Lifespan Research Institute

The Blog

Building a Future Free of Age-Related Disease

Public Longevity Group

Lifespan Research Institute Launches Public Longevity Group

[Mountain View, September 17, 2025]Lifespan Research Institute (LRI) today announced the launch of the Public Longevity Group (PLG), a new initiative focused on bridging the cultural gap between scientific breakthroughs in aging and their public acceptance. To kickstart its work, PLG has opened a crowdfunding campaign to develop tools that measure and strengthen public trust in longevity science.

While the science of longevity biotechnology continues to advance, skepticism and cultural resistance limit progress, with some studies showing that more than half of Americans would reject a safe, proven therapy to extend life. This hesitation poses risks of raising costs, delaying health-promoting regulation, and slowing the delivery of treatments that could combat age-related diseases and extend healthy lifespan.

“The breakthrough that unlocks all other breakthroughs is public trust,” said Sho Joseph Ozaki Tan, Founder of PLG. “Without it, even the most promising therapies may never reach the people they’re meant to help. PLG exists to change that.”

“Persuasion is a science too,” said Keith Comito, CEO of Lifespan Research Institute. “To bring health-extending technologies to the public as quickly as possible, we must approach advocacy with the same rigor as our research. With PLG, we’ll be able to systematically measure and increase social receptivity, making the public’s appetite for credible longevity therapies unmistakable to policymakers, investors, and the public itself.”

PLG is developing the first data-driven cultural intelligence system for longevity—a platform designed to track real-time sentiment, test narratives, and identify which messages resonate and which backfire. Early tools include:

  • The Longevity Cultural Clock: a cultural barometer mapping readiness and resistance across demographics and regions.
  • Sentiment Dashboards: real-time monitoring of public, investor, and policymaker perceptions.
  • Narrative Testing Tools: data-driven analysis that will enable robust pathways to public support.

The crowdfunding campaign will provide the initial $100,000 needed to launch these tools, creating the cultural foundation required for healthier, longer lives.

With a lean, data-driven team, the group aims to provide open-access cultural insights for advocates and policymakers while offering advanced analytics to mission-aligned partners.

Campaign Timeline:

  • Campaign completion: November 2, 2025
  • Dashboard development: Dec 2025 – Feb 2026
  • First survey deployment: Feb – Apr 2026
  • Beta dashboard launch: May 2026
  • First public insight report: June 2026

Supporters can contribute directly at: https://lifespan.io/campaigns/public-longevity-group/

The PLG campaign is sponsored by the members of LRI’s Lifespan Alliance, a consortium of mission-aligned organizations that believe in the promise of extending healthy human lifespan. Newly-joined members include OpenCures, AgelessRx, and Lento Bio.

About Lifespan Research Institute

Lifespan Research Institute accelerates the science and systems needed for longer, healthier lives by uniting researchers, investors, and the public to drive lasting impact. LRI advances breakthrough science, builds high-impact ecosystems, and connects the global longevity community.

Media Contact:

Christie Sacco

Marketing Director

Lifespan Research Institute

christie.sacco@lifespan.io

(650) 336-1780

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

A Single Gut Microbe Suppresses Weight Gain in Mice

Scientists have found that a single microbial species can blunt the negative effects of a high-fat diet due to the unique mix of lipids it produces [1]. They intend to identify its specific lipids in future work.

Good neighbors

The billions of gut microbes that we share our bodies with can profoundly influence our health. For instance, microbiome diversity is generally reduced in obese people [2]. Transferring microbiota from obese to lean mice causes the latter to put on weight [3], while entirely germ-free mice stay lean under a high-fat diet (HFD) [4], suggesting that some bacteria promote weight gain while others restrict it.

Scientists have known for a while that spore-forming (SF) bacteria support healthy metabolism and leanness. The team at the University of Utah that originally discovered this connection recently looked for particular bacterial strains that can produce an oversized effect, and they published their result in Cell Metabolism.

The lone hero

The researchers found that among all the SF species, the bacterium Turicibacter single-handedly improved the metabolic health of mice on HFD when supplied continuously, including lowering triglyceride levels more robustly than the entire SF community. Turicibacter also reduced weight gain and shrank white adipose tissue (WAT). It pushed down sphingolipid metabolism in the small intestine and lowered circulating ceramides. Ceramides, a subclass of sphingolipids, tend to rise on HFD and are often linked to insulin resistance and lipid overload.

“I didn’t think one microbe would have such a dramatic effect; I thought it would be a mix of three or four,” said June Round, PhD, professor of microbiology and immunology at U of U Health and senior author on the paper. “So when [we did] the first experiment with Turicibacter and the mice were staying really lean, I was like, ‘This is so amazing.’ It’s pretty exciting when you see those types of results.”

The team then used a human metagenomic database to compare Turicibacter levels across people categorized by obesity status. In that dataset, Turicibacter was markedly lower in individuals with obesity, which matches several prior studies.

As HFD in mice is also associated with reduced microbial diversity, the researchers hypothesized that diet might directly suppress Turicibacter, rather than the latter simply being a passive marker of obesity. To prove this, they used Turicibacter-monocolonized mice (germ-free mice colonized with Turicibacter alone), feeding them either HFD or normal chow. HFD almost eliminated Turicibacter from the small intestine and significantly reduced it in the lower GI tract, despite it being the only organism in the gut. Interestingly, palmitate, a major saturated fat in HFD, reduced Turicibacter growth in vitro.

These results suggest that HFD may promote weight gain in part by suppressing the bacteria that normally counteract it. Since HFD is hostile to stable colonization, in the in vivo experiments, the mice had to be not only monocolonized with Turicibacter but also constantly fed it to keep them exposed to the bacteria.

The secret is in the mix

Using bacterial lipidomics, the researchers showed that Turicibacter produces a highly specific mix of lipids dominated by galactolipids with relatively low phosphatidylcholine. They note, however, that 95% of the Turicibacter lipidome is unannotated, so it could be making sphingolipid-like molecules that current databases miss.

Importantly, experiments showed that lipids from Turicibacter can get into intestinal epithelial cells. Once there, they downregulate genes that support ceramide synthesis. The team suggests that this slowing down of ceramide production, which leads to reduced fatty acid uptake by epithelial cells, is likely a major contributor to the bacterium’s effect on weight gain. Apparently, HFD does not just reduce Turicibacter abundance, it also shifts its lipid composition, blunting its effect on ceramide synthesis.

Treating epithelial cells with Turicibacter-derived lipids in vitro recapitulated the bacterium’s effect on lipid uptake. When this lipid extract was fed to mice, the animals showed reduced weight gain, lower fasting glucose, lower WAT, and blunted sphingolipid-related gene expression.Bacterium lipid effects

Next, the researchers hope to identify the particular lipids responsible. “Identifying what lipid is having this effect is going to be one of the most important future directions,” Round said, “both from a scientific perspective because we want to understand how it works, and from a therapeutic standpoint. Perhaps we could use this bacterial lipid, which we know really doesn’t have a lot of side effects because people have it in their guts, as a way to keep a healthy weight.”

“With further investigation of individual microbes, we will be able to make microbes into medicine and find bacteria that are safe to create a consortium of different bugs that people with different diseases might be lacking,” said Kendra Klag, PhD, MD candidate at the Spencer Fox Eccles School of Medicine at the University of Utah and first author of the paper. “Microbes are the ultimate wealth of drug discovery. We just know the very tip of the iceberg of what all these different bacterial products can do.”

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Klag, K., Ott, D., Tippetts, T. S., Nicolson, R. J., Tatum, S. M., Bauer, K. M., … & Round, J. L. (2025). Dietary fat disrupts a commensal-host lipid network that promotes metabolic health. Cell Metabolism.

[2] Davis, C. D. (2016). The gut microbiome and its role in obesity. Nutrition today, 51(4), 167-174.

[3] Ridaura, V. K., Faith, J. J., Rey, F. E., Cheng, J., Duncan, A. E., Kau, A. L., … & Gordon, J. I. (2013). Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science, 341(6150), 1241214.

[4] Bäckhed, F., Manchester, J. K., Semenkovich, C. F., & Gordon, J. I. (2007). Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proceedings of the National Academy of Sciences, 104(3), 979-984.

Rejuvenation Roundup December 2025

Rejuvenation Roundup December 2025

We’re closer to 2050 than to 2000, and technology is advancing apace; this includes technologies that help us stop aging in all its forms. Here’s the highlights of what’s been accomplished last month.

Team and activities

Christmas-Editorial.pngA Year of Rejuvenation Research and Journalism: Winter is in full swing for those of us in the Northern Hemisphere. It is a time for cozy nights by the fire and a great time to catch up on what has been happening at Lifespan News and LRI.

Interviews

Maximina Yun on the Wonders of the Axolotl: Dr. Maximina Yun, principal investigator at Chinese Institutes for Medical Research in Beijing (CIMR), studies some of the most amazing animals in the world: salamanders, a group of amphibians that includes newts and species such as the universally loved axolotl.

Advocacy and Analysis

Close-look-at-documents.jpgAnalyzing the Quality of Preclinical Anti-Aging Research: Researchers have investigated the reporting quality of preclinical studies’ outcomes in anti-aging research. They analyzed how study quality changed over time, shortcomings in research, and the improvements that can be made in the future in order to yield as many valuable insights as possible.

Research Roundup

Arginine Reduces Signs of Alzheimer’s in Mice: The amino acid arginine shows promise in animal models of amyloid aggregation due to its ability to promote protein folding. This study’s researchers suggest that it could be useful for early prevention and treatment of Alzheimer’s.

Used cigarettesSecond-Hand Smoke Alters Protein Expression: A recent study investigated plasma proteins in people exposed to secondhand smoke and found that exposure affected multiple molecular processes, including immune, inflammatory, and tissue repair pathways.

Engineering Immune Cells to Fight Gut Senescence: In Nature Aging, researchers have published their finding that targeting urokinase plasminogen activator receptor (uPAR), a senescence-associated protein, restores gut function in mice.

Microglial cell on neuronMicroglia Replacement Already Working in Humans: A new review highlights the promise of microglia replacement, a strategy that made the leap from mouse studies to the first successful human trial in just five years.

Nasal Tissue Extracellular Vesicles Improve Health in Mice: Researchers have discovered that extracellular vesicles (EVs) derived from the nasal mucosa provide significant regenerative benefits to tissues throughout the body, including the brain.

Rhesus macaqueStem Cell-Derived Vesicles Improve Cognition in Aged Monkeys: In a new study, extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) improved spatial working memory in rhesus macaques, suggesting a possible reversal of age-related cognitive decline.

A Key Molecular Link Between Aging and Osteoporosis: In Aging Cell, researchers have described how an age-related deficiency in another compound leads the antioxidant FoxO1 to contribute to bone deterioration in osteoporosis by siphoning from a bone-building pathway.

Suspended stem cellsHuman Umbilical Cord Blood Metabolites Lengthen Worms’ Lives: A comparison of human umbilical cord blood with adult plasma revealed hundreds of metabolites whose abundances were age-dependent. Two different formulas, each a mix of a few metabolites, demonstrated anti-senescence properties in cell cultures and model organisms.

Molecule From Chocolate Linked to Slower Epigenetic Aging: A new observational study spanning two human cohorts has found a link between theobromine, a phytochemical abundant in cocoa, and slower epigenetic aging.

Obese mouseTargeting a Metabolic Regulator Reduces Fat in Mice: In Aging, researchers have described how they removed visceral fat from older male mice by targeting the metabolic regulatory protein CD47.

Senescence Exacerbated by Mitochondrial RNA Leakage: Scientists have investigated a little-known mechanism that fuels cellular senescence: mitochondrial RNA leaking into the cytoplasm. Targeting this mechanism showed promise in a mouse model of fatty liver.

GeneticsA Gene That Changes Function With Aging in Mice: In Nature Aging, researchers have discovered how growth differentiation factor 3 (GDF3), a cytokine that increases with aging, is related to more inflammatory macrophages in older animals.

Producing Thymic Factors in the Liver Rejuvenates Immunity: A new study proposes a novel approach to fighting immune system decline caused by thymic involution: making the liver produce proteins that support T cell development and function.

Auditory pathwayResearchers Find a Potential Target for Hearing Loss: In Aging Cell, researchers have identified bone marrow stromal antigen 2 (BST2) as a key protein in age-related hearing loss in a mouse model.

Bacterium From Frogs Completely Destroys Colon Cancer: Scientists in Japan have discovered that multiple strains of bacteria taken from frogs, newts, and lizards can be effective against human colon cancer in a mouse model. One particular microbe achieved a perfect response rate and survival.

SupplementsHow Multivitamins and Minerals Impact Health and Longevity: A recent review evaluated studies that included over 5.5 million participants to assess the impact of multivitamin and mineral supplements on different aspects of health.

Blood measure of neuronal death is exponentially higher with age, especially in females, and halted in Alzheimer’s disease by GM-CSF treatment: An exponential increase in neurodegeneration with age, accelerated by astrogliosis/inflammation, may underlie the contribution of aging to cognitive decline.

Pharmacologic reversal of advanced Alzheimer’s disease in mice and identification of potential therapeutic nodes in human brain: Forty-six proteins aberrantly expressed in advanced 5xFAD mouse brain and normalized by P7C3-A20 show similar alterations in human Alzheimer’s brain, revealing targets with potential for optimizing translation to patient care.

High- and Low-Fat Dairy Consumption and Long-Term Risk of Dementia: Evidence From a 25-Year Prospective Cohort Study: Higher intake of high-fat cheese and high-fat cream was associated with a lower risk of all-cause dementia, whereas low-fat cheese, low-fat cream, and other dairy products showed no significant association. APOE ε4 status modified the association between high-fat cheese and Alzheimer’s.

Long-Term High-Protein Diet Intake Accelerates Adipocyte Senescence Through Macrophage CD38-Mediated NAD+ Depletion: These findings establish macrophage-adipocyte NAD+ crosstalk as a central axis linking dietary protein excess to white adipose tissue aging, providing actionable targets for the prevention and treatment of age-related metabolic disorders.

Dietary fatty acids and epigenetic aging in US adults: results from the National Health and Nutrition Examination Survey: The researchers found consistent positive associations for saturated fat subtypes and negative associations for polyunsaturated fat subtypes with epigenetic aging; associations of monounsaturated fat subtypes varied.

Relationships among dietary patterns and heterogeneous biological aging at system and organ-specific levels and mortality risks: Weighted Cox regression models revealed that aging-related diet scores were more strongly associated with mortality risk than their respective diet scores alone.

Accelerometer-measured weekend catch-up sleep and incident dementia: A prospective cohort study: Moderate weekend catch-up sleep was linked to lower dementia risk, especially among individuals with less weekday sleep.

Oral sodium hyaluronate improves skin hydration, barrier function and signs of aging: a randomized, double-blind, placebo-controlled trial in 150 healthy adults: This approach improved multiple aspects of skin physiology, supporting its use as a functional food ingredient with measurable benefits for skin health and healthy aging.

An unbiased comparison of 14 epigenetic clocks in relation to 174 incident disease outcomes: Second- and third-generation epigenetic clocks show promise for disease risk prediction, particularly in relation to respiratory and liver-based conditions.

Small Extracellular Vesicles From Human Amniotic Membrane Mesenchymal Stem Cells Rejuvenate Senescent β Cells and Cure Age-Related Diabetes in Mice: According to the researchers, this establishes a framework for targeting cellular senescence in metabolic disorders.

Platelet-bioengineered hiPSC-sEVs achieve targeted repair of fibrotic sinoatrial node in preclinical SND models: This study establishes a targeted, cell-free nanotherapeutic platform for resolving fibrosis and electrophysiological dysfunction in sinus node disease.

Abrogation of aberrant glycolytic interactions eliminates senescent cells and alleviates aging-related dysfunctions: A new senolytic molecule that targets the PGAM-Chk1 interaction creates a specific vulnerability of senescent cells to potentially fight age-related diseases.

Enhanced non-enzymatic H2S generation extends lifespan and healthspan in male mice: These findings uncover the potential of enhanced hydrogen sulfide generation to promote healthy aging.

Long-term consequences of soft political repression on psychological well-being, systemic inflammation and cellular ageing: This study investigates the long-term sequelae of soft political repression in the former East Germany, which was linked to higher levels of interleukin-6, indicating increased systemic inflammation.

From sick care to healthspan: educating the longevity physician for health maintenance and health promotion: Education may represent one enabling factor in efforts to shift, where feasible, from predominantly reactive care toward more proactive approaches to health maintenance.

Why we age: The geroscience hypothesis, which states that intervention on the rate of aging should also modulate the incidence of age-related diseases, is likely to be correct.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Longevity investors

Longevity Investors Lunch 2026

Longevity Investors announces the upcoming Longevity Investors Lunch 2026, an exclusive, application-only gathering designed specifically for investors seeking exposure to the most compelling opportunities emerging at the intersection of longevity science, technology, and capital. Held during the World Economic Forum in Davos, the event convenes a highly curated group of global investors alongside select longevity scientists, researchers, and industry leaders shaping the future of health and aging.

Since its launch in 2022, the Longevity Investors Lunch has established itself as a trusted platform – one that bridges capital allocators with frontier science, applied longevity technologies, and academically-driven innovation moving toward commercialization. The event is purpose-built to translate scientific progress into investable insight, enabling informed capital deployment.

LIL2026 1

Inside the Longevity Investors Lunch in Davos — curated discussions bringing together investors, scientists, and industry leaders in an intimate setting.

The 2026 program will feature expert-led panels, focused insights, and curated networking sessions designed to foster meaningful dialogue and long-term collaboration between investors and the scientific community. Discussions will focus on areas including translational longevity science, computational biology, precision medicine, AI-enabled healthcare, and scalable interventions targeting aging and age-related disease. All discussions are shaped through an investor lens rather than purely academic exploration.

Announced Speakers for LIL 2026

LIL2026 2

Prof. Evelyne Yehudit Bischof, MD, PhD, MPH

Director, Sheba Longevity Center

Internationally recognized physician-scientist advancing translational longevity medicine, clinical innovation, and evidence-based health optimization.

Jordan Shlain, MD

Founder & Chairman, Private Medical

Physician, entrepreneur, and healthcare innovator known for advancing prevention-first medical care and long-term health strategy.

Dr. Liv Kraemer, MD, PhD

Founder, Dr. Liv Kraemer Skin Longevity Clinic

Dermatologist and longevity researcher specializing in skin biology, preventive dermatology, and personalized aesthetic-health integration.

Peter Fedichev

CEO, GERO.ai

Computational biophysicist and AI pioneer focused on aging biomarkers, systemic resilience, and data-driven longevity therapeutics.

Dr. Andrea Gartenbach, MD

Longevity Physician, Axmann/Gartenbach

Internal medicine specialist and longevity expert focusing on cardiometabolic prevention, functional and hormone-based therapies, and personalized performance-driven health optimization.

Dr. Neven Pičuljan, PhD

Co-Founder, Aion Longevity

AI engineer and technology entrepreneur applying machine learning to integrate wearable data, lab results, and longitudinal wellbeing signals—advancing personalized, data-driven longevity analytics and precision health insights.

“We are proud to host the fifth edition of our Longevity Investors Lunch in January 2026 in Davos as a side-event during the World Economic Forum. Our vision and ambition is still the same: to bring more money into the emerging longevity industry by attracting investors in Davos who support research and startups in the industry.“ says Marc P. Bernegger, Co-Founder and Host of Longevity Investors. “Longevity is developing at an extraordinary pace, yet many still wonder where the meaningful inflection points lie. LIL 2026 in Davos gives us the opportunity to filter out the noise, examine what the science really tells us, and connect investors with the innovators building solutions that can genuinely enhance healthspan.” says Dr. Tobias Reichmuth, Co-Founder and Host, Longevity Investors

LIL2026 3

Longevity Investors Co-Founders Marc P. Bernegger and Dr. Tobias Reichmuth, CEO Lucy Kupcova, and LIL 2025 speaker Dr. Deepak Chopra at the Longevity Investors Lunch in Davos.

LIL 2026 builds on a longstanding tradition of convening leaders who shape the future of healthspan, medicine, and scientific progress. As longevity science accelerates, the gathering offers an essential platform for investors and innovators to align on the breakthroughs, trends, and foundational work driving the next wave of global health transformation.

That dialogue continues with the seventh edition of the Longevity Investors Conference, taking place September 14–17, 2026, in Gstaad, Switzerland. Hosted in a private, invitation-only setting at a 5-star luxury hotel, the Longevity Investors Conference is the world’s most private investor conference dedicated to longevity, bringing together key opinion leaders, institutional and private investors, family offices, and funds to explore the scientific and investment foundations of human healthspan extension.

Together, the Longevity Investors Lunch and the Longevity Investors Conference reflect a shared conviction: as advances in biotechnology, medicine, and data-driven science accelerate, longevity is emerging as one of the defining investment opportunities of the decades ahead—and requires informed capital, rigorous dialogue, and long-term perspective.

Press Contact

Longevity Investors

info@longevityinvestors.ch

www.longevityinvestors.ch

https://www.longevityinvestorsnews.ch/

Follow us on social media: X: @Longevity_Inv IG: LongevityInvestors LinkedIn: Longevity Investors

Supplements

How Multivitamins and Minerals Impact Health and Longevity

A recent review evaluated studies that included over 5.5 million participants to assess the impact of multivitamin and mineral supplements on different aspects of health [1].

A big market

Multivitamin and mineral (MVM) supplements are widely used worldwide, and in the United States, around 40% of adults report taking MVMs regularly [2]. While there is scientific evidence regarding the beneficial effects of individual vitamins or micronutrients on health, the role of MVMs in lifespan and longevity remains to be evaluated.

Andrea Maier, whom we recently interviewed, and her team conducted a review to assess findings from 19 meta-analyses published in the last 25 years on the impact of MVMs on different aspects of health, with each study analyzing data from a few thousand to almost a million participants. While the analysis showed no effect of MVM supplementation on all-cause mortality, it did affect the risks and progression of various health conditions.

Impacting disease risk

The researchers began by examining the impact of MVM supplements on cancer. They noted that seven prospective cohort studies suggest a protective effect of MVM supplementation against colorectal cancer, with an 8% lower incidence among supplement users. However, no association was observed between MVM supplementation and breast cancer incidence, prostate cancer incidence, advanced/metastatic prostate cancer, or prostate cancer mortality.

Similarly, the pooled analysis of 16 cohort studies found no significant association between MVM use and cardiovascular disease mortality, coronary heart disease mortality, stroke mortality, or stroke incidence. This analysis, however, showed a lower risk of coronary heart disease incidence, but two randomized controlled trials (RCTs) didn’t support this association. Additional evidence for cardiovascular disease came from a set of 12 RCTs, which suggested “MVM intake lowers blood pressure in individuals with hypertension or chronic disease but has little effect in normotensive individuals and does not prevent hypertension.”

Differences by subgroup were also observed regarding infections. While no significant effect of MVM supplementation on infection rates was found in healthy people 65 and older, among people under 65, MVM supplementation was associated with fewer infections. Similarly, MVM supplementation for at least 6 months in older, undernourished people reduced the number of infections. Two RCTs that focused explicitly on COVID-19 infection found no effect of MVM supplementation on COVID-19 outcomes in hospitalized patients.

Regarding psychological health, the researchers reported positive effects of MVM supplementation, with studies showing that MVM supplementation reduces stress, mild psychiatric symptoms, anxiety, and fatigue in healthy adults, but not depression. The effectiveness was robust for supplements with high doses of B vitamins.

An additional positive impact of MVMs was observed on cognition and musculoskeletal health. Studies reported that MVM supplementation benefits global cognition and measures of cognitive performance, such as immediate free recall and episodic memory, and is associated with a lower risk of fragility-related hip fractures.

There was also one negative observation. When 13 RCTs of peopple aged 42-89 years old with varying stages of age-related macular degeneration were analyzed, the researchers reported that “MVM supplementation does not improve visual acuity and increases the risk of progression to late age-related macular degeneration.” Data from different vision-related studies were more positive, suggesting that MVM supplementation is associated with a lower risk of cataracts (according to observational findings). RCTs also suggest a protective effect, but only for one cataract subtype, nuclear cataracts, in older adults.

Pregnancy

While many studies exclude pregnant women, studies on MVM supplementation analyzed the MVM supplementation during pregnancy and its outcomes. They observed that there was no association between MVM supplementation containing folic acid and the risk of gestational hypertension or preeclampsia in observational studies. However, RCTs suggest a protective effect.

Studies suggest no significant effect of MVM supplementation on preterm birth, low birth weight, and stillbirth. Observational studies differed from RCTs regarding small-for-gestational-age births, in which observational studies, but not RCTs, suggest a protective effect of MVM supplementation. Observational studies also found a significant association between MVM supplementation and a lower risk of neural tube defects, congenital cardiovascular defects, urinary tract defects, and limb deficiencies, but not of Trisomy 21.

Additionally, children of mothers who took MVMs before or during pregnancy suggest that prenatal MVM supplementation is positively associated with reduced risk of specific pediatric cancers (leukemia, brain tumors, and neuroblastoma), and there is a possible protective effect of prenatal MVM use against autism spectrum disorder in offspring according to some, but not all, studies.

Supplement effects

The need for a standardized definition

The overall results indicate that MVM supplementation can be beneficial in specific populations or for particular conditions, suggesting that multiple variables, such as age, sex, health status, dietary intake, and food frequency, should be taken into account when recommending MVMs. The authors point out that nutritional deficiencies should receive special attention, as deficiencies in specific nutrients can lead to health problems and accelerated aging [3].

Other studies also suggest that “nutritional status may modify the response to supplementation,” and the benefits of supplementation can be more pronounced in individuals with lower dietary quality [4]. Stratifying study participants into subgroups based on different factors might help identify those most likely to benefit from supplementation. However, as the reviewers conclude, “the value of MVM supplementation in optimizing the health and healthspan of relatively healthy individuals remains uncertain.”

Analysis of multiple studies also highlighted general limitations and improvements needed when conducting MVM studies. One of the most significant problems in this field is the lack of standardized definitions of what qualifies for MVM supplements, which vitamins and minerals should be included, and in what quantities. Lack of definition can result in inconsistencies in study outcomes and makes comparing studies more challenging, since MVMs in various studies can have different compositions. Additionally, some studies use self-reported questionnaires to gather information, which often lack information about the type of supplement used.

There is also a possibility that some studies on MVMs, especially observational cohorts that recruit self-selected supplement users, suffer from “healthy user effect.” The cohorts in those studies might contain more health-conscious individuals, who are interested in taking supplements and engaging in other health-oriented behaviors, making the samples of MVM supplement users not representative of the general population. Such cohorts might be lacking participants who might benefit the most from MVM supplementation: less health-conscious people who might have nutritional deficiencies.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Lou, C., Wang, G., Xiong, Z., Jiang, Y. C., Li, Y., Zhu, M., Yang, H., Wang, L., He, L., Chang, H. M., Wang, J., Zhu, W., Dong, X., Li, T. Y., Yuan, S., Zhao, Y., & Mu, L. (2025). Association of female reproductive traits with altered aging trajectories: Insights from genetic and observational analyses. Cell reports. Medicine, 6(12), 102481.

[2] Knippen, K. L., Mahas, R., & Van Wasshenova, E. (2020). Outcome Expectancies, Health Information Seeking, and Cancer Beliefs Associated with Multivitamin/Mineral Use in a National Sample, HINTS-FDA 2015. Journal of the Academy of Nutrition and Dietetics, 120(8), 1368–1376.

[3] Mahadzir, M. D. A., Tan, S., Elena, S., Chin, E. M., Garg, V., Mantantzis, K., Péter, S., & Maier, A. B. (2025). Towards defining optimal concentrations of micronutrients in adults to optimize health. Mechanisms of ageing and development, 225, 112062.

[4] Rautiainen, S., Gaziano, J. M., Christen, W. G., Bubes, V., Kotler, G., Glynn, R. J., Manson, J. E., Buring, J. E., & Sesso, H. D. (2017). Effect of Baseline Nutritional Status on Long-term Multivitamin Use and Cardiovascular Disease Risk: A Secondary Analysis of the Physicians’ Health Study II Randomized Clinical Trial. JAMA cardiology, 2(6), 617–625.

Bacterium From Frogs Completely Destroys Colon Cancer

Scientists in Japan have discovered that multiple strains of bacteria taken from frogs, newts, and lizards can be effective against human colon cancer in a mouse model. One particular microbe achieved a perfect response rate and survival [1].

Bacteria against cancer

Recent research has illuminated the enormous impact of the microbiome on overall health. Microbes that live inside us can influence metabolism, modulate inflammation, tune hormone and neurotransmitter signaling, and alter drug breakdown and absorption [2]. Via their immunomodulatory action, bacteria can influence susceptibility not just to infections and autoimmunity but also cancer, affecting its initiation, progression, and responses to therapy [3].

In a new study published in Gut Microbes, the research team of Prof. Eijiro Miyako at the Japan Advanced Institute of Science and Technology (JAIST) ventured one step further from the human microbiome. They gathered and analyzed bacteria from three vertebrate species – two amphibians (Dryophytes japonicus and Cynops pyrrhogaster) and a reptile (Takydromus tachydromoides) – to see if they could find strains that actively suppress cancer.

100% kill rate

After an initial assessment, the team chose nine strains for further evaluation in a mouse colon cancer model. Immunocompetent mice were inoculated with human colon cancer cells subcutaneously, and after the tumors grew to 200 mm3, the study group received a single intravenous injection of a bacterial suspension. Tumor growth was then monitored for 40 days.

One strain produced no antitumor activity, which is still an important result, “indicating that not all gut bacteria possess intrinsic anticancer properties,” the paper says. The rest showed at least some slowing of tumor progression and survival improvement.

One particular bug stood out: Ewingella americana, taken from the guts of Dryophytes japonicus, the Japanese tree frog. Rarely do cancer studies produce such convincing results: E. americana achieved fast and complete tumor eradication in all mice. The entire study group survived until the end of the follow-up, while the entire control group was dead before day 30. When the control group was subjected to tumor rechallenge, none developed cancer, suggesting long-lasting anti-cancer immunity.multi

The researchers then compared E. americana’s efficacy to that of two established anti-cancer treatments: PD-1L checkpoint blockade and the chemotherapy drug doxorubicin. While the two treatments showed some efficacy, none of them came close to E. americana’s results.

The therapy was well-tolerated and safe. “Comprehensive analysis of hematological and biochemical parameters revealed no significant differences between E. americana-treated mice and PBS-treated control groups across all measured parameters,” the paper notes. Histology confirmed no detectable organ toxicity or structural damage.

Essentially, after the injection, E. americana colonized the tumor, quickly destroyed it, and then was itself cleared out by the immune system, leaving no trace except for the prolonged immunity. In any case, E. americana is known to respond well to antibiotics, so any lingering infection can probably be easily treated.

Direct cytotoxicity combined with immune recruitment

The authors then delve deeper into possible mechanisms of action. Interestingly, two more bacteria achieved initial tumor regression, but then cancer rebounded. Like E. americana, these two are facultative anaerobic bacteria, meaning that they can thrive in both oxygen-rich and hypoxic environments. The researchers note thhis finding is “consistent with established principles of bacterial cancer therapy, as anaerobic bacteria possess the unique capability to selectively accumulate and colonize within solid tumors due to the characteristically hypoxic and immunosuppressive tumor microenvironment.”

Working with an in vitro tumor model, the team found that E. americana killed lumps of cancer cells (cancer spheroids) directly by secreting cytolysins, toxins that create pores in cellular membranes. However, in vivo, the bacteria also recruited B cells, T cells, and neutrophils. This massive immune cell mobilization and accompanying increase in inflammatory signals, including interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), probably contributed a lot to the treatment’s effectiveness. The researchers confirmed tumor destruction using techniques for detecting cellular death by apoptosis.

“Collectively, these mechanistic investigations demonstrate that E. americana employs a multifaceted approach to achieve tumor elimination, combining direct bacterial-mediated cytotoxicity with robust activation of host immune responses,” the paper concludes. “The bacterium efficiently infiltrates and proliferates within tumors, where it exerts direct cytotoxic effects while simultaneously activating immune cells (particularly T cells, B cells, and neutrophils) to effectively eliminate cancer cells through complementary mechanisms.”

Like any study, this one had certain limitations. For instance, the colon cancer model was subcutaneous instead of gastrointestinal. However, as the authors note, such a model can be especially relevant for metastatic cancers, the deadliest variety. It also remains to be seen whether E. americana is effective against other cancers and how it fares in combination with other bacteria or treatments.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Iwata, S., Yamasita, N., Asukabe, K., Sakari, M., & Miyako, E. (2025). Discovery and characterization of antitumor gut microbiota from amphibians and reptiles: Ewingella americana as a novel therapeutic agent with dual cytotoxic and immunomodulatory properties. Gut Microbes, 17(1).

[2] Hou, K., Wu, Z.-X., Chen, X.-Y., et al. (2022). Microbiota in health and diseases. Signal Transduction and Targeted Therapy, 7, Article 135

[3] Dutta, D., & Lim, S. H. (2020). Bidirectional interaction between intestinal microbiome and cancer: opportunities for therapeutic interventions. Biomarker Research, 8(1), 31.

Auditory pathway

Researchers Find a Potential Target for Hearing Loss

In Aging Cell, researchers have identified bone marrow stromal antigen 2 (BST2) as a key protein in age-related hearing loss in a mouse model.

Neural myelination is necessary for function

Demyelination, the loss of neurons’ protective sheaths, is known to be fundamental in multiple serious neurological disorders, including both amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS). The proliferation and migration of Schwann cells (SCs) is critical in maintaining neuron myelination [1]; this includes the spiral ganglion neurons (SGNs) that govern hearing, and impairment of these cells leads to motor and sensory problems in mice [2].

Both ALS and MS are accompanied by an increase in BST2, and in a mouse model of experimental autoimmune encephalomyelitis (EAE), another demyelinating disease, suppressing BST2 in SCs halts its progression [3]. However, the precise relationship between BST2, SCs, and aging was “unclear in peer-reviewed literature” according to these researchers, and therefore, they performed this study to investigate it more closely.

Hearing loss is correlated with higher BST2 expression

In their first experiment, the researchers took a look at the SGNs of mice from 3 to 18 months old, with groups being 3 months apart. They noticed that even at only 6 months of age, the mice had very slightly less myelination; at 12 months, they had considerably less, and at 18 months, there was practically no myelin left. Similarly, the mice began to lose their hearing at 12 months, with many tones being far less audible.

Unsurprisingly, this was linked to an increase in BST2 in the SCs, with a direct correlation between hearing thresholds and BST2 expression. This was also accompanied by a decrease in N-cadherin and an increase in E-cadherin, which demonstrates that the SCs had a reduced ability to migrate.

The researchers then administered a BST2-suppressing adeno-associated virus (AAV) into the ears of group of 8-month-old mice. Compared to the untreated and empty-virus control groups, the treated mice enjoyed significantly improved hearing, although not to the level of 4-month-old mice. The myelin of their auditory nerves had been significantly restored, and further experimentation found that the treated mice had significantly fewer senescent Schwann cells.

A correlation with inflammation

SCs with elevated BST2 were also found to have elevations in the senescence- and inflammation-related NF-κB pathway, which led to a decrease in the crucial factor POU6F1. POU6F1 was found to be crucial to SC function, improving healing and cellular migration of SCs while increasing the myelin-promoting factor MPZ, and knocking out POU6F1 blunted the effects of knocking out BST2. Downregulating POU6F1 in 6-month-old mice impaired their hearing, and overexpressing it improved their hearing. This finding corroborates previous research demonstrating that POU6F1 promotes the growth of neurons [4].

The researchers noted that this experiment was performed on a very specific subset of Black 6 mice, C57BL/6J, and that C57BL/6N mice do not lose their hearing in the same way. While this paper reports that “the translational potential of targeting the BST2/POU6F1 axis is substantial,” it also recognizes that related experiments will have to be performed on other animal models and on human tissues before this line of research could proceed to clinical trials.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Alhamdi, A. A., Mackie, S., Trueman, R. P., & Rayner, M. L. (2025). Pharmacologically targeting Schwann cells to improve regeneration following nerve damage. Frontiers in Cell and Developmental Biology, 13, 1603752.

[2] Gambarotto, L., Russo, L., Bresolin, S., Persano, L., D’Amore, R., Ronchi, G., … & Cescon, M. (2025). Schwann Cell‐Specific Ablation of Beclin 1 Impairs Myelination and Leads to Motor and Sensory Neuropathy in Mice. Advanced Science, 12(5), 2308965.

[3] Manouchehri, Navid, et al. “CD11c+ CD88+ CD317+ myeloid cells are critical mediators of persistent CNS autoimmunity.” Proceedings of the National Academy of Sciences 118.14 (2021): e2014492118.

[4] McClard, C. K., Kochukov, M. Y., Herman, I., Liu, Z., Eblimit, A., Moayedi, Y., … & Arenkiel, B. R. (2018). POU6f1 mediates neuropeptide-dependent plasticity in the adult brain. Journal of Neuroscience, 38(6), 1443-1461.

Producing Thymic Factors in the Liver Rejuvenates Immunity

A new study proposes a novel approach to fighting immune system decline caused by thymic involution: making the liver produce proteins that support T cell development and function [1].

Bringing back the Ts

Immunosenescence, the gradual deterioration of the immune system, is a central aspect of aging. Research has tied it to increased cancer incidence, vulnerability to infections, weak vaccine responses, and so on [2]. Learning how to keep the immune system active and functional would be a huge leap towards meaningful life extension, although some recent studies suggest this would require simultaneously curbing autoimmunity, which also increases with age.

T cells, an important part of the immune system, mature in the thymus, a small organ near the heart. With age, the thymus experiences involution: the functional tissue gets replaced by fat, and the output falls dramatically. This leads to a loss of naive T cells, which are ready to be primed against a specific new pathoges, and a rise in memory and exhaustion-like T cell states, reducing immune resilience [3].

A new study by MIT researchers, published in Nature, suggests a novel approach to solving this problem. “Efforts to counter immune ageing have primarily focused on reversing thymic involution through hormones, cytokines, small molecules and heterochronic parabiosis, or by directly modulating haematopoiesis,” the paper says. “Although these strategies have provided valuable insights into immune ageing, they have been limited by effect size, toxicity or clinical feasibility.”

Instead of trying to rebuild the thymus, the team used the liver to produce factors that are usually made in the thymus and are central to T cell development.

“If we can restore something essential like the immune system, hopefully we can help people stay free of disease for a longer span of their life,” said Feng Zhang, the James and Patricia Poitras Professor of Neuroscience at MIT, who has joint appointments in the departments of Brain and Cognitive Sciences and Biological Engineering.

Increased thymic output

The authors first tried to pin down which thymic support signals actually fade with age. They profiled thymus tissue across many ages and used spatial assays to study cell–cell communication between thymocytes (immune cells maturing in the thymus) and thymic epithelial cells (TECs). The analysis pointed to age-linked weakening of Notch1/3 and IL-7 signaling programs, alongside reduced interstitial FLT3-L in aged thymus and classic involution metrics, such as a decrease in thymus weight.

Based on these findings, the researchers decided to put back the Notch ligand DLL1 in aged hosts along with FLT3-L and IL-7 to create a combined DFI treatment. They focused on the liver, because its protein-synthesis capacity is preserved at advanced ages and blood circulation, including T cells, passes through it.

The team used mRNA encapsulated in lipid nanoparticles (LNPs) to deliver “production instructions” to the liver, rather than simply flooding the blood with recombinant proteins. This was done for several reasons, including the rapid clearance of recombinant cytokines, toxicity issues associated with frequent dosing, and the fact that Notch ligands are transmembrane and normally require cell-cell contact. This means that simply putting them in the blood would not do the trick, as they must be expressed by cells on their surface, which hepatocytes can do.

The four-week DFI treatment of aged mice (~18 months) increased naive T cell counts and improved the naive-to-memory T cell ratio. Crucially, the team showed that these new cells were not just clones of a few old ones (“peripheral expansion”). Analysis of T cell receptor sequences instead supported the idea that more new T cells are being produced, signifying increased thymic output. None of the three factors alone showed the same effect.

Testing the concept

To determine if this actually makes the immune system work better, the researchers used a vaccination model based on ovalbumin, a harmless protein that the immune system can be primed against as if it were a pathogen. Aged mice normally generate fewer antigen-specific CD8+ T cells and show weaker vaccine responses.

Preconditioning with the DFI treatment improved vaccine-induced T cell responses in aged mice, increasing ovalbumin-specific CD8+ T cells significantly. It also preserved a higher naive T cell fraction post-vaccination, supporting more functional immunity.

Then came the big test: cancer. The team challenged aged mice with melanoma (B16-OVA) or colon carcinoma (MC38-OVA) cells and examined how well older animals could control tumors, including in the context of anti-PD-L1 checkpoint blockade, a current state-of-the-art immunotherapy. As expected, aged mice had faster tumor progression and worse survival, and PD-L1 blockade that controlled tumors in adults had little effect in aged cohorts.

In the melanoma model, DFI pre-treatment followed by anti-PD-L1 drove complete rejection in 40% of aged mice, while all controls died within about 3 weeks. In the colon carcinoma model, DFI pre-conditioning (with a short washout) improved endogenous tumor control, increasing spontaneous rejection rates and prolonging survival. In follow-up profiling, DFI was associated with a higher fraction of intratumoral CD8+ T cells and lower expression of exhaustion-associated markers.

Finally, the researchers checked whether DFI might increase autoimmunity. In a mouse model of type 1 diabetes, an autoimmune disease in which the immune system attacks beta cells in the pancreas, DFI didn’t raise blood sugar or make diabetes start sooner, and it didn’t increase self-reactive T cells. The team then ran tests in two additional autoimmunity models, supporting DFI’s immunological safety in those settings.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Friedrich, M. J., Pham, J., Tian, J., Chen, H., Huang, J., Kehl, N., … & Zhang, F. (2025). Transient hepatic reconstitution of trophic factors enhances aged immunity. Nature, 1-9.

[2] Liu, Z., Liang, Q., et al. (2023). Immunosenescence: Molecular mechanisms and diseases. Signal Transduction and Targeted Therapy, 8(1), 200.

[3] Liang, Z., et al. (2022). Age-related thymic involution: Mechanisms and functional impact. Aging Cell, 21(8)

Maximina Yun on the Wonders of the Axolotl

Dr. Maximina Yun, principal investigator at Chinese Institutes for Medical Research in Beijing (CIMR), studies some of the most amazing animals in the world: salamanders, a group of amphibians that includes newts and species such as the universally loved axolotl. On top of being cute, salamanders possess unparalleled regeneration abilities for vertebrates, being able to regrow organs and limbs.

Salamanders are also notoriously long-lived for their body sizes, with axolotls hitting around 20 years while demonstrating negligible senescence and cave olms sporting a mind-boggling maximal lifespan of over 100 years. Studying salamanders is not easy, but the potential rewards are enormous. We talked to Maximina about her interest in these animals and the ways that we can utilize their phenomenal adaptations for ourselves.

How did you become a geroscientist, and what does studying human aging mean to you personally?

I arrived in this area because of my interest in DNA repair. I did my PhD on mechanisms of genome stability and maintenance, and I had a strong interest in cellular plasticity. That is what brought me to do a postdoc in Jeremy Brockes’ lab and begin to understand the mechanisms of cellular plasticity commonly used during salamander regeneration.

Eventually, working with this model, I realized that there is a strong potential for using salamanders to understand the links between regeneration and aging. One of the reasons is that salamanders have long been considered organisms of negligible senescence. This is largely based on mortality studies indicating that the rate of death does not increase with age in salamanders, and actually this is seen in all species studied so far.

Moreover, if you go into the very early literature, you will find reports stating that we are unable to determine a salamander’s age. There is no such thing as differentiating a newt that is one year old or two years old versus one that is twenty years old. That presented a lot of potential, and I became interested from the biological side. Obviously, aging is a pressing challenge – probably the biggest challenge of the 21st century. But for me, as a biologist, what really pulled me into the field was the potential link with cellular plasticity and understanding how these two big processes, regeneration and aging, interplay.

I think salamanders are a phenomenal model. Tell me more about them – their regeneration abilities, the negligible senescence, and all the things they can do that we can’t.

Salamanders are a very special type of organism. They are amphibians, very close to Xenopus (frogs). But while Xenopus loses its regenerative ability through adulthood, salamanders keep it regardless of whether they go through metamorphosis or not. All salamander species reported so far are known to regenerate structures. It is thought that the ancestor of salamanders was able to regenerate. Actually, there is an area in Germany called Pfalz which has fossil records of ancient salamanders at different stages of regeneration. My postdoctoral mentor, Jeremy Brockes, used to have a couple of such fossils in his office and they never failed to leave me in awe.

This ability is remarkable among vertebrates. Particularly, they are the only tetrapods able to regenerate their nearly-full limbs as adults. They can regenerate parts of their brains – in experiments with newts, if you remove half the optic tectum, it will grow back. It takes a long time, over six months, but it will grow back.

They are also able to regenerate their ovaries, and work from my talented student Yuliia Haluza looking at thousands of matings over 15 years in the Dresden axolotl colony indicates that the axolotl retains fertility through lifespan, in keeping with their extreme aging resilience.

multi

Axolotl at Vancouver Aquarium. Photo: Arkadi Mazin

They can also regenerate up to a third of their heart, their tail including the spinal cord, maxillary bones, gills. This is really remarkable. They even regrow structures they don’t necessarily need. For example, the axolotl never undergoes metamorphosis, but it can regrow its lung even though it’s never going to use the lung for breathing on land. It’s not selected for utility, which is interesting. It’s an example of residual regeneration.

They are powerful at regenerating, but there is a catch: in all known cases of regeneration – except for the newt lens, which is a particular example – you always needed the remnant of the structure in order to regrow it. However, our lab has just discovered that the limits of regeneration can be extended further: axolotls are able to regrow their thymus de novo. This means they can regrow their thymus completely from scratch, and it’s the one example of a complex organ that can be fully regrown among vertebrates! This is truly exciting, as it present a completely different regeneration paradigm and further highlights the power of this model for regeneration research.

With regards to ageing, this raises key questions too: does the thymus involute with time in axolotls, as it does in mammals? Is its age-related homeostasis enhanced due to the regenerative abilities? Largely, how does a super regenerator age? Many exciting avenues ahead of us.

When they regenerate their brains, are memory and learning preserved?

We don’t know yet. Until recently, salamanders were not fully experimentally tractable. For example, the axolotl genome only came out in 2018, and the first chromosome assembly for the Spanish ribbed newt – the most tractable newt model – came out just in 2025, an effort led by the labs of Nick Leigh, Andras Simon and mine. Compared with other model organisms, this is all too recent. However, it means we can now truly exploit the wide range of tools existing for more traditional systems. There are memory paradigms currently being developed by a few research groups, both for axolotls and newts, and we are all looking forward to the results.

I think their genome is about ten times larger than the human genome, right?

It is ten times larger. There were a lot of complicatons because most salamanders exhibit genomic gigantism. Largely due to massive expansions of repetitive elements, not genome duplication (actually, the axolotl and the Iberian ribbed newt are both diploid). As they are extremely large and highly repetitive, standard sequencing approaches did not work well. Recent advances in genome sequencing tech, particularly PacBio long-read sequencing, made this possible.

So, for geroscience, it’s a fairly new model, and they also usually have long lifespans, correct?

That’s true. Although different salamanders have very different lifespans. For axolotls, the average is 10 to 13 years of age while their maximum lifespan is about 20 or 21. Newts are significantly longer lived, and then cave olms, such as Proteus anguinus, live well over 100 years! They are all lifespan outliers based on their body size.

But we don’t know for sure, say, from your colonies?

It is in fact from our colonies! We had a 21-year-old, one of the longest-lived axolotls. When the model is so long-lived, it’s difficult to do the studies. With newts for example, there have been a lot of capture-recapture studies in the wild. Critically, whenever a salamander species has been studied in terms of lifespan, one thing is obvious: there is no increase in mortality rate with age.

So, that’s negligible senescence.

Exactly. We classify them as having negligible senescence because this has been tested in several species and none of them exhibit mortality increase with age. But nothing is known at the molecular level, and this is where our lab jumps in: in one huge project, my lab is characterizing how these animals age, and how this is different from senescent species such as us.

I’m wondering if there is a relationship between regeneration and negligible senescence in these species.

That’s exactly what we are working on right now. The first attempt to address this was through building epigenetic clocks, together with Steve Horvath’s team. We found that we can build an epigenetic clock for up to four years of aging in the axolotl, which is the early life. Past that point, it seems to stabilize epigenetically, a phenomenon we have never seen before and could relate to negligible senescence traits.

With the part of the clock that works after four years of age, we used this to ask what happens to limbs or tails which have undergone several regeneration cycles. The answer is that if you compare a forelimb that has regenerated three times versus the contralateral limb that has never regenerated, the one that has regenerated three times is epigenetically younger by DNA methylation age than the contralateral limb.

This suggests there might be some rejuvenation events associated with regeneration, which is exciting because it’s in a natural setting. This organism does it naturally; it’s not reprogramming with Yamanaka factors, and it raises multiple questions which we’re trying to address. The epigenetic and other tools we are currently developing will help providing important answers with regards to the interplay between regeneration and aging.

If I remember correctly, regeneration in forlimbs is linked to cellular senescence.

Cell senescence is elicited every time you regenerate a limb. It appears at particular stages during regeneration, and it contributes to fueling the process, particularly by secreting factors – for example, Wnt signals – that will promote plasticity (in newts) or proliferation of the neighboring cells (in axolotls).

As regeneration progresses, you cannot have these processes turned on forever. We have seen that the number of senescent cells starts to decline as the limb progresses through regeneration. This depends on clearance by the immune system, in particular the macrophages.

We know that senescent cells play a role in wound healing in mammals. Is this some sort of a remnant of this mechanism?

It’s not clear. If you read Marco Demaria’s work – he and the late Judith Campisi uncovered the roles of senescence in wound healing – they are acting by secreting PDGF factors that promote the more rapid migration of the cells to close the wound. This is a different mechanism from what we see in salamanders, which fuels formation and proliferation of the blastema progenitors.

We don’t know exactly if they are related. Whether there are commonalities between this context of “beneficial” senescence that are different from the “negative,” pro-inflammatory senescence you see in aging is a very interesting question. We would love to explore this more with others in the field.

Salamanders also appear to have a superior clearance mechanism for senescent cells, right?

The clearance we see is very effective. Not only do the dynamics of regeneration lead to regenerative limbs that have no senescent cells – you can try to find them, but you won’t – but they never linger. Not even a couple, which is remarkable. We know that macrophages are required for their clearance, but little beyond this. There is a great opportunity here to probe the basis of efficient clearance mechanism, which I am convinced could inform strategies to improve clearance in mammalian systems.

Senescent cells are proposed to play roles in limb development, is it also the case in salamanders?

This is also different from development. If you see a developing limb bud, you won’t see senescent cells, but if you induce regeneration in that developing limb bud, then you will see senescent cells. I sometimes get asked if regeneration is similar to development. I would say it is not a full recapitulation. There are many differences. Senescent cell intervention and usage is different, and there are many other aspects. Of course, some of the patterning programs used to regrow the structure are conserved between development and regeneration, but there are many differences, especially at the beginning of the process.

There’s also dedifferentiation, maybe transdifferentiation of cells during regeneration. It’s a really interesting phenomenon, something like cell reprogramming in vivo.

What we know is that salamanders are masters at regulating cellular identity and plasticity. The cells know very well what they are, and when they go back, they never forget what they are. When a limb regenerates, connective tissue only gives you connective tissue; muscle only gives you muscle.

For example, in the newt, the muscle is generated by dedifferentiation. Even though the muscle comes from dedifferentiation, the resulting regenerative progenitors never fully lose their muscle marks and they only give you muscle.

Experiments with genetic tracing, single-cell analysis, and transplantations tell you the specific tissues overall maintain their identity. Dedifferentiation is also critical for axolotl limb regrow, whereby the connective tissue cells dedifferentiate to give you all CT-derived tissues. This is one of the most important principles when these animals regenerate.

Transdifferentiation is another mechanism these animals use. For example, in the newt lens: you can remove the lens of the eye completely, and the entire lens will come back by a transdifferentiation of the dorsal pigmented epithelial cells from the iris. When you remove the lens, these pigmented epithelial cells undergo transdifferentiation to form crystalline cells that form the lens. This is one example where pure transdifferentiation is used.

But in the context of many other complex structures, we haven’t seen much of this transdifferentiation. There are recent reports, such as an exciting preprint from Wouter Masselink, Elly Tanaka and Prayag Murawala, which indicates that the tail mostly regenerates via specific stem-like progenitors. These stem cell-like cells can give you different tissues in the tail, but it’s a bit different; it’s like playing with the potency of a stem cell, not necessarily direct transdifferentiation as in the lens case.

Your group is also working on thymus regeneration in salamanders. This is exciting since thymus involution clearly plays a role in human aging.

It is really amazing because, first, it’s something you can see with your own eyes. The thymus in the axolotl sits at the base of the gills, formed by three bilateral nodules. You can remove the entire nodules including the connective tissue surrounding them, and, in six to eight weeks, the entire organ will be back. It’s a complex organ because it’s formed by thymic epithelial cells, thymic progenitors, hematopoietic progenitors, dendritic cells, endothelium, macrophages, and obviously the different lymphocytes, and everything just reappears.

We found that when these animals regenerate the thymus, they recapitulate the morphology, cell populations, and function. One can do these very cool experiments where you transplant a regenerated structure from one salamander – a thymus nodule from a salamander that is fluorescently labeled – into a [transparently] white host. Then you can see if this regenerated thymus can support the hematopoiesis and lymphopoiesis of the host.

You can see that it does, because after a year or two, this thymus nodule will still have its original thymic epithelial cells fluorescently labeled, but it will be supporting the entire thymopoiesis of the host. It’s wonderful.

How can we translate insights from these species into humans? What are the main hurdles? I must confess that every time I speak with someone about long-lived or regenerating species, I get envious. I immediately want to have those abilities.

The first thing is to do solid basic science that gets deep into cellular and molecular mechanisms. Understand what really underlies these remarkable traits, and only then you can generate a blueprint for guiding interventions.

Regarding hurdles and timescales, these are intertwined. Primarily, you need to consider the physiology and biological differences between species. Sometimes you may have a target that seems sensible, but it’s actually tied to the particular species’ physiology.

In the case of salamanders, they are ectotherms. They live at different temperatures and control their energy differently. If they’re adapted to live at 20 degrees, can we adapt their mechanisms for an organism that lives at 37 degrees? We have to think about all this. Sometimes you can try to replicate these interventions by playing with the organism’s own proteome. That’s great because you are not subject to these differences.

The biggest hurdle is: do we need one hit or ten hits to reproduce this? But that is just trial and error, leveraging datasets and doing preclinical studies. We have started to take some of our thymus findings into other models to see if we can promote thymus rejuvenation or delay involution by giving them these molecular features that the axolotl has. We’re already doing it, and it’s very exciting.

It’s great to know that you are already looking at the translational angle.

Very much so. This goes hand-in-hand with generating a solid fundamental basis.

What about the evolutionary aspect? Do we know why salamanders have preserved regenerative abilities and mammals lost them almost completely?

It is not necessarily clear in the field of regeneration whether regeneration is a conserved trait of all animals or an acquired trait and an example of convergent evolution. A number of scientists, including my postdoctoral mentor Jeremy Brockes, sustained the latter, supported by the fact that there are salamander-specific proteins involved in regeneration. It is quite possible that regeneration has evolved in salamanders in a different way than in other organisms. For example, if you look at Hydra or planarians, they regenerate in very different ways. It’s not that all organisms that regenerate use the same mechanisms. Different organisms have different solutions.

So, it’s not necessarily settled that we lost the ability to regenerate. There are still many views regarding the evolution of regeneration. Among them is the cancer connection – that organisms that regenerate very well would be more prone to cancer, but that’s not the case in salamanders. They’re actually very resistant to cancer.

They seem to have all the bases covered somehow.

Exactly. And if you think of salamanders as an organism, they occupy almost every niche. They are really evolutionarily successful. For example, you can find salamanders in Siberia. You have newts in the tundra that get frozen and stay dormant in a block of ice for long periods, but when the ice melts they just walk away. There’s a lot of adaptability there.

Obviously, humans are a threat to salamanders worldwide. The axolotl is a good example; in Lake Xochimilco, human activity resulted in water contamination from rapid urbanization and the introduction of carps into the system. They started to wipe out axolotl populations in the natural environment to the point of near extinction in their natural environment (though they exist in high numbers in pet shops and colonies!). Right now, there are efforts towards bringing them back, and we are all rooting for this.

Do you have an opinion on Michael Levin’s bioelectricity-focused research into regeneration?

Bioelectricity is very important. There were early studies in the sixties and seventies showing that reverting currents in the water prevents for example, newt limb regeneration. So, we know that bioelectricity plays an important role. It’s a way in which cells communicate. I think Mike’s work is very interesting. For the field, we want to see this expanded into how the different molecular networks impact bioelectricity and how this is coordinated during regenerative processes. It’s a very interesting angle.

You moved to China recently. How do you see China’s place in the modern geroscience landscape, and how is the research and biotech climate different from Europe?

As in many other sectors, China’s influence is growing at speed. The trend is for science to be heavily influenced by China soon. I think the government has made very right steps in investing in science and deciding China should become the next scientific powerhouse. The environment here is certainly very welcoming, increasingly so towards internationals.

The current international context has led to a significant import of foreign scientists, which are in turn strengthening China’s position worldwide. There are significant funding opportunities here, for both basic research and the biotech sphere, combined with the chance to tap into a pool of talented and super hardworking students and staff.

Things happen at a very fast pace, and the regulatory frameworks for experimentation are much more science-friendly than in Europe. There is still perhaps too much focus placed on high-impact publications, but there is a clear effort towards promoting rigorous science. I’ve been here for four months, so we’ll see how things develop, but what I’m experiencing so far is very exciting.

Do you see the agility that is often invoked with regard to China – things being done faster, more efficiently?

Things are being done much faster than anything I have ever experienced. Sometimes if you have too much speed, that can be counterproductive. But institutes like the one I am in, Chinese Institutes for Medical Research (CIMR), are set up with more than 90% of the faculty coming from Europe and the US, so it’s a bit different.

Personally, I was attracted by the idea conceived by Yi Rao and others to create an HHMI-style institute in Beijing. Here, the support system and evaluation processes are based on the success stories that happen in the West, modeling institutes like the Laboratory of Molecular Biology in Cambridge, where I did my PhD, and Janelia Farm.

One of the features of such places is also the possibility to have resources and time to explore great, bold ideas, and I feel this component is important for doing significant science. Overall, there is a view that this will lead to success here.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Aging: Why Does Evolution Kill?

Why do we age at all, and why do different species age at such dramatically different rates? After decades of longevity research, biologists still disagree on the most basic questions: What is aging? Is it possible to stop or reverse it? And what strategies stand a real chance of working?

Most gerontologists explain aging as the gradual accumulation of cellular and molecular damage, much like rust in a car or the decay of a house. But this viewpoint struggles to explain a series of biological paradoxes. Tiny sparrows can live twenty years. Naked mole-rats survive twice as long. Hydra and planaria appear not to age at all. Queens in eusocial species can live fifty times longer than workers with the same genome. Some animals–jellyfish, comb jellies, and eusocial insects–can rejuvenate, yet they do so only under stress, not under ideal conditions. If youth is mechanistically available, why do they “choose” aging and death?

A new book, titled Aging: Why Does Evolution Kill?, written by Hong Kong–based professor Peter Lidsky, and published with the support of Open Longevity, offers a bold, non-orthodox answer. The book argues that classical evolutionary theories of aging are inconsistent with recent empirical and theoretical results–and develops instead a novel pathogen control theory of aging, in which aging is not just damage, but an evolved, adaptive program.

Early evolutionary thinking, dating back to August Weismann in the late 19th century, proposed that aging is a programmed process that removes maimed individuals from a population. This view fell out of favor because it relied on group selection: individuals supposedly sacrifice their own fitness for the good of the species, a mechanism later considered too weak to explain such costly traits. As a result, for much of the 20th century, theories of programmed adaptive aging were largely abandoned, and non-programmed, damage-based explanations took over.

Lidsky’s pathogen control theory revives the idea of programmed aging but grounds it in kin selection, the same well-accepted evolutionary force that explains parental care. As J.B.S. Haldane quipped, he would give his life “for two brothers or eight cousins”— a vivid illustration of kin selection, in which an individual may sacrifice even its life if this helps relatives, who share its genes, to survive and reproduce.

How, then, could death from aging ever help one’s “two brothers or eight cousins”? The book’s central claim is that the missing piece is chronic, sterilizing infections–pathogens such as syphilis or gonorrhea in humans, which do not kill quickly but prevent reproduction. Individuals carrying such infections become evolutionarily “worthless”: they cannot have offspring, and they can transmit these infections to relatives, harming their genetic interests. In this context, the early death of infected, non-reproductive individuals can be favored by kin selection.

Because the probability of acquiring such infections increases with time, evolution, according to the pathogen control theory, can favor mechanisms that remove older individuals as a function of age. In this view, aging is an immune strategy: a program that sacrifices older individuals to protect their kin from the infections they accumulate over long lives.

This perspective leads to a series of striking, testable predictions. If aging evolved to protect relatives from infection, then the population structure–who interacts and infects whom–becomes a major determinant of lifespan and aging patterns. The book argues that many “outlier” species fit this logic: eusocial insects, naked mole-rats, salmon, flying birds, and bats all have atypical population structures that can explain their unusual aging and death schedules.

In the closing chapters, Aging: Why Does Evolution Kill? explores the implications of the pathogen control theory for modern gerontology. It places aging squarely within the context of the immune system and outlines new research directions that could reshape how we think about interventions to slow or reverse aging. The book presents an ongoing research program: many of its hypotheses remain to be rigorously tested, and readers are invited to evaluate the theory critically.

Whether or not one ultimately accepts the pathogen control theory, this book offers a provocative rethinking of one of biology’s most fundamental problems. It will interest researchers, clinicians, and lay readers concerned with aging and longevity.

https://www.amazon.com/dp/B0G4R3DDH6

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.
Genetics

A Gene That Changes Function With Aging in Mice

In Nature Aging, researchers have discovered growth differentiation factor 3 (GDF3), a cytokine that increases with aging, is related to more inflammatory macrophages in older animals.

Fat and inflammation

We have recently reported on a potential treatment for visceral fat, as this kind of fat is known to generate health problems, including an increase in inflammation [1]. Macrophages are the cause of some of this inflammation, which also makes fat loss more difficult; this effect has been linked to the GDF3 signaling axis [2].

GDF3 has been heavily studied in multiple contexts, including aging. Unsurprisingly, other research has linked it to fat gain [3], but it has also been found to be effective against blood sepsis [4] and even encourages muscle regeneration in older organisms [5]. As a member of the TGF-β superfamily, it acts on SMAD, a group of molecules that modify gene expression and help to manage chromatin [6], which governs the availability of DNA.

GDF3 changes toxic shock response

In their first experiment, the researchers challenged young and old mice with lipopolysaccharide (LPS), a toxic compound that encourages inflammation. Unsurprisingly, the old mice reacted more strongly than the younger mice, reducing their body temperatures and increasing their numbers of inflammatory macrophages compared to other macrophage types. Gdf3, correspondingly, also substantially increased in the older mice.

The researchers then created a breed of mice that don’t express Gdf3. These mice had few differences from their unmodified counterparts, including in adipocytes, and there were no changes in metabolism. However, among older animals, their numbers of inflammatory macrophages were significantly lower than those of wild-type mice, and they did not have the inflammatory phenotype that makes fat burning difficult. Old Gdf3-knockout mice also appeared to have healthier responses to LPS than old wild-type mice; younger mice saw no benefit.

Animals that had Gdf3 knocked out of only their bone marrow (myeloid) cells had significant reductions in multiple inflammatory factors, including IL-1b and IL-6. In old age, these animals also had significant improvements in glucose metabolism, a better ability to burn fat, and less strong reactions to LPS.

The researchers then attempted to treat GDF3 in older mice by using JQ1 to inhibit BDF4, which binds to GDF3. Older mice treated with JQ1 did not develop hypothermia upon LPS injection the way younger mice did, and they had fewer inflammatory macrophages. These results suggest that GDF3 is treatable.

Changing what genes are accessible

These results were found to be directly related to SMAD. Increasing GDF3 levels also increased the phosphorlyzation of SMAD2/3, which was found to lead to the increased inflammation in macrophages. The researchers confirmed this by directly suppressing SMAD3, which stopped the negative effects of GDF3 in the macrophages of old mice but did not affect younger mice. This suppression also inhibited other gene expressions in young mice, but those effects were not found in the older mice, leading the researchers to conclude that SMAD3, and GDF3, affect different pathways with aging.

Further work found that chromatin remodeling was a significant part of this change. Comparing old murine macrophages with and without Gdf3 revealed significant differences in these cells’ chromatin, which significantly altered which genes were accessible. Not only was there a significant decrease in chromatin-related inflammation in the Gdf3-knockout group, there was significant overlap between genetic pathways that were more accessible in the Gdf3-expressing macrophages and genetic pathways related to aging.

The researchers admit their study’s limitations, most notably that this research only involved mice and murine cells, with no human cells being used. Additionally, GDF3 serves vital biological functions, including in the immune system, and its effects on human beings may be different from those on lab mice kept in a controlled environment. Further work will determine if people have the same age-related changes in GDF3/SMAD function as mice do, along with whether or not this compound can be targeted to fight inflammaging and help people live longer.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Carey, A., Nguyen, K., Kandikonda, P., Kruglov, V., Bradley, C., Dahlquist, K. J., … & Camell, C. D. (2024). Age-associated accumulation of B cells promotes macrophage inflammation and inhibits lipolysis in adipose tissue during sepsis. Cell reports, 43(3).

[2] Camell, C. D., Sander, J., Spadaro, O., Lee, A., Nguyen, K. Y., Wing, A., … & Dixit, V. D. (2017). Inflammasome-driven catecholamine catabolism in macrophages blunts lipolysis during ageing. Nature, 550(7674), 119-123.

[3] Wang, W., Yang, Y., Meng, Y., & Shi, Y. (2004). GDF-3 is an adipogenic cytokine under high fat dietary condition. Biochemical and biophysical research communications, 321(4), 1024-1031.

[4] Wang, P., Mu, X., Zhao, H., Li, Y., Wang, L., Wolfe, V., … & Fan, G. C. (2021). Administration of GDF3 into septic mice improves survival via enhancing LXRα-mediated macrophage phagocytosis. Frontiers in immunology, 12, 647070.

[5] Patsalos, A., Simandi, Z., Hays, T. T., Peloquin, M., Hajian, M., Restrepo, I., … & Nagy, L. (2018). In vivo GDF3 administration abrogates aging related muscle regeneration delay following acute sterile injury. Aging cell, 17(5), e12815.

[6] Bertero, A., Brown, S., Madrigal, P., Osnato, A., Ortmann, D., Yiangou, L., … & Vallier, L. (2018). The SMAD2/3 interactome reveals that TGFβ controls m6A mRNA methylation in pluripotency. Nature, 555(7695), 256-259.

Close-look-at-documents.jpg

Analyzing the Quality of Preclinical Anti-Aging Research

Researchers have investigated the reporting quality of preclinical studies’ outcomes in anti-aging research. They analyzed how study quality changed over time, shortcomings in research, and the improvements that can be made in the future in order to yield as many valuable insights as possible [1].

The need for quality

Aging research has grown substantially; however, conducting human trials in the aging field is time-consuming and requires substantial resources. Therefore, initial testing is done in preclinical models, such as mice, worms, fruit flies, and other model animals, as many genes, molecular processes, and aging mechanisms are conserved between those animals and humans [2]. To increase the likelihood of translating results from animal models to humans, high-quality studies are essential.

To assess the quality of preclinical studies in the anti-aging field, the researchers analyzed 667 studies published in peer-reviewed journals between 1948 and 2024, which included 720 experiments, from the DrugAge database. This is “a curated database of preclinical experiments investigating the effects of interventions on aging and lifespan in non-human animals.” The analyzed studies varied in the animal species they used, with a small fraction including more than one model organism. The researchers aimed to assess the quality of reporting, methodological rigor, the distribution of observed effect sizes, and the presence of biases in those studies.

Assessing quality

The researchers assessed the studies using the CAMARADES (Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies) score. This score, which usually involves scoring studies accordingly to a 10-item checklist, allows for assessing methodological quality and risk of bias.

The median CAMARADES score across the analyzed studies was 3, but the researchers observed differences depending on the species used. Only two assessed parameters were consistent across all studies. First, all studies went through peer review. Second, blinding was generally absent. Specifically, blinding to intervention was discussed in only 4% of studies and blinded assessment of outcomes in 3%.

Among the assessed parameters, the researchers noted that almost one-fifth of studies mention randomization. Randomization, along with sample size calculation, was rarely reported when Caenorhabditis worms and Drosophila fruit flies were used, and overall, it was uncommon, with only 6% of all studies reporting it. However, studies using Caenorhabditis and Drosophila almost always gave information regarding the temperature at which animals were undergoing experiments. Temperature information was also common across all experiments, regardless of species used, with over 90% of analyzed studies reporting it. Those who didn’t report it mainly used mice. However, mouse studies did better with other parameters assessed by the researchers.

Other measured parameters included animal welfare, reported in 13.9% of studies, and conflict of interest statements, reported in more than half of the studies.

Since the studies used in the analysis spanned eight decades, the researchers analyzed how reporting changed over time. They noted that reporting of some parameters, especially conflicts of interest, compliance with animal welfare regulations, temperature control, and sample size calculations, increased over time, contributing to an increase in the CAMARADES score. However, there was no significant increase in reporting of randomization and blinding.

multi

The critical parameters

The critical parameter for anti-aging interventions is the timing of initiation, since there is a need for effective interventions that can extend lifespan when taken in mid-life or in the elderly, and the intervention’s effect might differ depending on the start time. However, among the analyzed experiments, the vast majority (over 80%) begin early in life, while only around 8% start at 50% of average lifespan or later, a gap that future studies should address. The researchers also note that, in the pre-clinical studies analyzed, mammal experiments tend to start later in lifespan than non-mammal experiments.

Another critical component in aging research is the animal’s sex. It is well known that there are sex-dependent differences in aging trajectories, and interventions should be assessed in both sexes, as they may respond differently to the same treatment. However, among the experiments analyzed by the authors that included animals that reproduce sexually, fewer than half used both sexes; 35.7% used only males, 12.9% used only females, and some didn’t report the sexes used at all.

multi

Anti-aging compounds

The researchers noted that, among the studies in the DrugAge database, most compounds tested in non-mammalian models increased lifespan.

Additionally, the researchers compared the results between the mammalian and non-mammalian models. They noted that of 35 compounds tested in both mammalian and non-mammalian models, 21 significantly increased lifespan in non-mammalian models, but only one-third of those also significantly increased mammalian lifespan: curcumin, spermidine, epithalamin, D-glucosamine, estradiol, SKQ, and taurine. At the same time, two showed inconsistent results when compared to non-mammalian models, decreasing mammalian lifespan (quercetin and butylated hydroxytoluene). This suggests that in the case of those experiments, “non-mammal results do not seem to reliably predict mammal results, raising further concern for translation.”

The experiments in mammalian and non-mammalian models also differed in other parameters across compounds, including the median percentage increase in lifespan, which was smaller in mammalian models at 7.4% than in non-mammalian models at 17.5%.

Room for improvement

This study suggests that there is room for improvement in the way preclinical antiaging research is performed. The researchers noted that “important design features such as randomization, blinding of intervention, blinded assessment of outcome, compliance with animal welfare regulations, and sample size calculations were infrequently reported, despite evidence that the absence of such features can bias experimental results.” [3,4,5] Some of the most essential experiment design features, such as randomization and blinding, didn’t see substantial improvements over time. They conclude that “generally, most studies did not meet standard reporting guidelines for preclinical experiments.”

While this is not an excuse for failing to meet the standards necessary for high-quality research, those flaws are not limited to anti-aging research, as many studies addressing various diseases exhibit similar reporting and study design problems [6], suggesting a need for improvement.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Parish, A., Ioannidis, J. P. A., Zhang, K., Barardo, D., R Swindell, W., & de Magalhães, J. P. (2025). Reporting quality, effect sizes, and biases for aging interventions: a methodological appraisal of the DrugAge database. npj aging, 11(1), 96.

[2] Kenyon C. (2001). A conserved regulatory system for aging. Cell, 105(2), 165–168.

[3] Schulz, K. F., Chalmers, I., Hayes, R. J., & Altman, D. G. (1995). Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. JAMA, 273(5), 408–412.

[4] Schulz, K. F., & Grimes, D. A. (2002). Blinding in randomised trials: hiding who got what. Lancet (London, England), 359(9307), 696–700.

[5] Kringe, L., Sena, E. S., Motschall, E., Bahor, Z., Wang, Q., Herrmann, A. M., Mülling, C., Meckel, S., & Boltze, J. (2020). Quality and validity of large animal experiments in stroke: A systematic review. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism, 40(11), 2152–2164.

[6] Kilkenny, C., Parsons, N., Kadyszewski, E., Festing, M. F., Cuthill, I. C., Fry, D., Hutton, J., & Altman, D. G. (2009). Survey of the quality of experimental design, statistical analysis and reporting of research using animals. PloS one, 4(11), e7824.

Senescence Exacerbated by Mitochondrial RNA Leakage

Scientists have investigated a little-known mechanism that fuels cellular senescence: mitochondrial RNA leaking into the cytoplasm. Targeting this mechanism showed promise in a mouse model of fatty liver [1].

The new target

The increasing abundance of senescent cells with age has been linked to numerous diseases and is considered a hallmark of aging. Understanding why cells become senescent, and how we can either save them from this fate or mitigate the consequences, is an important target for geroscience.

It has been known that mitochondria in senescent cells leak mitochondrial DNA into the cytoplasm. The cells’ defense mechanisms often mistake it for viral DNA and trigger response mechanisms that exacerbate the cell’s senescent or pre-senescent state [2]. In a new study coming from Mayo Clinic and published in Nature Communications, a team of scientists focused on a different, much less-studied mechanism: the leakage of mitochondrial RNA.

Mitochondria have their own small circular genomes that encode several proteins essential for the organelle’s function. While the transcription and translation of mtDNA differ from those of nuclear DNA, the basics are the same: DNA is transcribed into RNA and then translated into proteins by ribosomes.

Mitochondrial RNA (mtRNA) can sometimes form double-stranded RNA (mtdsRNA), such as when complementary “sense” and “antisense” mitochondrial transcripts overlap. Because cytosolic dsRNA is a classic viral-like danger signal, its appearance outside mitochondria can trip the cell’s antiviral RNA sensors and set off an inflammatory “fire alarm”, which is similar in spirit to mtDNA leakage but occurs via a different sensing pathway [3].

The mtRNA-senescence connection

The team found that mtdsRNA levels are higher in the cytosol of senescent cells (specifically, fibroblasts), which sets off the RNA sensors RIG-I and MDA5. This happened across multiple senescence triggers (replicative, doxorubicin/etoposide) and cell lines. The levels of those RNA sensors also increase with age in multiple mouse tissues, along with the senescence markers p16 and p21 in addition to SASP factors.

To determine cause and effect, the researchers injected non-senescent fibroblasts with purified mitochondrial RNA, which likely contained and/or generated mtdsRNA. This boosted common SASP factors and RNA sensors, suggesting that the presence of mtRNA in the cytosol is enough to drive the SASP program.

They then did something roughly opposite, depleting mitochondria from already senescent cells. As a result, the cells stopped producing the SASP while still staying senescent. The researchers then added purified mtRNA to the cells to see what its impact would be in the absence of working mitochondria. In these mitochondria-depleted senescent cells, mtRNA add-back partially restored interferon/NF-κB inflammatory transcriptional programs – a key element of SASP regulation – rather than fully restoring SASP secretion.

The researchers then pharmacologically reduced mtRNA production in senescent cells by inhibiting mitochondrial RNA polymerase (POLRMT). This lowered cytosolic mtdsRNA, reduced RNA sensors and several SASP components, but did not lower p16 and p21, which can be interpreted as dampening SASP without reverting senescence. Interestingly, blocking STING to blunt mtDNA sensing reduced SASP more than blocking MAVS to blunt mtRNA sensing, and doing both didn’t help further, suggesting that these pathways overlap, with cGAS-STING likely being the main driver of SASP here.

The researchers suspected that mtRNA escape was driven by pores formed by the proteins BAX and BAK in a subset of mitochondria. Indeed, deleting both BAX and BAK reduced cytosolic mtRNA, lowered RNA sensors, reduced MAVS aggregation, and suppressed SASP components.

In vivo validation

The team validated their findings in a mouse model of metabolic dysfunction-associated steatohepatitis (MASH), a dangerous and increasingly prevalent subtype of fatty liver that is often triggered by obesity. They found increased RNA-sensing/SASP signals in livers of MASH mice and then showed that either hepatocyte-targeted Bax deletion or hepatocyte-targeted MAVS knockdown dampens inflammatory and fibrotic markers.

“Liver scarring and inflammation are hallmarks of MASH,” said Stella Victorelli, Ph.D., who is the lead author of the study. If left untreated, it can progress to liver cancer. This is why it’s so important to understand the mechanisms driving the disease so that we can prevent it or develop more effective treatments.”

“With age, we accumulate ‘zombie’ cells, which can lead to more disease,” added João Passos, Ph.D., senior author of the study. “Our idea is that if we can quiet these cells earlier, we can prevent runaway inflammation and the development of many age-related conditions, including liver disease. Understanding the mechanisms that drive disease allows us to target and delay those processes – potentially benefiting more than one condition.”

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Victorelli, S., Eppard, M., Martini, H., et al. (2025). Mitochondrial RNA cytosolic leakage drives the SASP. Nature Communications, 16, 10992.

[2] Victorelli, S., Salmonowicz, H., Chapman, J., Martini, H., Vizioli, M. G., Riley, J. S., … & Passos, J. F. (2023). Apoptotic stress causes mtDNA release during senescence and drives the SASP. Nature, 622(7983), 627-636.

[3] Dhir, A., Dhir, S., Borowski, L. S., Jimenez, L., Teitell, M., Rötig, A., … & Proudfoot, N. J. (2018). Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature, 560(7717), 238-242.

Obese mouse

Targeting a Metabolic Regulator Reduces Fat in Mice

In Aging, researchers have described how they removed visceral fat from older male mice by targeting the metabolic regulatory protein CD47.

A key regulator of metabolism

Visible fat carried around the body is subcutaneous fat. While this kind of fat is obviously not healthy to have in large amounts, it is not as immediately dangerous as visceral fat, which accumulates around organs and drives many metabolic diseases [1]. This metabolic damage is often associated with similar age-related problems [2], including sarcopenia, a loss of muscle mass that leads to frailty [3].

To combat visceral fat, the researchers focus on CD47, a multifunctional membrane protein that increases during aging and is known to drive several age-related disorders, including a loss of the ability to create new blood vessels [4]. Cancers also use it to protect themselves from the immune system [5].

Most relevant to this research, however, is the fact that excessive CD47 has been repeatedly found to lead to age-related metabolic disorders in animal models, including obesity and diabetes [6]. These researchers previously found that a CD47 deficiency leads to the browning of white fat, putting it into a state in which it can be burned for heat, and then encourages that burning [7]. Curiously, further work found that these results only appear to apply to male animals [8].

A potential treatment for fat generation

This work takes that previous research a bit further; while the previous work used modified mice, this uses an actual treatment: an antisense oligonucleotide that specifically targets CD47 (CD47 ASO). A group of 20-month-old wild-type Black 6 mice was injected twice a week, along with a saline-injected group and a control ASO group.

Overall body weight did not significantly change between the groups. Total fat mass, on the other hand, was significantly decreased in the CD47 ASO group compared to either of the controls, and this was accompanied by a significant loss of visceral fat. multiThere was also a marked improvement in diabetes-related biomarkers; the treated mice were better able to handle glucose, and HOMA-IR, a biomarker of insulin sensitivity, was improved. The fat cells themselves had become smaller with this treatment as well.

These findings were accompanied by marked changes in gene expression. While many genes related to fat usage were unchanged, genes related to the formation of fat were significantly downregulated in the CD47 ASO group. Additionally, there appeared to be upregulation of genes related to the anti-inflammatory M2 macrophage type, although most genes related to the pro-inflammatory M1 type were unaffected.

The lack of CD47 discouraging fat cells from forming was confirmed in a cellular study. The researchers grew and differentiated fat cells for 15 days, subjecting some of them to CD47 ASO. The cells so treated were not significantly affected for the first week, but by the end of this experiment, at which point the cells had become senescent due to replication, lipogenesis-related genes were significantly downregulated. A closer examination revealed a more nuanced finding: this approach appears to encourage cells to differentiate into fat cells while preventing senescent cells from accumulating fat.

The researchers noted the very specific effects of this treatment: while there was a slight improvement in the liver’s ability to metabolize glucose, CD47 ASO had no apparent effects on skeletal muscle and other tissues. These effects were sex-specific in mice, and it is not yet clear if they apply to human beings; a clinical trial would need to be done to determine if targeting CD47 could be effective in helping elderly men or women to lose weight.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Shuster, A., Patlas, M., Pinthus, J. H., & Mourtzakis, M. (2012). The clinical importance of visceral adiposity: a critical review of methods for visceral adipose tissue analysis. The British journal of radiology, 85(1009), 1-10.

[2] Tam, B. T., Morais, J. A., & Santosa, S. (2020). Obesity and ageing: Two sides of the same coin. Obesity Reviews, 21(4), e12991.

[3] Nishikawa, H., Asai, A., Fukunishi, S., Nishiguchi, S., & Higuchi, K. (2021). Metabolic syndrome and sarcopenia. Nutrients, 13(10), 3519.

[4] Ghimire, K., Li, Y., Chiba, T., Julovi, S. M., Li, J., Ross, M. A., … & Rogers, N. M. (2020). CD47 promotes age-associated deterioration in angiogenesis, blood flow and glucose homeostasis. Cells, 9(7), 1695.

[5] Sun, J., Chen, Y., Lubben, B., Adebayo, O., Muz, B., & Azab, A. K. (2021). CD47-targeting antibodies as a novel therapeutic strategy in hematologic malignancies. Leukemia Research Reports, 16, 100268.

[6] Maimaitiyiming, H., Norman, H., Zhou, Q., & Wang, S. (2015). CD47 deficiency protects mice from diet-induced obesity and improves whole body glucose tolerance and insulin sensitivity. Scientific reports, 5(1), 8846.

[7] Li, D., Gwag, T., & Wang, S. (2021). Absence of CD47 maintains brown fat thermogenic capacity and protects mice from aging-related obesity and metabolic disorder. Biochemical and biophysical research communications, 575, 14-19.

[8] Li, D., Gwag, T., & Wang, S. (2023). Sex differences in the effects of brown adipocyte CD47 deficiency on age-related weight change and glucose homeostasis. Biochemical and biophysical research communications, 676, 78-83.

Molecule From Chocolate Linked to Slower Epigenetic Aging

A new observational study spanning two human cohorts has found a link between theobromine, a phytochemical abundant in cocoa, and slower epigenetic aging [1].

A yummy geroprotector?

Popular wisdom has it that not many things in life are both healthy and delicious, but according to research, one of them is chocolate. Studies have repeatedly linked chocolate and cocoa consumption with improved health outcomes, including beneficial effects on cholesterol, inflammation, and cellular senescence [2].

Cocoa and coffee share a family of alkaloids called methylxanthines, which includes caffeine, theophylline, paraxanthine, theobromine, and 7-methylxanthine. Coffee is caffeine-heavy, while cocoa is theobromine-heavy.

Theobromine has been reported to extend lifespan in certain strains of C. elegans nematode worms [3] and has been linked to cardiovascular and other health benefits in observational human data [4]. A new study conducted by scientists from King’s College London and several German institutions, published in Aging, looks at theobromine in the context of human aging by analyzing the correlation between the chemical’s blood levels and biological age acceleration.

Two clocks confirm slower aging

The team used two metrics: GrimAge, one of the most robust methylation clocks and trained to predict mortality, and DNAmTL, a DNA methylation-based estimator of telomere length. The latter predicts telomere length in leukocytes from methylation at a specific set of CpG sites.

The researchers pulled together two population cohorts in which people had both blood metabolomics and DNA methylation data. TwinsUK, which includes 509 women with a median age of around 60, served as the discovery cohort. The results were then replicated in KORA, a cohort of 1,160 German adults of both sexes.

They started by measuring the association between the difference between the GrimAge reading and the person’s chronological age (clock acceleration) and six coffee/cocoa-related metabolites: five methylxanthines and the amino acid theanine.

Theobromine immediately stood out, showing the strongest negative correlation. In the researchers’ main model, higher theobromine levels were linked to roughly 1.6 years less GrimAge acceleration per standard step up in theobromine. Theobromine levels were also associated with higher DNAmTL readings, suggesting slower telomere attrition.

Testing the results

The researchers then extensively stress-tested these results. To see if this was just a generic “coffee drinker” signal, they added caffeine and its breakdown products to the model and asked whether the theobromine effect would disappear. It did not; the association with slower GrimAge acceleration stayed, becoming only slightly weaker.

Next, they used penalized regression models (LASSO and elastic net), which automatically shrink or drop less informative variables. Even under these harsher conditions, theobromine consistently remained one of the key predictors of GrimAge acceleration.

In TwinsUK, metabolomics and methylation measurements could be taken up to 5 years apart, so the researchers re-ran the analyses within narrower time windows to see how the association would change. The shorter the distance between the two readings (latency), the stronger the effect size became, strengthening their confidence that this was not just an artifact of long gaps between measurements.

In the KORA replication cohort, higher serum theobromine again tracked with younger epigenetic profiles. After adjustment for age, BMI, blood cell composition, technical factors, and the other methylxanthines, each standard step up in theobromine was linked to about one year of reduced GrimAge acceleration and to slightly longer telomeres.

Because the discovery cohort consisted entirely of women, the authors checked whether the pattern held in women from the KORA study and found a similar, slightly weaker association there, reinforcing their original discovery. In the full KORA cohort, which includes both men and women, the overall effect of theobromine on GrimAge was actually stronger than in women alone. This suggests that men also show a negative association between theobromine and epigenetic age that is at least as strong, if not stronger, than in women.

Dr. Ramy Saad, lead researcher at King’s College London, who is also a researcher at University College London and holds a doctorate in clinical genetics, said: “This is a very exciting finding, and the next important questions are what is behind this association and how can we explore the interactions between dietary metabolites and our epigenome further? This approach could lead us to important discoveries towards ageing, and beyond, in common and rare diseases.”

Dr. Ricardo Costeira, a Postdoctoral Research Associate from King’s College London, added: “This study identifies another molecular mechanism through which naturally occurring compounds in cocoa may support health. While more research is needed, the findings from this study highlight the value of population-level analyses in aging and genetics.”

Caveats and limitations

Being observational and cross-sectional, this study cannot prove a causal relationship between theobromine and aging or even the methylation-based aging metrics that the researchers used. While they adjusted for age, BMI, blood cell counts and several related metabolites, residual confounding by factors like overall diet, lifestyle, socioeconomic status, or other cocoa components, especially flavanols, remains a real possibility.

Both cohorts are European and middle-aged/older, and the discovery sample is composed entirely of female twins, which limits generalization to men, younger people, and other ancestries. Finally, epigenetic clocks and DNAm-based telomere estimates are useful but still imperfect proxies for biological aging. Nevertheless, it’s more encouraging news for chocolate lovers.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Bell, J., Saad, R., Costeira, R., Garcia, P. M., Villicaña, S., Gieger, C., … & Waldenberger, M. (2025). Theobromine is Associated with Slower Epigenetic Ageing. Aging.

[2] Arranz, S., Valderas‐Martinez, P., Chiva‐Blanch, G., Casas, R., Urpi‐Sarda, M., Lamuela‐Raventos, R. M., & Estruch, R. (2013). Cardioprotective effects of cocoa: Clinical evidence from randomized clinical intervention trials in humans. Molecular Nutrition & Food Research, 57(6), 936-947.

[3] Li, H., Roxo, M., Cheng, X., Zhang, S., Cheng, H., & Wink, M. (2019). Pro-oxidant and lifespan extension effects of caffeine and related methylxanthines in Caenorhabditis elegans. Food Chemistry: X, 1, 100005.

[4] Sharifi‐Zahabi, E., Hajizadeh‐Sharafabad, F., Nachvak, S. M., Mirzaian, S., Darbandi, S., & Shidfar, F. (2023). A comprehensive insight into the molecular effect of theobromine on cardiovascular‐related risk factors: A systematic review of in vitro and in vivo studies. Phytotherapy Research, 37(9), 3765-3779.

Suspended stem cells

Human Umbilical Cord Blood Metabolites Lengthen Worms’ Lives

Comparison of human umbilical cord blood with adult plasma revealed hundreds of metabolites whose abundances were age-dependent. Two different formulas, each a mix of a few metabolites, demonstrated anti-senescence properties in cell cultures and model organisms [1].

Young blood has benefits

Previous research has provided evidence that surgically connecting the circulatory systems of young and aged mice, known as parabiosis, in which older animals receive blood from young animals, can lead to reversal of age-associated deterioration in the older animal [2].

Going one step further (or rather, one step younger) is to investigate the properties of human umbilical cord blood and their anti-aging effects, something that has been done by the researchers who just published this study in Aging Cell.

The unique metabolites

Previous studies suggested that human umbilical cord blood has potential applications in anti-aging interventions [3, 4], with some studies showing that human umbilical cord plasma transfusion into aged mice improved their cognitive function [5]. However, no study has compared the metabolite composition of umbilical cord blood and adult blood. Those researchers filled that gap with an analysis of all the metabolites (metabolomics) in human umbilical cord blood, which they compared to adult plasma metabolites.

The researchers recruited 60 mother-offspring pairs with healthy pregnancies and 270 healthy adults, who were divided into groups based on age: young adults (18-25 years), middle-aged individuals (40-55 years), and elderly individuals (65-86 years), and they analyzed the metabolites in their blood. They identified 1092 compounds, with 662 showing significantly different abundances between cord plasma and adult samples.

multi

Further analysis of the identified metabolites revealed 43 molecular pathways that differed between cord plasma and adult plasma, suggesting extensive metabolic changes. Ten of the most differing pathways showed an increase in metabolite abundance in cord plasma. Some of those pathways were linked to amino acid metabolism, biotin metabolism, and pantothenate and CoA biosynthesis, which have been previously shown to positively impact health through their antioxidant properties, promoting wound healing and immune modulation [6-8].

There were 56 metabolites that showed at least a fourfold difference between cord and adult plasma, with 42 of them being more abundant in the cord plasma. Some of those 42 metabolites were also previously reported to have anti-aging properties. One example is inosine, whose antioxidant and anti-inflammatory properties were linked to improvements in learning and memory in aged rats [9]. The 56 metabolites were grouped into nine distinct functional groups, with metabolites that increased or decreased in cord blood forming separate clusters.

multi

Newborn plasma’s anti-aging potential

Previous research suggested that cord plasma metabolites might include some anti-aging properties. Given the apparent differences between cord and adult plasma metabolites, the researchers set out to identify such candidates. They searched for metabolites that were “characterized by significantly higher abundance in cord blood and a declining trend with age progression.” Their analysis yielded 211 potential candidates grouped into 34 distinct pathways, with the most notable related to amino acid metabolism. Some of those metabolites were previously reported to have anti-aging properties; however, many others have not been linked to aging, suggesting potential new approaches and research avenues.

Looking at broader cellular processes, the identified metabolites were grouped into six functional categories: inflammation, oxidative stress, energy and nutrition, proteostasis, DNA damage, and others. The five listed categories are well known to be implicated in aging processes.

To narrow down the list of 211 candidates based on their anti-aging potential, the authors ranked them based on two criteria: “(i) the fold change in abundance between cord and adult plasma, and (ii) the inverse correlation between their abundances and age.” Using those, they created a composite metric for antiaging potential. They identified 42 metabolites that ranked highest on both metrics, making them the most promising candidates. On the flip side, they noted 101 metabolites that had the potential to promote aging, as they were less abundant in cord blood than in adult blood and their levels increased with age.

multi

Validating candidates

The researchers experimentally validated the anti-aging potential of a selected group of metabolites that they identified using cell culture experiments and the model organism C. elegans, a nematode frequently used in aging research. First, they focused on carnosine, taurocholic acid, inosine, L-histidine, and N-acetylneuraminic acid, the five metabolites that ranked highest in their analysis (increased levels in umbilical cord plasma and decline with aging), and they created a formula they refer to as Cord-Formula 1 (CF1).

They treated human embryonic lung fibroblasts with CF1 after first inducing senescence using two different approaches: etoposide treatment, which causes DNA damage, and hydrogen peroxide-induced, oxidative stress-associated senescence. CF1 treatment reduced senescence markers and suppressed the upregulation of senescence-related genes, including SASP genes.

Similar anti-aging effects were seen when the same experiments (induction of senescence in lung fibroblasts) were conducted, but this time the cells were treated with a different formula: Cord Formula 2 (CF2), made of five short peptides. Those short peptides were also identified during the analysis, and their levels were increased in umbilical cord plasma and decreased with age. However, the researchers note that “they have no prior reports of antiaging activity.“

Treating C. elegans with either CF1 or CF2 significantly extended the animals’ lifespan. It also led to reduced markers of aging, improved motor ability (which declines with age), and enhanced stress resistance. There was no negative impact on the animal’s body length or fertility, suggesting a lack of toxicity.

multi

Developing future interventions

Those experiments, while they need to be confirmed in different systems and in humans, provide initial proof-of-concept that some metabolites from the umbilical cord plasma can indeed have anti-senescence and anti-aging activity. What’s more, since those are metabolites that naturally occur in plasma, there shouldn’t be concerns regarding their safety, which should make the development of potential interventions easier and faster.

The authors point out that while their analysis provided insightful new information, there is room for improvement. Metabolite levels have not yet been correlated with people’s broader health and aging profiles. Future studies could increase the number of participants with diverse aging trajectories, ranging from people with comorbidities to centenarians. These studies would allow researchers to assess “whether these metabolites are mere markers of aging or active modulators of healthy aging processes,” how they correlate with various health trajectories, and what kinds of interventions would be most beneficial to populations at different levels of health.

We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

Literature

[1] Liu, J., Jiang, S., Shen, Y., Wang, R., Jin, Z., Cao, Y., Li, J., Liu, Y., Qi, Q., Guo, Y., Wang, Y., Xie, B., Li, J., Cao, A., Wang, Y., Yan, C., Han, Q., Zhu, Y., Peng, J., Dong, F., … Xia, Q. (2025). Human Umbilical Cord Plasma Metabolomics Uncover Potential Metabolites for Combating Aging. Aging cell, e70295. Advance online publication.

[2] Ma, S., Wang, S., Ye, Y., Ren, J., Chen, R., Li, W., Li, J., Zhao, L., Zhao, Q., Sun, G., Jing, Y., Zuo, Y., Xiong, M., Yang, Y., Wang, Q., Lei, J., Sun, S., Long, X., Song, M., Yu, S., … Liu, G. H. (2022). Heterochronic parabiosis induces stem cell revitalization and systemic rejuvenation across aged tissues. Cell stem cell, 29(6), 990–1005.e10.

[3] Mei, Q., Mou, H., Liu, X., & Xiang, W. (2021). Therapeutic Potential of HUMSCs in Female Reproductive Aging. Frontiers in cell and developmental biology, 9, 650003.

[4] Bae, S. H., Jo, A., Park, J. H., Lim, C. W., Choi, Y., Oh, J., Park, J. M., Kong, T., Weissleder, R., Lee, H., & Moon, J. (2019). Bioassay for monitoring the anti-aging effect of cord blood treatment. Theranostics, 9(1), 1–10.

[5] Castellano, J. M., Mosher, K. I., Abbey, R. J., McBride, A. A., James, M. L., Berdnik, D., Shen, J. C., Zou, B., Xie, X. S., Tingle, M., Hinkson, I. V., Angst, M. S., & Wyss-Coray, T. (2017). Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature, 544(7651), 488–492.

[6] Miallot, R., Millet, V., Galland, F., & Naquet, P. (2023). The vitamin B5/coenzyme A axis: A target for immunomodulation?. European journal of immunology, 53(10), e2350435.

[7] Cararo, J. H., Streck, E. L., Schuck, P. F., & Ferreira, G.daC. (2015). Carnosine and Related Peptides: Therapeutic Potential in Age-Related Disorders. Aging and disease, 6(5), 369–379.

[8] Solana-Manrique, C., Sanz, F. J., Martínez-Carrión, G., & Paricio, N. (2022). Antioxidant and Neuroprotective Effects of Carnosine: Therapeutic Implications in Neurodegenerative Diseases. Antioxidants (Basel, Switzerland), 11(5), 848.

[9] Srinivasan, S., Torres, A. G., & Ribas de Pouplana, L. (2021). Inosine in Biology and Disease. Genes, 12(4), 600.