• Log in with Facebook Log in with Twitter Log In with Google      Sign In    
  • Create Account
  LongeCity
              Advocacy & Research for Unlimited Lifespans

Photo

Fight Aging! Newsletter, July 19th 2021


  • Please log in to reply
No replies to this topic

#1 reason

  • Guardian Reason
  • 1,101 posts
  • 241
  • Location:US

Posted 18 July 2021 - 01:40 PM


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

  • 20 Million in Donations Made to the SENS Research Foundation in the Past Few Days
  • Is Depletion of Soluble Amyloid-β the Reason Why Amyloid is Important in Alzheimer's Disease?
  • Why Do Some Older People Retain a Good Memory?
  • Stem Cell Transplantation to Treat Chronic Inflammation and Frailty
  • SENS Research Foundation is Hiring Scientists to Work on the Foundations of Human Rejuvenation
  • Assessing Risk of Age-Related Disease is a Hard Problem, as Presently Attempted
  • Towards Direct Reprogramming of Cardiac Cells to Induce Regeneration in the Heart
  • A Reminder that Merely Elevated Blood Pressure Still Increases Cardiovascular Disease Risk
  • Clearance of Senescent Cells as an Approach to Limit Scar Formation in Nerve Injury
  • Castration Delays Epigenetic Aging in Male Sheep
  • Dihomo-γ-linoleic Acid as a Basis for Senolytic Therapy
  • CD40L Inhibition as an Approach to Reduce Inflammation in Atherosclerosis
  • Chronic Inflammation Negatively Impacts Proteostasis in Aging Tissues
  • Ferroptosis in Aging
  • It is Easy to Produce Omics Data, Harder to Achieve Useful Progress Based on that Data
  • The Interaction of Senescent Cells and Macrophages in Fibrosis

20 Million in Donations Made to the SENS Research Foundation in the Past Few Days

The principals of a new high-profile blockchain chose to link participation in their launch process to donations made to the SENS Research Foundation, one of the most important scientific and advocacy organizations working on the foundations of future rejuvenation therapies. More than 20 million in charitable donations were made in the last few days, four times the present yearly budget. We live in interesting times!

Is Depletion of Soluble Amyloid-β the Reason Why Amyloid is Important in Alzheimer's Disease?
https://www.fightagi...eimers-disease/

The long years of failure to improve outcomes in Alzheimer's disease patients via the development of immunotherapies targeting amyloid-β has provoked a great deal of alternative theorizing and new exploration regarding the causes of the condition. The amyloid cascade hypothesis of the progression of Alzheimer's disease is being modified in numerous ways. In its original form, the formation of deposits of misfolded amyloid-β causes inflammation and other forms of disarray that sets the stage for later aggregation of tau into neurofibrillary tangles, which leads to the widespread death of neurons.

Some researchers believe that chronic inflammation, or persistent infection, or senescent cell accumulation, or all three, are in fact the primary drivers of the development of Alzheimer's, with amyloid-β aggregation as a side-effect. In this case, the amyloid-β stage of the amyloid cascade is replaced with one or more other mechanisms, with tau aggregation, neuroinflammation, and cell death remaining as the end stage of the condition. Other groups see the failing drainage of cerebrospinal fluid from the brain as a major contributing factor, allowing molecular waste such as aggregated amyloid-β to build up in the brain. Vascular dysfunction and consequent reductions in the supply of oxygen and nutrients to the brain is another contender as an important cause: outright vascular dementia does overlap significantly with Alzheimer's disease.

Here, researchers propose yet another modification to the amyloid cascade, which is that the real problem is a reduction in the levels of functional amyloid-β. Misfolding and aggregation of amyloid-β might be presumed to contribute to that issue, but given that some people exhibit both aggregates and sufficient functional amyloid-β, it seems more likely that other mechanisms are at work. It is worth remembering that amyloid-β is an antimicrobial peptide, a part of the innate immune defense against pathogens. The findings here may ultimately fit into models of Alzheimer's disease in which the disruptive influence of persistent infectious agents are an important driving factor.

Researchers question prevailing Alzheimer's theory with new discovery

Cognitive impairment could be due to a decline in soluble amyloid-beta peptide instead of the corresponding accumulation of amyloid plaques. To test this hypothesis, researchers analyzed the brain scans and spinal fluid from 600 individuals enrolled in the Alzheimer's Disease Neuroimaging Initiative study, who all had amyloid plaques. From there, they compared the amount of plaques and levels of the peptide in the individuals with normal cognition to those with cognitive impairment. They found that, regardless of the amount of plaques in the brain, the individuals with high levels of the peptide were cognitively normal.

They also found that higher levels of soluble amyloid-beta peptide were associated with a larger hippocampus, the area of the brain most important for memory. According to the authors, as we age most people develop amyloid plagues, but few people develop dementia. In fact, by the age of 85, 60% of people will have these plagues, but only 10% develop dementia, they say. "The key discovery from our analysis is that Alzheimer's disease symptoms seem dependent on the depletion of the normal protein, which is in a soluble state, instead of when it aggregates into plaques."

High cerebrospinal amyloid-β 42 is associated with normal cognition in individuals with brain amyloidosis

This cross-sectional study of 598 amyloid-positive participants in the Alzheimer's Disease Neuroimaging Initiative cohort examined whether levels of soluble Aβ42 are higher in amyloid-positive normal cognition (NC) individuals compared to mild cognitive impairment (MCI) and Alzheimer's disease (AD) and whether this relationship applies to neuropsychological assessments and hippocampal volume measured within the same year.

Higher soluble Aβ42 levels were observed in NC (864.00 pg/ml) than in MCI (768.60 pg/ml) or AD (617.46 pg/ml), with the relationship between NC, MCI, and AD maintained across all amyloid tertiles. Each standard deviation increase in Aβ42 was associated with greater odds of NC than AD (adjusted odds ratio, 6.26) or MCI (1.42). Higher soluble Aβ42 levels were also associated with better neuropsychological function and larger hippocampal volume. Thus, normal cognition and hippocampal volume are associated with preservation of high soluble Aβ42 levels despite increasing brain amyloidosis.

Why Do Some Older People Retain a Good Memory?
https://www.fightagi...-a-good-memory/

The myriad ways in which the brain changes with age are in some ways very well explored, but overall still a dark forest, little understood in fine detail. One approach to gain greater understanding of the processes that cause declining cognitive function with age is to compare people with good function and people with poor function, first categorizing, and then secondly assessing the properties of the brain, as best researchers are able to do so, given limited access to the inside of the cranium. Today's research materials are an example of this sort of research, focused on trying to better understand why some older people retain a good memory function, while their peers decline.

There are numerous possibilities, even looking at broad categories of potential mechanisms. This could be a matter of slower degeneration, that some people make good lifestyle choices throughout life and as a result take longer to reach critical thresholds of damage and dysfunction that impact memory. One might look at recent research that suggests the hippocampus is running right at the upper limit of its supply of nutrients, and thus better maintained physical fitness into later life ensures that blood supply remains sufficient. Alternatively, some people may be more resilient to specific mechanisms of damage that impact areas of the brain, such as the hippocampus, that are important to memory. Some individuals have a high burden of amyloid-β in the brain, but little to no sign of neurodegeneration, for example.

Lastly, it is possible that some people exhibit a better set of age-related compensatory changes in the brain. This is perhaps driven by a greater pace of neurogenesis, allowing the creation of new neural networks and the replacement of damaged or dead neurons in existing networks. Neurogenesis in the hippocampus is meaningfully affected by exercise and gut microbiome, via mechanisms that include those related to expression of BDNF. The storage and processing of memories in the brain appears quite dynamic, and it is possible to envisage processes whereby memory is continually shuffled around and preserved through the loss of specific neurons or alternations to neural networks.

Study reveals source of remarkable memory of "superagers"

As we age, our brains typically undergo a slow process of atrophy, causing less robust communication between various brain regions, which leads to declining memory and other cognitive functions. But a rare group of older individuals called "superagers" have been shown to learn and recall novel information as well as a 25-year-old. The superagers are participants in an ongoing longitudinal study of aging. "Using MRI, we found that the structure of superagers' brains and the connectivity of their neural networks more closely resemble the brains of young adults; superagers had avoided the brain atrophy typically seen in older adults."

In the new study, the investigators gave 40 adults with a mean age of 67 a very challenging memory test while their brains were imaged using functional magnetic resonance imaging (fMRI), which, unlike typical MRI, shows the activity of different brain areas during tasks. Forty-one young adults (mean age of 25) also took the same memory test while their brains were imaged. While the participants were in the scanner, the researchers paid close attention to the visual cortex, which is the area of the brain that processes what you see and is particularly sensitive to aging.

In the visual cortex, there are populations of neurons that are selectively involved in processing different categories of images, such as faces, houses or scenes. During aging, this selectivity, called neural differentiation, diminishes and the group of neurons that once responded primarily to faces now activates for other images. The brain now has difficulty creating unique neural activation patterns for different types of images, which means it is making less distinctive mental representations of what the person is seeing. That's one reason older individuals have trouble remembering when they may have seen a television show, read an article, or eaten a specific meal.

But in the fMRI study, the superagers' memory performance was indistinguishable from the 25-year-olds', and their brains' visual cortex maintained youthful activity patterns. An important question that researchers still must answer is whether "superagers' brains were always more efficient than their peers, or whether, over time, they developed mechanisms to compensate for the decline of the aging brain.

Greater Neural Differentiation in the Ventral Visual Cortex Is Associated with Youthful Memory in Superaging

Superagers are older adults who maintain youthful memory despite advanced age. Previous studies showed that superagers exhibit greater structural and intrinsic functional brain integrity, which contribute to their youthful memory. However, no studies, to date, have examined brain activity as superagers learn and remember novel information. Here, we analyzed functional magnetic resonance imaging data collected from 41 young and 40 older adults while they performed a paired associate visual recognition memory task. Superaging was defined as youthful performance on the long delay free recall of the California Verbal Learning Test. We assessed the fidelity of neural representations as participants encoded and later retrieved a series of word stimuli paired with a face or a scene image.

Superagers, like young adults, exhibited more distinct neural representations in the fusiform gyrus and parahippocampal gyrus while viewing visual stimuli belonging to different categories (greater neural differentiation) and more similar category representations between encoding and retrieval (greater neural reinstatement), compared with typical older adults. Greater neural differentiation and reinstatement were associated with superior memory performance in all older adults. Given that the fidelity of cortical sensory processing depends on neural plasticity and is trainable, these mechanisms may be potential biomarkers for future interventions to promote successful aging.

Stem Cell Transplantation to Treat Chronic Inflammation and Frailty
https://www.fightagi...on-and-frailty/

Today's open access commentary is a good companion piece to a recent paper covering the use of mesenchymal stem cell therapies to suppress age-related chronic inflammation. These first generation stem cell therapies have proven to be unreliable when it comes to the original goal of regeneration of organ function, but they do reliably reduce excessive inflammation for some months. Transplanted stem cells near all fail to survive and engraft. Some clinics report better results than others on this front, but there is little understanding at present as to why similar cells sources and methodologies can produce wildly different outcomes in different hands. Benefits in most cases arise due to transient signaling by the transplanted cells that changes the behavior of native cells for some time.

The chronic inflammation of aging and physical frailty go hand in hand. Inflammation is disruptive of normal tissue maintenance, both of muscle tissue and in vital organs. Controlling inflammation can produce patient benefits. A great many clinical trials, including those aiming to treat age-related frailty, have been based on this approach of targeting regulatory mechanisms of inflammation. Still, first generation mesenchymal stem cell therapies are not yet as widely used as they might be, most likely due to the continued issues with consistency of outcomes from clinic to clinic and patient to patient. Much remains to be explored regarding the reasons why this variability exists.

Inflammation, a common mechanism in frailty and COVID19, and stem cells as a therapeutic approach

Many research labs have developed cell therapies in search for tissue homeostasis improvement. To date, there are more than 38 clinical trials using stem cells against the effect of aging or frailty, although there are far fewer with mesenchymal stem cells (MSCs). Recently, randomized double blind studies showed that intravenous administration of allogeneic MSCs is safe, renders improved physical performance, and reduces inflammatory markers increased in frailty states.

In the first of these trials, 15 patients with mild to moderate frailty were treated with MSCs. This phase 1 study focused on safety evaluating severe adverse effects during 12 months after the injection of 20 to 200 million MSCs. Besides safety, the results showed improvements in physical activity, cognitive hallmarks, and bloodstream TNF-α levels. In phase II (random, double blind with placebo), 30 frailty patients were injected with 100 to 200 million MSCs resulting in positive results in activity hallmarks and several immune biomarkers 6 months after the injection. These studies, together the other trials, support the safety and efficacy of the intravenous injection of allogeneic MSCs from bone marrow against frailty.

The downside of these studies is the lack of consistency among the methodology used, with significant variations in the number of infused cells, cell origin, quality of the donor and their MSCs, and hemocompatibility, all common problems of MSC therapies. Tolerance to these treatments, demonstrated in hundreds of patients in multicenter trials, and the reversion of some parameters compromised in frailty make these therapies a well-founded hope. However, to progress in this approach, the field will need the establishment of new and consistent animal models, as well as a better and systematized diagnosis of frailty conditions through more sensible and validated biomarkers.

In summary, MSC research during the last decades has been a rollercoaster of promises, controversies, and unexpected discoveries, which have changed our perspective of their potential use as a therapy on different human conditions and diseases. We believe that although the original promises have not been met, the intense dedication to their study has opened new alternatives to use their less known paracrine properties on immunomodulation and aging to find new solutions to extremely important challenges of public health, such as the increasing incidence of frailty conditions.

SENS Research Foundation is Hiring Scientists to Work on the Foundations of Human Rejuvenation
https://www.fightagi...n-rejuvenation/

The SENS Research Foundation is hiring scientists! This is a chance to work at one of the hubs of the field of aging research, with a highly influential group of researchers and patient advocates. The SENS Research Foundation and its network of allies have played an important role in turning investigation of the mechanisms of aging from a toy field, in which intervention was never considered, into a serious field of translational research that has given rise to a growing biotech industry focused on slowing and reversing the processes of aging. In addition to advocacy, the SENS Research Foundation staff work to unblock slow-moving or underfunded areas of research that are nonetheless important to the development of future rejuvenation therapies.

The Strategies for Engineered Negligible Senescence (SENS) view of aging is a synthesis of the past century of data, focused on the accumulation of cell and tissue damage that arises as a result of the normal operation of a youthful metabolism. This is the root cause of aging, and periodic repair of this damage should be sufficient to produce meaningful rejuvenation. The first SENS position paper in 2002 included cellular senescence as a plausible contributing cause of aging and target for therapies, and today there are a dozen or more biotech companies working on senolytic therapies to clear senescent cells, while first generation senolytics have been shown to produce rejuvenation in mice, and are undergoing human trials for age-related conditions.

SENS Research Foundation Career Opportunities

Research Associate / Scientist - Boominathan Lab (MitoSENS)

The Boominathan lab at SENS Research Foundation is hiring highly motivated Research Scientists / Associates for a project geared toward translational therapies for mitochondrial dysfunctions. The successful candidate will use in vitro, in vivo, and stem cell models to address diseases due to mitochondrial DNA mutations. This research position is within a small but dynamic group that strives to develop a deep understanding and curative therapies using a gene therapy approach to treat mitochondrial myopathies.

Postdoctoral Research Fellow and Research Associate - Catabody Project

We seek a postdoctoral fellow to join our small but dynamic immunology team led by Dr. Amit Sharma. The project geared towards developing a novel way to remove abnormal tau aggregation. The project is potentially relevant for developing therapeutic mitigation of normal age-dependent cognitive decline, as well as for tauopathies like Alzheimer's disease and related disorders. This project involves utilizing enzymatic antibodies to target toxic tau aggregates. As part of the project we will explore ways of delivering antibodies into cells. We will use human induced pluripotent stem cell derived neuronal cells as a model system to test the catalytic antibodies and confirm tau degradation.

Postdoctoral Research Fellow and Research Associate - Senescence Immunology

We seek a postdoctoral fellow to join our small but dynamic immunology team led by Dr. Amit Sharma for a project geared toward investigating the mechanisms involved in the age-dependent decline in immune surveillance of senescent cells with the aim of finding promising interventions. There are three main projects currently for the postdoctoral fellow. One of the projects involves characterizing age-dependent phenotypes changes in the of Natural Killer cells and its implication on their ability to eliminate senescent cells in cell culture and mice models. The goal of the second project is to characterize the surface antigens on senescent cells and with the goal of developing CAR-NK cells with therapeutic application. The aim of the third project is to develop therapeutic interventions based on removal of these SASP proteins for enhancing immune surveillance of senescent cells.

Assessing Risk of Age-Related Disease is a Hard Problem, as Presently Attempted
https://www.fightagi...ntly-attempted/

As is discussed in today's open access paper, determining the risk of age-related disease is far from a solved problem. This is true even for cardiovascular disease, caused by degenerative processes that occur in every individual over the course of later life, and which would kill everyone in a world absent other fatal age-related conditions. Assessment of cardiovascular disease risk has received decades of sizable funding, large studies, and considerable attention from the research and medical communities. And yet it is still possible to write a lengthy paper on the very real shortcomings of present assessment approaches.

I believe that challenges in assessment of risk are a consequence of trying to assess risk based on factors that are only indirectly connected to causative processes. Aging is caused by forms of underlying molecular damage, and this damage creates a spreading network of downstream consequences, and further damage and problems caused by those consequences. A great deal of variability is present from individual to individual in this network, and so picking out parts of it may not be a good reflection of the actual burden of underlying, root cause damage. It would be better to assess that damage.

To take one example, senescent cell accumulation is an important contributing cause of aging. It isn't the only important contributing cause of aging, but it does appear to cause widespread dysfunction in tissues and systems throughout the body. Measuring senescent cell burden, once good non-invasive approaches are available to achieve this goal, should in principle be a better marker of disease risk than constructs based on lifestyle, diet, weight, and so forth. Once assays for cellular senescence exist, such as the blood sample microRNA approach under development by TAmiRNA, I'd imagine that we'll find out whether or not that is the case within a few years.

Cardiovascular risk and aging: the need for a more comprehensive understanding

The first half of the 20th century was marked by a shift in morbidity and mortality patterns in industrialized countries all over the world, moving from the leading role of infectious diseases to the increasing role of chronic, non-communicable diseases. In particular, cardiovascular disease (CVD) has been an important cause of morbidity and mortality, with coronary heart disease (CHD) being the leading cause of death. The primary reason for this transition was the discovery of antibiotics and vaccines, the widespread use of which has led to a decline in infectious diseases and increase in life expectancy and population aging.

CVD is a leading cause of morbidity and mortality worldwide, with the highest incidence and prevalence in an older population (> 60 years). Ever since the traditional CV risk factors were identified in the Framingham Heart Study, at the end of the 20th century, they have been used as the basis of risk-based strategies for predicting CVD and initiating drug therapy in primary CVD prevention. A number of predictive functions and score systems have been developed for CV risk assessment. Although there are some variations between the systems, most of them use the same limited set of variables, including age, sex, smoking, blood pressure, and cholesterol, to predict the ten-year absolute risk for developing CVD, or CVD-related death.

The current CV risk assessment systems have several limitations, which limit their implementation in practice and their efficacy in reducing the burden of CVD. Most systems perform well in the population from which they were derived but not in other populations. It is necessary to recalibrate the prediction equation for application in other populations, to allow for different CVD mortality rates and risk factor distributions. Another limitation is these systems' inability to represent the inter-individual variations in the CV risk accurately, so that a substantial proportion of individuals are wrongly classified, which can lead to either insufficient treatment or overtreatment. The variable age is the strongest CV predictor, and the current prediction models cannot distinguish between age and other risk factors.

Recent studies indicate that the effects of traditional CV risk factors attenuate among older individuals, and that other age-related factors, including comorbid conditions, become important for predicting CVD in older age. Based on the current knowledge, CVD develops concurrently with many comorbidities and other geriatric conditions, such as frailty, malnutrition, and sarcopenia, which share the common mechanisms and pathophysiology pathways as CVD. This is the reason why older people are very heterogeneous with respect to differences in their health status and functional performances, which makes CV risk prediction in older individuals complicated.

Towards Direct Reprogramming of Cardiac Cells to Induce Regeneration in the Heart
https://www.fightagi...n-in-the-heart/

Researchers have for some years proposed reprogramming of scar tissue cells in the injured heart as a way to produce a regrowth of healthy tissue, an outcome that does not normally occur. The heart is one of the least regenerative organs in mammals, and injury produces scarring and loss of function. A great deal of effort has gone towards the establishment of cell therapies to treat heart injuries, with some limited success, but reprogramming of native cells may prove to be a better option in the long term. As noted here, however, there is a great deal of work left to accomplish between the present state of the art and a future in which scar tissue in the heart can be safely reprogrammed into functional muscle.

The heart is composed of different types of cells, and cardiac function is carefully regulated, not only by cardiomyocytes, but also by other cells, such as vascular endothelial cells and fibroblasts. Cardiomyocytes account for approximately 30% of all cells in the heart, and at least 50% of the remaining cells are non-cardiomyocytes. Cardiomyocytes are terminally differentiated cells with no potential for self-renewal; cardiomyocytes that become necrotic due to myocardial infarction, heart failure, or other cardiac diseases are therefore replaced by proliferating fibroblasts. This situation results in scarring of the affected site due to the formation of fibrotic tissue. These fibrotic changes reduce the cardiac systolic function, and arrhythmia caused by scar tissue has a poor prognosis.

One promising approach to cardiac regeneration is to differentiate stem cells, such as induced pluripotent stem cells (iPS cells) into cardiomyocytes outside the body, and then transplant the differentiated cardiomyocytes into the body. However, generating the large numbers of cells required to replace as many as 1 billion cardiomyocytes lost to myocardial infarction or failing heart incurs enormous costs. It also poses other limitations, such as the presence of residual stem cells undergoing oncogenesis, and a low survival rate of transplanted cells

In 2010, we reported a novel strategy for the direct reprogramming of fibroblasts into cardiomyocytes. Based on these results, there are currently three possible pathways for the creation of cardiac muscle from fibroblasts. The three pathways can be summarized as follows: (1) full reprogramming of fibroblasts into iPS cells and subsequent cardiac differentiation, (2) partial reprogramming of fibroblasts into cardiac progenitor cells and subsequent differentiation, and (3) direct reprogramming of fibroblasts into cardiomyocytes. We proposed the concept of "direct cardiac reprogramming" in place of this conventional method of cell transplantation. This is a technique that converts cardiac fibroblasts, which are present in large numbers in the myocardium in cardiac direct reprogramming, into cardiomyocytes.

Three cardiogenic transcription factors: Gata4, Mef2c, and Tbx5 can induce direct reprogramming of fibroblasts into induced cardiomyocytes (iCMs), in mice. However, in humans, additional factors, such as Mesp1 and Myocd, are required. Inflammation and immune responses hinder the reprogramming process in mice, and epigenetic modifiers such as TET1 are involved in direct cardiac reprogramming in humans. Direct cardiac reprogramming needs improvement if it is to be used in humans, and the molecular mechanisms involved remain largely elusive. Further advances in cardiac reprogramming research are needed to bring us closer to cardiac regenerative therapy.

A Reminder that Merely Elevated Blood Pressure Still Increases Cardiovascular Disease Risk
https://www.fightagi...r-disease-risk/

The old guidelines for systolic blood pressure drew the line for increased risk of cardiovascular disease at 140 mmHg, with higher systolic blood pressure defined as hypertension. That dividing line was then moved down to 130 mmHg. In the past few years, further evidence has shown that elevated systolic blood pressure of 120 mmHg or above still produces increased risk, and that one shouldn't feel comfortable and safe in the 120-129 mmHg range. The risk of cardiovascular disease scales up with increasing blood pressure, and as noted here, also with the modern lifestyle choices leading to excess fat tissue, metabolic disease, and type 2 diabetes.

Blood pressure is written as two numbers. The first (systolic) number represents the pressure in blood vessels when the heart contracts or beats. The second (diastolic) number represents the pressure in the vessels when the heart rests between beats. Hypertension is diagnosed if, when it is measured on two different days, the systolic blood pressure (SBP) readings on both days is ≥140 mmHg and/or the diastolic blood pressure (DBP) readings on both days is ≥90 mmHg.

"The 2017 American College of Cardiology (ACC) / American Heart Association (AHA) BP guideline defined blood pressure ≥130/80 mm Hg as hypertension. This guideline showed that the normal level is less than 120/80 mm Hg and SBP 120-129 mm Hg and DBP < 80 mm Hg is elevated BP. However, little is known regarding whether elevated BP versus normal BP is specifically associated with a higher risk for coronary artery disease / cerebrovascular disease according to glucose tolerance status in real-world settings."

the authors addressed these research questions using a nationwide claims-based database that included information on 805,992 people enrolled with a health insurance provider for company employees and their dependents in Japan. In one arm of the study, they compared the cumulative incidence of coronary artery disease according to their SBP in individuals with normal, borderline, and elevated blood glucose, separately. The authors reported that, "a linear relationship was observed between cumulative incidence rates of coronary artery disease and SBP categories across all glucose tolerance status designations using SBP below 119 mmHg as the reference".

In another arm of the study, the investigators compared the cumulative incidence of cerebrovascular disease according to their SBP in individuals with normal, borderline, and elevated blood glucose, separately. Similarly, the authors observed a linear dose-response relationship between cumulative incidence rates of cerebrovascular disease and SBP categories across all glucose tolerance status. The study also found that combined together, the blood glucose status and blood pressure values had a synergistic effect on the incidence of coronary artery disease and cerebrovascular disease.

Clearance of Senescent Cells as an Approach to Limit Scar Formation in Nerve Injury
https://www.fightagi...n-nerve-injury/

Senescent cell behavior following injury is different and the clearance of these cells much more efficient in species like zebrafish and salamanders capable of regrowing organs. Researchers here suggest that senolytic therapies to selectively destroy senescent cells could be used in mammals to limit the scar formation that follows nerve injury, an important goal in enabling regrowth and restoration of nerve function. Their particular interest is spinal cord injury, their work should be applicable to the rest of the nervous system as well.

Mammals have a poor ability to recover after a spinal cord injury which can result in paralysis. A main reason for this is the formation of a complex scar associated with chronic inflammation that produces a cellular microenvironment that blocks tissue repair. Researchers have now shown that the administration of drugs that target specific cellular components of this scar can improve functional recovery after injury.

Researchers have been studying spinal cord injury using two different models: the zebrafish, where there is spinal injury recovery, and mammals that show poor recovery. The dense scar that forms at the lesion site has been of particular interest. In mammals, upon spinal cord injury, researchers observed that cells start to accumulate at the lesion periphery. But not any cells: "These cells are known as senescent cells. They have specific features and markers and are what we can call 'zombie cells', where growth and division is interrupted, but where the normal cell death program is not activated."

"While in zebrafish, the accumulation of these cells at the injury periphery is cleared out over time, in mammals, these cells persist and are important components of the dense scar observed. Because senescent cells have specific molecular markers, there are specific drugs that could be tested in this context. With the administration of different senolytic drugs, that specifically target these senescent cells, we have observed a progressive decrease of these cells, a decrease in the scar extension and lower levels of inflammation due to a decreased secretion of pro-fibrotic and pro-inflammatory factors. The observed changes at the molecular level underlie the improved locomotor, sensory, and bladder functions that we have also found."

Castration Delays Epigenetic Aging in Male Sheep
https://www.fightagi...-in-male-sheep/

Castration is known to extend life in male sheep. Researchers here show that epigenetic clocks constructed for this species show the expected slowing of epigenetic aging following castration. This is a way to dig deeper into the question of how it is that females live longer than males in mammalian species, an exploration of which mechanisms are important in determining that outcome. It is also a way to further explore how epigenetic clocks relate to biological aging. Since the clocks are constructed by machine learning approaches applied to epigenetic data, it remains far from clear as to what exactly they measure under the hood, meaning which of the processes of aging are driving changes in specific epigenetic marks used in the clocks.

In mammals, females generally live longer than males. Nevertheless, the mechanisms underpinning sex-dependent longevity are currently unclear. Epigenetic clocks are powerful biological biomarkers capable of precisely estimating chronological age and identifying novel factors influencing the aging rate using only DNA methylation data. In this study, we developed the first epigenetic clock for domesticated sheep (Ovis aries), which can predict chronological age with a median absolute error of 5.1 months. We have discovered that castrated male sheep have a decelerated aging rate compared to intact males, mediated at least in part by the removal of androgens.

Furthermore, we identified several androgen-sensitive CpG dinucleotides that become progressively hypomethylated with age in intact males, but remain stable in castrated males and females. Comparable sex-specific methylation differences in MKLN1 also exist in bat skin and a range of mouse tissues that have high androgen receptor expression, indicating that it may drive androgen-dependent hypomethylation in divergent mammalian species. In characterizing these sites, we identify biologically plausible mechanisms that explain how androgens drive male-accelerated aging.

Dihomo-γ-linoleic Acid as a Basis for Senolytic Therapy
https://www.fightagi...olytic-therapy/

This interview with a researcher working on the biochemistry of senescent cells notes the exploration of dihomo-γ-linoleic acid and derived compounds as potential senotherapeutics, capable of reducing the burden of senescent cells in old animals. At the end of the day there will be a very large number of such approaches, as the animal data for rejuvenation resulting from the clearance of senescent cells is impressive enough to drive a considerable growth in funding and interest. A sizable number of biotech companies are working on drugs to selectively destroy senescent cells, and many more programs are in earlier stages in academic labs.

There is a specific fatty acid made in small amounts in the body called dihomo-gamma-linoleic acid or DGLA. It's also present in tiny amounts in the diet. When I gave aged mice larger amounts of DGLA, they went from having quite a few senescent cells to having significantly fewer. This presents a new therapeutic target. I identified a candidate compound using the DGLA metabolic pathway that works at a dose that is over 1,000 times lower than fisetin, so you can imagine we're quite excited by these results.

Like many biomedical discoveries, it was accidental. DGLA makes anti-inflammatory lipids, which help alleviate conditions such as rheumatoid arthritis. I was studying this aspect of DGLA when I was surprised to discover that it killed senescent cells. My work is in its very early stages, and we've only studied a small number of mice, so it's too early for even tentative conclusions, although I'm obviously pleased that we've seen the elimination of a meaningful number of senescent cells in old mice. We'll be closely monitoring DGLA's positive effects as well as any negative effects on the mice.

First, we have to figure out how DGLA is killing senescent cells in mice. Again, not all studies with mice yield similar results in humans, so we are very careful about how we convey our findings and possible future actions. But I have met USDA researchers and nutrition scientists, and discovered that some of those folks were developing DGLA-enriched soybeans. In one scenario, you might go out for sushi and get a little bowl of DGLA-enriched edamame as a side. By the time you're done eating, you've helped reduce the odds of getting some age-related pathology. I don't know if it will play out that way, but it's an idea we're working toward. I also am working on therapies that elevate the amount of naturally occurring DGLA in senescent cells that I am very excited about, so this would be an alternative approach.

I am developing a quick and easy test to tell if senolytic therapy is working. Testing for senolytic effectiveness is not really being done now - you just look for improvement in symptoms or functioning and essentially conclude that it's due to the therapy. One way to solve this dilemma is to identify a biomarker, a measurable compound that consistently and reliably can confirm an intervention's effectiveness. For example, we know that a certain lipid, dihomo-15d-PGJ2, accumulates in large amounts inside of senescent cells. When we give a senolytic therapy that kills these cells in mice or human cells, this lipid is liberated. Detecting it in blood and urine is far less invasive, so that's what I'm working on now. Our aim is to be able to test people receiving senolytic therapy for the presence of dihomo-15d-PGJ2 in their blood and urine by the end of the summer.

CD40L Inhibition as an Approach to Reduce Inflammation in Atherosclerosis
https://www.fightagi...therosclerosis/

Atherosclerosis is an inflammatory condition. It is caused by dysfunction in the macrophage populations responsible for maintaining blood vessel walls, allowing fatty plaques to form, eventually leading to heart attack and stroke. This dysfunction is aggravated by a background of inflammatory signaling, and so there is some interest in finding ways to selectively interfere without preventing the beneficial activation of inflammation needed for normal immune function. That said, studies suggest that targeting inflammation in atherosclerosis is no more helpful than reductions in blood cholesterol, which is to say a modest reduction in mortality risk and only minimal reversal of existing lesions.

The protein CD40L is synthesized by, and expressed on the surface of specialized cells of the immune system. It is recognized by the CD40 protein, a membrane-bound receptor that is expressed on antigen-presenting cells. However, CD40L also binds to receptors on other cell types that have diverse physiological functions. Using a mouse model, researchers deleted the gene for CD40L specifically in T cells and platelets as well as its counterpart, CD40, on dendritic cells. They then crossed these mice with a strain that is particularly prone to develop atherosclerosis.

Secretion of interferon-gamma by T-cells is known to stimulate immune functions, but the CD40L-deficient T-cells were found to secrete less interferon-gamma than those in which the gene is intact. In addition, further experiments indeed showed that, in the absence of CD40L in T-cells, the atherosclerotic plaques that formed were smaller and more stable. This suggests that inhibition of CD40L could enhance the stability of atherosclerotic plaques, and thus reduce the incidence of heart attacks induced by the rupture of blood vessels.

Similar results were obtained in a mouse strain that was unable to produce CD40 in dendritic cells. Deletion of CD40L in platelets, on the other hand, had no effect on the incidence of atherosclerosis, but it was associated with a reduction in atherosclerosis-associated clot formation. Researchers are now extending their studies of the effects of CD40 und CD40L to other cell types, with the aim of developing drugs that can inhibit the functions of these proteins in a cell-specific fashion.

Chronic Inflammation Negatively Impacts Proteostasis in Aging Tissues
https://www.fightagi...-aging-tissues/

Proteostasis describes the steady state of a cell, maintaining an appropriate balance of various forms of protein machinery in order to enable continued normal function. With advancing age, proteostasis becomes disrupted in numerous complicated ways. This is a downstream outcome of underlying molecular damage, the reactions to that damage, and immediate consequences of that damage. When a machine becomes worn and broken, it functions poorly. That is a simple thing to observe in a simple machine, but a cell is an enormously complex machine, and exhibits enormously complex dysfunctions as it departs from the proteostasis that is normal for youthful tissues.

The progressive decline in the buffering capacity of the proteostasis network represents one of the molecular hallmarks of aging. However, the biological reasons why the proteostasis network deteriorates during aging are complex and not well understood. A progressive decrease in the activity and efficacy of the protein quality control systems, as well as in the mechanisms mediating the functional cooperation between them, could be the cause of these dysfunctions.

A growing body of evidence indicates a complex and bidirectional association between protein quality control systems and inflammation. For example, Th1 or Th2 cytokines stimulated or inhibited autophagy, respectively. Also, TNF-α modulated proteasome and autophagy function in human skeletal muscle cells. LPS-induced neuroinflammation produced ER-stress and altered proteasome and autophagy activity. Also, unfolded protein response (UPR) activation has been found to increase the production of inflammatory cytokines. UPR components can activate the transcription factor NFκ-B, which has a pivotal role in the onset of inflammation.

Because aging is associated with a low grade of chronic inflammation, the modulation exerted by inflammation on cellular proteostasis might be particularly relevant in aged cells. For example, the immunoproteasome, which is not expressed in cells from young animals, is expressed in rat and human aged cells from several tissues. Moreover, proteasome turnover is regulated by neuroinflammation. Most of the age-related alterations observed in cellular proteostasis are often reproduced in young animals following LPS injection. For example, LPS induced the expression of the immunoproteasome and decreased proteasomal activity leading to the accumulation of polyubiquitinated proteins in pyramidal neurons.

This data collectively indicates that inflammation and proteostasis alteration should be considered as synergistic negative factors that might increase cell vulnerability in aging. This is especially relevant in the context of some age-related pathologies such as obesity, hypertension, diabetes, and neurodegenerative disorders, all of them characterized by oxidative stress and inflammation. However, having in mind the complexity in the reciprocal influences between inflammation and the different protein quality control systems, as well as the cell specificity of these interactions, further studies in the context of aging will be necessary to better understand the synergistic negative effects of these two processes.

Ferroptosis in Aging
https://www.fightagi...tosis-in-aging/

Ferroptosis is a mode of programmed cell death that manages to be both fairly well explored in the broader research community and far less visible than other programmed cell death processes. It was first named and described about a decade ago, though of course researchers have long explored aspects of its biochemistry. There is some thought that ferroptosis may be connected to lysosomal dysfunction and accumulation of molecular waste in long-lived cells of the central nervous system, but in general it isn't much mentioned in the aging research field. This paper here provides an overview of why ferroptosis might be an interesting area of investigation, particularly in the context of neurodegenerative conditions.

Life is indeed continuously going through the irreversible and inevitable process of aging. The rate of aging process depends on various factors and varies individually. These factors include various environmental stimuli including exposure to toxic chemicals, psychological stress whereas suffering with various illnesses specially the chronic diseases serve as endogenous triggers. The basic underlying mechanism for all kinds of stresses is now known to be manifested as production of excessive ROS, exhaustion of ROS neutralizing antioxidant enzymes and proteins leading to imbalance in oxidation and antioxidant processes with subsequent oxidative stress induced inflammation affecting the cells, tissues, organs, and the whole body.

All these factors lead to conventional cell death either through necrosis, apoptosis, or autophagy. Currently, a newly identified mechanism of iron dependent regulated cell death called ferroptosis, is of special interest for its implication in pathogenesis of various diseases such as cardiovascular disease, neurological disorders, cancers, and various other age-related disorders. In ferroptosis, the cell death occur neither by conventional apoptosis, necrosis, nor by autophagy, rather dysregulated iron in the cell mediates excessive lipid peroxidation of accumulated lethal lipids. It is not surprising to assume its role in aging as previous research have identified some solid cues on the subject.

In this review, we will highlight the factual evidences to support the possible role and implication of ferroptosis in aging in order to declare the need to identify and explore the interventions to prevent excessive ferroptosis leading to accelerated aging and associated liabilities of aging.

It is Easy to Produce Omics Data, Harder to Achieve Useful Progress Based on that Data
https://www.fightagi...d-on-that-data/

The enormous reduction in cost and increase in capacity for analysis of living biochemistry over the past 20 years has led to vast warehouses of omics data: information on genomes, epigenomes, expression of transcripts and proteins, and more. Making something of this data in a reliable way is a more challenging proposition, and remains a work in progress. More data is almost always good in the long run, but the goal of science is understanding, not implementation. The data revolution in biotechnology may not greatly change the nature of the fastest path to human rejuvenation, which is to implement the SENS proposals for damage repair and see what happens. In the case of removing senescent cells, we can see that this produces rapid rejuvenation in mice, to a degree that is dramatic in comparison to any other approach to aging tested to date. If a tenth of the effort that goes into producing omics data went into furthering the SENS research agenda, we'd be much further along the road to radical life extension.

Biogerontologists are nowadays struggling with identifying actionable mechanisms of aging, with the goal of extending the time individual lives in good health, possibly delaying age-related diseases, and therefore reaching longevity. The issue is not simple to solve. In fact, although our understanding of aging biology in model systems has increased dramatically, thanks to the possibility to model the effect of single variants on the probability to extend our lifespan, Human aging and longevity are complex polygenic traits. They are influenced by the inheritance pattern of multiple genes/variants, each one with pleiotropic protective roles across several age-related diseases, and their interaction with environment. People can achieve older age while suffering major age-related diseases, because of their capability to survive those disorders, or they can escape entirely some of the most frequent causes of death and impairment, thus living not just a long but also a healthy life. The difference between these two aging trajectories and phenotypes is greatly discussed and investigated.

Biomedical innovation, and in particular research into "omics technologies," offers the promise of monitoring, preventing and treating age-related disabilities and diseases. Progress in genomics and functional genomics in the past decades have significantly supported our understanding of the molecular mechanisms associated with aging. However, it is nowadays clear that the complexity of aging requires a huge effort into data integration, building a broader omics profile, including genomics, proteomics, lipidomics or metabolomics, transcriptomics, etc.

Although the capacity to produce big data drastically increased over the years, integration, interpretation and sharing of high-throughput data remain major challenges. This seems even more challenging in the field of aging, because such an effort requires a more holistic view. Aging is not just the progressive decline of different functions, but rather a well-described phenotype, characterized by a complex remodeling across the whole organism. This is the key reason why omics technologies may greatly improve the definition of different aging phenotypes, and the classification of individuals with features ranging from the very frail, with a poor quality of aging, to the most extreme, the centenarian's phenotype, characterized by a long life.

The Interaction of Senescent Cells and Macrophages in Fibrosis
https://www.fightagi...es-in-fibrosis/

The interaction between senescent cells and macrophages is of great importance to wound healing. Differences in the behavior of these two cell types appear critical to proficient regeneration in species like salamanders versus poor regeneration and scarring in mammals. Fibrosis is a malfunction of tissue maintenance and regeneration, in which excessive scarring takes place, disrupting tissue function and structure. This too is connected to the presence and behavior of senescent cells and macrophages. In old individuals, there is a background of raised inflammatory signaling and a growth in lingering senescent cells. One way to look at fibrosis is that this chronic inflammation and persistence of senescent cells interferes in the normal signaling between transient senescent cells and macrophages in regeneration and tissue maintenance, leading to pathological outcomes.

Senescent cells are attractive candidates as drivers of age-related organ dysfunction. They are consistently seen in diseased and older tissues when compared with healthy age-matched controls, actively secreting pro-inflammatory and pro-fibrotic molecules capable of driving further (paracrine) senescence and propagating on-going tissue damage. This is potentially because they secrete pro-inflammatory cytokines in the senescence-associated secretory phenotype (SASP) which modify the surrounding environment.

Macrophages contribute to clearance of senescent cells by phagocytosis. This activity declines with age in multiple organ systems, including the kidney, as macrophages polarize from M1 to M2 in response to exogenous growth factors, and can potentially become 'senescent-associated' and possibly senescent themselves. This is followed by a concurrent increase in fibrosis with age, which negatively affects organ function.

New therapy strategies have been developed, both pharmaceutical and lifestyle changes that aim at reducing the burden of senescent cells and the SASP they generate, and reducing inflammation, aimed at removing blockades for macrophage polarity transitions essential for response to injuries. In this review, we examine senescent cells and the overlap between the direct biological impact of senescence and the indirect impact senescence has via its effects on other cell types, particularly the macrophage. The canonical roles of macrophages in cell clearance and in other physiological functions are discussed with reference to their functions in diseases of the kidney and other organs. We also explore the translational potential of different approaches based around the macrophage in future interventions to target senescent cells, with the goal of preventing or reversing pathologies driven or contributed to in part by senescent cell load in vivo.


View the full article at FightAging




0 user(s) are reading this topic

0 members, 0 guests, 0 anonymous users