Fight Aging! Newsletter, May 16th 2022

Fight Aging! publishes news and commentary relevant to the goal of ending all age-related disease, to be achieved by bringing the mechanisms of aging under the control of modern medicine. This weekly newsletter is sent to thousands of interested subscribers. To subscribe or unsubscribe from the newsletter, please visit: https://www.fightaging.org/newsletter/

Longevity Industry Consulting Services

Reason, the founder of Fight Aging! and Repair Biotechnologies, offers strategic consulting services to investors, entrepreneurs, and others interested in the longevity industry and its complexities. To find out more: https://www.fightaging.org/services/

Contents

  • A Reduction in Epigenetic Age is not at this Time Sufficient Proof of Slowed Aging
  • The Aging Gut Microbiome Negatively Influences the Brain via Inflammation
  • Few Evident Relationships Between Accelerated Epigenetic Aging and Cancer
  • Assessing Rejuvenation in Mice Produced by Fecal Microbiota Transplantation
  • Does Much of the Life Extension of Calorie Restriction Depend on Length of Fasting?
  • γδ T cells are Involved in the Generation of Chronic Inflammation by Excess Visceral Fat
  • The Adult Brain Contains Dormant Precursor Neurons, Slowly Used Up Over Life
  • SIRT6 in Aging, Immunity, and Cancer
  • Using Chaperones to Reduce Endoplasmic Reticulum Stress Improves Sleep and Cognition in Aged Mice
  • Reviewing What is Known of Mesenchymal Stem Cell Aging
  • Decreased Ribosomal Biogenesis in Some Long-Lived Individuals
  • An Interview with Researcher Thomas Kirkwood
  • Improving the Understanding of How Hypertension Results in Vascular Restructuring
  • Reviewing Mitochondria-Derived Peptides in Aging
  • The Concept of Cellular Exercise

A Reduction in Epigenetic Age is not at this Time Sufficient Proof of Slowed Aging
https://www.fightaging.org/archives/2022/05/a-reduction-in-epigenetic-age-is-not-at-this-time-sufficient-proof-of-slowed-aging/

Geroprotective therapies are those that slow aging. While true rejuvenation therapies capable of reversing aging may also fall under that broad umbrella, discussion of geroprotectors usually focuses on drugs such as mTOR inhibitors that can at least modestly slow aging in animal studies. One area of growing interest in the field is the use of epigenetic clocks to assess aging, and the degree to which the clock measurements are affected by potentially geroprotective interventions.

The challenge in epigenetic clocks - and other conceptually similar clocks - is that they are fitted to observed age-related changes in biochemical data without any understanding of what causes those changes. Perhaps characteristic epigenetic changes that take place with age reflect all of the underlying processes of aging, and perhaps they do not. Thus one cannot take any given result in the treatment of aging at face value until the specific clock has been calibrated to the specific intervention in life span studies, a lengthy prospect that entirely defeats the point of a simple measure of aging, and which has yet to be undertaken for any class of intervention.

As researchers point out here, this means that clocks, while interesting and meriting further study, must be relegated to the second tier of data for the foreseeable future. Whether or not a given intervention produces slowed aging, and is thus geroprotective, can only be assessed robustly at the present time via established measures of health and age-related disease.

Does Modulation of an Epigenetic Clock Define a Geroprotector?

The geroscience hypothesis, that the rate of aging can be changed, is indeed an exciting one, and one that will likely receive considerable attention in the future. Geroprotectors arising from studies exploring the geroscience hypothesis would undoubtedly revolutionize health care and result in dramatic societal changes, and for these reasons should be taken extremely seriously. However, the biomedical science community should be very sensitive to overenthusiasm concerning ways in which geroprotectors are vetted, since reliance on a solitary measure of aging, for example an epigenetic clock, to vet candidate geroprotectors might not be necessary. If geroprotectors, by definition, should improve health during the aging process, and health can be measured in myriad ways, then relevant trials should focus on these health measures directly.

In fact, as we have argued, it would be hard to make the case that a geroprotector that is only known or shown to modulate an epigenetic clock will extend health span or lifespan without impacting anything associated with health from traditional clinical perspectives. In addition, if one could show that a geroprotector actually does modulate age-related disease processes using routine and accepted clinical measures then the mechanism of action of that geroprotector is likely to be a key to an underlying universal aging clock. Ultimately, a purported geroprotector that has either no observable effect on many available common sense, well-accepted measures of health and vitality, or will only have an effect on health via some cryptic mechanism after the many years of use during which an individual is at typical risk for disease, is a tough sell.

Simply put, geroprotectors should provide overt health and disease prevention benefits but the time-dependent relationships between epigenetic clocks and health-related phenomena are complex and in need of further scrutiny. Therefore, studies that enable understanding of the relationships between epigenetic clocks and disease processes while simultaneously testing the efficacy of a candidate geroprotector are crucial to move the field forward.

The Aging Gut Microbiome Negatively Influences the Brain via Inflammation
https://www.fightaging.org/archives/2022/05/the-aging-gut-microbiome-negatively-influences-the-brain-via-inflammation/

A good deal of research of late has focused on the role of the gut microbiome in aging. One portion of this part of the field covers the interactions between the gut microbiome and the brain. For example, butyrate is a metabolite generated by the gut microbiome, in declining amounts with age as the balance of microbial populations shifts. Butyrate upregulates BDNF, which in turn upregulates neurogenesis in the brain, the production of new neurons. The consensus on neurogenesis is that more of it is a good thing, and many research programs are working towards safe ways to achieve this goal.

One of the more notable age-related changes in the gut microbiome is the growth of inflammatory populations, those that provoke the immune system and cause a meaningful fraction of the chronic inflammation that is characteristic of older individuals. This inflammation contributes to the onset and progression of all of the common age-related conditions. Since the immune system is responsible for gardening the microbiome and removing these inflammatory microbes, this is a bidrectional relationship. More inflammatory microbes degrade the effectiveness of the immune system, but the dysfunctions of immune aging allow these harmful populations to run amok.

What can be done about this? In animal studies, fecal microbiota transplantation from young individuals to old individuals has been shown to reset the gut microbiome, improve health, and extend life. There are other strategies with varying degrees of evidence to support their efficacy, but this one seems the most practical, given that fecal microbiota transplantation is already used in medical practice, and thus it would be a comparatively small step to adapt it to this new use case.

Gut Microbiota Interact With the Brain Through Systemic Chronic Inflammation: Implications on Neuroinflammation, Neurodegeneration, and Aging

It has been noticed in recent years that the unfavorable effects of the gut microbiota could exhaust host vigor and life, yet knowledge and theory are just beginning to be established. Increasing documentation suggests that the microbiota-gut-brain axis not only impacts brain cognition and psychiatric symptoms but also precipitates neurodegenerative diseases, such as Alzheimer's disease (AD).

How the blood-brain barrier (BBB), a machinery protecting the central nervous system (CNS) from the systemic circulation, allows the risky factors derived from the gut to be translocated into the brain seems paradoxical. For the unique anatomical, histological, and immunological properties underpinning its permeable dynamics, the BBB has been regarded as a biomarker associated with neural pathogenesis. The BBB permeability of mice and rats caused by GM dysbiosis raises the question of how the GM and its metabolites change BBB permeability and causes neuroinflammation and neurodegeneration (NF&ND) and brain aging, a pivotal multidisciplinary field tightly associated with immune and chronic systemic inflammation.

Gut microbiota-induced systemic chronic inflammation mainly refers to excessive gut inflammation caused by gut mucosal immunity dysregulation, which is often influenced by dietary components and age, is produced at the interface of the intestinal barrier (IB) or exacerbated after IB disruption, initiates various common chronic diseases along its dispersal routes, and eventually impairs BBB integrity to cause NF&ND and brain aging.

Few Evident Relationships Between Accelerated Epigenetic Aging and Cancer
https://www.fightaging.org/archives/2022/05/few-evident-relationships-between-accelerated-epigenetic-aging-and-cancer/

Epigenetic clocks are produced by identifying characteristic shifts in epigenetic marks with age, the decorations on the genome that control gene expression. It remains unclear as to the exact relationship between specific epigenetic marks and the underlying damage and dysfunction of aging, and so it remains unknown as to how comprehensively epigenetic clocks reflect the processes of aging: do all of the processes of aging contribute, or only some of them? If the latter, it will be hard to use epigenetic clocks to assess the quality of potential rejuvenation therapies. Removing that uncertainty will require a great deal of further work.

When epigenetic age is higher than chronological age, this is referred to as accelerated epigenetic age. It is thought to reflect a greater burden of the underlying cell and tissue damage that causes aging, but of course the uncertainty remains as to whether this is a full versus selective representation of the state of health for any given epigenetic clock - any given combination of epigenetic marks, in other words. Are there aspects of aging that contribute little to epigenetic age?

With that in mind, researchers here note that a first pass at analysis of cancer incidence and accelerated epigenetic age found little in the way of firm correlations. This is interesting, as (a) cancer risk is very robustly age-associated, (b) the risk of a number of other age-related conditions does correlate to accelerated epigenetic age, and (c) recent work suggests that incidence of serious mutational damage causes epigenetic change, so one might expect a greater pace of mutational damage to lead to both more cancer and more epigenetic aging.

Cancer: The aging epigenome

Age is a prominent risk factor for most types of cancer. Cancer risk increases with age, in part, because genetic mutations that arise from DNA replication errors and exposure to environmental carcinogens accumulate as we get older. Aging also alters the epigenome, the chemical marks spread across DNA that help switch genes on or off by altering how the genome is packaged. For instance, the addition of a methyl group to DNA can play a role in compressing the nearby DNA sequence so it can no longer be accessed by the cell's machinery. Epigenetic modifications, including DNA methylation, have also been shown to contribute to the development of cancer. However, the potential impact of age-related epigenetic changes on cancer development has not been fully characterized.

It has been hypothesized that people whose epigenetic age is greater than their age in years - a phenomenon known as accelerated aging - may be at higher risk of age-related diseases, including cancer. However, previous studies linking accelerated epigenetic aging and cancer have produced mixed results. Now a team has taken a different approach. Instead of associating a person's risk of cancer with epigenetic clock estimates, they correlated it against genetic variations that are known to influence these algorithms.

The results did not show many clear relationships between the epigenetic aging clocks and risk for the various types of cancer studied. The most promising finding was an association between the GrimAge clock and colorectal cancer. The GrimAge clock was not designed to predict age alone, but also reflects the effects of smoking and other mortality-related epigenetic features. Thus, the interpretation of this association is not straightforward, as this clock may capture the effects of environmental or lifestyle factors on the epigenome. One caution is that epigenetic clocks have largely been developed based on how aging affects DNA methylation in blood cells. Much less is known regarding aging and epigenetics in other tissue types, including those prone to cancer.

Assessing Rejuvenation in Mice Produced by Fecal Microbiota Transplantation
https://www.fightaging.org/archives/2022/05/assessing-rejuvenation-in-mice-produced-by-fecal-microbiota-transplantation/

Today's open access paper reports on an effort to measure the effects of microbial rejuvenation on tissue function in mice. Fecal microbiota transplantation can reverse the aging of the gut microbiome, at least when carried out in animal studies, and as measured by the detrimental shift in microbial populations that takes place with age. Transplanting microbes from a young gut into an old gut reverses many of the alterations in relative abundance of specific microbial species, and has been shown to improve health and extend life span in some species.

In an old mouse, or human, there are fewer microbes producing beneficial metabolites, and more inflammatory microbes that provoke the immune system. This contributes to declining tissue function and increased chronic inflammation. Interestingly, this shift may be largely due to the aging of the immune system, and a progressive failure to sufficiently garden the gut microbiome, but there may be other contributing causes as well. How large an effect on function is produced by the aging of the gut microbiome versus other issues in aging? The fastest way to answer that question is to restore a youthful gut microbiome and assess the results.

Fecal microbiota transfer between young and aged mice reverses hallmarks of the aging gut, eye, and brain

Altered intestinal microbiota composition in later life is associated with inflammaging, declining tissue function, and increased susceptibility to age-associated chronic diseases, including neurodegenerative dementias. Here, we tested the hypothesis that manipulating the intestinal microbiota influences the development of major comorbidities associated with aging and, in particular, inflammation affecting the brain and retina.

Using fecal microbiota transplantation (FMT), we exchanged the intestinal microbiota of young (3 months), old (18 months), and aged (24 months) mice. Whole metagenomic shotgun sequencing and metabolomics were used to develop a custom analysis workflow, to analyze the changes in gut microbiota composition and metabolic potential. Effects of age and microbiota transfer on the gut barrier, retina, and brain were assessed using protein assays, immunohistology, and behavioral testing.

We show that microbiota composition profiles and key species enriched in young or aged mice are successfully transferred by FMT between young and aged mice and that FMT modulates resulting metabolic pathway profiles. The transfer of aged donor microbiota into young mice accelerates age-associated central nervous system (CNS) inflammation, retinal inflammation, and cytokine signaling and promotes loss of key functional protein in the eye, effects which are coincident with increased intestinal barrier permeability. Conversely, these detrimental effects can be reversed by the transfer of young donor microbiota.

These findings demonstrate that the aging gut microbiota drives detrimental changes in the gut-brain and gut-retina axes suggesting that microbial modulation may be of therapeutic benefit in preventing inflammation-related tissue decline in later life.

Does Much of the Life Extension of Calorie Restriction Depend on Length of Fasting?
https://www.fightaging.org/archives/2022/05/does-much-of-the-life-extension-of-calorie-restriction-depend-on-length-of-fasting/

How much of the improvement in health and extension of life span produced by calorie restriction in mouse studies is due to lowered calorie intake versus length of time spent fasting between meals? In most past studies, calorie restricted mice were fed once a day, resulting in long periods of fasting and hunger-induced metabolic changes between meals. It may be that this timing is important, and of late researchers have started to run studies intended to assess this question.

Data obtained to date strongly suggests that, yes, time of fasting does matter and does contribute to health and longevity benefits in mice independently of reduced calorie intake. Today's research materials further support that conclusion. The authors report on a study in which calorie restricted mice are fed at different intervals. Allowing these mice to eat throughout the day reduces the gain in life span usually observed in calorie restriction studies.

Active phase calorie restriction enhances longevity

Timing feedings to match the active period of the circadian cycle extended the life span of lab mice more than three times as much as caloric restriction alone. Mice that ate as much and whenever they wanted lived nearly 800 days median life span - an average period for their species. Restricting calories but making food available around the clock extended their lives only 10% to 875 days despite restricting calories by 30-40%. Restricting this reduced-calorie diet to the inactive period of the circadian cycle boosted lifespan by nearly 20% to an average of 959 days. Offering the low-calorie diet only during the active period of the cycle extended their median life span to about 1,068 days, an increase of almost 35% over the unrestricted eaters.

Further investigation showed that the mice that lived the longest had significantly better metabolic health, with higher insulin sensitivity and blood sugar stability. They tended to get diseases that killed the younger mice, such as various forms of cancer, at far more advanced ages. Gene expression experiments showed fewer changes in the activity of genes associated with inflammation, metabolism and aging in the long-lived animals compared to the shorter-lived ones.

Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice

Caloric restriction (CR) prolongs lifespan, yet the mechanisms by which it does so remain poorly understood. Under CR, mice self-impose chronic cycles of 2-hour-feeding and 22-hour-fasting, raising the question whether calories, fasting, or time of day are causal. We show that 30%-CR is sufficient to extend lifespan 10%; however, a daily fasting interval and circadian-alignment of feeding act together to extend lifespan 35% in male C57BL/6J mice. These effects are independent of body weight. Aging induces widespread increases in gene expression associated with inflammation and decreases in expression of genes encoding components of metabolic pathways in liver from ad lib fed mice. CR at night ameliorates these aging-related changes. Thus, circadian interventions promote longevity and provide a perspective to further explore mechanisms of aging.

γδ T cells are Involved in the Generation of Chronic Inflammation by Excess Visceral Fat
https://www.fightaging.org/archives/2022/05/%ce%b3%ce%b4-t-cells-are-involved-in-the-generation-of-chronic-inflammation-by-excess-visceral-fat/

Visceral fat around the inner organs is a metabolically active tissue, and more of it is entirely detrimental to long-term health. These fat deposits interact with the immune system in a number of distinct ways to produce chronic inflammation, and that inflammation in turn drives the onset and progression of tissue dysfunction and all of the common age-related conditions. For example, visceral fat encourages the accumulation of senescent cells and their pro-inflammatory signaling, while visceral fat cells signal in ways that mimic infection, as well as producing DNA debris that activates the innate immune system. Given the number of known inflammatory processes, we might expect researchers to uncover further, novel mechanisms involved in generating inflammation in visceral fat, as is the case here.

γδ T cells are a unique and poorly understood class of lymphocytes generally regarded for their role in barrier protection with functionally distinct subpopulations residing in epithelial tissues, including those of the skin, gut, and lung. In addition to responding to antigen presentation via the T cell receptor, similar to conventional T cells (regulatory, helper, and cytotoxic T cells) of the adaptive immune system, γδ T cells can respond directly to cytokines and other intact proteins without antigen processing and presentation, and have the capacity to phagocytize much like innate immune cells.

In adipose tissues, contrary to their traditional function in infection control, γδ T cells appear to play major roles in maintaining homeostasis with respect to inflammation and insulin sensitivity. Adipose tissue γδ T cells have been shown to increase in number in mouse models of diet-induced obesity, where they promote macrophage accumulation, inflammation, and insulin resistance. More recently, they were reported to regulate adipose tissue regulatory T cell homeostasis and thermogenesis in adolescent and young-adult mice.

Here, we identified and characterized a population of γδ T cells, which show unique age-dependent accumulation in the visceral adipose tissue of both mice and humans. Diet-induced obesity likewise increased γδ T cell numbers; however, the effect was greater in the aged where the increase was independent of fat mass. Genetic deficiency of γδ T cells in old age improved the metabolic phenotype, characterized by increased respiratory exchange ratio, and lowered levels of IL-6 both systemically and locally in visceral adipose tissue. Decreased IL-6 was predominantly due to reduced production by non-immune stromal cells, primarily preadipocytes, and adipose-derived stem cells. Collectively, these findings suggest that an age-dependent increase of tissue-resident γδ T cells in visceral adipose tissue contributes to local and systemic chronic inflammation and metabolic dysfunction in aging.

The Adult Brain Contains Dormant Precursor Neurons, Slowly Used Up Over Life
https://www.fightaging.org/archives/2022/05/the-adult-brain-contains-dormant-precursor-neurons-slowly-used-up-over-life/

When it comes to better maintaining the aging brain, understanding the creation of new neurons in adult life is an important topic. If there are processes by which new neurons arise and are integrated into the brain, then they are targets for therapies intended to increase that output. It is generally agreed upon that a greater supply of new neurons is a good thing, and may even enhance function at all ages, not just in later life. In that context, the discussion here, regarding a precursor cell population that is slowly used up over a lifetime in order to generate new neurons, is most interesting.

Dormant non-proliferative neuronal precursors (dormant precursors) are a unique type of undifferentiated neuron, found in the adult brain of several mammalian species, including humans. Dormant precursors are fundamentally different from canonical neurogenic-niche progenitors as they are generated exquisitely during the embryonic development and maintain a state of protracted postmitotic immaturity lasting up to several decades after birth. Thus, dormant precursors are not pluripotent progenitors, but to all effects extremely immature neurons. Recently, transgenic models allowed to reveal that with age virtually all dormant precursors progressively awaken, abandon the immature state, and become fully functional neurons.

Compelling evidence implies that dormant precursors in the adult brain are physiologically relevant and may contribute to an overlooked form of late brain maturation. Intriguingly, our brain seems to use this resource sparingly throughout the whole course of life. To fully understand the contribution of dormant precursors integration, it will be crucial to identify the molecular mechanisms promoting or hindering maturation and the behavioral impacts.

Considering the possibility of a precursor-based contribution to learning and adaptation of input processing in the young individuals may shed a new light on development. The handful of dormant precursors still available up to an advanced age may also constitute a precious resource and understanding the mechanisms that promote their late integration could allow to recover every last bit of untapped potential, perhaps improving cognition and/or adaptation in the aging brain. Importantly, the number of dormant precursors is inherently limited by their non-proliferative nature and purposely promoting their integration in early life will lead to their premature exhaustion. Therefore, a comprehensive understanding of the relevance of dormant precursors in processes of brain maturation and adaptation along the different life phases constitutes a pressing need.

SIRT6 in Aging, Immunity, and Cancer
https://www.fightaging.org/archives/2022/05/sirt6-in-aging-immunity-and-cancer/

A lot of work has gone into better understanding the roles of sirtuin 1 (SIRT1) in aging, ultimately something of a dead end, not a large enough influence on relevant areas of cellular biochemistry to produce viable treatments to slow aging. Sirtuin 6 (SIRT6), on the other hand is less well explored, but somewhat more interesting, even though it is likely still only a path towards therapies that can do not more than modestly slow aging over time. Overexpression of SIRT6 extends life in mice. One of the possible mechanisms for that extension of life is promotion of DNA repair, and a startup biotech company is working on a SIRT6 gene therapy aimed at improving DNA repair in inherited DNA repair deficiency conditions. Nonetheless, what is presently known about SIRT6 is much less than we'd like to know, as noted in this review paper.

SIRT6 has a range of post-translational modification (PTM) capabilities and is widely involved in aging, immunity, and cancer regulation. SIRT6 is a longevity protein that prevents cells, tissues, organs, and the body from aging. Although the mechanisms underlying these effects are diverse, they all involve resistance of aging by promoting of DNA damage repair, maintaining of the normal telomere structure of chromosomes, regulating of glucose and NAD+ metabolic balance, and by regulating of the senescence-associated secretory phenotype (SASP). SIRT6 can also affect the differentiation and function of immune cells by regulating PTM affecting cells or the immunometabolism. However, the role of SIRT6 in immune regulation is complex.

Although most studies have shown SIRT6 to have anti-inflammatory activity, there is no lack of evidence regarding its pro-inflammatory potential. There has been insufficient research on how SIRT6 affects inflammation by regulating immune cells; SIRT6 has rarely been studied in many immune cells including granulocytes, monocytes, B cells, natural killer (NK) cells, and NKT cells. However, according to the recent research, the SIRT6-PTM or immunometabolism axes represent new directions with research potential.

The role of SIRT6 in cancer development is complex. SIRT6 shows differential expression in cancer tissues compared with normal tissues; its expression levels may also vary among different cancers, at different stages of the same cancer, and in different cell lines of the same tumor type. It also has both positive and negative effects on the regulation of cancer. Few studies have analyzed whether SIRT6 could achieve anti-cancer effects via regulation of immune cell function. This could represent a new direction for future research. For example, it may be possible to adjust the polarization of macrophages through SIRT6 to affect tumor progression.

Taken together, these findings indicate that SIRT6 will serve as an important target candidate for regulating immunosenescence and immune cell function. Drugs designed to target SIRT6 will also make an important contribution to the fight against chronic inflammation and cancer. SIRT6, as an important regulator throughout immunosenescence, inflammaging, and cancer, is a potential target for the regulation of the immune system.

Using Chaperones to Reduce Endoplasmic Reticulum Stress Improves Sleep and Cognition in Aged Mice
https://www.fightaging.org/archives/2022/05/using-chaperones-to-reduce-endoplasmic-reticulum-stress-improves-sleep-and-cognition-in-aged-mice/

4-phenyl butyrate can be delivered orally, and once inside cells it mimics some of the natural chaperone molecules that aid in protein folding in the endoplasmic reticulum. Improved the quality and pace of protein folding leads to better cell function, particularly given that rising levels of endoplasmic reticulum stress and impairment in the compensatory unfolded protein response are observed in aged tissues. Addressing this issue can improve the state of tissue function in aged animals, at least to some degree, as demonstrated in the research results noted here.

As the aging population grows, the need to understand age-related changes in health is vital. Two prominent behavioral changes that occur with age are disrupted sleep and impaired cognition. Sleep disruptions lead to perturbations in proteostasis and endoplasmic reticulum (ER) stress in mice. Further, consolidated sleep and protein synthesis are necessary for memory formation. With age, the molecular mechanisms that relieve cellular stress and ensure proper protein folding become less efficient.

It is unclear if a causal relationship links proteostasis, sleep quality, and cognition in aging. Here, we used a mouse model of aging to determine if supplementing chaperone levels reduces ER stress and improves sleep quality and memory. We administered the chemical chaperone 4-phenyl butyrate (PBA) to aged and young mice, and monitored sleep and cognitive behavior. We found that chaperone treatment consolidates sleep and wake, and improves learning in aged mice. These data correlate with reduced ER stress in the cortex and hippocampus of aged mice.

Chaperone treatment increased phosphorylated CREB (p-CREB), which is involved in memory formation and synaptic plasticity, in hippocampi of chaperone-treated aged mice. Further, hippocampal overexpression of the endogenous chaperone, binding immunoglobulin protein (BiP), improved cognition, reduced ER stress, and increased p-CREB in aged mice, suggesting that supplementing BiP levels are sufficient to restore some cognitive function. Together, these results indicate that restoring proteostasis improves sleep and cognition in a wild-type mouse model of aging.

Reviewing What is Known of Mesenchymal Stem Cell Aging
https://www.fightaging.org/archives/2022/05/reviewing-what-is-known-of-mesenchymal-stem-cell-aging/

Age-related decline in stem cell function is an important contributing cause of aging, and cellular senescence in stem cell populations and their supporting cells is a feature of this process. Mesenchymal stem cells are a well studied population that is not only relevant to tissue function but also widely used as a basis for stem cell treatments. These therapies also face challenges due to declining cell function and the onset of cellular senescence, occurring when stem cells are cultured and expanded for use in therapy.

Aging is a multifaceted and complicated process, manifested by a decline of normal physiological functions across tissues and organs, leading to overt frailty, mortality, and chronic diseases, such as skeletal, cardiovascular, and cognitive disorders, necessitating the development of practical therapeutic approaches.

Stem cell aging is one of the leading theories of organismal aging. For decades, mesenchymal stem/stromal cells (MSCs) have been regarded as a viable and ideal source for stem cell-based therapy in anti-aging treatment due to their outstanding clinical characteristics, including easy accessibility, simplicity of isolation, self-renewal, and proliferation ability, multilineage differentiation potentials, and immunomodulatory effects. Nonetheless, as evidenced in numerous studies, MSCs undergo functional deterioration and gradually lose stemness with systematic age in vivo or extended culture in vitro, limiting their therapeutic applications.

Even though our understanding of the processes behind MSC senescence remains unclear, significant progress has been achieved in elucidating the aspects of the age-related MSC phenotypic changes and possible mechanisms driving MSC senescence. In this review, we aim to summarize the current knowledge of the morphological, biological, and stem-cell marker alterations of aging MSCs, the cellular and molecular mechanisms that underlie MSC senescence, the recent progress made regarding the innovative techniques to rejuvenate senescent MSCs and combat aging, with a particular focus on the interplay between aging MSCs and their niche as well as clinical translational relevance. Also, we provide some promising and novel directions for future research concerning MSC senescence.

Decreased Ribosomal Biogenesis in Some Long-Lived Individuals
https://www.fightaging.org/archives/2022/05/decreased-ribosomal-biogenesis-in-some-long-lived-individuals/

Ribosomes in a cell are where proteins are assembled according to a messenger RNA blueprint. Like all cellular components, ribosomes are regularly created and recycled. Reduced production of new ribosomes is a feature of calorie restriction, slowed aging accompanied by a lower output of new proteins. Further, genetic alterations that force a reduction in ribosomal biogenesis also modestly slow aging in animal studies, so it is thought that the pace of protein production is a relevant mechanism in the connection between cellular metabolism and aging. Researchers here extend this line of research into humans, looking at ribosomal function and protein production in long-lived individuals.

As a paradigm of successful human aging, long-lived individuals (LLIs) achieve extreme old age without developing serious age-related diseases (e.g., cardiovascular disease, neurodegenerative disorders, and cancer). Gene expression is thought to have a close association with the activity of processes involved in healthy aging and longevity in LLIs. Here, to find the processes displaying reduced biological activities in long-lived people, we obtained and analyzed the transcriptomes of peripheral white blood cells from 193 female LLIs and 83 gender-matched spouses of LLI children (F1SPs) from two independent Chinese longevity cohorts.

Results showed that genes related to the ribosome pathway, especially ribosomal protein genes (RPGs), were significantly down-regulated in the LLIs. We also found that most of the RPGs were positively coexpressed with the ETS1 gene, which was down-regulated in LLIs. This gene encodes a transcription factor that binds to RPG promoters, and its down-regulation leads to the reduced RPG expression. Furthermore, knockdown of ETS1 alleviated cellular senescence and suppressed RPG transcription in human dermal fibroblast (HDF) and human embryonic lung fibroblast (IMR-90) cells.

Thus, these findings reveal that decreased ribosome biogenesis caused, at least in part, by the down-regulation of ETS1 exists in certain female LLIs and may contribute to healthy aging and life span extension in long-lived people.

An Interview with Researcher Thomas Kirkwood
https://www.fightaging.org/archives/2022/05/an-interview-with-researcher-thomas-kirkwood/

Here find a popular science interview with Thomas Kirkwood on his contributions to present thought on how and why degenerative aging evolved to be near universal in living organisms. At the high level, what we think that we know about the evolution of aging does to some degree inform the approaches taken to treat aging: in advance of firm data, should we expect one strategy to be better than another, and thus prioritize it?

"I wondered why cells allow damage to build up in the first place. And the idea came to me then, which was the realisation that it takes energy to combat the build-up of damage. There are maintenance and repair processes, proofreading mechanisms to make sure you don't do things wrong in the first place, and then clearance mechanisms to make sure you clean up your mistakes and get rid of them. And all of that costs energy."

"I started thinking that maybe this was the answer to question of why we age and die - because it was never evolutionarily worthwhile to invest enough in the maintenance and repair processes of the body to keep our cells from going on indefinitely. The essence of the theory is simply that, under the pressure of natural selection, organisms invest enough in the maintenance of somatic cells to keep them going for long enough to allow us to grow and reproduce and make the next generation, but it was never worthwhile for them to invest enough for those cells to last indefinitely."

"The theory says that aging will occur, because the whole repertoire of maintenance and repair systems will be tuned to a level that allows damage to build up. That has interesting implications in that it tells us that, from an evolutionary perspective, we should not expect there to be a single mechanism of aging. Very often you'll find groups of scientists that are championing one or other mechanism - so it's all telomeres, or it's all DNA mutations, or proteostasis collapse, or mitochondria, or cellular senescence. But the theory tells us is that it is not one mechanism versus another mechanism but that they work simultaneously, and they work in synergistic ways."

Improving the Understanding of How Hypertension Results in Vascular Restructuring
https://www.fightaging.org/archives/2022/05/improving-the-understanding-of-how-hypertension-results-in-vascular-restructuring/

The chronic raised blood pressure of hypertension is both (a) driven by the stiffening of blood vessel walls and (b) causes further detrimental restructuring of blood vessel walls, such as thickening of the intimal and medial layers. Researchers here explore the chains of cause and effect that lead to this outcome, mediated by increased inflammatory signaling and the presence of macrophages drawn into the blood vessel walls.

Persistent hypertension can cause long-lasting changes in the structure of vascular smooth muscle cells (the cells making up the walls of blood vessels) through a process called "vascular remodeling." If left unchecked, this restructuring can stiffen arterials walls, which lose their ability to adjust their size appropriately. This, in turn, leads to arteriosclerosis and increases the risk of cerebrovascular disease.

Why and how hypertension triggers vascular remodeling is not entirely clear. Scientists have shown that macrophages, a type of white blood cells that kill foreign bodies, are involved in the transformation. Specifically, the macrophages accumulate within blood vessel walls from outside the vessels and cause chronic inflammation. However, the underlying mechanism that orchestrates this process remains unknown.

A new study recently investigated a mechanism known as "excitation-transcription (E-T) coupling" in vascular smooth muscle cells. Although E-T coupling occurs in vascular smooth muscle cells after an influx of Ca2+ under high pressure, not much was known about how it happens, what genes are triggered, and the role it plays in our bodies.

By taking a detailed look at the genes promoted by E-T coupling and observing their effects when blocked or amplified, the researchers made some important discoveries. Firstly, some of these genes were related to chemotaxis, the phenomenon by which cells movement is triggered and directed by chemical stimuli. This helped explain the accumulation of macrophages in blood vessel walls from outside the vessels. Additionally, these genes promoted the remodeling of the medial layer of arteries, where vascular smooth muscle cells reside and control blood flow through contraction and expansion. "Taken together, our results explain how E-T coupling caused by high pressure in vascular smooth muscle cells can modulate macrophage migration and subsequent inflammation, altering the vascular structure,"

Reviewing Mitochondria-Derived Peptides in Aging
https://www.fightaging.org/archives/2022/05/reviewing-mitochondria-derived-peptides-in-aging/

Researchers have explored a number of mitochondria-derived peptides as a basis for treatments in the context of aging. These peptides are created from fragments of genes in the mitochondrial DNA, released from the cell, and appear to be involved in a range of mechanisms relevant to declining function in aging. Is it possible to supply such peptides as a therapy in order to produce benefits in an aged metabolism? A number of groups working towards that goal, on the basis of data in animal studies and humans patients.

The mechanisms that explain mitochondrial dysfunction in aging and healthspan continue to be studied, but one element has been unexplored: microproteins. Small open reading frames in circular mitochondria DNA can encode multiple microproteins, called mitochondria-derived peptides (MDPs). Currently, eight MDPs have been published: humanin, MOTS-c, and SHLPs 1-6.

MDPs have been extensively studied in the context of aging. Three MDPs have been studied in the context of age-related diseases: humanin, MOTS-c, and SHLP2. Humanin has been shown to mitigate Alzheimer's disease pathology in rodents, and its levels and genetic variation associate with age and cognition. MOTS-c has been described as an exercise mimetic and prevents muscle atrophy in mice, and its levels and genetic variation associate with age and type 2 diabetes (T2D). SHLP2 functions as a mitochondrial modulator and protein chaperone, and its levels associate with age and prostate cancer.

In addition to their ability to attenuate age-related diseases, MDPs have promoted lifespan and healthspan. Humanin is the best-conserved MDP and is found in as diverse species as humans, naked mole rats, and nematodes. Overexpression of humanin increased lifespan in nematodes, and this was dependent on FOXO. Additionally, humanin has increased autophagy in cells, and this increase in autophagy was also required for the lifespan extension in the transgenic worms. The second approach was to initiate a longevity experiment in mice in which we injected middle-aged (18-month-old) female mice with humanin twice a week. Although lifespan was not increased - likely because of humanin's short half-life of approximately 20 minutes - healthspan measures such as memory and metabolic parameters improved. Thus, humanin is sufficient to increase lifespan and healthspan in model organisms, and an optimized dosing of humanin may lead to increases in lifespan in more complex organisms.

The Concept of Cellular Exercise
https://www.fightaging.org/archives/2022/05/the-concept-of-cellular-exercise/

Researchers here coin a term, cellular exercise, to describe the benefits resulting from mild cellular stress and the consequent housekeeping responses. Increased cellular maintenance activities in response to mild stress lead to a net improvement in cell and tissue function. In short-lived laboratory species, interventions that provide chronic mild stress, such as calorie restriction, improve long term health and increase life span. Interventions based on this approach may be less interesting in long-lived species such as our own, however, given that, for example, calorie restriction provides up to 40% extension of life in mice, but at most a few years in humans.

"Cellular exercise" is a concept where low levels of cellular stress, induced by chronic calorie restriction or physical exercise, can lead to molecular adaptations on the cellular level that can protect the body from cancer and cardiovascular disease. An increase in reactive oxygen species induced by caloric restriction and physical exercise can produce improvements in redox equilibrium that can result in a more adaptive capable cell.

Insulin-like growth factor-1 has a dual effect wherein calorie restriction downregulates insulin-like growth factor-1 inhibiting pathways of carcinogenic proliferation and metastasis and physical exercise can upregulate insulin-like growth factor-1 to promote mitochondrial biogenesis and protein synthesis thereby strengthening healthy muscle against hypoxic ischemic damage and muscular regenerative properties. Transcription of Nrf2 is also upregulated to attenuate inflammation induced by nuclear factor-κB, AMPK upregulates genes through PGC-1α to prevent sarcopenia and induce lipolysis.

This molecular melody is the complex composition that explains the cellular adaption that occurs to strengthen the body from cognitive dysfunction, cardiometabolic failure and carcinogenic implantation and metastasis via mechanisms of redox equilibrium, oxidative protection, attenuation of inflammation, and attenuation of carcinogenic proliferation and growth.

Comment Submission

Post a comment; thoughtful, considered opinions are valued. New comments can be edited for a few minutes following submission. Comments incorporating ad hominem attacks, advertising, and other forms of inappropriate behavior are likely to be deleted.

Note that there is a comment feed for those who like to keep up with conversations.