×

The Blog

Building a Future Free of Age-Related Disease

Journal Club

Journal Club November 2022

The Journal Club returns at 12:00 Eastern time, November 29th. Dr. Medvedik will be taking a look at the recent paper entitled “Rapamycin treatment during development extends life span and health span of male mice and Daphnia magna“.

Abstract

Development is tightly connected to aging, but whether pharmacologically targeting development can extend life remains unknown. Here, we subjected genetically diverse UMHET3 mice to rapamycin for the first 45 days of life. The mice grew slower and remained smaller than controls for their entire lives. Their reproductive age was delayed without affecting offspring numbers. The treatment was sufficient to extend the median life span by 10%, with the strongest effect in males, and helped to preserve health as measured by frailty index scores, gait speed, and glucose and insulin tolerance tests. Mechanistically, the liver transcriptome and epigenome of treated mice were younger at the completion of treatment. Analogous to mice, rapamycin exposure during development robustly extended the life span of Daphnia magna and reduced its body size. Overall, the results demonstrate that short-term rapamycin treatment during development is a novel longevity intervention that acts by slowing down development and aging, suggesting that aging may be targeted already early in life.

Join the call using this link.

Rapamycin extends life span & health span in mice & Daphnia

The Journal Club returns at 12:00 Eastern time, November 29th on our Facebook channel. Dr. Medvedik will be taking a look at the recent paper entitled “Rapamycin treatment during development extends life span and health span of male mice and Daphnia magna”[1].

Abstract

Development is tightly connected to aging, but whether pharmacologically targeting development can extend life remains unknown. Here, we subjected genetically diverse UMHET3 mice to rapamycin for the first 45 days of life. The mice grew slower and remained smaller than controls for their entire lives. Their reproductive age was delayed without affecting offspring numbers. The treatment was sufficient to extend the median life span by 10%, with the strongest effect in males, and helped to preserve health as measured by frailty index scores, gait speed, and glucose and insulin tolerance tests. Mechanistically, the liver transcriptome and epigenome of treated mice were younger at the completion of treatment. Analogous to mice, rapamycin exposure during development robustly extended the life span of Daphnia magna and reduced its body size. Overall, the results demonstrate that short-term rapamycin treatment during development is a novel longevity intervention that acts by slowing down development and aging, suggesting that aging may be targeted already early in life.

Literature
[1] Shindyapina, A. V., Cho, Y., Kaya, A., Tyshkovskiy, A., Castro, J. P., Deik, A., Gordevicius, J., Poganik, J. R., Clish, C. B., Horvath, S., Peshkin, L., & Gladyshev, V. N. (2022). Rapamycin treatment during development extends life span and health span of male mice and Daphnia magnaScience advances8(37), eabo5482. https://doi.org/10.1126/sciadv.abo5482
Dividing cancer

Nicotinamide Riboside May Make Existing Cancer Worse

A paper published in the journal Biosensors and Bioelectronics has caused a stir in the longevity community by showing that nicotinamide riboside (NR), a precursor to NAD+, makes tumors more aggressive in a mouse model [1].

Using a novel diagnostic tool

In this new paper, the researchers employed a well-known technique called bioluminescence imaging [2] to quantify NR uptake by cells and tissues, and they meticulously described how this was done.

The researchers were able to demonstrate that their bespoke bioluminescence probe called BiNR can be used to monitor NR uptake in real time both in vitro and in vivo. The purpose of BiNR is to help scientists gain new insights into the activity and effects of NR, and the researchers used it to assess how NR supplementation affects cancer dynamics.

NAD+ has a complex relationship with cancer. On one hand, cancer cells are highly metabolically active and require a lot of energy to grow and divide. For energy production, most cancers switch from cellular respiration to aerobic glycolysis due to the Warburg effect, which leads to an increased demand for NAD+ [3]. Many types of cancer cells have been shown to depend on metabolic pathways regulated by NAD+ [4]. On the other hand, activated immune cells that fight cancer also consume more NAD+, and there is evidence that NAD+ supplementation fights cancer by boosting the immune system [5].

Sample size affects significance

For their experiments in vitro, the researchers used the BiNR probe to quantify NR uptake in four human breast cancer cell lines. The signal from the probe was four times more intensive in triple negative breast cancer (TNBC) cells than in non-TNBC cells. However, the researchers did not show how this compares to non-cancer cells.

Moving to the in vivo stage, the researchers divided several mice into two groups, putting one on a regular diet and the other one on an NR-rich diet. Two weeks later, both groups were inoculated with cells from the MDA-MB-231 TNBC cancer line. By week 10 of the experiment, 7 out of 10 mice in the NR group and 5 out of 9 mice in the control group had detectable tumors: a 27% difference. However, with such a small sample size, this was not statistically significant data, and even a single mouse could have changed the results.

The next experiment, in which the researchers assessed the effect of NR supplementation on the rate of tumor metastases’ formation by injecting MDA-MB-231 cells directly into the heart, was more clear-cut: 9 out of 11 mice in the study group and only 3 out of 12 in the control group developed metastases. This result easily clears the bar of statistical significance.

Interestingly, the researchers also demonstrated the anti-cancer side of NR by showing that its uptake is sharply increased in T cells upon activation. This suggests that both cancer cells and cancer-fighting cells need NAD+ to fuel their increased energy demands, and scientists might have to figure out the way to strike a good balance here.

Conclusion

Growing evidence suggests that NAD+ supplementation has many health benefits, but this study shows that its universal ability to spur cellular activity needs to be carefully studied and considered when devising treatments. It is possible that prior to cancer emergence, NAD+ supplementation fuels the immune system in ways that help prevent cancer, but after the disease is there, it fuels the cancer cells as well. Of course, more research is needed to either support or disprove this hypothesis.

Under normal circumstances, aggressive cancer cells do not suddenly appear in a living organism, and this study did not attempt to assess whether or not NR increases cancer risk in cancer-free mice. This should serve as a reminder that when it comes to science, we should read beyond the headlines and not rush to conclusions.

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] Maric, T., Bazhin, A., Khodakivskyi, P., Mikhaylov, G., Solodnikova, E., Yevtodiyenko, A., … & Goun, E. (2022). A bioluminescent-based probe for in vivo non-invasive monitoring of nicotinamide riboside uptake reveals a link between metastasis and NAD+ metabolism. Biosensors and Bioelectronics, 114826.

[2] Sadikot, R. T., & Blackwell, T. S. (2005). Bioluminescence imaging. Proceedings of the American Thoracic Society, 2(6), 537-540.

[3] Luengo, A., Li, Z., Gui, D. Y., Sullivan, L. B., Zagorulya, M., Do, B. T., … & Vander Heiden, M. G. (2021). Increased demand for NAD+ relative to ATP drives aerobic glycolysis. Molecular cell, 81(4), 691-707.

[4] Gujar, A. D., Le, S., Mao, D. D., Dadey, D. Y., Turski, A., Sasaki, Y., … & Kim, A. H. (2016). An NAD+-dependent transcriptional program governs self-renewal and radiation resistance in glioblastoma. Proceedings of the National Academy of Sciences, 113(51), E8247-E8256.

[5] Morandi, F., Horenstein, A. L., & Malavasi, F. (2021). The key role of NAD+ in anti-tumor immune response: an update. Frontiers in Immunology, 12, 658263.

LBF Logo

Longevity Biotech Fellowship Announces Launch

On Deck Longevity Biotech and the founders of LessDeath.org have collaborated to start the Longevity Biotech Fellowship, a new initiative focusing on bringing upcoming professionals into the longevity industry. The full press release is included below.

Would you like to help accelerate the biggest revolution in the history of medicine? Apply to the Longevity Biotech Fellowship (LBF) — a non-profit community for people to come together to build, join, or invest in revolutionary longevity biotechnology projects.

The longevity biotech industry has seen incredible growth over the past five years, but still represents less than 1% of total biotech market capitalization – and only 0.5% of NIH grants go towards aging biology. This despite the fact that aging is the root cause of most diseases, responsible for the large majority of physical suffering, medical expenses, and death.

To accelerate the growth of this industry, Nathan Cheng (program director for ODLB), Mark Hamalainen and Jun Axup (founders of LessDeath and Longevity Summer Camp) have joined forces to launch this new non-profit community.

The Longevity Biotech Fellowship’s mission is to help talented and mission-driven individuals to get involved in longevity and maximize their impact – and to work together to break bottlenecks to progress.

Our community is looking for talent in science, engineering, software, data, operations, automation, finance, and more! You don’t need a PhD in aging biology to make big contributions – We’ll help you fill the gaps in your knowledge, get connected, and get to work.

Over two dozen industry leading founders, investors, scientists, engineers and operators are volunteering their time as mentors for the Fellowship, including Joe Betts-LaCroix of Retro, Kristen Fortney of BioAge, Omri Amirav-Drory of Renewal Bio, and many more.

New members get an intensive orientation in-person retreat, 12-week online core curriculum, and a year of access to the community platform with weekly new online content, thriving online discussions, IRL meetups, job and startup fairs and more. Our team has a track record of helping people find jobs, make hires, meet co-founders, get funding, and make investments in longevity.

Best of all you’ll be joining a community of like-minded professionals working together to maximize healthy human lifespan.

The deadline for applications for the first cohort of the Longevity Biotech Fellowship is December 31st, with the in-person retreat kicking off the cohort on Jan 12th-16th in Stinson Beach, California — just one hour away from San Francisco.

Admissions are on a rolling basis and spots are limited. Candidates are highly encouraged to apply early at www.longbiofellowship.org!

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.
Brain clock

Correlation Between Cognitive Decline and Epigenetic Clocks

Researchers publishing in Aging have found a correlation between cognitive decline and measurements of epigenetic aging.

Familiar epigenetic clocks

For this study, the researchers used four familiar epigenetic clocks: the first-generation Horvath and Hannum clocks along with the second-generation PhenoAge and GrimAge clocks, which are geared towards predicting morbidity and mortality. They also employed principal component (PC)-based versions of these clocks, which compare systemic variation between epigenetic CpG sites to limit the noise caused by individual sites [1].

The DunedinPoAm and DunedinPACE clocks, which are estimates of the rate of change of biomarkers rather than comparisons to chronological age, were also used.

A midlife study

This study employed data from a total of 48 participants in the Adult Health and Behavior project. Half of the selected participants maintained their cognitive function between assessments taken roughly 16 years apart (Maintainers), while the other half notably declined (Decliners). While their average chronological ages proceeded inerrantly from 44 to 60 at the rate of one year per year, their epigenetic ages were less firm.

On average, the Horvath and GrimAge clocks returned roughly correct comparisons to chronological age, while the Hannum and PhenoAge returned values that were considerably too young: seven and ten years at baseline, respectively. All of the clocks, except PhenoAge, showed that the average participant aged at approximately 4/5 of a year per chronological year.

Interestingly, the PC versions of three of these four clocks showed far different results from the normal versions. While the Horvath clock was largely unaffected, PC-Hannum showed a full fifteen-year increase compared to regular Hannum for both assessments, and PC-PhenoAge and PC-GrimAge returned values ten years greater than their counterparts (placing PC-PhenoAge close to chronological age).

With these average values in hand, the researchers focused on their true metric: the differences between Maintainers and Decliners.

Cognitive decliners age faster

According to the PC-GrimAge clock, the Decliner group experienced two additional years of epigenetic aging compared to the the Maintainer group. The DunedinPoAm and DunedinPACE measurements supported this result. Even when controlling for immune system activity, these results remained statistically significant. The PC-Horvath, PC-Hannum, and PC-PhenoAge clocks did not show significant results.

The researchers then examined the relationship between specific cognitive tests and aging. The Stroop Color-Word test was found to be significantly correlated with PC-GrimAge, and the Trail A-B test was found to be significantly correlated with PC-GrimAge and the Dunedin measurements.

Poor performance on Trail A, Stroop Word, and Matrix Reasoning were also correlated with faster DunedinPACE but not with the other measurements. Working memory measurements did not seem to be correlated. Other measurements of executive function appeared to be correlated with measurements of epigenetic aging, but this data was not statistically significant.

Conclusion

While this study showed significant results, it seems probable that a larger cohort would have produced stronger evidence, providing more statistically significant data and minimizing the effects of statistical outliers. A clock specifically geared towards brain deterioration, such as PCBrainAge, might also have been informative.

Causality was not discussed in this study whatsoever. The effects that cognitive decline and epigenetic aging have on one another, and the existence of common factors that affect them both in similar ways, are still open questions that merit further study.

If the basic biological aspects of long-term cognitive decline can be thoroughly studied, this might lead to treatments that affect them at their root, thus leading to cognitive aging trajectories that are considerably below one year per year.

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] Higgins-Chen, A. T., Thrush, K. L., Wang, Y., Minteer, C. J., Kuo, P. L., Wang, M., … & Levine, M. E. (2022). A computational solution for bolstering reliability of epigenetic clocks: Implications for clinical trials and longitudinal tracking. Nature aging, 2(7), 644-661.

Quantum Healthy Longevity Mission Launch

New Initiative for a More Longevity-Friendly World

Quantum Healthy Longevity Innovation Mission is a new UK-based initiative touting a multi-pronged approach to push for longer lifespans and healthspans for everyone.

Aging is complex

A couple of decades ago, researchers began to realize that aging, like diseases, can be amenable to treatment, at least in model organisms. The modern field of geroscience was born.

Since then, geroscientists have had many successes and some setbacks, but most of all, the realization grew that aging is a multi-faceted phenomenon. While we are zeroing in on common mechanisms of aging that drive multiple age-related diseases at once, the accumulation of damage that underlines aging is caused by many factors and manifests itself in many ways. Air pollution, unhealthy diet, stress, overworking, poor sleep quality, and a myriad of other things accelerate aging. This begins much earlier than we previously thought, and it even occurs in the womb.

Another realization was that aging is a societal as much as a personal problem. The world’s population is quickly graying, and the diseases of aging create an immense burden on the healthcare system. In a paper from last year, the economic benefits from increasing healthspan for just one year by slowing aging were put at a staggering 38 trillion dollars.

Familiar faces, ambitious approach

In recent years, various initiatives have begun to appear that attempt to tie basic research, investment, regulation, policy, environmental issues, and advocacy into a tight knot of cooperation. For instance, we have previously written about the Longevity Biotechnology Association, which wants to bring together researchers, industry CEOs, and investors to forge a common vision.

This time, the announcement came from the UK with some familiar names attached to it. This new initiative, Quantum Healthy Longevity Innovation Mission, is led by people such as longevity social entrepreneur Tina Woods, professors Nir Barzilai, Bryan Kennedy, Nic Palmarini, and Andrew Scott, and Dr. Alex Zhavoronkov. The impressive scope of the project was outlined in an article published in Lancet Healthy Longevity:

We need to take a multipronged whole-of-life approach to tackle all the different, but interacting, drivers of health and disease. These include lifestyle factors, such as diet and physical activity; socioeconomic determinants, such as discrimination, early and lifelong education, training and skills, financial status, and social support; and, increasingly, the characteristics of physical environments, such as green spaces and air quality.

You can also watch the Mission’s launch video or check out the accompanying brochure.

Everything matters 

What do socioeconomic factors such as inequality have to do with longevity? In recent years, the rise in life expectancy that has been a source of pride for humanity (and one of the reasons the world population hit 8 billion just a few days ago) has slowed down, even when not accounting for the devastating effect of the COVID-19 pandemic. In some populations in the US, life expectancy is shrinking, which has been attributed to various environmental factors, from the opioid epidemic to unhealthy eating.

Inequality in life also means inequality in death. As the Lancet paper notes, in the UK, there is a 20-year difference in healthy life expectancy between the richest and the poorest (for the US, it’s 15.5 years). If you want people to live healthier for longer, you can invent a new drug, which would be commendable, or you can help bridge this gap.

This explains why the concept of the “exposome” – the aggregate of all external factors that influence health and well-being – is at the heart of the Mission’s approach. This organization believes that the fight against aging must be fought, to quote a famous Brit, “in the fields and in the streets” as well as in the laboratories. Those are not empty words: among the initiative’s stated goals are “Longevity Cities” (urban environments that are optimized to provide health and well-being for their inhabitants) and regional innovation clusters.

Longevity ecosystem

The initiative is planning to utilize some UK-specific strengths, such as the vast amount of longitudinal health data accumulated by the National Health Service (NHS). British health data repositories such as the UK Biobank are already widely used by geroscientists. Quantum Healthy Longevity aims to create a bank of aging biomarker data and an atlas of geroprotective interventions.

Of course, public policy is an essential ingredient in this all-encompassing formula. A push towards longevity requires favorable changes in regulation, and the Mission will be supporting more progress-friendly biotech and data regulation and finance reforms, making it easier for pensions to invest long-term capital in innovation, for example (which is a part of the “longevity ecosystem” as envisioned by the Mission’s leadership).

To sum things up, the Mission is an ambitious longevity-related and prevention-oriented enterprise aiming to create a strategy and an ecosystem that promotes health and longevity on virtually all levels, from research to regulation and investment to education. “The reason we’re launching this is that we have a health crisis, and not just in the UK, but around the world; we know we need some very different solutions to address this”, Tina Woods said at the launch event, and it looks like there is no shortage of proposed solutions in the Mission’s documents.

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.
Teeth and muscles

Muscle Strength Associated with Oral Health

Researchers publishing in The Lancet have found a relationship between poor oral health and weaker muscles in people who are at least 70 years old.

An established health link

We have previously reported on the relationship between disease and oral health, specifically diabetes, and research has discussed its relationships to walking speed [1] and vascular diseases [2]. While mentioning previous research that has discussed the relationship between oral health and frailty [3], these researchers state that this study is unique in its in-depth examination of the association.

An in-depth questionnaire

Drawing data from 612 participants in the British Regional Heart Study (BRHS) and 1,572 participants in the Health, Aging, and Body Composition (Health ABC) Study at both baseline and a roughly 7 1/2-year follow-up, this research used a panoply of both self-reported and objective measurements. Dentists had examined participants in both studies for markers of peridontitis, including gum pocket depth. Multiple oral health questions were asked, such as dry mouth, sensitivity to hot and cold foods, and difficulty eating.

Muscle strength was assessed with three measurements: sit-to-stand speed, gait speed, and grip strength. Confounding factors, such as socioeconomic status, smoking, alcohol consumption, regular exercise, and age were all considered in this study. While the two studies were not identical in the detail, approach, or timing, most of these measurements and confounding factors was addressed in some way. Unfortunately, only the BRHS had follow-up data relating to oral health, and that was a males-only study.

Substantial association

Even after adjustment for confounding factors, multiple markers of oral health were associated with both slower gaits and weaker grips in the BRHS cohort. The number of teeth remaining in the mouth was substantially associated with weaker grip strength, with people without any remaining teeth being significantly weaker on average. Difficulty eating was also associated with reduced strength.

People with dry mouth symptoms along with other oral health issues were also significantly more likely to have problems with frailty. People whose oral health had substantially deteriorated over the 7 1/2 years were significantly more likely to have problems with chair stand speed, and people with deep peridontal pockets were much more likely to have problems with both chair stand speed and walking gait speed.

Data from the Health ABC cohort corroborated these results. After adjustment, participants in that cohort with at least one oral health issue were more likely to have problems with frailty than people with none, and self-reported oral health was also found to be associated. In both studies, other related markers appeared to be suggestive of a relationship, although they did not meet the threshold for statistical significance.

Conclusion

The researchers hypothesize explanations for these results, suggesting a potential relationship between oral-related systemic inflammation and frailty or the possibility of nutrient deficiency due to problems with chewing. While a causal link remains unproven, this evidence shows that it is highly likely that maintaining good oral health is conducive to maintaining muscle strength.

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] Welmer, A. K., Rizzuto, D., Parker, M. G., & Xu, W. (2017). Impact of tooth loss on walking speed decline over time in older adults: a population-based cohort study. Aging Clinical and Experimental Research, 29(4), 793-800.

[2] Scannapieco, F. A., Bush, R. B., & Paju, S. (2003). Associations between periodontal disease and risk for atherosclerosis, cardiovascular disease, and stroke. A systematic review. Annals of Periodontology, 8(1), 38-53.

[3] Kotronia, E., Wannamethee, S. G., Papacosta, A. O., Whincup, P. H., Lennon, L. T., Visser, M., … & Ramsay, S. E. (2019). Oral health, disability and physical function: results from studies of older people in the United Kingdom and United States of America. Journal of the American Medical Directors Association, 20(12), 1654-e1.

Protein foods

Protein Content in an Optimal Diet

In a new study published in Nature Communications, researchers have designed guidelines for dietary amino acid consumption for optimal health [1].

Protein matters

Diet is an important component of a healthy lifestyle and is believed to play a major role in health status and longevity. It is also one of the most discussed topics among wellness enthusiasts, biohackers, and pretty much everyone else.

While the optimal diet is a complicated subject that is still being scientifically debated, various countries have developed official dietary guidelines. Although they differ in presentation, they all encourage increasing diversity and nutrient density in dietary choices.

Dietary guidelines also focus on reducing the consumption of added sugars and trans- and saturated fats. Meanwhile, there are no specific recommendations regarding proteins, which are viewed as a single entity despite being composed of different amino acids playing different physiological functions.

Moreover, similarly to carbs and fats, amino acids are categorized into subgroups: essential, nonessential, ketogenic, branched-chain, and glucogenic.

In this study, the researchers sought to explore the variability of amino acids in foods and diets. They also analyzed if the consumption of specific amino acids is associated with common age-associated chronic diseases.

Profiling foods and diets

First, the researchers made a total of 2,335 amino acid profiles of various foods such as eggs and pork. They show that glutamate/glutamine and asparagine/aspartate are the most abundant in human foods. On the other hand, cystine and tryptophan are the least abundant.

Interestingly, amino acid abundance variability in human foods is on par with that of fats and carbs. In addition, there is a high amino acid variability among plant- and animal-based foods. Some amino acids, such as methionine, vary greatly in different foods: they comprise 3.1% of the amino acids in eggs but only 1.3% in legumes.

Next, the researchers constructed amino acid profiles of 10 dietary patterns: Mediterranean, Japanese, vegetarian, plant-based, DASH, Paleo, ketogenic, Atkins, American (Western), and USDA (2015-2020 dietary guidelines for Americans).

They show that the amino acid signatures of these diets vary greatly. Surprisingly, even diets similar to each other, such as vegetarian and plant-based, have different amino acid profiles: a 30% difference in methionine abundance was found between these dietary patterns.

At the same time, plant-based diets are characterized by a lower abundance of lysine, histidine, and methionine compared to meat-heavy dietary patterns. Overall, the researchers show that the biggest difference across dietary patterns is not in fat or carb content but in amino acids instead.

Profiling individuals

Next, the researchers analyzed amino acid intakes of >30,000 people of diverse cultural and ethnic backgrounds based on the National Health and Nutrition Examination Survey (NHANES) 2007-2014. They supplemented self-reported data, such as protein intake, with corresponding blood metabolite measurements, such as urea nitrogen concentration.

Following a few more consistency checks, the researchers found that the amino acid intake correlated with age, but not other demographic features. They then analyzed the link between individuals’ amino acid intake profiles and the prevalence of such chronic diseases as hypertension, obesity, cancer, and diabetes.

The strongest association was shown to be between amino acid intake composition and obesity. For example, obese people were more likely to consume foods rich in methionine and poor in tryptophan. Surprisingly, the association between consumed amino acids and diseases was stronger than between diseases and carbs or fats.

Moreover, the researchers show that the prevalence of all four diseases can be predicted from an individual’s nutrient intake, particularly from the amino acid profile. Therefore, the authors use these results to suggest guidelines for an optimal diet.

The optimal diet

The researchers used an AI-based approach to develop amino acid intake guidelines. Such a ‘perfect diet’ would have to be low in methionine and glycine, and high in phenylalanine, tryptophan, and valine. Unexpectedly, none of the existing diets considered in this study satisfied these conditions.

The ultimate outcome of this study is an AI-guided tool that can design a personalized diet based on user-defined preferences and a reference ‘optimal’ combination of amino acids.

Abstract

Studies at the molecular level demonstrate that dietary amino acid intake produces substantial effects on health and disease by modulating metabolism. However, how these effects may manifest in human food consumption and dietary patterns is unknown. Here, we develop a series of algorithms to map, characterize and model the landscape of amino acid content in human food, dietary patterns, and individual consumption including relations to health status, covering over 2,000 foods, ten dietary patterns, and over 30,000 dietary profiles. We find that the type of amino acids contained in foods and human consumption is highly dynamic with variability far exceeding that of fat and carbohydrate. Some amino acids positively associate with conditions such as obesity while others contained in the same food negatively link to disease. Using linear programming and machine learning, we show that these health trade-offs can be accounted for to satisfy biochemical constraints in food and human eating patterns to construct a Pareto front in dietary practice, a means of achieving optimality in the face of trade-offs that are commonly considered in economic and evolutionary theories. Thus this study may enable the design of human protein quality intake guidelines based on a quantitative framework.

Conclusion

This groundbreaking study has revealed that the amino acid content of foods is an important aspect of an optimal diet. However, any causal link between the considered diseases and amino acid intake was not explored. Nevertheless, it could be insightful to use the tool developed in this study and get a personalized diet designed by AI.

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] Dai, Z., Zheng, W. & Locasale, J. W. Amino acid variability, tradeoffs and optimality in human diet. Nat. Commun. 13, 6683 (2022)

NMN pills

FDA Purges NMN From the List of Supplements

Reversing its own earlier decision, the FDA has informed an NMN manufacturer that the molecule can no longer be marketed as part of dietary supplements. Another company that is testing NMN as a drug probably contributed to the decision, which is not being enforced as of now.

NAD and NMN

NAD+ is a multi-role coenzyme central to metabolism. It facilitates energy production in cells and acts as a substrate for important enzymes, such as sirtuins and PARP. NAD+ levels decline with age, contributing to multiple age-related diseases [1]. Studies have confirmed various health benefits of NAD+ supplementation in animal models [2], although human trials are scarce.

NAD+ levels can be boosted via its precursors, which include nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN). NMN-containing supplements have been gaining popularity in the longevity community, even though not everyone agrees on their effectiveness or safety. NMN naturally occurs in foods such as edamame, broccoli, cabbage, cucumber, and avocado.

Allowed, then banned?

Considering NMN’s popularity, the news that the FDA has effectively banned its use in dietary supplements came as a shock, especially since this had nothing to do with the molecule’s efficacy or safety.

To market a compound in dietary supplements, a company must file a new dietary ingredient (NDI) notification with the FDA. Explicit approval is not required, but if the FDA raises objections, this precludes the compound from being used in supplements.

While NMN has been sold in some markets (mainly Japan) since at least 2014, it is a relative newcomer to the US. In the past two years, several companies have filed NDI notifications, and FDA did object to them on various grounds, mostly claiming that there is insufficient proof that NMN supplementation is safe.

In 2021-2022, new human NMN studies have been published [3], which might have alleviated FDA concerns. In May, the FDA acknowledged an amended NDI notification from the China-based company SyncoZymes without objections.

However, recently, responding to another NDI notification from the company Inner Mongolia Kingdomway, the FDA changed course and declared that ß-NMN (essentially NMN) cannot be marketed as a supplement due to it already being investigated as a drug.

The FDA decision was based on a provision in the Dietary Supplement Health and Education Act of 1994 (DSHEA) stating that a dietary supplement cannot contain an ingredient that has been “authorized for investigation as a new drug for which substantial clinical investigations have been instituted and for which the existence of such investigations has been made public”, unless the investigations began after the ingredient had been approved for use in supplements.

A request from a pharma company

The reason for the reversal is unknown, but as reported by Natural Products Insider, in December 2021, Metro International Biotech, a startup pharmaceutical company, wrote to the FDA: “As a company that has instituted publicly available clinical trials on ß-NMN, we request that FDA take the preclusion provision … seriously to protect the rights of companies that have spent significant time and research to develop drug products from competition from dietary supplements”. According to Insider, the FDA confirmed that it had considered Metro’s request.

Metro International Biotech, co-founded by Rajendra Apte and David Sinclair, is targeting several diseases with NAD+ precursors. It has developed a proprietary version of ß-NMN called MIB-626 and asked the FDA to approve its investigation as an investigational new drug (IND).

The earliest information about an MIB-626 trial found on ClinicalTrials.gov is dated September 2021. Apparently, at least two of the three criteria needed to trigger the exclusion clause were satisfied: MIB-626 is a subject of “substantial clinical investigations” that “has been made public”. However, it is not clear when exactly the drug was authorized for investigation. According to the FDA, this depends on when the IND authorization goes into effect, but surprisingly, the FDA does not disclose this information, and it also does not immediately become public.

Will the FDA enforce this?

The FDA can decide whether to enforce its own rulings on supplements. In a notable example, NAC (N-acetyl-L-cysteine), was excluded from the definition of a dietary supplement on similar grounds in 2021, but several months later, in response to an outcry that included citizen petitions and litigation, the FDA decided to exercise enforcement discretion with regard to NAC, effectively leaving its manufacturers alone for now.

While the law (DSHEA) does not provide a rationale for the exclusion clause, it appears like it is there solely to protect pharma companies that pour massive resources into investigating new drugs. However, some could see this as unfair towards supplement companies and their clientele. In this particular case, the companies were engaged in months of back-and-forth with the FDA, addressing various concerns and amending their proposals only to have them dismissed on entirely different grounds.

Currently, there is no sign of any NMN-containing supplements being taken off the shelves, and it is not clear whether the FDA will ultimately choose to enforce its ruling. Manufacturers of NMN supplements will likely continue pressing the FDA to exercise enforcement discretion, with Metro Biotech pulling in the opposite direction.

Metro Biotech did not respond to a request for comment. An FDA spokesperson contacted by Lifespan.io did not explain the rationale behind the exclusion clause, adding that the FDA “does not comment on whether or not it has or will commence enforcement actions.”

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] Imai, S. I., & Guarente, L. (2014). NAD+ and sirtuins in aging and disease. Trends in cell biology, 24(8), 464-471.

[2] Mills, K. F., Yoshida, S., Stein, L. R., Grozio, A., Kubota, S., Sasaki, Y., … & Yoshino, J. (2016). Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell metabolism, 24(6), 795-806.

[3] Yoshino, M., Yoshino, J., Kayser, B. D., Patti, G. J., Franczyk, M. P., Mills, K. F., … & Klein, S. (2021). Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science, 372(6547), 1224-1229.

Mice eating

Arginase Inhibitor Improves “Leaky Gut” in Old Mice

According to a new study, the age-related increase in intestinal permeability that drives inflammation can be alleviated by inhibiting the enzyme arginase, a regulator of nitric oxide production [1].

Gut feeling

While age-related sterile inflammation (inflammaging) is considered one of the hallmarks of aging and a major cause of age-related diseases [2], scientists still don’t fully understand what causes it. Inflammation is a part of the immune response, and “sterile” means that there is no obvious reason for the immune system to react that way.

Recently, studies began pointing to the age-related increase in intestinal permeability as a possible culprit [3]. The gut is a front line that is constantly attacked by pathogens from food, air, and the vast population of intestinal bacteria. Intestinal epithelial cells must be able to let nutrients in while keeping those pathogens out. Like many other bodily functions, this one declines with age. The gut becomes “leaky”, and pathogens enter circulation and cause an immune response, or so the theory goes.

More permeability with age

In this new study, the researchers investigated some aspects of this theory in humans and mice. First, they ran blood tests on 16 young healthy and 16 old healthy participants. Having healthy participants allowed the researchers to determine whether intestinal dysfunction appears prior to any known diseases of aging. None of the subjects suffered from obesity, cardiovascular diseases, type 2 diabetes, non-alcoholic fatty liver, chronic inflammatory diseases, or malignancies, and none of them were taking medications to control any of those.

Indeed, despite older subjects being healthy, several markers of intestinal permeability in their blood were significantly elevated compared to young controls, including bacterial endotoxins and the CD-14 protein, which enhances the body’s reaction to those toxins.

The same picture was seen in mice. In 24-month-old animals, bacterial endotoxins were more than twice as abundant than in younger controls. While intestinal morphology did not significantly differ between the two groups, both protein and mRNA levels of the tight junction proteins occludin and ZO-1 were significantly lower in the proximal small intestine of old mice. Tight junctions are connections between cells that hold them together, not unlike nails or rivets. Markers of cellular senescence were higher, and stem cell-related markers, such as telomerase, were lower in older animals. However, no such age-related differences were observed in the distal small intestine and colon.

No, it’s NO

Emerging research has attributed intestinal permeability to age-related changes in microbiotal composition, which indeed differed significantly between young and old mice. However, transplantation of microbiota from young to old mice failed to alleviate intestinal permeability in the latter. This stands in contrast with some recent studies in which various benefits of microbiotal transplantation were reported.

Nitric oxide (NO) is a simple but important molecule. Its healthy levels are associated with cardiovascular health, and it plays an important role in maintaining intestinal integrity [4]. With age, intestinal NO metabolism gets disrupted, while levels of arginase, a critical regulator of NO synthesis, go up, which is also what happened to the mice in this study.

Mouse intestinal barrier

Hence, downregulating arginase is a possible way to alleviate age-related intestinal damage. One previous study found that genetic deletion of arginase attenuates the onset of senescence and extends lifespan in mice [5]. This time, researchers used an arginase inhibitor rather than genetic engineering.

Arginase inhibition improves “leaky gut”

Treating old mice with the arginase inhibitor norNOHA for six weeks improved intestinal barrier function, lowered bacterial endotoxin levels, and decreased senescence markers in the liver while simultaneously increasing NO production.

Interestingly, despite those improvements, intestinal microbiotal composition was similar between old mice that had been treated with an arginase inhibitor and those that had not. The researchers hypothesize that “maintaining intestinal barrier function in aging may not depend solely on intestinal microbiota”, and that NO homeostasis in small intestinal tissue, regulated by arginase, is probably an important factor as well.

Here, we report that even in healthy older men low grade bacterial endotoxemia is prevalent. In addition, employing multiple mouse models, we also show that while intestinal microbiota composition changes significantly during aging, fecal microbiota transplantation to old mice does not protect against aging-associated intestinal barrier dysfunction in small intestine. Rather, intestinal NO homeostasis and arginine metabolism mediated through arginase and NO synthesis is altered in small intestine of aging mice. Intestinal arginine and NO metabolisms could be a target in the prevention of aging-associated intestinal barrier dysfunction and subsequently decline and ‘inflammaging’.

Conclusion

This study supports the idea that with age, the intestinal barrier loses integrity even in healthy people, which is likely to be a driver of inflammaging. Another important finding is that age-related changes in microbiota composition may not be the main cause of age-related intestinal dysfunction, and, consequently, microbiotal transplantation may not be the optimal solution. Slowing the age-related decrease in intestinal NO production via arginase inhibition, however, is an intriguing, novel strategy that deserves further research.

Literature

[1] Brandt, A., Baumann, A., Hernandez-Arriaga, A., Jung, F., Nier, A., Staltner, R., … & Bergheim, I. (2022). Impairments of intestinal arginine and NO metabolisms trigger aging-associated intestinal barrier dysfunction and inflammaging´. Redox Biology, 102528.

[2] Ferrucci, L., & Fabbri, E. (2018). Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nature Reviews Cardiology, 15(9), 505-522.

[3] Kavanagh, K., Hsu, F. C., Davis, A. T., Kritchevsky, S. B., Rejeski, W. J., & Kim, S. (2019). Biomarkers of leaky gut are related to inflammation and reduced physical function in older adults with cardiometabolic disease and mobility limitations. Geroscience, 41(6), 923-933.

[4] Mu, K., Yu, S., & Kitts, D. D. (2019). The role of nitric oxide in regulating intestinal redox status and intestinal epithelial cell functionality. International Journal of Molecular Sciences, 20(7), 1755.

[5] Xiong, Y., Yepuri, G., Montani, J. P., Ming, X. F., & Yang, Z. (2017). Arginase-II deficiency extends lifespan in mice. Frontiers in physiology, 8, 682.

Swimming mouse

The Effects of Niacin on Alzheimer’s in Mice

A study published in the Annals of Translational Medicine has shown the effects of niacin supplementation on a mouse model of Alzheimer’s and outlined the genes involved.

Niacin’s known effects

Niacin supplementation has been previously researched in its effects on neurological diseases: we have discussed its effects on Parkinson’s and brain cancer, and the related compound NMN has been shown to improve neurovascular coupling.

While dietary interventions have been examined for their potential in treating or preventing Alzheimer’s, such as the Mediterranean diet [1] and a ketogenic diet [2], niacin was not a prominent factor in that research. The researchers also note that most research related to NAD+ focuses on the salvage pathway of NMN and NR rather than the synthesis pathway of niacin.

A well-known transgenic model

To test their approach, the researchers employed six wild-type mice, six transgenic Alzheimer’s-prone mice given niacin supplements in drinking water, and six transgenic Alzheimer’s mice in the control group. All of these mice were 8 months old.

After six months of feeding, the mice were tested with the Morris water maze test, which tests the ability of mice to find and stand on a platform in water. While all the Alzheimer’s-prone mice performed similarly on the first day, the ones given niacin learned more quickly, having latency times more akin to those of wild-type mice.

These results matched the effects on gene expression. As expected of an NAD-related intervention, genes involving RNA and mRNA were affected, but many of the most prominent effects were on neuronal growth, development, and density. Signaling pathways were also affected.

One of the most prominent genes affected was Ctnnb1, a gene that is part of the Wnt signaling pathway. Mutations in this gene, and this pathway, are related to degenerative disorders. While the biology is complicated and the hypothesis is not proven, the researchers tie together this pathway and energy metabolism, which NAD+ has strong effects on. They suggest that energy metabolism, along with a reduction in inflammation and a reduction in amyloid beta, are the means by which niacin affects the progression of Alzheimer’s.

Conclusion

This research was done on a transgenic mouse model, as mice do not normally get Alzheimer’s disease. While specific genes were identified, whether this research can translate to humans is unclear. However, if niacin or another approach focused on NAD+ can affect the progression of Alzheimer’s disease, it will be a welcome gift to a community in desperate need of disease-modifying 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] Norwitz, N. G., Saif, N., Ariza, I. E., & Isaacson, R. S. (2021). Precision nutrition for Alzheimer’s prevention in ApoE4 carriers. Nutrients, 13(4), 1362.

[2] Neth, B. J., Mintz, A., Whitlow, C., Jung, Y., Sai, K. S., Register, T. C., … & Craft, S. (2020). Modified ketogenic diet is associated with improved cerebrospinal fluid biomarker profile, cerebral perfusion, and cerebral ketone body uptake in older adults at risk for Alzheimer’s disease: a pilot study. Neurobiology of aging, 86, 54-63.

Quantum Healthy Longevity

The Quantum Healthy Longevity Innovation Mission Launches

London. 14 November 2022.  The National Innovation Centre for Ageing and Collider Health launch the Quantum Healthy Longevity Innovation Mission with a keynote by Lord Bethell, former Parliamentary Under Secretary of State for Technology, Innovation and Life Sciences, to shift ‘sickcare’ to a new model based on proactive prevention, with the ever-growing recognition that wealth of nations is not possible without the health of populations.

We need healthcare not sickcare

It is no longer possible to afford a healthcare system based only on response to illness.  Society needs citizens to be more aware and supported in their health and behavioural choices across the life course, as well as a productive workforce equipped to respond to ongoing challenges and stresses. In response, leaders are coming together to radically reimagine short, medium and long-term responses at a global, societal and individual level and significantly invest in an interdependent ecosystem for science and innovation to accelerate healthy longevity at quantum scale and pace [1].

As we are progressively seeing with research on the psychology, sociology and biology of ageing, age-related chronic diseases are not inevitable or untreatable. We need to take a multipronged ‘whole of life’ approach to tackle all the different but interacting drivers of health and disease – lifestyle factors, including diet and physical activity, socioeconomic determinants, such as discrimination, early and lifelong education, training and skills, financial status, and social support; and, increasingly, the characteristics of physical environments, such as green spaces and air quality, need more attention.  In particular, continuous interactions with and cumulative exposures to the surrounding physical and social environments throughout life have vital roles in later-life outcomes.

The Quantum Healthy Longevity blueprint aims to harness such opportunities, address urgent needs and fulfil aspirations of the UK Innovation Strategy to tackle big, real-world problems in climate and health, by harnessing developments in the understanding of the exposome, the system of all external factors that influence our health and wellbeing trajectory [2].

We are on the cusp of transformational change with technologies that can give a massive step change in delivering healthy longevity for all, taking advantage of the UK’s unique strengths such as the world’s largest longitudinal cradle-to-grave dataset from NHS healthcare records. Insights from data outside the NHS dataset, such as genetics, biological, behavioural, social, environmental and financial data are also under-utilised and there are significant opportunities to use AI and multimodal learning to predict disease and incentivise healthier living using ‘life data’.

The mission aims to unlock the innovation economy and help drive UK’s aspirations to be a science and technology superpower, including helping biotech investment move from 2.0 (investing in ‘sickcare’) to 3.0 (investing in prevention) and leveraging the immense potential of intelligence to create the world’s leading test bed for health, powered by public and private sector investment to increase healthy longer lives for everyone.

“The time has come for action: to harness and develop a shared intelligence combining the intelligence of people, the intelligence of research and business, and the intelligence of data to transform what we all already know and accelerate what we have yet to understand about what impacts us across the course of our lives. At NICA we framed our mission in ‘adding intelligence to ageing and longevity’ and this is our contribution to the Quantum Healthy Longevity Mission, hoping that the many outstanding UK and international entities will join us to take the steps necessary for a quantum leap forward” said Nic Palmarini, Director of the UK National Innovation Centre for Ageing.

“There is clear citizen demand for this.  People are incredibly enthusiastic about the endless possibilities of science, technology and innovation and how this investment can massively help people and planet.  But they want action – and more, more join up, less waste.  They want to see solutions based on what they need, that they can afford and that can help them know what to do to live healthier better lives” said Professor Lynne Corner, COO of the National Innovation Centre for Ageing and Director of VOICE.

“The Mission takes an exponential  approach leveraging data from the ‘exposome’ – reflecting the external stresses and factors that influence our health- to understand how best humans can flourish in their environments.  The abundance of data that can be captured and analysed on how we behave and respond means we can better understand the complex patterns that link our environment, behaviours and health and yield critical insights into the underlying causes of age-related diseases- including dementia, the disease many of us fear most.” Says Tina Woods, CEO of Collider Health and Healthy Longevity Champion for the UK National Innovation Centre for Ageing.

Literature

[1] Woods, T; Palmarini N, Corner L et al. Quantum Healthy Longevity for healthy people, planet and growt. Lancet Healthy Longevity November 13 2022. https://doi.org/10.1016/ S2666-7568(22)00267-7

[2] Christopher Paul Wild; Complementing the Genome with an “Exposome”: The Outstanding Challenge of Environmental Exposure Measurement in Molecular Epidemiology. Cancer Epidemiol Biomarkers Prev 1 August 2005; 14 (8): 1847–1850. https://doi.org/10.1158/1055-9965.EPI-05-0456.

You can find more information here: https://issuu.com/nationalinnovationcentreforageing/docs/quantum_brochure_v.0.2_print

Please contact Tina Woods on tina.woods@colliderhealth.com for more information.

Protein

Protein Intake Increases Bone Density in Older Adults

Scientists have found that animal protein consumption positively correlates with bone density in older adults. Things become more complicated regarding plant protein [1].

 

Too little or too much?

Just how much protein people should consume has been a point of contention. Decades-old research, performed mainly in underdeveloped communities, shows that protein deficiency can be harmful [2]. Since then, the prevalence of this problem has greatly diminished, and the opposite, protein overconsumption, has come into the spotlight.

A widely cited study from 2014 showed that low protein intake is associated with a major reduction in cancer and overall mortality in people younger than 65. However, the opposite was true for older people [3]. This led the authors to suggest that “low protein intake during middle age followed by moderate protein consumption in old subjects may optimize healthspan and longevity.” The study also showed that the increase in mortality only correlated with animal protein intake.

Research in animals seems to support this hypothesis. Protein restriction has emerged as one of the few proven methods of extending lifespan in various model organisms, including rodents [4]. Clinical studies in humans are scarce, but one from last year showed that protein restriction can alleviate metabolic dysfunction [5].

Animal vs plant protein

Even if high protein intake might be overall unhealthy, protein is known to be important for bone health, which deteriorates with age, leading to osteoporosis and frailty. This new study looked for a connection between protein intake and bone density in elderly people by analyzing results of four earlier trials.

First, the scientists performed a cross-sectional analysis of the studies’ population: they analyzed baseline values of protein intake and bone mass density (BMD) for all 1570 participants. Participants had a median age of 71, and 56% were female. In their analysis, the researchers adjusted for several confounding factors: age, sex, physical activity level, smoking status, and alcohol intake.

Both total protein intake and animal protein intake were associated with higher total body and spine BMD, while plant protein intake showed a negative correlation. Interestingly, a stronger association was detected between total protein intake and spine BMD in females than in males, but the opposite was true for plant protein intake, which seemed to be more detrimental for spine BMD in males than in females.

In a subgroup analysis of participants with an adequate calcium intake and serum vitamin D levels, the association between total protein intake and BMD became stronger. According to the researchers, this could mean that animal protein improves bone density independently of calcium and vitamin D, as it might be more digestible and have a more complete amino acid profile (some plant-based foods, such as quinoa, also contain all essential amino acids).

However, the negative association between BMD and plant protein intake became insignificant after adjusting for calcium and vitamin D. This, in turn, could mean that plant protein isn’t inherently harmful for bone health, but its consumption negatively correlates with calcium and vitamin D intake.

Little effect from short-term interventions

The researchers then performed longitudinal analyses of the interventions used in the earlier trials. The largest of the four, NU-AGE, did not involve a ‘true’ protein intervention. Instead, its participants were asked to adhere to the Mediterranean diet. Consequently, it was excluded from the longitudinal analysis, which left the researchers with just 340 participants that did receive various protein interventions. However, none of those interventions, which spanned between 12 and 24 weeks in length, significantly improved BMD.

In one trial, after 24 weeks of protein supplementation plus resistance exercise, a noticeable but statistically insignificant improvement was observed. The researchers predict that “a larger sample size or a longer exposure to a protein and exercise intervention would increase the magnitude of effect”, but this, of course, requires proof.

In conclusion, we found an association between higher total and animal protein intake with higher total body and spine BMD. In contrast, higher plant protein intake was associated with a lower total body and spine BMD. Research is warranted to investigate further the added value of dietary protein alongside calcium and vitamin D for BMD improvement, especially in osteopenic or osteoporotic individuals. Furthermore, more research on the impact of a plant-based diet on bone health is needed.

Conclusion

While the design of this study has many limitations, it expands our limited knowledge of the relationship between protein intake and bone density in older adults. The current notion that older people should increase their protein intake (plus, probably, physical activity) seems to hold, but questions arise about the contribution of plant protein, which should be resolved by future, more rigorous, studies.

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] Groenendijk, I., Grootswagers, P., Santoro, A., Franceschi, C., Bazzocchi, A., Meunier, N., … & De Groot, L. (2022, September). PROTEIN INTAKE AND BONE MINERAL DENSITY: CROSS-SECTIONAL RELATIONSHIP AND LONGITUDINAL EFFECTS OF PROTEIN SUPPLEMENTATION IN A COMBINED COHORT OF FRAIL, MALNOURISHED AND HEALTHY OLDER ADULTS. In AGING CLINICAL AND EXPERIMENTAL RESEARCH (Vol. 34, No. SUPPL 1, pp. S270-S271).

[2] Edozien, J. C., Khan, M. R., & Waslien, C. I. (1976). Human protein deficiency: results of a Nigerian village study. The Journal of Nutrition, 106(3), 312-328.

[3] Levine, M. E., Suarez, J. A., Brandhorst, S., Balasubramanian, P., Cheng, C. W., Madia, F., … & Longo, V. D. (2014). Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell metabolism, 19(3), 407-417.

[4] Mirzaei, H., Raynes, R., & Longo, V. D. (2016). The Conserved Role for Protein Restriction During Aging and Disease. Current opinion in clinical nutrition and metabolic care, 19(1), 74.

[5] Ferraz-Bannitz, R., Beraldo, R. A., Peluso, A. A., Dall, M., Babaei, P., Foglietti, R. C., … & Foss-Freitas, M. C. (2022). Dietary protein restriction improves metabolic dysfunction in patients with metabolic syndrome in a randomized, controlled trial. Nutrients, 14(13), 2670.

Blood cancer

Harnessing Senescent Cancer Cells to Fight Cancer

New research published in Cancer Discovery has outlined a potential new method of using senescent cells to encourage the immune system to attack cancer [1].

Senescent cancer cells and immune clearance

Cellular senescence is a stress response [2], and cancer cells within tumors undergo significant stress: they lose access to oxygen and nutrients, they divide faster than they can sustain themselves, they accumulate reactive oxygen species, and they endure strong intercellular signals [2,3,4]. Many of these cells lose their ability to replicate, becoming senescent cancer cells.

The senescence-associated secretory phenotype (SASP) promotes inflammation, and previous work has shown that it encourages immune cells to clear out the senescent cells producing it [5]. Unfortunately, senescent cells can other compounds that inhibit the immune system, such as HLA-E, which discourages natural killer (NK) and CD8+ T cells from destroying them [6]. Most research in this area has been done with a focus on the innate immune system, particularly macrophages and NK cells; these cells, especially in the context of cancer, are considered the most important in destroying senescent cells [7].

The adaptive immune system’s role in senescence response is often considered less notable and has been less studied. However, one previous study showed that senescent liver cells in particular express MHC-II, a class of molecules that activate CD4+ T cells in this context [8]. This research pushes further in this direction, examining the effects of the SASP on CD8+ T cells.

Immune signals stimulate CD8+ T cells

Senescent cells are heterogenous, and the two principal ways in which they differ are their cell types and what has driven them to senescence. The researchers tested seven different combinations of type and driver, four taken from human cell lines and three from mice. Two of the cell types were cancerous. The marker SA-ß-gal and mRNA expressions of senescence-associated genes were used to confirm that the tested cells had actually become senescent.

All of the tested cells were found to have expressed increased levels of Type 1 interferons (IFNs), which are part of the SASP and stimulate production of the MHC-I immune signaling complex [9]. This study found that senescent cells of all the tested types, including cancer, increased MHC-I signals as a whole and that inhibiting IFN reduced these signals.

The next experiment was to determine if senescent cells would directly trigger a CD8+ T cell response. For this experiment, the researchers injected mice with either senescent fibroblasts or one of two controls: a negative control containing an empty vehicle and a positive control containing a known antigen.

The results were substantial. Living mice exposed to senescent cells formed a strong CD8+ T cell response against those cells, even when they were taken out of the mice. MHC-II was not expressed nearly as strongly, explaining the weaker response of CD4+ T cells.

The researchers found 74 peptides related to MHC-I that were only expressed by senescent cells. Ten out of those 74 were found to be substantially expressed by coding mRNA. Three of the prioritized ten, and three combinations of the remaining 64, were found to stimulate the murine cells that were trained against senescent fibroblasts.

A closer look at cancer

While highly informative, those experiments were not focused on cancer cells. To test cancer, the researchers injected mice with vehicle, untreated cancer cells, senescent cancer cells, and cancer cells undergoing immunogenic cell death (ICD). They also measured the damage-associated molecular patterns (DAMPs) of ICD cancer cells and senescent cancer cells, finding that they were similar.

However, the senescent cancer cells lasted much longer in mice. While the ICD cells died off and the cancer cells proliferated as cancer does, the senescent cells did little but provoke a substantial immune response among dendritic cells, which are part of the adaptive immune system and produce antigens. This response was even greater than against proliferating cancer.

Injecting the senescent cell-treated mice with cancer cells demonstrated that they were far better protected than ICD-treated mice or mice treated with dying senescent cancer cells. In the case of pancreatic cancer, none of the treated mice developed tumors at all. The immune cells of treated mice were also much better at infiltrating melanoma tumors that are usually resistant to immune attacks.

Co-culturing dendritic cells alongside senescent cancer cells showed a similar result: these cells were more effective at activating CD8+ T cells than their non-senescent counterparts.

Finally, and perhaps most importantly, these results were validated in a human cell experiment. Tumor-infiltrating lymphocytes were exposed to senescent and non-senescent human cancer lines, and the lymphocytes exposed to senescent cancer cells activated CD8+ T cells much more strongly.

Conclusion

With a wealth of strong preclinical data in hand, the researchers conclude that using senescent cancer cells to encourage human immune systems to destroy cancer is a potentially viable approach. As senescent cells, and cancer cells, differ wildly, a substantial number of human clinical trials will need to be conducted to determine if therapies built around this concept can improve survival rates and potentially defeat some types of cancer entirely.

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] Marin, I., Boix, O., Garcia-Garijo, A., Sirois, I., Caballe, A., Zarzuela, E., … & Serrano, M. (2022). Cellular senescence is immunogenic and promotes anti-tumor immunity. Cancer Discovery, CD-22.

[2] Calcinotto, A., Kohli, J., Zagato, E., Pellegrini, L., Demaria, M., & Alimonti, A. (2019). Cellular senescence: aging, cancer, and injury. Physiological reviews, 99(2), 1047-1078.

[3] Prata, L. G. L., Ovsyannikova, I. G., Tchkonia, T., & Kirkland, J. L. (2018, December). Senescent cell clearance by the immune system: Emerging therapeutic opportunities. In Seminars in immunology (Vol. 40, p. 101275). Academic Press.

[4] Muñoz-Espín, D., & Serrano, M. (2014). Cellular senescence: from physiology to pathology. Nature reviews Molecular cell biology, 15(7), 482-496.

[5] Wang, L., Lankhorst, L., & Bernards, R. (2022). Exploiting senescence for the treatment of cancer. Nature Reviews Cancer, 22(6), 340-355.

[6] Pereira, B. I., Devine, O. P., Vukmanovic-Stejic, M., Chambers, E. S., Subramanian, P., Patel, N., … & Akbar, A. N. (2019). Senescent cells evade immune clearance via HLA-E-mediated NK and CD8+ T cell inhibition. Nature communications, 10(1), 1-13.

[7] Xue, W., Zender, L., Miething, C., Dickins, R. A., Hernando, E., Krizhanovsky, V., … & Lowe, S. W. (2007). Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature, 445(7128), 656-660.

[8] Kang, TW., Yevsa, T., Woller, N., … & Lars Z. (2011). Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature, 479, 547–551.

[9] Raval, A., Puri, N., Rath, P. C., & Saxena, R. K. (1998). Cytokine regulation of expression of class I MHC antigens. Experimental & molecular medicine, 30(1), 1-13.

Walking Seniors

Astaxanthin Improves Endurance in Older Adults

In a new study published in International Journal of Environmental Research and Public Health, Japanese researchers have shown that astaxanthin supplementation improves endurance in healthy older adults [1].

Muscles succumb to oxidative stress

Aging leads to reduced muscle strength and endurance. Currently, exercising is the only method to slow down these processes, but it is often not feasible in older adults due to pain and other health issues.

On the molecular level, aging is accompanied by oxidative stress, which impairs energy metabolism and is connected with a decline in muscle strength. Therefore, taking antioxidant supplements in old age might be beneficial to offset muscle weakening and endurance decline, especially for people who can not perform continuous exercise.

In this study, the researchers focused on astaxanthin, as this antioxidant was previously shown to be effective at preventing age-related oxidative stress and mitochondrial dysfunction in muscles [2].

Astaxanthin is a member of the carotenoids, pigments that give the yellow, orange, and red colors to various foods. It is found in some algae and seafood, such as salmon, trout, and shrimp. Astaxanthin was previously shown to prevent muscle atrophy and increase endurance when combined with exercise [3].

Astaxanthin fights back

In order to explore if astaxanthin supplementation alone is effective at improving endurance in older adults, the researchers randomly assigned 24 healthy adults between the ages of 67 and 94 into an experimental group receiving 24 mg of astaxanthin and a control group receiving placebo for 16 weeks. The participants were living in nursing homes, which ensured that there was no variability in their diets. They were asked not to do any intense and unusual exercises throughout the trial.

First, the researchers conducted a reactive oxygen metabolites test and showed that astaxanthin supplementation reduced the participants’ oxidative stress. Next, the 6-minute walking test demonstrated that the participants from the astaxanthin group increased their walking distance. At the same time, astaxanthin supplementation did not improve muscle strength as shown by hand grip and knee extension tests.

In addition, the researchers measured the change in the participants’ lactic acid levels after the 6-minute walking test pre- and post-supplementation. Astaxanthin supplementation was shown to decrease lactic acid production in these participants.

Therefore, astaxanthin supplementation improved endurance by reducing oxidative stress, and lactic acid production was most likely decreased by inhibiting reactive oxygen species production and enhancing the activity of antioxidant enzymes.

Abstract

Oxidative stress is associated with deterioration of endurance and muscle strength, which are mostly accompanied by aging. Astaxanthin supplement has excellent antioxidant activity without any pro-oxidative properties. In this study, we investigated how astaxanthin supplementation affects walking endurance and muscle strength in nursing home residents. Healthy elderly individuals (age: 67 to 94) were divided into two groups: 13 subjects received a daily dose of 24 mg of astaxanthin for 16 weeks (astaxanthin group) and 11 subjects received a placebo (placebo group). These subjects were compared using body component measurements, serum d-ROM levels, the distance of 6-min walking, blood lactate levels after the 6-min walking test, and muscle strength. After supplementation, the levels of d-ROMs and blood lactate after the 6-min walking test in the astaxanthin group significantly decreased compared with the placebo group (p < 0.05). Additionally, the walking distance was significantly higher in the astaxanthin group than in the placebo group (p < 0.05), despite a significant reduction in lactate levels after 6-MWT (p < 0.05). However, no significant intergroup differences were observed in muscle mass and strength. Astaxanthin supplement for 16 weeks is effective to increase the endurance capacity of the elderly. Astaxanthin supplement suppresses d-ROMs at rest and lactic acid production after the 6-min walk test. In contrast, astaxanthin supplement did not show significant intergroup differences in the muscle mass and strength. Therefore, the effect was most likely accompanied by an increase in endurance instead of an increase in muscle strength.

Conclusion

This promising pilot study suggests a beneficial effect of taking astaxanthin supplements in older age, especially for people who do not engage in regular physical activity. The small size of the cohort along with the lack of individual response data limits the interpretability of these results. Nevertheless, it’s worth following up with future studies on astaxanthin supplementation in humans. Meanwhile, following a diet rich in vegetables and fruit along with low-intensity exercises is a proven way to keep oxidative stress down.

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] Nakanishi, R., Kanazashi, M., Tanaka, M., Tanaka, M., & Fujino, H. (2022). Impacts of Astaxanthin Supplementation on Walking Capacity by Reducing Oxidative Stress in Nursing Home Residents. International Journal of Environmental Research and Public Health, 19(20), 13492.

[2] Mocchegiani, E., Costarelli, L., Giacconi, R., Malavolta, M., Basso, A., Piacenza, F., … & Monti, D. (2014). Vitamin E–gene interactions in aging and inflammatory age-related diseases: Implications for treatment. A systematic review. Ageing research reviews14, 81-101.

[3] Liu, S. Z., Valencia, A. P., VanDoren, M. P., Shankland, E. G., Roshanravan, B., Conley, K. E., & Marcinek, D. J. (2021). Astaxanthin supplementation enhances metabolic adaptation with aerobic training in the elderly. Physiological reports, 9(11), e14887.