studies is no longer sufficient for determining outcomes.

Currently, we utilize surrogate markers or parameters, such as C-reactive protein (CRP), which is an inflammation marker. In some of my studies, I use CRP as a primary outcome, not necessarily as a longevity marker, but as an indicator of reduced inflammation. By demonstrating a decrease in CRP levels, we can refer to existing literature to understand the implications of such a change, such as the decreased likelihood of developing cardiovascular disease, for example. Additionally, we employ epigenetic clocks using methylation data in our trials. Instead of relying on a single clock, we use the median of multiple clocks since it is still uncertain which one is the most accurate. This approach allows for risk spreading, which is crucial since our trials have relatively short durations of two to three years. We need to consider outcomes that we would like to avoid in the long run, even if they might take a hundred years to manifest.

However, it is important to note that the long-term outcome parameters are more organ-specific and highly significant. In my research, the population I focus on for randomized control trials is much younger, typically between 30 and 60 years old. Therefore, using these specific endpoints would require significantly longer follow-up periods. It is also worth mentioning that I'm not primarily interested in dichotomous outcomes where individuals either develop a disease or not. Instead, I focus on continuous outcome parameters indicating the function of organs.

Ultimately, the decline of function in aging is a continuous process, and our goal is to determine how much decline we can prevent to make a clinically relevant impact. Currently, our field is in the process of understanding what is considered clinically relevant and within what timeframe such interventions need to take place.

As standardization can be a concern in human aging studies, can you elaborate on your ability to compare your findings with other similar studies conducted by various institutions, including academic and clinical ones?

This is a huge problem! It is a big mess at the moment and this brings me back to the toddler analogy. We haven't defined or standardized our approaches yet. I often find it challenging to conduct meta-analyses in systematic reviews due to the heterogeneity of studies. This serves as an alarm signal that standardization is necessary.

That's why I founded the Healthy Longevity Medicine Society (HLMS) to bring together healthcare professionals, scientists, and entrepreneurs. One of our major goals in HMS is to establish standards in research. This includes standardizing how we measure parameters like musculoskeletal or women’s health outcomes. Currently, there are numerous different ways to measure these parameters, leading to inconsistent outcomes.

We have learned from other fields, such as cardiology, that standardization is crucial. For example, we know how to measure blood pressure accurately by ensuring individuals rest for 3 to 5 minutes in a quiet room. We are now at the stage where we need to standardize our methods, what we measure, and when we measure it.

To address this issue, we established a consortium of publicly funded hospitals that are setting up longevity clinics. We are currently designing standardized protocols for measuring physiological systems in humans. By adhering to these protocols, we can ensure consistency. The next step will be certification, where practitioners receive a certificate for following the standardized protocols.

We need to accelerate the implementation and scalability of these standards to effectively reverse the aging process. Without standardization, we are just surrounded by noise caused by inconsistent methodologies.

When selecting interventions to test in humans, how much significance do you place on data derived from animal models, and what has been the level of translatability in your experience?

I always look at animal models as an initial step before starting a study. We conduct systematic reviews and analyse animal and human studies to inform our translational approach. Learning from past experiences, we have seen successful translation in the longevity field. For example, supplements that have shown positive effects in animal models often have a good success rate when translated to humans. The success depends on the mechanism being studied, as certain mechanisms are conserved across species.

However, it's important to note that we don't solely rely on animal data. If a therapeutic intervention shows safety in humans, and the animal data is not available or necessary for safety assessment, we may proceed directly to human trials. The primary consideration here is the safety aspect. Once we establish the safety, we can implement it in human studies to assess the effects.

Outro

Thank you for staying with us until the very end and as always we encourage you to reach out to us about content you’d like us to discuss in our next issues. See you next month!

Further Reading

Unbiased evaluation of rapamycin's specificity as an mTOR inhibitor

Mapping the trajectory of healthy aging: Insights from longitudinal cohort studies examining lifestyle factors

Novel Strategy in Searching for Natural Compounds with Anti-Aging and Rejuvenating Potential

Introduction Welcome to this month's edition of our newsletter, where we delve into the fascinating realm of gerontology - the scientific study of ageing - which has increasingly become a pivotal area of research, given the world's ageing population.
Read more
The Epigenetic Edge: Exploring Fitness, Aging, and Science with Prof. Nir Eynon during The Aging Science podcast by VitaDAO
June 6, 2023
Awareness
Longevity
The Epigenetic Edge: Exploring Fitness, Aging, and Science with Prof. Nir Eynon during The Aging Science podcast by VitaDAO

In this episode of the Aging Science podcast I talked with Prof. Nir Eynon (@nir_eynon). We touched on many topics, including elite athletes, how COVID affected working culture (for the better), the benefits of tabatas, epigenetic and muscle clocks and how they are affected by exercise, biases in research and the role of meta-analysis. I hope you will enjoy this episode as much as I did.

Short Bio – Nir Eynon

Nir earned his PhD degree with high distinction in 2010 from Porto University, Portugal. Early during his career, he was studying the genetics of elite athletes before switching to aging research and epigenetics.

His work on aging brought him to Victoria University where he became a program leader, at the Institute of Health and Sport (iHeS). Among other things, at Victoria he now runs the Gene SMART study on exercise through which his team aims to uncover the genetic and epigenetic responses to exercise.
https://www.vu.edu.au/institute-for-health-sport-ihes/research-areas-in-ihes/mechanisms-interventions-in-health-disease/muscle-exercise-movement/human-gene-smart-study

Let’s keep in touch - the Australian Physiological Society Meeting and more

Before we talk about epigenetics and exercise a few housekeeping notes.

Nir asked me to promote this year's AuPS Annual Scientific Meeting (@AuPhysiolSoc) which will be held in Melbourne on 26-29 November 2023. This, I am sure, would be a great opportunity to meet Nir personally so please attend if you can. I will also keep an eye on this meeting and hopefully there will be other Australian aging researchers attending. In particular I would be keen to meet with Rene Koopman who has published excellent work on iron dyshomeostasis and age-related muscle loss (Alves et al. 2021).

Would be lovely to run into other aging researchers or longevity enthusiasts there. In the meantime I suggest you can follow us on these channels if you have questions or speaker suggestions:

VitaDAO twitter account: @vita_dao
VitaDAO discord channel: https://discord.gg/vitadao
My own twitter account: @Aging_Scientist
VitaDAO telegram: https://t.me/vitadao

What is epigenetics?

The term epigenetics describes a form of gene regulation that relies on modifications to the (chemical) structure and organization of DNA and chromatin. This is in contrast to genetics which deals with changes to the DNA sequence itself.

Epigenetic marks are a type of modification that is attached to DNA that help genes to be expressed at the proper time. There are two major types of epigenetic marks, those found on histones and those found directly on the DNA. Histone modifications are highly diverse, whereas regulatory DNA modification is largely limited to the methylation of cytosines.

The different epigenetic marks are recognized by reader proteins that affect gene expression. Although these marks can have complex effects, some broad patterns are evident, like for example repression of transcription through CpG methylation.

Although methylation serves an important regulatory role, it turns out not all species have methylation at CpG sites. Bacteria have a different type of DNA methylation whereas C. elegans seems to have lost this feature altogether during evolution (2). That is one of the things I learned in this podcast!

The rise of epigenetic clocks

When we age so does our epigenome and one way to measure this epigenetic aging is through so called DNA methylation clocks. Many studies, initially pioneered by Steve Horvath, have shown that methylation at certain sites in the genome can be used to construct a clock that closely tracks the passage of time.

These clocks are exciting because they are much better at predicting chronologic and biologic age than the biomarkers we used to have. In fact, I still remember going to conferences and listening to talks that painted a rather disappointing picture of blood based aging biomarkers (e.g. based on the MARK-AGE project [2]).

These clocks are also exciting because they promise to become a surrogate endpoint for medium sized phase II-like studies. A surrogate endpoint is a cheap, easy-to-measure marker that substitutes for something else more expensive. The hope is that a slowing of epigenetic aging in small studies would be predictive of success in larger trials that focus on harder outcomes like health and survival. If true, this would push down the cost of trials by orders of magnitude because we could pre-select promising compounds before going into large studies. However, even the best surrogate endpoints like LDL or HDL cholesterol cannot substitute for large studies, as the countless failures and disappointments in the clinic show (e.g. CETP-inhibitors for atherosclerosis and the clinical failure of most non-statin drugs).

Another reason why clocks are important is their link with the biology of aging. Understanding why methylation at certain sites changes and what harmful effects these changes have, could improve our understanding of aging and allow us to find new therapies. To give an example of such advances, as mentioned in a prior podcast, the study of epigenetics has uncovered not only the importance of entropy in the process of aging, but also ways to reset some of these age-related changes (partial reprogramming). Nir is also interested in understanding sex differences in epigenetics as well as the effects of exercise on epigenetics. This could be very important since we still cannot fully explain why women live longer than men, much less on the epigenetic level.

However, there is a world of epigenetics beyond clocks, as Nir mentioned during the podcast. He is very interested in studying sites that are not commonly selected to build clocks because these could still have important functions that are being neglected.

Biases and problems in research

"Better take your chances when young" (Nir Eynon)

We discussed two large problems in research, one was the lack of diversity in the choice of our research subjects and the second one were the working conditions for scientists.

Historically, clinical trials have included predominantly Caucasian white males. This underrepresentation of minorities and women has obvious negative consequences because the therapies and clinical practices developed for white men may not translate to other groups (also known as lack of external validity). Interestingly, the same issues plague mouse research. Many studies use one strain of mice, therefore not representing the true genetic diversity of the species and in the past most studies used almost exclusively male mice of one common strain.

In his research Nir seeks to correct these biases by recruiting at least 50% female participants for his studies. This will allow him to better understand the epigenetic differences between men and women. 

As for the second point, poor working conditions, we specifically talked about the difficulties that parents and couples face. In fact, scientists are often expected to move across the globe in search of a lab that is a good fit for them and to further their career. Modern science is extremely specialized so only a couple of labs across the whole globe may be working in your niche. Therefore, it is almost impossible to be successful without moving from position to position and country to country.

Given this, it is almost surprising that people are still willing to work as scientists. Very few jobs in the world demand constant relocation, combined with low pay, long working hours, high pressure and mediocre job stability. In our conversation with Nir we explored ways in which this is improving and what career path might be most suited for those who cannot or do not want to relocate (I won’t spoil the answer). 

Exercise and Aging - facts, controversies and open questions

I think Nir is doing brilliant work trying to figure out how exercise affects the epigenetics of aging but I would like to provide an interesting perspective worth discussing.  Does exercise really slow aging? I think some of the data Nir mentioned himself is consistent with the idea that it does not.

TextDescription automatically generated

However, whether we believe that exercise actually slows aging or that it “only” has tremendous benefits for age-related health, we all agree that exercise is very important.

From a public health perspective exercise is still the holy grail, but from gerontologic perspective I think exercise is a bit oversold (2).

The other issue with exercise is compliance. No matter how healthy it is, many people do not have the time or motivation to actually go out and exercise. We need exercise in a pill and this is where Nir's research comes in and that of other exercise researchers. Although drugs will never fully recapitulate the benefits of exercise, we will nonetheless be able to reap some of the benefits of regular exercise if we gain an understanding of the molecular changes underlying exercise adaptation.

“I think at the end of the day, scientists that are working in aging, the differences in opinions between everyone are not that large. We want to think that they are very large and very big. But it's all nuances. Because at the end of the day, we want people to be healthy for a long time. That's the goal.” (Nir Eynon)

References

Weinhouse, Caren, et al. "Caenorhabditis elegans as an emerging model system in environmental epigenetics." Environmental and molecular mutagenesis 59.7 (2018): 560-575.

Alves, Francesca M., et al. "Iron accumulation in skeletal muscles of old mice is associated with impaired regeneration after ischaemia–reperfusion damage." Journal of Cachexia, Sarcopenia and Muscle 12.2 (2021): 476-492.

Notes

1/ There appears to be some genuine controversy whether C. elegans shows no methylation or whether they do have adenine N6 methylation (6mA).

Greer, Eric Lieberman, et al. "DNA methylation on N6-adenine in C. elegans." Cell 161.4 (2015): 868-878.

Also: https://pubpeer.com/publications/B024A9B1AFF2ED7E69DB745662382B?utm_source=Chrome&utm_medium=BrowserExtension&utm_campaign=Chrome

2/ The idea that exercise does not affect aging rate is based on studies in mice that fail to show lifespan extension with exercise and a rather pessimistic reading of the outstanding rat literature by John Holloszy. While in his studies he did usually find that exercise led to a modest extension of median lifespan in rats, this increase was lower than for caloric restriction. Furthermore, there was no improvement in maximal lifespan with exercise which led him to argue that exercise does not slow the aging rate. However, this might be over-interpretating the absence of a signal since maximal lifespan is notoriously difficult to measure due to dwindling sample sizes at old ages; and its use as a marker of aging rate remains somewhat controversial.

Garcia-Valles, Rebeca, et al. "Life-long spontaneous exercise does not prolong lifespan but improves health span in mice." Longevity & healthspan 2 (2013): 1-12.

Holloszy, John O. "Mortality rate and longevity of food-restricted exercising male rats: a reevaluation." Journal of Applied Physiology (1997).

3/ My intuition is that you can construct a clock using any type of dataset, be it epigenetic, transcriptomic, proteomic or using plasma proteins as was tried in the MARK-AGE study. However, the missing link was the combined use of multiple markers. No epigenetic clock uses a single site and it was a mistake to look for a single best blood biomarker of aging. The idea of using composite clocks is one of the big breakthroughs.

In this episode of the Aging Science podcast we talked with Prof. Nir Eynon (@nir_eynon). We touched on many topics, including elite athletes, how COVID affected working culture (for the better), the benefits of tabatas, epigenetic and muscle clocks
Read more
April Longevity Research Newsletter
May 16, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
April Longevity Research Newsletter

Introduction

Welcome back Vitalians! We are thrilled to celebrate our one-year anniversary of bringing you the latest research, insights, and developments in our field. Over the past year, we have covered a wide range of topics, from artificial intelligence and sustainability to genetics and aging research.

As we reflect on the past year, we are grateful for the support and engagement of our subscribers. Looking ahead, we are excited to continue bringing you the latest news and trends in our field, while also exploring new areas and expanding our coverage. Thank you for your support over the past year, and we look forward to continuing this journey with you for many more years to come. 

In this edition, we are excited to feature an interview with Dr. Shahaf Peleg, a renowned expert in the field of longevity research. His insights inspired us to consider some limitations the field might be facing. Despite the increasing interest and investment in the field of aging research, it has become progressively narrower in recent times. Many scientists concentrate their efforts on traditional aspects of aging research such as diet control, physical activity, and well known longevity interventions (e.g. Rapamycin), thereby disregarding less conventional areas that have greater risk. This tendency towards conventional approaches limits funding, publication, and progress in the field, and dominant concepts are unlikely to surpass the longevity ceiling, consequently decreasing the potential impact on healthy lifespan.

To overcome this barrier, it is essential to allot more funds and attention to novel, high-risk projects that have the potential for more significant impacts on health lifespan. These undertakings may be less orthodox, but they could be the means to unlocking new discoveries that could revolutionize the field of aging research. By promoting more daring and innovative thinking, we can identify new avenues of research and discover new treatments that could help us increase healthy lifespan beyond the current limit. This requires a shift in the mindset of the research community, and greater support from funders and policymakers who understand the significance of investing in unconventional, innovative, and even risky projects that can progress the field.

VitaDAO Funded Research Projects

Congratulations to the ImmuneAge team for receiving a 99.83% majority in the tokenholder vote to secure VitaDAO funding and support. 

ImmuneAge aims to rejuvenate aged hematopoietic stem cells to tackle age-related decline in the immune system, which is associated with numerous diseases due to pathological effects including chronic inflammation and loss of tumour surveillance.

Longevity Literature Hot Picks

Preprint Corner

VitaDAO’s very own longevity overlay journal - The Longevist - had its first on-chain vote to determine which preprints will receive a coveted place in the inaugural issue! Announcement of the preprints and journal launch coming in mid-May!

This month we are featuring 6 new preprints which are all available to review on our reviewing platform The Longevity Decentralised Review (TLDR) in return for a bounty of 50 $VITA each. Simply follow the above link to the TLDR page and get reviewing! What's more, we will be continuing the 50 $VITA bounty for reviewing any of the preprints featured in previous issues of this newsletter.

Five years later, with double the demographic data, naked mole-rat mortality rates continue to defy Gompertzian laws by not increasing with age

Development of a novel epigenetic clock resistant to changes in immune cell composition

Senescent cardiomyocytes contribute to cardiac dysfunction following myocardial infarction

OGG1 and MUTYH repair activities promote telomeric 8-oxoguanine induced cellular 

senescence

Evidence of a pan-tissue decline in stemness during human aging

Epigenetic fidelity in complex biological systems and implications for ageing

Published Research Papers

Ageing-associated changes in transcriptional elongation influence longevity

This Nature study found acceleration of RNA polymerase II (Pol II) is linked to aging and shorter lifespan as it becomes less precise and more error-prone with age. Mutations in insulin-signaling genes and low-calorie diets can slow down Pol II speed and increase lifespan. The study establishes a causal connection between Pol II speed and lifespan.

Dedifferentiation maintains melanocyte stem cells in a dynamic niche

Why does our hair turn grey with age? We know that it’s due to failure of the melanocyte stem cell (McSC) system, however the Ito lab has provided a mechanistic explanation as to why this happens. Unlike other stem cell populations, McSCs can switch between two different states (stem and transit amplifying) and have motility to travel between different compartments. However, some cells lose this motility, fail to dedifferentiate and melanocytes numbers diminish leading to a loss of pigment. 

A population of stem cells with strong regenerative potential discovered in deer antlers

Numerous animals such as lizards and lobsters have significant regenerative capability whereby whole limbs can be regrown, whereas regenerative capability in mammals is rare and less extreme, for example mice can only regenerate digit tips. However, deer have an amazing capability to regenerate their antlers each year, which the Qiu lab show is due to a population of cells called antler blastema progenitor cells (ABPCs). Interestingly, mice have a similar cell type in their regenerative tip.

Biological age is increased by stress and restored upon recovery

A study showing biological age - as quantified by transcriptomic, metabolic and epigenetic readouts - can increase as result of stresses such as heterochronic parabiosis in mice, or severe COVID-19 infection, which can then revert back following cessation of the stress and recovery. 

Lung adenocarcinoma promotion by air pollutants

Research showing that air pollutants contribute to lung cancer by eliciting an inflammatory response resulting in macrophages releasing the cytokine interleukin-1β, which in EGFR mutant lung alveolar type II epithelial cells can induce a progenitor-like state that drives tumourigenesis.

Cold temperature extends longevity and prevents disease-related protein aggregation through PA28γ-induced proteasomes

It has been shown that lowering body temperature can extend lifespan in numerous organisms such as nematode worms and fruit flies. The Vilchez lab now show that this effect is dependent on a protein involved in proteasome activation, which can allay age-associated protein aggregation. Interestingly, we recently featured research from John Speakman showing that body temperature impacts lifespan more than metabolic rate in 2 small mammals. Could these two findings be connected?

CISH impairs lysosomal function in activated T cells resulting in mitochondrial DNA release and inflammaging

We experience an age-related decline in immune function which can lead to chronic systemic inflammation, which is associated with numerous diseases. Here the Goronzy lab provide a mechanism whereby the proton pump V-ATPase is targeted for degradation, leading to faulty lysosomes and subsequent accumulation of amphisomes which undergo extracellular release of their mitochondrial DNA-containing cargos, which then elicit an immune response.

Reversal of liver failure using a bioartificial liver device implanted with clinical-grade human-induced hepatocytes

A bioartificial liver device employing human-induced hepatocytes has shown promise in treating post-hepatectomy liver failure in both animal and human studies. The treatment was well-tolerated and improved liver function, suggesting it could expand the population of eligible patients for liver resection.

Engineering longevity—design of a synthetic gene oscillator to slow cellular aging

By rewiring an endogenous toggle switch, sustained oscillations were created that increased the cells' lifespan by delaying the commitment to aging. This study provides a foundation for designing gene circuits that slow aging.

CRISPR-activated expression of collagen col-120 increases lifespan and heat tolerance

sgRNAs fed to C. elegans to CRISPR-activate the expression of collagen col-120 and col-10 in adulthood increase lifespan and heat tolerance. Results suggest collagen enhancement may promote longevity and stress resistance.

Genetic perturbation of mitochondrial function reveals functional role for specific mitonuclear genes, metabolites, and pathways that regulate lifespan

New evidence that cellular aging is fundamentally controlled by mitochondrial function.
This study identified a common mechanism that regulates lifespan by controlling cytosolic ribosomal protein abundance, actin dynamics, and proteasome function through mitochondrial genes. 

Body composition and mortality from middle to old age: a prospective cohort study from the UK Biobank

300,000 participants study found that being overweight didn't increase mortality risk, but obesity did, though the risk decreased with age. Additionally, lean mass didn't reduce mortality risk.

Spurious intragenic transcription is a feature of mammalian cellular senescence and tissue aging

Aberrant transcription occurs during cellular senescence and mouse aging, impacting cellular health and signal transduction pathways. This suggest that age-related spurious transcription promotes a noisy transcriptome and degradation of coherent transcriptional networks.

Published Literature Reviews

No rest for the wicked: Tumor cell senescence reshapes the immune microenvironment

Mitigating age-related somatic mutation burden

Recent clinical trials with stem cells to slow or reverse normal aging processes

Targeting the biology of aging with mTOR inhibitors

Job board

Retro biosciences are hiring across the board from fellows and research associates to project managers 

Retro are focusing on Cellular reprogramming, autophagy, & plasma-inspired therapeutics.

Rubedo are looking for Director of Program Management and Portfolio Strategy

Rubedo works on innovative therapies engineered to target cells which drive chronic age-related diseases and their drug discovery platform has engineered novel first-in-class small molecules designed to selectively target senescent cells.

Calico is now accepting applications for postdoctoral fellowships

They have openings across Protein Biochemistry and Structure Biology, Lung Homeostasis and Aging, Immuno-Oncology, and Mechanisms of Yeast Aging

The Hansen lab is inviting highly qualified, inquisitive scientists to apply for positions as a Postdoctoral Researcher

Buck Institute for Research on Aging, Novato, California

The Lapierre Lab is looking for motivated Ph.D. students and postdoctoral researchers!

To study how nucleo-cytoplasmic protein partitioning affects proteostasis, autophagy and aging using C. elegans.

News

People are taking a cheap drug to try reverse-age their ovaries — and cheat their 'biological clock'

Human trial - rapamycin for reproductive aging

King’s College and the LSF partner to advance human longevity education and research

Life Biosciences Presents Groundbreaking Data at ARVO Demonstrating Restoration of Visual Function in Nonhuman Primates

Introducing the Time Initiative with AFAR are empowering undergraduate leaders in aging research

Unity biotechnology announces positive 48-week results from phase 2 BEHOLD study of UBX1325 in patients with diabetic macular edema

FDA approves Biogen’s ALS drug tofersen, paving new road for accelerated pathway in neuro diseases

Drs. Sinclair, Huberman, and Brenner on the Future of NMN After the FDA Ban

Experimental Alzheimer’s drug slows cognitive declines in large trial, drugmaker Eli Lilly says

The antibody therapy in stage III clinical trial improved cognitive and functional decline by 35%

Longevity Investors Conference to showcase promising startups to high-net-worth investors

Resources

Computational Biology of Aging coursebook

The course covers computational biology methods for studying aging and offers hands-on experience in analyzing gene expression, constructing aging clocks, and using dynamical systems modeling to study the biology of aging.

Karl Pfleger: Announcing a categorization of 18 categories of aging/longevity companies & mappings between this, the SENS areas, Hallmarks & Pillars

Articles

Humans Can Stop—But Not Fully Reverse—Aging, Study Suggests

Why don’t whales get cancer? Cracking one of medicine’s greatest mysteries

Longevity for companion animals: why should we care

Possible ‘Steps’ to Revealing Super-Agers

The “stepping stone” approach to getting longevity drugs to market

Longevity is a dish best served cold

Ageing studies in five animals suggest how to reverse decline

Boosting body’s antiviral immune response may eliminate senescent cells

Benefits of 'zombie' cells: Senescent cells aid regeneration in salamanders

The science of reversing aging

Moderate Drinking Doesn’t Lower Mortality

Longevity Seekers Embraced This Drug. But Does It Actually Fight Aging?

Improving longevity literacy – and why digital health is the answer

“We Have 30 Extra Years”: A New Way of Thinking About Aging

Rapamycin's Role as a Molecular Brake for Cellular Hyperfunction and Runaway Cells

Women In Longevity And The Renaissance Of The SENS Research Foundation

The tech entrepreneur betting he can get younger

Conferences, Workshops and Webinars

Meet the most inspiring speakers and experts gathering to discuss their present research.

Pop-up city - Zuzalu

Longevity Events in May

May 6-9th: Longevity 0-1

May 13-14th: Longevity Industry Talks

The 2023 Harvard/Glenn Symposium on Aging

22nd of May, virtual

Authophagy UK

19-20th June, University of Birmingham

5th World Aging And Rejuvenation Conference

July 17-18, 2023 in Frankfurt, Germany.

BSRA Annual Scientific Meeting

British Society for Research on Ageing is pleased to hold its 2023 annual meeting at the University of Westminster

DGfA (German Society for Research on Aging) Annual Meeting

June 29-30th, 2023 at the Volkshaus Jena.

The Groningen-Jena Aging Meeting "G-JAM" 2023 

28-30th September 2023 at the European Research Institute for the Biology of Ageing (ERIBA) in Groningen, the Netherlands. 

Awards and Competitions

Maximon Longevity Prize

50 000 CHF for the winner

Deadline for applications: 30th of June

The Longevity Prize announces the winners of the Hypothesis Prize!

Tweets

Thought-provoking:

Anil Bushnan: Senescent cells are sentinel cells that coordinate an immune response. Immune surveillance with senescent cells is a mechanism for tissue homeostasis. There are no “good” and “bad” senescent cells-just the detrimental effects of failed immune surveillance leading to accumulation.

Controversial:

Elon Musk: Our lifespan is programmed into our genes, just like it is for fruit flies or pets. Fruit are already vegan & even if they do yoga every day & take every supplement, they still only live 50 days.

Elon Musk: The precision of our biological clock across trillions of cells is incredible. One never sees someone with an old left arm, but a young right arm. Aging can obviously be fixed. The real question is whether it should be.

Books

The Age of Scientific Wellness: Why the Future of Medicine Is Personalized, Predictive, Data-Rich, and in Your Hands

Taking us to the cutting edge of the new frontier of medicine, a visionary biotechnologist and a pathbreaking researcher show how we can optimize our health in ways that were previously unimaginable.

Outlive: The Science and Art of Longevity

“A groundbreaking manifesto on living better and longer that challenges the conventional medical thinking on aging and reveals a new approach to preventing chronic disease and extending long-term health, from a visionary physician and leading longevity expert.”

Podcasts and Videos

What they thought of aging in 1931 - The Sheekey Science 

What did scientists think about aging in 1931? That’s right. 1931, because that is the year the first biological textbook was published “The Science of Life”.

NewLimit are aiming to extend human lifespan, here is a progress update

Interview with Dr. Shahaf Peleg

Dr. Shahaf Peleg is a group leader at the FBN Institute at Dummerstorf, Germany, where he focuses on studying the role of metabolism-epigenetics connectivity during early aging, response to stress, and cognitive impairment. Through his research, Dr. Peleg aims to identify new and innovative therapeutic avenues that can help increase healthy human lifespan.

What inspired you to enter longevity research?

When I was in high school, I was thinking about what I should do later in life. Should I become a lawyer? Politician? Businessman? But the more I thought about it, the more vain it seemed. I wanted to tackle an interesting challenge in life that would make a difference. Studying aging immediately popped up in my mind as such an (impossible) challenge. Also, I noticed with sadness that people age and die and I knew from a young age that I would go on one day to study aging.

Which of the current theories of ageing do you think are the most convincing?

To be honest, I believe that a truly convincing theory of aging has yet to emerge.

How has the field changed since you started?

To put it in perspective, When I started my Masters in 2006, there was already an increasing interest in researching aging, leading to more investment, funding, and conferences in the field.

Advancements in technology, such as CRISPR, advanced omics, AI, and novel drugs, have also enabled more efficient aging research. Also, the rapid development of the internet and applications such as Twitter have facilitated collaborations among scientists and enable them to interact more efficiently than before.

We also observed that many scientists have started to question previous assumptions, results, and theories on aging, which were previously published, and this is followed by heated debates. As the field of aging grows and we collect more observations, it is likely we will disprove more previous theories on aging.

Unfortunately, the field has surprisingly become narrower. A handful of concepts such as diet, exercise, rapamycin, mtUPR, and a few others have become dominant – so dominant that they are over-studied, over-published, and importantly, over-funded. This poses dual disadvantages by inhibiting other promising ideas to be studied/funded/published and by the fact that it is likely that the current dominant ideas will not break the glass ceiling of longevity.

 

What mistakes do you think the longevity field has made?

I have alluded to it in the previous point. The field is too narrow and it’s easier to get funded and publish if the scientist chooses a ‘safe’ canonical aspect of aging research such as caloric restriction. But this is not the way to go. We need to encourage risky projects, out-of-the-box ideas and younger researchers (not always the big names). If we really wish to have a fair chance to increase life span beyond 120, the field needs to also pursue such novel ideas – the current canonical ideas may increase healthy lifespan, but unlikely to bring us beyond the 120 mark.

It is also concerning that some of the so-called-established results (leading to theories of aging) were based on data that is not reproducible. Considerable time, effort, and money were invested in follow-up studies, which resulted in failures and even discredited the field of aging to some extent. This may have been due to researchers taking other labs' results for granted without sufficiently questioning and thoroughly reproducing the published data.

Other than your own, what do you think have been the biggest/important discoveries in the field?

Naturally, the study by Cynthia Kenyon in 1993 showing that aging can be genetically modulated really pushed the field forward. I think many discoveries are important and big in the field, and it’s hard to pick just a few. However, I do find Darren Baker's work on targeting senescent cells interesting, and I believe the research on drugs such as Rapamycin and Metformin is crucial as they show potential in increasing healthy lifespan. Additionally, Alex Zhavoronkov's work on using AI to intervene with aging is novel and may be critical for the future of the field, as it's possible that AI could better assess the enormous amount of factors that contribute to aging. Nevertheless, there are many important discoveries in the field, and only time will tell which ones are promising.

What advice would you give to people currently working in longevity research?

To stay open-minded, respect the opinions of other colleagues (even when they differ from yours), be bold, think outside the box, and don't give up when things don't work. Longevity research is challenging, but that's precisely why we do it.

Which aspect of longevity research do you think requires more attention?

More attention should be given to data reproducibility and increasing funding, particularly for risky projects.

Is ageing a disease?

No, it’s worse. Studying aging is currently the second most unattainable and important quest of mankind (I’ve ranked space travel as first). Aging is more complicated, inevitable, and lacks any treatment in comparison. Moreover, it is the primary cause of a large number of diseases, and slowing down aging can delay many of them. Aging is a fundamental process that gradually 'kills' us.

 

Your research covers a broad range of topics, including epigenetics, metabolism, and various dietary interventions. What is the biggest impact of consuming certain foods and drinks on epigenetic age?

 

While I am not certain about the impact of epigenetic age (and in fact, what epigenetic age tells us in terms of aging really), maintaining a healthy diet and exercising regularly can significantly improve your healthy lifespan and delay the onset of certain diseases. However, it is important to note that these practices alone may not extend lifespan beyond the 120 mark.

 

Your latest study used optogenetics to increase mitochondrial membrane potential in C. elegans. Can you explain the basics of optogenetics and how you were able to use this technique to modulate mitochondrial function? Do think your finding might have translational potential in the context of human aging and longevity?

 

I'll start with the second question - yes, I think it has potential for translation to human aging, otherwise, I wouldn't be interested in it. Our goal is never to make yeast/worms/flies/fish/mice live longer - we always aim for human aging!

I'll leave the explanation of basic optogenetics to more qualified researchers. What we're doing is not really modulating mitochondrial function or even classical optogenetics in that sense. The idea, at least from my current view, is that we use an engineered light-activated proton pump and place it inside the mitochondria. This tool was developed by Brandon Barry at Andrew Wojtovich's lab. By taking this approach, we enable animal cells to harness the energy of light into chemical energy. We theorize that this can lead to several benefits in terms of aging. For example, it's currently assumed that mitochondrial function declines with aging, and as such, our approach may compensate for impaired metabolic function and increase cellular energy levels simply by exposure to light. Please read our Nature Aging paper that we published earlier this year. We're just at the beginning, and time will tell if this risky idea will work for us.

 

Looking ahead, what are your upcoming research projects or collaborations that you are particularly excited about? Can you give us a sneak peek into the direction of your future work?

I would like to continue the optogenetics project as I am very excited about its potential. As for a sneak peek… I would like to start a company!

Outro

Thank you for staying with us till the very end and as always we encourage you to reach out to us about content you’d like us to discuss in our next issues. See you next month!

Further Reading

Telomere-to-mitochondria signalling by ZBP1 mediates replicative crisis

A hyper-quiescent chromatin state formed during aging is reversed by regeneration

Compartmentalization of the SUMO/RNF4 pathway by SLX4 drives DNA repair

Defining regorafenib as a senomorphic drug: therapeutic potential in the age-related lung disease emphysema

Rhythms in barriers and fluids: Circadian clock regulation in the aging neurovascular unit

Effects of intermittent fasting on cognitive health and Alzheimer’s disease

Brain insulin responsiveness is linked to age and peripheral insulin sensitivity

Contextual modifiers of healthspan, lifespan, and epigenome in mice under chronic social stress

Flow Cytometry-based Method for Efficient Sorting of Senescent Cells

The multi-tissue landscape of somatic mtDNA mutations indicates tissue-specific accumulation and removal in aging

A p21-GFP zebrafish model of senescence for rapid testing of senolytics in vivo

The effects of caloric restriction on adipose tissue and metabolic health are sex- and age-dependent

Reviews

Exploring the Causal Relationship Between Telomere Biology and Alzheimer’s Disease

Aged brain and neuroimmune responses to COVID-19: post-acute sequelae and modulatory effects of behavioral and nutritional interventions

Gene Therapy Strategies Targeting Aging-Related Diseases

Fungi as a source of bioactive molecules for the development of longevity medicines

Obesity: an evolutionary context

Cellular senescence in skin-related research: Targeted signaling pathways and naturally occurring therapeutic agents

To keep up-to-date with the latest breakthroughs in longevity research, exclusive insights, and exciting updates from our team, make sure to subscribe to our newsletter.

We are thrilled to celebrate our one-year anniversary of bringing you the latest research, insights, and developments in our field. Over the past year, we have covered a wide range of topics, from artificial intelligence and sustainability to genetic
Read more
VitaDAO Letter: Prizes and Research!
May 9, 2023
Sarah Friday
Awareness
VitaDAO Letter: Prizes and Research!

VitaDAO Letter: Prizes and Research!

Welcome back to the VitaDAO Newsletter, your monthly source of insights into the VitaDAO community. Inside this Newsletter, find:

  • Zuzalu: pop-up city community
  • A recap of VitaDAO’s 3rd DeSci & Longevity Symposium
  • Info on VitaDAO’s job openings
  • Summary of VitaDAO’s recent proposals
  • Meet DAO member: Laurence Ion

🚨Anotha One: A 3rd Research Symposium

An event so good, we did it a third time! On April 5th, Max Unfried and Eleanor Sheekey hosted a third VitaDAO research symposium. Garnering over 700 viewers, the event was a day packed with education and learning about the future of rejuvenation therapies and longevity. Speakers from around the world provided valuable insights into the fascinating world of aging, including talks on reproductive aging, AI and aging, and longevity drug discovery. Notable speakers included Jackie Han, Zhongwei Huang, Kamil Pabis, Dr. Arturo Bujarrabal, Prof. Karsten-Henrich Weylandt, Vadim Gladyshev, and Aubrey de Grey.

🏆Hypothesis Prize Winners

The Longevity Prize is a DeSci initiative aiming to accelerate research efforts. Coordinated by VitaDAO, Foresight Institute, Methuselah Foundation, and Lifespan.io, the Longevity Prize aims to highlight and accelerate progress in longevity. The first prize, the Hypothesis Prize, was awarded to individuals who formulated a hypothesis in an undervalued area of longevity. First-place winner, Carlos Galicia, proposed the study of rejuvenation during embryogenesis as a solution to age-related decline. Second-place winner, Rakhan Aimbetov, proposed the study of proteostasis disruption in age-related diseases. Third-place winner, Shahaf Peleg, proposed the exploration of mitochondrial dysfunction in aging through the use of a novel approach of external energy replacement using mtON technology. You can watch a recording of the Award Ceremony, here.

The Longevity Prize is a crowdsourced prize funded through the Gitcoin community and matched by Vitalik Buterin to fund longevity research projects. Want to learn more? Listen to Allison Deuttman, president of Foresign Institute, and Eleanor Sheeky discuss this ecosystem generator.

🌎Zuzalu & Creating A Longevity Network State

Zuzalu is a first-of-its-kind pop-up city community initiated by Vitalik Buterin & frens in Montenegro, from March 25 to May 25, 2023. Taking place in Montenegro, Zuzalu has brought 200 core residents brought together through a shared desire to learn, create, live longer and healthier lives, and build self-sustaining communities.

VitaDAO is supporting every aspect of longevity at Zuzalu, including various events on biotech innovation and jurisdictions for longevity, as we’re exploring evolving VitaDAO towards a Longevity Network State, and building a special jurisdiction for medical innovation with a better regulatory framework.

We will dive into how biotech innovation can bring aging under medical control, and how new cities and states can enable better regulatory pathways for innovating in longevity therapeutics.

During the first week of Zuzalu, we introduced residents to longevity, starting with the lifestyle aspects, then an overview of how we can really bring aging under medical control, with biotech innovation and special jurisdictions with better regulations We then delved deeper into why Zuzalu is exciting to our mission — building new cities, jurisdictions, and states for longevity and medical innovation, exploring how people congregating physically can unite & foster groundbreaking advancements in healthcare and human lifespan.

And then we got to learn from many founders of new cities and network states — while exploring the intersection of longevity and network states on a panel with Adam Gries, Balaji, Laurence Ion and Niklas Anzinger

Sad you’re unable to join Vitalik and many others in person? Watch sessions live through the Zuzalu Livestream.

🧪The Longevist: A Curation of Longevity Research

Drum roll please🥁…. introducing The Longevist, an overlay journal curating the most impactful longevity research every quarter. You might be wondering to yourself, how exactly does this journal work? Each quarter, a mix of industry and academic experts in the longevity space vote on preprints to feature. With a focus on quality, The Longevist aims to select 10–20 articles a year, showcasing top articles from servers like BioRxiv, MedRxiv, and Arxiv.

This month, The Longevity launched 2023 Q1 voting. With over 20 voters, the top three selections were more than clear:

  1. Gene Therapy Mediated Partial Reprogramming Extends Lifespan and Reverses Age-Related Changes in Aged Mice
  2. p16-dependent upregulation of PD-L1 impairs immunosurveillance of senescent cells
  3. Multidimensional proteomics identifies molecular trajectories of cellular aging and rejuvenation

Want to join in on the action? Help fund the dissemination of longevity research. Connect with tim@vitadao.com or rhys@vitadao.com to vote, build, or contribute.

🗣️VitaDAO in the Wild

- Laurence Ion and Vincent Weisser joined the Epicenter Podcast to speak about the purpose of DeSci, the advantages of DeSci, and ways VitaDAO is accelerating the space.

- Coindesk wrote about VitaDAO’s new initiative, VitaTech, a for-profit company that will be created to secure and distribute funding for longevity research.

-VitaDAO Governance Lead, Gavin Singh, spoke at DeSci Tokyo about governance in the DeSci space.

🏛️VitaDAO Governance: Season 0 of Governance Recap

Earlier this year, the VitaDAO community voted to improve the governance process by “batching” governance into seasons. Expanding from February 20th to April 3, the VitaDAO community had its first round of seasonal governance: what a learning experience! It was exciting to see community engagement when discussing governance goals and during the creation of proposals.

Season 1 aimed to pass proposals that focused on increasing community size, increasing the number of token holders, accelerating research assets, and increasing researcher engagement. It is with excitement to share four proposals passed as part of VitaDAO’s first season of governance:

  • SG 0.3a: This proposal will establish the Commercialization Squad, a group of individuals whose goals are to increase the $VITA token utility value and commercialize VitaDAO’s assets.
  • SG 0.3c: This proposal will establish VitaTech, a US-based for-profit company to license longevity technologies from US universities and research institutions and raise non-dilutive funding to commercialize assets.
  • SG 0.3b: This proposal approved Molecule’s offer to pay for IP services in sweat equity as opposed to cash.
  • SG 0.1a: This proposal defined the creation of an Ambassador Program, a collection of people appointed to be Ambassadors and perform outreach on behalf of the DAO.

📣Community Approved: What you voted for!

VDP-63 Passed! VitaDAO and the Gorbunova Lab are launching Matrix Bio, a cutting-edge research venture leveraging the anti-cancer and pro-longevity effects of high molecular weight hyaluronic acid from naked mole rats to humans. VitaDAO will fund up to 300k to support the screening and development of small molecule inhibitors of hyaluronidases.

VDP-74 Passed! This vote was an assessment of Etheros Pharma, a preclinical biotech that is pioneering a new small-molecule drug class to extend mammalian lifespan and neural healthspan.

VDP-80 Passed! This vote was an assessment of ImmuneAGE, the first drug discovery platform for immune system rejuvenation. ImmuneAGE is developing compounds that target the hematopoietic stem cells (HSCs) of the bone marrow.

VDP-85 Passed! This vote was an assessment of HDAX, a preclinical-stage drug discovery company targeting a class of proteins implicated in neuropathies and inflammation.

VDP-86 Passed! This vote was an assessment of Humanity, an app that uses wearable technology and quantified-self methods to measure one’s rate of aging.

VDP-94 Passed! This vote re-allocates the remaining tokens already minted under VDP-11 and deploys them in partial satisfaction of VDP-72 obligations; to defer minting more tokens in the near-term.

💪Exercise Your Right to Vote on VitaDAO’s Governance Forum:

Find all of VitaDAO’s pending live proposals on Discourse, VitaDAO’s governance hub for proposals before they are moved on Snapshot. These proposals are open for engagement, voting, questions, and conversation!

💸We’re Hiring!

VitaDAO has six job openings! If you are a well-organized and dynamic person, curious about the world of longevity, crypto, and open-source communities, then we’re looking for you! Open roles include Legal Lead, Onboarding Lead, Governance Lead, Community Moderator, Company Builder, and Operations Champion. Come get rewarded with competitive compensation while simultaneously helping shape the future of VitaDAO!

🧑‍💻Ch-ch-ch-changes

Haven’t noticed? VitaDAO has made new changes to its website: https://www.vitadao.com/. Find updated descriptions of projected funded by VitaDAO and even explore VitaDAO’s new treasury dashboard.

👋Meet Longevity Dealflow Steward: Laurence Ion

Laurence Ion is an entrepreneur and investor working to “rejuvenate” the biotech industry. With a tech background and a keen passion for the scientific method, he’s helping translate science into startups, accelerating progress in medical innovation, on a mission to bring aging under medical control. He now serves as Steward of the Longevity Dealflow Working Group and as Director of Vitality Healthspan Foundation, a nonprofit organization that supports healthspan research.

Why longevity?

I am working in service of life (vita, in Latin), helping end aging.

Because I love life, it makes sense to tackle aging, the biggest hindrance to life (and the root cause of all age-related, chronic diseases).

Unlike most young people, I know what it’s like to lose my health and VITAlity (ok I’ll stop with the Vita puns). I know what it’s like to feel frail, to feel old — I’ve spent a lot of time in hospitals, I’ve had many surgeries.

How did you first get involved in the longevity space?

While I’ve had a big passion for biology and biomedical innovation, I was disillusioned with the current medical approach, so I was in tech entrepreneurship and investing, but about 5 years ago I decided to switch to the longevity biotech industry because progress was slower than expected.

I started as an angel investor, syndicating and working on a longevity VC fund. I realized that the bottleneck in the field is the lack of diversity of startups. Capital was needed earlier, to create more startups.

You were involved in VitaDAO from the earliest stages. How did you meet the team and what drew you to VitaDAO?

I had been growing my involvement in the crypto space since 2013 and was inspired by Griff Green to use the power of decentralized communities and cryptoeconomic incentives to fund longevity labs, especially given the alignment between the crypto and longevity fields.

When I came across the IPNFT framework, enabling DAOs to hold real-world IP assets, I reached out to Tyler and decided to drop everything and help in any way I can, because I wanted to fund early-stage research and accelerate it to commercialization, creating startups that wouldn’t otherwise exist, bringing that much-needed diversity, as well as scalable democratic participation.

What is the greatest challenge of being VitaDAO’s Longevity Dealflow Steward?

Keeping up with all the amazing things going on and all the requests from members, while also finding time to focus and innovate, myself.

So far, we, the VitaDAO community, have demonstrated we can act at a world-class level, with impressive, scalable, collaborative, deal flow, deploying $4M+ into 17+ projects, and bringing on the most fitting Strategic Members, including big pharma, crypto VCs, angels. This, while keeping things decentralized, requires tremendous coordination, but it’s worth it! I think that being a Steward in VitaDAO is the best way to achieve my mission of serving life… and it’s fun! Otherwise, I wouldn’t have dedicated my life to this, doing what was needed, as well as flying & living wherever needed.

While I don’t care about the title, people need to refer to me somehow, and I’m usually doing whatever is most important for VitaDAO at any point, but making sure deal flow runs smoothly while also empowering members and keeping it non-hierarchical, is definitely a challenge.

What does the future of VitaDAO look like?

Growing this highly-aligned community, a lot, with improving our capacity for action, such that we are scaling enough that the community has significant force. And with scale, as well as physical presence (special economic zones/jurisdictions/new cities, states, etc) the will of the people can be much more effective.

We can become as big and resourceful as a country. We can remove any barrier and achieve progress much faster than people think.

Any closing thoughts for readers?

Don’t hesitate to reach out, join us on Discord, hang out, and you’ll figure out how you can add value and potentially have the most significant role in VitaDAO. Think outside the box, be resourceful!

🗓️Upcoming Event

VitaDAO @ Longevity Med Summit — May 4–5, 2023- Come join VitaDAO in sunny Lisbon, Portugal at the 2023 Longevity Med Summit. Here, join other longevity enthusiasts, in learning about the development of anti-aging science, the exploration of new regenerative and longevity therapeutics, and the limiting R&D challenges faced in therapy development.

Longevity Zero to One @Zuzalu May 7–10 — Dive into the science, get career guidance from experts, and create your impact roadmap.

Longevity Biotech Conference @Zuzalu — May 12–14 — Discover breakthroughs in rejuvenation & anti-aging biotech.

🤝Get Involved

Have a special skill set? Contribute to VitaDAO and receive $VITA in return. Join the VitaDAO community on Discord and stay up to date on Twitter! As of February, VitaDAO’s Discord is now token-gated, requiring a minimum of 10 $VITA to gain access to basic contributor channels.

Inside this Newsletter, find: - Zuzalu: pop-up city community - A recap of VitaDAO’s 3rd DeSci & Longevity Symposium - Info on VitaDAO’s job openings - Summary of VitaDAO’s recent proposals - Meet DAO member: Laurence Ion
Read more
Beginners Guide to the VITA Token
April 18, 2023
Tokenomics
Beginners Guide to the VITA Token

Holding VITA tokens is an important step if you are looking to become an active contributor to VitaDAO's governance. VITA is the governance token our contributors use to vote on longevity research proposals, and direct the overall course of the DAO. 

VITA holders have voting rights and are active stakeholders that determine how the DAO advances its longevity mission. We'll be walking you through all you need to know about the VITA token, how you can get some, and how you can begin contributing to our governance processes today!

Understanding the VITA Token and Its Purpose

VITA is an ERC-20 token. If you're not familiar with blockchain lingo, ERC-20 is simply a standard for tokens built on top of the Ethereum blockchain. The token is available both on the Ethereum network and the Gnosis chain (which is a sidechain built on the Ethereum blockchain). 

As a governance token, VITA is used for voting purposes within the DAO. This includes decisions around what projects are funded, how they are funded, treasury management decisions, and other governance activities that help improve the DAO's processes. VITA holders vote based on the number of tokens held in their wallets. In other words, the more tokens held in a wallet, the weightier the vote from that wallet.

The supply of VITA is currently capped at 64,298,880 VITA. This represents the number of minutes lived by Jeanne Louis Calment (the longest-lived person in history). The supply cap may only increase if someone lives longer than that. At Genesis, 70% of this supply was left unminted in the DAO's treasury while the remaining 30% was allocated to the community.

While ownership of VITA grants access to governance processes, it does not give holders ownership rights to any of the IPs held by VitaDAO. However, VITA holders can determine how the IP is monetized and other decisions around its usage and application.

How to Get VITA Token

If you are new to cryptocurrency and you'd like to purchase VITA, here's a quick run-down with a brief explanation on how you can get started:

Create a Wallet

We recommend you use MetaMask, which is a popular wallet option for Ethereum and Gnosis networks. You can add the extension to your web browser on your personal computer. If you're more likely to interact on your mobile device, consider downloading the app on your play store or app store.

Buy Ether

Since the VITA token runs on the Ethereum network, you'll need to buy some ETH to be able to pay transaction fees on the network and swap for VITA. Note, VITA can only be purchased (swapped) with other cryptocurrencies. If you are on the mobile app, you can buy ETH directly on MetaMask using your credit card or other payment options available. On the browser extension for PC, you'll be directed to MetaMask's supported providers like Coinbase to buy cryptocurrencies. 

Alternatively, you can buy ETH on a centralized exchange and send it to your wallet address on MetaMask. If you don't want to go through the route of a centralized exchange, Ramp provides the option of buying ETH directly to your wallet address by paying with your fiat currency using available payment methods.

Go Ahead to Buy VITA

Now that you've got some ETH, you can go ahead and buy VITA on supported decentralized exchanges. Here's how it works: you have to swap one token for the other in order to buy VITA on these decentralized exchanges (DEXs). The most popular would be to swap ETH for VITA. A step-by-step process to do this is as follows:

  • Buy the ETH equivalent of VITA using any of the methods earlier described
  • If you are using the mobile app, open the dApp browser on the app, go to any of the DEXs listed below, and connect your wallet
  • If you are on your PC, simply go to any of the DEXs below on your internet browser and connect your wallet 
  • After connecting your wallet, swap ETH for VITA on the decentralized exchange.

You can use any other supported cryptocurrency on the DEX to swap for VITA instead of ETH. Here is a list of several decentralized exchanges where you can easily buy VITA: 

Alternative Ways to Get VITA

Besides the option of buying, another way to get VITA is to earn the token through your contributions to the DAO. Contributors at the different working groups are compensated with VITA tokens. If you are looking to earn some, hop into the Discord channel and try to get plugged in to see how you can use your skills to help drive progress at the DAO. 

VITA Token in VitaDAO Governance

Governance at VitaDAO happens in two forms: Soft Governance, which involves governance decisions made without an on-chain vote; and on-chain governance, where on-chain voting is required to make decisions. Either of these governance mechanisms is used depending on the type and impact of decisions to be made. 

Governance processes are based on three phases. Here's an outlook of how decisions are made and how we decide which of the governance forms are used in decision-making:

Phase 1: Idea

This is where we receive proposal ideas for consideration. Anyone is invited to propose an idea. If an idea receives positive support from five or more people, it moves to phase 2.

Phase 2: Proposal Development and Approval

Here, the idea is shaped into a proposal using a defined template. Based on the funds requested, the proposal is either passed via a soft governance approach or on-chain voting. Here's how we decide that:

  • A proposal requesting up to 2,500 USD only requires approval from two working group representatives.
  • If the proposal requests up to 10,000 USD, it will require approval from a majority of all the working group stewards.
  • For proposals that ask for up to 50,000 USD, a public Discourse poll is set up and a majority vote is required for approval.
  • Proposals with required funds greater than 50,000 USD require a majority vote on a public Discourse poll and it is moved to phase 3.

Phase 3: On-chain Voting

The following are the kinds of decisions that require an on-chain vote for approval:

  • Upon having a majority vote on a public Discourse poll, proposals with a ask greater than 50,000 USD are moved to Snapshot for on-chain voting. Here, VITA holders carry out gasless (without transaction fees) voting. Proposals must achieve a quorum and a majority vote before they are approved.
  • Proposals that involve the operating rules of the DAO, changes to such rules, membership, and enforcement of norms all require on-chain voting before they can be passed. This is done regardless of the proposal's budgetary threshold.

How to Vote on Snapshot Using Your VITA Token

When proposals are posted on Snapshot, VITA holders are required to vote on those proposals to determine whether they will be approved or not. To vote on Snapshot, click the voting link and connect your wallet.

Having connected your wallet, you can go ahead to vote on the current proposal. VitaDAO uses shielded voting on Snapshot so everyone can make their choice without being influenced by current voting results. 

Conclusion

We are democratizing longevity research through decentralized and community-governed processes. VITA token holders are an essential part of these governance processes as they help steer the course of progress at the DAO. Be a part of the voices that determine how VitaDAO is run by becoming a token holder today.

Holding VITA tokens is an important step if you are looking to become an active contributor to VitaDAO's governance. VITA is the governance token our contributors use to vote on longevity research proposals, and direct the overall course of the DAO.
Read more
‍Unlocking Longevity: Autophagy, Mitochondria, and The Radical Pursuit of Life Extension during The Aging Science podcast by VitaDAO
April 13, 2023
Awareness
Longevity
‍Unlocking Longevity: Autophagy, Mitochondria, and The Radical Pursuit of Life Extension during The Aging Science podcast by VitaDAO

Check out the podcast here. 

I (@aging_scientist) had a marvelous conversation with Assoc. Prof. Viktor Korolchuk (@VIKorolchuk) about autophagy, mitochondria, senescence, the importance of modelling and human desire for radical lifespan extension. Originally from Ukraine where he received his PhD, Viktor worked in Cambridge as a postdoctoral fellow and later moved to Newcastle upon Tyne in the beautiful north of the UK to pursue his work on aging.

He has received a grant from VitaDAO to identify novel autophagy activators against aging and lysosomal storage disorders.
https://www.vitadao.com/projects/korolchuk-lab

Lab page: https://www.ncl.ac.uk/medical-sciences/people/profile/viktorkorolchuk.html

Below we will review some of the concepts we discussed in the podcast.

Mitochondria – multifaceted players in aging

These organelles are often called the powerhouse of the cell but, apart from being cliched, this metaphor does not do them justice. If we stick to that “urban” metaphor, the mitochondria are maybe like a manufacturing plant, a port, a power plant and a minor geopolitical center all in one. Given the complex responsibilities of mitochondria it should not come as a surprise that they have diverse failure modes during aging.

For example, during aging we see clonal expansion of point mutations and deletions which leads to tissue mosaicism, meaning that one and the same tissue ends up having stretches of totally different, often more or less dysfunctional, mitochondrial populations.

There is also the classic mitochondrial (mt) free radical theory of aging. In one more modern interpretation of this theory, production of radical oxygen species (ROS) at mitochondria is a constant source of macromolecular damage irrespectively of whether this mtROS production increases with aging or not. This is consistent with and supported by findings of reduced mtROS production (Lambert et al. 2007) and higher membrane lipid stability in long-lived species.

In some aging tissues we observe reduced mitochondrial mass and decreased mitochondrial biogenesis (1). Based on this we would expect increased mitochondrial biogenesis to improve healthspan or lifespan, and, indeed, PGC1a is consistently upregulated in long-lived rodents and overexpression of this gene extends fly lifespan (Tyshkovskiy et al. 2019, Ozkurede et al. 2019, Rera et al. 2011).

Although we do observe decreased mitochondrial mass in some tissues, in other tissues there may be a vicious cycle of defective mitochondria that proliferate out of hand. The latter is a theory which is bolstered by the work of Aiken and McKenzie (Herbst et al. 2022) as well as work by Prof Doug Turnbull from Newcastle (Vincent et al. 2018).

Some other theories suggest that aging mitochondria lead to increased susceptibility to cell death, and that the mitochondrial network becomes disorganized with impaired fusion-fission dynamics.

And this is just a small sample of mitochondrial aging theories!

Rapamycin, mTOR and its effects on aging

Rapamycin is an approved immunosuppressant drug that is produced by a type of bacteria from the Streptomyces genus. The drug was first isolated from a soil sample found at the island of Rapa Nui hence the name.

Most of its effects are mediated via inhibition of the mTOR pathway. Generally speaking, this pathway is turned on during cellular growth when nutrients are abundant and need to be utilized for growth, and it is turned off during starvation when the cell needs to focus on survival, efficiency and recycling.

This relatively obscure drug has become the superstar and a major workhorse in biogerontology because it extends mouse lifespan and also leads to many physiologic changes that appear beneficial. 

These include increased autophagy, reduction of the senescence associated secretory phenotype (SASP), reduced cell growth and protein synthesis etc. Interestingly, rapamycin is not primarily senolytic, i.e. it does not kill senescent cells, rather it reprograms them to be less harmful. It remains a matter of debate which downstream effects of rapamycin are responsible for the lifespan benefits. Nonetheless, the efficacy of rapamycin does provide some evidence for the importance of autophagy during aging.

Senescence

Senescence is an irreversible non-proliferative state. This means senescent cells exit the cell cycle and stop growing, instead they secrete cytokines and growth factors, many of which are harmful. Senescence is considered anti-tumorigenic and senescent cells are involved in wound healing and possibly other physiologic processes like insulin secretion or structural integrity of tissues. Perhaps some of these secreted growth factors serve a purpose and therefore the removal of senescent cells poses a health risk.

However, given that ablation of senescent cells improves healthspan and extends lifespan in mice this suggests that, on balance, these cells are harmful. This alone does not explain the hype around senescence and senolytics (therapies that remove senescent cells).

The topic of senescence has received a lot of attention in the field, specifically because a single ablation treatment in mid-life can have lasting health benefits in mice. This is basically a dream therapy come true if it translates to humans. One time treatments are cheap and safer than chronic treatments, making them perfect for clinical use.

Senescence can be triggered by DNA damage, oxidative stress, telomere erosion, viral infection and many other stressors. This links senescence as a kind of downstream “damage” with many modern aging theories, like the stress resistance theory of aging, for example.

Autophagy and aging

"some of the data with these [autophagy-inducing] molecules looks almost magical" (Viktor Korolchuk)

The word autophagy describes a process of self-eating and self-recycling that almost every cell in the body utilizes. This process is necessary for a cell in order to adapt to changing environmental conditions. Organelles and proteins that are not needed in a given situation are broken down into their components, amino acids or lipids, for example, which are used to assemble whatever the cell needs at that moment. Another key function of autophagy is quality control and maintenance through the removal of damaged and dysfunctional organelles or proteins. 

Especially during starvation autophagy is needed to fuel essential cellular processes and to use cellular proteins for the production of energy (ATP).

We can distinguish several types of autophagy based on their molecular characteristics. The three key types are macroautophagy, microautophagy and chaperone mediated autophagy (CMA). As the name suggests macroautophagy degrades large, bulky structures whereas the other two do not. Chaperone mediate autophagy uses so called chaperones to help unfold proteins and import them into the lysosome, where they will be degraded. There is also crosstalk between the different types which makes it hard to study them individually due to compensatory mechanisms.

Macroautophagy has attracted a lot of attention because it is easy to measure and may remove damaged organelles and proteins believed to accumulate with aging. Macroautophagy itself is also subdivided into bulk and selective autophagy, the latter includes the autophagic degradation of organelles like mitochondria (mitophagy), peroxisomes (pexophagy), etc.

We have a lot of evidence which suggests that increased autophagy can delay aging. For example, there are comparative studies showing that cells from genetically longer-lived mice show more prolific autophagy. There is also a hint of increased autophagy in long-lived species more broadly.

The most specific and direct evidence comes from invertebrates, however, where it is possible to show that autophagy genes are necessary for the effects of caloric restriction. These things are always harder to show in mammals. However, we do have several genetic gain-of-function studies in mice suggesting that increased autophagy does indeed extend lifespan (e.g. Fernández et al. 2018), although they have their own limitations.

Other than that, there is a lot of indirect evidence. We know that many lifespan extending strategies in mice have autophagy in common, e.g. rapamycin and caloric restriction both induce autophagy.

Interestingly, autophagy deficient mice die early during life, whereas autophagy deficient cells appear fine at first glance. However, Viktor mentions that they have problems when they are forced to respire and utilize their mitochondria, which is when they start dying. This is why, in his VitaDAO project, he can use a viability assay in autophagy deficient cells to find novel autophagy inducers.

The philosophy of lifespan extension

Towards the end of the podcast, we had an interesting foray into the important question of “how long would you want to live?” and both Viktor and I agreed that: "an average person...would choose to live indefinitely if they were in good health, mental and physical".

However, this question is very sensitive to the phrasing and there are certainly people who would like to choose differently. If you are interested in understanding the psychology and philosophy behind this question, I have written extensively on this topic before and one of my recent blog posts might be a good primer for this kind of debate.
How long do you want to live? (June 22, 2022)
http://biogerontolgy.blogspot.com/2022/06/how-long-do-you-want-to-live.html

Important Reading (selected references)

Herbst, Allen, et al. "Age-and time-dependent mitochondrial genotoxic and myopathic effects of beta-guanidinopropionic acid, a creatine analog, on rodent skeletal muscles." Geroscience (2022): 1-13.

Ozkurede, Ulas, and Richard A. Miller. "Improved mitochondrial stress response in long‐lived Snell dwarf mice." Aging Cell 18.6 (2019): e13030.

Tyshkovskiy, Alexander, et al. "Identification and application of gene expression signatures associated with lifespan extension." Cell metabolism 30.3 (2019): 573-593. webpage: http://gladyshevlab.org:3838/Gentervention/

Rera, Michael, et al. "Modulation of longevity and tissue homeostasis by the Drosophila PGC-1 homolog." Cell metabolism 14.5 (2011): 623-634.

Gonzalez-Freire, Marta, et al. "Reconsidering the role of mitochondria in aging." Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences 70.11 (2015): 1334-1342.

Lambert, Adrian J., et al. "Low rates of hydrogen peroxide production by isolated heart mitochondria associate with long maximum lifespan in vertebrate homeotherms." Aging cell 6.5 (2007): 607-618.

Fernández, Álvaro F., et al. "Disruption of the beclin 1–BCL2 autophagy regulatory complex promotes longevity in mice." Nature 558.7708 (2018): 136-140.

Vincent, Amy E., et al. "Subcellular origin of mitochondrial DNA deletions in human skeletal muscle." Annals of neurology 84.2 (2018): 289-301.

(1) Regarding reduced mitochondrial biogenesis and mitochondrial mass during aging: I remain somewhat unconvinced by this aging theory, although I have not looked at this in much detail. If decreased biogenesis was a main driver of aging it should occur in both sedentary and exercised individuals, since both of them do show muscle aging, but this is not the case:

“However, it has become increasingly clear that most of the declines in mitochondrial biogenesis, turnover, and function are a consequence of physical inactivity. Indeed, when physical activity levels are matched between young and elderly people, or physical activity is otherwise taken into account, most investigations do not find any age-related changes in mitochondrial enzyme activities, mitochondrial respiration, or ATP flux” (Barbieri et al. 2017)

On the other hand, given this evidence, it appears that aging increases the susceptibility to muscle loss, including reductions in mitochondrial mass and biogenesis. This by itself does appear to be a proper aging phenotype -- consistent with the concept of reduced resilience during aging. 

I (@aging_scientist) had a marvelous conversation with Assoc. Prof. Viktor Korolchuk (@VIKorolchuk) about autophagy, mitochondria, senescence, the importance of modelling, and human desire for radical lifespan extension.
Read more
March Longevity Research Newsletter
April 9, 2023
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
March Longevity Research Newsletter

March Longevity Research Newsletter

Introduction

Welcome back Vitalians!

In recent years, autophagy has emerged as an important topic in the field of aging biology and is now considered a major Hallmark of Aging.

Autophagy is a fundamental process that plays a critical role in the maintenance of cellular homeostasis. This process is involved in the degradation and recycling of cellular components, including damaged organelles, misfolded proteins, and intracellular pathogens.

Autophagy has been shown to become dysregulated with age, contributing to the aging process by impairing cellular homeostasis and increasing cellular damage. This is thought to play a role in the pathogenesis of several diseases, including cancer, neurodegeneration, and metabolic disorders. 

Understanding the role of autophagy in aging biology has important implications for the development of interventions to promote healthy aging. By enhancing autophagy, it may be possible to improve cellular function and delay the onset of age-related diseases. This has led to growing interest in the development of drugs and other interventions that target autophagy as a means of promoting healthy aging. 

VitaDAO are supporting autophagy research in several ways, including funding Dr. Viktor Korolchuk's lab to pursue the discovery of novel autophagy activating drugs. We had the pleasure of interviewing Dr. Korolchuk for this month’s newsletter - so don’t miss this insightful interview which touches on autophagy drug development potential, new theories of aging and the state of the longevity field.

Longevity Literature Hot Picks

Preprint Corner

This month we are featuring 7 new preprints which are all available to review on our reviewing platform The Longevity Decentralised Review (TLDR) in return for a bounty of 50 $VITA each. Simply follow the above link to the TLDR page and get reviewing! What's more, we will be continuing the 50 $VITA bounty for reviewing any of the preprints featured in our January and February research newsletters.

Multidimensional proteomics identifies molecular trajectories of cellular aging and rejuvenation

Modelling the dynamics of senescence spread

The intensities of canonical senescence biomarkers integrate the duration of cell-cycle withdrawal

Stochasticity explains non-genetic inheritance of lifespan and apparent trade-offs between reproduction and ageing

Mitochondrial haplotype and mito-nuclear matching drive somatic mutation and selection through aging

Heterogeneous aging across multiple organ systems and prediction of chronic disease and mortality

Mitochondrial H2O2 release does not directly cause genomic DNA damage

Published Research Papers 

The DREAM complex functions as conserved master regulator of somatic DNA-repair capacities

Björn Schumacher’s team have discovered the first master regulator of somatic DNA repair capacities - The DREAM complex. This complex transcriptionally represses essentially all DNA-repair systems and inhibiting this can lead to improved DNA damage repair and resistance to various types of genotoxic insults.

Centenarian clocks: epigenetic clocks for validating claims of exceptional longevity

Horvath’s team develops a centenarian epigenetic clock to validate claims around exceptionally old age based on samples from centenarians, semi-supercentenarians (aged 105 +), and supercentenarians (+110, the oldest person being 115).

TFEB-ependent lysosome biogenesis is required for senescence

Cellular senescence is associated with dysfunctional lysosomes which the cell compensates for by undergoing lysosomal biogenesis. Here the Carroll lab show that upon senescence induction TFEB accumulates in the nucleus and regulates lysosomal biogenesis. Furthermore, inhibiting this process results in the premature death of senescent cells due to diminished degradative ability as a result of impaired lysosomes and inability to produce more to compensate. 

Long-term intensive endurance exercise training is associated to reduced markers of cellular senescence in the colon mucosa of older adults

Senescence markers are high in middle aged and older overweight adults than young sedentary people, but this was significantly reduced in aged matched runners suggesting that high volume high intensity endurance exercise prevents senescent cell accumulation. The resulting decrease in senescence associated inflammation is especially important in cancer prone tissues such as the colon mucosa.

Aging disrupts MANF-mediated immune modulation during skeletal muscle regeneration

Skeletal muscle regeneration is disrupted with age. An immune modulator MANF is responsible for muscle injury repair in young animals but is not induced in old ones, thus impairing regeneration. Restoring MANF levels could be a viable strategy to preserve skeletal muscle regenerative function with age.

Four anti-aging drugs and calorie-restricted diet produce parallel effects in fat, brain, muscle, macrophages, and plasma of young mice

The Miller lab previously published that genetic mouse models of slow ageing show similar changes in a number of pathways. They now observe similar changes with 4 lifespan-extending drugs and calorie restriction. Changes include an increase in anti-inflammatory macrophages and decrease in inflammatory macrophages, and differential expression of numerous proteins involved in age-related diseases such neurodegenerative, metabolic and inflammatory diseases. 

Heterochronic parabiosis reprograms the mouse brain transcriptome by shifting aging signatures in multiple cell types

Single cell transcriptomic profiling was employed to measure changes in young and old mice brains after parabiosis. Analysis showed that multiple hallmarks of aging in the brain were regulated by heterochronic parabiosis in a cell type specific manner.

Habitual daily intake of a sweet and fatty snack modulates reward processing in humans

Continued consumption of high fat, high sugar snacks in normal weight individuals resulted in decreased desire for low fat, low sugar food. The changes were independent of body weight and metabolic alterations.

Sex differences in adult lifespan and aging rate across mammals: a test of the ‘Mother Curse hypothesis’

The Mother's Curse hypothesis suggests that as mitochondria are maternally inherited, there is no selective pressure to remove mitochondrial genome mutations which are exclusively deleterious to males. Here the authors study over a hundred mammal population across 104 species and concludes that Mother's Curse hypothesis doesn't affect mammalian ageing rated between male and females

Published Literature Reviews

The autophagy–NAD axis in longevity and disease

Ageing as a software design flaw

Impact of Cellular Senescence on Cellular Clocks

BCL-2 proteins in senescence: beyond a simple target for senolysis?

Cholesterol drives inflammatory senescence

Cellular Senescence: beneficial, harmful and highly complex

An aging, pathology burden, and glial senescence build-up hypothesis for late onset Alzheimer’s disease

Putting aging on ICE

AI Technology for Anti-Aging: an Overview

Job board

Joe Nassour is joining the University of Colorado as an Assistant Professor in summer 2023! His lab will study how telomeres interact with autophagy and innate immunity to prevent cancer initiation. Reach out to him to explore opportunities to join his team!

Fabrizio d'Adda di Fagagna is looking for a new lab member to join his group working on telomere biology studies in cancer

Research Lab Technician II (Mouse) - Benayoun lab/USC

The Benayoun Lab is seeking a mouse research lab tech II to join the team to help them understand sex differences in aging at USC in southern CA!

Marco Demaria’s lab, working on aging and cellular senescence, has an available postdoc position. ERIBA - European Research Institute for the Biology of Ageing UMCG research

BioAge is hiring a talented Senior Data Scientist and a Research Associated! BioAge’s mission is to deeply analyse human longevity data to discover drug targets for diseases of aging

News

First longevity clinical study design fetches FDA approval

Rubedo Life Sciences Presents In Vivo Efficacy Data Targeting Senescence Cells to Treat Dermatological Diseases

Sam Altman invested $180 million into a company trying to delay death

Regenerative cell biotech Thymmune launches with backing from George Church, John Maraganore

NVIDIA Unveils Large Language Models And Generative AI Service To Advance Life Sciences R&D

Resources

Spannr 2022 Longevity Funding Report

Open Problems in Longevity Science

An online comprehensive compilation of open problems in longevity science for creating a coordinated scientific effort for finding cures for aging. Created by João Pedro de Magalhães, Hamid Hossein Alfatemi and Angelo Talay.

Articles

A ridiculously fluffy rodent may hold the secret to longevity

Who wants to live for ever? Quite a lot of people

Ants Live 10 Times Longer by Altering Their Insulin Responses

Want to live to 150? The world needs more humans.

If they could turn back time: how tech billionaires are trying to reverse the ageing process

Retro: A longevity biotech triple-threat?

Fetal exposure to Great Depression economic hardship linked to accelerated aging

Ageing is a disease, and Christian Angermayer is here to cure it

Are you a rapid ager? Biological age is a better health indicator than the number of years you’ve lived, but it’s tricky to measure

Loyal’s latest milestone: the first longevity clinical study design supported by the FDA

Removing Senescent Cells May Slow, Halt, or Reverse Diseases of Aging

People are desperately trying to live forever. Here are the biggest anti-aging trends sweeping the nation

Four 80-year-old men finished a 100-mile race. Here’s how they did it

Everything Wrong with Mouse Studies (Kinda)

Conferences, Workshops and Webinars

Longevity Med Summit - Lisbon

4-5th May, 2023 in Lisbon, Portugal.

The main goal of Longevity Med Summit is to provide and share knowledge to the medical community aimed at helping patients live longer and delay age-related health issues.

5th World Aging And Rejuvenation Conference

July 17-18, 2023 in Frankfurt, Germany.

Meet the most inspiring speakers and experts gathering to discuss their present research.

Pop-up city - Zuzalu

Longevity Events in May

May 6-9th: Longevity 0-1

May 13-14th: Longevity Industry Talks

Podcasts and Videos

Life Blood: Live Better and Longer with Dr. Charles Brenner

LifeBlood: We talked about how to live better and longer, what NAD is and how it impacts metabolism, how to become more physically resilient, and how to restore damaged cells, with Dr. Charles Brenner, Chief Scientific Advisor at ChromaDex, researcher and healthy aging advocate.   

The Tim Ferriss Show

Interviewing Dr. Peter Attia — The Science and Art of Longevity, Optimizing Protein, Alcohol Rules, Lessons from Glucose Monitoring with CGMs, Boosting Your VO2 Max, Preventing Alzheimer’s Disease, Early Cancer Detection, How to Use DEXA Scans, Nature’s Longevity Drug, and More (#661)

Interview with Prof Viktor Korolchuk

An Associate Professor at Newcastle University, UK, Dr. Viktor Korolchuk received his PhD from the Institute of Biochemistry, National Academy of Sciences, Ukraine, and conducted postdoctoral research at the University of Cambridge, UK. Dr. Korolchuk's laboratory is presently dedicated to investigating the molecular mechanisms of ageing with the aim of discovering interventions that could potentially promote cellular rejuvenation and extend the healthy human lifespan. VitaDAO is proud to support his project on autophagy activators.

What inspired you to enter longevity research?

When choosing the directions for my independent research over ten years ago I considered the biggest remaining questions and challenges in the field of life sciences. Upon extensive reflection I decided that philosophically there is no other problem as important as ageing.  Being able to contribute to our understanding of this process, to delay or prevent it all together, would have the greatest impact on human life, society, and the world as we know it. This was the reason for applying for an academic position at the first and most established Ageing research centre in the UK based at Newcastle University.

Which of the current theories of ageing do you think are the most convincing?

There are a lot of interesting developments in this area. Some are based on the idea of ageing as a programmatic process, e.g. a software design flaw by Joao Pedro de Magalhães (doi: 10.1186/s13059-023-02888-y) or a selective destruction concept by my colleague and friend James Wordsworth (e.g. doi: 10.1016/j.mad.2022.111709). Both could also potentially explain metabolic slowdown with ageing. We see experimental evidence of this age-associated sluggishness at every level of biological complexity, from individual cells isolated from aged human donors to whole organisms. Trying to explain the reasons and significance of these changes is very important but we also should consider that cellular metabolism could in turn affect the gene expression and the ageing clocks. This is where autophagy could also play the role and our working hypothesis that activating autophagy could restart metabolism and may even slow down the ticking of the clock.

However I think that neither programmatic or damage accumulation-based theories alone would be completely sufficient to explain why we become old and die. The obvious truth is that this is a very complex process and will probably require a theory that would integrate multiple mechanisms rather than attempting to use a reductionist approach. Developing a convincing theory is extremely important as it would lay the ground for probably equally complex anti-ageing interventions.

How has the field changed since you started?

Biogerontology is no longer a fringe science. With the increased awareness of the socioeconomic and healthcare problems associated with ageing it became a mainstream field. Ageing research centres sprung around the world and a lot of funding has been channelled into this area. There is excitement for the future at the moment and one can only hope that this trend will continue for the years to come.

What mistakes do you think the longevity field has made?

Working in silos, using the same reductionist approaches that work so well for simpler biological problems, forming and maintaining tribes which protect own ideas/theories. These are common problems in any area of biomedical sciences but particularly damaging for biogerontology which should ultimately explain and cure ageing, the most complex biological process of all.

Other than your own, what do you think have been the biggest/important discoveries in the field?

The first example that comes to mind is cellular senescence, which was previously seen as no more than a curious artefact in a petri dish. Now senescence is recognised as an important driver of ageing and a target for rejuvenation therapies. The idea of rejuvenation by reprogramming has also taken off, although a lot of practical hurdles remain to be solved before we would be able to capitalise on its potential. However, the most dramatic development for me was the rise of longevity biotech, which was brought by the realisation (or a mere hope, time will tell) that ageing is a malleable process and that even the smallest advances in this area could bring a huge benefit for humankind.

What advice would you give to people currently working in longevity research?

Learn everything that we know about human biology, think outside the box, keep an open mind and challenge any established preconceptions.

Which aspect of longevity research do you think requires more attention?

Fundamental studies of biological processes underlying ageing, particularly using human-centric models. Only when we understand why we become old will be able to do something about it.

Is ageing a disease?

If we are ever able to delay or cure human ageing, it will become a disease from the past for generations to come.

How do caloric restriction and intermittent fasting affect cellular autophagy in vivo? Do we have evidence of this process in humans and are benefits evident?

Most of our evidence is based on experiments using animal models. We are still far away from being able to monitor autophagy and its benefits in a living human, particularly in tissues that are not easily accessible for testing. Detrimental effect of impaired autophagy on human health is very clear (e.g. caused by mutations in autophagy-related genes), but the benefit of autophagy upregulation is largely an assumption. This being said, autophagy is probably one of the most promising target for interventions considering its homeostatic and metabolic functions.

What disease indications would autophagy enhancers impact the most? For example if your VitaDAO funded project for discovering novel autophagy activators was successful, what disease model would you use to validate it first?

The choice of initial disease indication is purely practical, based on the cost and the time required to demonstrate safety and efficacy. Rare monogenic diseases such as lysosomal storage disorders could be one potential application as these are associated with an accumulation of damaged cellular components. Activation of autophagy could be beneficial in this situation, and we see evidence of this in neuronal models of these diseases. A range of other age-related diseases could also be targeted depending on the type of pathology and drug pharmacokinetics.

Do you see autophagy activation as a longevity treatment? Is there evidence it helps healthy aging adults (i.e. in the absence of disease), reduces disease risk or lowers any biological age clocks/biomarkers?

It is certainly our hope that autophagy activators will be a part of anti-ageing strategies. As mentioned above, this is largely based on laboratory models where genetic or pharmacological (e.g. spermidine) interventions activating autophagy increase lifespan. There is also ample evidence for anti-ageing (as for example estimated using epigenetic clocks) effects of rapamycin which among other things activates autophagy. It is still early days for human trials of autophagy activators, for example rilmenidine has recently been found to be safe to use in patients with age-related neurodegenerative disease but larger scale randomised controlled trials are now needed. Before we could go into healthy people, we need specific and safe small molecules, which is what we and others are working on.

Tell us about your latest paper on the relationship between NAD+ metabolism and autophagy. What is the key take-away?

The paper is based on over seven years of our effort trying to understand the connection between autophagy and NAD. The key message is that when autophagy (or mitophagy) fails NAD levels also plummet down, which is not a good thing if you want to stay alive. The implications are not only for those specific pathological states where autophagy is impaired, but also for ageing in general. Autophagy is believed to go down with age and according to our model it would also bring down NAD levels thus resulting in a metabolic decline. 

Outro

Thank you for staying with us till the very end and as always we encourage you to reach out to us about content you’d like us to discuss in our next issues. See you next month!

Further Reading

Acute Effects of Coffee Consumption on Health among Ambulatory Adults

The Timing Sequence and Mechanism of Aging in Endocrine Organs

Welcome back Vitalians! In recent years, autophagy has emerged as an important topic in the field of aging biology and is now considered a major Hallmark of Aging.
Read more
VitaDAO Letter: ’Tis the Season (Of Governance)
March 28, 2023
Sarah Friday
Awareness
VitaDAO Letter: ’Tis the Season (Of Governance)

VitaDAO Letter: ’Tis the Season (Of Governance)

Inside this Newsletter:

  • VitaDAO’s Transition to Seasonal Governance
  • Should VitaDAO Form A Longevity Network State?
  • Steward Election Results!
  • Meet DAO Member: Gavin Singh

A Lot Has Happened Since the Last Newsletter!

Early this year, VitaDAO announced a $4.1 million fundraising round, that included strategic members and longevity enthusiasts such as Pfizer Ventures, Shine Capital, L1 Digital, Beaker DAO, Balaji Srinivasan, and Joe Betts-LaCroix. Since this announcement, VitaDAO has been busy! Inside this Newsletter, find a recap on some of the things that have been going DAOn.

DAO News

The Results are In!

Earlier this year, several changes were made to VitaDAO’s governance structure. Most notable was the condensation of VitaDAO’s prior working group structure into three working groups: Longevity Dealflow, Community and Awareness, and Coordination. Following this structural change, VitaDAO voted to elect stewards for each of the three working groups (VDP-78.1, VDP-78.2, VDP-78.3). Elections are held every six months, with future elections to be held in September 2023. Congratulations to VitaDAO’s newest facilitators:

Creating A Longevity Network State?

From March 25th- May 25th, VitaDAO is partaking in a pop-up mini-city experiment in Montenegro. This experimental mini-city, taking place at a resort, will offer mini-conferences, co-working spaces, tracking of biological age, and group exercise- all with a longevity focus! There are inefficiencies within VitaDAO’s current jurisdiction. This experiment is VitaDAO’s first step in exploring the possibilities of a highly aligned community with the capacity for collective action. Interested in learning more? Dig into some of the interesting conversations about the future of network states and the importance of sovereignty taking place on VitaDAO’s #Vita-State Discord Channel. Explore Balaji’s Twitter insight into VitaDAO’s evolution towards a network state. TLDR, he says network states should:

  1. Start with 1–2 month experiments around the world
  2. Recruit curious people
  3. Make sure participant costs are reasonable
  4. Scale if effective

A New Initiative: The Aging Podcast

VitaDAO started a podcast, more specifically, “The Aging Science Podcast”! In the first episode, host Kamil Pabis talks with expert Alaattin Kaya about challenges in research funding and the replication crisis in aging research. Alaattin Kaya, a research investigator who recently started a lab at Virginia Commonwealth University, uses yeast as a model to study the basic mechanisms of aging. When asked about yeast as a model organism: “Of course, yeah, everybody loves yeast.” Listen to the first episode of The Aging Science Podcast to hear about recent breakthroughs in the field, the difficulties of funding risky research, and the importance of overexpression genetic screens in aging research. Read highlights from the podcast on VitaDAO’s blog!

Vita Governance

Approved: Brain Tissue Replacement Therapy

VDP-10 Passed! This proposal was an assessment of BE Corp, a spinout company out of Jean Hebert’s lab at Einstein College of Medicine. Jean Hebert is a leading expert on the subject of brain aging. He is working on engineering transplanted stem cells that can replace damaged brain tissue, of particular use in stroke patients. Senior reviews voiced that this proposal has robust experimental data, a strong founding team, and a reasonable timeline!

Tis the Season(al Governance)

The VitaDAO community voted to improve the governance process by “batching” governance into seasons, each season consisting of a Governance Phase and an Execution Phase. As a result, VitaDAO has now divided operations into seasons, with each season being 4 months long. We are excited to say that from February 20th to April 3, the VitaDAO community is starting its first round of seasonal governance. We are currently voting on goals, creating budgets, and forming project teams. The goals and objectives of Season 1 include increasing community size, increasing the number of token holders, accelerating research assets, and increasing researcher engagement.

To get started in VitaDAO’s Seasonal Governance, visit our discourse for an overview of the process, sign up for an account, and join the conversation! Want to learn more? Watch a recording of a Live Twitter Space hosted by Catthu, Todd White, and Gavin Singh.

Other initiatives within Season 1 include:

  • Creation of an Ambassador Program: A proposal to create a collection of individuals to perform outreach on behalf of VitaDAO. This proposal outlines general criteria for ambassadors, ambassador reimbursement, and goals for ambassador events.
  • Creation of a Commercialization Squad: A proposal to establish a Commercialization Squad within VitaDAO to directly increase token utility value and commercialize VitaDAO’s assets.

Live on VitaDAO’s Governance Forum — discuss and vote!

Find all of VitaDAO’s pending live proposals on Discourse, VitaDAO’s governance hub for proposals before they are moved on-chain. These proposals are open for engagement, voting, questions, and conversation! Below, read through summaries of two proposals currently on VitaDAO’s Discourse:

  • VDP-87 [Assessment] Zoe Biosciences: This is an assessment of an early-stage therapeutic biotech that has two programs centered on targets that are well-validated both for longevity and aging-related diseases. The first program is PAI-1 biologic inhibitors with high target selectivity and affinity, relevant for diseases including elevated FGF23 syndromes and metabolic disorders. The second program is APJ small-molecule agonists for similar age-related indications.
  • VDP-88 Creating VitaTech: This proposes the creation of VitaTech, a US-based for-profit company to license longevity technologies from US universities and research Institutions and, raise non-dilutive funding to develop commercialized assets.

Zuzalu — The first pop-up mini-city experiment 25March — 25May

We’re exploring evolving VitaDAO towards a Longevity Network State, and building a special jurisdiction for medical innovation with a better regulatory framework.

Advances in aging research are being translated into medicine, in clinical trials. We will dive into biotech to bring aging under medical control, and into medical innovation jurisdictions to enable better regulatory pathways for innovating in longevity therapeutics.

Join us to embark on a journey of meaningful dialogue and build lasting connections

If you would like to join the longevity events at Zuzalu, you can expect:
• workshops and initiatives to improve personal longevity
• engage in insightful discussion around biotech and medical innovation through the use of technology
• ideate around creating a new jurisdiction and regulatory environment to foster innovation

Apply to visit: https://vitadao.com/zuzalu

Meet Coordination Working Group Contributor: Gavin Singh

Gavin Singh is an active contributor to VitaDAO’s Coordination Working Group and Governance Squad. Since joining the DAO in August, he has worked to organize VitaDAO’s Discourse and is currently assisting in VitaDAO’s transition to seasonal governance. Outside of VitaDAO, Gavin graduated from McGill University in 2021 and has since worked in private equity as part of an impact-focused family office fund to build communities at several start-ups.

How did you first get involved in web3?

Surprisingly enough, it was my first venture in preventative healthcare right out of university. I was looking for ways to revolutionize the traditional healthcare system. This led me to web3 as it was the values behind the tech that made me realize the possibilities to empower people, not just patients, are endless. That sparked me to delve into DeSci, contribute to the Governance WG in VitaDAO, co-host events and spaces with VitaDAO, DeveloperDAO, and Gitcoin — and before you knew it, I was halfway across the world in Devcon Bogota assimilating with my tribe.

What attracted you to the VitaDAO community?

I was introduced to longevity at a young age. I remember watching a 90s Spider-Man TV series episode called “Neogenic Nightmare”. The villains were fighting over the ancient “tablet of time,” which promised a younger, stronger, and healthier life. To think that there was a community out there within the marriage of existing ideas around longevity and emerging tech with web3, just being a part of it was attractive enough and a dream come true alone.

Can you walk me through what role you have taken on in VitaDAO?

What got me to start to contribute was finding the rare Schelling point of all my passions combined — longevity, public goods, and meta-governance. The rest is history. I started by doing starter tasks like developing the VitaDAO constitution on our discourse forum and my continued involvement for more than half a year now eventually led me to take on the role of governance lead for the DAO.

Have you found that there are gaps in infrastructure and tooling that could enable the DeSci community?

The constant gap has always been onboarding the next billion. With DeSci, the battle against the disrupting the ancient structure of centralized science itself is hard to make researchers understand, as it seems almost alien to the pathways they’ve been led to know and experience. let alone, innovations in human coordination with meta-governance and DAOs. My goal is to bridge the gap with the rest of the community but more so in meta-governance tooling curated for academics as much as DeSci tooling as a whole. That is where my passion lies.

What has surprised you the most about VitaDAO?

The sheer passion within the contributors to empower everyone in our community regardless of their knowledge, background, and skills. I think that alone aside from our numbers went a long way in our recent fundraising round.

What excites you most about VitaDAO’s future?

I’m most excited about how long VitaDAO will be here. Not to take it for granted, but being in the midst of people that share my passions till the unforeseeable future feels like I’ve found my tribe. On the notion of longevity, VitaDAO’s future is an ever-extending one as we improve the lifespan of humans, our community, and the world.

Any closing thoughts for readers?

As a servant leader, my driven purpose of existence is to immortalize my ideas for the betterment of public goods. I’m a longevity and anti-aging enthusiast on a mission to attain optimal coordination with new paradigms in meta-governance and to revolutionize the world with unworldly regenerative innovations. I’d love to connect with anyone and everyone willing to see these goals flourish as much as I do. I’m most active on Twitter/Discord/Telegram: @consigli3re

Upcoming Events

The first pop-up mini-city experiment with longevity at heart — 25 March — 25 May — We’re exploring evolving VitaDAO towards a Longevity Network State, and building a special jurisdiction for medical innovation with a better regulatory framework. Apply to visit.

3rd VitaDAO DeSci & Longevity Symposium — April 5th — Sign up and learn to learn more about the science behind slowing down aging and the development of rejuvenation therapies.

VitaDAO @ Longevity Med Summit — May 4–5, 2023- Come join VitaDAO in sunny Lisbon, Portugal at the 2023 Longevity Med Summit. Here, join other longevity enthusiasts, in learning about the development of anti-aging science, the exploration of new regenerative and longevity therapeutics, and the limiting R&D challenges faced in therapy development.

Join Us!

Have a special skill set? Contribute to VitaDAO and receive $VITA in return. Join the VitaDAO community on Discord and stay up to date on Twitter! As of February, VitaDAO’s Discord is now token-gated, requiring a minimum of 10 $VITA to gain access to basic contributor channels.

Early this year, VitaDAO announced a $4.1 million fundraising round, that included strategic members and longevity enthusiasts such as Pfizer Ventures, Shine Capital, L1 Digital, Beaker DAO, Balaji Srinivasan, and Joe Betts-LaCroix.
Read more
The Mouse Longevity Chronicles: Unraveling Aging with Dr. Richard Miller during The Aging Science podcast by VitaDAO
March 23, 2023
Awareness
Longevity
The Mouse Longevity Chronicles: Unraveling Aging with Dr. Richard Miller during The Aging Science podcast by VitaDAO

The Mouse Longevity Chronicles: Unraveling Aging with Dr. Richard Miller during The Aging Science podcast by VitaDAO

Check out the podcast here.

In this podcast, I (@aging_scientist) spoke with Dr. Rich Miller about the ins and outs of mouse aging.

The specific topics we discussed, roughly in this order, included the Interventions Testing Program, rapamycin, mecilizine and other lifespan-extending drugs. We talked about the importance of genetically heterogenous mouse stocks and the issues with fast-aging progeroid mouse models. We also covered his more recent work on aging rate indicators, and the difference between classic biomarkers and aging rate indicators. Finally, we talked about the importance of lifespan research, misuses of the word healthspan and the emerging use of frailty indices in mice.

I did not make it easy for Rich asking some difficult questions, most of which he answered quite well, even if I may disagree here or there. All in all, it was great to interview one of my favorite aging researchers!

About Rich Miller – short biography

Rich Miller got his BA degree at Haverford College, and then an MD and PhD at Yale.  He received postdoctoral training at Harvard and Sloan-Kettering and was a faculty member at Boston University before moving to Michigan in 1990.

Now he is a principal investigator at the University of Michigan where his lab studies long lived mice and tries to understand why mice and other animals live as long as they do. He has been instrumental in facilitating the birth of the interventions testing program (ITP) which was the program that first discovered that rapamycin extends the lifespan of mice. 

If you want to know one cool piece of trivia about Rich. I once checked and it appears that he has coauthored more mouse longevity studies than a small European country like the Netherlands. This attests to both his experience but also to the lack of mouse research in Europe.

http://www.richmillerlab.com/

Scientific background

Let us briefly talk about the importance of mouse lifespan studies and why healthy mice are an irreplaceable scientific tool. I will also explain some of the concepts that we discussed in the podcast. 

"It's not simple" (Rich Miller)

About the Interventions Testing Program (ITP)

“The Interventions Testing Program (ITP) is a peer-reviewed program designed to identify agents that extend lifespan and healthspan in mice. Investigators at any university, institute, company, or other organization are invited to recommend interventions for testing by submitting an application before the February deadline each year. Testing is carried out in the genetically heterogeneous UM-HET3 mouse stock at three sites — the Jackson Laboratory, the University of Michigan, and the University of Texas Health Science Center at San Antonio.”

Several features of the ITP make it stand out as a very unique project. One is that almost anyone can propose a compound through a simple, short application. Scientists have been calling for simpler grant applications for a while so it is refreshing to see this implemented in practice. Hopefully other institutions will follow suit.

Another feature is the sheer success of the ITP in finding new interventions.

“the ITP has identified nine agents that significantly increase median lifespan — acarbose (Harrison 2014, Strong 2016, Harrison 2019), aspirin (Strong 2008), canagliflozin (Miller 2020), captopril (Strong, 2022), glycine (Miller 2019), nordihydroguaiaretic acid (NDGA) (Strong 2008, Strong 2016), Protandim® (Strong 2016), rapamycin (Harrison 2009, Miller 2011, Wilkinson 2012, Miller 2014) and 17α-estradiol (Harrison 2014, Strong 2016, Harrison 2021).”

Although, all these worked in a subset of mice, I think this list is slightly overstating the success of the ITP. Some of these drugs are much more effective than others. Some of these are in a league of their own. It is hard to compare rapamycin, perhaps the biggest breakthrough of the 21st century (so far), with compounds that produce minor benefits in one gender like e.g. protandim. However, even if the ITP had only discovered rapamycin, it would have been a success in my eyes.

Almost as important as the successes are the failures. The ITP has refuted or weakened the idea that several putative longevity compounds actually slow aging, e.g. metformin and resveratrol. While these could still benefit humans and improve health, the ITP provided strong evidence that they fail to slow aging in mice.

On the importance of good mouse husbandry and healthy mouse strains

"Inbred mice are an inappropriate model for most kinds of aging research" (Rich Miller)

Although in this case Rich was quoting other researchers, I think this does sum up the message he wanted to convey. While I would not go quite that far, I think he is totally right when he says that we need to use healthy, genetically heterogenous mouse models for aging research. As much as possible, whenever feasible.

Humans are diverse and we need diverse models to better represent our biology. Even more importantly, inbred mice often used in research have limited heterogeneity in what they die of. Thus, preventing this one idiosyncratic disease that is killing all mice of an inbred strain could appear to slow aging and extend lifespan when it was just mitigating this one condition. However, in defense of our traditional model the inbred “Black 6” (B6) mouse – and it is indeed a mouse with black fur, hence the name – I would like to note that genetically heterogenous mice still predominantly die from cancer (Lipman et al. 2004) not much different than B6 mice. On top of that, B6 mice are actually quite long-lived for a mouse.

The ITP uses UM-HET3 mice, which are a heterogenous four-way cross, to mitigate the issue of limited genetic diversity. I would like to champion the UM-HET3 mouse as a replacement of B6 and other strains whenever possible. It is a really amazing mouse strain. Please use it in your resarch and when you read a paper featuring UM-HET3 consider this a “bonus point” in favor of that work.

Aging Rate Indicators

The lab of Rich Miller is developing so called aging rate indicators, which could be used to predict how quickly a mouse or a cohort of mice will age. The idea is to find such indicators and to screen a battery of novel compounds against these, before testing them in large, expensive lifespan studies like the ITP. 

In the analogy he used the aging rate indicators are a kind of “speedometer”, whereas conventional biomarkers measure the damage that mice accumulated during aging akin to an “odometer”. A mouse that ages at a slower speed will accumulate less damage and hence live longer.

The controversial bits and pieces – progeroid models, frailty and healthspan

Rich is not the biggest fan of senolytics, progeroid models and frailty indices used in mice. We talked about the latter two in the podcast. I think he does a better job explaining this than I could so I will instead just explain the science necessary to understand our podcast.

So called progeroid mouse models show something akin to accelerated aging. They are employed by aging researchers because they die quickly allowing them to test more interventions in a shorter period of time. Whether these models faithfully mimic aging remains a matter of debate in the literature and both sides have good arguments. On the one hand, many of these models develop similar changes to aged mice due to mutations in pathways we think are involved in aging (e.g. DNA repair). On the other hand, most progeroid mouse models develop extreme phenotypic changes that are never seen in aging mice to the same extent.

Frailty indices were developed in order to measure something resembling health, healthspan or frailty in mice – whatever your preferred term is. Basically, functional decline during aging. Indeed, such functional decline almost always precedes mortality and the two are closely linked. However, measuring lifespanis very expensive as it requires many animals. So one of the reasons why mouse frailty indices are gaining popularity is the hope that they could serve as a replacement for expensive lifespan studies. While commonly employed frailty indices do seem to capture functional decline with aging, many parameters included in these indices are not very sensitive to aging (Schultz et al. 2020) and they have not been thoroughly validated across different mouse strains yet. Furthermore, it is also very difficult to observe frailty in mice non-invasively. Old mice often die spontaneously while still looking healthy.

My (minor) disagreements with Rich Miller – ITP compound selection

Asking hard questions is the job of a good journalist and being critical of conventional wisdom is the job of a good scientist. I am trying to be both here.

My major criticism, which Ì tried to explain, was that the ITP is somewhat biased towards approved drugs that target age related diseases, or diseases of affluence, instead of more experimental bona fide anti-aging drugs. As biogerontologists, we always say that we must target the root causes of aging. I do not think that approved most drugs are doing that. Certainly not statins and most blood pressure lowering medications, several of which were tested in the ITP.

As far as I can tell, Rich’ counterargument is that every suggested drug was evaluated on its own merits. Only if the proposal was persuasive then it was accepted. Maybe for some of these traditional drugs arguments can be made why they might slow aging. Indeed, rapamycin itself is an approved drug “traditionally” used for immunosuppression and the proposal certainly included a rationale why it could be repurposed to fight aging based on genetic data from yeast (Kaeberlein et al. 2005).

Furthermore, the proposed rationale and why a drug works do not need to be the same. For example, in the case of acarbose the mechanism of action in mice is subtly different from the one that was proposed, as we discuss in the podcast.

Nevertheless, in my opinion, there remains a bias towards “playing it safe” in the ITP. Just to give an example, I have a really hard time finding a rationale why simvastatin might have slowed aging in mice – because it so specifically targets atherosclerosis, an age-related disease that mice fail to develop. Statins do not work particularly well if you have low blood cholesterol levels and do not reduce all-cause mortality in these populations (Singh et al. 2020), hence the failure in mice was perhaps also predictable.

My (minor) disagreements with Rich Miller – does rapamycin slow or postpone aging?

My second disagreement concerns rapamycin. I think Rich and others are too hastily discounting the idea that rapamycin postpones aging rather than slowing aging, or that something else strange is going on with rapamycin. However, that is not to say that rapamycin would fail to extend lifespan in humans. It may, but the mechanism could be different than we think.

In fact, I believe that rapamycin is the best drug we have right now and I am on the record saying that it should be in the TAME study instead of metformin. No one can claim I am not an optimist about this drug.

I used to think the same way as Rich does: if a drug extends median and maximum lifespan in healthy mice and slows age-related decline then it slows aging per definition.

However, we need to be a bit more precise and distinguish between slowing and postponing aging. In the long term this will be very important.

Different papers studying mortality patterns using Gompertz and other mathematical approaches find that rapamycin behaves differently than caloric restriction and provide evidence for postponing rather than slowing aging (Garratt et al. 2016): “Reduced mTOR signalling extends lifespan in nematodes by strongly reducing the degree to which mortality rates increase with age (aging rate). By contrast, life extension in mice and yeast occurs largely by pushing back the onset of aging, but not altering the shape of the mortality curve once aging starts. Importantly, in mice, the altered pattern of mortality induced by reduced mTOR signalling is different to that induced by dietary restriction, which reduces the rate of aging”

The benefits of rapamycin in the ITP do not scale very well with age of onset. When treatment is started at 600 days of age (20 months), then, expressed as increase in mean lifespan, the effect sizes were +9% for males and +13% for females in the pooled data set (Harrison et al. 2009). For comparison, when treatment was started at 270 days of age or (9 months) then median survival was extended by an average of +10% in males and +18% in females (Miller et al. 2011).

Importantly, one recent paper found that an mTOR hypomorphic allele leads to immediate improvements in multiple age related parameters even in young mice (Xie et al. 2022). This suggests we are measuring age related decline incorrectly without including a young control group. Xie et al. concludes that: “Many [interventions] influence phenotypes long before the onset of detectable age-dependent change, but, importantly, do not alter the rate of phenotypic change. Hence, these [these interventions] have limited effects on aging.”

Hopefully we can revisit this debate some day

Rich Miller in other media

If you still cannot get enough aging science content after having finished our podcast, you can check out the below interviews with Rich.

On Peter Attia’s famous Drive podcast:
https://peterattiamd.com/richardmiller/

On the Live Longer World podcast, which is one of my favorite aging podcasts:
https://www.youtube.com/watch?v=42PzfNs9egA

On the The Sheekey Science Show, which is one my favorite aging channels on youtube:
https://www.youtube.com/watch?v=hHlDTiFWbbg
(you can follow Sheekey on twitter @EleanorSheekey)

On the amazing NUS Healthy Longevity webinar, as published on youtube:
https://www.youtube.com/watch?v=UPqstwgr7x0
(you can register for the webinar here so as not to miss any live sessions: https://medicine.nus.edu.sg/continuing-education/knowledge-hub/healthy-longevity/)

References

Xie, Kan, et al. "Deep phenotyping and lifetime trajectories reveal limited effects of longevity regulators on the aging process in C57BL/6J mice." Nature Communications 13.1 (2022): 1-29.

Harrison, David E., et al. "Rapamycin fed late in life extends lifespan in genetically heterogeneous mice." nature 460.7253 (2009): 392-395.

Miller, Richard A., et al. "Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice." The Journals of Gerontology: Series A 66.2 (2011): 191-201.

Garratt, Michael, Shinichi Nakagawa, and Mirre JP Simons. "Comparative idiosyncrasies in life extension by reduced mTOR signalling and its distinctiveness from dietary restriction." Aging Cell 15.4 (2016): 737-743.

Singh, Bishnu M., et al. "Role of statins in the primary prevention of atherosclerotic cardiovascular disease and mortality in the population with mean cholesterol in the near-optimal to borderline high range: a systematic review and meta-analysis." Advances in preventive medicine 2020 (2020).

Yourman, Lindsey C., et al. "Evaluation of time to benefit of statins for the primary prevention of cardiovascular events in adults aged 50 to 75 years: a meta-analysis." JAMA internal medicine 181.2 (2021): 179-185.

Schultz, Michael B., et al. "Age and life expectancy clocks based on machine learning analysis of mouse frailty." Nature communications 11.1 (2020): 1-12.

Kaeberlein, Matt, et al. "Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients." Science 310.5751 (2005): 1193-1196.

Lipman, Ruth, et al. "Genetic loci that influence cause of death in a heterogeneous mouse stock." The Journals of Gerontology Series A: Biological Sciences and Medical Sciences 59.10 (2004): B977-B983.

In this podcast, I (@aging_scientist) spoke with Dr. Rich Miller about the ins and outs of mouse aging.
Read more
‍February Longevity Research Newsletter‍
March 10, 2023
Rhys Anderson & Maria Marinova
Awareness
Longevity
Newsletters
‍February Longevity Research Newsletter‍

Introduction

Welcome back Vitalians!

Following our successful fundraise of $4.1 million (check out the Forbes article), a new wave of projects are in the pipeline to be funded. First up in 2023 is Brain Tissue Replacement Therapy with Jean Hebert - see his latest paper on rebuilding neocortical tissue in our hot picks below.

The VitaDAO community also voted to develop a brand new overlay journal - The Longevist - a curation of the most impactful longevity research every quarter, as voted on by a large body of key opinion leaders. Stay tuned as we aim to launch our 1st issue in April!

There are a plethora of different theories as to why we age, often with overlapping ideas that make it hard to separate the relative contributions of each factor. This month, we are excited to bring you an interview with Prof. John Speakman - a world leader on energy expenditure - who recently published his “Live cold, die old” paper, with some elegant experiments providing evidence that body temperature is a greater driver of ageing than metabolic rate in two species of small mammals. Check out the interview as Prof. Speakman discusses this and how it affects our understanding of calorie restriction interventions, free radical damage, and how temperature and entropy could be key to understanding biological ageing (recall our previously featured preprint from Dr. Peter Fedichev: Aging clocks, entropy, and the limits of age-reversal). Enjoy!

Longevity Literature Hot Picks

Preprint Corner

This month we are featuring 7 new preprints which are all available to review on our reviewing platform The Longevity Decentralised Review (TLDR) in return for a bounty of 50 $VITA each. Simply follow the above link to the TLDR page and get reviewing! What's more, we will be continuing the 50 $VITA bounty for reviewing any of the preprints featured in January's longevity research newsletter.

p16-dependent upregulation of PD-L1 impairs immunosurveillance of senescent cells

NLRP1 inflammasome modulates senescence and senescence-associated secretory phenotype

Gene body DNA hydroxymethylation restricts the magnitude of transcriptional changes during aging

Insulin-mTOR hyperfunction drives C. elegans aging opposed by the megaprotein LPD-3

The human pathome shows sex specific aging patterns post-development

Aging Atlas Reveals Cell-Type-Specific Regulation of Pro-longevity Strategies

Biological Age Estimation Using Circulating Blood Biomarkers

Published Research Papers 

Photoactivatable senolysis with single-cell resolution delays aging +

A senolytic strategy integrating multiple technologies delays aging (research briefing)

Senolytics have been shown to promote longevity but precise and trackable senolysis is still a challenge. An interesting approach to tackle this is described here - a photosensitive senolytic drug targeting the enzyme substrate of SA-β-gal with fluorescence tag for the precise tracking.

An In Vivo Platform for Rebuilding Functional Neocortical Tissue

While advances in stem cell transplantation have been made, the functionality of transplants remains a challenge. This in vivo transplant platform resulted in functional graft with differentiated layers of neurons, vascularization within a week, electrophysiological activity in a month and it responded to visual stimuli.

Transgenerational inheritance of acquired epigenetic signatures at CpG islands in mice

The study provides evidence for transgenerational epigenetic inheritance, a long debated subject. They show the CpG island reprogramming in the parental generation was maintained and transmitted across multiple generation alongside the phenotypic traits.

Effect of long-term caloric restriction on DNA methylation measures of biological aging in healthy adults from the CALERIE trial

The DNA methylation of 220 subjects with obesity randomized to 25% CR or ad libitum for 2 years were assessed but resulted in inconclusive results. There was a striking difference between DNAm tests with DunedinPACE showing slowed pace of aging, while PhenoAge and GrimAge showed no change in biological age.

Unique progerin C-terminal peptide ameliorates Hutchinson–Gilford progeria syndrome (HGPS) phenotype by rescuing BUBR1

HGPS is a premature aging disease and there has been a growing body of evidence that mitotic defects play a role. A core spindle assembly protein BUBR1 had decreased levels and the remaining protein was anchored. A unique peptide prevented binding and increased expression, showing potential for progeria therapeutics. 

Correlated evolution of social organization and lifespan in mammals

Phylogenetic comparative analysis of over 1000 species compares solitary vs group-living species and concludes that group-living species generally live longer than solitary-living, suggesting correlated evolution of social organisation and longevity.

Telomere-to-mitochondria signalling by ZBP1 mediates replicative crisis

Dysfunctional telomeres were shown to activate immune responses as a mechanism for telomere mediated tumour suppression. The process is dependent on mitochondrial telomeric-repeat-containing RNA (TERRA) transcripts that are synthesized from dysfunctional telomeres forming oligomers with ZBP1 (Z-DNA binding protein).

The multi-tissue landscape of somatic mtdna mutations indicates tissue specific accumulation and removal in aging

Mitochondrial DNA mutation rate and accumulation significantly differed across aged tissues. This was not correlated to tissue function and mitochondrial content. An unexpected lack of mutations linked to oxidative damage were found, suggesting dynamic clearance.

Job board

XPRIZE Healthspan Lead

A two-part role including being the Executive Director of XPRIZE’s Health Domain and Director of the upcoming “Healthspan XPRIZE”

Lab Assistant for Killifish Husbandry and Research at The Department of Genetics at Stanford University, US

To help with research on aging and age-related diseases.

SenNet Consortium Underrepresented Student Program Summer 2023 Application

Summer undergrad research internships at the NIH cellular senescence research consortium! Deadline 15th March 2023.

A Postdoc (Research Fellow, RF) and Ph.D. position in Biology of aging are available in the Sorrentino laboratory at NUS in Singapore. The projects explore the interconnectivity and possible therapeutic targeting of the cellular hallmarks of the aging process, with a particular focus on mitochondrial dysfunction, protein aggregation and alterations of NAD+ metabolism. 

Are you interested in cardiovascular research and aging? Apply for a PhD position and join a research team part of the VASC-HEALTH Flagship Project at the Medical University of Graz

News

A4LI is proud to announce the founding of the Congressional Caucus for Longevity Science. 

The formation of the “Longevity Science Caucus” is a transformational moment for the longevity biotechnology industry and the movement to increase healthy human lifespans.

Targeted degradation: putting autophagy to work

Autophagy inducing biotech company Casma Therapeutics closed a $46 million round with the aim to develop novel treatments for cancer, inflammation, neurodegeneration, and metabolic disorders

Articles

Antiaging talk: Is red wine good for the skin?

Global longevity investment hit $5.2 billion in 2022

The puzzling gap between how old you are and how old you think you are

Mapping out the longevity investment landscape

Anti-ageing scientists extend lifespan of oldest living lab rat

Restrict calories to live longer, study says, but critics say more proof is needed

Young female mice exposed to scents from older females live longer

Just Getting Started at 75

Study reveals how exercise turns back the clock in aging muscles

Conferences, Workshops and Webinars

Young ICSA Cambridge, UK 2023

Two-day cellular senescence symposium at Cancer Research UK - Cambridge Institute! A great opportunity for PhD students and postdocs.

Workshop: Midlife Stress and the Hallmarks of Aging

Researchers at all career stages from academia, industry, and government with an interest in the impacts of stress on aging in human populations and animal models of stress and/or aging.

Podcasts and Videos

Bryan Johnson's Blueprint, Spotify Enters Health and VitaDAO Raises $4M

Increased Lifespan from Beans

The intake of legumes—beans, chickpeas, split peas, and lentils—may be the single most important dietary predictor of a long lifespan. But what about concerns about intestinal gas?

Interview with Prof. John Speakman

Professor John Speakman is a leading expert on metabolic activity and energy expenditure and was instrumental in the development of doubly-labelled water (DLW) technique. He has made numerous significant contributions to our understanding of factors that govern variation in food intake and energy expenditure, and the consequences for fat storage (obesity) and ageing. He currently runs 2 research laboratories and is Head of Integrative Physiology at the University of Aberdeen, and a professor at the Chinese Academy of Sciences in Shenzhen. 

What inspired you to enter longevity research?

My PhD in the 1980s was to study energy balance and metabolic rates in wild animals. At the time there was a lot of interest in among species scaling relationships for all sorts of things – one of which was lifespan. Bigger animals live longer, and one idea why that was the case was because they have lower metabolic rate: the ‘rate of living theory’. This was also a potential reason why calorie restriction exerts its effects – by lowering metabolic rate. It was a pretty old idea from the 1920s and in the 1950s it got a boost by the idea that higher metabolism leads to greater production of free-radicals and therefore the reason we age is because of free-radical damage that stems from our metabolism. By the 1990s this idea had enormous traction among biologists studying ageing. Many of them were taking large amounts of vitamin C and E daily to quench their free-radical damage. Since I knew something about metabolism, I got interested in this idea that lifespan depends on metabolism and free-radicals, and that we have a fixed amount of lifetime expenditure of energy. I also started taking antioxidant vitamins daily. At the time in the 1990s however almost all the work on the idea was correlational and inter-specific. I figured that we could probably do intra-specific experiments to test the ‘rate of living/free-radical damage’ idea, by making animals expend more or less energy, and giving them anti-oxidants, and then looking at how long they live. I applied for money to the UK BBSRC to do such experiments as part of their first special topic on ageing and was successful. I recruited an amazing post doc (Colin Selman) who eventually became a great gerontology professor, and we did some interesting work together. The general outcome of our work was that the rate of living/free-radical damage model was not sustainable. Voles given antioxidants for example lived shorter lives. It had at least one direct human consequence, in that I personally stopped taking antioxidant vitamins. These studies eventually led to my groups work on calorie restriction which has dominated my work on ageing ever since.

Which of the current theories of ageing do you think are the most convincing?

I quite like the idea from the Nobel prize winning physicist Erwin Schrodinger in the 1930s that he presented in a short book called ‘what is life?’. In it he posited that organisms are incredibly complex low entropy systems that naturally have a tendency to increase in entropy because it needs a constant and large flow of energy to sustain their low entropy state. For a while it is advantageous in terms of reproduction for us to invest in holding back the increase in entropy in our soma, and we do so, but then eventually it starts to accumulate (ageing). This becomes self-reinforcing because the cost to repair the system starts to get larger and larger. Eventually entropy accumulates to the point where the system is overwhelmed, and that overwhelmed state is what we call death. Many other theories of ageing are potentially just restatements of the entropy principle – eg mutation accumulation, free-radical damage, senescent cell accumulation. What I like about the entropy idea is that it is very non-specific, so the reasons people die are probably all slightly different and depend on the stochastic nature of the system falling to bits. This not just explains why living things age and die but why any complex system fails. If you go around a scrap yard and look at the cars in there they are all in there for slightly different reasons but fundamentally for many of them they are there because investing more money in stopping them falling to bits was too expensive. I think what this tells us is that chasing down a single problem (like accumulation of senescent cells for example) is unlikely to be very effective as an overall anti-aging strategy because there will always be another problem that arises reflective of the increasing entropy. In other words as we all know there are multiple hallmarks of ageing. Ultimately, then, I think the solution will be to increase the activity of the systems that keep entropy low when we are young. This will really mean intervening when that system starts to decline which is probably in our twenties or thirties, not our sixties and seventies. By then its probably already too late.

How has the field changed since you started?

I guess the major recent change in the field is the fact that the boomer generation has finally realised that they are all going to die and they are not happy about it. So now there are very large sums of money being poured into ageing research by very wealthy people in their late 50’s early 60’s and that has given ageing research a tremendous boost. The money invested in Calico by Google, the Hevolution project and into Altos labs by Bezos and colleagues is enormous. Whether they will see a practical return on this investment in their lifetimes is an interesting question. Most of them in my view have left it too late already.

A second big practical difference is the interventions testing program in the USA. I think that was a real landmark achievement to set up an integrated multi-lab platform to test compounds that might have lifespan impacts in rodents. This program has saved a lot of time and effort chasing down false leads that would have been based on underpowered small studies.

What mistakes do you think the longevity field has made?

For cost purposes medical science in general has placed enormous efforts into understanding model organisms like yeast, C. elegans, Drosophila and mice. I think this has generally led to some important insights in many fields, but the benefit to cost ratio of understanding what causes ageing in C. elegans or yeast is very low. The translational benefits of working on ageing in mice are already not great because mice are not little people. Once you get into ectothermic animals, and single cell organisms that reproduce a-sexually, with fundamentally different physiology (eg ability to go into Dauer states in C. elegans or generate ethanol in yeast) the translational potential of such work is effectively zero. A recent review by Bene and Salmon in Geroscience showed that there was virtually no translation from C. elegans to mouse for life extending therapeutics. Studies of ageing in these organisms have led us down several expensive blind alleys, perhaps most notably the sirtuin story which is now starting to completely unravel.

The key issue though is that as you get closer to humans and the translational potential increases there are increasing ethical issues. Moreover, as the lifespan of the organisms under study get longer, the required experiments get longer as well, until we get to a point where nobody can do the required experiments within a single career of one scientist and progress is phenomenally slow. It is a hard nut to crack in a world where people want answers yesterday. I think this impatience for answers is basically what continues to fuel C.elegans work on ageing irrespective of its likely utility.

Other than your own, what do you think have been the biggest/important discoveries in the field?

I have found it really intriguing the very simple observation that if you transfuse blood from a young mammal to an older one you can slow down its ageing. I think this will turn out to be an incredibly important and practically significant observation. Is it because the young blood contains life promoting compounds, as some recent work suggests, or does it just dilute life shortening compounds in the old blood. Either way I think this has the most exciting potential for an immediate practical intervention.

What advice would you give to people currently working in longevity research?

Apart from finding out what is going on with young blood, if someone was just starting out a career then I think my advice would be to focus on ageing of the brain. You can envisage that for pretty much all our bodily functions it will eventually be possible to replace them with mechanical substitutes. In fact this is already true for many of our organs and musculo-skeletal systems. Recently exoskeletons were developed to augment declining muscle strength, thereby allowing elderly people to retain mobility and avoid falls. These days as people age we routinely swap out parts of their failing bodies and replace them with man-made mechanical alternatives – like heart valves and hip replacements. Although hip arthroplasty has a history dating back to the 1800s it only became a widely used and available procedure with minimal complications in the 1970s. Prior to that if your hips wore out you were basically crippled or bed-ridden and had a very low quality of life until you prematurely died. Nowadays, understanding the processes that lead to our hip joints failing as we get older is pretty much pointless because that problem has already been solved as far as practical aspects of hip ageing is concerned – although may give us insights into bone degeneration more generally. That will become increasingly the case as more and more mechanical replacements are developed. The one system, however, that I don’t see us ever being able to replace mechanically is the brain. That is the fundamental thing in your body that harbours what we identify as you. If it was possible to combine the head of a person grafted onto the body of a different person, we would identify the resultant being from the head (brain), not the body. Therefore, preserving the brain and preventing brain ageing is really the whole key to preventing ageing in the very long term. That, and inventing mechanical substitutes for all the other systems.

Which aspect of longevity research do you think requires more attention?

Brain ageing – see above.

Is ageing a disease?

As far as we understand it, from an evolutionary standpoint, ageing and death happen as part of an adaptive program. We stop reproducing at a certain point in our lives, and there is no evolutionary selective pressure to invest in effective somatic maintenance programs to preserve us into old age. Mutations in genes leading to improvement in those features in later life never get passed on. Without that selective pressure to evolve physiological processes that sustain our systems they slowly fall apart. Should we call that a disease? To be honest I don’t think it matters. It is something that negatively affects all of us, creates enormous personal, economic and societal costs, and therefore there will always be an impetus to try and stop it. Interestingly the first ever written document (the story of Gilgamesh) is about discovery of anti-ageing therapeutics. We have always been interested in avoiding ageing and extending lifespan. Whether we call it a disease or not won’t change that. There is an argument that if it isn’t classed as a disease then the FDA would not grant a licence for drugs that aim to retard ageing in general, rather than specific components of it. Calling it a disease then is just a pragmatic solution to a practical hurdle. It’s not something that I think scientists should spend their time agonising over. If (when?) an effective anti-aging drug appears that needs approval, a billionaire will pay a very well paid lawyer to argue the case that ageing is a disease, and then the path will be open for FDA approval.

You recently published your “live cold - die old” paper - would you be able to summarise your findings?

In general when endothermic animals like humans reduce their metabolic rate they reduce their heat production and this can lead to them having a lower body temperature. During calorie restriction for example it is pretty widely agreed that metabolic rate goes down (whether on a whole animal or normalised for body size basis) and that body temperature is also lower. Formally then because these two things go in tandem its not possible to separate which (if any) is a causal factor in the observed life extension. To separate the effects of metabolic rate and body temperature you need a situation where they change in opposite directions. One such situation in small rodents is when you change the housing temperature from below thermoneutral (around 20 oC) to the top of the thermoneutral zone (32.5 oC). When you do that metabolic rate goes down at the same time that body temperature goes up. Looking at that effect on lifespan in mice and hamsters at these temperatures allowed us to separate the effect of body temperature from that of metabolic rate. If lower metabolic rate was important then they should live longer when hotter, if body temperature drives the effect they should live shorter. The answer was that when you made the animals hotter their lifespan declined. We then showed that if you blow air over the animals in hot conditions you can prevent the rise in body temperature without altering metabolic rate, and when you do that you can rescue the lifespan effect. It seems that body temperature is more important than metabolic rate. In the press release we used the bumper sticker ‘live cold, die old’ to promote the work. Interestingly this may be a much more general principle than just organismal ageing. For example, the duration that transistors ‘live’ until failure is related negatively to their operating temperature. I suspect temperature is related to entropy change. When complex things are hotter entropy accumulates faster and they fail more quickly.

Do you envisage that cooler environments / increased convection would benefit humans? / Do you have any plans to test this?

Living in a colder environment will generally not reduce your body temperature much compared with the environments we currently seek out just below thermoneutral so that isn’t probably going to work. Similarly increased convection probably won’t be very effective either. It worked in the mice and hamsters to cool them down from a much higher temperature. We have recently completed some preliminary tests getting people to drink cold water and monitoring the impact on their body temperature as a pilot study to get the necessary data to perform power analysis for a randomised clinical trial. I like the idea but I didn’t start drinking cold water everyday myself yet.

Which other non-therapeutic interventions do you think hold promise for improving human healthspan/lifespan?

Although fraught with some ethical issues I think blood transfusion from young to older people holds a lot of promise based on the mouse work.

Outro

Thank you for staying with us till the very end and as always we encourage you to reach out to us about content you’d like us to discuss in our next issues. See you next month!

Further Reading

The association between body height and longevity: evidence from a national population sample

High Investment in Reproduction Is Associated with Reduced Life Span in Dogs

Following our successful fundraise of $4.1 million (check out the Forbes article), a new wave of projects are in the pipeline to be funded. First up in 2023 is Brain Tissue Replacement Therapy with Jean Hebert
Read more
Exploring the Frontiers of Science: Breakthroughs, Funding Challenges, and Replication Crisis with Alaattin Kaya during The Aging Science podcast by VitaDAO
March 9, 2023
Awareness
Longevity
Exploring the Frontiers of Science: Breakthroughs, Funding Challenges, and Replication Crisis with Alaattin Kaya during The Aging Science podcast by VitaDAO

Exploring the Frontiers of Science: Breakthroughs, Funding Challenges, and Replication Crisis with Alaattin Kaya during The Aging Science podcast by VitaDAO

 
Check out the podcast here.

In this podcast, I (@aging_scientist) had the pleasure of interviewing Asst. Prof Alaattin Kaya (@akaya_lab).


We talked about recent breakthroughs in the field, the difficulties of getting funding for risky and novel research, funding agencies, “fishing expeditions”, the importance of overexpression genetic screens in aging research, novel mechanisms of action, and the replication crisis.

We also talked about yeast as a model organism and had a clear consensus:

“Of course, yeah, [it’s] everybody loves yeast.” (Alaattin Kaya)

It was a fun podcast! Since we really liked Alaattin’s overexpression screens (see below for more) we want to help him fund his amazing research. So if you know of any philanthropists or funding agencies that are willing to fund moonshots in aging research, please do reach out to us or directly to him.
link: https://vimm.vcu.edu/laboratory-groups/alaattin-kaya-phd/

Scientific background

Let me give a bit of scientific background on our main topics before we start. If you are quite familiar with this topic you can also just jump right into the podcast. Even if the below explanations are a little handwavy, I do hope they will be useful.

Overexpression of essential genes in yeast screens

The idea of genetic screening in model organism is quite simple. Models like yeast, worms or flies are less complex than humans, have a short lifespan and reproduce quickly making them facile models to study aging. Researchers can test thousands of compounds or change the expression of thousands of genes in a matter of weeks, in order to see whether these extend lifespan. Often they will find genes or pathways that are conserved between species, meaning they could also work in humans. One famous example of a conserved pathway is the Insulin/IGF-1 signalling pathway. Cynthia Kenyon and others discovered and validated this pathway in worms, showing that loss of daf-2 — which is related to human insulin and IGF-1 — leads to drastic lifespan extension in the worm. The importance of this pathway was later confirmed in mice.

Essential genes are those that are necessary for the survival of an organism. It stands to reason that these are important and often involved in tissues maintenance or repair. Given that “damage theories” of aging are well supported this makes essential genes attractive targets to study. What is more, essential genes will be often conserved between species. So if we found an essential longevity gene in yeast, for example, it would be more likely to do the same in humans than a non-essential gene.

Intriguingly, during evolution the duplication of (essential) genes often yields genes with novel functions, some of which are important for aging. Indeed, I worked myself on one such gene family. We suggested that cytoprotective metallothioneins might have undergone duplications that allowed for higher expression in longer-lived species (Pabis et al. 2021). While the evidence is not perfect, it is quite striking that humans have a dozen metallothioneins while short-lived mice have only three and worms have two metallothioneins. Alaattin mentions another example of an important gene duplication during the podcast.

Longevity probably evolves via gain of function (and change of function) rather than loss of function, which we usually study in the lab.

In contrast, essential genes are hard to study, because knocking them out, as is often done in genetic screens, would be lethal to the organism. Therefore most genetic screens focus on the knock-out of non-essential genes and in pharmaceutical screens inhibitors are easier to find than activators. Here is where Alaattin’s idea comes in. He decided to study the over-expression of essential genes to test whether these promote longevity in yeast, which are the perfect model for this since you can overexpress genes using so called plasmids.

If you want to know more about the project, Matt Kaeberlein explored this topic in his ARDD talk titled “The dark matter of bio-aging” (5). For those who are keen to read the paper itself, Alaattin’s preliminary work funded by Impetus grants, among others, has been published (Oz et al. 2022).

We scientists often call such screening experiments jokingly “fishing expeditions” because you never know what you will dig up. While a valid criticism of this approach, it is not a convincing one. During the podcast we discussed how reviewers take this criticism too seriously, favouring work with a clear mechanistic rationale instead and how this affected Alaattin’s research.

Lack of novel mechanisms?

“I think in aging everything is controversial at this point to be honest” (Alaattin Kaya)

Without getting into too much detail, reviewers who decry fishing expeditions could not be more wrong. While, yes, we do have some very outlandish ideas in the pipeline (reprogramming, parabiosis), by and large the field is almost exclusively dominated by quasi caloric restriction(CR)-mimetics. Treatments that target anabolic pathways and slow growth in one way or another are currently the only interventions that lead to robust mouse lifespan extension. Even worse, we still do not understand aging well enough and hence we need to feed the translational pipeline from the bottom up with the most novel and outlandish things we can find during our fishing expeditions — if we want to make any progress against aging in the longterm. Overexpression screens in model organisms would be great for that.


Bio and research focus — Alaattin Kaya

Alaattin Kaya

Alaattin is a young investigator who recently started his own lab at the Virginia Commonwealth University. He worked as a PhD and postdoc under the great Vadim Gladyshev and is a close collaborator of Matt Kaeberlein. His focus is on using yeast as a model to study basic mechanisms of aging. On his lab webpage Alaattin makes a good argument for using yeast in aging research:

“Current evidence suggests that many of the aging mechanisms and related genes are conserved among eukaryotes, from yeast to mammals. Each model system provides key advantages and challenges. Due to a variety of factors — notably including ease of genetic manipulation and a physiology similar to that of humans — the mouse has become the pre-eminent mammalian model organism in aging biology. However, in light of the high housing costs and relatively long lifespan of mice, large-scale unbiased screening to identify anti-aging medicines is not feasible in this organism. With the realization that many aging-related pathways are evolutionarily conserved, even among widely divergent species, short-lived invertebrate models have instead been employed for such screening.”

You can find more information under kayalab.org or on twitter @akay_lab.

References

1. Evidence that conserved essential genes are enriched for pro-longevity factors.
Oz N, Vayndorf EM, Tsuchiya M, McLean S, Turcios-Hernandez L, Pitt JN, Blue BW, Muir M, Kiflezhgi MG, Tyshovskiy A, Mendenhall A, Kaebverlein M, Kaya A. (2022) GeroScience https://doi.org/10.1007/s11357-022-00604-5

2. Kenyon, Cynthia, et al. “A C. elegans mutant that lives twice as long as wild type.” Nature 366.6454 (1993): 461–464.

3. Malavolta, Marco, and Kamil Pabis. “Elevated metallothionein expression in long-lived species.” Aging (Albany NY) 14.1 (2022): 1.

4. Pabis, Kamil, et al. “Elevated metallothionein expression in long-lived species mediates the influence of cadmium accumulation on aging.” GeroScience 43.4 (2021): 1975–1993.

5. Matt Kaberlein at ARDD2022: The dark matter of bio-aging
https://www.youtube.com/watch?v=kkozY2QlNZs&t=176s

Further reading

Kaya, Alaattin, Alexei V. Lobanov, and Vadim N. Gladyshev. “Evidence that mutation accumulation does not cause aging in Saccharomyces cerevisiae.” Aging cell 14.3 (2015): 366–371.


Read more
January Longevity Research Newsletter
February 3, 2023
Maria Marinova & Rhys Anderson
Longevity
Awareness
Newsletters
January Longevity Research Newsletter

Introduction

Happy new year Vitalians and welcome back!

The Longevity field is starting the year strongly with new updated Hallmarks of aging, exactly 10 years after the original paper was published. So time to update your introduction slides with the figure we will see in every presentation for the next 10 years!

For the first month of the year we want to focus on one of the original and major hallmarks of aging - genome instability. DNA damage is a well-known driver of aging. It comes in many flavours - including single and double strand breaks, base modifications and crosslinking. DNA lesions are very common, occurring by the tens of thousands every day in every cell of our body. Luckily, we have evolved extremely efficient DNA damage repair machinery to cope with this. These mechanisms, while effective, succumb to age-associated decline, allowing DNA damage to accumulate in later life. Many environmental factors can contribute to DNA damage, from UV to toxins and radiation. Genome stability and epigenetics are tightly intertwined as we’ll see in a few of our hot pick papers this month. To demystify this and other DNA damage queries you may have, we have interviewed Prof Björn Schumacher, an internationally recognised expert in the area.

Longevity Literature Hot Picks

Preprint Corner

The rise of preprints has reduced the time it takes for new scientific discoveries to be disseminated to the world prior to publication in a peer-reviewed journal. For this reason we are introducing Preprint Corner to keep you even more up-to-date with the Longevity literature. This month we are featuring 6 new preprints which are all available to review on our reviewing platform The Longevity Decentralised Review (TLDR) in return for a bounty of 50 $VITA each. Simply follow the above link to the TLDR page and get reviewing! We will also be awarding a 500 $VITA prize to the review which receives the most upvotes out of the 6 preprints listed! 

Reviewing bounty for these preprints available until 28th February, with upvotes counted and the prize winner announced 2 weeks later!

We are also excited to announce that voting is now open on Snapshot for our proposed overlay journal - The Longevist - aiming to be a curated collection of the most impactful longevity research every quarter, as voted on by a large body of Key Opinion Leaders.

Accurate aging clocks based on accumulating stochastic variation

A dual MTOR/NAD+ acting gerotherapy

Gene Therapy Mediated Partial Reprogramming Extends Lifespan and Reverses Age-Related Changes in Aged Mice

All-optical spatiotemporal mapping of ROS dynamics across mitochondrial microdomains in situ

Learning accelerates the evolution of slow aging but obstructs negligible senescence

Genetic perturbation of mitochondrial function reveals functional role for specific mitonuclear genes, metabolites and pathways that regulate lifespan

Published Research Papers 

Inheritance of paternal DNA damage by histone-mediated repair restriction

Paternal, but not maternal, exposure to ionising radiation leads to embryonic lethality and the mechanisms are still unclear. Depletion of histone and heterochromatin proteins could reverse embryonic lethality by reducing histone methylation and enabling repair, ultimately improving offspring viability. 

Loss of epigenetic information as a cause of mammalian aging

In the works for over 13 years, this massive study by Sinclair’s team presents an aging model - ICE (inducible changes to the epigenome) mice. DSB repair was reported to induce epigenetic deterioration earlier in life, which translated to physiological changes associated with aging. OSK reprogramming of ICE mice resulted in a lower epigenetic age.

Cell-cell metabolite exchange creates a pro-survival metabolic environment that extends lifespan

Metabolism is highly associated with ageing, with many longevity interventions targeting key metabolic pathways. Here the authors show that yeast cells can exchange metabolites across generations with the nature of these interactions capable of determining cellular lifespan. 

OxPhos defects cause hypermetabolism and reduce lifespan in cells and in patients with mitochondrial diseases

Patients with mitochondrial disease have increased resting energy expenditure, or hypermetabolism, which is thought to accelerate biological aging. Disrupting OxPhos (oxidative phosphorylation) either genetically or pharmacologically doubles the energy expenditure in the cell. More studies are needed to understand how these processes interact and link.

Intravascularly infused extracellular matrix as a biomaterial for targeting and treating inflamed tissues

Decellularized and enzymatically digested extracellular matrix with pro healing properties was infused intravascularly in rats and pigs post injury. Substantial improvements were seen in ventricle volumes and wall-motion scores as well as molecular level changes indicating tissue repair processes suggesting translational potential for this technology to heal the heart “from the inside out”.

Sphingolipids accumulate in aged muscle, and their reduction counteracts sarcopenia

Sarcopenia is a major issue with the quality of life of older adults that does not have current treatment options. Inhibition of sphingolipid synthesis showed promise in the alleviation of age-related decline and improved functional parameters like exercise capacity and strength.

Lithium treatment extends human lifespan: findings from the UK Biobank

Lithium has been used in the clinic for many years as a management strategy for bipolar and similar psychiatric disorders. A number of recent studies have shown it can increase lifespan in multiple animal models but now a correlation with human longevity is found. An observational study with over half a million people reveals that lithium was linked to decreased mortality and 3.6 times lower chance of dying when compared to users on other antipsychotic drugs.

Genome-wide RNA polymerase stalling shapes the transcriptome during aging

Whilst gene expression analysis has identified that many genes are differentially regulated with ageing, little has been known about the mechanisms driving these changes. This research shows that endogenous DNA damage can lead to RNA polymerase stalling which lowers transcriptional output and acts in a gene-length dependent manner. 

Optogenetic rejuvenation of mitochondrial membrane potential extends C. elegans lifespan

It’s been around half a century since Denham Harman updated his Free Radical Theory of Ageing to implicate mitochondrial ROS production in driving age-associated decline. Here the authors show that increasing mitochondrial membrane potential can decelerate ageing and extend lifespan in nematodes. This is achieved with an optogenetic approach with a light-activated proton pump!

Protein lifetimes in aged brains reveal a proteostatic adaptation linking physiological aging to neurodegeneration

The study reveals how the lifetimes of proteins change in the aging brain. The altered rates of synthesis and degradation point to a metabolic adaptation prior to neurodegeneration.

Published Literature Reviews

Hallmarks of aging: An expanding universe

Here we have the above mentioned updated hallmarks of ageing publication!

Insights into Alzheimer’s disease from single-cell genomic approaches

A complex interplay of numerous brain cell types is behind the Alzheimer’s dementia pathology. Cell type specific alterations and five main pathways were uncovered using single-cell profiling across five cell types. The highlighted pathways could be a important targets for therapeutic development.

Unraveling female reproductive senescence to enhance healthy longevity

Reproductive longevity is still largely understudied. The review summarises potential intervention that can prolong fertility in females and delay the onset of menopause which comes along with increased risk of a plethora of age-related diseases.

Testing the evidence that lifespan-extending compound interventions are conserved across laboratory animal model species

Concern about the predictive value of testing therapeutics in lower life forms is raised after surprisingly little predictive value for identifying drugs in worms that extend lifespan in mice. While not conclusive in making C. elegans obsolete as a model, the study raises the question of model accuracy and relevance to translation to humans. 

Clinical Trial Updates

Geron Announces Positive Top-Line Results from IMerge Phase 3 Trial of Imetelstat in Lower Risk MDS

FDA approves Alzheimer’s drug lecanemab amid safety concerns

Job board

VitaDAO company builder

VitaDAO is looking for a relentless, resourceful, well-organized, and dynamic person to advance the projects in our portfolio and spin them out as a co-founder or EIR.

PhD and Postdoc positions in the newly minted Correia-Melo Lab - Leibniz Institute on Aging - Fritz Lipmann Institute (FLI)!

The new “Microbiome and Metabolism” research group studies the metabolic crosstalk between cells in health and disease, with particular focus on aging. 

PhD positions available at the Computational Biology group at the FLI - a multi-disciplinary group composed of bioinformaticians, statisticians, biochemists, and biologists. Projects focus of using machine learning to uncover more about epigenetic changes during cancer and aging. Applications accepted until January 31.

Postdoctoral positions are open at the McAlpine lab, Icahn School of Medicine at Mount Sinai. If you want to explore the role of immune cells in cardiovascular and neurodegenerative diseases, please reach out to the group.

Gero is looking for a data scientist to help them in their mission to develop new therapeutics and biomarkers against aging and complex diseases.

Retro is now offering a Graduate Research Fellowship, which is a great opportunity for undergraduates who do not want to do a PhD, but want to become scientists. Retro works on partial reprogramming, blood factors and autophagy for cellular rejuvenation.

News

Eisai’s approach to US pricing for Leqembi (Lecanemab), a treatment for early Alzeimer’s disease, sets forth our concept of “societal value of medicine”in relation to “price of medicine”

The study with more than 800 patients (early Alzheimer’s) showed Leqembi slowed physical and mental decline by 27% over an 18-month period. The average cost of the drug will be $26,500 per year. Eisai defended the price based on the drug’s value to society, but it is likely to cause concern and draw scrutiny because of its potential wide use.

Hevolution Foundation Appoints William Greene, MD as Chief Investment Officer

Dr. Greene Draws on a Deep Reservoir of Experience as a Venture Capitalist, Impact Investor and Biotech Entrepreneur

Former Aging Cell editors establish new journal - Aging Biology

After a dispute resulting in the resignation of editors-in-chief from the journal Aging Cell, the former editors have now founded a new journal - Aging Biology - publishing research in the field of aging. 

Pfizer Takes First Small Step Into Longevity Field With Gero Collaboration

Big pharma dipping its toes in longevity with Peter Fedichev’s Gero collaboration

Articles

The start-ups seeking a cure for old age

As a Doctor, I See Aging Differently

A Drug to Treat Aging May Not Be a Pipe Dream

Two research teams reverse signs of aging in mice

DAOs Might Be Cure for Biotech Startups and New Drug Development

Dietary Restriction Works in Lab Animals, but It Might Not Work in the Wild

U.S. life expectancy continued to fall in 2021 as covid, drug deaths surged

Robust Mouse Rejuvenation project details announced

Urine test hope for early liver cancer diagnosis

Inside the $3bn mission to reverse signs of ageing

Can ageing be cured? Scientists are giving it a try

This biotech startup says mice live longer after genetic reprogramming

The Rich’s Search for the Key to Immortality Is Harmful to the Rest of Us

I left my CTO job to work on longevity

The Buck Institute, where the promise of aging research isn’t longevity

How to Be 18 Years Old Again for Only $2 Million a Year

Conferences and Webinars

The first ever DeSci London Conference was a resounding success! Hosted over 2 days in January at the esteemed Francis Crick institute in London, it featured talks from the who's who of the DeSci world, with Ethereum founder Vitalik Buterin even dialling in to share his thoughts on DeSci. Live recordings from the conference coming soon….

Podcasts and Videos

Why Aging is a Disease With David Sinclair | EP #18 Moonshots and Mindsets

Repairing Cells and Longevity Myths with Dr. Charles Brenner

Remarkable Science: Understanding lifespan and why aging might be reversible with Dr. David Sinclair

Lessons From The World's Longest Happiness Study - Dr Robert Waldinger | Modern Wisdom Podcast 578

Longevity Tweet of the Month

Sebastian J. Hofer:

I asked #ChatGPT how to achieve #healthy #aging. Can't argue with its reasoning. Next, I asked which interventions should be combined to achieve maximal #lifespan extension in mice (see comment). So, who will try this? And who will fund it? 

@impetusgrants @agingdoc1

Follow the link to see ChatGPT’s response!

Longevity Resources

João Pedro de Magalhães hosts an educational and information resource on the science of aging: https://www.senescence.info/

A great resource showing Longevity biotech companies and their investors: https://agingbiotech.info/investorsXcompanies/

.

Interview with Prof Björn Schumacher

Prof Schumacher is a director of the Institute for Genome Stability in Ageing and Diseases (IGSAD) at CECAD Research Centre of the University of Cologne, a president of the German Society for Ageing Research (DGfA), a Vice President of the German Society for DNA Repair (DGDR) and serves on several editorial boards.

What inspired you to enter longevity research?

That happened during a biology class at high school, when I realised how little we knew about our own biology. We are all subject to the invariable fate of ageing but we don’t understand that process at all. I knew I had to study the biology of ageing even though that wasn’t even a research field back then. So I decided to study biology and over time I would find out what would be the most important aspects of biology to study in order to illuminate ageing.

Which of the current theories of ageing do you think are the most convincing?

The theory of ageing is pretty much settled. It is really clear that ageing is a consequence of the lack of natural selection to indefinitely maintain the soma after the genes have been passed on to the subsequent generation. This is reflected in Tom Kirkwood’s disposable soma theory but also in the mutation accumulation and the antagonistic pleiotropy theories. The soma is only the vehicle for the indefinite maintenance of the germline throughout the generations.

How has the field changed since you started?

The field has really expanded phenomenally in the past two decades. When I started there was some telomere research that was super exciting and the genetic work in C. elegans that was really driven into the spotlight by Cynthia Kenyon. The most important development in the field over the past two decades was that it attracted scientists from diverse fields. This has made the field much stronger because ageing is affecting about every biological process.

What mistakes do you think the longevity field has made?

Some of those big claims and great expectations have had an inflationary effect on expectations. “Ageing reversal”, “rejuvenation” are big words with very little factual biology to back it up as true physiological reversal of ageing. While it is great to inspire with exciting concepts, it is also important to realise that ageing is complex and we have only grasped the tip of the iceberg.

Other than your own, what do you think have been the biggest/important discoveries in the field?

There were some real breakthrough discoveries. Conceptually, Tom Johnston’s and Cynthia Kenyon’s discovery of genetic mechanisms of ageing in the early 1990s, and Jan Hoeijmaker’s demonstration that DNA repair defects promote pretty much the entire set of ageing phenotypes in the early 2000s were really ground-breaking. The transformative discoveries of stem cell reprogramming by Shinya Yamanaka’s and Steve Horvath’s pioneering work on ageing clocks probably have the most practical consequences when it comes to interventions.

What advice would you give to people currently working in longevity research?

It is very important to study ageing with open eyes and receptive minds. The biological processes that impact ageing are intimately interconnected. We must further thrive to understand the physiology of ageing, which is why organisms are so important as study models. Often people stay within their own silos and are ignorant of other aspects of biology.

Which aspect of longevity research do you think requires more attention?

I think the integration of model organism and human data need to be much more proactively pursued. Cancer research has shown us how difficult it is to transfer interventions that work in mice to humans. Transferring geroprotective interventions will be hugely more difficult because ageing is an integration of long-lasting trajectories that are impacted by all those distinct genetic variants and epigenetic effects that each individual human is subjected to but are unaccounted for in laboratory animals.

Is ageing a disease?

Ageing is a physiological process but not a disease. However, it is the cause of all chronic diseases for which age is the primary risk factor. Therefore, we need to target the ageing process itself to prevent age-related diseases. Given the demographic change this is our only option for a future with 2 billion elderly whose active participation in society to a significant degree depends on their health. It is as urgent as fighting climate change to fight the multimorbidity that is affecting a growing proportion of the population. We need to boost ageing research now!

Can you discuss the relative importance and potential causative relationship between DNA damage and epigenetic alterations in aging with examples from your own work?

DNA damage is occurring all the time. Our genome is inherently unstable and requires constant repair. The DNA in our cells not only encodes all information but in contrast to all other molecules, the genome cannot be replaced. The response to DNA damage affects about every process in the cell. We found that the longevity assurance mechanisms that regulate lifespan respond to DNA damage and increase the tolerance of DNA damage accumulation. Epigenetic mechanisms are also a very important regulatory response to DNA damage. We recently found that epigenetic regulators mediate the recovery of protein homeostasis following the repair of DNA damage that interferes with transcription. This finding links three causal hallmark processes of ageing: Genome instability, epigenetics, and protein homeostasis.

Although there has been a lot of interest in epigenetic aging in recent years, there has been less excitement and success stories around DNA damage and repair. Why do you think this is the case? What are the biggest challenges in the field?

There are two important factors here: One is the complexity of the DNA repair machineries and the second one is technology. Epigenetic modifications are very easy to detect and quantify. DNA damage is much harder to detect because of the sheer number of distinct lesion types. There has been tremendous progress just in the past year to detect mutations, which are consequences of DNA damage and it has become clear that the rate of somatic mutations is highly correlated with the lifespan of a species. Despite the complexity, targeting ageing at the most fundamental level involves developing strategies for augmenting genome stability.

What do you think are the most viable therapeutic avenues for targeting DNA damage?

For human ageing DNA repair is absolutely fundamental as the pathologies of congenital disorders caused by mutations in DNA repair genes clearly demonstrate. DNA repair mechanisms are as complex as the plethora of lesion types in the genome. We are working on mechanisms that can augment the entire set of DNA repair machineries. The key lies in the germ cells, because they repair far more effectively than somatic cells as their genomes are maintained indefinitely. Our concept is to confer germline-like repair efficiency to somatic cells.

And how likely are they to increase healthspan and lifespan from what we know in animals?

The realization that both healthspan and lifespan could be increased in organisms is one of the defining concepts of ageing biology. The plasticity of lifespan is quite distinct in species. The nematode C. elegans is the gift that keeps on giving because of its vast plasticity of lifespan, which is an adaptation to the boom and bust economy in the ecological niche of this species. Amid ample food they reproduce exponentially and upon starvation they can outlive a normal developmental cycle at least ten times. Such a degree of plasticity doesn’t exist in more complex animals. Nonetheless, some of the centenarians are healthy until the last year before their death at very old age. So also in humans, it is perfectly possible to extend healthspan to the 120 years that are currently thought to be the upper limit of human lifespan. This would be truly transformative for human health and prerequisite for a demographically changed society that is inclusive and harmonious.

Outro

Thank you for staying with us till the very end and as always we encourage you to reach out to us about content you’d like us to discuss in our next issues.

Further Reading

SIRT6 is a key regulator of mitochondrial function in the brain

Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration

Gut microbiota as an antioxidant system in centenarians associated with high antioxidant activities of gut-resident Lactobacillus

Alcohol consumption and epigenetic age acceleration in young adults

Biochemistry and Cell Biology of Ageing: Part III Biomedical Science

Convergent genomics of longevity in rockfishes highlights the genetics of human life span variation

Midnight snacks might shorten your life - lifespan and healthspan advantages of eating less and at the right time

The importance of reaction norms in dietary restriction and ageing research

Unravelling Effects of Anti-aging Drugs on C. elegans using Liposomes

Is Aging a Disease? The Theoretical Definition of Aging in the Light of the Philosophy of Medicine

Mechanisms of spermidine-induced autophagy and geroprotection

Meta-hallmarks of aging and cancer

Age Discrimination in the Context of Motivation and Healthy Aging

ERCC1 mice, unlike other premature aging models, display accelerated epigenetic age

A concerted increase in readthrough and intron retention drives transposon expression during aging and senescence

The Longevity field is starting the year strongly with new updated Hallmarks of aging, exactly 10 years after the original paper was published. So time to update your introduction slides with the figure we will see in every presentation for the next
Read more
VitaDAO Closes $4.1m Fundraising Round With Pfizer And Shine Capital
January 30, 2023
Awareness
Longevity
VitaDAO Closes $4.1m Fundraising Round With Pfizer And Shine Capital

2023 kicks off with strong support for longevity research and a huge step for decentralized science(DeSci) 

We are excited to announce that we have closed a $4.1m fundraising round from strategic members including Pfizer Ventures, Shine Capital, L1 Digital; decentralised science and web3 organisations Beaker DAO and Spaceship DAO; and longevity enthusiasts including Balaji Srinivasan (former CTO of Coinbase and General Partner at a16z) and Joe Betts-LaCroix (Retro Biosciences) and many others.

The proceeds from the fundraise will be used to fund further longevity research projects and precede the spinout of VitaDAO's first biotech startups, scheduled in the first quarter of 2023, as well as further investment in commercialization and licensing of its IP-NFT assets through its partnership with Molecule DAO.

“2022 was a highly successful year for VitaDAO, and the quality of research we have seen has been impressive,” says Laurence Ion, Steward of the Dealflow Working Group. “We’ve also seen some of our projects move rapidly to the next stage in development -- such as entering into clinical trials with Mantis Photonics and new data from the Morten Scheibye-Knudsen Lab at the University of Copenhagen and the Victor Korolchuk Lab at the University of Newcastle. This has further proven that a highly aligned community can contribute to advancing the science and clinical innovation in longevity research.”

The fundraising is an important milestone for VitaDAO and will allow the organisation to accelerate its mission of extending human life and healthspan. With these new funds, VitaDAO will be able to fund cutting-edge longevity research, commercialise new technologies that target human aging, and build out its global community of experts and enthusiasts.

Pfizer is the first pharma to vote on DAO proposal (within VitaDAO), and also participates in VitaDAO in many other ways, including dealflow, scientific evaluation, incubation, and commercialization.

In 2022, we grew our community to include over 9000 enthusiasts, researchers, and contributors, as well as deployed over $3.5m in more than 15 projects researching the diseases of ageing, and the repair of damage associated with the hallmarks of ageing. 

All researchers are welcome to apply for funding, including professors, post-docs, research scientists, and even students. 

VitaDAO wishes to thank its contributors for their support.  

How to get involved

VitaDAO is defined as an active and diverse community of contributors.  Everyone can contribute to VitaDAO. We’re looking for researchers to identify and assess longevity projects and non-researchers to help with tech, legal, awareness, and operations If you’d like to get involved, a great first step is to join our Discord.

Do you want to stay up to date with VitaDAO? Subscribe to our newsletter, add the vitadao calendar, and visit our twitter

For any media requests, reach out to dao@vitadao.com and check out our press kit.

2023 kicks off with strong support for longevity research and a huge step for decentralized science(DeSci)
Read more
VitaDAO Letter: Year in Review + Interview with VitaDAO Co-initiator, Tyler Golato
January 17, 2023
Sarah Friday
Awareness
VitaDAO Letter: Year in Review + Interview with VitaDAO Co-initiator, Tyler Golato

VitaDAO Letter: Year in Review + Interview with VitaDAO Co-initiator, Tyler Golato 

What a year it has been! 2022 was a year full of growth, science, and a whole lot of funded longevity research. In the 1.5 years since VitaDAO’s historic Genesis auction, VitaDAO has funded 15+ longevity projects, gathered 9000+ community members, and continued to lead the decentralized science movement. Here at VitaDAO, as a new year begins, we’re filled with gratitude. Travel with us as we highlight some of the craziness that was 2022!

 

New Here?

VitaDAO is a decentralized organization made up of individuals from across the globe working to fund early-stage aging research and spin out longevity biotech startups. VitaDAO is focused on maximizing healthy human lifespan. 

 

In the coming year, we hope you’ll join our mission. Want to get involved in the longevity field? Contribute to VitaDAO and receive $VITA tokens in return. Join the VitaDAO community on Discord and stay up to date on Twitter! Want monthly updates on what’s happening at VitaDAO? Subscribe to this Newsletter to stay up-to-date. 

2022 in Review

Below, read about what went DAOn in 2022. Buckle up! Even though this is not an extensive list, it’s still a lot!

January

 February

March

April

  • VitaDAO hosted its first Crypto meets Longevity Symposium. This was the first-ever conference held with the intent to explore the integration of longevity and web3 and consisted of over eighteen scientists, blockchain experts, and longevity professionals. 
  • VitaDAO hosted three panel discussions:

May

June

July

  • Multichain voting went live! As a result, DAO members can vote on both the Ethereum and Gnosis chains.
  • VitaDAO launched the Longevity Prize - a crowdsourced longevity prize to unlock progress in longevity research and recruited 3 top organizations as partners: Foresight Institute, Methuselah Foundation, and Lifespan.IO.  We raised over $250k in prizes through the Gitcoin Longevity Round we initiated, with support from thousands of community members, whose donations were matched by VitaDAO, Vitalik Buterin, and Stefan D. George.
  • Laura Minquini and Ines Silva hosted a VitaDAO panel about Gender Biases in Research.

August

 September

October

 November

  • VitaDAO’s Institutional Genesis Raise was a huge success. VitaDAO gained amazing strategic contributors committed to building a public good and participating in the governance of VitaDAO using $VITA tokens. Below, find a list of the contributors and their contributions: 
  • Pfizer Ventures - $500k + dealflow, commercialization & pharma awareness
  • L1 Digital - $500k + tokenomics & governance support
  • ShineCapital - $1M + positioning, fundraising, partnerships & tokenomics support
  • BeakerDAO - $75k + dealflow, incubation of spinouts, awareness & connections
  • and more (including Balaji Srinivasan, SpaceshipDAO, and Retro Biosciences)

December

  • We implemented shielded voting. Now, votes can be private during the voting period and revealed with each proposal closure.
  • The VitaDAO community approved delegated voting. VitaDAO members can delegate their voting rights to a delegate and wallet of their choice.
  • Top contributors in the Longevity-Dealflow Working Group were allocated 900,000 VITA tokens over 4 years.

VitaDAO Numbers: The Low Down

$3.5M+ Research Funded

250+ Projects Sourced

1300+ Token Holders

9000+ Community Members

15,000+ Twitter followers 

Meet: VitaDAO Co-initiator, Tyler Golato

Tyler Golato graduated from Stockton University before completing research fellowships at Columbia University and the National Institute on Aging. As a researcher, he has a background in experimental therapeutics and biogerontology. As an entrepreneur, in February 2021, Tyler Golato co-initiated VitaDAO. Today, Tyler also serves as Chief Scientific Officer at Molecule AG, a platform created to facilitate decentralized, modular, and collaborative drug development. 

Can you speak to your interest and journey in biogerontology?

 

I started out my career aiming to be a physician. As I progressed in that, I became aware of how primitive medicine is as an applied practice. Beyond communicable disease, there is not much that we can cure. Most conditions are simply managed, at best. 

 

I had an interest in oncology, and in particular, intractable cancers such as glioblastoma and pancreatic cancer. As an oncologist, you mostly apply a standard of care and the outcomes fall along a predictable statistical distribution. Most prognoses are poor, and we have not advanced much regarding cancer therapy, across all cancers, over the last 50 years. Thus, I became more interested in research and trying to understand how I could advance the field.

 

I joined an experimental therapeutics laboratory at Columbia University where I worked with Dr. Robert Fine. I found the work very interesting and rewarding, but most of the developments and wins in pancreatic and brain cancer were focused on stabilizing or shrinking tumors and extending overall survival by months. I was really much more interested in how we might prevent cancer, or even mutation, altogether. 

 

With this in mind, I became more interested in biogerontology. When you realize how primitive medicine is and how poorly our reactive, treatment-focused health care system treats conditions (sick care, not health care), it becomes more attractive to consider how we might prevent disease. In my mind, biogerontology was the closest thing that existed to preventative medicine as a research domain. Most chronic disease and mortality is age-related. If we could understand the driving causes of aging, perhaps we could affect them and therefore also age-related diseases. Further, I was interested in the metaphysical aspects of fighting aging as a conquest. This always inspired a sense of awe in me, as I imagine it does for most people, and certainly has for humanity throughout millennia. 

 

When I finished at Colombia, I joined the National Institute on Aging in the Laboratory of Molecular Gerontology under David M Wilson and Vilhelm Bohr. My focus was on DNA damage and repair, and segmental progerias, which recapitulate some segments of the aging process. I really enjoyed my time at the NIA, but ultimately decided I wanted to focus on something that could improve the overall system of science funding and healthcare at the incentive level, as opposed to being a researcher. 

 

Before founding Molecule and later, VitaDAO, were you involved in Web3/decentralized technologies?

 

I interacted with Bitcoin as early as 2013, and found the technology curious. I was also interested in Ethereum from 2015 or so, but primarily around innovations in governance the ideas that were brewing around The DAO. After that failed, I remained skeptical about the general maturity of the technology. While I found it intriguing, I didn’t see many near-term relevant use cases. It was only in 2017/2018, when I met Paul Kohlhaas, my co-founder at Molecule, and the technology started to mature, that I became seriously interested. I was mostly interested in novel approaches to mechanism and incentive design in science and drug development, specifically. Web3 provided a clear architectural framework where one could create a sandbox environment to play with incentive and governance designs. This excited me a lot. 

 

How did you first come up with the idea of VitaDAO? 

 

VitaDAO is really an amalgamation of ideas. Before VitaDAO, Molecule had been working on the premise of creating a decentralized drug development protocol. We always thought about this through the lens of using IP ownership as a distributed incentive. What if, instead of a company owning a patent, a group of disparate collaborators - patients, researchers, VCs, pharmas, academic labs, biotechs - all worked together and contributed to the development of a drug in exchange for partial ownership in IP? This idea of turning IP ownership into a fractional incentive was really exciting to me, given how modular drug development had become, but also given the fact that there was often a missing incentive for collaboration. 

 

With this idea in mind, we began working on an IP-NFT framework for funding research projects. This is at the core of Molecule’s and VitaDAO’s technology stack today. 

 

VitaDAO was really the first working proof of concept to bring those principles together in a unified structure, focused on biogerontology, which is my passion. We believed that by bringing together a community of diverse stakeholders from all aspects of biogerontology, we could build a more robust organization than a biotech company, a nonprofit, a university, or a fund. While it is still a work in progress, I believe we are beginning to see the validation of that thesis. 

 

What do you think is VitaDAO's greatest strength? 

 

VitaDAO’s biggest strength is its community, and the fact that anyone capable of adding value can easily become a core part of that community. VitaDAO has a lower barrier of entry than a startup or a traditional org - you can show up in the Discord and if you have something the organization needs, you can become productive quickly. This is incredibly unique and enables a constant influx of talent and dealflow. Additionally, I think VitaDAO’s unique structure - one focused on creating a sustainable organization that is not distinctly for or non-profit - gives the organization the ability to succeed and rise up in a field currently bogged down by poorly designed incentives and a lagging status quo. 

 

What has surprised you the most about VitaDAO?

 

The thing that has surprised me most about VitaDAO is the extent to which it has been responsible for catalyzing the broader DeSci movement. There has been an explosion of new bioDAOs and DeSci orgs seeking to do something similar to VitaDAO, but in different therapeutic areas or with slightly different design parameters. This has been hugely exciting and rewarding to watch (and participate in), as it creates a real sense of legacy for one’s work. I take a lot of joy and comfort knowing that independent of the success of any of these organizations, the spirit of DeSci and making science more open, accessible, collaborative, and democratic is virtually unstoppable at this point. 

 

How do you see VitaDAO evolving in the future? - What is something you believe today that you didn't believe ten years ago?

 

VitaDAO is going to become laser-focused on translating its assets to market. Our first year was really focused on funding projects. Now, we will become more focused on incubating these projects, spinning them out, and really proving our thesis end-to-end. My hope is that the organization will continue to grow and thrive, and that we will be able to fund total moon shots - things that traditional organizations wouldn’t touch, where the risk is too high (but the reward also asymmetric). I believe VitaDAO will become one of the top 5 go-to sources for funding in biogerontology, and that we are at the beginning of a journey towards DAOs becoming the default organizational structure for funding and incubating assets. 

 

There are many things that I believe today that I did not believe 10 years ago. Technology is advancing so quickly that anyone who believes they can see 10 years into the future is likely wrong. 10 years ago, I couldn’t see the extent to which technology - things like web3, AI, etc - would completely transform how science is conducted. 

 

I’ve recently written about my vision for the future of science and drug development here: https://www.molecule.to/blog/the-scientific-singularity-our-vision-for-the-future-of-research-with-ip-nfts. This is a future I could not have fully imagined even 5 years ago, but I now have a high degree of confidence we will see this reality soon. We are already beginning to, and VitaDAO is playing a major role. 

Upcoming Events

Hackathon: Jan 20th - 22nd - Join VitaDAO’s Hackathon - a remote, cross-disciplinary hackathon focusing on the future of longevity. Registration is now open! 

 

VitaDAO @ Longevity Med Summit: May 4-5, 2023 - Come join VitaDAO in sunny Lisbon, Portugal at the 2023 Longevity Med Summit. Here, join other longevity enthusiasts in learning about the development of anti-aging science, the exploration of new regenerative and longevity therapeutics, and the limiting R&D challenges faced in therapy development.

Check out VitaDAO’s calendar and view all our upcoming events: https://vitadao.com/calendar 

What a year it has been! 2022 was a year full of growth, science, and a whole lot of funded longevity research. In the 1.5 years since VitaDAO’s historic Genesis auction, VitaDAO has funded 15+ longevity projects and gathered 9000+ community members
Read more
December Longevity Research Newsletter
December 20, 2022
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
December Longevity Research Newsletter

Introduction

As the end of the year is fast approaching we wanted to look back and appreciate all the effort and hard work of the researchers in the VitaDAO community - from students, postdocs and fellows to principal investigators who have been awarded VitaDAO funding - this month, in addition to our hotpicks, we bring you a collection of 2022 papers from our Vitalians.

We are fortunate enough to work with experts from across the aging field and in this issue we are delighted to share with you an interview with VitaDAO Steward, Asst. Prof @ WashU and BIOIO founder - Tim Peterson.

Longevity Literature Hot Picks

Aging is associated with a systemic length-associated transcriptome imbalance

https://www.nature.com/articles/s43587-022-00317-6

Transcript size matters. Transcriptome analysis reveals the length of the transcripts alone can account for most age related transcriptional changes in both mice and humans. There is lower abundance of long transcripts with age and the longest transcripts enrich for longevity related genes. Importantly this abundance issue could be counteracted by interventions like rapamycin, FGF21 and others.

Deep phenotyping and lifetime trajectories reveal limited effects of longevity regulators on the aging process in C57BL/6J mice

https://www.nature.com/articles/s41467-022-34515-y

We have discovered numerous interventions which extend lifespan, but do any of these actually affect the ageing process per se, or do they affect specific pathologies? This study performs deep phenotyping on numerous putative anti-ageing interventions (PAAIs) and finds that many have no effect on the ageing phenotype.

In vivo partial reprogramming by bacteria promotes adult liver organ growth without fibrosis and tumorigenesis
https://www.cell.com/cell-reports-medicine/fulltext/S2666-3791(22)00379-2

The fascinatingly odd paper about bacteria with natural partial reprogramming capability in armadillos shows in vivo de novo organogenesis. This bacterial hijacking induced reprogramming to progenitor state and drove liver growth, with preserved function, architecture and without tumorigenesis.

Association of spermidine plasma levels with brain aging in a population-based study

https://pubmed.ncbi.nlm.nih.gov/36321615/

While other studies show that spermidine supplementation might be beneficial in healthy aging, this population-based study raises some concerns. Increased levels of spermidine in plasma can potentially be an early biomarker for AD as it showed association with advanced brain aging.

Manipulation of the diet–microbiota–brain axis in Alzheimer’s disease

https://www.frontiersin.org/articles/10.3389/fnins.2022.1042865/full

More and more evidence is uncovered that microbiome is connected to everything in our body, including the brain via the gut-brain axis. It is unclear if the altered microbiota in the gut is a cause, a consequence of AD or both via feedback loops, we do see that certain probiotic treatments can prevent AD onset. 

Rapamycin improves the quality and developmental competence of in vitro matured oocytes in aged mice and humans

https://www.aging-us.com/article/204401/text

Beneficial effects of rapamycin have been described across the body, and now even down to the oocyte level. Culturing oocyte cells from mice and humans in rapamycin supplemented medium increased in vitro maturation efficiency and oocyte quality as shown by reduced ROS, chromosome aberrations and DNA damage markers (γ-H2AX).

CSF proteome profiling across the Alzheimer’s disease spectrum reflects the multifactorial nature of the disease and identifies specific biomarker panels

https://www.nature.com/articles/s43587-022-00300-1

Cerebrospinal fluid (CSF) is the liquid filling the space in and around the brain. It may be used, this study suggests, as a biomarker and diagnostic for AD. Analysing the proteins from CSF samples can not only identify disease state, but even help scientists distinguish between mild cognitive impairment with amyloid, AD dementia and non-AD dementia.

Could aging evolve as a pathogen control strategy?

https://www.sciencedirect.com/science/article/abs/pii/S0169534722001987?dgcid=author

There are numerous theories as to why organisms age, with most suggesting ageing is either evolutionary programmed or an accumulation of damage. Here the authors propose the pathogen control hypothesis suggesting that “defense against infectious diseases may provide a strong selection force for restriction of lifespan”. 

Biological Age Predictors: The Status Quo and Future Trends

https://www.mdpi.com/1422-0067/23/23/15103

To validate the efficacy of longevity interventions, it will be important to develop robust biomarkers for ageing. Here the authors provide a thorough review on currently used ageing biomarkers and what the future may hold.

A Glb1-2A-mCherry reporter monitors systemic aging and predicts lifespan in middle-aged mice

https://www.nature.com/articles/s41467-022-34801-9

A new mouse model in which beta-galactosidase is tagged with a fluorescence marker. The authors observe an increase of this reporter signal with age and is associated with cardiac hypertrophy and decreased lifespan.

A short history of saturated fat: the making and unmaking of a scientific consensus

https://journals.lww.com/co-endocrinology/Fulltext/9900/A_short_history_of_saturated_fat__the_making_and.42.aspx

A thought-provoking review arguing that the supposed link between saturated fats and heart disease was based on poor evidence, yet it became an unchallenged dogma. This resulted in policy making which, even in light of evidence to the contrary, has become difficult to alter due to bias and financial conflicts of interest.

2022 Papers from the VitaDAO Community

LipidClock: A Lipid-Based Predictor of Biological Age https://www.frontiersin.org/articles/10.3389/fragi.2022.828239/full

Max Unfried is Vita core and a key member of VitaDAO Awareness. He’s also a PhD candidate in the labs of Brian Kennedy and Morten Scheibye-Knudsen. Here is some of his work on LipidClocks, which can predict survival curves in agreement with lifespan experiments.

Beer, wine, and spirits differentially influence body composition in older white adults https://onlinelibrary.wiley.com/doi/10.1002/osp4.598

Tovah Wolf is an active contributor and also a scientist and a consultant. A UK Biobank study from her team reveals that while beer and spirits could contribute to adipogenesis, however red wine may be protective of it. White wine even predicted higher bone density. Finally some good news, but we’d be very curious to see this teased apart and what mechanisms might be at play.

New hallmarks of ageing : a 2022 Copenhagen ageing meeting summary

https://ruj.uj.edu.pl/xmlui/handle/item/303202

Tomas Schmauck-Medina and Adrian Moliere are active contributors who not only published this summary of the ARDD conference in Aging but also an article on rapamycin for VitaDAO. They are students in the lab Evandro Fang, who was one of the recipients of Vita funding. Their team has been particularly productive so here are a few more papers from the lab:

Mitophagy and neuroinflammation: a compelling interplay

https://europepmc.org/article/med/35762540

WIPI2 positively regulates mitophagy by promoting mitochondrial recruitment of VCP

https://www.tandfonline.com/doi/full/10.1080/15548627.2022.2052461

And even a book chapter - Molecular linkages among Aβ, tau, impaired mitophagy, and mitochondrial dysfunction in Alzheimer’s disease

https://www.sciencedirect.com/science/article/pii/B9780323899062000071

Clinical Trials Targeting Aging
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9261384/

Nuclear morphology is a deep learning biomarker of cellular senescence

https://www.nature.com/articles/s43587-022-00263-3

Two impactful papers from the lab of our first funding recipient and Vita supporter Morten Scheibye-Knudsen. 

Short senolytic or senostatic interventions rescue progression of radiation-induced frailty and premature ageing in mice

https://elifesciences.org/articles/75492

Activation of autophagy reverses progressive and deleterious protein aggregation in PRPF31 patient-induced pluripotent stem cell-derived retinal pigment epithelium cells

https://onlinelibrary.wiley.com/doi/full/10.1002/ctm2.759

NDP52 acts as a redox sensor in PINK1/Parkin-mediated mitophagy

https://www.embopress.org/doi/full/10.15252/embj.2022111372

Another one of our grantees, Viktor Korolchuk, has published numerous papers across the hallmarks of aging with a few highlighted examples here.

Clinical Trial Updates

BioAge Announces Positive Topline Results for BGE-105 in Phase 1b Clinical Trial Evaluating Muscle Atrophy in Older Volunteers at Bed Rest

https://www.businesswire.com/news/home/20221205005201/en/BioAge-Announces-Positive-Topline-Results-for-BGE-105-in-Phase-1b-Clinical-Trial-Evaluating-Muscle-Atrophy-in-Older-Volunteers-at-Bed-Rest

BioAge is pleased to announce positive topline Phase 1b data showing that their apelin receptor agonist BGE-105 was able to attenuate muscle atrophy in healthy volunteers aged ≥65 years following 10 days of bed rest. 

Funding Opportunities

British Society for Research on Ageing

https://bsra.org.uk/funding/

1. Small Research Grant (equipment & publication costs)

2. Travel Grant (attending biology of ageing conference)

3. Summer Studentship (for undergraduates)

Scholarship for the Longevity Biotech Fellowship

https://www.longbiofellowship.org/scholarships

VitaDAO funded a $2k grant for people who can't afford, but would greatly benefit from attending https://longbiofellowship.org - its a non-profit community for people to come together to build, join, or invest in revolutionary longevity biotechnology projects.

Job board

The Kane lab at the Institute for Systems Biology is seeking a highly motivated Postdoctoral Fellow

http://isbscience.hrmdirect.com/employment/job-opening.php?req=2101816&&#job

The research project focuses on understanding, measuring and predicting heterogeneity in aging, and particularly identifying determinants and biomarkers of frailty. And apparently “postdoc salaries in Washington can't be beat!” ISB pays postdoctoral fellows on a scale based upon experience: $65,478 - $91,711 annually

Deciduous Therapeutics are looking to hire a research scientists and associates

https://www.deciduoustx.com/careers/

Deciduous Therapeutics are an early stage company working on endogenous immune activation to treat age-related diseases, via senescent cell elimination. Based in San Francisco (on site role)

News

Congratulations to Marco Demaria for being appointed Editor in Chief of npj Aging!

https://www.nature.com/npjamd/

The Longevity Biotech Fellowship

https://www.longbiofellowship.org/

The Longevity Biotech Fellowship (LBF) is a non-profit community for people to come together to build, join, or invest in revolutionary longevity biotechnology projects. LBF kicks off their first cohort in Jan 2023.

Peter Attia’s long-awaited longevity book “Outlive: The Science and Art of Longevity” will be released on 28th March 2023

https://peterattiamd.com/outlive/

Available to pre-order now wherever books are sold.

Cajal Neuroscience launches with $96M Series A, tackling Alzheimer's and Parkinson's

https://endpts.com/cajal-neuroscience-nets-96m-series-a-and-is-looking-to-go-after-neurodegeneration-targets/

Articles

Why is progress in biology so slow?

https://www.sam-rodriques.com/post/why-is-progress-in-biology-so-slow

A thought-provoking article on why Biology research is slow compared to other fields such as AI. The reasons proposed are: speed, knowledge and talent.

Funding science through blockchain technology and cryptocurrencies

https://from.ncl.ac.uk/funding-science-through-blockchain-technology-and-cryptocurrencies

A blog post from Newcastle University TTO Matt Abbott on Viktor Korolchuk’s VitaDAO-funded project

You may have heard about NAD but not really know what it is

https://post.news/article/2I5moQs7DrHbuc00sDLMQbuiTdY

A brief introduction to nicotinamide adenine dinucleotide (NAD) by Prof. Charles Brenner

Sarcopenia: What a Waste

https://neo.life/2022/12/sarcopenia-what-a-waste/

“Like memory loss, sarcopenia was traditionally seen as a normal part of aging, but is now recognized as a disease. It still lags far behind dementia in terms of research funding and everyday familiarity, partially because it’s ill-defined and poorly understood.”

Conferences and Webinars

Ageing Research at King's (ARK) Longevity Week event

https://www.kcl.ac.uk/events/longevity-week-2022-data-driven-longevity

You can see all recorded taks here:

https://www.youtube.com/@ageingresearchkings9013

Featuring our very own, newsletter co-author and research associate at King’s College London, Rhys Anderson

https://www.youtube.com/watch?v=FWC8M5lgq5c

DeSci London

15th-16th January - The Francis Crick Insitute, London, UK

https://www.desci.london/

Don't miss the 1st ever DeSci London Conference! Vitalik Buterin will be joining via Zoom to share his thoughts on DeSci.

Travel grants available: 

https://docs.google.com/forms/d/e/1FAIpQLSfDY2PJfAka82ick6NB-K3UFEyt1UrJ45naANim33U4lGDJPg/viewform

Drugs that slow ageing in mice | Prof Richard A. Miller

https://www.youtube.com/watch?v=UPqstwgr7x0

Podcasts and Videos

Evelyne Bischof: 100 Questions with Longevity Legends

https://www.youtube.com/watch?v=RjUg_VAwuZg

Infinite Monkey Cage Podcast: Can we cure ageing?

https://www.bbc.co.uk/sounds/play/p0dcn524

Peter Attia: Chris Hemsworth on Limitless, longevity, and happiness

https://peterattiamd.com/chrishemsworth/?utm_source=twitter&utm_medium=social&utm_campaign=221212-pod-chrishemsworth&utm_content=221212-pod-chrishemsworth-tw

Translating Aging: Hacking the Complex System of Aging (Peter Fedichev, GERO)

https://podcasts.apple.com/ua/podcast/hacking-the-complex-system-of-aging-peter-fedichev-gero/id1569628505?i=1000590133981

Longevity Tweet of the Month

Prof. Charles Brenner:

“animals have homeostatic functions that allow them to repair themselves

their capacity to do so declines after they reach reproductive prime

if you want to quantify aging, quantify repair capacity

if you want to test aging interventions, test maintenance of repair capacity”

https://twitter.com/CharlesMBrenner/status/1588549823183015937

Longevity Resources

Join Talent Bench - a database from the Norn Group (who brought us the Impetus Longevity Grants) to help people of all levels build out labs, companies, and careers!

https://norn.group/talent-bench

Longevity Apprenticeship Journal Club

https://docs.google.com/document/d/16edJ1vw5mfHKC2IQmcj-T73OjDLL8FK-mn7pxV375Lc/edit#

The Norn Group provides another public resource intended for people trying to learn about ageing biology. This document is the culmination of their year-long journal club from their Longevity Apprentices.

And here’s another journal club, written by Morgan Levine on Michael Rose’s early work on fecundity/longevity selection

https://www.nature.com/articles/s41576-022-00560-y

Interview with Tim Peterson

Tim Peterson has made some significant contributions to our understanding of longevity, especially in elucidating mechanisms of how the mammalian target of rapamycin (mTOR) protein functions. Tim is currently an assistant professor at Wash U, founder of BIOIO, and a VitaDAO Steward.

What inspired you to enter longevity research?

Several things. Aging encompasses most of human health, so everyone would benefit from the research. It's universal. It is a hard problem. 

Which of the current theories of ageing do you think are the most convincing?

That there are multiple causes.

How has the field changed since you started?

A move away from silver bullet thinking to a greater appreciation of how complicated biological networks are.

What mistakes do you think the longevity field has made?

Focusing on specific explanations as to why people age and get disease (Amyloid hypothesis, etc). Also I think categorising molecular biology into the hallmarks of aging has ossified our thinking.

Other than your own, what do you think have been the biggest/important discoveries in the field?

Yamanaka reprogramming and CRISPR seem to be the most impactful discoveries.

What advice would you give to people currently working in longevity research?

We need more people developing therapies. There’s always more knowledge to gain, but I feel like not enough people are trying to translate the knowledge to therapies.

Which aspect of longevity research do you think requires more attention?

Aging is a decline in function. Most small molecules inhibit function. For these reasons I don’t think small molecules will have a significant effect on lifespan. We need combination therapies involving manipulating multiple genes, such as epigenetic reprogramming. We wrote about it recently on our VitaDAO blog post titled “Synthetic Vitality”.

Is ageing a disease?

I get that this matters to the FDA, but I’m not sure I have much to add to this debate other than people should have more rights to try new medicines.

You have made some significant contributions to our understanding of the role of the mTOR pathway in ageing. Do you think rapamycin treatment is likely to be a successful longevity intervention for humans?

Any single agent is unlikely to make a significant effect. Even rapamycin, which is a best case scenario seems to max out at ~20%. 20% is phenomenal and the world will benefit, but we can do better.

What do you think is the biggest challenge in bringing rapamycin to the clinic to treat and prevent age-related diseases?

I think it’s coming. Some rapalogs look promising!

You have developed some novel senolytic drugs – which disease indications do you intend to target first and why?

We feel our senolytics should work in ALS and we have the tools and team to test them there, but we admit ALS is a challenging indication.

Which other interventions do you think hold promise for improving human healthspan/lifespan? 

Epigenetic reprogramming. Anything that would have synergistic effects.

You and VitaDAO have recently launched a new reviewing platform – The Longevity Decentralised Review (TLDR) (https://longevity.review/). Could you explain what this platform is and why people should get involved? 

TLDR is an on-demand peer review service. We built it to help people get feedback on their work ahead of submitting to journals. We also wanted to provide an option for peer review while people are waiting at journals. We want to separate peer review from journal brand.

VitaDAO have had an impressive 2022 with over 3 million USD allocated to fund projects, along with new initiatives such TLDR, The Longevity Fellowship, and The Longevity Prize. What does 2023 have in store for VitaDAO?

Besides funding hopefully ~10 projects, we are going to be fractionalizing IP-NFTs, and are launching an on-chain overlay journal, The Longevist.

Outro

Thanks for reading and staying with us until the last issue of the year! We hope to see you again next year and as always, feel free to reach out with requests on what you’d like to see explored or featured here, even any new year resolutions for the newsletter are welcome. 

We wish you a happy holiday season and hope you enjoy a well deserved break!

Further Reading

The impact of population-level HbA1c screening on reducing diabetes diagnostic delay in middle-aged adults: a UK Biobank analysis

https://link.springer.com/article/10.1007/s00125-022-05824-0

The Significance of NAD+ Biosynthesis Alterations in Acute Kidney Injury

https://pubmed.ncbi.nlm.nih.gov/36411195/

mTOR inhibition attenuates chemosensitivity through the induction of chemotherapy resistant persisters

https://www.nature.com/articles/s41467-022-34890-6

https://twitter.com/marc_dema/status/1594712799124529155

Cell division drives DNA methylation loss in late-replicating domains in primary human cells

https://www.nature.com/articles/s41467-022-34268-8

Molecular mechanisms of exercise contributing to tissue regeneration

https://www.nature.com/articles/s41392-022-01233-2

Sexual identity of enterocytes regulates autophagy to determine intestinal health, lifespan and responses to rapamycin

https://www.nature.com/articles/s43587-022-00308-7

Interaction of aging and Immunosenescence: New therapeutic targets of aging

https://www.sciencedirect.com/science/article/pii/S1567576922008815

Consumption of coffee and tea with all-cause and cause-specific mortality: a prospective cohort study

https://bmcmedicine.biomedcentral.com/articles/10.1186/s12916-022-02636-2

High-confidence cancer patient stratification through multiomics investigation of DNA repair disorders

https://www.nature.com/articles/s41419-022-05437-w

Watch this fun video telling the story behind the paper: https://www.youtube.com/watch?v=hLXsIveTbpc

As the end of the year is fast approaching we wanted to look back and appreciate all the effort and hard work of the researchers in the VitaDAO community - from students, postdocs and fellows to principal investigators
Read more
Synthetic Vitality
November 29, 2022
Tim Peterson
Awareness
Longevity
Synthetic Vitality

Therapies that act synergistically are needed to significantly promote longevity

The math behind the expression “aging is inevitable” is the Gompertz-Makeham law. This law describes the probability of death as a function of age. It reaches 99.9+% by the time we are 100. One doesn’t need to know math to realize this, but because you’re here the underlying formula is an exponential function.

“Aging is a math problem — multiplicative therapies is the answer.”

Exponents involve multiplication. Another way this can be understood is that multiple factors synergize to make us die. Factors like smoking, drinking, poor sleep, bad diets have multiplicative effects. They don’t add together. Like an evil 401K, they compound to kill us. Aging is a math problem — multiplicative therapies is the answer.

“Understanding that it is factors that synergize to cause us to die will be key to fixing how we make therapies.”

Understanding that it is factors that synergize to cause us to die will be key to fixing how we make therapies. For many decades now, since the rise to prominence of rational drug design, there has been a fixation on identifying silver bullet therapies. That is, if we can just find the right target and then drug it strongly and selectively enough, we can cure disease. The invention of Gleevac, which targeted the gene mutation BCR-ABL for the cancer type CML, added fuel to this fire. (You too can cure disease if you can just make your drug as good — potent, selective — as Gleevac!).

From the joke above, you can tell the CML example created a false expectation. CML is a monogenic disease. Targeting the single mutant gene with a single agent worked there, but it has nothing to do with polygenic diseases that are most diseases we care about (Alzheimer’s, most other common cancers, heart disease, etc.). The case is similar with two of the blockbuster drugs of the past decades — statins and ACE inhibitors. Those drugs have one job — to move a number down (cholesterol or blood pressure, respectively). That is not what’s being asked of most other drugs.

“Rapamycin is a start, but it is unlikely to promote the transformative longevity gains we are aiming for.”

For longevity, the current best case drug is rapamycin. Rapamycin has a 10–20% effect on lifespan in animals. Do the exercise and review the literature for other small molecules for any disease model. You’ll see that 20% improvements are a best case scenario in general. 20% is not good enough, especially because animal experiments are done in highly controlled conditions that are optimized to find an effect if there is one to be detected. This won’t be the case in people. All this is to say — Rapamycin is a start, but it is unlikely to promote the transformative longevity gains we are aiming for.

The longevity community needs to learn from the cancer community. More recently, the cancer community is focusing on synthetic lethality. “Synthetic” is a terminology geneticists use to describe what I’m referring to as synergistic or multiplicative.

What is a multiplicative therapy? I mean agents that have profound effects on physiology when combined. Their effects are not additive. They are multiplicative. 20 X 20, not 20 + 20. The former is 10X the effect of the latter. The Yamanaka four factors are perhaps the first bona fide example of a multiplicative longevity therapy. Combining four genes that can convert any cell to an embryo! Fortunately, people are already starting to leverage this technology to turn back the clock of old cells. Other strategies involving combinations of small molecules are emerging too.

“The radical view through the Overton window is that the FDA should require that a therapy hits multiple targets in order to be approved.”

The recently approved ALS drug, which is the only disease modifying drug for this fatal condition, is a combination of two agents. It’s not even known molecularly how they work! Considering one of the most valuable drugs ever Aspirin has 23 putative targets, I’m not sure why we haven’t done more to normalize ‘irrational’ drug design. The radical view through the Overton window is that the FDA should require that a therapy hits multiple targets in order to be approved. Otherwise I think if one soberly surveys what gets approved these days the cost/benefit analysis won’t be good. Even the aforementioned ALS drug, despite how promising it might be, is not sustainably priced at $158,000 per year.

Statins and blood pressure meds are what I call ‘counting’ medications. All we care about is the number going down. We’re rarely doing addition. We need to stop counting and start doing multiplication. Just as cancer needs synthetic lethality, aging needs synthetic vitality.

“Just as cancer needs synthetic lethality, aging needs synthetic vitality.”

At VitaDAO we are looking for multiplicative therapies. Rejuvenome is too. Here’s a starting list of projects doing multiplicative therapies. We’d love for you to add to the list.

Therapies that act synergistically are needed to significantly promote longevity
Read more
November Longevity Research Newsletter
November 15, 2022
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
November Longevity Research Newsletter

Introduction

A multitude of discoveries and inventions throughout history have been made by people who were initially trained in one domain, but were able to make a significant contribution to a different field due to their ability to look at a problem with a unique perspective. Would Charles Darwin still have conceived the Theory of Evolution if it were not for his background in Geology and appreciation that for geological processes to have occurred, Earth must have been around for a vast amount of time!

The contributions of physicists in particular to our understanding of biology have been immensely valuable over the past century. Great thinkers such as Szilard, Schrödinger and Crick, compelled to find out what life is and how it works, were able to provide humanity with novel ideas and trailblazing discoveries in molecular biology. 

Take this month’s issue as a tribute to the novel insights provided from physics with a few newly published papers, pre-prints and an interview with Dr. Peter Fedichev - highlighting the power of cross-disciplinary thinking to help advance longevity research! 

Longevity Literature Hot Picks

After another month of overwhelming amounts of great longevity research being published,  we hope to make it easier for you to keep up with it all by presenting some of our favourite papers from the field (and remember our Further Reading section at the bottom of this page for even more new longevity papers).

While we normally pick peer-reviewed published papers, there were a few interesting pre-prints this month which sparked a lot of debate that we couldn’t ignore!

Unsupervised learning of aging principles from longitudinal data

https://www.nature.com/articles/s41467-022-34051-9?fbclid=IwAR0nILUom4ulJUGm3RY_uKcmCFTAaJuqX9NuSLsSYSUUYZTbZoKJdfC8q4Q

What is the relationship between physiological changes with age and lifespan? To answer this question analytical and machine learning tools were employed to design deep artificial neural networks with auto-regression model, identifying a relationship between physiological state during aging and a variable named “dynamic frailty index” or dFI. dFI increased exponentially with age and predicted lifespan from mouse blood samples. It also responded accordingly to know life-shortening and life-extending interventions.

Aging clocks, entropy, and the limits of age-reversal

https://www.biorxiv.org/content/10.1101/2022.02.06.479300v2

A single variable called thermodynamic biological age (tBA) tracks entropy produced, and hence information lost, with age. As tBA increases with age it reduces resilience, and drives the exponential acceleration of chronic disease incidence and death risks, which is a linear and irreversible drift, setting severe constraints on age reversal possibilities. But wait, not all hope is lost. We might be able to figure out how to “cool down” the organism, control entropy and reduce the rate of aging in humans.

F1F0 ATP Hydrolysis is a Determinant of Metabolic Rate, a Correlate of Lifespan, and a Weakness of Cancer

https://www.biorxiv.org/content/10.1101/2021.10.28.466310v3

F1F0 ATP hydrolysis generates metabolic heat in an organism. A drug that can selectively inhibit ATP hydrolysis, but not synthesis, is explored here, and hypothesised that it could extend lifespan by slightly lowering body temperature. In 12 different species, decreased F1F0 ATP hydrolysis correlates with greater maximal lifespan. Anti-cancer properties were also observed in vitro.

Dietary restriction fails to extend life in stressful environments

https://www.biorxiv.org/content/10.1101/2022.10.17.512576v1

Dietary restriction has been shown to extend lifespan in numerous model organisms, however here the authors show that this effect is lost in fruit flies which are also under stress due to cold or hot living environments. This raises the question of whether laboratory-tested interventions will translate to animals living in more natural environments. 

Epigenetic reversal of hematopoietic stem cell aging in Phf6-knockout mice

https://www.nature.com/articles/s43587-022-00304-x

Aging leads to the accumulation of hematopoietic stem cells with reduced regenerative potential. The mechanisms behind this are unclear but this study shows that inactivation of the plant homeodomain factor 6 (PHF6), a single epigenetic regulator, rejuvenates mouse aged hematopoietic stem cells.

The commentary on the article:

Is the philosopher’s stone to rejuvenate blood stem cells an epigenetic regulator?

https://www.nature.com/articles/s43587-022-00305-w

Repurposing SGLT-2 Inhibitors to Target Aging: Available Evidence and Molecular Mechanisms

https://www.mdpi.com/1422-0067/23/20/12325/htm

A new take on CR mimetics with Sodium-glucose cotransporter 2 inhibitors (SGLT2-i), which lower glucose by elimination through urine, and actually induce a net loss of calories, fostering ketones and fatty acids utilization as glucose-alternative substrates. This process modulates major nutrient-sensing pathways held to drive aging, such as mTOR and resembles CR. Preliminary data also shows that it can inhibit cellular senescence and inflammaging.

Circadian transcriptional pathway atlas highlights a proteasome switch in intermittent fasting

https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01403-6

Intermittent fasting has gained traction among the longevity community as a safe lifestyle intervention hoping to improve healthspan and increase lifespan. The authors set out to discover if there was an internal "timer" to respond to fasting duration. They found that in mice, the hepatic proteasome switches leading to transcriptional resonance which is reversed upon re-feeding. It will be interesting to investigate if a similar mechanism is conserved in humans and what fasting duration is required to induce the switch. 

The role of gut microbiota in liver regeneration

https://www.frontiersin.org/articles/10.3389/fimmu.2022.1003376/full

Unlike other human organs, the liver has an amazing ability to regenerate itself. 

Check out this review exploring how gut microbiota play a role in liver regeneration through regulating the liver immune microenvironment thus modulating inflammatory signalling at different stages of the regenerative process.

Combining stem cell rejuvenation and senescence targeting to synergistically extend lifespan

https://www.aging-us.com/article/204347/text

The 2012 Nobel Prize for Physiology or Medicine was awarded for showing that differentiated cells could be reverted back to stem cells by activation of 4 transcriptional factors, now eponymously known as Yamanaka factors. This led to huge interest in the field including the founding of the multi-billion start-up Altos Labs. Another longevity target are senescent cells, the removal of which by either genetic or pharmacological means can increase lifespan in mouse models. Here the authors show that combining Yamanka activation with senescence removal has a synergistic effect on lifespan, with the largest effect resulting from transient treatment of both interventions.

Clinical Trial Updates

A Randomized Clinical Trial showing that a sustained low-carbohydrate diet might be a useful dietary approach for preventing and treating type 2 diabetes.

https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2797714

Meta analysis of aerobic exercise improving intelligence and cognitive function in patients with Alzheimer’s disease

https://journals.lww.com/md-journal/fulltext/2022/10210/meta_analysis_of_aerobic_exercise_improving.15.aspx

UNITY Biotechnology Announces Positive 24-Week Data from Phase 2 BEHOLD Study of UBX1325 in Patients with Diabetic Macular Edema

https://www.globenewswire.com/news-release/2022/11/01/2545288/0/en/UNITY-Biotechnology-Announces-Positive-24-Week-Data-from-Phase-2-BEHOLD-Study-of-UBX1325-in-Patients-with-Diabetic-Macular-Edema.html

News

JPMorgan Launches Life-Sciences Venture Group

https://www-wsj-com.cdn.ampproject.org/c/s/www.wsj.com/amp/articles/jpmorgan-launches-life-sciences-venture-group-11667300402

Crypto entrepreneur Justin Sun, founder of Tron, donate’s 51k USD to the Longevity Prize 

https://www.longevityprize.com/

Journal eLife eliminates reject/accept decisions from their review process

https://elifesciences.org/articles/83889

Articles

MIT Technology Review - The Mortality Issue

https://www.technologyreview.com/magazines/the-mortality-issue/#features

Featuring:

The debate over whether aging is a disease rages on

https://www.technologyreview.com/2022/10/19/1061070/is-old-age-a-disease/

Why the sci-fi dream of cryonics never died

https://www.technologyreview.com/2022/10/14/1060951/cryonics-sci-fi-freezing-bodies/

How scientists want to make you young again

https://www.technologyreview.com/2022/10/25/1061644/how-to-be-young-again/

Modern Vampirism: “Young Blood” Transfusions

https://peterattiamd.com/young-blood-transfusions/?utm_source=twitter&utm_medium=social&utm_campaign=221030-newsletter-blood-transfusion&utm_content=221030-newsletter-blood-transfusions-tw

Blood from young mice has rejuvenating effects on old mice, but it’s too early to translate these results into treatments for humans.

Upcoming conferences

A lot of us are conferenced-out for this year but there is one more meeting with an impressive line up we’d recommend:
The Longevity Summit

December 6-7 | Buck Institute for Aging | Novato, CA

https://longevitysummit.io/

While we’re waiting for more exciting conferences, make sure you’re caught up with the recordings from ARDD and VitaDAO’s own crypto-longevity symposium.

https://www.youtube.com/channel/UClOnplI2mzpJlwdX3vlOMJA

https://www.youtube.com/watch?v=-70SgGJPzTo

Evotec Webinar series focusing on 3 key areas:

  • Therapeutic approaches for aging and age-related disease
  • Aging and precision medicine
  • Why older adults should not be underrepresented in clinical trials. 

https://news.evotec.com/a-spotlight-on-aging-webinar-series

Funding Opportunities

Hevolution Foundation Announces Pilot Grants Program for Top Healthspan-Related Research in the United States

https://hevolution.com/en/w/hevolution-foundation-announces-pilot-grants-program-for-top-healthspan-related-research-in-the-united-states

Job board

The Schumacher lab is looking for a postdoc to join them at the Institute for Genome Stability in Ageing and Disease, CECAD-Cluster of Excellence in Aging Research, University of Cologne

https://schumacher.cecad-labs.uni-koeln.de/open-positions

Vita therapeutics, a cell engineering company using iPSCs, have two open positions - research associate and a quality assurance director:

https://www.vitatx.com/careers#openpositions

Head of Ops for Longevity Fund - $150K+ salary, higher if you’re significantly experienced. Looking for someone smart and capable with 3+ years of operating at a fast-paced startup or VC firm

email laura@longevity.vc if a fit! $10K referral bonus if you find someone great :)

Longevity Tweet of the Month

Even if you don’t use Twitter, it must have been near impossible to escape the news and debate surrounding Twitter’s new owner Elon Musk and his plans to charge for verification. The author Stephen King took umbrage to this and threatened to leave the platform. Alex Zhavoronkov had an amazing response, leading to the inauguration of our new ‘Longevity Tweet of the Month’:

Alex Zhavoronkov: “I will pay @StephenKing's $20/month twitter for the rest of his life if he writes his scariest book titled "The Biology of Aging" (the concept is so horrifying that after 40, you do not need a costume for Halloween anymore). With @davidasinclair as the savior protagonist  🎃👻💀”

https://twitter.com/biogerontology/status/1587477303671816192

Interview with Dr. Peter Fedichev

Dr. Peter Fedichev describes himself as a "physicist in drug discovery land". With a background in condensed matter physics, biophysics and bioinformatics he has been able to provide a unique perspective to longevity research, for example the 2 papers highlighted in this month's issue. He is currently the CEO and co-founder of the longevity biotech company GERO.

What inspired you to enter longevity research?

I am a physicist by training and did my studies in the field of strongly interacting and correlated systems. I am fascinated with phase transitions and emergence - the ability of complex systems to develop properties that do not exist on the level of their constituent parts. “More is different,” in P. Anderson's words, and we find novel interesting phenomena whenever it applies. Living systems and their gene regulatory networks fall into this category. Somebody brought my attention to negligible senescence (slow or no-aging in some species). I immediately recognized that such aging-resistance might also be an example of emerging property. 

Since then, we followed this lead and developed a set of ideas casting the “normal” Gompertzian aging and negligible senescence as two distinct phases of gene regulatory networks differentiated by dynamic stability properties. I am increasingly convinced that negligible senescence is the most important discovery in the field of aging studies. I also see that physics lends great tools for understanding this phenomenon and that achieving negligible senescence is a great scientific, humanitarian, and technological goal. 

Which of the current theories of ageing do you think are the most convincing?

Let me answer this question by bringing up the following analogy. In physics, there is the Standard Model. This is by far the best theory ever made. Until very recently (2018), there’s been no single experiment contradicting the theory. Even now, with only a few experiments in neutrino physics falling from the scheme, the discrepancy between the theory and the experiment is abysmal. Nevertheless, the Standard Model is a model, not a theory. This is because everyone knows it is inconsistent and thus can not be a complete theory. 

Compare this to the situation in aging sciences. We have a dozen of “theories” of aging. Such an impressive number of “theories” tells us that we do not have a theory of aging. It may look bad, but it is exciting too. Other fields of science lived through the times of such a  “zoo” of theories. The physics literature was full of unrelated forces and particles not long ago. We need the grand unified theory of aging, and it will come in due course. 

How has the field changed since you started?

The field matures. People do clinical trials. More people in science and the general public believe that lifespan is modifiable. We have more and increasingly better-trained people with diverse scientific and entrepreneurial backgrounds coming into the field (think, for example, of physicists, AI engineers, and crypto-activists joining biologists and medics). 

What mistakes do you think the longevity field has made?

There’s been tremendous progress both in understanding aging and translating the growing fundamental knowledge into preclinical and clinical trials. However, most currently developed solutions will produce small effects on par with the effects of lifestyles. For many, this strategy seems to reduce risks. But any working solution in biotech takes 10 years at least to develop fully. Working on weak interventions is a waste of resources and years of life. I would also think of hopes and expectations here. The public and policymakers have high hopes and may end up frustrated if the longevity biotech ends up with “statistically significant”  and, at the same time, miserable results. 

Other than your own, what do you think have been the biggest/important discoveries in the field?

To my taste, the discovery of negligible senescence - the discovery of advanced animals, such as mammals, that defy Gompertz mortality law is the biggest discovery in the field of aging. This is a very important demonstrator proving that aging is not evitable. Reducing mortality acceleration to almost zero may increase human lifespan a few fold. 

What advice would you give to people currently working in longevity research?

Think big - avoid solving unimportant problems even if those lend you a paper in a good journal.

Which aspect of longevity research do you think requires more attention?

Negligible senescence

Is ageing a disease?

This is a hard question that is probably more politically and emotionally charged than scientifically justified. From what we see in human and animal data, I believe aging can be slowed down and even stopped, not reversed. In this sense, this “disease” can not be fully cured but may be prevented. A huge entropic component in human aging makes aging more like a syndrome (multiple diseases leading to the same symptoms). To me, this does not matter. 

I am trying to avoid this conversation since it quickly gets murky and is dangerously close to scholastics. I believe that aging must be stopped, and once this is demonstrated, the technological solution will be covered by governments and insurance regardless of whether it addresses a disease. The potential upside is huge. It is so huge that new business models will emerge if the existing payers fail to recognize the opportunity.  If I am correct on this, we must forget about politics and focus on demonstrating an effective solution.

Can you explain what you’ve found in your research to be the major difference in the ways different species age (e.g. mice, naked mole-rat (NMR), humans)?

This is a great question. We published two Nature Comms works (a week ago, Unsupervised learning of aging principles from longitudinal data | Nature Communications,  and a year ago, Longitudinal analysis of blood markers reveals progressive loss of resilience and predicts human lifespan limit | Nature Communications) suggesting that humans and mice belong to two distinct classes of regulatory networks function. Mice are dynamically unstable; their organisms amplify damage, the damage does more damage, and this vicious loop leads to the exponential accumulation of damage. We found that the damage accumulation rate is the same as the mortality rate doubling time, and therefore the inability of the regulatory systems to control damage is the cause of exponential mortality acceleration. On the positive side, aging in mice appears to be mostly reversible.

On the other hand, humans control the damage very well for the most time up until late in life. The transition from the stable to the unstable phase occurs stochastically at some age around 60 due to the gradual loss of resilience, the ability of the organism to regain its homeostatic equilibrium after a shock. 

If this is true, aging in mice is the model of late-life morbidity and mortality in humans. Drugs acting in mice and extending their lifespan will improve lifespan beyond healthspan. Since resilience is lost late in life, the effects of such drugs on the human lifespan will be limited. 

To develop drugs that could let people live in good health for 150–200 years, we must confront the loss of resilience. This is especially challenging since the resilience is already lost in mice by the age of 25 weeks, if not earlier. This forms what we would call a “preclinical trap”: to convince investors and peer scientists, you have to select drugs that work best in mice. Such interventions, however, could only help people late in life. Unfortunately, the same drugs will provide small effects in healthy people and likely would not affect resilience. 

What are the fundamental principles and mechanisms behind negligible senescence in some animals (like NMR) and what causes them to die if there is no increased risk of mortality or frailty over time?

This is an open question. Our models suggest that NMRs and other negligibly senescent animals die of the same diseases. Just the incidence of those diseases (and hence death) does not grow exponentially as we age. Our last publications hint that humans are already almost negligibly senescent. In contrast to mice, humans control the hallmarks of aging for dozens of years until resilience is lost, and all hallmarks of aging and chronic diseases start appearing simultaneously. I believe that humans and NMR belong to the same class of aging systems. NMRs lose their resilience way slower than us, and I want to learn how they do it. Or, better, how to make humans stop losing their resilience.

Could human biology be modified in a way that it utilises those underlying principles of negligible senescence?

The short answer is yes. People are working to find out how. This is what Gero does, and answering this question is my personal scientific goal.

What do you consider “true aging”? And in order to extend healthy life, should we intervene before cellular and organismal loss of resilience? 

As I said, we observe that humans are very resilient. More is different, we are not talking at the level of cells. We are describing and measuring the resilience on the level of the organism as a whole. We work with very large datasets of human data and find that it’s very hard to die before resilience is lost. Curiously, according to our measurements, the number of people demonstrating the loss of resilience increases in the population exponentially and doubles every eight years, exactly as fast as the mortality rate doubles. Once the resilience is lost, like in mice, you have just a few years of life left.

Therefore, the loss of resilience is the most fundamental aging phenotype in humans. We call it “true aging” and it is different from the classic  hallmarks of aging driving late-life mortality and morbidity. Our worst nightmare came true: this year, we put up a preprint, Aging clocks, entropy, and the limits of age-reversal | bioRxiv, suggesting that the loss of resilience is driven by a thermodynamically irreversible process. If this is true, true aging in humans can not be fully reversed but only stopped. This is the least fortunate outcome of our research, and we had to be very careful trying for a few years to disprove this conclusion.  

Our approach is human-centric. We are applying modern AI/ML tools for identifying markers and genetic determinants of true aging in human data. In this way, we aim at the disentanglement of aging and diseases.  This helps in two ways. First, it helps people who look for cures for specific diseases. We work with pharma companies to identify novel targets, targeting reversible processes leading to transformative medicines. 

Most importantly, we track true aging in real-world medical and genetic data to find genetic factors modifying the rate of true aging. We are making good progress and hope to initiate experiments to test the theories in practice. 

Our goal is to make humans negligibly senescent species.

Outro

Thanks for reading our November issue of VitaDAO's Monthly Longevity Newsletter!

Once again, if there is anything you would like us to feature in future issues, please get in contact. 

This time we leave you with a thought provoking article in Science from 1972 by Nobel prize winner in physics Philip Anderson - More is Different because even though it’s 50 years old it’s still relevant today. There Prof. Anderson challenges the hierarchical structure of sciences - from physics to biology - and argues that “at each level of complexity entirely new properties appear”:

https://cse-robotics.engr.tamu.edu/dshell/cs689/papers/anderson72more_is_different.pdf

Further Reading

Healthy Aging: Strategies to Slow the Process

https://www.sciencedirect.com/science/article/abs/pii/S0030666522000809

Association of sleep duration at age 50, 60, and 70 years with risk of multimorbidity in the UK: 25-year follow-up of the Whitehall II cohort study

https://journals.plos.org/plosmedicine/article?id=10.1371/journal.pmed.1004109

Does Abdominal Obesity Increase All-Cause, Cardiovascular Disease, and Cancer Mortality Risks in Older Adults? A 10-Year Follow-Up Analysis

https://www.mdpi.com/2072-6643/14/20/4315/htm

A long-term obesogenic high-fat diet in mice partially dampens the anti-frailty benefits of late-life intermittent fasting

https://link.springer.com/article/10.1007/s11357-022-00678-1

Lipid hydroperoxides and oxylipins are mediators of denervation induced muscle atrophy

https://www.sciencedirect.com/science/article/pii/S2213231722002907?via%3Dihub

The time is now: Regular exercise maintains vascular health in aging women

https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP282896

One-year aerobic exercise increases cerebral blood flow in cognitively normal older adults

https://journals.sagepub.com/doi/abs/10.1177/0271678X221133861?journalCode=jcba&

Mitochondria dysfunction and impaired response to oxidative stress promotes proteostasis disruption in aged human cells

https://www.sciencedirect.com/science/article/pii/S1567724922000861?via%3Dihu

The use of progeroid DNA repair-deficient mice for assessing anti-aging compounds, illustrating the benefits of nicotinamide riboside

https://www.frontiersin.org/articles/10.3389/fragi.2022.1005322/full#.Y1SIjLKuaWo.twitter

Attenuation by Time-Restricted Feeding of High-Fat and High-Fructose Diet-Induced NASH in Mice Is Related to Per2 and Ferroptosis

https://www.hindawi.com/journals/omcl/2022/8063897/

A multitude of discoveries and inventions throughout history have been made by people who were initially trained in one domain, but were able to make a significant contribution to a different field due to their ability to look at a problem with a uni
Read more
VitaDAO Letter: New collab with Research Hub, Symposium Recap & more
November 14, 2022
Sarah Friday
Awareness
Longevity
VitaDAO Letter: New collab with Research Hub, Symposium Recap & more

Inside:

  • A Recap of VitaDAO’s 2nd “Crypto meets Longevity Symposium” 
  • VitaDAO Fellowship Update
  • New VitaSpotlight Series: Rapamycin as a Promising Aging Intervention 
  • How to contribute to VitaDAO and ResearchHub’s Community Driven Paper
  • Meet DAO Member Paolo Binetti

Upcoming Events

Community Call: November 24th at 9 AM EST-  Swing by VitaDAO’s monthly community call and learn more about what is going on in VitaDAO behind the scenes! 

The Longevity Summit December 6-7 | Buck Institute for Aging | Novato, CA -  THE FUTURE OF MEDICINE: ELIMINATING DISEASES OF AGING. Use the code "VitaDAO" for 15% off admission to the conference on the Eventbrite.

Hackathon: 13-15th Jan and 20-22- Join VitaDAO’s Hackathon, an online hackathon focusing on longevity. Registration is now open! 

DAO News

Symposium Recap: Exploring the Intersection of Longevity and Web3

On October 20th, 2022, Max Unfried and Eleanor Sheekey hosted the 2nd VitaDAO Crypto meets Longevity Symposium. The symposium consisted of twenty-two scientists, blockchain experts, and longevity professionals from internationally recognized institutions who spoke on the intersection of longevity and the blockchain.

Their talks highlighted updates in longevity medicine, strategies in drug development, discussions surrounding DAO economic ecosystems, and more!

Missed the conference? Catch up on VitaDAO’s Youtube

The VitaDAO Fellowship: Funding the next generation of Longevity Enthusiasts

VitaDAO received over 200 applications for a 3rd VitaDAO Fellow Cohort. These funds were made possible by generous donors on Gitcoin, in addition to the funding provided by VitaDAO (VDP-57). 

By providing grants up to 3,000 USD, the fellowship financially supports upcomers in the field to attend longevity conferences, fund research, and afford tuition to longevity programs.

Two cohorts, totaling 48 VitaDAO fellows, have been funded with over $45,000 USDC! Current VitaDAO Fellows have taken active roles in VitaDAO and are part of the exclusive VitaDAO Fellows network. 

Learn more: http://vitadao.com/fellowship 

New VitaSpotlight Series: Exploring Rapamycin - a Promising Aging Intervention?

Vita Fellow Adrian Moliere and Tomas Schmauck-Medina explore Rapamycin’s mechanism of action and its potential. Additionally, Professor Mikhail Blagosklonny weighs in on the idea that inhibiting pathways responsible for growth also slows aging: 

“Exploring Rapamycin: Most Promising Anti-aging Compound?

Find out more about how genetic knockouts or interventions showing the greatest increase in lifespan are targeting growth-, and not damage repair signaling pathways.

Exciting collab with Research Hub:
Contribute to a Community Driven Review Paper 

What are the most reliable longevity biomarkers, and how can they be evaluated? Alongside ResearchHub, VitaDAO is offering a bounty of 3,000 $VITA + 50,000 $RSC to be split among individuals who contribute to a community-driven review paper exploring current biomarkers for longevity.

Are you interested in exploring the pros and cons of different longevity biomarkers? Join the discussion on VitaDAO’s Discourse.

Governance: Funding Vita Fellows and Governance Updates

October’s Passed Proposals

VDP-57: $10k for the next VitaDAO Longevity Fellowship Batch 

As a result of this passed proposal, VitaDAO will donate $10,000 USDC to fund VitaDAO Longevity Fellows. VitaDAO Fellows can apply to receive micro-grants of up to $2,000 USDC.

VDP-59: VitaDAO Governance Amendment #3

As a result of this passed proposal, seven changes will be made to VitaDAO’s governance framework. These changes include a time limit of three months for phase 2 proposals, a mandatory seven-day waiting period for phase 2 proposals after the last significant change to the proposal, a formal on- and offboarding process for managers and authors on VitaDAO’s Snapshot space, changes to the on- and offboarding process of becoming a signer on VitaDAO’s multi-signature wallets, introduction of “Stakeholders” as a new user type, and amendments to the Code of Conduct.

Live on the Vita Governance Forum - discuss and vote!

Find all of VitaDAO’s pending live proposals on Discourse, VitaDAO’s governance hub for proposals before they are moved on-chain. These proposals are open for engagement, voting, and questions! This month, several new proposals were added to Discourse:

This proposal suggests governance improvement by “batching” VitaDAO’s governance into seasons, with each four-month season consisting of a Governance Phase and an Execution Phase. Within the season, the Governance Phase lasts 6 weeks and the rest is the Execution Phase where ideas proposed during the Governance Phase are executed.

This proposal requests $300k USDC to fund the discovery and development of a novel antiglycation and anticross-linking agent aimed at decreasing ECM stiffness.

  • VDP-62: Budget Allocation to Sponsor Longevity and Aging Conferences 

This proposal requests 20,000 USDC to sponsor eight months of Longevity and Aging Conferences and Events and requests that the SciComm and Awareness WG be given the power to decide which events to sponsor. 

Meet Key Longevity WG Contributor: Paolo Binetti

Paolo Binetti is one of our top VitaDAO Longevity/ Deal flow Working Group contributors. Outside of VitaDAO, he is a venture fellow for Healthspan Capital, a bio-expert for Capital Cell, an On Deck Longevity Biotech fellow, and an investor. Paolo holds a PhD in Controls, Robotics, and Bioengineering as well as a Bioinformatics specialization and has industry experience in R&D project management and business development.

How did you first get involved with the VitaDAO community?

Adding healthy years to our life will be beneficial for everyone. So everyone should be able to contribute to longevity biotech, either by funding it or working on it, but this is not happening, at least not as massively as it should. I want to solve this problem, VitaDAO could be one way to do it, so I decided to join.

What does your role as a Longevity/DealFlow Working Group Member entail?

Sourcing and reviewing research proposals and helping to mature them before they get to senior reviewers and community vote. I love it because every day I work with amazing researchers and founders on new cutting-edge ideas that could one day extend our healthspan and lifespan. I also help improve our infrastructure to make it even more effective.

What has surprised you the most about VitaDAO?

First, the amount of incredibly talented and competent people that I have come across, all committed to the same goal. Second, a rich infrastructure to collaborate in a decentralized way. Third, the flexibility to evolve toward ever more efficient ways of working. And the community’s welcome was phenomenal!

What are you most excited about in VitaDAO’s future?

Building a portfolio of solid projects with therapeutic potential, in collaboration with our new institutional partners in venture capital and pharma, and with an increasing contribution from the public. Everyone should have VITA tokens in their pockets, shaping the future of healthcare and reaping the benefits of the longevity biotech boom.

Any closing thoughts for readers?

I am always looking for new ideas for our deal flow, so if you have a great one that could generate valuable intellectual property and eventually translate to the clinic, please reach out!

Join Us! 

Have a special skill set? Contribute to VitaDAO and receive $VITA in return. Join the VitaDAO community on Discord and stay up to date on Twitter!  

Enjoyed this newsletter? Subscribe to stay up-to-date. 

Inside: A Recap of VitaDAO’s 2nd “Crypto meets Longevity Symposium” VitaDAO Fellowship Update New VitaSpotlight Series: Rapamycin as a Promising Aging Intervention How to contribute to VitaDAO and ResearchHub’s Community Driven Paper Meet DAO Membe
Read more
Rapamycin is the most promising aging intervention we currently have
October 25, 2022
Adrian Molière & Tomás Schmauck-Medina
Longevity
Science
Awareness
Rapamycin is the most promising aging intervention we currently have

It was in 1975 when scientists from Ayerst (now Pfizer) discovered a novel compound called rapamycin (also known as Sirolimus) in bacteria on Rapa Nui(Easter Island) in Chile. In 1999 rapamycin obtained FDA approval for the prevention of acute rejection of renal transplant. Unknown at the time, rapamycin would become the most potent anti-aging drug that humans currently hold.

This is the first article of a two-part series on rapamycin.

Rapamycin extends lifespan in all model organisms tested

The profound effect rapamycin has on lifespan was first observed in yeast cells, and later confirmed in every model organism tested, including the nematode C. elegans, fruit flies, and mice.

The results in mice were carried out by the Interventions Testing Program (ITP), the gold standard for lifespan experiments in mice.

The first rapamycin experiment of the ITP was particularly interesting. Originally, the plan was to test the compound from young adulthood onwards. However, due to bioavailability problems, the mice were treated with rapamycin when they were already 600 days old, roughly corresponding to a 60-year-old human. Many thought that there was no way a drug can have an effect on aging when treatment starts at such a high age, but to everyone’s surprise, rapamycin still extended lifespan by up to 14%.

The effect of rapamycin on extending lifespan in mice has subsequently been replicated numerous times, and to this day rapamycin is the most effective drug at extending the lifespan of healthy mice, with increases of up to 26% when started earlier in life.

And since the dose and regimen for mice are not yet optimized, the maximum possible lifespan increase is probably even greater.

Another promising feature of rapamycin is that it works in both sexes, while many other interventions have beneficial effects either solely or predominantly in males, with little effect in female mice.

So, what is rapamycin doing to confer this longevity-promoting effect?

Rapamycin inhibits mTORC1 in a highly specific manner

Rapamycin works by inhibiting the function of an enzyme called mTOR (mechanistic target of rapamycin). mTOR is a master regulator of cell growth and metabolism.

mTOR is so critical to these essential processes that it can be found in all eukaryotic organisms. Furthermore, there is relatively little difference between mTOR in yeast and in humans, explaining the effectiveness of rapamycin at inhibiting mTOR in all animals tested. mTOR is an enzyme belonging to the class of kinases. Kinases work by adding phosphate groups to their substrates, which can cause conformational changes to the protein structure to regulate their function.

Rapamycin does not inhibit mTOR directly, but rather by forming a complex with a protein from the class of immunophilins called FKBP12. The resulting complex in turn binds mTOR and inhibits it allosterically. Allosteric inhibition means that the inhibitor slows down the enzyme, mTOR, without blocking its active site. The advantage of this mechanism is that rapamycin is highly specific to mTOR, compared to inhibitors that block the kinase function at the active site of mTOR directly, which tend to inhibit other kinases as well.

mTOR is part of two functionally distinct complexes, mTORC1, and mTORC2. Rapamycin only inhibits the mTORC1 complex directly, however, after chronic treatment with rapamycin mTORC2 levels decline as well. mTORC2 and mTORC1 functions are different. While the role of mTORC1 is well understood, many aspects of mTORC2 signaling remain elusive. We will focus on mTORC1 in this article, as it is the main target of rapamycin. We will cover the differences between mTORC1 and mTORC2 inhibition and its consequences in more detail in part two of this series.

Figure based on http://dx.doi.org/10.1016/j.cmet.2014.01.001

mTOR is a master regulator of nutrient sensing and growth signaling

The mTOR complexes are the main signaling nodes integrating environmental signals into cellular responses. If the conditions are favorable for anabolic processes, the mTOR pathway will initiate growth and proliferation.

To facilitate this, mTORC1 responds to nutrient availability, energy, oxygen status, and growth signals. If, for example, there is an abundance of amino acids, then mTORC1 will get activated and anabolic processes will get initiated.

As a result, active mTORC1 increases protein, lipid, and nucleotide synthesis, promotes mitochondrial biogenesis and energy production, and inhibits autophagy, among other processes. Basically, active mTORC1 is telling the cell to go full throttle. By inhibiting mTORC1, rapamycin tricks the body into reacting as though nutritional resources are scarce, thus activating protective pathways and mechanisms, like autophagy.

For that reason, rapamycin was initially seen as a mimetic of caloric restriction, one of the most potent lifespan-extending interventions. However, even though mTOR inhibition is crucial for the lifespan extension of caloric restriction, rapamycin and caloric restriction are still distinct in their effect.

Rapamycin positively influences all hallmarks of aging

One of the most influential papers in the field of aging is a 2013 review titled “The Hallmarks of Aging”. Here 9 hallmarks are attributed to aging, such as telomere attrition, epigenetic alterations, genomic instability, and others. Rapamycin influences all of them.

Fig from https://doi.org/10.1016/j.tma.2017.09.004

The hallmarks of aging do not necessarily reflect a causal explanation for aging. Still, they touch on most aspects of molecular changes seen with age and if a compound manages to influence all of them, then there is a good chance that itwill have an effect on healthspan/lifespan.

mTOR regulates mitochondrial processes, such as ATP production and translation of mitochondrial proteins. And indeed, rapamycin treatment has been shown to increase mitochondrial efficiency.

That rapamycin improves the hallmark of nutrient sensing is unsurprising, given that mTOR is the central nutrient sensor in the cell. Similarly, improved proteostasis is a logical conclusion due to the upregulation of autophagy by rapamycin.

Regarding epigenetic alterations, rapamycin has been shown in vitro to rejuvenate epigenetic markers of age. In animals, this connection is less clear. While rapamycin treatment reduces epigenetic age in mice, this was not observed in marmosets, a type of primate.

Rapamycin has also been characterized as a senomorphic, meaning it blocks the conversion of cells into senescent cells and reduces inflammatory phenotypes in senescent cells.

Beyond that, rapamycin rejuvenates hematopoietic stem cells, reduces DNA damage, and counteracts telomere attrition. It furthermore remodels the microbiome, which is proposed to be a novel hallmark of aging.

The improvements of many facets of aging upon rapamycin treatment thus support the idea that it targets aging itself. But why is mTOR inhibition seemingly more effective than many other interventions that also target age-related cellular changes?

Development might hold the answer to the anti-aging effects of rapamycin

An interesting observation is that the most potent anti-aging interventions are targeting pathways that are highly relevant to growth and development, like mTOR, Insulin-IGF, and AKT signaling. There seems to be a general trend that inhibiting pathways responsible for growth also slows aging. This suggests that the same pathways that promote growth actually cause aging in some way. We are joined by Professor Blagosklonny, one of the main proponents of this idea.

Interview with Prof. Mikhail Blagosklonny

AM: Why is it important to develop theories that try to explain the cause of aging?

MB: Well, it’s important because a correct theory can allude to the correct treatment of aging. Many theories of aging have been proposed over the years and a lot of them state that aging is solely caused by an accumulation of molecular damage, for example by free radicals. Therefore, antioxidants are widely used in the general population to slow down aging despite animal studies contradicting that free radicals cause aging and clinical trials showing no benefit. In 2006 I predicted based on the hyperfunction theory that rapamycin will extend the lifespan of animals, which was later confirmed.

So this is why it’s important: the theory can be translated into clinical practice.

AM: So does the accumulation of molecular damage play no role in aging?

MB: Molecular damage accumulates with age and after a long time it would kill the organism, but the organism generally dies from normal (mTOR-driven) aging before that happens. Molecular damage is not life-limiting. Accumulation of molecular damage could be artificially accelerated in animals. For example, by damage in enzymes that repair molecular damage. But in these cases, symptoms are different from symptoms of normal aging.

AM: If an accumulation of molecular damage is not the cause of aging, then what is, and how is it connected to mTOR (Target of Rapamycin)?

MB: The basis of the hyperfunction theory is that aging is a continuation of growth and development. Genes are highly regulated during development but fall into an evolutionary selection shadow post development and reproduction. Thus many genes show antagonistic pleiotropy.

Antagonistic pleiotropy describes the idea, that the same gene can have multiple effects (pleiotropic) that can be both beneficial and detrimental to the organism. Genes are selected for their beneficial traits at the beginning of life, even though they might be detrimental later in life. Evolutionarily, this detrimental effect later in life is not selected against as it only occurs after reproduction, thus lying in a selection shadow.

This means that the program responsible for growth and development, mainly the mTOR pathway, doesn’t get properly switched off after development, the pathway becomes hyperfunctional. This unwanted continuation becomes harmful and leads to the development of age-related diseases and aging. This explains why the genetic knockouts or interventions showing the greatest increase in lifespan are targeting growth-, and not damage repair signaling pathways. That hyperfunctional growth pathways drive age-related diseases is not just speculation. Involvement of mTOR was described in all human age-related diseases.

But while the hyperfunction theory is in complete agreement with evolutionary concepts like antagonistic pleiotropy, it is not an evolutionary but a mechanistic theory, since it can also give a mechanistic explanation of aging on the cellular level.

When cells cannot divide anymore they can enter a state of cellular senescence, which is characterized by hypertrophy (the cells become enlarged) and a hypersecretory phenotype. When I was working on cell growth and cellular senescence we found that cellular senescence is a continuation of cellular growth, when the cell cycle is arrested. Hyperfunctional growth signaling leads to a geroconversion from reversible cell cycle arrest into a senescent state. One of the main growth-signaling pathways is mTOR, and we showed that rapamycin can stop cells from becoming senescent. This cellular geroconversion is one of the mechanistic explanations for aging on an organismal level as well.

AM: How is mTOR regulated at different life stages and why does it become hyperfunctional?

MB: This is related, as mentioned, to antagonistic pleiotropy. Evolutionarily, it only matters what is beneficial early in life, for reproduction, so natural selection doesn’t regulate mTOR activity in later life. So, natural selection keeps mTOR activity optimal for fitness early in life, but too high for longevity late in life.

When the mTOR pathway is inhibited early in life, during development, it results in slow growth and decreased reproduction. These observations are made in laboratory animals, such animals will die in the wild of course, due to decreased fitness. But in laboratory conditions, they survive and live longer. Just recently it was published that inhibition of mTOR with rapamycin in young animals also extends lifespan.

But of course, this is not translatable to humans, we cannot treat infants or children with rapamycin. To slow their growth so they live longer, that would be completely absurd.

But fortunately, we don’t need to do this. You don’t need to inhibit development. But you can turn mTOR down pharmacologically in adults after development, when it becomes hyperfunctional, which I think holds great promise.

Disclaimer & Disclosure

DISCLAIMER: THIS ARTICLE DOES NOT PROVIDE MEDICAL ADVICE. No material in this article is intended to be a substitute for professional medical advice, diagnosis, or treatment. The text, images, and other material contained in this article are for informational purposes only.

VitaDAO has funded a clinical trial by Dr. Brad Stanfield on exercise and Rapamycin dosing. Pfizer has recently joined VitaDAO as a strategic contributor.

It was in 1975 when scientists from Ayerst (now Pfizer) discovered a novel compound called rapamycin (also known as Sirolimus) in bacteria on Rapa Nui(Easter Island) in Chile. In 1999 rapamycin obtained FDA approval for the prevention of acute reject
Read more
October Longevity Research Newsletter
October 13, 2022
Maria Marinova & Rhys Anderson
Awareness
Longevity
Newsletters
October Longevity Research Newsletter

Introduction

Welcome back to VitaDAO’s monthly longevity research newsletter!

Some species in the animal kingdom live extraordinarily long lives. They seem to be somehow protected from all the damage that we would expect to accumulate in aged tissues. Many are very large, like bowhead whales, and there is an established correlation between mass and lifespan, often attributed to slower metabolism. There are also animals like the naked mole rat, which live many times the life of a rodent of the same size with no increased risk of disease or decline of physiological function. We have a lot to learn from such species and Prof. Vera Gorbunova is doing just that - her lab focuses on understanding the underlying mechanisms of aging and how they differ between long-lived mammals and humans. We interviewed her for this month’s issue, so make sure you don’t miss this! (and if you just can’t wait, it can be found towards the bottom of this page!)

Longevity Literature Hot Picks

As so many great longevity papers are being published every month, we’ve decided to keep our newly introduced Further Reading section at the end of the newsletter! But first, here are our hotpicks of the month:

Sex- and age-dependent genetics of longevity in a heterogeneous mouse population

https://www.science.org/doi/10.1126/science.abo3191

This substantial body of work, done on the largest-ever mouse longevity study unravels the distinct genetic determinants of male vs female longevity. The group has compiled their data with other sources to produce an interactive research tool, which is quite fun to play around with: https://lisp-lms.shinyapps.io/itp-longevity-app/

Brown-fat-mediated tumour suppression by cold-altered global metabolism

https://www.nature.com/articles/s41586-022-05030-3

Cancer cells are known to utilise glycolysis to provide cellular energy - a process reliant upon glucose uptake. Non-shivering thermogenesis of adipose tissues also requires glucose uptake. Interestingly, Seki et al., show that cold exposure can lead to a non-shivering thermogenesis-induced decrease in blood glucose levels which can attenuate tumour growth!

Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions

https://www.cell.com/cell/fulltext/S0092-8674(22)01127-8

A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors

https://www.cell.com/cell/fulltext/S0092-8674(22)01173-4

For years research has implicated a role for bacteria in cancer - now two papers published in Cell provide extensive evidence that fungi are also found in numerous cancers and can even be associated with different prognoses. Certain fungi and bacteria were even found to often co-exist in some cancer types. This adds an additional level of complexity to our understanding of cancer, and could provide new markers for diagnostic and prognostic testing.

Insulin signaling in the long-lived reproductive caste of ants

https://www.science.org/doi/10.1126/science.abm8767

There is a trade-off between reproduction and lifespan in most organisms, but the queen ant is an exception. When a caste ant switches to a reproductive status of a pseudo-queen, her lifespan is extended 5x. During this switch, genes expression patters of insulin, MAPK, AKT is altered, consistent with the observed longer life.

SIRT3 deficiency decreases oxidative metabolism capacity but increases lifespan in male mice under caloric restriction

https://onlinelibrary.wiley.com/doi/10.1111/acel.13721

Contrary to previous hypotheses that the mitochondrial SIRT3 is responsible for caloric restriction (CR) mediated lifespan extension, this study shows that deletion of SIRT3 actually extends the life of mice under CR conditions. This points to SIRT3 being dispensable for CR induced lifespan extension but it is required for aerobic capacity and oxidative metabolism.

Ketogenic diet prevents chronic sleep deprivation-induced Alzheimer’s disease by inhibiting iron dyshomeostasis and promoting repair via Sirt1/Nrf2 pathway

https://www.frontiersin.org/articles/10.3389/fnagi.2022.998292/full

An interesting interplay between sleep diet and Alzheimer’s has been described. Ketogenic diet seemed to be preventative of cognitive deficiency, amyloid deposition and hyperphosphorylated tau induced by chronic sleep deprivation by inhibiting ferroptosis and alleviating oxidative stress. 

Is “cellular senescence” a misnomer?

https://link.springer.com/article/10.1007/s11357-022-00652-x

David Gems and Carina Kern argue that considering cellular senescence has been shown to play roles in healthy physiological processes such as acute wound healing and embryogenesis, perhaps the word ‘senescent’ is a misnomer as the dictionary definition implies that something is old. Instead they propose the term should be replaced with one more descriptive of the phenotype - “remodelling activation” - which would consequently replace SASP with RASP. Whilst it’s unlikely that the word will change any time soon (historically used terms have sticking power) it’s an important idea to keep in mind when thinking about what exactly a senescent cell is.

Rapamycin treatment during development extends life span and health span of male mice and Daphnia magna

https://www.science.org/doi/10.1126/sciadv.abo5482

Another study convincingly shows how tightly connected development and aging are. Only 45 days of rapamycin treatment during early development (right after birth) were sufficient to increase lifespan by 10% in male mice. The cost of this was smaller size and slowed down development. 

Drugs, clocks and exercise in ageing: hype and hope, fact and fiction

https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP282887

This review takes a broad look at the field and compares the effects of pharmacological vs non-pharmacological interventions and the accuracy and predictive power of functional measurements vs aging clocks. Roadblocks in the advancement of the field and potential future strategies are also discussed.

Aging and cancer epigenetics: Where do the paths fork?

https://onlinelibrary.wiley.com/doi/full/10.1111/acel.13709?campaign=wolearlyview

There is a clear association between aging and cancer on molecular and eidemiological level but can this also be explained via epigenetic mechanisms? While there are considerable differences between the two processes, epigenetic clocks could be key to unravel the intricacies of this relationship, especially if integrated with genetic evidence of DNA damage.

News

Celebrity Strategy Consultant Predicts What Will Be The Most Impactful Area In The Pharmaceutical Industry

https://www.forbes.com/sites/alexzhavoronkov/2022/09/19/celebrity-strategy-consultant-predicts-what-will-be-the-most-impactful-area-in-the-pharmaceutical-industry/?sh=7c19492316b4

Read Dr. Alex Zhavoronkov’s article on Dr. Michael Ringel’s ARDD lecture titled “The Emerging Commercial Landscape for Aging Biology-Based Therapeutics”.

The US House of Representatives held a hearing on geroscience and how it drives age-related disease and disability

https://science.house.gov/hearings/the-fountain-of-youth-the-quest-for-aging-therapies

Speakers include: Dr. Jay Olshansky, Dr. Laura Niedernhofer and Dr. Steve Horvath.

The Healthy Longevity Medicine Society Official Launch

https://hlms.co/

Led by President Prof. Andrea Maier and Vice President Dr. Evelyne Bischof,  the HLMS vision is “to develop longevity medicine as a respected and independent medical speciality that extends the healthspan of ageing individuals, tackles ageing mechanisms and optimizes an individual’s performance.”

Methuselah Foundation Launch the ELONgevity Protection Project 

https://www.mfoundation.org/news/2022/9/29/life-insurance-only-pays-out-after-you-die-we-want-you-to-live

To promote human anti-aging efforts and provide members diagnosed with terminal diseases access to experimental therapies.

1st NIH SenNet Consortium meeting in Rockville

https://sennetconsortium.org/

“The purpose for the Cellular Senescence Network (SenNet) is to catalyze the development of a framework for mapping cellular senescence and its associated secretory phenotype at high resolution, to provide atlases of cellular senescence in multiple tissues and under diverse conditions, including early development, and across the lifespan.”

Launch of Biotech DAO Accelerator Bio.xyz 

https://www.bio.xyz/

Biotech DAO accelerator and meta-governance layer for the DeSci ecosystem! Expect resources, courses, frameworks and $100k USDC grants to enable a new generation of talented builders in DeSci to build and launch.

Hevolution Foundation Matches Funding for Impetus Grants to Accelerate Research in Neglected Areas of Healthspan Science

https://hevolution.com/en/w/hevolution-foundation-matches-funding-for-impetus-grants-to-accelerate-research-in-neglected-areas-of-healthspan-science

Upcoming conferences

2nd VitaDAO Crypto meets Longevity Symposium

October 20th, Online

Check out our 1st symposium here: https://www.youtube.com/watch?v=GJ-rJAfjhBE

VitaDAO Hackathon 2022

Ideation weekend: 28th-30th October

Hackathon weekend: 4th-6th November

Register here: https://vitadao.com/hackathon

The Longevity Summit 2022

December 7th-8th, Buck Institute for Aging, US

https://longevitysummit.io/

Funding Opportunities

The Hevolution/AFAR New Investigator Awards in Aging Biology and Geroscience Research

https://www.afar.org/imported/AFAR-Press-Release_HevolutionAFARNewInvestigatorsAwardandConferencesFund_08.31.22.pdf

Providing up to 18, three-year awards of US $375,000 each and will support research projects in basic biology of aging or geroscience.

Job board

Joao Pedro de Magalhaes’ lab is looking for a researcher/developer to work on online resources for research on ageing. https://www.linkedin.com/jobs/view/3311250129/?refId=TBaRILN%2FQN6S%2FH88SMOZaA%3D%3D

Postdoc position is available at Antebi lab (molecular genetics) at the Max Planck Institute for Biology of Aging. Apply through the portal:

https://www.age.mpg.de/career-education/open-positions

Cambrian Biopharma is looking for a product manager for their biomarker development program - Project Ordo. The position could be project based, part-time or full-time.

Please contact prashant@cambrianbio.com if interested.

Gordian Bio is hiring PhDs, postdocs and technicians for two positions in San Francisco:

Molecular biologist: https://jobs.lever.co/gordian-bio/174715cc-62c5-4408-9053-51331b3f39b1

Single cell scientist: https://jobs.lever.co/gordian-bio/814a6528-4f4c-446b-9ea1-3711f1210b34

Vincere Bio is looking for 1-2 mitochondrial biologists to test mitophagy enhancers on myocardial senescence or Parkinson’s and visualise mitophagy in vivo using mouse models. Boston, MA

Contact: recruiting@vincerebio.com

DataBETA (Database for Epigenetic Evaluation for Treatment of Aging) is a non-profit, looking for a project director to hire as a contractor. More info at dataBETA.io
Reach out to Josh Mitteldorf, including why is this job best next step for your career and how will you launch the project with simultaneous fundraising: aging.advice@gmail.com

Articles/Blogs

Inside Saudi Arabia’s $20 Billion Bet On Longevity Biotechnology

https://www.forbes.com/sites/alexzhavoronkov/2022/09/28/inside-saudi-arabias-20-billion-bet-on-longevity-biotechnology/?sh=357610364aec

Can You Increase Longevity? The Science Behind Living Longer

https://blog.insidetracker.com/increase-longevity-science-behind-living-longer?utm_campaign=september%202022&utm_content=220638130&utm_medium=social&utm_source=twitter&hss_channel=tw-425907208

Hide and seek - Cancer cells hide inside each other when the immune system attacks

https://elifesciences.org/digests/80315/hide-and-seek

Original research: Transient cell-in-cell formation underlies tumor relapse and resistance to immunotherapy: https://elifesciences.org/articles/80315

DNA clocks suggest ageing is pre-programmed in our cells

https://www.newscientist.com/article/2338033-dna-clocks-suggest-ageing-is-pre-programmed-in-our-cells/

Book Review

A science-based review of the world's best-selling book on aging

https://www.sciencedirect.com/science/article/pii/S0167494322002126?via%3Dihub

Podcasts

Investing in Longbio True Believers - Sebastian Brunemeier, Healthspan Capital & ImmuneAge Pharma

https://player.captivate.fm/episode/2fb68713-bea4-4706-ac3d-b19b8b53a811

Check out Sebastian Brunemeier discussing how the science of human longevity is transforming the way we treat disease.

Should Aging Be Called A Disease?

https://www.youtube.com/watch?v=9kd5mQCfIfg

Listen to longevity heavyweights Prof. Nir Barzilai and Prof. David Sinclair discussing whether aging should be classified as a disease and the associated pros and cons. 

The Sheekey Science show with Prof. Charles Brenner

https://www.youtube.com/watch?v=opMijdJ6z9Q

Catch up on this interesting discussion on NAD+, aging and fertility.

Michael Levin on the Lex Fridman Podcast

https://www.youtube.com/watch?v=p3lsYlod5OU

Biology, Life, Aliens, Evolution, Embryogenesis & Xenobots.

TV Series

Limitless with Chris Hemsworth | Disney+

https://www.disneyplus.com/en-gb/movies/limitless-with-chris-hemsworth/4MIMDuD9nF2s

Trailer: https://www.youtube.com/watch?v=SJPnK_NgHVI

“Chris Hemsworth explores the different ways humans can live better for longer by taking on physical challenges such as diving in ice or climbing skyscrapers.”

Just for fun

Wordle for Genes!

https://andrewholding.github.io/gene-wordle/

Get Involved

The Norn Group: http://norn.group (responsible for the Impetus Longevity Grants) will soon be launching a program to empower people across the globe to impact the Longevity field.

https://docs.google.com/forms/d/e/1FAIpQLSdrigugaS0W5sgtbQ9l15XeOsr7OkDaNHBIKt8614ksPtxPBQ/viewform

If you train and/or employ people to work on aging, the Norn group would love to hear your ideas for what a CompBio aging course should cover.

Interview with Professor Vera Gorbunova

Prof. Vera Gorbunova is a Professor of Biology at the University of Rochester and a director of the Rochester Aging Research Center. Her research is centred around aging, cancer and genome stability. Her lab aims to understand the basic mechanisms underlying these processes by studying multiple model organisms, especially long-lived mammals. 

What inspired you to enter longevity research?

This is the most fascinating problem in biology and medicine… How could I work on anything else?
Longevity research addresses a fundamental problem of biology and has relevance to every living human.

Which of the current theories of ageing do you think are the most convincing?

I find the damage accumulation theory the most convincing. However, the nature of this “damage” is a subject of debate. Most likely, this is a combination of DNA damage, mutations and epigenetic dysregulation.

How has the field changed since you started?

I witnessed tremendous progress. We now have the tools we did not have before. Genomes of various species, various other omics tools became widely available and used in everyday research. Conserved pathways of aging had been identified in model organisms.

What mistakes do you think the longevity field has made?

Too much focus on short-lived model organisms. These organisms are helpful to identify conserved aging pathways, but they are not suitable to find longevity adaptations that evolved in long-lived species.

What advice would you give to people currently working in longevity research?

Do more bold and out of the box things. We need novel approaches and new research models. When studying long-lived organisms, do not just compare young to old. Obviously, the long-lived organisms age slower. What is most important is to find the mechanisms that make them age slower.

Is ageing a disease?

From the evolutionary standpoint aging is not a disease. However, we have to approach it as a disease to develop treatments and interventions.

You have done research across the animal kingdom. How do long-lived animals benefit longevity research? What are the most promising organisms to study that are currently underexplored?

Long-lived animals are essential for longevity research as they hold the clues for long lifespan. I am particularly interested in long-lived mammals because they are phylogenetically closer to people and the discoveries would be more translatable. There are many long-lived mammals and each may have evolved unique longevity mechanisms. That is why it is important to study all of them. My group works on naked mole rats, bats, bowhead whales and many other mammalian species. 

What are the most interesting insights from naked mole rat studies and what are potential ways to translate them?

There were many interesting discoveries made on naked mole rats. The most interesting ones come when researchers look at novel mechanisms not merely testing the same pathways that were shown to be important in short-lived species. Here I can name our discovery of abundant hyaluronic acid and unique ribosomal structure with split 28S rRNA that results in highly accurate protein synthesis.

Could you explain hyaluronic acid anti-tumour and pro-longevity effects you have observed, and do you see this as a potential intervention in humans?

Naked mole rat tissues are saturated with hyaluronic acid of very high molecular weight (HMW-HA). Such hyaluronic acid has antiproliferative properties and arrests proliferation of premalignant cells leading to lower cancer incidence. HMW-HA is also a potent anti-inflammatory molecule reducing inflammation in the body and through this having an anti-aging effect.

Outro

Thanks for your interest in VitaDAO's Monthly Longevity Newsletter!

We hope to keep you up to date in this rapidly expanding field. Once again, if there is anything you would like us to feature in future issues, please get in contact. 

If you have been wondering what has been going on in the VitaDAO community, here are some links below to some of our highlights from the past month. We look forward to seeing you again next month!

The Longevity Decentralized Review (TLDR) by VitaDAO | With Tim Peterson

https://www.youtube.com/watch?v=MDH7ZmMgp4I

VitaDAO IP-NFT Transfer Ceremony with Molecule & ApoptoSENS

https://www.youtube.com/watch?v=nUOhVGtb3zw

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with Longevity Research.

Further reading

Is early-onset cancer an emerging global epidemic? Current evidence and future implications

https://www.nature.com/articles/s41571-022-00672-8

The emerging neuroprotective roles of exerkines in Alzheimer’s disease

https://www.frontiersin.org/articles/10.3389/fnagi.2022.965190/full

Monocyte subsets display age-dependent alterations at fasting and undergo non-age-dependent changes following consumption of a meal

https://immunityageing.biomedcentral.com/articles/10.1186/s12979-022-00297-6

Reduced endosomal microautophagy activity in aging associates with enhanced exocyst-mediated protein secretion

https://onlinelibrary.wiley.com/doi/10.1111/acel.13713?utm_campaign=R3MR425&utm_content=LifeSciences&utm_medium=paidsearch&utm_source=google

The lysosomal proteome of senescent cells contributes to the senescence secretome

https://onlinelibrary.wiley.com/doi/10.1111/acel.13707

Psychological factors substantially contribute to biological aging: 

evidence from the aging rate in Chinese older adults

https://www.aging-us.com/article/204264/text

An intercellular transfer of telomeres rescues T cells from senescence and promotes long-term immunological memory

https://www.nature.com/articles/s41556-022-00991-z

Metabolic changes in aging humans: current evidence and therapeutic strategies

https://www.jci.org/articles/view/158451

Transcriptional Heterogeneity of Cellular Senescence in Cancer

https://www.jci.org/articles/view/158451

Relationship Between 5 Epigenetic Clocks, Telomere Length, and Functional Capacity Assessed in Older Adults: Cross-Sectional and Longitudinal Analyses 

https://academic.oup.com/biomedgerontology/article/77/9/1724/6473288?login=false

Dermal Tattoo Biosensors for Colorimetric Metabolite Detection

https://onlinelibrary.wiley.com/doi/10.1002/anie.201904416

Sirtuins are Not Conserved Longevity Genes

https://academic.oup.com/lifemeta/advance-article/doi/10.1093/lifemeta/loac025/6711379?login=false

Umbilical cord plasma concentrate has beneficial effects on DNA methylation GrimAge and human clinical biomarkers

https://onlinelibrary.wiley.com/doi/10.1111/acel.13696

Sexual activity and successful aging

https://link.springer.com/article/10.1007/s41999-022-00694-6

Some species in the animal kingdom live extraordinarily long lives. They seem to be somehow protected from all the damage that we would expect to accumulate in aged tissues. Many are very large, like bowhead whales, and there is an established correl
Read more
VitaDAO Letter: VitaDAO's first cell therapy funding: ApoptoSENS!
October 11, 2022
Sarah Friday
Awareness
VitaDAO Letter: VitaDAO's first cell therapy funding: ApoptoSENS!

Happy DeSci Fall! Inside this month’s newsletter:

  • VitaDAO's first cell therapy funding: ApoptoSENS
  • A New Publication on Decentralized Technology Transfer
  • The Longevity Decentralized Review Explainer
  • Meet DAO Member: Estèfano Pinilla 

DAO News

1/4 Million Research Funding for ApoptoSENS

On September 13th, VitaDAO celebrated the funding of its fifth longevity research project funded via IP-NFT! VitaDAO is supporting ApoptoSENS’s research with $253,000 as they develop CAR-NK cells to target senescent cells in vivo. ApoptoSENS will explore ways to make the elimination of senescent cells more precise and safe. You can watch a recording of the live-streamed IP-NFT Transfer Ceremony with Molecule and ApoptoSENS on VitaDAO’s Youtube. 

Publication: A New Scientific Incentive Structure

In “Decentralized Technology Transfer: A Modern Framework to Empower Scientific Innovation,” Zack West writes about a modern incentive structure for individual researchers, labs, and publishers that diverts from the traditional Technology Transfer Office (TTO) process. TTOs actively block the progression of IP getting to market. Ideally, the introduction of decentralized tech transfer can enable researchers to operate outside of their institutions and leverage other institutions’ resources.

New VitaDAO Initiative: The Longevity Decentralized Review (TLDR) 

The Longevity Decentralized Review (TLDR) is an on-demand peer review service, founded by VitaDAO’s Tim Peterson. TLDR enables researchers to have proof of their peer reviews such that they don’t need to rely on journals for this proof. Want to learn more about TLDR? Watch Tim’s new video explainer on VitaDAO’s Youtube and check out the TLDR proof of concept website

Governance: Parter & DAO2DAO Development

Updates: VitaDAO's Institutional Genesis Raise 

Last month, Pfizer Ventures, L1 Digital, and Shine Capital applied to participate in VitaDAO’s Institutional Genesis Raise to contribute capital and participate in the governance of VitaDAO using $VITA tokens. This month, SpaceshipDAO, BeakerDAO, and Healthspan Capital applied to VitaDAO's Institutional Genesis Raise.

VDP 54.4: SpaceshipDAO Expression of Interest

SpaceshipDAO, a member-owned investment DAO supported by Tribute Labs, proposed to contribute $50,000 to participate in the governance of VitaDAO. They proposed to support VitaDAO with access to deal flow, domain expertise, awareness, and commercialization efforts of IP. 

VDP 54.5: BeakerDAO Expression of Interest

BeakerDAO proposed to contribute $75,000 to participate in the governance of VitaDAO. They proposed to support VitaDAO with community support and expertise in the DeSci space.

VDP 54.6: Healthspan Capital Expression of Interest

Healthspan Capital proposed to contribute $100,000 to participate in the governance of VitaDAO using $VITA tokens. Healthspan Capital proposed to support VitaDAO beyond capital contribution with deal flow sharing, diligence notes comparing, support for newco formation and incubation, IP diligence, and industry partnering.

Live On Snapshot: Funding VitaDAO Fellows

VDP-57 proposes that VitaDAO donate $10,000 USDC to fund VitaDAO Longevity Fellows with micro-grants of up to $2k. VitaDAO’s fellowship program supports individuals who desire to dive deeper into learning about longevity. Previously, VitaDAO’s Fellowship was made possible by donors on Gitcoin. Due to the positive feedback from the broader community, as well as hearing from unfunded applicants, VDP-57 requests financial support for a new batch of fellows.

Key Working Group Contributor: Estèfano Pinilla

Estèfano Pinilla is a long-term VitaDAO working group contributor, who first joined VitaDAO in May 2021. He is a postdoctoral researcher, studying Cardiovascular Pharmacology, at Aarhus University in Denmark. His interests include longevity, extracellular matrix, and fibrosis. 

How did you first get involved with the VitaDAO community/ in DeSci?

I first stumbled upon VitaDAO on Twitter, a month before the initial token auction. I wasn’t very interested in the blockchain back then, I only saw the financial applications and it seemed too self-referential to catch my interest. However, I am familiar with trying to bring research from the lab to the patients and with the limitations of the current patent system. As a researcher, I immediately could see the potential of the IP-NFT concept, and the idea of a community of scientists, professionals, and longevity enthusiasts getting together with the mission of extending healthy life sounded extremely exciting. I was impressed with how honestly the project was communicated as an experiment, and I decided to jump right in and started contributing. I’m always in for a cool experiment! 

Can you walk me through what your position in VitaDAO entails?

As a Longevity Working Group contributor, I have shepherded several projects as well as assisted other shepherds as a squad member. Being a shepherd consists in serving as a bridge between the community and the applicants, coordinating the evaluation of the projects, putting all the information together, and writing the proposal for the community to vote on. In the Comms and Awareness Working Group, my main activity has been organizing the second season of the Longevity Journal Club, where we discussed the latest research papers in Longevity in an accessible way for the non-technical audience.

What has surprised you the most about VitaDAO?

What has surprised me the most is the diversity of backgrounds and expertise of the members of our community. It’s great to be able to lurk around the Discord group and see what is being discussed in the chats from the different working groups, I have learned a lot and accessed information that I wouldn’t have otherwise just by doing that. Also, it’s amazing to see ideas from community members growing into full-fledged projects in real-time. The worst part is not having the bandwidth to participate in all of them!

What are you most excited about in VitaDAO’s future?

There is so much potential and so many exciting projects on the horizon that it’s difficult to pick a single thing. One of the things I’m most excited about is the possibility of seeing biotech companies coming out of the projects we fund, companies trying to bring medicines to market in which the researchers have more upside and a diverse community of stakeholders have much more control over the assets

Any closing thoughts for readers? 

I would like to encourage other researchers to join the DAO and apply for funding, we are always looking for cool new ideas with the potential to extend healthy life and reach the patients. The collective knowledge of the community is impressive and we have a lot of passion to bring projects forward!

VitaDAO in the Media: Vitalik discusses VitaDAO

Should VitaDAO be a DAO? In a recent essay titled “DAOs are not corporations: Where decentralization in autonomous organizations mattersVitalik Buterin, co-founder of Ethereum, explores DAOs as a first-order organization and discusses VitaDAO. 

Theo Beutel spoke on behalf of VitaDAO as a part of Tally's video interview series. He covered the power of crypto funding, VitaDAO’s governance, and VitaDAO’s IP NFT mechanism. 

At VitaDAOxPolygonDAO AMA, VitaDAO members Laurence Ion, Catthu, Jesse, and Time Peterson discussed VitaDAO’s achievements, the challenges of scientific funding, and VitaDAO’s future plans. 

Upcoming Events: A Second VitaDAO symposium & more 

October 10-13th at 11 AM EST- Our members, Tim Peterson, Max Unfried & Laurence Ion,  will be speaking at Targeting Metabesity 2022 Virtual Conference

October 17th at 11 AM EST- Swing by VitaDAO’s monthly community call, hosted on TwitterSpace. Learn more about what is going on in VitaDAO behind the scenes! 

October 19th at 10 AM EST- (Recurring weekly on Wednesdays)- Join a VitaDAO onboarding session! Here, you can ask questions about VitaDAO, meet others interested in the DAO, and learn about VitaDAO’s working groups. 

October 20th at 9 AM EST- Join hosts Max Unfried and Eleanor Sheekey at VitaDAO’s second VitaDAO Crypto meets Longevity Symposium. The symposium will serve as a chance for scientists, blockchain experts, and longevity professionals from internationally recognized institutions to connect and learn more about the intersection of Web3 and longevity

October 28-30th & 4-6th Nov  - VitaDAO Hackathon - hackathon focusing on longevity is a strong requirement for the future of the longevity field. VitaDAO Hackathon registration is now open! Register here

Join Us! 

Have a special skill set? Anyone can contribute to VitaDAO and receive $VITA in return. Join the VitaDAO community on Discord and stay up to date on Twitter!  

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with monthly VitaDAO updates. 

Happy DeSci Fall! Inside this month’s newsletter: VitaDAO’s 5th IP-NFT Funding Ceremony A New Publication on Decentralized Technology Transfer The Longevity Decentralized Review Explainer Meet DAO Member: Estèfano Pinilla
Read more
Longevity Research Newsletter September 2022
September 9, 2022
Maria Marinova & Rhys Anderson
Awareness
Longevity
Longevity Research Newsletter September 2022

Welcome back to our monthly newsletter! This month we are excited to bring you an interview with Dr. Marco Demaria who has made some significant contributions to our understanding of cellular senescence. 

For those of you not well-versed in senescence, here's a little intro….

It used to be thought that given the correct culture conditions human cells would be able to proliferate indefinitely, however some pioneering experiments from Leonard Hayflick showed this was not the case and eventually cells would permanently  stop dividing - this phenomenon was coined 'cellular senescence'. Traditionally thought of as a tumour suppressor mechanism, research has shown that senescence is involved in a range of functions from development, to wound healing (Marco Demaria!). However, senescent cells have also been shown to elicit a pro-inflammatory response (senescence-associated inflammatory response - SASP) thought to attract immune cells to clear them. But as we age, an increase in the number of cells entering senescence, coupled with a decline in the ability of the immune system to clear them, leads to an accumulation of senescent cells and a chronic SASP which can paradoxically lead to a pro-tumourigenic environment and is associated with numerous diseases. Genetic and pharmacological removal of senescent cells has been shown to attenuate age-related diseases and extend lifespan in mouse models. Efforts to target/suppress senescent cells in humans (senolytic/senotherapy) have therefore gained significant traction both in academia and industry.

For some up-to-date perspectives and cutting edge research on cellular senescence (as well as other topics) check out our hot picks below!

Longevity Literature Hot Picks

So many fascinating papers were published this month that, in addition to our hot picks, we’ve had to include a Further Reading section for you at the bottom of this issue - enjoy! 

Nuclear morphology is a deep learning biomarker of cellular senescence

https://www.nature.com/articles/s43587-022-00263-3

A universal marker for cellular senescence has yet to be discovered. However, research from the Scheibye-Knudsen laboratory has utilised machine learning to detect senescent cells by nuclear morphology alone, with up to 95% accuracy in human fibroblasts, when compared to a cocktail of canonical senescence markers.

Cellular senescence: the good, the bad and the unknown

https://www.nature.com/articles/s41581-022-00601-z

Senescence and cancer — role and therapeutic opportunities

https://www.nature.com/articles/s41571-022-00668-4

Cellular senescence: a key therapeutic target in aging and diseases

https://www.jci.org/articles/view/158450#.YuhXTn-A0M4.twitter

Cellular senescence and senolytics: the path to the clinic

https://www.nature.com/articles/s41591-022-01923-y

Above are 4 reviews on senescence which each give unique perspectives ranging from our mechanistic understanding of the senescent phenotype, to its role in cancer and other diseases, to how we can target senescent cells and efforts to translate this to the clinic.

Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis

https://www.cell.com/cell/fulltext/S0092-8674(22)00790-5?rss=yes&utm_source=dlvr.it&utm_medium=twitter

Dysregulated mitochondrial homeostasis can result in switching cell death modalities and direct immune outcomes. The inflammasome is activated in Lrrk2 gain of function macrophages. Mitochondrial ROS directs the pore-forming protein gasdermin D to mitochondrial membranes, leading to inflammasome activation.

Naked Mole-Rat Hyaluronan Synthase 2 Promotes Longevity and Enhances Healthspan in Mice (under review)

https://papers.ssrn.com/sol3/papers.cfm?abstract_id=4185135

High molecular weight hyaluronic acid (HMW-HA) is known to aids cancer resistance and possibly healthspan the naked mole-rat. Overexpressing hyaluronan synthase 2 resulted in lower cancer incidence, increased lifespan and improved healthspan, likely by affecting inflammation, response to ROS and enhancing the gut barrier with age. 

Warburg-like metabolic transformation underlies neuronal degeneration in sporadic Alzheimer’s disease

https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00313-8#.Yv_gSYZ1KU8.twitter

With the aim to study the drivers of Alzheimer’s disease, the study identifies a metabolic switch to aerobic glycolysis in induced neurons from AD patient fibroblasts. Cancer associated PKM2 isoform was essential for this change and if chemically modulated to prevent nuclear translocation, neuronal metabolism was restored.

Astrocytic traffic jams in the aging brain

https://www.nature.com/articles/s43587-022-00270-4

Three distinct clusters of astrocytes were identified in aged mice hippocampi. One of them is novel and previously unrecognised, which was characterised with disrupted proteostasis. This contributed to a defective astrocyte morphology, synaptic maintenance and protein trafficking.

Cyanidin-3-O-glucoside promotes stress tolerance and lifespan extension of Caenorhabditis elegans exposed to polystyrene via DAF-16 pathway

https://www.sciencedirect.com/science/article/abs/pii/S0047637422001051?via%3Dihub

Microplastics are a growing pollutant in our environment and food chain. The effects and toxicity of a common microplastic polystyrene (PS) were explored in worms. As expected PS was toxic and decreased lifespan, which was mitigated by cyanidin-3-O-glucoside (C3G) treatment.

Chill out: environmentally relevant cooling challenge does not increase telomere loss during early life

https://www.sciencedirect.com/science/article/pii/S0016648022001332?via%3Dihub

Stressors early in life can have both positive (damage resistant) or negative (damage promoting) effects later in life. Timing and context is crucial for deciding the effect of environmental challenges. In birds, cooling has no effect on telomeres, but brood size and growth rate during nesting stage affect telomere shortening.  

Dose-response association between the daily step count and all-cause mortality: A systematic review and meta-analysis

https://www.tandfonline.com/doi/full/10.1080/02640414.2022.2099186

Getting in your 10,000 steps a day has become as popular a health target as eating your “5 a day” of fruit and vegetables. However, metrics such as these are often criticised for being too arbitrary. Now a meta-analysis of all-cause mortality shows a non-linear inverse relationship between the number of daily steps and chances of dying - so keep moving!

Measuring biological aging in humans: A quest

https://onlinelibrary.wiley.com/doi/10.1111/acel.13080

Biomarkers to quantify biological age are required in order to test the efficacy of interventions aimed at delaying the onset of age-related diseases. This review highlights the known hallmarks of ageing, their link to morbidity and how being able to quantify biological age could lead to a therapeutic revolution. 

Promotion of Hair Regrowth by Transdermal Dissolvable Microneedles Loaded with Rapamycin and Epigallocatechin Gallate Nanoparticles

https://www.mdpi.com/1999-4923/14/7/1404

Rapamycin - a drug which inhibits mTOR (one of the central regulators of metabolism) has been shown to be able to extend lifespan in numerous model organisms. New research shows that injection of rapamycin-containing nanoparticles can increase hair growth in mice in just 7 days, evidenced by increased hair follicle density and higher hair shaft growth rate. 

Clinical Trial Updates

Unity Biotechnology

Unity announced positive data from their Ph 2 BEHOLD study of UBX1325 in diabetic macular edema. The 12- and 18-week results are a promising step in validating their senolytic platform and supports the potential of UBX1325 in retinovascular diseases.

https://ir.unitybiotechnology.com/news-releases/news-release-details/unity-biotechnology-announces-positive-data-phase-2-behold-study

New / Active Rapamycin Human Clinical Trials (related to Aging)

https://www.rapamycin.news/t/new-active-rapamycin-human-clinical-trials-related-to-aging/2650

News

Peter Adams and collaborators at UCSD been awarded $10.6M from @NIH_CommonFund to join SenNet, an initiative to create a comprehensive atlas of how and where aging cells accumulate

Read more: https://bit.ly/3vVCTwd

LongevityTech.fund II launches – now accepting new investors

https://longevity.technology/news/longevitytech-fund-ii-launches-now-accepting-new-investors/

BioAge Partners With Age Labs to Decipher Healthy Longevity Using Samples and Data From a Preeminent Northern European Biobank

https://www.businesswire.com/news/home/20220823005823/en/BioAge-Partners-With-Age-Labs-to-Decipher-Healthy-Longevity-Using-Samples-and-Data-From-a-Preeminent-Northern-European-Biobank

Longevity Resources

Age-related Disease Spreadsheet

https://docs.google.com/spreadsheets/d/19wPcWmW868mmMvugL_N4DJjMLt0WpnLB3o7D6RaiGP8/edit#gid=0

The Norn Group has curated this useful resource containing an overview of age-related diseases with data on biology, epidemiology, clinical landscape, animal models and market research!

Articles

Decentralized investor communities gain traction in biotech

https://www.nature.com/articles/s41587-022-01459-z

Epigenetic ‘Clocks’ Predict Animals’ True Biological Age

https://www.quantamagazine.org/epigenetic-clocks-predict-animals-true-biological-age-20220817/

As billionaires race to fund anti-aging projects, a much-discussed trial goes overlooked

https://www.statnews.com/2022/08/09/anti-aging-projects-funding-much-discussed-trial-overlooked/

Martin O’Dea: longevity is burning bright in Ireland

https://longevity.technology/news/martin-odea-longevity-is-burning-bright-in-ireland/

40,000 pet dogs, one big question

https://www.theatlantic.com/science/archive/2022/07/senior-dog-aging-project-brains/671008/

These Six Biotechs are Winning the Race to Get AI-Designed Drugs to the Clinic

https://www.insideprecisionmedicine.com/artificial-intelligence/these-six-biotechs-are-winning-the-race-to-get-ai-designed-drugs-to-the-clinic/

The Opposite of Death Is Youth

https://tomhyde.substack.com/p/the-opposite-of-death-is-youth?r=hf6xj&utm_medium=ios

With advances in medicine, could 80 become the new 40?

https://www.cbc.ca/radio/spark/with-advances-in-medicine-could-80-become-the-new-40-1.6427495

Lengthening a woman's fertility may extend her life as well, research finds

https://edition.cnn.com/2022/08/20/health/extending-fertility-aging-life-itself-wellness/index.html

Book 

Methuselah's Zoo: What Nature Can Teach Us about Living Longer, Healthier Lives

Book by Steven N. Austad

Newly published book delving into the biology of long-lived organisms and what they can teach us about the biology of ageing. Check out an article written by the author Steven N. Austad in ‘The Atlantic’: 

https://www.theatlantic.com/science/archive/2022/08/clams-bivalves-long-lifespan-ming/671107/

Also, a review of the book by Prof. Charles Brenner: 

https://www.science.org/doi/full/10.1126/science.add9130?fbclid=IwAR0HCk6DOpclwY27gR9x0m0de3lA30sXwfQjdum7bKDdHxdzphjcpE2jhrk

Upcoming conferences

Longevity Summit Dublin 2022

September 18th-20th, Dublin

https://longevitysummitdublin.com/

Longevity Investors Conference

September 28-30, 2022, Gstaad, Switzerland

https://www.longevityinvestors.ch/

7th International Cell Senescence Association (ICSA) Conference

September 29th - October 1st, Groningen

https://www.icsa2022groningen.nl/

The Rejuvenation Startup Summit 

October 14-15, 2022, Berlin, Germany

https://forever-healthy.org/summit/

2nd VitaDAO Crypto meets Longevity Symposium

October 20th, Online

Check out our 1st symposium here: https://www.youtube.com/watch?v=GJ-rJAfjhBE

The Longevity Summit 2022

December 7th-8th, Buck Insitute for Aging, US

https://longevitysummit.io/

Funding Opportunities

Life Extension Ventures

https://www.lifex.vc/

A new $100 million fund that is focused on life extension for "people and planet."

Job board

Research Associate Position, Ochre Bio, New York, US

Ochre Bio works on chronic liver health challenges, from reducing cirrhosis complications through metabolic stress, metabolism, and regeneration programs.

https://jobs.lever.co/ochre-bio/c8da319b-f300-4945-9a1d-1cfc97fc9c29

Post-doc Position in Dr. Marco Demaria’s Lab - ERIBA Labs, Groningen, Netherlands

https://www.linkedin.com/feed/update/urn:li:share:6963462684604252160

The Demaria lab are looking for a motivated postdoc to lead a project on chemotherapy-induced senescence.

Post-doc Position in David James’ and Mark Larance’s Lab - Charles Perkins Centre, University of Sydney, Australia

Interested in healthy ageing? David James and Mark Larance are looking for a motivated postdoc with experience in proteomics.

Please apply to David.james@sydney.edu.au

PhD Studentship in Dr. Ina Huppertz’s Lab - Max Planck Institute for Biology of Ageing, Cologne, Germany

https://www.ageing-grad-school.de/phd/host-lab-proposals/dr-ina-huppertz

Seeking a PhD student with a strong interest in RNA biology and metabolism.

Interview with Dr. Marco Demaria

Dr. Demaria gained his PhD in Molecular Medicine from the University of Torino, Italy where he researched the role of cellular metabolism and chronic inflammation on tumour progression. He then joined Prof. Judith Campisi's lab at the Buck Institute where he developed tools and methods to analyse senescent cells in vivo and made the important discovery of the role of transient cellular senescence in wound healing. He is now an Associate Professor at the ERIBA Labs and Group Leader of the Cellular Senescence and Age-Related Pathologies laboratory.

What inspired you to enter longevity research?

From a personal perspective, growing up living in close contact with my grandparents exposed me to the understanding of the ageing process very early in life. From a professional perspective, I began to do biomedical research on cancer. Looking for potential cancer drivers in the tumor microenvironment I encountered cellular senescence. I decided to join the laboratory of Judith Campisi to study it, and learned how senescence might be implicated in virtually every age-associated disease, not only cancer. This eventually inspired me to focus on the possibility that we might be able to target age-related mechanisms to extend quality of life and healthy longevity.

Which of the current theories of ageing do you think are the most convincing?

I have always been a supporter of antagonistic pleiotropy. I find it quite logical that evolution selected traits related to optimal fitness at young age without caring of the detriment that they could provoke in post-reproductive stages. I think the main topic of my research, cellular senescence, well fits that theory. While less convinced by the experimental evidence, I do not discard the idea of programmed aging. This is mainly driven by the fact that certain ageing mechanisms seem to follow a very regulated and reproducible trajectory.

How has the field changed since you started?

When I started in the field, pharmacological approaches to reduce age-associated features and to extend healthy longevity were very sporadic. Only when the hallmarks of aging started to become popularized, mainly thanks to the review in Cell in 2013, the pharma industry realized there were specific mechanisms to target. Also, more funding opportunities from public and private sources became available to researchers, thus boosting aging research. The field is exponentially growing, and we are just scratching the surface of understanding how ageing works. Exciting times.

What mistakes do you think the longevity field has made?

I think the longevity field has been too quick to move interventions forward, and there are too many parallel efforts in this direction with no standards and minimal exchange of knowledge. We risk to find that targeting individual hallmarks of ageing is only minimally impactful, and also to be unable to compare different studies. But making mistakes is not a problem if we learn something from them. In this case, a failure would direct towards a better standardization of how we read out ageing and towards development of combinatorial and synergistic approaches.

Other than your own, what do you think have been the biggest/important discoveries in the field?

Several discoveries from outstanding laboratories worldwide related to understanding basic mechanisms have been instrumental to improve our knowledge on how organisms age. In terms of therapeutic approaches, besides the concept that elimination of senescent cells can delay onset and progression of tissue dysfunctions, the idea that we can reprogram cells has been revolutionary for its potential in regenerative medicine.

What advice would you give to people currently working in longevity research?

First, try not to rush to conclusions. We are just starting to understand how various age-associated traits interact, and we should invest more resources for fundamental research in this area. Moving too quickly to interventions is risky and might jeopardize the current interest in the field. Second, let’s be more collaborative and collegial. I’m starting to see too many people pushing their ideas and not sharing with peers. Too many scientists talking about compound a and compound b without disclosing any detail. This can harm progress and knowledge.

Which aspect of longevity research do you think requires more attention?

As mentioned before, the formal description of the hallmarks of ageing popularized longevity research. However, this is an oversimplification. We should not think we have fully understood how ageing works, and we should invest more resources in doing basic science, not only in developing therapeutics. Finally, it is important to note that the current pharmacological approaches are mainly derived from what we are learning on the mechanisms engaged by good lifestyle choices – mainly diet and physical exercise. We need to think that these approaches might not work on individuals that are already taking good lifestyle choices. More attention should be taken in understanding what can further delay ageing on already long-lived organisms, and support more research in this area.

Is ageing a disease?

No, aging is a condition that increases our predisposition to develop diseases.

You have made significant advances to the field of cellular senescence. Could you summarise your discovery of the role that senescent cells play in wound healing and what the potential therapeutic implications could be?

Senescent cells are transiently induced during tissue damage and they promote optimal tissue repair. Currently, we know this phenomenon is happening in the heart, liver, kidney and skin but we think it might be an event occurring in every tissue. Because of this function, pro-senescence therapies, in particular interventions that can switch cell fate from death to senescence, can have an enormous potential to improve recovery from acute tissue injury and to avoid pathological sequelae.

There still lacks a universal marker for senescence – what do you think are the most robust markers discovered so far?

The three most senescence-associated markers remain activation of the lysosomal enzyme bgal, induction of the cell cycle inhibitor p16 and loss of the nuclear protein LaminB1. All these markers are not 100% specific, so combinations of them have the highest chance to reveal senescence in biospecimens. It is becoming more and more clear that different subtypes of senescent cells express subset-specific markers. Studies on senescence should start to incorporate such markers to identify the exact subpopulations.

Although senescence can be thought of as a tumour suppressor mechanism, research from you and others has highlighted the detrimental effect senescent cells can have. What do you think are the most promising senolytic strategies and what are the potential negative impacts of removing senescent cells?

There are several strategies to eliminate senescent cells, most of them leading to activation of apoptosis. Most of these approaches are based on re-purposed small molecules or natural compounds. The advantage is that they normally have a well-characterized safety profile in humans, and thus are more ready to enter clinical trials for a new indication. The disadvantage is that these compounds have senolytic properties but also additional senescence-independent on-target effects. This might lead to unwanted side effects and adverse reactions. Senolytics with increased targeted activity are under development, and promising results are shown by using senescence-specific peptides and gene therapy strategies. The potential negative impacts of targeting senescent cells might be reduced tumor immunosurveillance and impaired ability to repair acute tissue damages.

Outro

Thanks for your interest in the space and joining us yet again for VitaDAO’s Monthly Longevity Newsletter!

Don’t hesitate to contact us to let us know what you want to see next and give any feedback. We would love to hear from you! Until next time and if you’ve missed this month’s highlights, check this out on YouTube:

The Longevity Prize Opening Ceremony | with VitaDAO, Foresight Institute & the Methuselah Foundation - YouTube

Further Reading

Cellular senescence in neuroinflammatory disease: new therapies for old cells?

https://www.cell.com/trends/molecular-medicine/fulltext/S1471-4914(22)00183-6

Age and life expectancy clocks based on machine learning analysis of mouse frailty

https://www.nature.com/articles/s41467-020-18446-0

Exercise as an Aging Mimetic: A New Perspective on the Mechanisms Behind Exercise as Preventive Medicine Against Age-Related Chronic Disease

https://www.frontiersin.org/articles/10.3389/fphys.2022.866792/full

Resistance Exercise Training as a Primary Countermeasure to Age-Related Chronic Disease

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6563593/

A distinct astrocyte subtype in the aging mouse brain characterized by impaired protein homeostasis

https://www.nature.com/articles/s43587-022-00257-1

Senescence-associated morphological profiles (SAMPs): an image-based phenotypic profiling method for evaluating the inter and intra model heterogeneity of senescence

https://www.aging-us.com/article/204072/text

New horizons in life extension, healthspan extension and exceptional longevity

https://academic.oup.com/ageing/article/51/8/afac156/6653481?login=true

Transformed cells after senescence give rise to more severe tumor phenotypes than transformed non-senescent cells

https://www.sciencedirect.com/science/article/pii/S0304383522003342?via%3Dihub

Somatic mutations in single human cardiomyocytes reveal age-associated DNA damage and widespread oxidative genotoxicity

https://www.nature.com/articles/s43587-022-00261-5

Plasma proteome profiling of healthy individuals across the life span in a Sicilian cohort with long-lived individuals

https://onlinelibrary.wiley.com/doi/10.1111/acel.13684

Living in endemic area for infectious diseases accelerates epigenetic age

https://www.sciencedirect.com/science/article/pii/S0047637422000951

Metabolic changes in aging humans: current evidence and therapeutic strategies

https://www.jci.org/articles/view/158451

Developmentally-programmed cellular senescence is conserved and widespread in zebrafish

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7585104/

Autophagy-nutrient sensing pathways in diabetic complications

https://www.sciencedirect.com/science/article/abs/pii/S104366182200353X?via%3Dihub

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with Longevity Research.

Welcome back to our monthly newsletter! This month we are excited to bring you an interview with Dr. Marco Demaria who has made some significant contributions to our understanding of cellular senescence. 
Read more
VitaDAO Newsletter Issue No 9 September 2022
September 7, 2022
Sarah Friday
Awareness
VitaDAO Newsletter Issue No 9 September 2022

Dear Vitalians, We are glad you are here! 

Inside this month’s newsletter:

  • Pfizer, L1 Digital and Shine Capital Contribution Proposals
  • Passed VitaDAO Proposals 
  • Meet DAO Member: Hamza Qureshi
  • Upcoming Events, including a 2nd VitaDAO Crypto meets Longevity Symposium

DAO News

ARDD 2022: Annual Aging Research and Drug Discovery Meeting

From August 29th to September 2nd, longevity industry leaders gathered in Copenhagen, Denmark for the 9th Annual ARDD Meeting. VitaDAO served as an event sponsor, helping to bring together researchers and investors from across the world to discuss advances in aging and drug discovery. Many VitaDAO contributors were present as both attendees and panel speakers at the event. Missed the event? You can find recordings of the Longevity Medicine workshops on ARDD’s Youtube

VitaDAO Core Member and Dealflow Steward, Laurence Ion, spoke as part of the “Investing in Longevity Biotechnology” panel at AARD 2022. 

Pfizer Ventures Application

Pfizer Ventures applied to VitaDAO's Institutional Genesis Raise, proposing to contribute $500,000 USD to VitaDAO and participate in the governance of VitaDAO using $VITA tokens. If selected, Pfizer Ventures plans to support VitaDAO beyond the capital contribution with access to deal flow, the ability to support incubation and commercialization efforts of the intellectual property (IP), and the ability to raise awareness for VitaDAO’s mission. 

L1 Digital Application

L1 Digital applied to VitaDAO's Institutional Genesis Raise, proposing to contribute $500,000 USD to VitaDAO and participate in the governance of VitaDAO using $VITA tokens. If selected, L1 Digital plans to support VitaDAO beyond the capital contribution with on-chain and off-chain Market Makers to help improve the liquidity of $VITA, access to an extensive network of limited partnerships, assistance with token economic design, experience with dealflow, and support of VitaDAO’s inner initiatives. 

Shine Capital Application

Shine Capital applied to VitaDAO's Institutional Genesis Raise, proposing to contribute $1,000,000 USD to VitaDAO and participate in the governance of VitaDAO using $VITA tokens. If selected, Shine Capital plans to support VitaDAO beyond the capital contribution by providing strategic guidance surrounding project selection, leveraging financial and business acuity to help build a durable contributor framework, and contributing a network of capital partners, contributors, and go-to-market partners. 

VitaDAO Longevity Research Newsletter

Authors Maria Marinova and Rhys Anderson released an August VitaDAO Longevity Newsletter. Inside this newsletter, find longevity literature hot picks, read about longevity clinical trial updates, and meet Professor Matt Kaeberlein. This newsletter is a valuable resource for keeping up to date with longevity research happening around the globe! 

Governance

VDP-54 Passed!

As a result of VDP-54, approximately 6% of the total VITA token supply will be allocated to new strategic contributors admitted as institutional members of VitaDAO. Through an Institutional Genesis Raise, VitaDAO aims to raise at least 4 million USD in mission-aligned funding by auctioning VITA to strategic contributors selected by the VitaDAO community. As part of the auction, an individual or entity can craft a proposal for VitaDAO to become a strategic contributor with a written application to the VitaDAO community. This addition of institutional members will extend VitaDAO’s reach, advance VitaDAO’s mission, and enable additional longevity research funding, deal flow, and community awareness! 

VDP-53 Passed! 

VDP-53 supports the Longevity Decentralized Review (TLDR), an on-demand service that funds peer review using funds from those who submit their research to be reviewed. TLDR aims to grow VitaDAO’s treasury, grow VitaDAO longevity knowledge, and augment VitaDAO’s longevity dealflow by creating a larger more decentralized community evaluating longevity research. 

The main goal of TLDR is to enable researchers to have proof of their peer reviews such that they do not need to rely on journals for proof. VitaDAO is funding 20,000 $VITA tokens to pay out to reviewers.

Live on Discourse

Find all of VitaDAO’s pending live proposals on Discourse, VitaDAO’s governance hub for proposals before they are moved on-chain. These proposals are open for engagement, voting, questions, and conversation! This month, several new proposals were added to Discourse. Two are outlined below:

  • VDP 55: VitaDAO <> AthenaDAO Partnership
VDP-55 proposes a “DAO2DAO relationship” between VitaDAO and AthenaDAO, a decentralized collective to fund women’s reproductive health research and drug discovery. This relationship would involve the exchange of 50k $VITA for either 3% of $Athena or 1.2x the monetary value in $ATHENA, once a token has been launched. 

Community Member Highlight: Hamza Qureshi 

How did you first get involved in web3?

I am not particularly enamored with how the employment contracts in general - and the non-compete clauses in particular - work, at least in North America. A typical non-compete clause basically prohibits the employee from engaging in any business, profession, or project activity that is in competition with the employer’s business, after the employment relationship ends. This clause is time and geography dependent, and in a number of cases, hard to enforce. I believe that everyone - if they can manage - should engage in side consulting projects, etc. all the time because this is good personal and professional exposure. There is usually a net positive for all parties involved at the end of the day. I joined the web3/DAO space because it allows this kind of cross-pollination more readily. The efficacy of the collaborative web3 work is yet to be determined, however.

Also, I heard people make money in web3.

What attracted you to VitaDAO?

After discovering NFTs, I scoured the space for projects that were embedding NFTs in spaces/industries that were going to be around in 5, 10, or more years. That is when I stumbled upon Molecule and found that VitaDAO was the best channel to get involved in the mission through. A lot has changed since then - for the better

What is your current role in VitaDAO? 

Operations, Project Management, Onboarding and Community, a little bit of Governance. I get to see large swathes of the VitaDAO pipeline, which is exhilarating or exhausting depending on the day. I like to say VitaDAO is increasing lifespan but discord is decreasing lifespan, so we are back to square 1. 

 

What does the future hold for VitaDAO?

VitaDAO is a risky but exciting experiment that is slowly gaining traction. What I cannot wait to see - like a lot of people in and outside the organization - is whether this experiment becomes financially self-sustaining at some point in the future. As VitaDAO is a trailblazer in the space, the answer to this question will probably affect the validity and optics of the entire DeSci industry. I am extremely optimistic about the future social impact of the organization regardless of the finances.

Any closing thoughts for readers?

I have asked biologists about this but haven't received a satisfactory answer. How 'decentralized' are organizations such as ant colonies and bee hives? How do biologists measure decentralization and what is considered optimal? If it is different in different contexts, why? I would invite the readers to think about this and let me know their thoughts. 

Another thing to think about is decentralization in the context of psychology. When I am working with people - in a supposedly flat organization - with different skills, backgrounds, and perspectives, I naturally create hierarchies in my head. For instance, if there is an exceptional marketer in our midst, they become the head of the marketing department in my head. It turns out navigating life is not possible without such hierarchical structures in place. This makes me wonder if 100% decentralized organizations are really a good idea. Where is the sweet spot in this context?

VitaDAO in the Media

VitaDAO was featured in a Nature Biotechnology News Feature, “Decentralized investor communities gain traction in biotech”. In this exciting feature, learn about VitaDAO’s origin story and read about VitaDAO’s role in decentralizing research funding.

VitaDAO co-founder, Paul Kohlhaas, spoke on (DAO,DAO)’s Twitter Space about Decentralized Science

In a Neo.Life article by Richard Sprague, read about VitaDAO’s role in disrupting science, as we know it. 

Inside their biotech analysis, BowtiedBiotech touches on Pfizer's interest in VitaDAO. 

Upcoming Events 

September 14th at 1 PM EST- (Recurring Bi-Weekly on Wednesdays)- Join a VitaDAO onboarding session! Here, you can ask questions about VitaDAO, meet others interested in the DAO, and learn about VitaDAO’s working groups. 

September 13th at 12 PM EST- Celebrate alongside VitaDAO and Molecule at VitaDAO’s fifth public IP-NFT Ceremony to commemorate the funding of the ApoptoSENS project. 

October 20th- Join hosts Max Unfried and Eleanor Sheekey at VitaDAO’s second VitaDAO Crypto meets Longevity Symposium. The symposium will serve as a chance for scientists, blockchain experts, and longevity professionals from internationally recognized institutions to connect and speak about the intersection of longevity and the blockchain. 

Thanks for Reading! 

VitaDAO is defined by an active and diverse community of contributors. To learn more about the community, join our Discord and follow us on Twitter!  

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with monthly VitaDAO updates. Already subscribed? Forward this newsletter to a friend!

Dear Vitalians, We are glad you are here! Inside this month’s newsletter: Pfizer, L1 Digital and Shine Capital Contribution Proposals Passed VitaDAO Proposals Meet DAO Member: Hamza Qureshi Upcoming Events, including a 2nd VitaDAO Crypto meets Long
Read more
Set up MetaMask and receive your first VITA tokens
August 25, 2022
Maria Marinova
Awareness
Tokenomics
Set up MetaMask and receive your first VITA tokens

1. Make sure you are using a Chromium-based browser, such as Chrome, Brave, Firefox or Edge and install the browser extension MetaMask, a reputable and easy-to-use Ethereum wallet.

2. When prompted, allow the extension to be added to your browser and on the next screen continue with “Get Started”.

3. Click on “Create a Wallet” and, on the next screen, choose whether you would like to provide MetaMask with optional analytical data.

4. Create a secure password and accept the Terms of Use. 

We recommend you think of your MetaMask wallet like a bank account. Make it a secure password and store it in a safe place, such as a password manager.

5. Very important: Follow the instructions on your screen carefully in order to securely store your “backup phrase”. This, and only this phrase, guarantees you access to your wallet and all tokens contained inside. DO NOT share this phrase with anyone and keep it safe (ideally on paper). If lost, you cannot restore your account and lose your tokens. 

6. Confirm your phrase and continue.

7. MetaMask is set up successfully if you see a big “Congratulations” on your screen. Great - click “All Done”! Dismiss the release notes and make yourself comfortable with your newly created Ethereum wallet.

8. You can find your wallet address under the label “Account 1”. Move your cursor on top of it and click  to copy your public Ethereum address to your clipboard and save it for later. We need this address in order to send you VITA tokens.

9. You will not be able to see your VITA even if it has been sent, before you add it as a custom token. Click on “Import Token” at the bottom of the wallet page and then “Custom Token”. Add the Token Contract Address, which is 0x81f8f0bb1cB2A06649E51913A151F0E7Ef6FA321

Click on “Next”. Click on “Add Tokens”. Now, you see your VITA balance.
Congratulations, you now have acquired VITA tokens and become a member of VitaDAO with full voting rights.

Set up MetaMask and receive your first VITA tokens
Read more
Longevity Research Newsletter August 2022
August 11, 2022
Maria Marinova & Rhys Anderson
Longevity
Awareness
Longevity Research Newsletter August 2022


Introduction

We can all agree that the age-associated decline in physiological functionality and increased risk of developing diseases is terrible, but what can we do to treat, slow down or even prevent it?

Many people in the longevity space would agree that from the interventions explored so far, rapamycin is the one holding the most promise. This inhibitor of one of the central regulators of metabolism — mTOR — has shown positive and consistent results in extending healthy life across multiple experimental models. Research from Prof. Matt Kaeberlein’s lab showed that even transient treatment of rapamycin in middle-aged mice was enough to extend remaining lifespan by up to 60%! This would equate to around an extra 20 years of life for a 50 year-old human, however clinical trials are needed to ascertain how translatable this would be.

To highlight the importance of rapamycin, in this issue we interviewed Prof. Matt Kaeberlein and picked an interesting hot-off the press paper that focuses on the connection between development and aging, showing how even a short treatment with rapamycin in early life can also lead to a significant extension of lifespan in both flies and mice.

VitaDAO-funded Research Projects

This is the first month since starting the Longevity Newsletter that we have not funded a new project (although there are many in the pipeline) — so thought it would be a great opportunity to introduce you to the newly launched Project Pages which summarise previously funded projects and provide all the necessary links to learn more.

https://www.vitadao.com/projects/scheibye-knudsen-lab

https://www.vitadao.com/projects/turn-biotechnologies

https://www.vitadao.com/projects/rubedo-life

https://www.vitadao.com/projects/korolchuk-lab

https://www.vitadao.com/projects/evandro-fang

Longevity Literature Hot Picks

Transient rapamycin treatment during developmental stage extends lifespan in Mus musculus and Drosophila melanogaster

https://www.embopress.org/doi/full/10.15252/embr.202255299

Administration of a short rapamycin treatment during a specific time window in early life extends Drosophila lifespan, showing again the tight link between development and ageing. A potential suggested mechanism is the upregulation of sulfotransferases during early rapamycin treatment in both newborn Drosophila and mice.

Single-cell analysis of the aging female mouse hypothalamus

https://www.nature.com/articles/s43587-022-00246-4

Specific hypothalamic cell type ageing signatures were found for neuronal subtypes, microglia and astrocytes by single nuclei RNA sequencing. As expected, changes in those cell types were linked to metabolism, body composition and cognition. A surprising finding was the female specific ageing pattern driven by elevation of Xist, driver of X inactivation.

Age-dependent impact of two exercise training regimens on genomic and metabolic remodelling in skeletal muscle and liver of male mice

https://www.nature.com/articles/s41514-022-00089-8

Adult and old mice were assigned to three exercise regimens: sedentary, moderate intensity and high intensity training. The highest improvement in muscle strength, body composition and fasting blood glucose were observed in the moderate intensity aged group, while the effects for the old high intensity training were much less pronounced.

Age-associated differences in the cancer molecular landscape

https://www.cell.com/trends/cancer/fulltext/S2405-8033(22)00135-2

Certain somatic mutations occur more often in old or young cancer patients. A lot of the age-related gene expression and epigenetic changes regulate tissue microenvironment via extracellular matrix organisation, immune processes and angiogenesis with profound impact on cancer development.

A life-course approach to cardiovascular disease prevention

https://www.nature.com/articles/s41591-022-01870-8

Preventing atherosclerotic cardiovascular diseases, which is still the biggest killer in the western world, should start focusing on long-term strategies starting from an early age. New public health approaches will be needed to incentivise families with children to mitigate risk factors across the lifespan.

Is ageing a disease?

https://www.thelancet.com/journals/lanhl/article/PIIS2666-7568(22)00154-4/fulltext

An international group of clinicians have disputed the proposed revision to WHO International Classification of Disease to include the term “old age” as a diagnostic category. This term was retracted and replaced with “ageing associated decline in intrinsic capacity”.

New intranasal and injectable gene therapy for healthy life extension

https://www.pnas.org/doi/10.1073/pnas.2121499119

High-capacity cytomegalovirus (CMV) was used to deliver telomerase and follistatin both internasally or as an injectable gene therapy.This resulted in median life extension of 41.4% and 32.5% for each respective gene. Healtspan indicators such as glucose tolerance and physical performance were improved. Telomeres and mitochondrial structure were protected.

Targeting the “hallmarks of aging” to slow aging and treat age-related disease: fact or fiction?

https://www.nature.com/articles/s41380-022-01680-x

The authors highlight some weaknesses in the theory that targeting hallmarks of aging can treat age-related disease. It is difficult to make conclusions because of the overreliance on lifespan as a marker of aging, study designs that cannot clearly estimate intervention effect, and models with ambiguous relevance.

Telomeric 8-oxo-guanine drives rapid premature senescence in the absence of telomere shortening

https://www.nature.com/articles/s41594-022-00790-y

Oxidative damage did not induce premature telomere shortening as expected but it did, however, result in p-53 dependent senescence, telomere fragility and impaired replication by oxidative base lesions (8oxoG).

Systemic induction of senescence in young mice after single heterochronic blood exchange

https://www.nature.com/articles/s42255-022-00609-6

Blood from aged mice induced senescence in the young animals after a single heterochronic exchange. This effect was attenuated when the old mice were treated with a senolytic prior to blood exchange, indicating old blood rejuvenation by senescent cell clearance.

Senolytic treatment reduces cell senescence and necroptosis in Sod1 knockout mice that is associated with reduced inflammation and hepatocellular carcinoma

https://onlinelibrary.wiley.com/doi/full/10.1111/acel.13676

Sod1KO mice exhibiting accelerated aging phenotype were treated with dasatinib and quercetin (D+Q). Senescence marker p16 and numerous SASP factors ware reduced in the liver after 7 months of treatment. Expression of liver cancer and necroptosis genes was also reduced but fibrosis markers were unaffected.

A complex systems approach to aging biology

https://www.nature.com/articles/s43587-022-00252-6

The new theoretical framework is built on foundations of the principles of homeostasis, evolutionary and network theories. It proposes that organismal aging is dependent on the disruption of regulatory pathways at different hierarchical scales.

Exercise Counters the Age-Related Accumulation of Senescent Cells

https://journals.lww.com/acsm-essr/Abstract/9900/Exercise_Counters_the_Age_Related_Accumulation_of.8.aspx

Exercise attenuates multiple molecular pathways associated with aging. It can also activate an immune response in order to drive senescent cell clearance.

Histone deacetylase 4 reverses cellular senescence via DDIT4 in dermal fibroblasts

https://www.aging-us.com/article/204118/text

The role of histone deacetylase 4 (HDAC4) in cellular senescence and skin aging was investigated by RNA sequencing after OE and KD of HDAC4. DNA damage-inducible transcript 4 (DDIT4) was identified and a molecular target of HDAC4 and senescence associated factor. Its OE reversed SASP and aging-related gene expression.

Senescence: An Identity Crisis Originating from Deep Within the Nucleus

https://www.annualreviews.org/doi/10.1146/annurev-cellbio-120420-013537

Senescence is discussed as a cell identity shift and a gain-of-function phenotype, rather than loss-of-function. The cellular functionality and epigenetic features of senescent cells are defined, highlighting their heterogeneity.

The Achilles’ heel of cancer survivors: fundamentals of accelerated cellular senescence

https://www.jci.org/articles/view/158452

Cytotoxic cancer treatments induce senescence in cancer survivors and so accelerate aging and the onset of chronic disease. This review highlights senescence as a mechanism of this phenotype and provides preclinical and translational evidence of the potential of senolytics to mitigate this.

News

Longevity Foundation to Fund Geroscience Research with €860M

https://www.labiotech.eu/trends-news/geroscience-longevity-foundation/

Decentralising biotech — Molecule raises $12.7 million in seed funding

https://longevity.technology/decentralising-biotech-molecule-raises-12-7-million-in-seed-funding/amp/

Deep Longevity Granted The First Microbiomic Aging Clock Patent

https://www.linkedin.com/pulse/deep-longevity-granted-first-microbiomic-aging-clock-patent

Insilico Medicine’s AI uncovers 28 new potential drug targets for ALS

https://www.fiercebiotech.com/medtech/insilico-medicines-ai-uncovers-28-new-potential-drug-targets-als

Articles

Why longevity matters to everyone: Living longer lives in the world of Web3

https://cointelegraph.com/news/why-longevity-matters-to-everyone-living-longer-lives-in-the-world-of-web3

The Aging Research Is On The Rise

https://www.linkedin.com/pulse/aging-research-rise-andrii-buvailo-

How sure are we that physical activity makes us live longer?

https://blogs.bmj.com/bjsm/2018/03/20/how-sure-are-we-that-physical-activity-makes-us-live-longer/

More Life — Decoding the Secret of Aging

https://m.dw.com/en/more-life-decoding-the-secret-of-aging/av-62441589?maca=en-Twitter-sharing

Aging is Still One of Biology’s Greatest Mysteries

https://www.discovermagazine.com/health/aging-is-still-one-of-biologys-greatest-mysteries

Should We Consider Aging a Disease?

https://quillette.com/2022/07/28/whats-in-a-disease/

Upcoming conferences

Ending Age-Related Diseases

August 11–14th, virtual

https://www.lifespan.io/ending-age-related-diseases-2022/

9th Aging Research and Drug Discovery (ARDD) Meeting

August 29th — September 2nd, Copenhagen

http://www.agingpharma.org/

Longevity Summit Dublin 2022

September 18th-20th, Dublin

https://longevitysummitdublin.com/

7th International Cell Senescence Association (ICSA) Conference

September 29th — October 1st, Groningen

https://www.icsa2022groningen.nl/

2nd VitaDAO Crypto meets Longevity Symposium

Coming this fall (date TBC), Online

Check out our 1st symposium here: https://www.youtube.com/watch?v=GJ-rJAfjhBE

Clinical Trials

Insilico announces first patient dosed in phase I study in China of its first AI-discovered anti-fibrotic drug candidate with novel target.

https://www.eurekalert.org/news-releases/960247

Job board

Interested in pursuing a scientific career against ageing?

Check out https://rejuvenomicslab.com/ for opportunities in Prof. João Pedro de Magalãhes Lab

Research associate/Senior research associate wanted!

Dorian Therapeutics works on senoblockers to rejuvenate cells and tissues and treat age-related diseases. More about Dorian: https://www.doriantherapeutics.com/

Position application link: https://www.linkedin.com/jobs/view/research-associate-senior-research-associate-at-dorian-therapeutics-3162063900/?fbclid=IwAR3TJ0fI7fHmgBq8aOgjLRAzwHGN7TpYSkm7X0bkEFvQP1AGuDe4uRD9sPQ

Funding

Fellowship applications are open!

Scientists, engineers, and entrepreneurs in longevity, molecular machines, neurotech, computing, space, and more; we want to support you working on flourishing futures!

https://foresight.org/foresight-fellowships/

Prizes

We’re excited to bring you a new section to the newsletter this month — PRIZES!

The Longevity Prize

https://www.longevityprize.com/

VitaDAO, the Foresight Institute and the Methuselah foundation have joined forces, with the help from donors such as Vitalik Buterin to create and fund the Longevity Prize — launching with the Hypothesis Prize of $20k for person who provides the best answer for what they perceive to be the most promising but underappreciated/obscure area of longevity science that should be pursued. More prizes will also be launching soon for addressing various aspects of longevity research.

Maximon Longevity Prize

https://www.maximon.com/longevityprize

“The Maximon Longevity Prize for Translational Research awards outstanding breakthrough research in the field of longevity that can be translated for clinical or non-clinical applications and has the potential to substantially increase health span or life span of humans.”

Interview with Professor Matt Kaeberlein


Prof. Matt Kaeberlein carried out his PhD in Prof. Lenny Guarente’s lab at MIT where he began, and has continued to make, significant contributions to our understanding of the evolutionary conserved mechanisms of ageing. Currently a professor at Washington University, Prof. Kaeberlein has won numerous prizes and been elected to several gerontology/scientific societies. He also co-directs the pioneering Dog Aging Project (https://dogagingproject.org/) which works at increasing our understanding of ageing, whilst aiming to help both dogs and humans live longer, healthier lives.

What inspired you to enter longevity research?

I attended a talk by Prof. Lenny Guarente during my first year in graduate school at MIT where he discussed how his lab was using genetics, molecular biology, and biochemistry to study the biology of aging. Up to that point, I thought I would do structural biology or x-ray crystallography for my PhD, but I was so inspired by Lenny’s talk that I joined his lab and the rest is history.

How has the field changed since you started?

I’d say there have been a few big changes, mostly positive. One negative change in my view is that the field has somewhat narrowed in scope. It used to be that many people were taking unbiased approaches to identify new mechanisms and determinants of longevity. Almost nobody is doing that anymore, despite the fact that we’ve only explored a small fraction of the “intervention space” in any organism. We don’t know what we don’t know, but unfortunately very few people are looking for what we don’t know. This concerns me.

On the positive side, we now accept conserved mechanisms of aging exist and we understand a fair amount about those conserved mechanisms of aging. When I started in this field, I remember many people arguing that nothing about aging in a worm or a yeast or a fly would be shared with a mouse or a human. It is also now commonly accepted that it’s possible to intervene in aging biology during middle age, which greatly facilitates translational geroscience. It was really the Harrison et al. rapamycin study in 2009 that launched this change in perception. Before that, most people thought you would need to start early to have any significant effect in a mammal.

The field has also gained some credibility in the mainstream scientific community, larger funding sources seem to be coming online although we still lag badly in federal funding, and we are starting to see a proliferation of longevity biotech, which is fantastic.

What mistakes do you think the longevity field has made?

I think the field in general has been too slow to correct bad science in the literature. Irreproducible and overhyped studies have resulted in the waste of hundreds of millions of dollars, stained the reputation of the field, and slowed the entry of biotech and big pharma into the field.

Other than your own, what do you think have been the biggest/important discoveries in the field?

I think the Harrison et al. 2009 rapamycin study was a big one because it showed for the first time that a drug could significantly increase lifespan when treatment was started during middle age. The fact that this was an accident just makes it a better story. The discovery of senescent cells as a major in vivo contributor to age-related functional decline and disease has also obviously been hugely important and provided several opportunities for translational applications. Epigenetic and other “clocks” have had a big impact in popularization of the science and have the potential to be transformative for personalized geroscience. Reprogramming might be the breakthrough of the century — time will tell.

What advice would you give to people currently working in longevity research?

Practice healthy skepticism, of your own work and others. Be careful not to fall into the trap of confirmation bias. Always seek to identify where your model is weakest and try to break it, because that’s the only way you’ll make it better.

Which aspect of longevity research do you think requires more attention?

First, I think we need more quality clinical trials. There has been an uptick in geroscience clinical trials, which is great, but not enough well-powered and well-designed clinical trials. That’s not a knock on anyone, but a reflection of the lack of funding put towards this.

Second, I’d like to see more effort put on unbiased approaches to figure out what we’ve missed so far. In many ways the Hallmarks of Aging have been great for the field, but they have also contributed to a narrowing of scope where people have a hard time thinking outside of the hallmarks and it’s very difficult to get a grant to study anything that doesn’t fit that framework. There is a huge intervention space we have not probed in order to identify new longevity factors, which could seed future mechanistic and translational discovery.

Is ageing a disease?

That’s the wrong question. It just distracts from what’s important. Aging biology is modifiable, and we can treat it to prevent disease and maintain health.

What do you think is the biggest challenge in bringing rapamycin to the clinic to treat and prevent age-related diseases?

Money. If sufficient resources were put toward demonstrating safety and efficacy for the many indications where it seems likely rapamycin would work, the other roadblocks would fall away.

What is the most important experiment/trial you would do right now to advance the field, if there were no financial, regulatory or other restraints?

I’m not sure if it’s the most important, but one thing I would do is invest the resources to create an infrastructure that would allow for geroscience intervention testing in companion animals at the highest ethical and safety standards. We’ve created a template for how to do this with the Dog

Aging Project rapamycin trial, but it is still extremely difficult to recreate this, and nobody is really able to carry out a similar trial right now, despite the fact that there are probably 5–10 interventions that could be tested today. The benefits of showing efficacy for a geroscience intervention (or more than one) in companion animals are enormous for the field, the time frame (3–4 years) is very reasonable, and the cost is small compared to human clinical trials. It seems like a no-brainer to me.

Which other interventions do you think hold promise for improving human healthspan/lifespan?

I know it’s boring, but exercise, particularly resistance training, is the sure bet for most people to get biggest bang for the buck.

I actually think the interventions with the most promise are just sitting out there waiting to be discovered, but almost nobody is looking for them. Which is why I feel pretty excited to start exploring the intervention space at scale.

Among those we know about now, epigenetic reprogramming probably has the most upside, but in part that’s because it’s still early in development, and my intuition is that while some useful clinical applications will eventually arise, it will never achieve the effect size people are hoping for. I hope I’m wrong about that. In terms of non-lifestyle interventions, rapamycin is the safest bet right now for significant, although perhaps not huge, effect size for healthspan and lifespan benefits in people.

Outro

Thanks for your interest in the space and joining us yet again for VitaDAO’s Monthly Longevity Newsletter!

Don’t hesitate to contact us to let us know what you want to see next and give any feedback. We would love to hear from you! Until next time and if you’ve missed this month’s highlights, check them out on YouTube:

Another VitaDAO IP-NFT Transfer Ceremony with Molecule & Jonathan An

https://www.youtube.com/watch?v=JgwC2prEtSM

And a few episodes of Meet The Vitalians | Imagining the future of VitaDAO with key members of the VitaDAO community:

Laurence Ion

https://www.youtube.com/watch?v=gtmaWgnYJ8Q

Sebastian Brunemeier

https://www.youtube.com/watch?v=4JddKx2E32M

Paul Kohlhaas

https://www.youtube.com/watch?v=yEpHpCu2sk8

We can all agree that the age-associated decline in physiological functionality and increased risk of developing diseases is terrible, but what can we do to treat, slow down or even prevent it?
Read more
VitaDAO Newsletter Issue No 8 August 2022
August 8, 2022
Sarah Friday
Awareness
VitaDAO Newsletter Issue No 8 August 2022

Dear Vitalians, happy DeSci-Summer!

Inside this month’s newsletter:

  • Longevity Prize in collaboration with Foresight and Methuselah Foundation
  • Multichain Voting 
  • VitaDAO’s Fourth IP-NFT Ceremony
  • Upcoming Events

DAO News

Longevity Prize

The VitaDAO Longevity Prize is a collaboration with Foresight Institute and Methuselah Foundation to encourage novel approaches and breakthroughs in the realm of longevity research. One key challenge with current prize models is that academics and biotechs only perform experiments when they have funding secured. To combat this issue, our first round of prizes will be given out for hypothesis generation.

These hypotheses will be used to define the second round of larger prizes. The first prize, the “Hypothesis Prize”, will reward individuals who identify an under-appreciated or obscure area of longevity science. The first place proposal will win $20,000, with other submissions receiving prizes as decided by jury votes. All are welcome to apply! Watch a recording of the Opening Ceremony to meet the judges and learn more about the Longevity Prize! 

POPR- Proof of Peer Review

A VitaDAO squad has started a peer-review service. While still early in its conception, they have been focused on creating a decentralized journal that focuses on providing peer-review for projects that need peer-review for publication or funding purposes. 

As the “Uber of peer review,” this journal would build an ecosystem where individuals are compensated for peer review. On Discourse, VDP-53 further describes the intricacies of how VitaDAO would grow its treasury by taking in revenue from POPR, and how VitaDAO would grow longevity knowledge as a result of POPR. If you’re interested in joining the movement, head to VitaDAO’s Discord and join the conversation in the “Peer-Review-Journal” channel. 

Meet the Vitalians

VitaDAO contributor Victoria Forest has been working on the series, “Meet the Vitalians”. It is composed of interviews picking the brains of core contributors & leadership. 

This month's highlight: DAO contributor Sebastian Brunemeier. Head to Youtube to meet Sebastian Brunemeier, and listen to him talk about longevity, the Web3 space, and his experience as a member of VitaDAO’s community. 

Vote with your VITA tokens on Gnosis Chain

Our tech team updated VitaDAO's Snapshot space so that you can now vote with your VITA tokens on Gnosis Chain using the multichain strategy module. Previously, you could only use your VITA token balance on Ethereum mainnet. This change will also allow expanding the voting process to other chains (Polygon, other L2s) in the future. Gnosis Chain, formerly known as xDai chain, is an Ethereum sidechain (L2) that is also known as the "DAO chain" due to its low transaction fees. Click here, for more information on how to acquire VITA on Gnosis Chain and for more information on how to bridge your tokens from Ethereum mainnet to Gnosis Chain. Once you have a VITA balance on Gnosis Chain, simply open VitaDAO’s Snapshot space, connect a MetaMask account, and Snapshot will automatically consider your token balance on both Gnosis Chain and Ethereum mainnet. Note, you can only vote with your token balance as of 12 hours before a proposal has opened for voting. If you acquire tokens during an ongoing vote, they are not counted towards your voting balance.

IP-NFT Transfer Ceremony

On August 2nd, VitaDAO hosted its fourth IP-NFT Transfer Ceremony 🎉. The ceremony celebrated the funding of Jonathan An’s research exploring geroscience-based treatments for periodontitis. Watch a recording of the IP-NFT Transfer Ceremony and learn more about the connection between periodontitis and longevity on VitaDAO’s Youtube channel

Governance

VDP-52 Passed! 

As a result of VDP-52, steward elections will be delayed until a consensus is reached between VDP-36 and VDP-51, with elections being held no later than September 15th. VDP-36 and VDP-51 are proposals written with the intent to improve upon the original steward election process outlined in VDP-19. This extra time allows for consensus to be reached within the VitaDAO community regarding which approach is most beneficial and widely desired within the VitaDAO community. 

Live on Discourse

Find many proposals live on Discourse, VitaDAO’s governance hub for proposals before they are moved on-chain. These proposals are open for engagement, voting, questions, and conversation. Head over to Discourse to check out open governance proposals, peruse live funding proposals, and add to the conversation! 

Community Member Highlight: Sarah Friday

How did you first get involved in web3?

In college, a friend introduced me to blockchain technology and smart contracts. After learning more about use cases for blockchain in healthcare and scientific research, I began to seek out communities and individuals at the forefront of decentralized science. VitaDAO, a community-owned collective funding early-stage longevity research, stood out. In December 2021, I connected with Niklas Rindtorff on Twitter and became a VitaDAO contributor shortly after!

What attracted you to VitaDAO? 

After taking a course on the biology of aging in college, I fell down the longevity rabbit hole. I found that VitaDAO combined two of my side interests: research of lifespan expansion and the intersection of web3 and science. 

Additionally, I was attracted to the community at VitaDAO. VitaDAO is comprised of a unique group of international researchers, longevity enthusiasts, and entrepreneurs. I appreciate that individuals involved in VitaDAO openly embrace change. 

What is your role within VitaDAO? 

I am a Communication and Awareness Working Group contributor. Since December 2021, I have helped author VitaDAO's monthly newsletter. More recently, I have also taken on the role of providing weekly updates in VitaDAO's Discord. I see myself as a newswoman, of sorts, as I help disseminate community announcements and recap past events. 

What has surprised you most about VitaDAO? 

The speed. VitaDAO's framework eliminates the traditional timeline associated with biotech funding. It has been amazing to see what can be accomplished and how quickly new ideas can be implemented when such highly motivated individuals are focused on a central goal. 

VitaDAO in the Media

Molecule hosted its first-ever “DeSci Podcast.” The inaugural podcast focused on VitaDAO’s first year of existence. Listen to Tim Peterson, Laurence Ion, and Sebastian Brunemeier talk about the future of biotech

https://www.youtube.com/watch?v=-z76r78jqno

"The first decentralized community to fund real-world research. We focus on funding longevity research to create therapeutics that target aging”.

Forefront author Esha Bora wrote on DeSci movements and the future of scientific research and progress. In the piece, “The Decentralized Science Movement,” VitaDAO’s token-based ecosystem was highlighted! 

When discussing the role of Web3 in the rise of DeSci, VitaDAO is mentioned in the Investor Times article “DeSci: the Web3-based science movement”. 

Upcoming Events 

August 17th at 7 PM CET- (Recurring bi-weekly on Wednesdays)- Join a VitaDAO onboarding session! Here, you can ask questions about the DAO, meet others interested in the DAO, and find out more about VitaDAO’s working groups. 

August 11th at 1 PM CET- The NUS Yong Loo Lin School of Medicine, together with Professor Brian Kennedy and Professor Andrea Maier are proud to be hosting a Healthy Longevity webinar series. Tune into this virtual series on August 11th to listen to VitaDAO-funded Evandro Fang! 

 Aug 29th-Sept 2nd- Join the ARDD 9th Aging Research and Drug Discovery Meeting. VitaDAO is one of the many sponsors of this event, happening in Copenhagen, Denmark.

Thanks for Reading! 

VitaDAO is defined by an active and diverse community of contributors. To learn more about the community, join our Discord and follow us on Twitter!  

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with monthly VitaDAO updates. Already subscribed? Forward this newsletter to a friend!

Dear Vitalians, happy DeSci-Summer! Inside this month’s newsletter: Longevity Prize in collaboration with Foresight and Methuselah Foundation Multichain Voting VitaDAO’s Fourth IP-NFT Ceremony Upcoming Events
Read more
Announcing "The Longevity Prize"
July 12, 2022
Longevity
Announcing "The Longevity Prize"

VitaDAO is excited to announce The Longevity Prize in collaboration with Foresight Institute.

A series of prizes to honor and accelerate progress in longevity and rejuvenation. 

The longevity ecosystem is growing rapidly. But the problem is vast and we’re running out of time. The longevity prize encourages novel approaches for turning back our aging clocks.

You may be familiar with standard prize models that set a fixed amount to target a specific scientific goal with exact criteria. Those are great!

This prize series is different. Its aim is to generate an avalanche of proposals, experiments, and collaborations on undervalued areas.

This can include smaller bets growing into larger sums, innovating with novel prize voting mechanisms, or even a series of workshops or hackathons to develop promising ideas. 

Common to all these prize experiments is their goal to support a growing longevity ecosystem, connecting those who generate proposals for progress with those who want to help execute them, and drive high-trust collaboration toward solving them. 

We love to collaborate. The first round of $180k in prizes was fundraised through Gitcoin, supported by community members, whose donations were matched by VitaDAO, Vitalik Buterin and Stefan George.Thank you for your generous support! If you have an idea for a prize you’d like to sponsor, we’d love to hear from you!

Together with our judges, we are excited to announce our first round of prizes, the Hypothesis Prize:

  1. Hypothesis Prize to be given for best proposal for undervalued and non-obvious prizes in the longevity space: 

One key problem with the prize model is that academics and biotechs will only perform experiments when they have the money in the bank. That’s why our first round of prizes will be given out for hypothesis-generation to define the second round of larger prizes.

You can achieve a lot by reading the literature – over a century of all the world’s biological knowledge is available to anyone. There are cases where key discoveries are made in the past, but forgotten for long periods of time – only to be rediscovered. 


We would like to hear from you: what is the most promising but under-appreciated area of geroscience and LongBio that we should pursue? Review the literature (and you’re welcome to include your own unpublished data), explain why this area is undervalued, generate a hypothesis for making progress, and propose an experiment to further investigate this approach. The more concrete, e.g. including people, resources, and time required for next steps, the better. 

Proposal length limit: A 1-pager would be great, 3-pages maximum.

Who can apply: No need to be a bench scientist or PI to apply – anyone can submit a proposal. 

Prize incentives: Up to $20k for this round! Finalists will be invited to present their proposal to the judges. Excellent proposals will be moved to the next phase, where they will be eligible for follow-on funding.

Apply: You can find examples of the prizes and information on how to apply on the prize website: https://longevityprize.com/

Prize deadline: End of 2022

We are excited to be supported by an excellent panel of judges:

  • Alexandra Stolzing (Loughborough University)
  • Brian Kennedy (Buck Institute) 
  • Celine Halioua (Loyal)
  • Christine Peterson (Foresight Institute) 
  • Daniel Ives (Shift BioSciences) 
  • David Furman (Buck Institute) 
  • Eleanor Sheekey (Sheekey Science, Cambridge) 
  • Joe Betts-Lacroix (Retro) 
  • Jose Luis Ricon (Rejuverome)
  • Jean-Hebert (Albert Einstein School of Medicine) 
  • Jamie Justice (Wake Forest School of Medicine) 
  • Karl Pfleger (Agingbiotech.info) 
  • Michael Snyder (Stanford University) 
  • Nathan Cheng (On Deck Longevity Biotech)
  • Nir Barzilai (Albert Einstein School of Medicine)
  • Petr Sramek (Longevity Tech Fund) 
  • Reason (FightAging) 
  • Sebastian Brunemeier (ImmuneAGE & Healthspan Capital)
  • Sonia Arrison (100 Plus) 
  • Tim Peterson (WashU, VitaDAO, Healthspan Tech)
  • Tyler Golato (VitaDAO, Molecule)
  • Vadim Gladyshev (Harvard University) 

Questions: Please contact your prize Co-Initiators:

  • VitaDAO: Vincent Weisser, Cat-Thu Nguyen-Huu, Sebastian Brunemeier, Tim Peterson, Tyler Golato,
  • Foresight Institute, a non-profit institute to support ambitious science and technology development in longevity, molecular machines, neurotech, space, and computing. Contact Aaron King, Research Director, to learn more. 

We are announcing The Longevity Prize in collaboration with Foresight.‍ A series of prizes to honor and accelerate progress in longevity and rejuvenation. ‍The longevity prize encourages novel approaches for turning back our aging clocks.
Read more
Longevity Research Newsletter July 2022
July 8, 2022
Maria Marinova & Rhys Anderson
Longevity
Newsletters
Longevity Research Newsletter July 2022

Introduction

Why do we age? Is aging a result of a genetic program encoded within each of our cells, or is it simply a consequence of damage accumulation throughout life? Do we stand a chance in intervening or is it futile and physically impossible to stop the ever increasing entropy? Many theories of aging have been proposed which aim to answer these questions.

One side of the story is the accumulation of damage. This can take many forms. From the wear and tear hypothesis to accumulation of DNA mutations, free radicals, protein crosslinks or aggregates. The other side suggests there are programs in place to regulate cell life and turnover. This could include anything from telomeres getting shorter with each cell division, to cellular senescence perturbing tissue homeostasis and driving inflammation, to your favourite longevity gene, that if altered might impact on lifespan.

This is of course not an exhaustive list. You can see our previous issue for Charles Brenner’s take on why ageing is inextricably linked to development and how a decline in repair capacity is a major contributor to aging. And of course the highlight of this month’s newsletter is our interview with Professor Tom Kirkwood, who formulated the disposable soma theory of aging, which addresses evolutionary trade-offs between investment in reproduction vs maintenance of our somatic cells.

VitaDAO-funded Research Projects

The VitaDAO community have voted, with a 71.22% majority, to fund Repair Biotechnologies - a preclinical-stage biotech company developing a first-in-class universal cell therapy for atherosclerosis (the main cause of cardiovascular disease). The aim will be to engineer macrophages, to express a protein capable of degrading excess cholesterol, which can then be delivered to patients.

https://snapshot.org/#/vote.vitadao.eth/proposal/0x843c0e546ecfdcfb850ba00e88bcfd8c4a1e9204f838342dbbf98a81606a50f6

Longevity Literature Hot Picks

While we’re on the topic of theories of aging, we need to mention last month’s hot pick, which contributes to the discussion.

The relationship between epigenetic age and the hallmarks of ageing in human cells https://www.nature.com/articles/s43587-022-00220-0

The paper bridges together the deterministic epigenetically programmed aging theory and the stochastic “wear and tear” one. Multiple mechanisms are playing out synchronously but independently as seen by the disconnect between the hallmarks of aging and epigenetic clocks. More on this in our previous issue and now back to this month’s picks hot off the press.

Diverse partial reprogramming strategies restore youthful gene expression and transiently suppress cell identity

https://www.sciencedirect.com/science/article/abs/pii/S240547122200223X

Partial reprogramming restored youthful expression in two cell types - adipogenic and mesenchymal stem cells, but it also suppressed somatic identity programs temporarily, as demonstrated by single cell genomics. The study tests multiple subsets and combinations of Yamanaka factors and compares how they restore youthful expression and to what degree they suppress somatic identity.

Lipid metabolism dysfunction induced by age-dependent DNA methylation accelerates aging

https://www.nature.com/articles/s41392-022-00964-6

The changes in the epigenome and cell metabolism interact and contribute to aging. The Elovl2 gene is found to correlate strongly with age and it’s involved in lipid metabolism regulation. If its function is impaired an aging phenotype occurs due to endoplasmic reticulum stress and mitochondrial dysfunction.

Dietary restriction and the transcription factor clock delay eye aging to extend lifespan in Drosophila Melanogaster

https://www.nature.com/articles/s41467-022-30975-4

Visual senescence and altered light perception can negatively affect the circadian rhythm which is associated with decreased lifespan. Dietary restriction reduced the photoreceptor activation, thus amplifying circadian rhythm and mitigated the shortened lifespan phenotype.

Measuring biological age using omics data

https://www.nature.com/articles/s41576-022-00511-7

The review pulls together recent advancements in high-throughput omics and how the data can be used to build reliable aging clocks by harvesting the power of machine learning. This can not only integrate the information from the epigenome, transcriptome, proteome and metabolome but it has also shown the capability to identify novel aging biomarkers.

Molecular mechanisms of exceptional lifespan increase of Drosophila melanogaster with different genotypes after combinations of pro-longevity interventions

https://www.nature.com/articles/s42003-022-03524-4

The combination of multiple pro-longevity strategies indeed lengthens Drosophila lifespan by impacting epigenetics, nutrient sensing, autophagy, immune response, lipid metabolism and cellular respiration. There was a trade off of locomotion for longevity in the flies.

Skin Aging in Long-Lived Naked Mole-Rats is Accompanied by Increased Expression of Longevity-Associated and Tumor Suppressor Genes

https://www.sciencedirect.com/science/article/pii/S0022202X2200402X

While there are a number of similarities between human and naked mole rat (NMR) skin aging such as decrease of epidermal thickness, keratinocyte proliferation, and a decline in the number of Merkel cells, T-cells, and expression levels of dermal collagens, there are some contrasts in the expression of certain longevity-related and tumour-suppressor genes in NMR skin that likely protect it from damage and skin cancer.

Inference of age-associated transcription factor regulatory activity changes in single cells

https://www.nature.com/articles/s43587-022-00233-9

Transcription factors (TFs) are vital for cell function and cell differentiation, which is why it is important to understand their role in the aging process. This study reveals age-associated macrophage dedifferentiation across tissues, with a single cell resolution.

Comparative transcriptomics reveals circadian and pluripotency networks as two pillars of longevity regulation

https://www.sciencedirect.com/science/article/pii/S1550413122001383

Transcriptomic analysis of 26 diverse mammalian species with varying lifespans shows that genes for inflammation and energy metabolism are associated with lower lifespans compared to expression of genes for RNA transport, microtubule organisation and DNA repair correlate with longer lifespans.

Autophagy at the intersection of aging, senescence, and cancer

https://febs.onlinelibrary.wiley.com/doi/10.1002/1878-0261.13269

An interesting review from Narita lab discussing the roles of autophagy in the maintenance of stem cell populations and prevention of cellular senescence, along with how stress-induced senescence might rely upon autophagy. They also describe evidence suggesting autophagy can have a tumour suppressive or promoting effects in early vs late stage tumorigenesis respectively.

The Less We Eat, the Longer We Live: Can Caloric Restriction Help Us Become Centenarians?

https://www.mdpi.com/1422-0067/23/12/6546/htm

Calorie restriction is one of the most studied interventions in longevity research - here the authors review the literature on the metabolic pathways involved and potential for lifespan extension in humans.

And last but not least, an exciting pre-print from Calico:

The complete cell atlas of an aging multicellular organism https://www.biorxiv.org/content/10.1101/2022.06.15.496201v1

Aging in C. elegans is described to be a tightly coordinated process involving mostly metabolic and stress-response genes. The signatures of aging in different cell types were distinctly different with one common factor of decreased energy metabolism.

Clinical Trial Updates

A small clinical trial for rectal cancer observes remission in 100% of patients!

https://www.nejm.org/doi/full/10.1056/NEJMoa2201445

Administering the monoclonal antibody Dostarlimab, an inhibitor of programmed death 1 (PD-1), to patients with a subset of rectal cancer characterised by a DNA mismatch repair deficiency was able to clear all observable cancer in every patient.

News

Saudi Arabia plans to spend $1 billion a year discovering treatments to slow aging

https://www.technologyreview.com/2022/06/07/1053132/saudi-arabia-slow-aging-metformin/

The Hevolution Foundation’s mission is to make ageing healthier by supporting innovation in life sciences and medicine with grants and investments that focus on targeting the biology of ageing itself, rather than specific diseases.

Insilico raise $60M in Series D funding

https://endpts.com/no-humans-allowed-alex-zhavoronkov-adds-robotic-lab-to-his-ai-drug-discovery-play-as-insilico-unveils-60m-raise/

Upcoming conferences

Ending Age Related Diseases

August 11-14th, virtual

https://www.lifespan.io/ending-age-related-diseases-2022/

9th Aging Research and Drug Discovery (ARDD) Meeting

August 29th - September 2nd, Copenhagen

http://www.agingpharma.org/

Longevity Summit Dublin 2022

September 18th-20th, Dublin

https://longevitysummitdublin.com/

Job board

Lab technician or early career scientist at Vincere

https://www.indeed.com/cmp/Vincere-Biosciences,-Inc/

Vincere is working to slow or stop Parkinson's and other age-related diseases. They are hiring a lab tech or early career scientist to work at the bench in Boston, MA running cell culture assays related to mitochondrial pathways.

Postdoctoral fellowships at University of New Mexico School of Medicine, U.S

https://hsc.unm.edu/medicine/education/reo/training-programs/asert/

The McCormick Lab - working on delaying ageing - are an eligible lab for this call and are welcoming people to get in contact!

Postdoctoral Research Fellow in Dr. Adam Antebi’s Lab - Max Planck Institute for Biology of Ageing, Cologne, Germany

https://www.age.mpg.de/career-education/open-positions

A great opportunity for a post-doc who has expertise in fish biology to work on a project to understand ageing in killifish.

Interview with Professor Thomas Kirkwood

Prof. Tom Kirkwood received his PhD from Cambridge University and went on to have a prolific career in gerontology research. Most notably, he formulated the “disposable soma theory of ageing”. Prof. Kirkwood has also made great contributions to the promotion of ageing research, including publishing the popular science books “Time of Our Lives” and “The End of Age: Why Everything About Aging Is Changing”. He was appointed Commander of the Order of the British Empire (CBE) in the 2009 New Year Honours.

What inspired you to enter longevity research?

A mixture of chance and curiosity. I was working on something totally different when a colleague, the distinguished molecular geneticist Robin Holliday, happened to ask for my thoughts on a question concerning replicative senescence. It sounded interesting, we began a productive collaboration, and my curiosity just grew and grew.

How has the field changed since you started?

The field was tiny when I started in the mid-1970s. People had speculated about ageing for a very long time, but labs dedicated to longevity research were then very rare. It would be several years before I got a job that actually included my work on ageing, but I was lucky that my growing interest was tolerated by my then employer. Today, the extent of the network of longevity labs is amazing.

Other than your own, what do you think have been the biggest/important discoveries in the field?

The discovery of replicative senescence in the 1960s was a milestone, though it took many years to reach our present understanding. So many big discoveries have been made about molecular and cellular aspects of ageing and longevity. Among the biggest are the role of signalling pathways, genetics and epigenetics, and the significance of a systems approach.

What advice would you give to people currently working in longevity research?

Enjoy the challenge – this is an exciting time to be in the field. Ageing is complex – respect the complexity. A Nobel Prize-winning physicist once said “one should neither seek nor avoid complexity in addressing the problem at hand”. I’ve found this advice very helpful. Make time to read and think widely. Unexpected connections can arise that lead to breakthroughs.

Which aspect of longevity research do you think requires more attention?

Addressing complexity via systems approaches is certainly one. Ageing plays out via multiple mechanisms at multiple levels. Personally I’m very keen on using the power of evolutionary analysis to connect the big Why? and How? questions. Also, a striking feature of ageing is its inherent variability – this needs some more serious attention.

Is ageing a disease?

For me, ageing isn’t a disease in the sense I would usually apply to this term. It’s a normal process. But by its very nature it involves the generation of molecular and cellular abnormality. This feeds into multiple kinds of dysfunction. When a particular kind of dysfunction – which may have causes additional to ageing – passes a threshold recognised by clinicians, it gets a diagnosis of disease.

You are most well-known for proposing the Disposable Soma Theory of Ageing? Could you summarise this in a couple of sentences?

The disposable soma theory (DST) proposes an answer to the question: how much of its energy should an organism invest in the long-term maintenance of its somatic tissues? The answer is that, whereas it is important for the germ-line to be immortal, the soma needs only to be maintained well enough to remain in sound condition for as long as the individual might reasonably expect to still be alive. In nature, most deaths result from external risks, so the deleterious consequences of limited maintenance don’t count much in the evolutionary struggle for survival.

How well do you think the disposable soma theory has aged?

The essential predictions of DST are (i) somatic cells should be less well protected than germ cells, and (ii) species longevity should be regulated by selection to raise or lower the level of somatic maintenance in relation to the hazard rates in the species’ ecological niche. Both of these predictions are very well supported.

There are interesting questions about how the resources freed up by limiting the investment in maintenance should be invested to maximise Darwinian fitness, for example, by enhancing reproduction and/or growth rate. In other words, should we see trade-offs between longevity and reproduction (or other life history traits)? Much evidence supports the existence of such trade-offs, but exceptions have been found. These, together with growing data on the diversity of ageing across the tree of life, have prompted some interesting developments. These are important for our understanding of longevity science but the DST has the flexibility to accommodate them. So to answer the question, I think the DST has aged well. Like all of us, it’s been challenged by new experience and become older and wiser along the way.

Are there any criticisms (such as women being longer lived compared to men, or longevity benefits of caloric restriction) which you think hold merit / are there amendments you would make to the theory now?

Critical challenge of a theory is always welcome, but neither of these is a serious problem.

Regarding sex differences, females in many species make more direct investments in reproduction than males, so it might seem strange that they are appear also to be better maintained. But, one must not overlook the big investments made by males in reproductive competition, which can be very costly. Since the integrity of the female soma is essential to nurture the next generation, it might indeed be less disposable than the male soma.

Regarding CR, how can less nutrition cause longer lives? One suggestion, made by others, is that famine may cause an animal to suspend reproduction, which might allow maintenance to be temporarily boosted to preserve integrity for when the famine is over. We have shown this to be at least theoretically possible, but other possibilities exist. The DST provides a framework within such questions can be considered, but they don’t constitute critical predictions of the theory.

The DST framework, based as it is on the principle of evolutionary optimisation of resource allocation strategies, has relevance for very diverse organisms, as others as well as I have recognised. These extend well beyond the case originally in mind of a multicellular animal with a strict distinction between germline and soma. In this sense, the theory offers exciting opportunities for further adaptation or amendment.

Outro

Thanks for sticking with us for another issue of VitaDAO’s Monthly Longevity Newsletter!

As always, we would love to hear your feedback and suggestions for content you want to see. For now we will leave you with our highlights for the past month and we hope to see you again for our next issue!

Meet The Vitalians | Imagining the future of VitaDAO | With Tim Peterson

https://www.youtube.com/watch?v=O2xZZR1sydA

VitaDAO IP-NFT Transfer Ceremony with Molecule & Evandro Fang

https://www.youtube.com/watch?v=ZcUXGzgm8_k

MenoAGE - Community Call

https://www.youtube.com/watch?v=KH2qRMm-QY0

Why do we age? Is aging a result of a genetic program encoded within each of our cells, or is it simply a consequence of damage accumulation throughout life?
Read more
VitaDAO Newsletter Issue No 7 July 2022
July 7, 2022
Sarah Friday
Awareness
Newsletters
VitaDAO Newsletter Issue No 7 July 2022

June was a month of celebration for VitaDAO! This June, VitaDAO celebrated its first birthday, held a ceremony for the transfer of VitaDAO’s third IP-NFT, and completed another Gitcoin Grant Round. In this newsletter, learn about VitaDAO’s involvement in various community conferences and read about the DAO’s active and past voting proposals.

Community News

IP-NFT Transfer Ceremony

VitaDAO hosted an IP-NFT Transfer Ceremony to commemorate VitaDAO’s third longevity research project funded via IP-NFT🎉. This transfer celebrated the funding of the Evandro Fang lab. The Fang lab is working to identify novel mitophagy inducers for healthy brain aging. If you missed the ceremony, you can watch a video of the transfer to the Evandro Fang project. Interested in learning more about the Fang project? Find more information on Snapshot.

Celebrate Good Times

June 18th marked one year since VitaDAO’s Gnosis Token Auction🎊. In just one year, VitaDAO funded over 10 longevity research projects, sourced over 200 projects, grew to over 5000 community members, and garnered over 1000 token holders. Additionally, over the year, VitaDAO launched the Longevity Prize via Gitcoin grants, funded over 40 Vita Fellows with over $45k, and hosted its 1st VitaDAO Crypto meets Longevity Symposium. Here’s to another year of funding longevity research, growing a vibrant community, and continuing to revolutionize scientific funding!🥳

Early Bird Gets The POAP

This month, VitaDAO released a special edition POAP for VitaDAO genesis auction participants. POAPs are digital collectibles that serve to commemorate contributions to web3 events. If you took part in VitaDAO’s genesis auction, you can claim your POAP!

Meet the Vitalians

VitaDAO contributor Victoria Forest has been working to re-ignite the series, “Meet the Vitalians.” This video series is composed of interviews picking the brains of core contributors to the DAO. This month, head to YouTube to meet DAO member, Tim Peterson. In this chat, learn about Tim’s thoughts on longevity, the web3 space, the VitaDAO community, decentralization, and more! If you missed previous “Meet the Vitalians” episodes, catch up on DAO members Todd White, Theo Beutel, and Audie Sheridan.

Longevity Newsletter

VitaDAO released its second Monthly Longevity Research Newsletter! 🔥 Inside this newest edition, Authors Maria Marinova and Rhys Anderson highlight hot longevity literature, longevity clinic trial updates, and interview Charles Brenner. Find June’s Longevity Research Newsletter, here.

Another Successful Gitcoin Round

VitaDAO completed Gitcoin Grant Round 14! 👩‍🚀 VitaDAO supports public goods by organizing Gitcoin rounds where projects have the opportunity to receive quadratically matched funding. In this most recent round, VitaDAO’s Gitcoin round had over 1500 donors and VitaDAO’s Longevity Prize Project reached the max matching pool amount of $25,000. 🚀

Crypto Meets Longevity featuring Qiao Wang

Max Unfried chatted with AllianceDAO core contributor Qiao Wang. You don’t want to miss their conversation discussing potential future roles of crypto in biotech, barriers to decentralized science, and more!

A Conversation with Brian Kennedy

Max Unfried and Eleanor Sheekey hosted Professor Brian Kennedy to discuss the decentralization of healthcare through geroscience. Professor Brian Kennedy is an internationally recognized researcher in the aging field and the Director at the Centre for Healthy Longevity at NUS Medicine 🥼. In this presentation, Professor Brian Kennedy talks about the need for longevity research and provides an overview on the basics of how longevity research is conducted!

Ch-ch-ch-ch-changes

VitaDAO passed two proposals on Snapshot in June! You can follow other proposals and add to the conversation before votes are added to Snapshot on the VitaDAO’s Discourse. There are currently multiple proposals being discussed on Discourse, including:

  • VDP-26.1: Dealflow Structure & Incentives
  • VDP-29: Exosomes for Treating Systemic Inflammation
  • VDP-36: Stewardship Process Amendment #1
  • VDP-37: VitaDAO Governance Amendment #2
  • VDP-44: Dispute Resolution Process
  • VDP-47: Immunis Biomedical
  • VDP-48: Ikaria Therapeutics assessment
  • VDP-49: MenoAge — A Glycomics-based Blood Biomarker for Female Aging

VDP-45 Passed!

VDP-45 (Decentralized Tech Transfer) will help to create a decentralized tech transfer network that can connect VitaDAO directly to researchers. Currently, tech transfer offices act as a major bottleneck in advancing research. In offering a more decentralized setup to connect researchers with VitaDAO, VDP-45 has the potential to revolutionize research funding.

VDP-46 Passed!

VDP-46 (Repair Biotechnologies) proposal has successfully passed! Repair Technologies is a preclinical-stage biotech company developing a first-in-class universal cell therapy for atherosclerosis🫀. They aim to solve macrophage dysfunction with novel allogeneic cell therapy, “Cholesterol-Degrading Platform.” To learn more about this proposal, watch a Community Call with Repair Bio or read the full proposal.

VitaDAO in the Wild

This month, VitaDAO contributors represented VitaDAO at conferences across the world:

- VitaDAO Contributors Max Unfried and Jane Chuprin attended a Gordon Research Conference focusing on Systems Aging. At this conference, VitaDAO sponsored the “Best Poster Prizes” from the many posters focused on systemic processes, omics approaches, and biomarkers in aging.

- VitaDAO Steward Theo Beutel held a panel at DAO NYC on Impact DAOs.

- Theo Beutel also spoke at Funding the Commons about a user-centric approach to DeSci and Tokenomics.

- VitaDAO hosted a Longevity x Web3 dinner event and talk at the Collision Conference. This event brought together VitaDAO contributors and individuals interested in longevity from across the world.

What’s Going DAOn

July 28 at 3PM CET- Listen in on a VitaDAO Community Call. Recordings of past community calls can be found on VitaDAO’s Youtube.

July 28th at 10AM CET- Take part in a VitaDAO IP-NFT Fractionalization Workshop.

Aug 29th-Sept 2nd- Join the ARDD 9th Aging Research and Drug Discovery Meeting. VitaDAO is one of the many sponsors of this event, happening in Copenhagen, Denmark.

DAOn’t Forget…

The Operations Working group has been hard at work improving VitaDAO onboarding. This ongoing effort to improve VitaDAO onboarding includes a more streamlined onboarding process. Head over to the VitaDAO Discord, introduce yourself and follow the instructions on the “👋ㆍstart-here” channel.

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with monthly VitaDAO updates. Already subscribed? Forward this newsletter to a pal interested in web3, science, or longevity! 💛

June was a month of celebration for VitaDAO! This June, VitaDAO celebrated its first birthday, held a ceremony for the transfer of VitaDAO's third IP-NFT, and completed another Gitcoin Grant Round.
Read more
Longevity Research Newsletter June 2022
June 13, 2022
Maria Marinova & Rhys Anderson
Longevity
Awareness
Newsletters
Science
Longevity Research Newsletter June 2022

Welcome back to VitaDAO’s monthly longevity research newsletter — we hope you enjoyed our maiden issue!

The longevity field remains one of the most controversial areas of science. There are a number of common objections which arise to the concept of trying to understand ageing and increasing lifespan, such as: overpopulation, wealth inequality, long-lived dictators, slower progress, longer decrepitude, unsustainable pensions, being against the “natural order” and many others. For a great discussion of all these points with interesting counter-arguments, check out: https://agingbiotech.info/objections/.

Another problem is that the field has often been infiltrated with quacks, snake oil and sensationalist headlines — often claiming to have discovered the “elixir of life” or the “fountain of youth”. Sadly, this issue will only get exacerbated as interest in ageing research grows and gains more popularity in the public eye. As longevity gets more “trendy” and becomes more of a buzzword even greater caution must be taken with anti-aging pills salesmen.

It is therefore important that the longevity space doesn’t become an echo chamber, but rather an open and transparent community which welcomes intellectual discourse. That’s why this month we reached out to Prof. Charles Brenner, a prolific critic of numerous ideas within the longevity field, for his take on things. He kindly provided us with some important words of caution as well as interesting thoughts on the inextricable link between development and ageing. Prof. Brenner also participated in a no-holds-barred debate with Prof. Aubrey de Grey (on the Let’s Talk Longevity Podcast — featured below) on the idea of Longevity Escape Velocity and how near/realistic it is.

At VitaDAO we are devoted to supporting projects with strong scientific integrity which aim to tackle age-related diseases with the hope of increasing healthspan and genuinely improving peoples’ lives.

VitaDAO-funded Research Projects

This month VitaDAO has funded a 4 year doctoral research program to probe DNA quadruplexes in age-related proteostasis

https://snapshot.org/#/vote.vitadao.eth/proposal/0x9b40b1e3ae9a0d7510502dad67c299bd720d8ef4727e8f54c95d3becebdff24c

Prof. Tim Peterson has been awarded 231,000 USD to design new drugs to build upon chemistry which he discovered can act as a “cellular soap” to simultaneously target both pathogens and senescent cells:

https://snapshot.org/#/vote.vitadao.eth/proposal/0xa419160d43e53c659936d98d4b9339231a3b1586b8dc84fab2f3ffd30b9a3aaf

Longevity Literature Hot Picks

Once again, the abundance of great longevity research that was published this month has been hard to keep up with, but here are some of our favourite papers to keep you up-to-date with the field.

Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17

https://www.nature.com/articles/s41586-022-04722-0

Infusing cerebrospinal fluid from young mice into the brains of aged mice was shown to boost oligodendrocyte progenitor cell (OPC) proliferation and differentiation in the aged hippocampus and improve memory. They identified fibroblast growth factor 17 (Fgf17) as a protein in the young serum which confers these beneficial effects by activating the transcription factor — serum response factor (SRF).

Senescence-induced endothelial phenotypes underpin immune-mediated senescence surveillance

http://genesdev.cshlp.org/content/early/2022/05/25/gad.349585.122.long

Senescence surveillance mechanisms allow for the recruitment of immune cells to specifically remove senescent cells. Here the authors show that senescent cells induce local endothelium to become an organising centre for the adaptive immune response during senescence surveillance.

Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice

https://www.science.org/doi/10.1126/science.abk0297

Caloric restriction has been observed to increase healthspan/lifespan in a number of model organisms. However, trying to unravel the relative contribution of calorie reduction versus fasting proves difficult as calorie restricted animals tend to consume their food quicker and thus spend a longer time fasting. Here the authors show that calorie restriction increases lifespan more when there is a fasting period of 12 hours as opposed to feeding regularly throughout the day. They also observed a further increase in lifespan if the food was restricted to the time corresponding to the active phase of their circadian rhythm, although further research is required to determine if this difference is due to altered sleeping patterns in mice from the opposing feeding schedule.

Calorie restriction and calorie dilution have different impacts on body fat, metabolism, behavior, and hypothalamic gene expression

https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00608-8

Another consideration to understanding the role of diet on lifespan is discerning the relative effects that the food content itself versus the process of ingestion has on the ageing process. Here the authors compare calorie restriction to calorie dilution (ad lib diet with indigestible components) and find that hunger signalling is a key process mediating the benefits of CR.

Dietary intervention improves health metrics and life expectancy of the genetically obese Titan mouse

https://www.nature.com/articles/s42003-022-03339-3

The Titan mouse line has previously been thought of as a model for healthy obesity, but here it is shown that they have numerous metabolic dysfunctions and are short lived. However, switching to a energy-reduced feed was able to restore some of these dysfunctions and significantly increase their lifespan.

The importance of aging in cancer research

https://www.nature.com/articles/s43587-022-00231-x

An interesting editorial highlighting that although cancer is generally an age-associated disease, most animal research is performed in relatively young animals and in human clinical trials, patients over the age of 75 years old are under-represented.

Fecal microbiota transfer between young and aged mice reverses hallmarks of the aging gut, eye, and brain

https://microbiomejournal.biomedcentral.com/articles/10.1186/s40168-022-01243-w

Fecal microbiota transplants, or stool transplants, have been gaining traction in recent years. In this paper, the authors show that transplanting intestinal microbiota from old to young mice can result in an acceleration of both age-associated central nervous system and retinal inflammation, whereas transplanting microbiota from young to old can attenuate these effects.

The relationship between epigenetic age and the hallmarks of ageing in human cells

https://www.nature.com/articles/s43587-022-00220-0

Epigenetic clock scores have been linked to several age-related processes and have shown accuracy as mortality predictors, however further research has been needed to understand the underlying mechanisms. Here the authors show that epigenetic ageing is associated with stem cell composition, mitochondrial activity and nutrient sensing, however independent from genomic instability, telomere attrition and cellular senescence.

Clinical Trial Updates

Curcumin and Function in Older Adults (SPICE)

https://clinicaltrials.gov/ct2/show/results/NCT03085680?recrs=e&rslt=With&type=Intr&cond=Aging&rfpd_s=01%2F01%2F2022&rfpd_e=05%2F24%2F2022&draw=2&rank=4

Resveratrol and Exercise to Treat Functional Limitations in Late Life

https://clinicaltrials.gov/ct2/show/results/NCT02523274?recrs=e&rslt=With&type=Intr&cond=Aging&rfpd_s=01%2F01%2F2022&rfpd_e=05%2F24%2F2022&draw=2&rank=1

Dementia Risk and Dynamic Response to Exercise (DYNAMIC)

https://clinicaltrials.gov/ct2/show/results/NCT04009629?recrs=e&rslt=With&type=Intr&cond=Aging&rfpd_s=01%2F01%2F2022&rfpd_e=05%2F24%2F2022&draw=2&rank=2

News

Oviva announces $11.5M in Seed Financing to improve ovarian health and extend female healthspan through therapeutic innovation

https://www.ovivatx.com/news-and-press/oviva-announces-11-5m-in-seed-financing-and-exclusive-license-with-mgh

Upcoming conferences

4th Annual Longevity Therapeutics Summit

28–30th June 2022, San Francisco, USA

https://longevity-therapeutics.com/

5th International Symposium on Neurobiology & Neuroendocrinology of Aging

15–19th July 2022, Bergenz, Austria

http://www.neurobiology-and-neuroendocrinology-of-aging.org/

5th Annual Ending Age-Related Diseases Conference

11–14th August 2022, virtual meeting

https://www.lifespan.io/ending-age-related-diseases-2022/

9th Aging Research and Drug Discovery Meeting (ARDD)

29th August — 2nd September 2022, Copenhagen, Denmark

http://www.agingpharma.org/

Funding Opportunities

Merck Grants to Stimulate Innovative Research

Drug discovery — 3 grants comprising up to 500,000 €/year for 3 years with the option of extension for applicants with an interest in approaches interfering with the molecular hallmarks of ageing to promote healthy longevity.

https://www.merckgroup.com/en/research/open-innovation/research-grants.html

The San Diego Nathan Shock Center (SD-NSC) Pilot Grant program

Seeking projects that explore the heterogeneity of aging.

Proposals are due by August 24 2022.

https://www.salk.edu/science/research-centers/san-diego-nathan-shock-center/pilot-grant-program/

Job board

Open Research Assistant position in Manning Lab — Harvard University, US

https://www.hsph.harvard.edu/molecular-metabolism/academic-position-openings/

To explore how the PI3K-mTOR signaling network is altered in cancer, diabetes, and aging, and how targeting components of the signaling network might lead to new insights into how to treat or prevent these conditions

Post-doc position available in Alan Attie’s Lab — University of Wisconsin-Madison, US

To work on transcription factors that affect beta-cell function and diabetes susceptibility — learning genetics/genomics and beta-cell biology. Apply to: adattie@wisc.edu

PhD Studentship in Kristina Kirschner’s Lab — Beatson Institute, University of Glasgow, UK

Working on senescence heterogeneity and liver cancer

https://www.beatson.gla.ac.uk/Education/Studentships/

Senior Post-doc position in Gavin Richardson’s Lab — Newcastle University, UK

To investigate senescence and cardiovascular disease.

https://jobs.ncl.ac.uk/job/Newcastle-Research-Associate/795078901/

Five Group Leaders Positions — University of Coimbra, Portugal

The Multidisciplinary Institute of Ageing will soon be opening for applications for group leader positions in the biology of ageing. Watch this space: www.uc.pt/mia

Postdoctoral and technician positions in the Lamming Lab — University of Wisconsin-Madison, US

To investigate how what, when, and how much we eat regulates healthy aging using mouse models: https://lamminglab.medicine.wisc.edu

Head of Engineering (co-founder), Head of Biosimulation (co-founder), Product Manager, CTO, Backend, Frontend, Scientific Developer and multiple other roles at Formic Labs: https://apply.workable.com/formic-labs/

Formic Labs is building an AWS-inspired platform for life sciences R&D that alleviates many of the bottlenecks to integrating knowledge.

Bioinformatician — Teal Omics

Teal Omics is an aging company with a mission to transform healthcare by developing a deep, personalized, and data-driven understanding of age-related diseases, based on research by the Wyss-Coray lab at Stanford University. Contact: alex@tealomics.com

Lab tech or early career scientist — Vincere, Boston, MA, US

https://www.indeed.com/cmp/Vincere-Biosciences,-Inc/

Vincere runs cell culture assays related to mitochondrial pathways. Experience with cell culture is a must, and experience with mitochondrial readouts would be a plus.

Debate: Aubrey de Grey vs Charles Brenner — Longevity Escape Velocity

The gloves were off in this eagerly anticipated debate between Aubrey de Grey and Charles Brenner, on the topic of how realistic/near we are from longevity escape velocity (LEV) — the idea that if the rate of lifespan expansion from medical advancements outpaces the actual passage of time, that we would achieve biological immortality. Just in case you had any doubts about which of the debaters thought we are nearing LEV, here are a few snippets to whet your appetite:

Brenner: “You don’t have any evidence that if you can remove these types of damage that any animal with longer”

DeGrey: “I’m saying we have a 50:50 chance of reaching longevity escape velocity in 15 years from now”……………”

Brenner: “and I’m saying you don’t”

Brenner: “You don’t consider this fantastical?”

Brenner: “Your approaches are so naive”

Check it out here:

S01 E01 — On Longevity Escape Velocity with Aubrey de Grey & Charles Brenner

https://rss.com/podcasts/letstalklongevity/480365/

Interview with Charles Brenner

Charles Brenner received his PhD from Stanford University and has made several important contributions to biochemistry and molecular biology — including the discovery of nicotinamide riboside (NR) as a nicotinamide adenine dinucleotide (NAD) precursor vitamin. He is currently a professor and department chair at City of Hope and also known for his public criticism of many aspects within the ageing field.

Ageing rates and lifespans vary hugely within the animal kingdom — what are your thoughts on trying to understand the mechanisms underlying these differences in hope of translating this knowledge to increasing human healthspan/lifespan?

Different animals reach reproductive maturity at different rates. In large part, animals that take a long time to reach reproductive maturity are longer lived. For sure, there’s discoverable knowledge in the rates of animal development.

Which of the current theories of ageing do you think are the most convincing?

To my mind, aging starts at fertilization and is inextricably linked to development. The fact that the most powerful monogenic longevity genes are loss of function alleles of genes conserved for growth is telling us a powerful lesson. In fact, they are not conserved as longevity genes because their normal functions, which are also essential for reproduction, are growth. It’s also quite evident that all vertebrates exhibit a loss of repair capacity after they are reproductively mature. I don’t have much use for terms like the information theory of aging or the seven types of damage that constitute aging.

Do you think trying to understand the mechanisms driving ageing, with a view to targeting all age-related diseases simultaneously, is a viable parallel strategy to researching these diseases individually? And do you have concerns about the scientific rigour about current attempts at this approach?

I find it astonishing that some investors and newcomers have been brought into this field who have been swayed into thinking that aging is a simply correctible engineering flaw and not a basic part of everyone’s life course. If it weren’t for aging, a fertilized egg could not develop into a born animal, and there would be no growth, puberty or maturation. The most powerful genes ever found that control aging are involved in all of these processes.

If the goal is to help more people lead healthier lives, I’m all for that. The pitch is usually much more extravagant though.

I’m not going to make a blanket critique of approaches but I have been quoted as saying that the SENS approach is structurally unsuitable to make significant progress; in vivo reprogramming is going to be much harder that some people made it out to be; and CRISPR doesn’t change very much because there isn’t a dominantly acting longevity gene that everyone needs more copies of.

Do you think ageing is treatable?

You mean do I think that we can prevent post-reproductive decline or do I think we can age better? I think we can age better. I don’t think that we can prevent post-reproductive decline but it is surely possible to stay in good form for a long time.

What do you think is the most promising longevity intervention?

High physical and mental activity is the single most important intervention. There’s nothing even close to that.

If a prophylactic drug were shown to delay the onset of numerous/all age-related diseases — would you support this? And how would you define/characterise this in terms of FDA disease indications?

Sure, why not? I’m sure you know that it would have to be approved for a disease or condition. If its mechanism of action were such that it delays the onset of other diseases, it would likely be used off label. This is already the case for drugs that have not been shown to delay the onset of age-related conditions in healthy older people. Here I’m referring to rapamycin and metformin.

I also do not think there’s a nefarious plot of biotech or pharma to not develop anti-aging drugs. I think the incentives are already present in our system for people to discover and test important new molecules and for the most part, drugs are approved when the safety and efficacy data support their approval.

What do you think have been the biggest/important discoveries in the field?

I think more people should read the work of Michael Rose on the polygenic nature of longevity and the fact that longevity has generally not been under selective pressure in animals. It is out of this work, that I realized that longevity is an emergent property of other key measures of fitness such as the ability to obtain food, avoid predation, attract a mate, reproduce and take care of young ones. Animal gene sets can then move toward longevity when mothers and fathers are able to reproduce multiple times but it’s not like there is a magical dominantly acting longevity gene. It’s more like nearly every gene has to promote the function of all the organs to do all the things over time.

What are your thoughts on the concept of “biological age” and what do you think the most accurate way to determine this is?

There’s something to it but the kits have zero value to consumers. Subjectively, I agree with the statement “you’re as young as you feel” and in the context of something like dating or running a mile, a 30, 40, 50, 60 or 70 year old person can perform more or less youthfully than their chronological age.

There are also DNA methylation marks that track aging and have some value in predicting future mortality. But we’ve recently read that the GrimAge score goes up during infection and then goes down as the infection is resolved, so I have a concern that some of what is being measured is inflammation. The whole biological age program is a research project to obtain biomarkers that could potentially be used in interventional trials. But while you are trying to figure out if a lower GrimAge score, for example, means that a person is healthier and you don’t yet know that, you can’t score the result of an intervention simply based on their GrimAge score going down. I have publicly stated that we know enough about growth hormone to consider it a pro-aging intervention such that for GrimAge to go down in the TRIIM trial suggests that GrimAge is not measuring what we think it is measuring.

What mistakes do you think the longevity field has made?

Oh gosh, where to begin? Longevity has been a non-evidence based and unfulfillable promise since time immemorial full of potions, vacation spots, magic pills and requests for funding.

Although there is real science to aging, there are individuals in the field who represent themselves as scientists, who are really just promoters. They are actually pretty easy to spot because their ideas are not falsifiable. Their talks, books and appearances do not teach critical thinking. Instead, they are longevity gurus who convince people that they have found the path. These individuals have outsized influence particularly in public spheres, which has seriously damaged the field and I categorically reject the notion that they have helped the field by bringing in money. The consumers and investors attracted by gurus are being damaged and the field of aging science is damaged by hypesters. Hype hurts.

What would you do differently to the approaches currently taken? / What changes would you like to see within the field?

I’m pretty clear that we have to shun longevity gurus. We need to realize we are living in the real world in which we haven’t identified dominantly acting longevity genes, Yamanaka factors are oncogenic and teratogenic, and our gene set has been selected for us to reproduce and provide care to our offspring but has never been under selective pressure to maintain our fitness into a tenth decade. I plan to test some approaches to support repair capacity as we age — I think that NR may have activity in this respect.

What advice would you give to people currently working in longevity research?

Learn and respect developmental biology. Make sure that lab staff are blinded to treatment. Learn from people with falsifiable hypotheses. Steer clear from influencers and promotional review articles.

Outro

Thanks for reading our second issue of VitaDAO’s Monthly Longevity Newsletter!

Once again, if there is anything you would like us to feature in future issues, please get in contact.

Next month we will be highlighting some of the prominent theories of ageing and featuring an interview with Professor Emeritus Thomas Kirkwood — formulator of the Disposable Soma Theory of Ageing — not to be missed!

If you are keen to see what else has been going on in the VitaDAO community, we have provided links below to some of our highlights from the past month — enjoy and we look forward to seeing you next month!
Subscribe here for more!

The Science of Biostasis and Cryopreservation with Kai Micah Mills & Dr. Emil Kendziorra

https://www.youtube.com/watch?v=X9mH1punHx4

Applied Longevity Medicine with Prof. Evelyne Bischof, Prof. Andrea B. Maier and Weronika Prusisz

https://www.youtube.com/watch?v=uGvF3Q-IJf0

Pay for success x IP NFT Pilot — A New Financial Model for Repurposing Off Patent Rapamycin

https://www.youtube.com/watch?v=1j4zMH-4vqE

Bridging IP into Web3 with IP-NFTs

https://www.youtube.com/watch?v=TjUuC9lL_i4

Crypto Meets Longevity with Qiao Wang

https://www.youtube.com/watch?v=BhHeBCM1lbU

Decentralising Healthcare through Geroscience with Prof. Brian Kennedy

https://www.youtube.com/watch?v=VHlKFXJ-lgA

Last but not least, we invite you to watch the full recording of DeSci.Berlin I Advancing Scientific Collaboration, or find the individual talks of some incredible speakers at Molecule’s YouTube channel

https://www.youtube.com/watch?v=sMgNQakX0QA&t=10449s

Welcome back to VitaDAO’s monthly longevity research newsletter — we hope you enjoyed our maiden issue!
Read more
VitaDAO Newsletter Issue No 6 June 2022
June 6, 2022
Sarah Friday
Awareness
Newsletters
VitaDAO Newsletter Issue No 6 June 2022

They say April showers bring May flowers. Instead of flowers, we hope we rose to the occasion with June’s newsletter to show how VitaDAO flourished this May. Inside this month’s newsletter, learn about VitaDAO’s time at DeSci Berlin, catch up on last month’s panel discussions, meet DAO member Max Unfried, and learn about the DAO’s active and past voting proposals.

Community News

DeSci Berlin: The Intersection of Web3 and Science

VitaDAO assisted in running a two-day Decentralized Science conference in Berlin. At this conference, participants learned about the principles of decentralized web-native collectives and their use to coordinate, fund, and execute scientific research🧪. Sad you missed the conference? No need to have FOMO! There are livestream recordings of both day one of the conference and day two of the conference.

DeSci.Berlin: Livestream Day

ANOTHER Newsletter?!

🚨There is another newsletter on the block🚨. This month, VitaDAO launched the first Monthly Longevity Research Newsletter! Authors Maria Marinova and Rhys Anderson highlight hot longevity research papers, upcoming scientific conferences, and job openings. Find May’s Longevity Research Newsletter, here.

The Science of Biostasis and Cryopreservation: A Panel Discussion

Watch a recording of moderator Max Unfried, Kai Micah Mills, and Dr. Emil Kendziorra discussing how cryopreservation fits into the longevity field, examples of cryopreservation in nature, and more!

On Applied Longevity Medicine: A VitaDAO Panel Discussion

In this VitaDAO Panel Discussion, moderator Max Unfried talks with professor Evelyne Bischof, professor Andrea B. Maier, and Weronika Prusisz about the applications of longevity medicine. You don’t want to miss their conversation on biological age testing, confronting the food industry🥗, and optimizing lifestyle over performance.

VitaDAO Panel Discussion: Applied Longevity Medicine

Vote For a POAP

If you vote, claim your POAP! On the majority of recent Snapshot proposals, one can claim a POAP after voting on Snapshot. POAPs are digital collectibles that help record participation in VitaDAO’s votes. Claiming on Snapshot is free, gasless⛽️, and fun!

Longevity Hackers NFT

As a result of VDP-39, VitaDAO is funding up to 100,000 USD to support a documentary by Filmmaker Michal Siewierski. This documentary will feature the longevity eco-system, and subsequently highlight VitaDAO. Find a link to the NFT representing this partnership, here!

Hot Off the Press: Japanese Explainer

VitaDAO published an explainer on VitaDAO basics for our Japanese audience! We are excited about the opportunity to continue to expand VitaDAO’s reach in Asia. Take a peek at the medium article, here.

Ch-ch-ch-ch-changes

Below, find summaries of May’s four passed proposals. More in-depth descriptions can be found on Snapshot! As always, you can follow proposals and add to the conversation before votes are added to Snapshot on the VitaDAO’s Discourse. There are currently multiple proposals being discussed on Discourse, including VDP-46 (Repair Biotechnologies), VDP-44 (Dispute Resolution Process), VDP-45 (Decentralized Tech Transfer), VDP-29 (Exosomes for treating systemic inflammation), VDP-36 (Stewardship Process Amendment #1), VDP-37 (VitaDAO Governance Amendment #2), and VDP-26.1 (Dealflow structure & incentives).

VDP-43 Passed!

VDP-43 initiates a partnership with BanklessDAO. As a result of this proposal, VitaDAO and BanklessDAO will token swap $25,000 worth of $VITA for $25,000 of $BANK taken at the 30-day moving average. VitaDAO is excited about this partnership as it means further treasury diversification and increased DAO awareness! 🤝

VDP-40 Passed!

This proposal allocates $231,000 to Dr. Tim Peterson’s project to develop a cellular soap to simultaneously block both pathogens and senescent cells. It has been appreciated that pathogens age humans by driving cell senescence. This research project offers a promising strategy to thwart pathogens and different aging pathologies.

VDP-42 Passed!

VDP-42 confirms an additional $580,000 arm’s-length loan from VitaDAO to VHF, secured against the underlying equity in companies VHF in turn funds, and a $100,000 donation to VHF to support VHF and VitaDAO’s aligned non-profit initiatives. VitaDAO, through the secured loan agreement for $580,000, will help VHF fund 10 projects within the $50–100K range.

VDP-22 Passed!

VDP-22 provides 50,000 USD of funding to a 4-year doctoral research program at the Institute of Chemical Biology, Imperial College London. The program plans on probing DNA quadruplexes in age-related proteostasis. In exchange for supporting this doctoral program, VitaDAO expects to have access to IP licensing rights (potentially related to therapeutic compounds or biologics) derived from the research program via an IP-NFT via Molecule.

Community Member Highlight: Max Unfried

What is your role at VitaDAO?

​​I’m part of the Longevity and Awareness WG — and help with project sourcing and evaluation, and science communication by hosting events to educate the crypto community on longevity and aging research.

How did you first get involved in web3?

I’ve been following crypto since 2014 from the side-lines after a friend told me about Ethereum, but got seriously involved in some projects from 2019 onwards.

What was it that attracted you to VitaDAO?

The opportunity to be part of a parallel system to fund and promote rejuvenation research.

What excites you most about VitaDAO’s future?

VitaDAO is still in its early innings — but I am very excited for the first community funded and owned rejuvenation therapeutic that makes it to market.

What is something you believe in now that you didn’t ten years ago?

10 years ago I was under the impression that science and technology were advancing rapidly. Nowadays, I’m under the impression that meaningful science and technology are not advancing fast enough — especially in the world of atoms — as too many resources are spent on projects that won’t make much impact even if successful.

Any closing thoughts for readers?

Do not take life too seriously, laugh a lot, and do your part to create a better tomorrow.

VitaDAO Discover Monthly

VitaDAO was featured in multiple audio formats throughout May:

  • Laurence Ion and Todd White joined @DogelonWarriors on Twitter for a Twitter Live. Missed the Twitter live? Listen to a recording.
  • Tyler Golato and Laurence Ion joined the Translating Aging Podcast. Listen to their conversation on the essence of VitaDAO, project funding, and the intersection of biotech, blockchain, and longevity.
  • Laurence Ion and Theo Beutel joined the Campfire podcast, hosted by Jackson Steger. In this episode on Democratizing Longevity Research, they discuss IP within a DAO, VitaDAO’s current projects, and more.

What’s Going DAOn

June 2nd at 6 PM CET- Join a VitaDAO Dealflow Onboarding presentation by clicking, here.

Aug 29th-Sept 2nd- Join the ARDD 9th Aging Research and Drug Discovery Meeting. VitaDAO is one of the many sponsors of this event, happening in Copenhagen, Denmark.

DAOn’t Forget…

VitaDAO still has several open roles! These roles are best fit for individuals who are interested in longevity, well organized, and passionate about open source communities. Positions available include Longevity Dealflow Manager, Longevity Dealflow Principal, and Longevity Venture Associate.

Between co-organizing DeSci Berlin, releasing a new Monthly Longevity Research Newsletter, and four passed proposals, VitaDAO had a busy May. In the upcoming month, be on the lookout for new governance proposals, more panel discussions, and Evandro Fang’s project IP-NFT transfer ceremony.

Enjoyed this newsletter? Subscribing is the best way to guarantee you stay up to date with monthly VitaDAO updates. Know someone that is interested in longevity science, web3, or both? Forward this newsletter to them! 💛

They say April showers bring May flowers. Instead of flowers, we hope we rose to the occasion with June’s newsletter to show how VitaDAO flourished this May.
Read more
VitaDAOへようこそ
June 6, 2022
Hiroaki Hamada & Satoshi Hirano
Awareness
VitaDAOへようこそ

老化克服や寿命延長を目的とした医薬品開発を自律分散型組織(decentralized autonomous organization, DAO)によって主導するVitaDAOについて日本の読者の方々にご紹介します。Wikipediaによれば、DAO とは、「中央集権的なリーダーシップが介在しないメンバー保有のコミュニティであり、DAOの金融取引記録やプログラムルールがブロックチェーンによって管理されている」と定義されています。

VitaDAO Japanese Explainer Banner

This article is presented as an introductory resource for existing and future Japanese members of the VitaDAO community. The discussion here is focused on introducing readers to current issues in the field of longevity research, the VitaDAO ecosystem, and how to get involved in the VitaDAO community. The discussion here is broad and readers are encouraged to check out the VitaDAO discord and VitaDAO website for more detailed information.

Introduction

VitaDAOはバイオテックの研究とブロックチェーンを組み合わせた、全く新しいタイプのサイエンスコミュニティです。中でもVitaDAOは老化克服や寿命延長を目的とした、longevity research(長寿研究)に特化しています。VitaDAOのミッションは、長寿研究を加速し、それによって人類の健康寿命や寿命そのものを延長することです。さらに、ブロックチェーン技術によって、ファンディングや研究データをデジタル化することで、このミッションをより効率的に追求しようとしています。

VitaDAOでは、知的財産(IP)の活用する権利を得る代わりに長寿研究に資金提供しています。このVitaDAOが保有している特許を将来的には製薬会社等へライセンスアウトすることで、そこで得られた利益によって更なる資金提供も可能になると考えています。

すでに10以上のプロジェクトに資金提供が開始され、投資総額も200万ドルを超えました。メンバーが議論しているDiscordアプリには5000人以上が参加するなどコミュニティも活発になっています。

この記事では、VitaDAOを多くの皆さんに知ってもらうためにVitaDAOが挑戦している課題、VitaDAOの解決策、現在のエコシステムについてご紹介したいと思います。

  1. これまでの長寿研究の課題
  2. VitaDAOのエコシステム
  3. VitaDAOに参加する方法

これまでの長寿研究の課題

VitaDAOは長寿研究のどのような課題に挑戦しているのでしょうか?

加齢に伴う様々な病気は多くの国で健康問題のみならず財政的な問題になっており、長寿研究は地球規模で重要なテーマになっています。すでに、人類にとって加齢は一種の病気であるという考え方も浸透し始めており、幹細胞やミトコンドリア、ゲノム編集などをターゲットにした研究開発が進んでいます。

しかし、多くの医薬品研究開発は、数億ドルにものぼる莫大なコストと効果検証のための長い月日が必要なため、大規模な研究所やビリオネアなどの大富豪などが加齢研究を中央集権的に管理している現状があります [DiMasi et al., 2016]。したがって、仮に老化を治療できる夢の薬ができたとしても、一部の富裕層にしか渡らずに、誰にとっても手頃な価格にはならないかもしれません。

歴史的に見ても、近年の医薬品開発は、製薬会社による独自の特許の保有によって、研究開発が一部の資金が豊富な研究所などに限られ、高額になった治療法には一部の限られた人しかアクセスできないという問題があります。これに対してVitaDAOは、長寿研究の支援をオープンで、共創的なコミュニティでシナジーを生み出すことでこの問題を解決するために組織されました。

バイオテックにおける新たなライセンスは近年、大学や研究所などのアカデミアから生まれてきたものも多く(2018年には最も販売された医薬品の半数以上がアカデミアから創出されました。[Haung et al., 2021])、アカデミアのシーズをバイオテック産業に生かす仕組みづくりがさらに進めば、研究開発のさらなる効率化が期待できます。

現在、アカデミアの研究シーズの多くはライセンス化されておらず、資金が注入されないため開発がそれ以上進められない、いわゆる「死の谷」が存在しています。VitaDAOでは、この早期の研究に投資を行うことで、オープンな研究開発を促進することを目指しています。

また、既存のVCとの違いや利点はどこにあるのか疑問に思う方もいらっしゃるでしょう。VitaDAOのもう一つの特徴は、商業化まで時間のかかるような早期の技術にも投資できることです。通常のVCはファンドの運用期間が約10年間と決まっており、その期間内での投資回収を求められます。したがって、研究者がスタートアップを起業し、VCから資金を調達するには、10年以内に投資回収できるシナリオを示さなければなりません。しかし、バイオテックのベンチャーは臨床試験に時間がかかります。特に、長寿研究の成果検証には時間がかかることが想定されるため、10年間というVC側のファンド期限が大きなハードルとなり得ます。これに対しVitaDAOは、通常のVCのような時間制限が無いため、長期的な目線で投資を行うことが可能になります。

つまり、VitaDAOでは、①長寿研究によって得られる利益を一部の富裕層だけでなく、多くの人へ開放する、②アカデミアシーズの「死の谷」をオープンな共創によって解決する、③期限に縛られず長期的な投資を行うことを目指しているのです。

VitaDAOという自律分散型組織による解決策

VitaDAOはこれまで長寿研究が抱えていた課題の解決策として下記の3つを提案しています。

  1. IP-NFTsと呼ばれる知的財産(IP)のNFTによる運営費の管理と獲得
  2. Ocean marketplaceなどのようなデータ取引によるDAOの運営費の獲得
  3. VITAトークンによるVitaDAOのガバナンス参加

以下、順を追ってご紹介します。

NFTを活用した知的財産(IP)の運用

VitaDAOが提案しているNFTの使い方に知的財産権(Intellectural Property, IP)をDAOで活用するという方法があります。

VitaDAOでは研究プロジェクトへの出資条件として、プロジェクトで生み出されたIPをVitaDAOが保有することを求めています。またVitaDAOは、IPをNFT化したIP-NFTsを活用することで研究室のIPをブロックチェーン化します。このIP-NFTsを製薬会社などに貸し出したり、売買することで、売却益や取引手数料などを得る仕組みとなっています。かつて、通貨の概念で金本位制という仕組みがありましたが、VitaDAOはさながら「知財」本位制のような仕組みを目指しています。

データ資産の活用

VitaDAOが出資した研究で、DNA発現などの大規模な実験データが得られる場合もあります。このデータ自体もライセンス契約による貸出や売却をすることで、資産としての価値が出ることが期待されています。

VitaDAOはOcean Protocolが提供するOcean Data Marketというデータ取引のプラットフォームと提携しており、データを安全に販売やライセンス利用することができるようにしています。

VITAによるガバナンス

VitaDAOはVITAトークンを発行しており、ワーキンググループに参加しVitaDAOに貢献することでVITAトークンを獲得することができます。また、トークンを直接購入することもできます。トークンを保有しているメンバーはVitaDAOのいくつかの意思決定に参加することができます。

VITAトークンをもったメンバーは、

1. どのIPがDAOから資金提供を受けられるかの調査

2. プロジェクトの営利化の方法の決定

3. VitaDAOのガバナンス

4. 資金のマネジメント

などの意思決定に参加することができます。VITAの保有者が適切に参加することで、有望な研究開発が行われ、そこで得られたIPが適切に売り買いされることで、VitaDAOはその価値を高めていきます。このようにしてVitaDAOが持続的に成長していくことが可能となるのです。

VitaDAOは、これらIP-NFTs、データ資産、VITAトークンによって創薬研究への資金提供を持続的に行うためのシステムを作り上げているのです。

Diagram illustrating the VitaDAO IP-NFT and data ecosystem

VitaDAOのチームと現在のエコシステム

VitaDAOの現在のチームはどのようになっているのでしょうか?

VitaDAOではすでに、900万ドルの資金を持ち、5000人以上のDiscordメンバーが活動しています。また、30以上のプロジェクトがすでに評価され、総額150万ドルの長寿研究へのファンディングが行われました。

ここではVitaDAOを支えるチームを、VitaDAOのワーキンググループと支援の仕組みなどの観点から紹介していきます。

ワーキンググループ

ファンディングを含めた活動や、データ資産の活用、IP-NFTsを活用を進めるために現在7つのワーキンググループ(WG)があり、それぞれVitaDAOを支える重要な役割をはたしています。

法制度: LEGAL

法制度WGでは、VitaDAOにおけるコミュニティと運用に関する法制度に関する理解を高めたり監督しています。法的観点からコミュニティのIPと資産を守りつつWeb3での法制度に関するトレンドを確立していきます。

広報: Awareness

広報WGでは、VitaDAOをさまざまなメディアを通じて持続的なコミュニティの成長のためにVitaDAOの認知を高める活動に専念しています。メンバーは、web3や老化研究者のコミュニティに目に留まるようなコンテンツの作成を担います。

長寿研究サポート: longevity

長寿研究WGでは、有望なシーズ研究を探し出し、評価します。メンバーは、アカデミックな研究者からシーズ募集し、ファンディングのためにプロジェクト提案、評価、モニタリングします。

DAOガバナンス: governance

ガバナンスWGでは、VitaDAOのコミュニティのガバナンスや意思決定に関するインフラストラクチャーを担当し改善します。メンバーは、コミュニティの透明性、インクルーシビティ、効率性を確立します。

コミュニティ運営: OPS

コミュニティ運営のWGでは、VitaDAOで現在進行形で進んでいる活動の計画、実行、モニタリングをサポートしています。メンバーは、マイルストーンをチェックし、効率性や、タスクのインセンティブを担当します。

技術: TECH

技術WGでは、コミュニティのための技術的な機能を開発したり、維持することを担当します。メンバーは、コミュニティのgithubアカウントなどのウェブサイト開発、スマートコントラクト開発、その他コミュニティの要望に技術的に対応することなどを担当します。

トークノミクス: TOKENOMICS

トークノミクスWGでは、トークンエコノミーの管理と生成を担当しています。メンバーは、コミュニティの目的に沿うような行動のインセンティブを最適化するために$VITAトークンのモニタリングを行っています。

すでに、VitaDAO公式アカウントは1万人以上のTwitterのフォロワーがおり、認知度も高まりつつあります。プロジェクトの支援も実際にはじまっており、多くの研究者との連携も活発になってきています。

研究者との連携

VitaDAOは、ステイクホルダーとしてのアカデミックの研究者たちと協力しながらファンディングを進めています。

2021年には、コアメンバーであるTyler GolatoがARDD 2021においてVitaDAOを紹介する[Meron et al., 2022]したり、定期的にYoutubeで研究者とディスカッションを行うなど連携を深めています。

2021年7月にはファンディングが開始され、最初にコペンハーゲン大学のScheiby-Knudsen准教授のグループの健康寿命を延ばす化合物候補を検証するプロジェクトに対して25万ドルのファンディングが行われました。ファンディングは、VitaDAOのメンバーの保有者の投票によって決まります。現在までに、60つのプロジェクトが検討され、10以上のプロジェクト(VDP-5, VDP-8, VDP-15, VDP-16など)への資金提供が行われました。

VDP-5 Scheibye-Knudsen Lab Funding Proposal

また、このファンディングではIP-NFTを介して契約が行われています。対応するIP-NFTをVitaDAOが保有する代わりに対象の研究室が支援を受けるようになっています。

👇 Scheiby-KnudsenグループとVitaDAOで交わされたIP-NFT

https://opensea.io/assets/0x42d2354ef0b54279516f5799791086f2f499086e/77379318712039290824426798626037872787628160616774329410851863732077774590161/

これらの意思決定やサービスを一つのDAOだけで開発するには多くのリソースが必要になるため他のDAOとの連携が活発になっています。VitaDAOにおいても、すでにIP-NFTのプロトコルを提供するMoleculeやDAO支援のツールを提供するPrimeDAOといったDAOとの連携が発表されており、今後も持続可能な組織運営になるように連携を進めています。

VitaDAOに参加する方法

現在、VitaDAOに参加する方法は3つあります。

①研究者としての参加

研究者のためのファンディングは随時受け付けています。

所属する組織との同意のもと、IPをVitaDAOが所有することと引き換えに、最大で3000万円(25万ドル)ほどの直接経費と10%ほどの間接経費が受けられます。

👉ファンディングへの応募

さらに長寿研究を行う人たちに見返りを求めない資金提供も行っています。研究するため、学会に参加するためやOn Deck Longevity Biotechのプログラムに参加するためなど多様な用途で利用が可能です。

👉VitaDAOフェローシップ

②ワーキングメンバーとしての参加

先に述べたようにVitaDAOには複数のワーキンググループがあり、メンバーの募集を随時行っています。メンバーとして参加するためには、以下の手順に従ってDiscordアプリにご参加ください。

  1. Discordアプリをこちらからダウンロードしていただき、プロフィールを作成してください
  2. VitaDAOサーバーへご参加ください
  3. 自己紹介を「introductions」でお願いします。また日本語話者の場合は、「thread」ボタンをクリックしていただき、「japanese-chat thread」を開いてください。

詳しくはこちらの記事をご覧ください。

③支援者としての参加

さらに、VitaDAOを支援する方法はVITAトークンを購入したり、Discordアプリを通じて長寿研究のプロポーザルやVitaDAOの施策等について議論に参加するなどの方法もあります。またトークンの保有者はDAOで提起されたプロポーザルに意思決定(例えば特定の研究プロジェクトを支援するのに賛成か反対かなど)を下す権利も保有できます。

👉 VITAトークンの入手 (CowSwap)

他にもSNSやYoutubeなどから情報を随時更新していますのでぜひチェックいただきたいです。

SNS & Youtube

👉 Gov

👉 Medium

👉 Telegram

👉 Twitter

👉 Youtube

Twitterで活動しているメンバーたちもいます。ぜひフォローして日々の情報発信もチェックしてほしいです。

VitaDAOはさまざまなDAOと連携するだけなく、さまざまな研究者たちと連携しつつ、ミッションを追求しています。

VitaDAOに興味がある方はぜひご連絡ください。

執筆者

濱田太陽 株式会社アラヤ、国立研究開発法人量子科学技術研究開発機構(QST)で神経科学者として活動。Consciousness Club Tokyoの主宰。研究テーマは好奇心の神経メカニズムの解明。また、web3を通じて、教育や科学のオルタナティブな方法を追求している。

Satoshi Hirano 日本の官民ファンドのINCJに勤務。長寿研究のバイオテクノロジーに強く関心がある。長寿研究に焦点を当てたMediumブログに公開している。ODLB2フェロー。

Ariella Coler-Reilly ワシントン大学セントルイス校のMD・PhDコースに在籍し、老化の遺伝学について研究。サイエンスライター、イラストレーターとして活動する傍ら、現在はVitaDAOブログのマネージングエディターとしても活動中。公共教育、多様性と包括性、科学とWeb3の横断に情熱を注いでいる。

参考文献

  1. DiMasi, J. A., Grabowski, H. G., & Hansen, R. W. (2016). Innovation in the pharmaceutical industry: New estimates of R&D costs. Journal of health economics, 47, 20–33. https://doi.org/10.1016/j.jhealeco.2016.01.012
  2. Huang, S., Siah, K.W., Vasileva, D. et al. (2021). Life sciences intellectual property licensing at the Massachusetts Institute of Technology. Nature Biotechnology. 39, 293–301. https://doi.org/10.1038/s41587-021-00843-5
  3. Meron, E., Thaysen, M., Angeli, S., Antebi, A., Barzilai, N., Baur, J. A., Bekker-Jensen, S., Birkisdottir, M., Bischof, E., Bruening, J., Brunet, A., Buchwalter, A., Cabreiro, F., Cai, S., Chen, B. H., Ermolaeva, M., Ewald, C. Y., Ferrucci, L., Florian, M. C., Fortney, K., … Scheibye-Knudsen, M. (2022). Meeting Report: Aging Research and Drug Discovery. Aging, 14(2), 530–543. https://doi.org/10.18632/aging.203859

Thanks to Hiro Taiyo Hamada

老化克服や寿命延長を目的とした医薬品開発を自律分散型組織(decentralized autonomous organization, DAO)によって主導するVitaDAOについて日本の読者の方々にご紹介します。Wikipediaによれば、DAO とは、「中央集権的なリーダーシップが介在しないメンバー保有のコミュニティであり、DAOの金融取引記録やプログラムルールがブロックチェーンによって管理されている」と定義されています。
Read more
Longevity Research Newsletter May 2022
May 17, 2022
Maria Marinova & Rhys Anderson
Longevity
Awareness
Newsletters
Science
Longevity Research Newsletter May 2022

Welcome Vitalians to the inaugural issue of VitaDAO’s Monthly Longevity Research Newsletter!

Whether you are a longevity neophyte or a seasoned professor, we hope this will be an interesting and valuable resource to keep you up-to-date with the most important longevity research activity happening around the globe.

Why Longevity? Humans have long desired eternal life, with reports dating as far back as the first emperor of China, Qin Shi Huang (260–210), sending hoards of people out in search of the fabled Elixir of Life. But putting dewy-eyed dreams of immortality to one side, let’s consider that for many of the leading causes of death worldwide — cancer, heart disease, neurodegeneration — the biggest underlying risk is age. A school of thought has emerged proposing that, in addition to trying to tackle these diseases individually, we should also be investing in trying to understand the mechanisms which drive ageing itself in the hope that this knowledge could allow us to tackle all of these diseases simultaneously.

Why do we think ageing can be targeted? It is clear from looking at the animal kingdom that ageing rates are not universal — with lifespan ranges from a few years in small rodents to a staggering 200 years plus for bowhead whales. Some organisms such as sea hydras are even thought to be biologically immortal. But even comparing animals with similar sizes and metabolic rates such as mice and bats we observe a 10-fold difference in lifespan, with bats able to live over 30 years. Even among humans we observe disparities of over a decade in healthspan dependent upon postcode alone! Examples such as these provide evidence that ageing is influenced both by genetics and environment.

Experimental evidence? By mutating just a single gene in the microscopic nematode C. elegans, scientists were able to extend lifespan by around 10-fold. In mammals, a 50% increase in mice lifespan has also been achieved, which if translated to humans would mean some people living beyond 160 years old. Interventions such as calorie restriction and rapamycin dosing have also been shown to extend lifespan in numerous organisms.

Treating Ageing. Hitherto, ageing is not recognised by the FDA as a disease indication that can be treated. However the trail-blazing Targeting Aging with Metformin (TAME) trial, spearheaded by Dr. Nir Barzilai, hopes to achieve just that by conducting a 6-year study following over 3000 65–79 year olds to assess whether metformin can delay the onset and progression of age-related diseases https://www.afar.org/tame-trial

If successful, this will be a landmark moment in the history of medicine, however, conducting several year long trials in elderly populations for every new intervention is unfeasible and thus it will be imperative for more robust biomarkers of ageing to be developed so efficacies of interventions can be quantified much more quickly and cheaply.

VitaDAO’s Mission. Over the past few decades biogerontology research has gained significant traction in both academia and industry, however there is still a lack of funding for early stage research projects — VitaDAO aims to bridge this “valley of death” and accelerate R&D into the extension of both human healthspan and lifespan.

VitaDAO-funded Research Projects

This month we have seen two great projects receive approval for VitaDAO funding — with Jonathan An receiving up to 300k USD to tackle periodontitis (gum disease) with gero-science based treatments: https://snapshot.org/#/vote.vitadao.eth/proposal/0x8464713774628fa0af6262478f7c1ecccb0a3272cc72f6324183f598e9f29007 and ApoptoSENS receiving 253k USD to engineer Chimeric Antigen Receptor (CAR-NK) cells to precisely and safely eliminate senescent cells: https://snapshot.org/#/vote.vitadao.eth/proposal/0xcafd8183c8ac4990d99d90b3bd669de2c86376aa361c2384b12d578a49f686c0.

We will be keeping you up-to-date with each new project that gets funded as well as providing updates on existing projects when the data starts to roll in!

Longevity Literature Hot Picks

There is now such a plethora of longevity research being published on a daily basis that we would need a significant lifespan extension to read everything! So to help we have provided you with a selection of our hotpicks of the month — enjoy!

Somatic mutation rates scale with lifespan across mammals

https://www.nature.com/articles/s41586-022-04618-z

Somatic mutations are well known to cause cancer, but they also occur in normal tissue — for example skin cells. With age these mutant but morphologically normal cells multiply but no tumour develops. This happens in multiple tissues and the number of mutant cells dramatically increases with age. The Nature study shows that mutation rates grow linearly with age across species and the calculated mutation rates per year differ significantly between species. The organism mutation rate has a strong reverse correlation to lifespan.

Don’t forget to check out our interview with Alex Cagan — the first author of this paper — below!

Multi-omic rejuvenation of human cells by maturation phase transient reprogramming

https://elifesciences.org/articles/71624

Cellular reprogramming has definitely been a hot topic in the aging field and this new paper from Reik lab shows a new method to achieve this. The novel technique outperforms other reprogramming protocols. It was demonstrated to be highly effective in rejuvenating the epigenome of human skin cells from middle aged donors and turning the clock back 30 years according to a new transcriptomic clock.

FGF21 is required for protein restriction to extend lifespan and improve metabolic health in male mice

https://www.nature.com/articles/s41467-022-29499-8

Protein restricted diets have been shown to extend lifespan in numerous organisms — here Hill et al., show that beneficial health effects of protein restriction in mice are dependent on the metabolic hormone FGF21.

Early SRC activation skews cell fate from apoptosis to senescence

https://www.science.org/doi/full/10.1126/sciadv.abm0756

Upon severe genotoxic insult, cells undergo either apoptosis or senescence — here the authors show that inhibition of the kinase SRC can cause cells to preferentially apoptose, thus highlighting a potential therapeutic avenue to target senescent cells in aged or damaged tissues.

Cell senescence, rapamycin and hyperfunction theory of aging

https://pubmed.ncbi.nlm.nih.gov/35358003/

Prof. Mikhail Blagosklonny provides an interesting perspective that hyperfunctional signalling pathways, such as mTOR, can drive both cellular senescence and organismal ageing whilst arguing that the two are not dependent on each other.

Nutrition, longevity and disease: From molecular mechanisms to interventions

Longo and Anderson review over a century of research, adopting a multi-pillar approach, to begin formulating an optimal longevity diet:

https://www.cell.com/cell/fulltext/S0092-8674(22)00398-1

Clinical Trial Updates

Whilst ageing is not (yet) recognised as a disease indication, there are numerous ongoing trials aiming to tackle age-related diseases.

A team from Osaka University, led by Prof. Koshi Nishida, have announced they have successfully improved vision in near-blind patients by transplanting iPSC-derived corneal tissue.

https://english.kyodonews.net/news/2022/04/c8af6b7913b2-japan-team-proves-ips-based-cornea-transplant-safe-in-world-1st-trial.html

BioAge Labs announced that the first cohort has been dosed in a Phase 1b trial of their drug BGE-105 — an inhibitor of the apelin receptor, aimed at tackling muscle atrophy.

https://bioagelabs.com/

Upcoming conferences

Now that worldwide travel restrictions from the COVID-19 pandemic are easing, we can all look forward to meeting up again in person to discuss longevity research.

May 17–20, 2022, San Antonio, Texas, fully integrated hybrid (in person-virtual)

https://www.americanagingassociation.org/annual-meeting

Bay Area Aging Meeting (BAAM)

20th Bay Area Aging Meeting Tuesday

May 17, 2022, UC Berkeley, Li Ka-Shing Center, Room 245

https://agingmeeting.org/registration/

Omics Approaches and Biomarkers in Aging

Systems Aging Gordon Research Conference Systemic Processes

May 29 — June 3, 2022, Newry, ME, United States

https://www.grc.org/systems-aging-conference/2022/

The Reproductive Aging Conference
Jointly hosted by FASEB and the American Aging Association (AGE)
June 5–9, 2022, Palm Springs, CA

https://www.faseb.org/meetings-and-events/src-events/the-reproductive-aging-conference

Funding Opportunities

After making a seismic shake-up to the research funding process with their short application form and an astonishing turnaround of just 3 weeks from submission to decision, the Impetus Longevity Grants are back! This time the criteria are more focused, so get your thinking caps on and good luck: https://impetusgrants.org/

New Science are on a mission to create an alternative avenue for basic research to be carried out independently of academic institutions, with donors including Jaan Tallinn (co-founder of Skype) and Vitalik Buterin (creator of Ethereum) amongst others. Applications for their first 1 year fellowship closed on May 1st, but definitely worth keeping an eye on for the future. https://newscience.org/

Job board

Modulo Bio

Modulo Bio is a neuroimmunology company that harnesses the power of machine learning, patient iPSC derived cellular modeling and large scale CRISPR screening to develop therapies for neurodegenerative diseases like ALS and Frontotemporal dementia

Senior Research Associate or Research Associate

Please contact jobs@modulo.bio for more information.

BioAge

Platform developing therapeutics to extend human healthspan and lifespan

Multiple roles: https://jobs.lever.co/bioage

Early-stage stealth longevity company is hiring interns for this summer (2–3 months):

In-person / hybrid in Redwood City, CA for soon-to-be or recent graduates

  • Scientific Research interns (“aging clocks” ) — Biology / Medical majors
  • Medical Research interns (aging interventions) — Biology / Medical majors
  • Product / Market Research interns (Productization) — Business / Economics / Entrepreneurship / CS majors

Email your resume and 4–5 sentences which answer, “Why do you want to intern at our early stage biotech / longevity focused startup?” to our Recruiting Lead aqil@longevity-co.com. Deadline until May 31st, 2022

VitaDAO

We are collectively funding, and advancing longevity research in an open and democratic manner. Longevity dealflow is currently looking for three positions: manager, principal and venture associate.

https://vitadao.notion.site/43a4a6a8889a4f7190ddc06c453fc3d5?v=12750ada30364c2eb9645d9627e7d818

Newlimit

Newlimit are a new company hoping to tackle age-related diseases and extend lifespan by targeting epigenetic drivers of ageing. They currently have numerous roles available across a range of levels:

https://www.newlimit.com

SiPhox

YC company SiPhox https://siphox.com/ is looking for someone to fill a FT or consulting d2c growth role. They are looking for someone who is interested in aging, longevity, and blood biomarkers who has marketing/startup/growth experience and can help design some branding and UX

Retro

Retro Biosciences https://retro.bio/careers/ are focusing on extending human healthy human lifespan via cellular reprogramming, autophagy & plasma-inspired therapeutics. Thy are recruiting multiple roles, the majority of which are science based.

Post-doc (assistant/associate) position in Alberto Sanz’s lab, University of Glasgow

The aim is to investigate why mitochondria produce more ROS as we age, with a view to implementing strategies to tackle mitochondrial ageing.

https://www.nature.com/naturecareers/job/research-assistantassociate-university-of-glasgow-757335

Advertise Here!

If you would like us to advertise any of your longevity-related positions from PhDs, to postdocs, to start-up hires etc then please get in contact and we will be glad to include them in next month’s issue.

Interview with Dr. Alex Cagan

After studying Anthropology at the University of Cambridge followed by a PhD in Evolutionary Genomics of Animal Domestication at the Max Planck institute, Dr. Cagan now works as a scientist at the Wellcome Sanger Institute — utilising the power of comparative genomics to better understand the processes of somatic evolution and how it relates to cancer and ageing. He also works as a scientific illustrator and is passionate about the combined power of art and science.

How do you think your latest paper on somatic mutations will impact the field of ageing research and what are the remaining questions?

In our paper we looked at cells in the intestinal crypt across 16 mammalian species and found a strong inverse correlation between somatic mutation rate and lifespan across. In other words the longer the lifespan of a species the slower the DNA mutation rate in the cells of that species, such that a mouse and a human end their lifespan with a similar number of somatic mutations in their intestinal cells. These findings are consistent with a role for somatic mutations in the aging process, something that has been theorized for decades. What the results don’t tell us is through what mechanisms, if any, somatic mutations are contributing to aging. This remains a fascinating open question. Our results also suggest that longer lived species than humans may have found ways to lower their mutation rate beyond our own. Studying these species and understanding how they achieve this, potentially through higher fidelity mechanisms of DNA repair, could be of great benefit.

What inspired you to enter longevity research?

Aging fascinates me because it is one of the fundamental biological processes that still remains relatively mysterious. As an evolutionary geneticist I am fascinated by exploring the relatively uncharted landscape of somatic evolution, the process by which mutations in the DNA occur and spread throughout the human body as we age. It’s long been known that this process causes cancer but due to technical limitations it has been challenging to study this process in normal tissues as we age. Now that genome sequencing methods have improved we are able to study this hitherto inaccessible process and are learning about how mutations in our cells radically alter the molecular landscape of our bodies as we age. I am excited to discover to what extent these processes may be contributing to a range of age-related diseases and potentially even aging itself.

How has the field changed since you started?

There has been an enormous surge in interest in this field since I started, both within academia as new technologies have been developed that enable us to quantify the aging process in new ways and from industry, as the potential of breakthroughs to improve the human healthspan is enormous.

Other than your own, what do you think have been the biggest/important discoveries in the field?

Because of the nature of aging as a multifactorial process there have been a multitude of important discoveries. As we have yet to fully understand which processes are the key drivers of aging it is challenging to assess their relative importance, they all contribute to our understanding. I would say that the discovery of epigenetic clocks, that telomere shortening and proteome turnover rates correlate with species lifespan and our growing understanding of cellular senescence are among the most important discoveries in the field.

What advice would you give to people currently working in longevity research?

I would say that it’s an incredibly exciting time to be working in this field and that given the multifactorial nature of aging a central challenge now is to quantify to what extent different molecular mechanisms contribute to specific phenotypic changes that occur during aging.

Which aspect of longevity research do you think requires more attention?

There are already species that have far longer lifespans than our own. More energy could be devoted to studying these species in order to understand the mechanisms by which they have extended their lifespans. In some senses evolution has already solved the problem of aging in these species and we would probably do well to try and learn from these solutions that are already sharing the planet with us.

Is ageing a disease?

Aging certainly raises the risk for a number of diseases. Whether or not we define aging itself as a disease I would consider largely a semantics issue, though with implications for how we think about our lives and how we fund aging research. I enjoy considering different perspectives on aging, whether it is something we should ‘accept’ or ‘conquer’ or ‘cure’. Such conversations remind me of Sophocles’ lines about humanity from Antigone ‘Numberless are the world’s wonders, but none more wonderful than man; …from every wind he has made himself secure — from all but one: In the late wind of death he cannot stand.” It is awe inspiring and humbling to think that there is a realistic prospect that aging itself could be overcome through human ingenuity. Though we are currently far from reaching this point I think it’s important that as a society we begin to have conversations about what the implications of this would mean.

Outro

Thanks for reading VitaDAO’s first ever Monthly Longevity Newsletter!

We are constantly striving to provide valuable content for our community, so if there is anything you would like us to feature in future issues, please get in contact.

If you are keen to see what else has been going on in the VitaDAO community, we have provided links below to some of our highlights from the past month — enjoy and we look forward to seeing you next month!
Subscribe here for more!


Phage Directory: How a Decentralized Network of Researchers Find Cures For Drug-resistant Infections

https://www.youtube.com/watch?v=F0xYTYg48qU

First VitaDAO Crypto meets Longevity Symposium

https://www.youtube.com/watch?v=GJ-rJAfjhBE

Decentralizing Science — Using Blockchain to Reinvent the Scientific Process

https://www.youtube.com/watch?v=dzJUDgwzxPA

Novel Mechanisms to Fund Longevity Science

https://www.youtube.com/watch?v=E8YN6-tNkcQ

Heterogeneity in Senescence: from Mechanisms to Interventions with Prof Marco Demaria

https://www.youtube.com/watch?v=hnZELlseRyY

How Crypto Can Help Science & Medicine by Tim Peterson

https://vitadao.medium.com/what-crypto-means-for-science-medicine-4b52703b5935


Welcome Vitalians to the inaugural issue of VitaDAO’s Monthly Longevity Research Newsletter!
Read more
VitaDAO Newsletter Issue No 5 May 2022
May 1, 2022
Sarah Friday & Hossam Zaki
Awareness
Newsletters
VitaDAO Newsletter Issue No 5 May 2022

Hello Vitalians! April Fools might have started the month, but we promise this Newsletter is no joke! Many people in VitaDAO were able to attend ETHAmsterdam for one of the first ever DeSci days, featuring talks about all things DeSci. In this month’s newsletter, learn more about new proposals, job openings, votes, community member Ariella Coler-Reilly, and other things happening around the DAO.

Community News

Crypto x Longevity Symposium

April contained the first VitaDAO crypto meets longevity symposium. The Symposium featured talks from longevity researchers, members from LabDAO and Molecule, and of course, VitaDAO. We had over 300 attendees for the Symposium, and we hope to do this again in the future! Missed it? Catch a recording of the symposium.

Refer to VitaDAO, Make $$

Know of anyone doing interesting work in the longevity space? Do you like getting money for advancing scientific research? Then refer a project to VitaDAO! For each relevant project you introduce us to, you can get 500 $USD or 1000 $VITA. In exchange, the project can get funding and VitaDAO’s support to advance its mission. Have a researcher in mind? Fill out the form here.

Bridging IP into Web3 with IP-NFTs

IP is a difficult and confusing process, and our good friends at Molecule are working hard to simplify it, while also enabling new fundraising and collaboration strategies. They recently released a thread on this topic, which you can find here, as well as an article by Molecule and VitaDAO Cofounder Tyler Golato, here.

Apply for a VitaDAO Fellowship

We’re still accepting applications for our VitaDAO fellowship! This fellowship funds fast grants of up to $3k to fund you to do research, take a break to focus on longevity, attend a conference, or join a program. This fellowship has NO strings attached, with no expectations or claim of any research resulting from the fellowship. We have supported 37 people so far, with funding supporting attendance at longevity conferences, conducting research, and joining the On Deck Longevity Fellowship. Want to apply? Check it out here.

DeSci Day at ETHAmsterdam

People all over the world working in DeSci met up in Amsterdam for DeSci day. We had panels discussing IP-NFTs, governance, diversity, partnerships and onboarding in DeSci DAOs, as well as panels from our friends at Molecule and LabDAO! Didn’t get the chance to go to Amsterdam in person? No worries! There’s a recording of the day.

Upcoming DeSci Conference in Berlin

DeSci day at ETHAmsterdam was a massive success…. so it only makes sense to have a whole DeSci Conference! We are planning a DeSci conference in Berlin on May 23–24th. We are currently planning our speaker schedule, but expect a lot of talk on IP-NFTs, DeSci DAOs, research funding, and DeSci execution. Register or apply to be a speaker!

Open Roles at VitaDAO

If you are a well-organized and dynamic person, curious about the world of longevity, crypto, and open-source communities, then we’re looking for you! We’re hiring 3 positions: Longevity Dealflow Manager, Longevity Dealflow Principal, and a Longevity Venture Associate. Interested? Check out this thread with each position here.

New Agent of VitaDAO

VDP23IP, a Swiss Association (Verein), was formed! It will serve as VitaDAO’s IP holding company following our vote on VDP-23. Stay tuned for news about how to attend VDP23IP’s inaugural General Meeting during DeSci Berlin! There it will appoint members and set the roadmap for the next year. Register your interest in becoming a member of VDP23IP by messaging Jesse or Savva in the Legal Working Group. We’re filling spots for a Council of 9.

Hot off the Press

Tim Peterson, Victoria Perdue, and Zach West released a new article, “How Crypto can Help Science and medicine.” In this article, they write about how DeSci can improve both clinical medicine and research, by funding more projects as well as prioritizing going-to-market plans, instead of the standard of patents. If this sounds interesting to you, make sure to give the article a read.

What’s been going DAOn

We passed a few proposals, and currently have others open for voting on Snapshot! As always, you can follow proposals and add to the conversation before votes are added to Snapshot on the VitaDAO’s Discourse.

VDP-18 Passed! Towards Reversing Periodontal Disease using Geroscience

Periodontal disease is a chronic oral disease with inflammation of the tissues supporting the teeth. The disease’s greatest underlying risk factor is age, highlighting the importance of studying periodontitis within the lens of geroscience studies. This passed proposal provides up to 300,000 USD to fund an 8-week mouse-model study that will use small molecule inhibitors of the PI3K/NFkB/mTOR pathway to treat periodontal disease.

VDP-38 Passed! ApoptoSENS — Senolytic CAR-NK cells

We are funding more longevity research projects! This proposal provides 253,000 USD to fund SENS Research Foundation’s ApoptoSENS project to develop CAR-NK cells to precisely and safely eliminate senescent cells. This is a mouthful, so let’s break it down. Over time, some cells experience damage, enter senescence, and can secrete proinflammatory factors leading to tissue dysfunction. By creating Chimeric Antigen Receptor Natural Killer (CAR-NK) cells to eliminate these senescent cells, we hope to be able to prevent this tissue damage.

VDP-22 Passed! Probing DNA Quadruplexes in Age Related Proteostasis

We voted to contribute 50,000 USD to a 4 year doctoral program at the Institute of Chemical Biology, Imperial College London. The goal will be to determine the role of DNA Helicases as regulators of proteostasis and to understand more about their impact on declining function before the onset of age-related proteostasis.

VDP-42 Continued Funding of VHF Passed!

VDP-42 proposes contributing $680,000 to the Vitality Healthspan Foundation. This would fund an additional arm’s-length loan of $580,000 from VitaDAO to VHF, secured against the underlying equity in companies VHF in turn funds, and a $100,000 donation to VHF to support VHF and VitaDAO’s aligned non-profit initiatives. This is an active proposal, ending in early May!

VDP-40 Passed! Cellular ‘Soap’ For Infectious Disease Accelerated Aging — Dr Tim Peterson

VitaDAO will allocate $231,000 to Tim Peterson’s project developing a cellular “soap” (sphingolipid mimetics) to thwart pathogens and senescent cells.

Community Member Highlight: Ariella Coler-Reilly

How did you first get involved with the VitaDAO community/ DeSci?

I studied the genetics of aging at WashU under Dr. Tim Peterson, who is one of the stewards of VitaDAO’s deal-flow group. At first, I had no interest in VitaDAO because I imagined a group of finance bros and blockchain developers, which is definitely not me. However, I ultimately realized that all kinds of people with all kinds of skills are needed to run a biomedical DAO. In August 2021, I joined the DAO as a science communication contributor, beginning my rapid descent into the crypto rabbit hole, and I have never looked back!

How do you balance your work as a very active DAO community member and full-time Ph.D. student?

Frankly it is very difficult, and I sometimes struggle to establish boundaries and allocate my time appropriately. That said, an important part of the Ph.D. experience is participating in career development opportunities to network and explore options for the future, and VitaDAO has given me more of these opportunities than I ever could have imagined.

Can you walk me through what your position in VitaDAO entails?

My primary position is Managing Editor of our longevity science blog (https://vitadao.medium.com/). I help onboard new science communication (#SciComm) contributors and guide them through the process of writing articles that match their areas of expertise. It is critical that all of our community members feel informed about the foundational biomedical principles underlying aging and longevity biotech.

What has surprised you most about VitaDAO?

I was surprised by the sheer volume of people from all over the world with various professional backgrounds who have self-aggregated here in our community. The tools we use are native to the gaming and crypto realms but completely foreign to others — and yet, so many people have taken a leap of faith and created their first crypto wallet and Discord messaging account because we all share this common goal of advancing research to extend healthy life.

What are you most excited about in VitaDAO’s future?

There is a lot to be excited about, but one highlight for me is the potential for DAOs to disrupt the broken drug development pipeline by realigning financial incentives around the interests of researchers, physicians, and patients. The biotech valley of death has killed so many potentially groundbreaking initiatives, and I believe VitaDAO is leading the way in building a bridge to finally cross that valley.

Any closing thoughts for readers?

We always need more science writers and illustrators. If you are a science communicator interested in the aging and longevity field, please get in touch!

VitaDAO in the Wild

A16Z posted an article written by Jocelynn Pearl outlining the current landscape of DeSci. It is an excellent read to know more about what’s going on. Peep the VitaDAO shoutout!

Balaji Srinivasan mentioned us and Molecule on Shane Parrish’s Podcast, “The Knowledge Project.” Check out the clip here.

Last month, VitaDAO partnered with Dr. Peter Diamandis, founder of Abundance360 community. Every A360 member became a $VITA token holder! This month, VitaDAO was shouted out by Peter H. Diamandis in a Twitter thread.

Check out an amazing podcast by BioAgePodcast with Tyler Golato and Laurence Ion about the essence of VitaDAO, changing incentive structures around drug development, and more!

Laurence Ion and Vincent Weisser joined DefiDad to talk about VitaDAO and decentralized science and biotech more broadly.

Upcoming Events

May 12th at 6PM CET- Attend a VitaDAO Panel Discussion On Applied Longevity Medicine with host Max Unfried and speakers Prof. Evelyne Bischof, Prof. Andrea Maier, and Weronika Prusisz. Sign up here

May 18th at 9 PM CET- (Recurring Every Wednesday) — Pop into VitaDAO’s weekly onboarding session on Discord with Niklas Rindtorff and Alex Dobrin. Join the VitaDAO Discord server to learn more about the DAO, how to get involved, and find schedules for specific working group meetings.

May 23–24th- DeSci Conference in Berlin! Make sure to register if you’re interested in attending! If you are unable to make it in person, you can watch the live stream on Youtube! A link will be available on our Twitter.

Aug 29th-Sept 2nd ARDD 9th Aging Research and Drug Discovery Meeting

DAOn’t Forget…

Have questions, concerns, or inquiries? Join the Discord channel and introduce yourself! VitaDAO is a friendly community and its Discord functions as the heartbeat of the DAO. Our operations team is great at onboarding and directing newcomers to roles that fit their skills.

Want to know more about VitaDAO’s funded projects? Check out the newly revamped VitaDAO website! Kudos to our Tech Working Group!

If you enjoyed reading this newsletter, make sure to subscribe! Forward this to people you think would enjoy this- no April fools here or May showers to come, we promise!

Hello Vitalians! April Fools might have started the month, but we promise this Newsletter is no joke! Many people in VitaDAO were able to attend ETHAmsterdam for one of the first ever DeSci days, featuring talks about all things DeSci
Read more
How Crypto Can Help Science & Medicine
April 23, 2022
Tim Peterson
Science
Awareness
How Crypto Can Help Science & Medicine

Science & medicine are a monopoly

It might be hard to believe but science & medicine are caught in an innovator’s dilemma. This concept describes how an incumbent loses its edge to a disruptor because the incumbent focuses too much on what makes it successful now rather than what will make it successful in the future.

How can this be with so many advances happening all the time? It’s because the “customers” of science & medicine — the general public — are losing out. In the innovator’s dilemma, the value to customers when plotted as a function of time is a S-curve. Late in the innovation cycle there isn’t enough value being delivered to enough customers so the curve flattens out. As the COVID pandemic made clear, the problem is there is a monopolistic “company” that charges too much money and makes it too difficult on its “customers”.

What is the monopoly I’m referring to? It’s a cabal of entities largely in the US. Its regulatory agencies such as FDA and CDC, insurance companies such as United Healthcare, funding agencies like the NIH, and publishers such as NEJM and the Nature journals. Though there are sibling entities in other countries — let’s be honest — the US dictates much about what happens in science & medicine around the world.

by Victoria Padure

Besides unsatisfied customers, how do we know U.S.A., Inc. has existential problems? Look no further than its own “employees”. Complaints by US scientists and doctors (disclosure: I am one) about the monopolistic practices of our “employer” have grown hopelessly long. We have spilled too much ink on the abuses to be able to summarize here. Why don’t these complaints change anything? This is why the innovator’s dilemma exists. Science & medicine is a monopoly too locked into its own “success”.

DeSci is anti-trust

Herein I argue if there were an alternative financial path for scientists and doctors where our time would be better spent, we would follow it. That path is DeSci. DeSci stands for decentralized science. Today it’s a nascent idea. But it’s an idea that has the potential for scientists and doctors to have a bigger impact because we’d be bringing our talents to the whole world. The whole world to fund our research. The whole world to benefit from our treatments.

by Victoria Padure

Decentralized = alpha

DeSci takes its name from the first industry disrupted by crypto a.k.a. web3, finance. Decentralized finance (DeFi) applications use smart contracts and cryptocurrencies to perform a variety of financial services. DeFi is powerful because these crypto tools significantly improve upon traditional financial (tradFi) services. Take for example the tradFi staple — a savings account. Banks provide less than 1% yield on savings accounts whereas DeFi applications often provide 5–50% yields.

In finance, one’s returns or yield relative to those of the market are described as “alpha”. The whole point of “decentralized” is to maximize yield by not being restricted by geography. What does yield look like for DeSci? Basic research and clinical medicine have different goals so let’s consider them separately.

How the “De” in DeSci can improve clinical medicine

Yield in clinical medicine means more shots on goal — more new drugs and more clinical trials. For DeSci to make these a reality we need a few things:

1. Demonstrating the advantages of decentralized clinical trials (DCT). DeSci pioneers need to adopt a willingness to test a new drug wherever in the world can get the trial done the quickest and most economically, yet also safely. This will almost assuredly not be in the US. The FDA already has a huge backlog of projects to deal with. Only those with the most resources can get through it. Instead, we need to look to people and organizations elsewhere in the world who have a greater sense of urgency and where barriers to entry are lower. For example, perhaps a DCT trial for a new ALS treatment could involve doctors and patients in both New Zealand and Uruguay because of the high prevalence of this condition in these countries? Besides more quickly connecting more doctors around the world to the latest treatments, to make DCTs work we also need to develop a remote-first mindset in trial design. Meaning, we must strongly consider whether trial participants need to go get expensive and burdensome testing in the hospital as is typically done in the US-based trials. Or rather, whether at-home monitoring e.g., using more accessible approaches like smart phone apps or low-tech solutions could suffice.

2. Focusing on going-to-market over patents. In the incumbent system the costs of drug development are prohibitively high ($billions and 5–10+ years) and growing all the time. Patents are needed in this system to protect one’s investment because it takes so much time and money until gains can be realized. But what if patents aren’t necessary? As said above, DCTs will make the main cost driver of drug development — clinical trials — cheaper and take less time. At some point if a therapy is cheap enough and enough doctors and patients to prefer it, other drug companies won’t be able to undercut it — patent or no patent. Going-to-market first will win out over protectionism like it does in other sectors of the economy.

With crypto, profits also aren’t necessary. There’s no shareholders to pay dividends to. There’s no CEO pushing an agenda. Crypto represents a fundamental change in how people align incentives where contributors and value creators are compensated without the burden of having to pay middlemen and corporate overlords. So how does this profit-less system work? It works because with crypto the overhead is significantly less and at the same time it has significantly more tools than tradSci does in its financial toolbox to sustain itself and reward its participants. Crypto projects have tokens, can tap into DeFi, and can use tokenomics. The point is these things can be engineered to ensure funds last and can go back into funding more trials. Not only that, they can be engineered out in the open so that anyone who wants to participate can. It’s a more fair system for everyone.

How the “De” in DeSci can improve basic research

Yield in basic research means more funding for research and less time spent by researchers on non-research activities. To make DeSci a reality in basic research we need a few things:

1. Achieving funding levels that rival those offered by tradSci grant agencies. The DeSci ecosystem has to be capable of functioning independently of legacy systems. In basic research this means being able to financially support researchers fully so they don’t need to also rely on tradSci. In the aging/longevity field, this is happening quicker than expected. Three DeSci initiatives have emerged in the last year, VitaDAO, Impetus Grants, and Gitcoin that are funding in the $100K-$1M range. These funding levels are already about half of NIH’s shorter-term and longer-term grant mechanisms, R21 and R01, respectively. The goal for DeSci should be to continue to move from being a supplementary source of funding to being a main one.

2. Deprioritizing academic journals. DeSci funding organizations can help the researchers more than just with money. They can help scientists save considerable time. How? The simplest but biggest first step would be to make sure researchers interested in getting DeSci funding need only to have their research posted on preprint servers (e.g., BioRxiv, MedRxiv, and Arxiv) rather than also at academic journals like Nature, Science, and Cell. Why would DeSci funding agencies do this? It’s because academic journals waste huge amounts of researchers’ time on things that have little impact on the research. Things like having researchers format and reformat their work many times and respond to endless reviewer comments. Often it takes years to move from initial submission to the public release of the work in an academic journal. Also, academic journals profit disportionately off of the work of researchers because we not only produce the research and pay the journal to publish it, but we also review others’ work for the journal for free! If a preprint only requirement was made, researchers could post their results free of cost and with little time investment and move on to do more science. (TradSci grant agencies also waste huge amounts of researchers time, but until DeSci has enough money flowing through it researchers will still need to rely on it.)

Both clinical medicine and basic research will require strong coordination between DeSci organizations. DeFi applications seamlessly integrate with each other because they share the same database, the blockchain. The same needs to happen with DeSci. We need coordination between multiple DAOs to forge this new system independent of the tradSci oligarchy. As noted above, as soon as researchers can get their entire money from crypto, they won’t need the legacy system. One early example of inter-DAO coordination is LabDAO performing de-risking drug mechanism-of-action studies for the longevity therapies VitaDAO funded. These two organizations can trade tokens rather than transact in USD, saving both parties money while at the same time generating upside for each other.

How does Innovation happen?

Zooming out, let’s remember innovation doesn’t typically involve incumbents. Incumbents slow down the disruptor. Look at the handling of crypto by the SEC. The SEC held up crypto projects for years, played favorites, and won’t disclose its employees’ own crypto holdings. Fortunately, crypto pioneers have ignored the SEC’s indecision and bad faith negotiation and created ecosystems such as DeFi a $40B industry in less than two years (which rivals the budget that it took 80 years for NIH to have). We propose the same thing needs to happen with DeSci. Instead of waiting on the FDA, NIH, or anyone else to bless DeSci, crypto pioneers should just make it happen without the US.

DISCLAIMER: This is an opinion piece that represents the views of the author, Tim Peterson, and not those of VitaDAO.

By focusing anywhere but the US, DeSci can solve the hardest problems with science & medicine.
Read more
VitaDAO Newsletter Issue No 4 April 2022
April 7, 2022
Sarah Friday & Hossam Zaki
Awareness
Newsletters
VitaDAO Newsletter Issue No 4 April 2022

Hello Vitalians! Another month, another newsletter- and we couldn’t be more excited about it. Many of the core contributors to the DAO were able to meet up in sunny ☀️ Lisbon this month to organize, bond, and brainstorm in person. In this month’s newsletter, learn about the community’s first treasury 💰 report, find a review of proposals and votes that passed in the DAO 🗳, and find out more about what’s going DAOwn inside the DAO!

Community News

2021 Wrapped in VitaDAO

Forget about Spotify Wrapped, the rave is all about VitaDAO’s newly released 2021 Community and Treasury Report (aka the enDAOment report). In this report, find a summary of the workings of VitaDAO’s six working groups (Awareness, Longevity, Governance, Legal, Technical and Product, and Operations) and a comprehensive tokenomics overview. If you’re interested in learning more about what VitaDAO did in 2021, this is something you’ll want to read!

Rapamycin & Healthspan Trial with Dr. Brad Stanfield

In a March community call, Dr. Brad Stanfield chatted with VitaDAO contributors Jason Colasanti, Tim Peterson, and Todd White about the effect of regular exercise and intermittent rapamycin dosing on muscle performance in older adults. Dr. Stanfield has designed a randomized, double-blind, placebo-controlled trial to test the role of rapamycin in muscle performance💪. If you missed this call, you can find it on VitaDAO’s Youtube channel!

Longevity Hackers Film — VitaDAO Community Call

As if one community call wasn’t enough! In this community call, learn about the Longevity Hacker’s Film🎥. The Longevity Hackers is a feature-length documentary centering on longevity and radical life extension.

Extra! Extra! Read All About Epigenetic Clocks! ⏰

Do biological aging tests work? Author Lindsay Ciocco’s medium article explores different DNA methylation clocks, their applications, and the utility of home biological age testing.

Interested in buying a Glycan Age test? Buy one using $VITA and use VitaDAO’s community discount (code: VITADAO) to save 50%!

Journal Club: Can We Modify Immune Cells to Fix Broken Hearts?

In this journal club, hosts Jane and Estefano dive into the use of immune cells to fix a cardiac injury🫀. In this paper, scientists engineer immune cells to try to treat heart cells undergoing wound healing. Missed the journal club? Listen to it on Twitter Space or Anchor.

Journal Club: Can Young Gut Bacteria Help You Act Younger?

Hosts Ariella and Estefano explore the role of gut microbiota 🦠 in aging. Does transferring poop from a young mouse to an older mouse convey any benefits? It’s not too late to take a peek at the Nature article before listening to a recording of the Journal Club on Twitter Space.

We raised money! (again)

Once again, VitaDAO teamed up with Gitcoin to launch quadratically matched longevity research donations🤑. And once again it was successful! Check out a list of projects funded and funded amounts, here.

A New Partnership

VitaDAO is partnering with Dr. Peter Diamandis! Peter is involved in health initiatives such as XPRIZE, Abundance360, Celularity, Vaxxinity, and HumanLongevity, Inc. This partnership merges Peter’s Abundance360 community with our community as each A360 member will become a $VITA token holder! Here’s to funding longevity research with even more friends!

Ch-ch-ch-ch-changes

In case you too haven’t caught on yet, VDP stands for VitaDAO Proposal. In March, there were many active VDPs voted on and passed. Let’s go through them.

VDP-26 Dealflow Structure and Incentives

VDP-26 was voted on to provide clarity on the Longevity Dealflow Working Group operations. Deal flow is involved in sourcing projects, shepherding projects, and sharing information with the DAO. Want to learn more? Take a look at a visual representation of the new deal flow process.

VDP-30 VitaDAO Gitcoin Quadratic Longevity Funding Round 2 + VitaDAO Longevity Fellowship

VDP-30 funded $40,000 USDC to VitaDAO’s Gitcoin quadratic donation round and $15,000 to VitaDAO’s Longevity Fellows. This enables VitaDAO to continue to support smaller Longevity-centric enterprises!

VDP-31 Supporting of Clinical Trial- “Effect of Regular Exercise and Intermittent Rapamycin Dosing”

VDP-31 supports the funding of $50,000 USD for a clinical trial by Dr. Brad Stanfield on exercise and Rapamycin dosing. This study will look to establish if intermittent Rapamycin use can delay frailty and muscle mass loss.

VDP-32 Hyperspectral imaging for early diagnosis of Alzheimer’s Disease

VDP-32 financially supports Mantis Photonics’ camera technology for early screening of Alzheimer’s Disease. This camera technology uses the imaging of an individual’s eye to look for peptides associated with Alzheimer’s disease. Using the eye to look for neurological changes could provide an inexpensive and minimally invasive window into one’s central nervous system!

VDP-33 Better Together- A PrimeDAO Partnership

With VDP-33, we are embarking on a new partnership with PrimeDAO! This proposal initiates a partnership between PrimeDAO and VitaDAO through a token swap of $D2D and $VITA. The first of many DAO2DAO relationships! We hope this will encourage more DAOs to participate in VitaDAO and while also allowing VitaDAO to take part in other DAOs as well.

VDP-34 Defining VitaCore

VDP-34 proposal elucidates confusion and better defines VitaCore as consisting of four stakeholder groups who are accountable to VITA token holders. These four groups include Working Group Stewards and Co-Stewards, ambassadors of service providers, ambassadors of partner DAOs, and strategic contributors.

VDP-39 Longevity Hackers Film Participation

VDP-39 funds up to 100,000 USD to co-fund Longevity Hackers, a documentary by Filmmaker Michal Siewierski.This documentary will expand VitaDAO’s community reach through the production of an independent film to be released later this year!

VitaDAO in the Wild

CoinMarketCap posted a Youtube video defining and exploring Decentralized Science. Fast forward to 10:15 to see VitaDAO’s highlight!

Our friends at Lifespan posed and answered the question: “Why is VitaDAO a DAO and what does this mean in practice?”.

In an article about longevity research, the Methuselah Foundation gave VitaDAO a shoutout!

We have open roles!

If you’re a resourceful, proactive, well-organized, and dynamic person, curious about the world of longevity, crypto, and open source communities with a hunger for impact — we’re looking for you! Then one of these positions might be right for you! As someone involved with VitaDAO, you will have the chance to interact with members across all of the working groups of the DAO, and most importantly, inject good vibes into the DAO. Check out the links below for more information on specific open positions:

Upcoming Events

April 7th at 9 PM CET- You don’t want to miss VitaDAO’s first-ever Women of Vita Panel on Female Longevity! Head over to VitaDAO’s Twitter and join the Twitter Space to listen in on and contribute to a conversation about female reproductive longevity!

April 13th at 3:30PM CET — Join us for a full day of fun at a VitaDAO Crypto meets Longevity Symposium. The symposium is free of charge and will be live-streamed on Youtube and other social media platforms.

Sign up here: https://www.meetup.com/vitadao/events/285057395/

What’s Going DAOwn

Have questions, concerns, inquiries? Join the Discord channel and introduce yourself! VitaDAO is a friendly community and its Discord functions as the heartbeat of the DAO. Our operations team is great at onboarding and directing newcomers to roles that fit their skills.

Want to know more about VitaDAO’s funded projects? Check out the newly revamped VitaDAO website! Kudus to our Tech Working Group!

If you enjoyed reading this newsletter, make sure to subscribe! If you want some St. Patty’s day 🍀 good luck, share this newsletter with your luckiest friend! Until next month 😊

Hello Vitalians! Another month, another newsletter- and we couldn’t be more excited about it. Many of the core contributors to the DAO were able to meet up in sunny ☀️ Lisbon this month to organize, bond, and brainstorm in person.
Read more
VitaDAO x PrimeDAO partnership
March 25, 2022
Tokenomics
Governance
VitaDAO x PrimeDAO partnership

PrimeDAO is building tools that turn DeFi into a cooperative ecosystem. DAO2DAO products and services, such as DAO launchpads, deals, ratings, pools and much more. 

We want to work closely with Prime as a power user, to give input on which tools would be most useful to DAOs such as VitaDAO.  VitaDAO and PrimeDAO share a common purpose to push the abilities of DAOs to fund and advance human potential. 

VitaDAO and PrimeDAO exchanged $50k worth of VITA and $50k worth of D2D by 30-day average as of the end of the vote in both DAOs. 

Why? 

The partnership aims to align the interest of the parties to intensify the collaboration between the organisations.

PrimeDAO provides DAO tools which we want to further explore with VitaDAO, and provide input on which tools would be most useful for us and our ecosystem.

This also creates mutual awareness in both DAOs through the introduction of this proposal, as well as governance alignment of VitaDAO being able to govern PrimeDAO through its governance votes, and vice versa. 

Learn more about PrimeDAO on https://prime.xyz and make sure to follow them @PrimeDAO_ 

PrimeDAO is building tools that turn DeFi into a cooperative ecosystem.
Read more
Epigenetic Clocks: A Simple Lab Test Measuring Biological Age
March 22, 2022
Lindsay Ciocco
Longevity
Science
Epigenetic Clocks: A Simple Lab Test Measuring Biological Age

Increases in biological age correspond to the hindrances of aging, the concept symbolizing the overall health of an organism relative to its chronological age. In contrast to chronological aging, objective measurement of biological aging has been elusive: epigenetic clocks have subsequently emerged as innovative lab tests for quantifying aspects of biological aging.

What is an Epigenetic Clock?

Most commonly, the term epigenetic clock refers to a DNA methylation clock. These clocks track the presence of methyl groups, small molecules composed of carbon and hydrogen, at regions of DNA called CpG sites. Methylation patterns contribute to the regulation of gene expression and are known to shift in relation to environment, lifestyle, and aging. Researchers can use CpG methylation as a proxy to uncover patterns of gene expression in relation to aging or disease¹.

Another more complex example of epigenetic control of gene expression is histone modification. In histone modification, a few different types of chemical groups can attach to histone tails of DNA coils called nucleosomes; as such, gene expression is altered based on the type of chemical group and location of its attachment. Although histone modifications are also known to adjust with age, DNA CpG methylation is better studied in its relation to aging² ³.

Image By: Victoria Padure

What are the Differences Between Various Clocks?

Methylation clocks have been constructed in numerous ways, and multiple versions currently exist. Clocks can differ in the number and location of sites selected, tissue types sampled, and methods used for computation⁴. As such, they can diverge in displaying different aspects of aging, enabling them to be tailored for distinct purposes. Some clocks are connected tightly to chronological age, rendering them less useful for tracking biological age but valuable for use in forensics and animal conservation⁵ ⁶. Alternatively, they can be constructed to associate with traits of aging and disease ³.

Early clocks were primarily based on correlation with chronological age. One of the first generations of clocks developed was the Hannum Clock, developed by Gregory Hannum Ph.D. et. al, which identified 71 sites predictive of age. In this clock, sex and genetic differences were determined to be primary influencers of the aging rate, with men aging 4% faster on average⁷. However, many clocks have since been developed, sampling different sites and have found various novel elements associated with aging.

“As models of human aging improve, it is conceivable that biological age, as measured from molecular profiles, might one day supersede chronological age in the clinical evaluation and treatment of patients.”

- Hannum et. al (2013)

The Horvath Clock, perhaps one of the most well-known original clocks, was developed by Steve Horvath, Ph.D., Sc.D., a prominent researcher studying human genetics and biostatistics. It utilized 353 CpG sites from multiple tissues, finding an epigenetic age close to zero for embryonic cells and age acceleration in cancer cells⁸. Horvath has since developed other clocks, including GrimAge, which is said to be accurate in predicting mortality. PhenoAge, another clock developed by Horvath along with Morgan Levine, Ph.D., was trained on established blood biomarkers and has been found to be predictive of survival, healthspan, and physical function⁹.

Both Horvath and Levine continue to research methylation clocks. Horvath recently developed a clock based on changes in multiple tissues in several mammalian species, which suggests a component of aging is evolutionarily conserved¹⁰. Meanwhile, Levine created a meta-clock by combining similarities detected in different methylation clocks. This meta-clock revealed common patterns associated with decreased metabolism, immune function, increased chromatin modifications, and autophagy dysregulation⁴.

What are the Potential Applications of Methylation Clocks?

There is still much to be explored in the research of methylation clocks, but the most obvious application is biomarkers for evaluating aging interventions. This could allow the effectiveness of a given intervention to be determined within a time frame realistic for a clinical trial. Their utility also does not need to be limited to just aging; they could similarly be tailored for quantifying the efficacy of treatments for many diseases. This could further the aim of precision medicine, allowing therapies to be tested and tailored to the treatment response of an individual.

Is Rejuvenation by Resetting the Clock Possible?

There is speculation that epigenetic markers are not only useful for measuring aging but — akin to a stopwatch — their reset might turn back the clock, reversing significant cellular aspects of aging itself¹¹ ¹². Caution is presently warranted on this topic since the research supporting it in vivo is scarce; however, there is some evidence that the alteration of epigenetic markers is associated with the restoration of youthful attributes in cells. This is supported by studies investigating epigenetic reprogramming, which can return aged cells to a stem cell state, and partial reprogramming, which according to a few reports has resulted in rejuvenated tissues¹³ ¹⁴. Additionally, a few studies, with a limited number of participants, have reported reversal of epigenetic clocks in humans undergoing different sets of pharmaceutical or lifestyle interventions¹⁵ ¹⁶.

At the same time, there is still a debate over how reliable epigenetic clocks are, and large human trials to confirm these results do not exist. There is some variance in the epigenetic age of an individual between tissue types and even between cells from the same tissue sample. Likewise, even the exact same sample can vary on repeat assessment due to lack of consistency in DNA methylation measurement; one study found a range in variance of up to 3 to 9 years in a survey of 6 different clocks¹⁷ ¹⁸. Furthermore, there are still many questions about how exactly methylation patterns result in the cell and tissue changes associated with aging¹⁹.

What are the Considerations When Purchasing Tests for Biological Age?

Healthcare, like other systems within modern societies, is trending towards decentralization and personalization, with direct-to-consumer lab tests and wearable health trackers being prime examples. Along with this trend, larger numbers of people will likely aspire to track their own health using such methods, with quite a few “biological age” tests already commercially available. Home testing of biological age may empower people to take control of their own health, but also comes with important considerations.

Although several manufacturers claim that these tests can track “age-reversal” the current evidence does not entirely support this. Still, these tests might be worthwhile, allowing an individual to incrementally trial different lifestyle interventions to determine how much impact each has on the test result, ideally translating the benefits to improved health. For example, individuals could trial different diets (adjusting macronutrients, micronutrients, caloric intake, or fasting schedules) to see what impact they might have. Other interventions within reach could focus on the amount and type of exercise, hot or cold stressors, meditation, or sleep.

Like genetic tests or other lab assays, epigenetic testing constitutes personal health information and can inform much about disease risk and personal history.

A few issues should be considered when pursuing home biological age testing. Foremost, it would be best if the variance between tests or reliability is known and reported. A high degree of variation would render the test unreliable for trialing any interventions in plausible time frames; a test that varies in reliability by a few years would probably be meaningless. An increase in testing frequency could help balance out small amounts of variability and confirm a trend. However, most of the currently available tests are priced at a few hundred dollars per test, which could make testing on a regular basis unrealistic for many people.

Additionally, having some background knowledge on the aspects of aging captured by such tests would be helpful. This type of transparency would be useful both for interpreting the results and helping to expose biases. For example, a company could sell a test along with a supplement advertised to reduce aging. The supplement might have an impact on the test, giving the appearance of reversing age, but might not have any association with aging or only correlate with a single aspect of aging like inflammation. Although inflammation plays an important role in aging, sometimes referred to as “inflammaging”, other signals could be more ominous. We cannot rule out the possibility that a test may capture a signal shared by both rejuvenation and cancer growth, like cell proliferation or loss of cell type.

Consumers should also consider privacy in addition to reliability, cost, and value. Like genetic tests or other lab assays, epigenetic testing constitutes personal health information and can inform much about disease risk and personal history. For example, both smoking history and alcohol use have been ascertained in methylation studies²⁰ ²¹. Consumers will need to determine their level of risk tolerance for privacy when submitting such data. Understanding the testing methods, reliability, and privacy terms would aid consumers in determining if such testing is appropriate and useful to them.

Final Thoughts on Biological Aging Tests

Advances in defining and measuring biological age hold promise for detecting interventions with the greatest impact on improving health. While tests are currently available to consumers, they can be pricey, and questions remain about their reliability.

Laboratory research into epigenetic clocks for measuring biological age continues to progress and appears encouraging for exploring underlying mechanisms of aging and investigating the efficacy of therapies. Someday, this type of testing might even be added to traditional clinical biomarkers of health, aiding in the realization of precision preventative medicine.

DISCLAIMER: THIS ARTICLE DOES NOT PROVIDE MEDICAL ADVICE. No material in this article is intended to be a substitute for professional medical advice, diagnosis, or treatment. The text, images, and other material contained in this article are for informational purposes only.

Lindsay Ciocco is a practicing optometrist pursuing a Master of Arts in science writing at Johns Hopkins University. She earned a Bachelor of Science in evolution and ecology, and a Doctorate of Optometry from The Ohio State University. She has an interest in writing about biotechnology, especially advances for treating ocular disease.

Victoria Padure is an illustrator and graphic designer passionate about science and longevity. With her artwork, she hopes to bring more awareness around these fields and see them thriving

Ariella Coler-Reilly is an MD Ph.D. candidate at Washington University in St. Louis studying the genetics of aging. She moonlights as a science writer and illustrator, currently working as a managing editor for the VitaDAO blog. She is passionate about public education, diversity & inclusion, and the intersection of science & web3.

References

1. Salameh Y, Bejaoui Y, el Hajj N. DNA Methylation Biomarkers in Aging and Age-Related Diseases. Frontiers in Genetics. 2020;11:171. doi:10.3389/FGENE.2020.00171/BIBTEX

2. Yi SJ, Kim K. New Insights into the Role of Histone Changes in Aging. International Journal of Molecular Sciences. 2020;21(21):1–20. doi:10.3390/IJMS21218241

3. Bergsma T, Rogaeva E. DNA Methylation Clocks and Their Predictive Capacity for Aging Phenotypes and Healthspan. Neuroscience Insights. 2020;15. doi:10.1177/2633105520942221

4. Liu Z, Leung D, Thrush K, et al. Underlying features of epigenetic aging clocks in vivo and in vitro. Aging Cell. 2020;19(10):e13229. doi:10.1111/ACEL.13229

5. Montesanto A, D’Aquila P, Lagani V, et al. A New Robust Epigenetic Model for Forensic Age Prediction. Journal of Forensic Sciences. 2020;65(5):1424–1431. doi:10.1111/1556–4029.14460

6. Barratclough A, Smith CR, Gomez FM, et al. Accurate Epigenetic Aging in Bottlenose Dolphins (Tursiops truncatus), an Essential Step in the Conservation of at-Risk Dolphins. J Zool Bot Gard. 2021;2021:416–420. doi:10.3390/jzbg2030030

7. Hannum G, Guinney J, Zhao L, et al. Genome-wide Methylation Profiles Reveal Quantitative Views of Human Aging Rates. Molecular Cell. 2013;49(2):359–367. doi:10.1016/J.MOLCEL.2012.10.016

8. Horvath S. DNA methylation age of human tissues and cell types. Genome Biology. 2013;14(10):R115. doi:10.1186/GB-2013–14–10-R115

9. Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging for lifespan and healthspan. AGING. 2018;10(4). Accessed March 11, 2022. www.aging-us.com

10. CONSORTIUM MM, Lu AT, Fei Z, et al. Universal DNA methylation age across mammalian tissues. bioRxiv. Published online January 19, 2021:2021.01.18.426733. doi:10.1101/2021.01.18.426733

11. Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: Resetting the aging clock. Cell. 2012;148(1–2):46–57. doi:10.1016/j.cell.2012.01.003

12. Noroozi R, Ghafouri-Fard S, Pisarek A, et al. DNA methylation-based age clocks: From age prediction to age reversion. Ageing Research Reviews. 2021;68:101314. doi:10.1016/J.ARR.2021.101314

13. Lu Y, Brommer B, Tian X, et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature. 2020;588(7836):124–129. doi:10.1038/s41586–020–2975–4

14. Wang C, Rabadan Ros R, Martinez-Redondo P, et al. In vivo partial reprogramming of myofibers promotes muscle regeneration by remodeling the stem cell niche. doi:10.1038/s41467–021–23353-z

15. Fahy GM, Brooke RT, Watson JP, et al. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell. 2019;18(6):e13028. doi:10.1111/ACEL.13028

16. Fitzgerald KN, Hodges R, Hanes D, et al. Potential reversal of epigenetic age using a diet and lifestyle intervention: a pilot randomized clinical trial. Aging. 2021;13(7):9419–9432. doi:10.18632/aging.202913

17. Sugden K, Hannon EJ, Arseneault L, et al. Patterns of Reliability: Assessing the Reproducibility and Integrity of DNA Methylation Measurement. Patterns. 2020;1(2). doi:10.1016/J.PATTER.2020.100014/ATTACHMENT/A01126CF-8D98–4563–9807–6641905A764B/MMC2.XLSX

18. Higgins-Chen AT, Thrush KL, Wang Y, et al. A computational solution for bolstering reliability of epigenetic clocks: Implications for clinical trials and longitudinal tracking. bioRxiv. Published online April 19, 2021:2021.04.16.440205. doi:10.1101/2021.04.16.440205

19. Unnikrishnan A, Freeman WM, Jackson J, Wren JD, Porter H, Richardson A. The role of DNA methylation in epigenetics of aging. Pharmacology and Therapeutics. 2019;195:172–185. doi:10.1016/J.PHARMTHERA.2018.11.001

20. Maas SCE, Vidaki A, Wilson R, et al. Validated inference of smoking habits from blood with a finite DNA methylation marker set. European Journal of Epidemiology. 2019;34(11):1055. doi:10.1007/S10654–019–00555-W

21. Liu C, Marioni RE, Hedman AK, et al. A DNA methylation biomarker of alcohol consumption. Molecular Psychiatry 2018 23:2. 2016;23(2):422–433. doi:10.1038/mp.2016.192

Humans can instinctively recognize if someone appears older or younger than their age; so then, it should be apparent that many species, including humans, develop age-related changes at different rates.
Read more
VitaDAO Newsletter Issue No 3 March 2022
March 3, 2022
Sarah Friday & Hossam Zaki
Awareness
Newsletters
VitaDAO Newsletter Issue No 3 March 2022

We know we say it every month, but the VitaDAO community has been busy. We’ve been told to shorten these newsletters, but in all honesty, all that happened in VitaDAO this month makes that difficult! February was packed full of journal clubs, new medium articles, active votes, and a growing community. In this newsletter, you can find recaps of February’s important events, updates on votes happening in the DAO, and upcoming events.

New here?

Welcome👋 TLDR: VitaDAO is a decentralized organization made up of individuals from across the globe🌎 working to fund early-stage longevity research. At its core, VitaDAO functions as an ecosystem with three moving parts: the DAO, the researchers, and their IP. Check out the VitaDAO Whitepaper for more information on VitaDAO governance. Learn more about IP-NFTs in this article written by Molecule here (and see an example of an IP-NFT here!). Want to purchase $VITA’s governance token? Read through the VitaDAO guide on how to purchase $VITA.

Community News

$VITA Goes Gnosis Chain

We added $VITA liquidity pools on Gnosis Chain! This will allow you to buy $VITA with decreased gas fees. Gnosis Chain is now the network with the 2nd largest liquidity pool for $VITA! Find a tutorial on how to buy $VITA on the Gnosis chain here.

Not Just Any Prize, The Longevity Prize!!

VitaDAO and the Foresight institute have launched the Longevity Prize (https://twitter.com/longevityprize)! This is a crowdsourced prize funded through the Gitcoin community and matched by Vitalik Buterin👽 to fund longevity research projects. In the upcoming months, roughly $185k donated will be distributed to projects advancing longevity research!

First Round of VitaDAO Fellows Selected!

The VitaDAO fellowship provides funding to individuals who want to get more deeply involved in longevity research📚. In the past month, VitaDAO has funded the first round of VitaDAO fellows to attend conferences, engage in programs like On Deck Longevity, and kickstart longevity projects. Interested in being included in the second round of awards? Apply here: https://www.vitadao.com/fellowship.

New Partnership Between Molecule, Apollo, and VitaDAO

Molecule and VitaDAO have partnered with Apollo Health Ventures, a leading longevity-focused VC fund. We hope that this partnership will allow us to help more longevity scientists turn their ideas into real-world applications. We are excited to introduce a traditional VC firm to crypto/web3, continuing to revolutionize the financing of drug development.

say hello to FRENS

VitaDAO has developed the FRENS framework for fractionalizing IP-NFTs via sublicenses. FRENS, which stands for Fair, Reasonable, Ethical, Nondiscriminatory Sublicense, aims to make drug development more like software development and distribute power into the hands of the community. Read specific details on FRENS on the VitaDAO medium.

Community Plug🔌 : Women of VitaDAO

Are you a woman who is interested in meeting other women involved in VitaDAO? VitaDAO is excited to have a new women-only Discord channel where women in VitaDAO can provide support, feedback, and attend community calls with other women in the DAO community! We love to see #Web3WomeninScience supporting #Web3WomeninScience 🤝

Scientific Publishing

Watch hosts Jocelynn Pearl’s and Vincent Weisser’s conversation with Ben Hills and Darren Zhu discussing the power of Web3 to revolutionize scientific publishing. This podcast explores how web3 can decrease the cost and barriers of traditional scientific publishing, and also touches upon proposals of future diversification of research models and means of research financing. VitaDAO x UltraRare collaborative podcasts feature conversations at the intersection of science and web3. Check out the podcast here.

Panel Discussion: Long Live the Worm- Studying Aging with Invertebrates

Learn about the use of creepy-crawly 🪱🪱 invertebrates in the study of longevity with speakers Prof. Dr. Björn Schumacher, Prof. Jan Gruber, Prof. Eugene Berezikov, and host Max Unfried.

https://www.youtube.com/watch?v=9EOpATmuvyY

Introducing…. LabDAO!!!

We are so happy to support a new arm of the DeSci ecosystem, LabDAO. LabDAO was created by our great fren Niklas Rindtorff as an idea sprouting from the previously existing #decentralized biotech VitaDAO Discord channel. LabDAO aims to be the AWS of science.To learn more about LabDAO, check out this video of Niklas presenting LabDAO at EthDenver, as well as their Twitter. Expect more updates about them soon!

Journal Club: Can Blood from Youngsters Make You Young

Listen to a Twitter Space recording discussing heterochronic parabiosis, an old scientific technique where the circulatory systems of two animals of different ages are connected. This journal club examines if the benefits of “young blood” in an old animal remain if the old animal stops receiving young blood. In an interesting figure of the discussed Zhang et al paper, gene expression changes due to heterochronic parabiosis are found to be similar to gene expression changes seen with other better known life-extending interventions like calorie restriction!

Journal Club: Doggos, Doggerinos & Their Lifespan

Interested in learning about the use of dogs as model organisms for longevity research? Have no fear! You can play a Twitterspace recording of the journal club and learn about a new insulin-like growth factor 1 (IGF-1) variant found in dogs. Inhibition of the IGF-1 pathway is one of the most reliable life-extending interventions in lab animals. This paper explores the finding that the newly discovered IGF-1 variant predicts dog size.

Never want to miss a journal club? Subscribe here.

Article: The Science Behind NAD-Boosting & Anti-Aging

Author Maria Marinova recently published an article on NAD+. This article explores the interaction of NAD+ with sirtuins, changes in NAD+ levels with aging, potential benefits to increasing NAD+, and methods of raising NAD+. Our humble review of the article: “Very informative! 10/10 recommend 👏🏻”.

✅We Voted✅

A Third Research Project Funded?! (VDP-21)

The Evandro Fang lab from the University of Oslo will be the third research lab to have a project funded by VitaDAO and IP-NFTs! 🎉 The lab is aiming to identify and characterize new drug candidates for activation of mitophagy with a positive impact on Alzheimer’s disease by investigation of underlying molecular mechanisms.

Forming an IP Holding company! (VDP-23)

VitaDAO voted to form an IP holding company for 2 reasons: to empower VitaDAO to hold existing soft IP (copyrights, logo trademarks) and to empower VitaDAO to hold future patents related to IP-NFTs. It is important to note that the IP holding company is formed only to hold IP which it cannot, for one regulatory reason or another, hold as IP-NFTs. Generally speaking, having an IP holding company will enable VitaDAO to obtain stronger legal protections for its IP and, crucially, more easily bring lawsuits against violators when needed.

Special-Purpose Vehicle (SPV) as a Bridge for Strategic Contributors! (VDP-24)

VitaDAO voted to create a special-purpose vehicle (SPV) to allow institutional actors such as pharmaceutical companies and funds to participate in governance of VitaDAO through a corporate agent. The SPV will be a Swiss association and will hold $VITA, allowing its shareholders to vote on VDPs. The SPV will not be required to vote as a single block. Rather, its members will be able to vote with their pro-rata portions of $VITA held in the SPV.

Open For V-V-V-V-Voting

Funding for Gitcoin Quadratic Longevity Round and VitaDAO Fellowship (VDP-30)

VDP-30 proposes the donation of $40,000 USDC to match donations for longevity research initiatives in Gitcoin’s March 2022 quadratic donation round and $15,000 USDC to fund the VitaDAO Fellowship program. Peruse the proposal and vote on Snapshot.(No tokens needed to vote!)

VitaDAO in the Wild

With DeSci trending, VitaDAO has been talked about and typed about by many members of the crypto community in February. Below, find a few times where VitaDAO was plugged by others in the crypto-community.

Upcoming Events

March 4th at 6 PM CET- Join a security law workshop run by DAO member Jesse. This workshop has been created as a result of conversation sparked with VDP-27. Hop on and learn more about how VitaDAO is moving forward with funding equity and IP-NFT deals.

March 9th at 9 PM CET (Recurring Every Wednesday) — Pop into VitaDAO’s weekly onboarding session on Discord with Niklas Rindtorff and Alex Dobrin. Join the VitaDAO Discord Server to learn more about the DAO, how to get involved, have any lingering questions answered, and find schedules for specific working group meetings.

Interested in meeting other members from VitaDAO in person? Make sure to check out the #socialize-vita channel on Discord.

April 13th — There is a proposed VitaDAO Symposium occurring on April 13th, 2022. See event details, vote on the event proposal, or comment any questions on the VitaDAO Discourse.

Until Next Time

Between funding a third research project, bridging $VITA to the Gnosis chain, and publishing lots of new scientific communication content, VitaDAO had a very busy February. In the upcoming month, be on the lookout for more information on opportunities to meet other DAO members at global conferences, new proposals to vote on, and interesting research projects to fund..

We hope you enjoyed this month’s newsletter as much as we enjoyed writing it 😊. If so, we challenge you to subscribe and forward this Newsletter to a friend 💛.

151


We know we say it every month, but the VitaDAO community has been busy. We’ve been told to shorten these newsletters, but in all honesty, all that happened in VitaDAO this month makes that difficult!
Read more
$VITA goes Gnosis Chain
February 12, 2022
Tyler Green
Tokenomics
Governance
$VITA goes Gnosis Chain

Part 1: Get started on Gnosis Chain

  1. Get a wallet that is compatible with Gnosis Chain, such as MetaMask, Coinbase Wallet, Status, a hardware wallet (via MetaMask or WalletConnect) or a Gnosis Safe.
  2. Add Gnosis Chain to your wallet. If you are using MetaMask, just navigate to Chainlist, connect your wallet, scroll down to Gnosis Chain (ID: 100) and click on “Add to MetaMask”.
  3. Add the addresses of tokens that you want to transact with to your wallet (that is VITA). If you are using MetaMask, simply open the corresponding page on Blockscout, find the line where it says “Contract” and click on the MetaMask icon on the right-hand side. Here is the link for VITA. XDAI is added by default.

Part 2a: Get VITA on Gnosis Chain with Fiat

  1. Use Ramp Network to buy XDAI by following this guide and pay in EUR, USD or GBP via credit card or bank transfer.
  2. Exchange XDAI for VITA by swapping the desired amount on Honeyswap.

Part 2b: Get VITA on Gnosis Chain with Mainnet Tokens

  1. Swap your tokens for DAI on Cowswap. This only applies if you are using Ethereum Mainnet assets other than DAI, such as ETH.
  2. Convert Mainnet DAI to Gnosis Chain XDAI by using xDai Bridge following this guide.
  3. Exchange XDAI for VITA by swapping the desired amount on Honeyswap.

Note: only get VITA for the purpose of using it in VitaDAO.

Part 2c: Bridge Mainnet VITA to Gnosis Chain

  1. Convert Mainnet VITA to Gnosis Chain VITA by using OmniBridge following this guide.

Note on XDAI for gas: Each of the options in Part 2 should provide you with at least some XDAI tokens which are being used to pay for transaction fees (gas) on Gnosis Chain. Just 0.01 XDAI (equivalent to 0.01 USD) will get you a long way. While it is obvious in Part 2a and 2b, Part 2c should provide you with 0.01 XDAI sponsored by OmniBridge. If you, for any reason, do not have XDAI in your wallet and struggle to obtain some, try using a faucet, ask a friend or each out on Discord in the #token-talk channel and post your Gnosis Chain public key/wallet address — someone will be there to help you out.

Part 3: Use your VITA on Gnosis Chain

Governance: We are working on allowing VITA tokens on Gnosis Chain to be used for voting on Snapshot. Stay tuned for news on our Discord.

Providing liquidity: If you would like to use your VITA tokens to provide liquidity, see this guide for an explainer on liquidity pools on Honeyswap.

DISCLAIMER: This guide is intended only for people who want to get VITA for use in VitaDAO.

Resources


Save gas fees by using Gnosis Chain (formerly xDai Chain) — DISCLAIMER: This guide is intended only for people who want to get VITA for use in VitaDAO. Part 1: Get started...
Read more
The Science Behind NAD-Boosting & Anti-Aging
February 11, 2022
Maria Marinova
Science
Awareness
The Science Behind NAD-Boosting & Anti-Aging

If you have been following the longevity field for even just a little a while, you have probably heard of NAD+… a lot. So what is NAD+, and why are people excited about it?

The Importance of NAD+ in Cellular Health

Nicotinamide adenine dinucleotide or NAD+ is essential for the fundamental biological processes of cellular health like energy production, metabolism, DNA repair, mitochondrial function, and more. NAD+ is vital for glycolysis, the citric acid cycle, and the electron transport chain, which is how the cells produce their energy. Moreover, NAD+ plays an important role in deacetylation, which is the removal of a specific molecular fragment called an acetyl group, driving numerous metabolic processes [1].

NAD+ is a Helper Molecule for Sirtuins and PARPs

NAD+ is a cofactor, or a “helper molecule,” for a large class of deacetylases called sirtuins [2]. These enzymes require a non-protein chemical cofactor like NAD+ to function. The effects of sirtuins go well beyond metabolism. They play key roles in inflammation, transcription of genes, programmed cell death, and — of course — aging. Different sirtuins are localized differently within the cell and have varying targets [3]: DNA modulation, energy homeostasis, and cell cycle control, just to name a few.

The sirtuins residing in the nucleus of the cell function as histone deacetylases and are vital for genome integrity and stability. Histones are proteins that DNA wraps around such that only the necessary bits are accessible and the rest is safely packed away. If there is a lack of NAD+, their function is reduced [4], the DNA loosens, and the histones are exposed to damaging external agents. Protecting genome integrity is essential for every cell to maintain its regular functions and characteristics.

Image by Dani Bergey

Another important family of proteins that require NAD+ are the DNA repairing proteins called PARPs [5] (poly (ADP-ribose) polymerase). DNA damage can accumulate in our cells as we age, and our cells try to do everything in their power to protect their DNA since it is the code book that regulates all life. More damage collected overtime would also require more repair, which is a problem in itself.

PARPs similar to sirtuins can also play a role in protein post translational modifications [6] via adding ADP-ribose units to certain proteins [7], a process called parylation. At the moment, much less is understood about how parylation impacts gene expression and protein function than acetylation/deacetylation, but it is expected that this part of the NAD+ field will grow rapidly in the coming years.

NAD+ Levels Decline with Age

Our genome guardians — the PARPs — use NAD+ in large amounts. As we age, there is a lot of DNA damage, which can start to exhaust the NAD+ supplies. This could be one reason why we see NAD+ decline as life progresses. Another reason is a well-known NAD+ consumer enzyme called CD38 [8]. We know that inhibiting CD38 [9] leads to an increase in available NAD+.

We find ourselves in this cellular Catch-22: there’s more DNA damage as we age, and we need NAD+ to repair it, but there is less NAD+ available with age, and we exhaust it even further when we repair DNA.

The availability and balance of NAD+ within the cell are crucial for health, and they are dependent on the rate of degradation and production of the molecule. Recent studies [10] show that NAD+ production is maintained with age but is used up more quickly as animals get older, leading to a 30% median decline in the tissues of 25-month-old mice (approximately equivalent to 80-year-old humans). While the mysteries behind this age-related decline are not yet completely understood, PARPs and CD38 are the main suspects.

This drop in NAD+ levels has proven to have detrimental consequences throughout the body [11] observed, across the brain, liver, muscles, vasculature, heart, kidney, immune and reproductive systems. A lot of the decline has been shown in studies to be reversible. We know we can fight the dropping levels by supplying the organism with NAD+ precursor molecules. This way we tip the scales in favor of NAD+ production and offset the increased degradation.

Potential Benefits of Boosting NAD+ Levels

Benefits from NAD+ boosting supplements may be seen across various systems in the body. Administration of NAD+ precursors has the potential to reverse neural degeneration [12], improve synaptic plasticity and memory [13] in Alzheimer disease animal models, and delay the senescence [14] of neural stem cells in old mice. In the liver, boosting NAD+ levels might be useful to prevent or treat diseases such as alcoholic and non-alcoholic steatohepatitis (NASH) [15], obesity [16], and more, as the evidence from animal studies suggest. In the muscles, human studies suggest it could be helpful against sarcopenia [17] (muscle wasting and weakness) as it appears to aid mitochondrial function, energy ATP production, inflammation reduction, and ultimately results in functional benefits such as increased endurance for running.

In the vascular systems [18] of multiple animal models, an increase of NAD+ levels seems to be effective at improving blood flow, restoring density of the capillary network and improving endothelial function for artery stiffness. In one large study on elderly mice, an NAD+ precursor supplementation showed a promising increase in blood flow, capillary density, and endurance [19]. In the heart [20] itself, boosting NAD+ has been shown to attenuate the progression of heart failure and specifically cardiomyopathy in mouse models.

A study suggests that NAD+ boosters could also act like anti-inflammatory agents, as they inhibit the expression of inflammatory markers and reduce chronic low-grade inflammation [21] (known as inflammaging) in mice. This decrease of inflammation, especially in certain tissues like the adipose tissue (fat), could mean a reduction in insulin resistance and obesity [22].

Lastly, beneficial effects are seen in the fastest aging system — the female reproductive system [23] — where NAD+ booster supplementation improves oocyte quality [24], ovulation rate, and as a functional outcome, the number of live births in aged mice.

Raising NAD+ Levels with Diet and Exercise

Some non-pharmacological ways to increase your NAD+ are diet and exercise [25]. Aerobic and resistance training for the duration of 12 weeks both were shown to ameliorate the age-induced NAD+ decline by increasing levels of the enzyme nicotinamide phosphoribosyltransferase (NAMPT.). These regimes also improved functional health measurements such as BMI, lean body mass, and glucose infusion rate.

NAD+ is synthesized via three different pathways from nicotinic acid, tryptophan, or precursors like NMN (nicotinamide mononucleotide) and NR (nicotinamide riboside). These molecules are bioavailable, and consuming a healthy balanced diet can provide a sufficient amount for a young person. Foods particularly rich in NAD+ precursors are milk, fish, yeast, green vegetables and whole grains. This, however, might be insufficient to battle age-related decline.

When compared to other bioavailable precursors like NA (nicotinic acid) and NAM (nicotinamide), NR is by far the most efficient. But how does it compare to NMN — the rising star in the NAD+ field?

Raising NAD+ Levels with Supplements: NMN vs NR

Both NMN and NR are available as supplements, and it is often debated which one is more effective. The current debate on whether one is better than the other is based on two main points: which one is more readily taken up by the cells and which one is more efficiently utilized.

One main difference is that NMN is a bigger molecule with an extra phosphate group, which means it is harder for NMN to enter the cell. NMN is the one, however, which directly produces NAD+. On the other hand, NR more easily enters the cell, but then it needs to be converted to NMN before it becomes NAD+.

Some studies [26] show that NMN needs to be converted to NR in order to cross the cell membrane, at which point the NR is converted back to NMN and subsequently to NAD+. Recently, a new NMN transporter [27] was found in the gut of mice, suggesting NMN can be directly transported into cells after all. According to genomic datasets, this transporter is also expressed in humans [28]. Its expression levels vary between different tissues, and closer study is required to determine what portion of NMN, if any, passes through that transporter in humans. The evidence that this transporter is actually trafficking NMN has been disputed [29].

Other studies propose that NMN has to become NR only to enter certain types of cells [30], while it is readily accepted by others. It has been seen to enter the mouse small intestine as is, even with the bulky phosphate group. We still do not know how this translates to humans.

While there are differences, there are also many similarities. As described above, there are numerous age-related health conditions in which NAD+ levels are reportedly low. In animal models, supplementing with NMN or NR seems alleviate these disease states [31]. Mostly, the end results are quite comparable in animals, though more studies are needed in humans. Both molecules and their derivatives are in clinical trials for several diseases, such as heart failure [32], sarcopenia (muscle wasting) [33], insulin resistance [34], and kidney disease, [35] to name a few, and so far have been found to be safe in humans [36].

Consumer Caution and Due Diligence

As with any other bioactive molecule, NMN and NR must be administered with caution and precision, as high levels might have potentially harmful effects. For example, an increased ratio of NMN to NAD+ was shown to activate a metabolic sensor in neurons associated with axon degeneration [37]. Another study showed that NMN enhances the inflammatory environment [38] in a mouse cancer model and that proinflammatory pathways can be induced by NAD+ metabolism.

Importantly, none of those negative effects have been observed yet in human clinical trials. Moreover, as described above, other studies have suggested the exact opposite: that NAD+ supplementation can fight neural degeneration and promote anti-inflammatory effects. Still, it is important that consumers be aware the science behind NAD+ supplementation is still actively under investigation.

Both NMN and NR are available for purchase as dietary supplements. It is important to note that regulations on supplements are very lax compared with pharmaceuticals, so consumers must exercise caution and due diligence when purchasing. For example, a recent report [39] showed that a number of NMN supplements sold on Amazon contained a concentration of NMN far below the one indicated on the label, and many had no detectable NMN whatsoever. This of course is not evidence against NMN itself, but rather evidence of unreliable retailers. It should be noted that this report was initiated by a competitor brand selling NR, and NR supplements were not tested. We will focus on considerations for supplement quality in an upcoming piece, since this is a hot topic in the community.

Final Thoughts

In closing, there is a wealth of data suggesting that healthspan can be extended by maintaining youthful NAD+ levels in the body. Studies have explored this over a number of preclinical models, and we are expecting data from human clinical trials very soon. We hope further studies will help us determine which supplement is the superior NAD+ booster and how it is best utilized in people.

DISCLAIMER: THIS ARTICLE DOES NOT PROVIDE MEDICAL ADVICE. No material in this article is intended to be a substitute for professional medical advice, diagnosis or treatment. The text, images and other material contained in this article are for informational purposes only.

Maria Marinova is a molecular biology graduate and a PhD candidate in reproductive medicine (OBGN). Her work focuses on ovarian damage caused by aging or chemotherapy and exploring the potential of NAD+ boosting molecules on protecting against or reversing these negative effects. Her interests extend beyond the ovary and towards extending women’s healthspan and lifespan by means of delaying menopause or preventing premature menopause. Some of her work on this topic has been published (co-author) in Cell Reports demonstrating how NAD+ repletion extends fertility in aged animals.

Dani Bergey, MS is a professional medical illustrator based in Seattle, WA, USA. Her focus is in the strategic storytelling and visualization of cell and molecular biology. She has an academic background in neurobiology, mathematics, medical illustration, and multimedia art; and she is a founding member of UltraRare, a web3 collective at the intersection of science and art.

Ariella Coler-Reilly is an MD PhD candidate at Washington University in St. Louis studying the genetics of aging. She moonlights as a science writer and illustrator, currently working as a managing editor for the VitaDAO blog. She is passionate about public education, diversity & inclusion, and the intersection of science & web3.

References

[1] Zhao et al. Regulation of cellular metabolism by protein lysine acetylation. Science. 2010

[2] Satoh et al. The Role of Mammalian Sirtuins in the Regulation of Metabolism, Aging, and Longevity Handb Exp Pharmacol. 2013

[3] Yamamoto et al. Sirtuin Functions in Health and Disease. Molecular Endocrinology. 2007

[4] Imai et al. It takes two to tango: NAD+ and sirtuins in aging/longevity control. Aging and Mechanisms of Disease. 2016

[5] Chaudhuri and Nussenzweig. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nature Reviews Molecular Cell Biology. 2017

[6] Ryu et al. Metabolic regulation of transcription through compartmentalized NAD+ biosynthesis. 2018. Science

[7] Gupta et al. PARPs and ADP-Ribosylation: recent advances linking molecular functions to biological outcomes. Genes Dev 2017

[8] Wu and Zhang. CD38-expressing macrophages drive age-related NAD+ decline. Nature Metabolism

[9] Tarrago et al. A Potent and Specific CD38 Inhibitor Ameliorates Age-Related Metabolic Dysfunction by Reversing Tissue NAD+ Decline. 2018. Cell Metab

[10] McReynolds et al. NAD + flux is maintained in aged mice despite lower tissue concentrations. Cell Systems. 2021

[11] Zapata-Perez et al. NAD+ homeostasis in human health and disease. EMBO Mol Med. 2021

[12] Hikosaka et al. Implications of NAD metabolism in pathophysiology and therapeutics for neurodegenerative diseases, Nutritional Neuroscience. 2019

[13] Lautrup at al. NAD+ in Brain Aging and Neurodegenerative Disorders. 2019. Cell Metab

[14] Zhang et al. NAD⁺ repletion improves mitochondrial and stem cell function and enhances life span in mice. 2016. Science

[15] Gariani et al. Eliciting the mitochondrial unfolded protein response by nicotinamide adenine dinucleotide repletion reverses fatty liver disease in mice. Hepatology. 2016

[16] Canto et al. The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high‐fat diet‐induced obesity. Cell Metab. 2012

[17] Migliavacca et al. Mitochondrial oxidative capacity and NAD + biosynthesis are reduced in human sarcopenia across ethnicities. Nat Commun. 2019

[18] Jahan and Bagchi. Enhancing NAD+ Metabolome in Cardiovascular Diseases: Promises and Considerations. Front Cardiovasc Med. 2021

[19] Das et al. Impairment of an Endothelial NAD +-H 2 S Signaling Network Is a Reversible Cause of Vascular Aging. 2019. Cell

[20] Diguet et al. Nicotinamide Riboside Preserves Cardiac Function in a Mouse Model of Dilated Cardiomyopathy. Circulation. 2017

[21] Mills et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 2016

[22] Berg and Scherer. Adipose tissue, inflammation, and cardiovascular disease. 2005. Circ Res

[23] Bertoldo et al. NAD+ repletion rescues female fertility during reproductive ageing. Cell Rep. 2020

[24] Miao et al. Nicotinamide Mononucleotide Supplementation Reverses the Declining Quality of Maternally Aged Oocytes. 2020. Cell Reports

[25] De Guia et al. Aerobic and resistance exercise training reverses age-dependent decline in NAD+ salvage capacity in human skeletal muscle. Physiol Rep. 2019

[26] Ratajczak et al. NRK1 controls nicotinamide mononucleotide and nicotinamide riboside metabolism in mammalian cells. Nature Comm. 2016

[27] Grozio et al. Slc12a8 is a nicotinamide mononucleotide transporter. Nat Metab. 2019

[28] SLC12A8 solute carrier family 12 member 8 [ Homo sapiens (human) ] Gene ID: 84561

[29] Schmidt and Brenner. Absence of evidence that Slc12a8 encodes a nicotinamide mononucleotide transporter. 2019. Matters Arising

[30] Formentini et al. Detection and pharmacological modulation of nicotinamide mononucleotide (NMN) in vitro and in vivo. Biochem Pharmacol. 2009

[31] Yoshino et al. NAD+ Intermediates: The Biology and Therapeutic Potential of NMN and NR. Cell Metab. 2018

[32] Obrien et al. (2016, May — 2019, June). Nicotinamide Riboside in Systolic Heart Failure. Identifier: NCT03423342

[33] Seldeen et al.(Ongoing 2021). Impacts of Nicotinamide Riboside on Functional Capacity and Muscle Physiology in Older Veterans (NR-VET). Identifier: NCT04691986

[34] Schrauwen et al. (2016, July — 2019, March) Nicotinamide Riboside in Metabolic Health. Identifier: NCT02835664

[35] Kestenbaum and Roshanravan. (2018, July — 2021, November). Trial of Nicotinamide Riboside and Co-enzyme Q10 in Chronic Kidney Disease (CoNR). Identifier: NCT03579693

[36] Irie et al. Effect of oral administration of nicotinamide mononucleotide on clinical parameters and nicotinamide metabolite levels in healthy Japanese men. Endocr J. 2020

[37] Figley et al. SARM1 is a metabolic sensor activated by an increased NMN/NAD+ ratio to trigger axon degeneration. 2021. Neuron

[38] Nacarelli et al. NAD+ metabolism governs the proinflammatory senescence-associated secretome. Nat Cell Bio. 2019

[39] ChromaDex. Quantitative Analysis of Twenty-Two NMN Consumer Products. 2021

If you have been following the longevity field for even just a little a while, you have probably heard of NAD+… a lot. So what is NAD+, and why are people excited about it?
Read more
VitaDAO Newsletter Issue No 2 Feb 2022
February 1, 2022
Sarah Friday & Hossam Zaki
Awareness
Newsletters
VitaDAO Newsletter Issue No 2 Feb 2022

The VitaDAO community quickly got to work after watching the ball drop. In this January edition of the VitaDAO Newsletter, you can learn about VitaDAO proposals that passed over the last month, get to know DAO rockstar Alex Dobrin, and more!

New Year, new you? What we can say with confidence is — New Year, same fantastic VitaDAO. Decentralization of science is taking off and VitaDAO is here to fund more science and spread good vibes while doing so 🥳. We knew we were reaching the masses when John Cena followed our Twitter (as of now, this isn’t a joke). As we roundup the VitaDAO 2021 and highlight some upcoming events, remember to Join the Discord and hop into one of our orientation “Ask Me Anything” calls. You can find links to the Discourse, Discord, and Twitter here.

Community News

2021 was an incredible year for VitaDAO filled with milestones, exciting developments within the longevity space, and vibrant growth within the community. Below is a summary of notable events that happened in the community during 2021, notable Governance measures, and impactful discussions going on right now!

2021 in Review

It is hard to believe VitaDAO’s Gnosis auction ended just over six months ago on June 23, 2021. In the months since then, our community has been growing and gaining traction and recognition within the crypto community. As of January 2022, VitaDAO has:

  • Funded 2 IP-NFTs
  • Evaluated 35+ Research Projects evaluated
  • Communicated with over 150 researchers/ potential projects
  • Funded $1.5 million worth of longevity research!!
  • 4000+ Discord Members
  • 8 Very Active Working Groups
  • 80+ Working Group Contributors
  • Published 17 Medium Articles
  • Published 31 Youtube Videos
  • Gained over 9000 Followers on Twitter

VitaDAO is always looking for new cutting-edge longevity research projects to fund. In fact, we have a bounty out until February 15 for 500 U.S. dollars or 500 VITA for each relevant project you introduce us to! Click here to submit a project for consideration.

Voting is now LIVE on Snapshot

In great addition to our current system, Tier 3 Governance has moved to Snapshot, making voting completely gasless aka FREE🤑. This will encourage more participation among $VITA holders and reduce the friction associated with voting. If you want to vote, make sure you un-stake your $VITA! Click here to vote.

Note: this doesn’t replace our current Tier 1 and Tier 2 governance. Tier 1 and Tier 2 will still occur on Discord and Discourse, respectively.

VitaDAO Liquidity Pools on L2

On the topic of gas fees, there will now be VitaDAO Liquidity Pools on L2! We are starting with bridges to Polygon and GnosisChain, the most used L2s. In the future, we may potentially expand to Arbitrum, Fantom, Optimism, Zksync, and others. This will allow for cheaper ways to participate in VITA and its governance. Be on the lookout for announcements coming up about this, and links to swaps! Check out VDP-20 for more info!

Additional 10% Token Mint for Aligned Partners

The passing of VDP-11 allows VitaDAO to mint an additional 10% of tokens for treasury and strategic contributors. VitaDAO has been approached by many biotech and web3 entities who have previously been unable to purchase VITA on DEXs at the volume they’d require. By growing the DAO treasury, we will be able to include these entities who can bring value to VitaDAO. Check out the results on VDP-11 here.

Now that we’ve decided to mint more tokens, what happens next? Follow the continued discussions on VDP-11 here.

New Steward Onboarding/Offboarding Process Approved

Working groups are the backbone of VitaDAO. They work behind the scenes to make the DAO as best as it can be (not that it’s not already great☺). Each working group is led by one or two stewards. We haven’t previously formalized the process of onboarding one… until now!!!

Now, a steward is nominated by another steward and is voted in by working group members. When a steward’s time is done, there are a few ways a steward can be offboarded. They can choose to leave with a two weeks notice or the community can vote for a change of leadership.

Check out the results of VDP-19 here.

Introducing the VitaDAO Fellowship!

VitaDAO is funding individuals with need-based grants to enable one to dive deeper in longevity (amounts starting from $100 to $3k). The grants can fund your research, allow you to take a break to explore longevity, attend a conference, or join a program like On Deck Longevity. To learn more, and apply, check our website here.

The fellowship is funded by generous donors from our Gitcoin project “Longevity Fellowship Grants” and matched by Vitalik Buterin.

We have open roles!

Are you an organized and dynamic person, who is interested in web3 x bio, and looking to get more involved in VitaDAO? Then one of these positions might be right for you! As someone involved with VitaDAO, you will have the chance to interact with members across all of the working groups of the DAO, and most importantly, inject good vibes into the DAO. Check out the links below for more information on specific open positions:

  • Operations Champion
  • Community and Alignment Manager
  • Twitter and Social Media Growth Lead

Panel Discussion: On the Quest to Quantify Biological Age

Check out the video below to learn more about biomarkers in January’s panel discussion “On the Quest to Quantify Biological Age” with speakers Dr. Morgan Levine, Nikolina Lauc, Dr. Joris Deleen, and host Max Unfried.

Voting Matters

Part of the strength held by the DAO model is the right of community members to vote in governance. In this section, you’ll find a rundown of notable Governance protocols that are active in the VitaDAO community. Remember, initial voting takes place on our Discourse forum before going on-chain via Snapshot!

VDP-21: Discovery of Novel Mitophagy Activators

The accumulation of damaged mitochondria, a result of compromised mitophagy, is a hallmark of aging and age-related neurodegeneration. Upregulating mitophagy could improve brain health, healthspan, and lifespan.

The lab has identified a robust mitophagy inducer that also overrides a central feature of Alzheimer’s pathology. The project will aim to validate the observed healthspan and memory improvements in mice and will be investigating the underlying molecular mechanisms.

The DAO would purchase all of the IP of the data and background IP related to the project, and is estimating that we can submit the IP for patenting before the end of 2022. The total cost of the project would be between $500k — $650k.

Cast your vote for VDP-21 here

The DeSci Revolution

The intersection of web3 and science is revolutionary. Decentralization takes the power from the few to the many. VitaDAO is a prime example of this. By allowing the community to vote on research projects to fund, we are now able to support more creative and ambitious projects that wouldn’t have otherwise been funded. It also brings together people from a variety of backgrounds and disciplines to work on amazing projects instead of being siloed into various labs. Want to learn more about DeSci, check out this thread that I (Hossam) wrote about it.

Community Member Highlight: Alex Dobrin

How did you first get involved with the VitaDAO community?

I entered an onboarding call in May 2021 after I did not hear back on my application for the Awareness Working Group. Not long after that, I started helping the awareness efforts, coming up with ideas, and executing them, including editing and distributing a press release for the genesis auction. The rest is history.

Have you always been interested in longevity research?

Ever since I was a child I was fascinated by the idea of life extension and immortality, but I did not know what to do about it. A few years ago, when I became financially free I thought about what’s meaningful in life and I decided to change my career and figure out how to best get involved in longevity.

Can you walk me through what your role inside of VitaDAO entails?

I lead the community building and social media efforts, support the working groups with operations and do whatever needs to be done.

There is no precedent for how a community like VitaDAO functions. What has surprised you most?

The fact that we can actually get stuff done without a hierarchy and the huge number of people that want to get involved every day to help the future of longevity.

What excites you most about VitaDAO’s future?

Turning a lot of early-stage aging research projects into startups that can advance longevity therapeutics to market.

Any closing thoughts for readers?

Think you can’t help the longevity field? There’s something to do for everyone. Join our community and you’ll see.

VitaDAO in the Wild

Many core members of the DAO were featured on the Idea Machine Podcast! Listen to it or watch a recording of it.

Our friends at Messari Crypto also wrote a Twitter thread and article about us! Another great summary of VitaDAO’s role in revolutionizing the longevity research industry.

Upcoming Events

February 2nd at 9 PM CET (Recurring Every Wednesday) — Swing by a VitaDAO onboarding session on Discord with Niklas Rindtorff and Alex Dobrin. Join the VitaDAO Discord Server to learn more about the DAO, how to get involved, and have any lingering questions answered.

February 13th at 2 PM CET — VitaDAO Game Night on @gather_town. Pop by to meet some fellow longevity enthusiasts. Click here to come to Game Night.

We know you didn’t think we could top the last newsletter. Well, we tried our best… Show some love and forward this newsletter to some of your friends (so that they keep letting us write these every month🙏)!

In this January edition of the VitaDAO Newsletter, you can learn about VitaDAO proposals that passed over the last month, get to know DAO rockstar Alex Dobrin, and more!
Read more
VitaDAO Newsletter Issue No 1 Jan 2021
December 31, 2021
Sarah Friday & Hossam Zaki
Awareness
Newsletters
VitaDAO Newsletter Issue No 1 Jan 2021

The VitaDAO community has been hard at work this year pioneering the future of longevity research and IP-NFTs. These past few weeks have been particularly exciting! In this December edition of the VitaDAO Newsletter you will find updates on what has happened!

It’s officially the holiday season🎄. We are very excited to begin to bring you a monthly VitaDAO newsletter! There is so much going on in the VitaDAO community. By putting things in one place, we hope it will be easier to keep up with all of VitaDAO’s moving pieces (there’s a lot!). You can expect updates on VitaDAO community news, journal club updates, summaries of active proposals, and upcoming community events. We welcome any feedback, comments, or suggestions✏️.

What is VitaDAO?

VitaDAO is a decentralized organization made up of individuals from across the globe working to fund early-stage longevity research. At its core, VitaDAO functions as an ecosystem with three moving parts: the DAO, the researchers, and their IP.

Members of the DAO govern the community and agree on projects that should be funded. Researchers within and outside the community then perform their non-profit research projects and generated IP is digitized in the form of IP-NFTs. Any proceeds added to the treasury by selling IP-NFTs to for-profit entities are used to fund future research.

Check out the VitaDAO Whitepaper for more information on VitaDAO governance and IP-NFT technicals.

Members can join VitaDAO by purchasing $VITA tokens or earning them through contributions of work or Intellectual Property. Want to purchase $VITA’s governance token? Read through the VitaDAO guide on how to purchase $VITA.

Community News

This month was a big month for VitaDAO. There were proposals, a historical second IP-NFT was issued, and exciting things are stirring for $VITA holders. Below is a summary of all the many things that happened in the VitaDAO community this month.

VDP-16 Passed

We have our second project funded by Vita DAO 🚀🚀 This is a historic moment in Vita DAO history and longevity history as a whole. The Korolchuk lab will be the second research project that will be funded through Vita DAO and IP-NFTs.

The Korolchuk lab is researching how to identify novel bioactive autophagy inducers to be used in drug discovery. Full details of the VDP-16 proposal are publicly available and can be read on the VitaDAO governance forum.

Watch a recording of the public celebration of the IP-NFT transfer:

Vita DAO VDP-14 passed!

This means that Tier 3 Governance is moving to Snapshot, making voting free! Current proposals in the VitaDAO community must pass through three tiers. Tier 1 occurs on Discord and relies on informal votes in the form of reactions and comments.

Tier 2 occurs on the Discourse governance forum and relies on off-chain votes. Tier 3 had previously taken place on-chain. If you have done anything on-chain recently, you know the struggle of gas fees these days⛽️😫. Snapshot can take a snapshot of $VITA holders and quantities at different blocks on the Ethereum Mainnet.

In taking this snapshot, staking $VITA will no longer be required to vote and governance will be easier, cheaper, and more accessible to all $VITA hodlers. Such an exciting change in the voting structure!

VitaDAO VDP-15 passed!

This vote verifies agreement regarding the Longevity Working Group’s assessment of the Rubedo Life Science Company. Rubedo Life Science Company shows promise in its ability to develop a senolytic prodrug and is composed of a talented team of scientists.

VitaDAO VDP-17 passed!

This vote approves financing a feasibility study to establish a new kind of model for drug development that also leverages IPNFTs and smart contracts. Crowd Funded Cures’ mission is to implement pay-for-success smart contracts. Pay-for-success smart contracts are an on-chain version of Social Impact Bonds, the funding of effective social services through performance-based contracts.

VDP-17 proposed providing $40,000 to Crowd Funded Cures to commission a feasibility study from a leading consulting firm on this new model of research funding. This is relevant to VitaDAO as this analysis will back a financial model VitaDAO can use in the future to fund off-patent clinical trials. Ultimately, this might allow Vitalians to support a future Generic Longevity Drug Repurposing IP-NFT, which would benefit VitaDAO and the broader community.

VitaDAO stopped selling $VITA….for a few hours.

On December 13th, a Genesis contributor was tricked out of 500k worth of $VITA. In response, the VitaDAO team moved fast, as members of the VitaDAO community sold $VITA and pulled liquidity from exchanges.

There was no hack. There was no rug pull. The VitaDAO multisig has since restored liquidity and volatility is back under control!🚀🚀🚀 Out of this experience, one important lesson has surfaced: always be careful with anyone who contacts you privately to buy your $VITA or other tokens.

Fundraising at its finest via Gitcoin!

VitaDAO collaborated with LifespanIO and Vitalik Buterin to quadratically match funding in the human longevity space. In VDP-13, the DAO voted to donate $64,299 USDC to match longevity research initiatives on Gitcoin. Thank you to everyone who donated!

Listen to Keith Comito and Vitalik Buterin’s thoughts on the longevity research funding round and future of decentralized research:

Note: The relevant segment starts around the 35-min mark

Want to learn more about public goods funding? Listen to a conversation between Gitcoin Co-Founder, Scott Moore, and VitaDAO contributor Lea Degen:

STA-STA-STA-Staking!

$VITA will soon be available on the Bancor Network for individuals to provide liquidity and earn rewards! Stay on the lookout for a VitaDAO medium article outlining how to stake your $VITA token and the intricacies of the process.

Article: Pay-for-Success Models

Savva Kerdemelidis and Ariella Coler-Reilly recently published an article on the new pay-for-success model of conducting drug development. This article compares traditional means of drug development with pay-for-success contracts using unique graphics and a hypothetical scenario involving rapamycin to treat aging.

Article: Public Investment Yield

Check out VitaDAO’s medium to read an article written by VitaDAO Longevity Working Group Steward, Tim Peterson, on ways VitaDAO is improving the yield of existing public investment and enabling democratic participation in longevity projects.

Blockchain for Science

Check out the video below for a recap of December 8th’s Blockchain for Science & VitaDAO MiniCon 2021:

Exercise Your Right to Vote

Among the most exciting aspects of VitaDAO is that anyone holding $VITA can vote on proposals! Here are two notable proposals nearing phase 2 of voting on Discourse:

VDP-11: New 10% VITA Mint for Treasury & Strategic Contributors

This proposal was recently updated for clarification. In layman’s terms, the proposal suggests minting an additional 10% of tokens to be issued to contributors (voted on by the DAO) in exchange for treasury funds and expertise from the selected entities.

Due to the low volume of decentralized exchanges, strategic contributors have reached out to VitaDAO with the inability to purchase desired quantities of $VITA. This proposal would enable five new enthusiastic contributors, who have been voted on by VitaDAO, to apply to 5 whitelisted addresses for a 10% vested fresh VITA mint. Addresses would have a one, two, or five-year vesting period during which their locked tokens could earn APY and the ability to vote.

There is a lengthy discussion surrounding the pros and cons of the proposal on Discourse. If you have an opinion, thoughts, or anything to add to the conversation, engagement is encouraged!

VDP-18: Jonathan An- Towards Reversing Periodontal Disease using Geroscience

Periodontal disease is a chronic oral disease with inflammation of the tissues supporting the teeth. The disease’s greatest underlying risk factor is age, highlighting the importance of studying periodontitis within the lens of geroscience studies.

This proposal involves $250,000.000 funding to support an 8-week mouse-model study that would use small molecule inhibitors of the PI3K/NFkB/mTOR pathway to treat periodontal disease. The study would be composed of 5 drug interventions and rapamycin as a positive control. Many age-related diseases follow a path of low-grade, chronic inflammation without infection. This process is referred to as “inflammaging.”

In evaluating the pathways that target “inflammaging,” this proposal suggests a potential gero-science based treatment of periodontitis and the potential for a positive impact on age-related cognitive decline. Discourse voting has not yet opened, but the full proposal is able to be viewed on the VitaDAO Governance Discourse forum.

Help Build the Future on Longevity Research

Currently, anyone (even people without $VITA tokens!) can vote on phase 1 and phase 2 of proposals taking place on Discord and Discourse! By voting, you are helping the DAO make important decisions and playing a role in directing the future of the DAO.

That being said, if you do hold $VITA, we encourage you to vote in all three phases of voting! The transition to Snapshot will require no gas fees, no staking of tokens, and provide more DAO members with an actionable way to shape the future of the DAO.

VitaDAO In The Wild

If you can get past the paywall, see if you can find VitaDAO mentioned in this Nature article on decentralized science (the paywall is a little ironic, right?): https://www.nature.com/articles/d41586-021-03642-9

All this discussion of IP-NFTs, exciting new research proposals, and the developing blockchain technology can be tough to follow. It’s a running joke that 1 month in crypto is like 1 year in traditional tech fields! Check out this deep dive on how VitaDAO addresses current issues in scientific funding and dissemination of novel information.

How to Get Involved

The most streamlined way to get involved is to join the Discord and hop into an orientation “Ask Me Anything” call. You can find links to the Discourse, Discord, and Twitter here: https://linktr.ee/vitadao

Upcoming Events

January 5th at 9 PM CET (Recurring Every Wednesday)- Swing by a VitaDAO onboarding session on Discord with Niklas Rindtorff and Alex Dobrin. Join the VitaDAO Discord Server to learn more about the DAO, how to get involved, and have any lingering questions answered.

January 7th at 4 PM CET (Recurring Every Other Friday)- Once involved, hop onto a call for Working Group Stewards and all Working Group Members. Where? On the VitaDAO Discord Working Group Office — Voice channel.

January 9th at 5–7 PM CET (Recurring Every Subsequent Sunday)- VitaDAO Game Night on @gather_town. Pop by to meet some fellow longevity enthusiasts. Click Here to Join Up on Game Night.

January 27th at 3:30 PM CET- Join a panel discussion with VitaDAO contributor and AI guru Max Unfried discussing the intersection of Longevity and Crypto.

Here’s to living longer, living healthier, and perhaps a holiday season full of grapes 🍇

The VitaDAO community has been hard at work this year pioneering the future of longevity research and IP-NFTs. These past few weeks have been particularly exciting! In this December edition of the VitaDAO Newsletter you will find updates on what has
Read more
Public Health @ VitaDAO: Using Democratized Wealth To Democratize Health
December 21, 2021
Tim Peterson
Science
Public Health @ VitaDAO: Using Democratized Wealth To Democratize Health

On the wealth and health of nations

There is a striking correlation between the health and the wealth of nations. For example, as life expectancy rises in a country so do economic measures like its per capita gross domestic product (GDP). These increases have been in tandem over the past centuries and are causally related to our investment in our health via sanitation, antibiotics, vaccines, etc. Meaning, it has taken our wealth to improve our health.

Many of the healthiest and wealthiest nations are democracies. A democracy is an organization governed by people that reside on land owned by the democracy. Though people living under democracies have higher living standards than ever in human history, the plateauing of life expectancy gains despite continued per capita GDP gains demonstrate the limitations of existing forms of government. In a dramatic example, in the US, in what some consider democracy in its strongest form, life expectancy is increasingly lagging behind other high GDP countries to the point where it’s now even declining.

Health and wealth for individual countries over the last 200+ years

To pinpoint the reasons for life expectancy plateauing in countries we need to look both within and beyond their borders. First, within the borders of many successful democracies the health and wealth gains are disproportionately accumulated by specific groups. The well known expression “socialized losses, privatized profits” summarizes the view. For example, certain ethnic groups lack access to health care as other groups have. Also, organizations owned by a few, e.g., pharmaceutical companies, capture a disproportionate percentage of the value of work funded by the many, e.g., NIH. Second, the health-wealth creation is disproportionately restricted to those within the borders of those who create it. See for example the COVID pandemic where the data suggests that vaccine-producing countries are prolonging the pandemic for themselves by not distributing it to those who don’t have access. These issues demonstrate the failings of health and wealth access being based on the land on which one is born.

Health care, arguably more than any other industry, sheds light on the core inefficiency of meatspace (a.k.a., land-based, real world) democracies — their difficulties in managing their assets. Fortunately, over the last decade a new type of democracy based on blockchains has emerged. The blockchain equals country analogy makes sense because a country is, in essence, a collection of assets. With a blockchain everyone’s assets fit neatly on a computer spreadsheet where everyone can easily agree on who owns what. Whereas with a country everyone’s assets are scattered over physical distance and it can often involve large time and financial costs for everyone to maintain their ownership.

Digital > Physical

Just like we’ve seen computers do with many things in the physical world — e.g., online stores vs. brick-and-mortar stores — blockchains have several advantages over countries. A key advantage of blockchains is their accessibility. They allow significantly more opportunity and control than what traditional databases managed by centralized organizations can provide. Blockchain assets are highly ‘liquid’ — they ‘flow’ where they are most needed, i.e., where the most upside is available. Blockchains are also permissionless, which means anyone can use them. Contrast this with traditional asset managers, which typically restrict their access to already wealthy people. Thus, blockchains present a new way to democratize wealth and this potential is increasingly appreciated. The role of Bitcoin in El Salvador and Nigeria are notable examples here. Behind the scenes the peer-to-peer architecture of blockchains enable individuals to transact value independent of where they are on earth without needing to trust potentially unreliable third-parties such as banks or government treasuries.

Enter DAOs

While peer-to-peer transactions are at the core of blockchains, most of the progress of our species hasn’t been made by individuals. It has been made by people taking collective action. Humans thrive because we pool and then allocate our resources. Said another way, we govern ourselves. Blockchains make possible several new types of governance structures. Notably, a decentralized autonomous organization (DAO) is a new type of blockchain-based governing body. In a DAO, code is law and consensus rules.

The efficiencies of computer-augmented organizations over human-only organizations are well documented. Think of how organizations benefited from replacing paper documents with office productivity software. The code that drives DAOs goes the key step further by turning its collective knowledge into collective action. In doing so, it tackles a fundamental issue of meatspace governance — mis-alignment of people’s resources. In meatspace, people vote on intermediaries (e.g., representatives) who then vote on allocating resources. Setting aside the substantial time and cost sink of voting on intermediaries, it’s not possible for an intermediary’s decisions to accurately represent each person’s wishes. This mis-alignment of interests is solved by a DAO because with a DAO: 1) each person themselves votes on how resources are allocated and most importantly 2) each person votes with their own resources. Thus, there are no intermediaries, and the more people put their resources behind a project, the more likely the DAO will do that project. DAOs enable what no meatspace can do — everyone simultaneously putting their money where their mouth is.

Introducing VitaDAO

Most DAOs to date have focused on financial applications, and thus on wealth creation. Given that wealth leads to health, can we imagine a DAO where people use their wealth to improve their health? The goal of VitaDAO is just that. Specifically, VitaDAO enables anyone in the world to fund and participate in projects that promote life expectancy gains, a.k.a., longevity.

VitaDAO wants to steepen the slope of the health-wealth curves for all countries. Currently, funding for biomedical research and drug development comes from investment firms, non-profits, and companies. How can VitaDAO improve upon what these organizations do? Recognizing that innovation usually builds upon existing frameworks, the most near-term solution for VitaDAO is to work hand-in-hand with these organizations. This means investing in improving their existing infrastructure.

VitaDAO Aim 1. Improve the yield of existing public investment in longevity projects

The point of time and place where technology fails is often referred to as the “valley of death”. This is the point where no-strings-attached funding fails to link up with profit-motivated funding to bring a project to market. Surprisingly, one of the largest valleys of death lies around our world’s universities. Currently a huge percentage of university developed technologies go unutilized. If one could more efficiently bridge non-profit and for-profit funding, this could improve the yield of existing public investment.

What would this bridge look like in practice? At the center of new medical technology are patents. Biotech patents exist because it is expensive and time-consuming to develop new treatments. Patent holders need to protect their investment of time and money until they are able to make a return on them. Though, what if universities could profit off their patents from the time of invention or better yet before the invention was made? This is now possible using a IP-NFT marketplace (intellectual property — non-fungible token) built by Molecule. Traditionally, intellectual property (patents and data) sit on computers waiting to be monetized. Unfortunately, selling anything takes time and skills that inventors often don’t have. Universities aren’t natural sales people either. Among other tools to fund research, VitaDAO intends to leverage Molecule’s IP-NFT marketplace to help these groups monetize their assets. Some early VitaDAO enabled university IP-NFTs from the University of Copenhagen (Denmark) and Newcastle University (UK) are described here and here, respectively.

The above approach can be thought of as building bridges to fix existing roads. These are efforts in series. In parallel, one can also imagine building new roads. With more roads, more people can participate in improving their health as well as the health of others.

VitaDAO Aim 2. Enable democratic participation in longevity projects

While patents are critical to modern medicine, historically many important medical advances, such as antibiotics and vaccines, were made possible without patents. We need to make a distinction between new technology, where inventors rightfully benefit from their efforts, and existing technology where incremental advances drive much more profit than patient benefit. The latter is what we want to avoid because of its flattening effect on health-wealth curves. A famous example of this involves insulin. The original insulin patent was given away such that it could be made cheaply and widely available. Unfortunately, over the last several decades pharma manufacturers have creatively generated new insulin patents and lobbied governing bodies, which significantly increased insulin prices to the point where many people no longer can afford this life-saving medicine. Thus, tragically we now have a crisis for something that had been a solved problem for almost 100 years now.

VitaDAO will step in here by facilitating initiatives where intellectual property isn’t relevant. Researchers and pharma companies focus on patents, but there are other participants in healthcare who don’t view success through the lens of patents: namely, patients and health care providers and insurers. For patients, what a patent-less scenario can look like is crowdfunding where the R&D work is funded by those interested in the treatment. For providers and insurers, what a patent-less scenario can look like is social impact bonds (SIB, a.k.a., pay-for-success financing), where organizations pay because their patients get healthier and this makes their performance better (providers) or reduces their payouts (insurers). An early VitaDAO SIB initiative enabled by Crowd Funded Cures is described here. NFTs might not make sense in either of the case of crowdfunded research or SIBs, but VitaDAO can still drive success, e.g., by distributing its $VITA tokens to those based on their contributions.

Fix the money, fix the world

Internet pioneer Marc Andressen famously said, “Software is eating the world”. This was a powerful slogan that catalyzed the ambitions of many Web2 entrepreneurs. Unfortunately it also anthropomorphized software as greedy. Greed gorging on tech that resembles fast food has not gotten us where we, Marc, and Peter Thiel (“We wanted flying cars but instead we got 140 characters.”) want to go. The time now is for software that is healthy for us. This is what blockchains and Web3 represent and VitaDAO will do its part.

Author: Tim Peterson, VitaDAO Longevity Working Group Steward

Illustration: Tim Peterson and Si Maclennan

Read more
A New Financial Model for Repurposing Off-Patent Longevity Medicines
December 16, 2021
Savva Kerdemelidis
Awareness
A New Financial Model for Repurposing Off-Patent Longevity Medicines

Presented by VitaDAO and Crowd Funded Cures

Under the current system, patent protection is critical for developing a new therapy. The large clinical trials required for obtaining regulatory approval can cost tens of millions of dollars, and patent royalties are how this investment is recouped and rewarded. Thus, if a drug is off-patent or “generic”, then it has almost no chance of being repurposed to treat a new disease, even if it would be safer and more effective than a new, patented drug. This is because pharma companies cannot charge a monopoly price for a generic drug as they are not the sole manufacturer. Doctors can prescribe drugs for unapproved (i.e. off-label) indications based on their knowledge of small, lower-quality studies; however, they are often reluctant to do so for fear of putting patients at risk.

In this article, we will contrast the old way of conducting drug development (based on patenting new molecules and charging a monopoly price) with a new financially innovative “pay-for-success contract” model. Under this new system, health insurers pay a subsidised price for generic drugs based on the promise of overall cost savings through improved health and reduced hospitalisations. This incentivises investors to repurpose generic drugs to treat new diseases, which can happen up to 100x cheaper and 10x faster than developing novel drugs under the old patent model. The reason it is so cheap and fast is because there are already years or even decades of existing safety data on generic drugs, giving clinical trials a huge head-start. Here we will provide a head-to-head comparison of these two models using generic rapamycin to treat aging as a hypothetical example.

Generic Drug Repurposing under the Current Patent Model

A researcher discovers in a small study that an off-patent drug, rapamycin, is especially effective at improving aging biomarkers at a specific dose and in patients with a specific genetic profile and age group. Unfortunately, as rapamycin is off-patent, the researcher will struggle to obtain funding for the large clinical trials required to obtain regulatory approval. Centralised grant and philanthropic funding is competitive with other more prestigious research organisations being more likely to obtain the funds. Grant funders also do not want to take on the political and financial risk of large clinical trials failing. For this reason, clinical trials for off-patent rapamycin are unlikely to get funded and it can only be prescribed off-label by doctors, who are reluctant to do so based on smaller trials. Philanthropic funding is also difficult because of the high risk and expense of large clinical trials.

The researcher approaches their university’s tech transfer office to see if they can get any industry attention. The only chance is for industry to work with patent attorneys to try to patent “rapalogs,” which are tweaked versions of the off-patent rapamycin. The tweaked rapalog molecule also has less safety data than off-patent rapamycin, so there is a potential risk to patients. One pharma company decides to fund a tweaked rapalog version of rapamycin that can be patented and raises hundreds of millions of dollars. It obtains regulatory approval and charges billions of dollars in monopoly prices from health insurers. To make matters worse, after a few years, doctors discover that the patented rapalog is equivalent or less safe than generic rapamycin and stop prescribing the rapalog. For this reason, industry is reluctant to fund further studies into rapamycin to determine optimal treatment protocols and dosing. In short, everyone loses.

Generic Drug Repurposing under the Pay-for-Success Model

A researcher discovers in a small study that an off-patent drug, rapamycin, is especially effective at improving aging biomarkers at a specific dose and in patients with a specific genetic profile and age group. Under the pay-for-success model, the researcher does not need to rely on patent protection to raise funds to conduct clinical trials. This is because health insurers have agreed to pay a subsidised price of up to $30m for a successfully repurposed generic drug, on the basis that it will save over $100m in costs due to reduced hospitalisations overall. This conclusion is supported by a pharmacoeconomic analysis validated in a feasibility study (lower cost per quality-adjusted-life-year or QALY).

In this hypothetical example, let’s imagine a scenario where the VitaDAO community takes on the role of the investor. In order to improve efficiency, access to liquidity, and leverage tokenomics and market forces, the pay-for-success model is implemented in a trustless manner using smart contracts and Molecule’s IP-NFT framework. This allows fractionalised ownership of the generic drug repurposing clinical trial data by token holders, such as VitaDAO members who collectively provide $10m in funds towards an “Open-Source Rapamycin” IP-NFT.

Regardless of who takes on the role of the investor and generates the funds, let’s imagine that we now have $10m to spend. This $10m is used to pay an independent contract research organisation (CRO) to conduct the clinical trials, and — if successful — obtain regulatory approval and a new label for a “branded” generic rapamycin. If the rapamycin clinical trials are successful, the health insurer agreed to pre-order $20m of the branded rapamycin at a subsidised price and to purchase $10m more, assuming that clinical efficacy is maintained. The proceeds go to the investors, i.e., the IP-NFT owners, under an arrangement with the generic drug manufacturer.

The investors (in this case, the IP-NFT owners) may then fund additional IP-NFTs for clinical trials to optimise the treatment protocol for rapamycin and create better formulations. In return, they will of course receive additional outcome payments under subsequently negotiated pay-for-success contracts with the original health insurer and others. Doctors and patients globally benefit from the clinical trial data validating the efficacy of the optimal treatment protocol involving low-cost generic longevity medicines using this innovative open-source approach.

Conclusion

Using pay for success contracts, it is possible to avoid the irrational sole reliance on the patent system to fund biopharma R&D and clinical trials. Information about which off-patent or generic medicines are safe and effective to treat new diseases or improve healthy lifespan and their optimal dosing regimes is a quintessential public good. By establishing this new pay-for-success model, we can ensure researchers are empowered to follow the science rather than forcing a patentable approach. Researchers will be able to obtain funding for large clinical trials that determine which off-patent treatment protocols help patients, without reliance on patents.

Addendum for Advanced Readers

What if you cannot find an insurer to back a pay-for-success contract or there are insufficient outcome payments available to attract an impact investor? In this case, it is possible to crowdfund outcome payments from philanthropy or the public from issuance of Open Source Pharma Longevity NFTs. These funds can be locked into a smart contract, and only payable if clinical trials are successful, as determined by a Results Oracle maintained by the CRO, using a similar method to Retroactive Public Goods Funding. It is also possible to bootstrap the pay-for-success contract using a trade secret strategy: encrypting the clinical trial results in an IP-NFT until a price is negotiated with insurers or crowdfunded from the public (via NFTs or otherwise) to unlock access to the Open Source Pharma IP-NFT and “open-source” the successful treatment protocol.

How can you learn more and get involved?

Read more about VitaDAO’s quest to decentralize ownership in biomedical longevity research and Molecule’s IP-NFT framework on their websites or on Medium, such as this announcement of the first biopharma IP-NFT Transaction and this description of the technical framework for IP-NFTs.

Read more about Crowd Funded Cures’ generic drug repurposing pay-for-success model on Medium.

If you are already a VitaDAO token-holder, please see the current VitaDAO Crowd Funded Cures partnership proposal and consider voting on-chain for VDP-17 to provide US$40k to support a Feasibility Study for a Generic Drug Repurposing Social Impact Bond (a form of Pay for Success Contract) on the basis of cost savings for health insurers. A Feasibility Study is like an investment memorandum showing that both insurers and impact investors can benefit financially from signing a pilot pay-for-success (PFS) contract to repurpose generic drugs or nutraceuticals. For example, by providing $30m to back a PFS contract to successfully repurpose a generic drug to treat a longevity-related illness, an insurer can save $100m in reduced hospitalisations. On the other side of the PFS contract, an impact investor that successfully repurposes a generic drug can make a potential $30m return by taking on the risk of a $10–15m investment in clinical trials. We aim for the Feasibility Study to be conducted by Guidehouse, who were formerly the public consulting arm of PriceWaterhouseCoopers.

About the Authors

Savva Kerdemelidis is a Commercial/IP Consultant Legal Counsel and a NZ and Australian Patent and Trade Mark Attorney with 18+ years experience advising in relation to IP, commercial law and crypto. He conducted his LLM thesis on alternatives to the patent system for developing medicines.

Ariella Coler-Reilly is an MD PhD candidate at Washington University in St. Louis studying the genetics of aging. She moonlights as a science writer and illustrator, currently working as a managing editor for the VitaDAO blog. She is passionate about public education, diversity & inclusion, and the intersection of science & web3.

Under the current system, patent protection is critical for developing a new therapy.
Read more
On the Origins of Cellular Senescence
September 28, 2021
Jason Colasanti
Longevity
On the Origins of Cellular Senescence

Originally described as gradual deterioration over time, senescence has taken on a much more intricate meaning in the past few decades. What used to be a hand-wavy term to describe the increased rate of mortality with age is now a term used often by molecular biologists to describe cellular dysfunction due to DNA damage and other environmental stressors. This radical shift came in 1961 when Leonard Hayflick, now a titan in the field of aging research, observed that senescence occurs on a cellular level, and not just on an organismal level as previously believed.

“Senescence is an inevitability. All we can do is try to strike the right balance between graceful acceptance and raging against a dying light.” — Marty Nemko, PhD.

Before Hayflick’s discovery, it was believed that cells in culture could grow indefinitely. This idea was fueled by work done from the 1910s to 1940s by Alexis Carrel, a Nobel-prize winning French surgeon, in which he observed that fibroblasts isolated from chicken hearts could grow in culture for over 30 years [1]. This finding sent shockwaves through the scientific community, especially since chickens have a maximum lifespan of only ~10 years. This work led researchers in the field to believe that cells were essentially immortal in vitro and would only succumb to aging in an in vivo setting.

The end of immortality

In 1958, the Wistar Institute hired Hayflick as a cell culture expert, and he was tasked with generating large quantities of human cells for purposes such as vaccine development. At the time, lab technicians had been struggling, unable to successfully maintain a culture of healthy (non-cancerous) cells without these cultures degrading over time. Hayflick set out to resolve this problem using a technique known as serial passaging. With serial passaging, once the cells start outgrowing their culture dish and running out of food (called “culture media”), a small portion of cells is transferred to a clean dish with fresh media. Though he achieved success using a rigorous application of this method to grow healthy human cells for many months, there was still a limit: eventually, his cells too began to degrade and stop replicating.

Convinced that he had made an error at some point in the protocol, he conducted a series of experiments with Paul Moorhead, a skilled cytogeneticist, to investigate this issue [2]. The duo conclusively determined that healthy cells indeed undergo a finite number of divisions — enough to undergo only about 40–60 passages — after which they will replicate no more. This finding represented a major break from Carrel’s earlier work, which was still widely accepted at the time. Hayflick and Moorehead’s observations suggested for the first time that aging occurs on a cellular level, as healthy cells eventually cease to divide and enter a state of cellular senescence.

It is now believed that the work originally done by Carrel was deeply flawed [3]. There are a few theories as to how this may have occurred: one popular theory is that the original population of cells contained pluripotent stem cells; another theory is that new cells were being added to the original population with every addition of fresh cell culture media. Despite Carrel’s lauded “discovery” at the time, his experiments with cell division have been irreproducible to this day.

Hunting for an internal counter

The discovery that somatic cells undergo a finite number of cell divisions was replicated in numerous cell types across a plethora of species, and this constraint became known as the Hayflick Limit. Hayflick speculated that cells could somehow “count” or keep track of their population doublings and began investigating this hypothesis. He observed that cryogenically preserved cells retained a “memory” of past cell divisions and adhered to the Hayflick Limit even after being frozen for years. This finding suggested that the mechanism responsible for the Hayflick Limit was not based on the passage of time, but on an unknown biological phenomenon.

Dubbed the “replicometer”, Hayflick and his lab set out to find the mechanism responsible for restricting the maximum number of cell divisions. The first mystery to solve was where the cellular machinery underlying this mechanism was located, and from there they planned to decipher which organelles and molecules were involved. They postulated that there were two plausible scenarios: either the machinery was located in the cytoplasm or the nucleus. If located in the cytoplasm, the counting mechanism would most likely be due to the progressive accumulation of damaged organelles; if in the nucleus, then the explanation could be that senescence is a programmed genetic event.

In 1975, Woodring Wright, a graduate student in Hayflick’s lab, answered this question by fusing together cells from different rounds of population doublings [4]. Wright took cells and removed their nuclei by treating them with cytochalasin B. This process leaves two distinct parts of biological material: cytoplasts (cells without nuclei) and nucleoplasts (free nuclei). For his experiments, he took nucleoplasts from young cells (10–15 rounds of population doublings) and inserted them into the cytoplasts of old cells (40–50 population doublings). He also did the reverse: he inserted nucleoplasts from old cells into the cytoplasts from young cells. Wright found that the number of population doublings exhibited by these hybrid cells depended entirely on the donor nucleus. In other words, cells with young nuclei exhibited many more population doublings before becoming senescent, while the cells with old nuclei reached senescence after far fewer doublings. This experiment showed that the internal counting mechanism takes place in the nucleus and not in the cytoplasm.

Solving the end replication problem

Though the location of this internal counter had been identified, the molecular mechanism remained a mystery. Alexey Olovnikov, a Russian theoretical biologist studying DNA replication, proposed an answer [5]. He knew that each time a cell divides, it first duplicates its DNA so as to pass on one copy of its genome to each of the two daughter cells. He also knew that DNA replication machinery was unable to access the very end of the DNA strand, known as the 3’ end. Thus, he postulated that with each cell division, the DNA strands must become shorter and shorter; given enough of these divisions, the cell will eventually lose important genetic information, leading to dysfunction or death. This is known as the end replication problem.

To get a sense of how this occurs, imagine a construction worker standing on a brick wall. His job is to lay down bricks while walking on the wall in a single direction. He places one brick down, then takes one step back, then places another brick down, takes another step back, etc. He is able to successfully carry out his job for almost the eternity of the wall, except he is unable to lay the very last brick because he has nothing left to stand on, and so he falls off, leaving the wall incomplete. This is the end replication problem in a nutshell, wherein the new layer of bricks is the new DNA strand, and the construction worker is the DNA replication machinery.

Construction worker analogy for the end replication problem.

Both the end replication problem and the cellular senescence phenomenon raised important questions about the nature of life itself. Namely, if the DNA in cells continues to shrink with each generation, then would all life not eventually die out? Similarly, if cells are capable of a finite number of cell divisions before becoming senescent, then how does life continue to persist? Since life has been thriving on our planet for over 4 billion years, Olovinikov hypothesized in 1971 that there must be mechanisms responsible for protecting genetic information as well as lengthening DNA. Despite these thought-provoking hypotheses, Olovinikov never conducted the laboratory experiments necessary to support this idea.

Fortunately, scientists in the United States were making significant progress to support the existence of both a mechanism to protect genetic information and a mechanism to lengthen DNA. In 1978, while conducting experiments on tetrahymena, a single-cell organism, Elizabeth Blackburn and Joseph Gall noticed something peculiar about its DNA. They observed that the ends of each DNA strand had repeated sequences of six nucleotides, TTGGGG. These redundant regions of the DNA, known as telomeres, repeat 20–70 times [6]. This finding provided evidence that telomeres act as buffers to protect important genetic information from being eliminated. Telomeres, or some variant of them, exist in many different types of organisms ranging from fungi to animals.

In 1985, a mechanism for telomere lengthening was co-discovered by Elizabeth Blackburn and Carol Greider when they identified the enzyme telomerase, which is responsible for adding telomeric repeats onto the ends of DNA [7]. Telomerase is active in germline and stem cells, but not somatic cells, which is why Hayflick and others observed that cells like dermal fibroblasts have finite replicative lifespans. In 1990, Blackburn and Greider demonstrated that telomeric DNA length decreases with each cell division, both in vitro and during natural aging in vivo [8]. Finally in 1998, Woodring Wright, now running his own laboratory, showed that telomere shortening was sufficient for cells to enter senescence [9]. Thus, the mechanism responsible for cells reaching the Hayflick Limit was finally elucidated. Elizabeth Blackburn would go on to win the 2009 Nobel Prize in Physiology or Medicine and share it with Carol Greider and Jack Szostak for these findings.

Timeline of important discoveries on the road to understanding replicative senescence (1940–1998).

These seminal discoveries raised important questions in the field of cellular biology. What is the purpose of cellular senescence? Is senescence a mechanism that has evolved to confer a survival advantage to a species, or is it merely an accident that evolution has failed to eradicate? In this article, I have described the initial discovery of cellular senescence, the mechanism behind replicative senescence, and the great minds responsible for these findings. Still, this is only a small fraction of the vast array of captivating insights and outstanding questions within the field of cellular senescence. To learn more about cellular senescence, including the relationship between senescence, aging, and longevity, stay tuned for future articles in this series.

By Jason Colasanti, Author
and Ariella Coler-Reilly, Editor and Illustrator

Timeline graphic created using BioRender.com.

Note: This article was updated on Oct 5, 2021 to more precisely describe Hayflick’s motivations and observations.

References

  1. Carrel, A. & Ebeling, A. H. Age and multiplication of fibroblasts . J. Exp. Med. 34, 599– 606 (1921). doi: 10.1084/jem.34.6.599
  2. Hayflick, L., & Moorhead, P. S. (1961). The serial cultivation of human diploid cell strains. Experimental cell research, 25(3), 585–621. doi: 10.1016/0014–4827(61)90192–6
  3. Hayflick, Leonard. “Mortality and immortality at the cellular level. A review.” Biochemistry-New York-English Translation of Biokhimiya 62.11 (1997): 1180–1190. PMID: 9467840
  4. Wright, W. E., & Hayflick, L. (1975). Nuclear control of cellular aging demonstrated by hybridization of anucleate and whole cultured normal human fibroblasts. Experimental cell research, 96(1), 113–121. doi: 10.1016/s0014–4827(75)80043–7
  5. Olovnikov A. M. (1971). Printsip marginotomii v matrichnom sinteze polinukleotidov [Principle of marginotomy in template synthesis of polynucleotides]. Doklady Akademii nauk SSSR, 201(6), 1496–1499. ResearchGate Article
  6. Blackburn, E. H., & Gall, J. G. (1978). A tandemly repeated sequence at the termini of the extrachromosomal ribosomal RNA genes in Tetrahymena. Journal of molecular biology, 120(1), 33–53. doi: 10.1016/0022–2836(78)90294–2
  7. Greider, C. W., & Blackburn, E. H. (1985). Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. cell, 43(2), 405–413. doi: 10.1016/0092–8674(85)90170–9
  8. Harley, C. B., Futcher, A. B., & Greider, C. W. (1990). Telomeres shorten during ageing of human fibroblasts. Nature, 345(6274), 458–460. doi: 10.1038/345458a0
  9. Bodnar, A. G., Ouellette, M., Frolkis, M., Holt, S. E., Chiu, C. P., Morin, G. B., … & Wright, W. E. (1998). Extension of life-span by introduction of telomerase into normal human cells. science, 279(5349), 349–352. doi: 10.1126/science.279.5349.349

Originally described as gradual deterioration over time, senescence has taken on a much more intricate meaning in the past few decades.
Read more
The First Longevity Drugs might be already among us — Drug Repurposing to Fight Aging
August 30, 2021
Estefano Pinilla
Longevity
The First Longevity Drugs might be already among us — Drug Repurposing to Fight Aging

We are getting quite good at extending lifespan, or at least we are getting quite good at extending the lifespan of laboratory animals as complex as mice.

If you follow the progress in anti-aging research, you will read about an arsenal of molecules, strategies and pathways that seem to make animals live longer (sometimes by several folds) and healthier. These results make us believe that increasing our years of healthy life is possible. Although, as nothing can be that easy, the benefits of these strategies seem to decrease with the complexity of the animal models. If we could show beyond reasonable doubt that any of these interventions works in humans, even by extending our healthy life by only a couple of years, that would be a huge milestone for the anti-aging field that would greatly accelerate progress.

Aging is not currently considered a disease by any regulatory agency, and therefore testing treatments to extend healthy lifespan is highly disincentivized.

There are several obstacles separating us from that first step, though. Aging is not currently considered a disease by any regulatory agency, and therefore testing treatments to extend healthy lifespan is highly disincentivized, since no drug can be approved on that basis. Additionally, drug development is difficult, with more than 90% of new molecules failing before they reach clinical trials due to high toxicity and/or efficacy. Things get a little bit easier when our molecule has reached human trials and failure rates drop significantly, being the lack of efficacy for the chosen disease the main reason for compounds to be dropped at this stage. As the drug gets closer to market, the trials get more and more expensive in order to answer the demands of regulatory agencies, and economic reasons start having more weight in the decision of dropping the development of new treatments.

This process already takes 10–12 years and costs between $2–3 billion for drugs with recognized indications… imagine the timescales and costs for drugs aimed to increase healthy lifespan. Any time that can be saved could have a huge impact and greatly reduce costs. For example, at the level of clinical trials we could find good surrogate biomarkers, such as the so-called epigenetic clocks, instead of waiting for years until participants die. But what would happen if some of the drugs that are used on a daily basis by doctors everywhere had the potential to extend lifespan and we only needed to tweak them a bit? This is the idea behind drug repurposing.

Starting the Race Half Way Closer to the Finish Line

Drug repurposing consists of using already existing drugs for other purposes than the one for what they were originally created, and it can be done with molecules that went all the way through their regulatory approval for a different use, or with those that were left in the way for different reasons. Since these molecules at least would have demonstrated to be decently safe and bioavailable, it is not difficult to see the convenience of repurposing when we consider the failure rates in early drug discovery that we have previously mentioned. If drugs that are currently in use are found to be repurposable, we start the race half way closer to the finish line and what is left is to demonstrate that our molecule is effective for its new use.

Of course this is no small feat, but it can shorten a 10–12 years process to a 3–8 years one, saving millions of dollars and what is more important, a comparable amount of lives.

Figure from Kane, Anil. Whitepaper: Drug repurposing trends and strategic approaches for shortening timelines (https://patheon.com/resource-library/whitepapers/drug-repurposing-trends-and-strategic-approaches-for-shortening-timelines/)

If this is such a great strategy, why are we not seeing Big Pharma pursuing it more? Well, the pharmaceutical industry has been repurposing drugs successfully for years; sildenafil citrate (aka. Viagra) being probably the most well-known example. The blue pill was initially conceived as a treatment for angina pectoris, a heart condition, but interactions with important drugs in the management of this disease and the fact that it had to be taken three times a day made Pfizer drop that line of testing. Men participating in the clinical trials, however, experienced interesting side effects that convinced the pharmaceutical company to put the drug to a better use rather than abandoning it completely, resulting in the blockbuster drug for erectile dysfunction that we all know.

The issue with drug repurposing in the pharmaceutical industry is that it has been the result of serendipity, as the example of Viagra illustrates, rather than of a systematic approach.

AI and Decentralized Governance for Drug Repurposing

Given the potential benefits, there has been an increasing interest by the pharmaceutical industry on establishing repurposing programs, although the big players seem to encounter some challenges unlocking the full potential of drug repurposing and mainly focus on reusing their own old compounds. Some of these challenges include lack of good drug repositories and data, lack of collaboration within the industry and with academia, and maybe most importantly lack of financial incentive to find new uses for off-patent molecules.

That is why collaborative groups of academic researchers have taken over the task of repurposing generic drugs, lack of funding being their main limitation. All these pitfalls might explain why, despite the efforts, all the attempts to use already existing drugs to treat the SARS-CoV-2 infection have failed so far. One cannot avoid thinking if we would not have obtained better outcomes with more funding of independent researchers and transparent collaborations between companies in the industry.

Although traditional models of funding, IP and collaboration in the pharmaceutical industry do not seem to be particularly well-suited for this approach, a lot of exciting innovation has occurred in the repurposing space tackling both lack of data and the lack of incentives. Additionally, several companies have leveraged the power of AI to create interesting platforms for drug repurposing. Intellectual property sharing between stakeholders and crowdsourcing have also been proposed as possible solutions. At VitaDAO we are excited about the enabling potential that the IP-NFT framework has in this regard, in addition to the effects of decentralized governance on the creation of better incentives.

Metformin and Rapamycin: Old-style Repositioning Against Aging

But let’s bring the focus back to our mission: longevity. Drug repurposing is not an unkown approach in the anti-aging field: two of the most well studied compounds to extend lifespan in laboratory animals, metformin and rapamycin, were designed with very different aims.

Metformin has been used for decades to lower blood glucose in type 2 diabetic patients, being one of the most commonly prescribed drugs, and rapamycin is an immunosuppressant widely used to prevent organ rejection after transplants. Funnily enough, the current use of rapamycin is itself repurposing after the drug failed as an antifungal agent because it caused potent immune suppression as a side effect.

Metformin and rapamycin seem to work by acting on two different enzymes that function as energy sensors in cells: AMP-activated protein kinase and mammalian target of rapamycin (AMPK and mTOR for short) respectively. The activating effect of metformin on AMPK and the inhibitory effect of rapamycin on mTOR mimic some molecular aspects of calorie restriction and have been consistently shown to increase lifespan in model organisms.

Metformin and rapamycin are just the most well studied cases, but other molecules such as acarbose or the combination of growth hormone and dehydroepiandrosterone are in the candidate list.

Although large and comprehensive studies by the National Institute of Aging Intervention Testing Program cast doubt on the effectiveness of metformin to increase lifespan in mice, some claim that large clinical trials in non-diabetic patients are justified given the broad safety margin of the drug. Moreover, small trials in elderly patients have shown promising effect in markers associated with healthy aging. In fact, the Targeting Aging with Metformin (TAME) trial is currently raising funds in order to launch and hopes to make the treatment of aging an indication, which would be revolutionary in its own right.

Rapamycin, on the other hand, has a more narrow safety profile compared to metformin but has been consistently shown to extend lifespan in all animals where it has been investigated, and a clinical trial in humans has been recently crowdfunded to test its effect on several markers of aging, including epigenetic clocks.

Metformin and rapamycin are just the most well studied cases, but other molecules such as acarbose, used to inhibit the digestion of starch in diabetics, or the combination of growth hormone and dehydroepiandrosterone used for thymus regeneration are in the candidate list of compounds that could be repurposed to fight aging.

Can Serendipity be Systematized?

Besides these classical interventions, researchers are also trying to use more systematic approaches to find candidates for longevity drugs among our existing arsenal of compounds. A common way of doing this is to use databases of proteins or genes with a well-known role in aging and virtually screen libraries of compounds to predict which existing molecules will interact with the desired target.

Of course, these computational methods are just the first step in the repurposing path and need to be validated by experiments in model organisms, so one could argue that these approaches look similar to starting from scratch. That is why some researchers decide to skip the computational part and directly test a bunch of compounds in simple model organisms where you can get data on a quite large population and measure directly how they improve different parameters, including lifespan, with relative ease and without needing to care about the mechanism at first.

Some researchers decide to skip the computational part and directly test a bunch of compounds in simple model organisms as the worm Caenorhabditis elegans

A good example of this is the use of the worm Caenorhabditis elegans, which offers a convenient model of a small multicellular organism that shares some age-associated features with mammals. The problem with model organisms is that, obviously, they are not humans and sometimes drugs just do not work in the same way when you change species. This issue is called lack of translatability, and to tackle it some researchers try to directly use data from human patients and start from there to make predictions.

An interesting approach in this direction is to use existing data to look at how the expression of genes changes with aging in a certain tissue, and then look in existing databases for drugs that induce opposing changes, or similar changes if we suspect that they are adaptive, in cells. This has been successfully done for the human brain with a couple of promising molecules popping up after the analysis, including among them well known anti-aging drugs such as rapamycin. All these approaches have their advantages and disadvantages, and can certainly be combined to obtain more robust candidates, but all rely on the existence of available repositories with good quality data.

The Longevity Molecule: pushing for the anti-aging revolution

This takes us to the first project funded by VitaDAO: The Longevity Molecule by the Scheibye-Knudsen lab. Using exclusive access to the incredible wealth of real-world data contained in the medical and prescription records of the Danish Healthcare System, the research group led by Morten Scheibye-Knudsen has analyzed data from 4.8 million patients, including 1.04 billion prescriptions of 3500 different drugs over 40 years trying to find drugs that were correlated with a longer life.

Of course, people that get prescription drugs are not particularly healthy and how much they live depends more on their diagnosis than on the drugs they are taking. To avoid this “diagnosis bias” the Scheibye-Knudsen lab quantified the survival benefit of prescribed drugs compared to other drugs given for the same indication. Using this method, they found more than 10 medications that correlated strongly with lifespan in long-lived populations, and selected the three with a stronger correlation.

This project has the potential to uncover new mechanisms driving longevity

For the first part of the project, they will confirm the effects of these drugs in controlled laboratory experiments on human cells and on the well-known aging model Drosophila melanogaster (aka the fruit fly). If these experiments are successful, they will optimize them and begin testing on mice for the second phase of the project. Besides the goal of getting at least one candidate to start trials on human patients, this project has the potential to uncover new mechanisms driving longevity that could be targeted in the future.

If any of these approaches succeed, the next step under the current framework would be to choose an age-related disease to demonstrate an effect on human healthspan in the eyes of the regulatory agencies. Otherwise the framework should be changed to accept aging as a disease, but that is a different fight. We are still a long way from that, but only a few years of extra healthy life would be enough to change public perception and spark an anti-aging revolution. It is hopeful to think that the molecule able to do that could be already among us, hiding in plain sight.

Sources:

We are getting quite good at extending lifespan, or at least we are getting quite good at extending the lifespan of laboratory animals as complex as mice.
Read more
How to acquire VITA on Uniswap
June 23, 2021
Awareness
Tokenomics
How to acquire VITA on Uniswap

This article will guide you through the process of using Uniswap to obtain VITA, VitaDAO’s token. For more information on VitaDAO itself, see this blog post as well as further links at the bottom of this article. If you have any question, do get in touch on Discord, Telegram or Twitter.

💡 Be sure to obtain authentic VITA

The original and only token contract address of the VITA token is 0x81f8f0bb1cb2a06649e51913a151f0e7ef6fa321. Any token with another contract address — even if also called “VITA” — must be fake. Make sure that you obtain authentic VITA by using this and only this pool on Uniswap.

1. Prerequisites

In order to obtain VITA on Uniswap, you will need

1️⃣ the public address of your Ethereum wallet

and

2️⃣ Etherum tokens (ETH) or USDC equivalent to the amount you would like to acquire VITA tokens for, plus additional resources for gas costs (that is, transaction fees).

In case you already have both 1) and 2) in place, you may skip to section 3 “How to obtain VITA on Uniswap”.

2. What does this mean and what do I need to do?

1️⃣ The public address of an Ethereum wallet of yours

Unless you already have an Ethereum wallet on which you would like to hold your VITA tokens, you will need to create a new one. Please note, an exchange account does not work and cannot be used to obtain VITA tokens from Uniswap.

While there are many Ethereum wallet options, we recommend the following:

1. Install a Chromium-based browser, such as Chrome, Brave, Firefox or Edge.

2. Install the browser extension MetaMask, a reputable and easy-to-use Ethereum wallet.

3. When prompted, allow the extension to be added to your browser.

4. Click on “Get Started”.

5. Click on “Create a Wallet” and, on the next screen, choose whether you would like to provide MetaMask with optional analytical data.

6. Choose a secure password and agree to the Terms of Use. In terms of password security, we recommend you think of your MetaMask wallet like a bank account. Make it a secure password and store it in a safe place, such as a password manager.

7. Very important: Follow the instructions on your screen carefully in order to back up your “backup phrase”. This phrase, and only this phrase, guarantees you access to your Ethereum wallet and your VITA tokens.You must not disclose this phrase to others. If you lose this phrase, you cannot restore your account and lose your tokens. Make sure you keep it safe.

8. Confirm your phrase and continue.

9. Now, if you see this screen, MetaMask is set up successfully.

10. Dismiss the release notes and make yourself comfortable with the overview page of your newly created Ethereum wallet.

11. Finally, move your mouse cursor to the top where it says “Account 1”, click left to copy your public Ethereum address to your clipboard and save it for later. You will need it in the next section in order to top up ETH.

2️. ETH or USDC equivalent to the amount you would like to buy VITA tokens, plus additional ETH for “gas costs”

1. Go to an exchange of your choice. Any exchange where you can acquire ETH will work, including popular exchanges like Coinbase or Binance. Depending on your jurisdiction, your options may be limited. Note that registering a new account with any exchange may take a few days of processing time, especially due to Know Your Customer (KYC) procedures.

Note: On Uniswap, you can use ETH or any ERC-20 token like USDC. The fees are the lowest for USDC, which is why this is the optimum asset to acquire VITA with. However, using ETH is more convenient and only slightly more costly in terms of fees, which is why the following steps only explain how to acquire ETH.

For simplicity, we recommend one of the following:

or

  • Wyre Widget, which you can use directly in MetaMask (click here for their FAQ)

2. Acquire ETH of

  • the amount that you would like to buy VITA tokens for

and

  • approximately 0.02 ETH or the USDC equivalent additionally, in order to account for all possible transaction fees

3. Transfer your ETH to your wallet using the public Ethereum address you noted down in step 11 of the last section. When making a withdrawal/transfer, always double check your public Ethereum address is correct. If you enter the wrong address, your funds will be irreversibly lost. Transferring ETH to your own wallet may take several minutes to confirm.

Note: Depending on which exchange you chose in step 1, your ETH may transfer directly to your Ethereum wallet, e.g. when using Ramp or Wyre. In that case, this step is irrelevant.

4. Finally, open MetaMask and ensure the account balance in your Ethereum wallet is as you expect it to be. If you just withdrew your ETH from an exchange, it may take several minutes for your funds to arrive.

5. Now, you are all set to acquire VITA on Uniswap.

3. How to obtain VITA on Uniswap

Step 1 : Import VITA token

Go to the pool of the official VITA token on Uniswap v3 by following one of the links mentioned below. First, you will need to manually import the VITA token. Since VITA is a relatively new token that has been existing only for a few days, it is not yet listed on what is known as Uniswap Lists. In the meantime, you need to manually import VITA but be mindful of fake tokens.

Only use one of the following links:

  • If you want to exchange ETH for VITA, click here.
  • If you want to exchange USDC for VITA, click here.
  • If you want to exchange another ERC-20 token for VITA, click here and later manually your desired token.

To be sure, compare the Uniswap URL to this Etherscan URL of the official token contract. Both must refer to the address 0x81f8f0bb1cb2a06649e51913a151f0e7ef6fa321.

If everything seems fine, click on “Import”.

Double check that the address is 0x81f8f0bb1cb2a06649e51913a151f0e7ef6fa321 and click “Import”.
Step 2: Connect your wallet

In the top right-hand corner, click on “Connect to a wallet”.

Select a wallet of your choice by clicking on it. Any of the given options will work. In this guide, we will be using MetaMask.

Next, a pop-up will appear. Select the wallet you earlier transferred USDC and/or ETH. Click “Next”.

Finally, click “Connect”.

The pop-up will close automatically and you will see the Uniswap page again.

Step 3: Enter the desired amount

Now, you can specify the desired amount of USDC or ETH you would like to spend on acquiring VITA tokens. Alternatively, you may also enter the desired amount of VITA you would like to acquire and let Uniswap calculate the corresponding amount in USDC or ETH respectively.

[If applicable] Step 4: Approve tokens

If you are using Uniswap for the first time, you will likely have to approve the use of your tokens first. To do so, click the blue button. You will then need to confirm the transaction in your MetaMask wallet.

Step 5: Confirm swap

After you have specified the amount, click on “Swap”.

Double check that everything is correct and click on “Confirm Swap”. Follow the usual steps in your MetaMask wallet to confirm.

Before the transaction is completed, it needs to be processed which may take up to a few minutes. You may click on “View on Explorer” to track the progress on Etherscan.

Step 6: Add VITA token to MetaMask

While waiting for your transaction to confirm, click on “Add VITA to MetaMask” and follow the steps.

Step 7: Verify transaction

As a last step, wait for the transaction to clear. You will be notified through a banner in the upper right corner, as displayed here:

Finally, open your MetaMask wallet by clicking on the little fox icon on the top right-hand side. Next, click on “Assets” and verify that your desired amount of VITA has been added to your wallet.

If you cannot see VITA, follow these steps:

[If applicable] Step 8: Add VITA as custom token

Click on “Custom Token” and add the Token Contract Address, which is 0x81f8f0bb1cB2A06649E51913A151F0E7Ef6FA321 (see also on Etherscan). The remaining two fields are filled automatically.

Click on “Next”.

Click on “Add Tokens”.

Now, your MetaMask shows your balance of VITA tokens.

Congratulations, you now have acquired VITA tokens and become a member of VitaDAO with full voting rights.

Stay tuned for another guide how to take part in VitaDAO’s governance processes and how to vote on proposals.

We hope to see you soon on Discord and our forum. VitaDAO is off to a great start and we cannot wait to get going. The real work begins only now!

This article will guide you through the process of using Uniswap to obtain VITA, VitaDAO’s token. For more information on VitaDAO itself, see this blog post as well as further links at the bottom of this article.
Read more
Announcing the First VitaDAO Research Project: The Longevity Molecule
May 7, 2021
Awareness
Projects
Longevity
Science
Announcing the First VitaDAO Research Project: The Longevity Molecule

1.04 billion prescriptions
4.8 million individuals
50 years of The Danish National Health Service Prescription Database

We’re excited to introduce the opening project to be funded by VitaDAO — the world’s first decentralized intellectual property collective. The research project is led by Morten Scheibye Knudsen and carried out by the Scheibye-Knudsen Lab in Copenhagen.

Who is VitaDAO?

VitaDAO is a new type of entity in the biotech research space, and its members will own the intellectual property assets that result from the projects it supports and funds. Its mission is to extend the human lifespan by researching, financing, and commercializing longevity therapeutics in an open and democratic manner.

Introducing The Scheibye-Knudsen Lab

The Scheibye-Knudsen lab focuses on developing interventions for aging and age-associated diseases using in silico, in vitro, and in vivo approaches.

The lab discovered that DNA damage leads to changes in certain metabolites and that replenishment of these molecules may alter the rate of aging in model organisms.

These findings, and the ones from other groups, suggest that normal aging and age-associated diseases may be malleable to similar interventions. The hope is to develop interventions that will allow everyone to live healthier, happier, and more productive lives.

Take a short tour of the lab with the founder, Morten:
https://www.youtube.com/watch?v=kkmXBOe3Wy4

The Scheibye-Knudsen Lab team comprises a diverse group of distinguished biotech scientists working together in the lab in Copenhagen.

Research Project Overview: The Longevity Molecule

In an unprecedented effort, using advanced machine learning technologies, the Scheibye-Knudsen lab has analyzed 1.04 billion prescriptions from 4.8 million individuals over 50 years in The Danish National Health Service Prescription Database and correlated this with the survival of individuals prescribed certain drugs. They received exclusive access to carry out this work.

The Scheibye-Knudsen Lab has identified 10+ FDA-approved medications that appear to have a strong effect on lifespan following analysis.

This project will focus on optimizing, repurposing, and re-formulating the three drugs with the strongest and most significant impact on the human lifespan.

Preparing the fly cultures for testing.

Project Significance

Since aging is the largest risk factor for most diseases, discovering compounds able to extend the healthy lifespan could have profound implications not only on disease progression but also on society. Indeed, this project may be transformative for healthcare and everyone’s well-being and is particularly attractive from a commercial standpoint considering the current 200 billion+ USD anti-aging market.

Project Timeline

Pre-Clinical Studies 1: Testing in Fruit Flies and Human Cells — 12–24 Months (IN PROGRESS)

The first step is testing the identified drug’s ability to attenuate features of aging in a controlled laboratory setting. Here, we will test the molecules in both human cells and the animal model Drosophila melanogaster (fruit flies).

We are testing the drugs in human cells with urgency to understand how the molecules affect human cellular aging. Fruit flies are used extensively in biomedical research and specific aging research as they have a short lifespan of about 60 days. We have exclusive access to a deep-learning-based technology (Tracked.bio) that can investigate aging in thousands of flies at once.

Fruit Fly (Drosophila melanogaster)

Pre-Clinical Studies 2: Testing in Mouse Models — 12–18 Months

After successful testing in fruit flies and cells, we will perform compound optimization in cell lines and proceed to test the compound for age-attenuation in mice. We will utilize medicinal chemistry for compound optimization, allowing better efficacy, solubility, stability, and tolerability. We will specifically test toxicology on optimized compounds for the mice and how age-associated features, such as hearing, are affected.

Clinical Trials 1: Human Trials — TBD

We will transition to clinical trials and test the age-attenuating effect of the compound in healthy volunteers. Currently, the Scheibye-Knudsen lab is running three clinical trials for aging interventions in humans. Fortuitously, Morten Scheibye-Knudsen also runs the largest recruitment platform for volunteers for clinical trials in Denmark.

Introducing the founder

“For Dr. Scheibye-Knudsen, associate professor at the University of Copenhagen, aging is the most significant risk factor for most diseases, and damage to our genome is likely the cause of aging.

His focus is on exploring the physiological consequences of DNA damage and what we can do to intervene in genotoxic insults. His goal is to understand, modulate, and treat aging and age-related diseases.

In addition to his academic activities, Morten has founded several companies, including Forsoegsperson.dk, Mitodb.com, Tracked.bio, and Soosys.com. He is a board member at the Longevity Vision Fund and serves as Chief Editor at Frontiers Media.”

Morten Scheibye-Knudsen, Founder of the Scheibye-Knudsen Lab in Copenhagen (Reference — https://www.dataforgood.science/tag/morten-scheibye-knudsen/)

If you would like to join the VitaDAO community, get in on the conversation, stay up to date and find ways to contribute — visit our website and community links below!

— Website (vitadao.com)
— Twitter (twitter.com/vita_dao)
— Discord Community & Working Groups (discord.gg/vitadao)

— Telegram Community (t.me/vitadao)
— Would you like to participate? You can fill out our contact form (https://airtable.com/shrgouAxrM9jsjUlF)

We’re excited to introduce the opening project to be funded by VitaDAO — the world’s first decentralized intellectual property collective.
Read more
Announcing VitaDAO — The first publicly owned decentralized IP collective
April 3, 2021
Awareness
Announcing VitaDAO — The first publicly owned decentralized IP collective

Hello world!
Hello VitaDAO

We’re a decentralized cooperative that lives in the meta-verse.

We have a single purpose:
Accelerate and democratize research into longevity therapeutics.

As humans, longevity is our final frontier
Our mortality is what separates us from the machines that we create. Biotechnology has entered a phase of exponential progress that few of us grasp — redefining what it means to be human. Web3 is at a similar point, accelerating at a superhuman pace and promising the greatest breakthroughs in open collaboration humanity has witnessed.

Photo by Massimo Virgilio https://unsplash.com/photos/pv5dSOM1UCY

Beyond curing diseases, researchers across the globe are redefining aging and developing therapeutics that may enable you to read this very article in a hundred years from now and chuckle.

This research progress is happening right now. And there is a race to centralize it in the hands of the few. History likes to repeat itself unless we act.

VitaDAO is your chance to act

We are a new cooperative vehicle for community-governed and decentralized drug development.

Our core mission is the acceleration of R&D in the longevity space and the extension of human life- and health-span. Today, the biopharma industry is booming with unprecedented late-stage investment inflows, particularly in the longevity space. However, critical early-stage funding is severely lacking and incentives between patients, researchers and industry are misaligned.

Think of yourself as a Schelling point in this great coordination game we call life
VitaDAOs goal is to attract the best and earliest longevity research, acquire it, and democratize it. In that sense, VitaDAO is like a self-governing ETF composed of hundreds of early-stage longevity projects. Many will fail, but the few that succeed may change your future forever.

To align incentives and vitalize early-stage funding in longevity biopharma, VitaDAO utilizes a combination of novel governance frameworks and decentralized autonomous organizations (DAOs), non-fungible tokens (NFTs), and financial engineering tools such as algorithmic automated market makers (AMMs) that run on the Ethereum blockchain.

VitaDAO Token Economy Overview

At its core, biopharma value creation is composed of intellectual property (IP) rights and research data

Research and development has become prohibitively expensive and siloed, partially due to how IP business models incentivize monopolization of innovation through creation of thickets of patents in protected portfolios. These IP frameworks prevent the open sharing of research data, inherently disincentivizing collaboration and transparency. They prevent the public and patients from having any real ownership in biopharma IP, even though their tax dollars fund much of the early-stage development. Outside of grants to fund basic research, early-stage funding for drug development is extremely limited. When drugs do finally make it to market, there are strong incentives for price gouging.

VitaDAO solves some of these problems, and in the long run perhaps all of them by creating entirely new open IP business models. Your vision is audacious and bold. But if you won’t, who will?

As an open cooperative that anyone can join and support, VitaDAO’s goal is to acquire, support, and finance new therapeutics and research data in the longevity space, and will directly hold IP rights in novel early-stage therapeutics while looking to grow a portfolio of both IP and data assets. These can be made available and monetized in novel data marketplaces such as Ocean Marketplace, promoting both open science and new open data business models.

Ownership and governance of VitaDAO is relegated through VITA tokens

VITA can be obtained by contributing work, or funds, or other resources such as data and IP to VitaDAO. Ownership of VITA allows the holder to participate in the democratic governance of VitaDAO, directing its research, accessing and monetizing its data repositories, and managing its IP portfolio.

Right now, we are a child. Learning to crawl, speaking our first words. Over time, we will walk, read and write, then we will run. Remember your first thoughts, that first summer as a child, and make those thoughts your own. You are VitaDAO. And life will happen slowly, then all at once.

Learn more and read the 1st whitepaper draft: vitadao.com/about

Join the discussion!
Discord Community & Working Groups: discord.gg/SMz2RRYCY5

You can also get involved here: https://airtable.com/shrgouAxrM9jsjUlF

Telegram Community: t.me/vitadao

Stay up to date: twitter.com/vita_dao

Hello world! Hello VitaDAO
Read more
Thank you! Your submission has been received!
Oops! Something went wrong while submitting the form.