×

The Blog

Building a Future Free of Age-Related Disease

Human Pilot Study Results for Senolytics Published

The results from a human pilot study that focused on treating idiopathic pulmonary fibrosis with senescent cell-clearing drugs has been published. The drugs target aged and damaged cells, which are thought to be a reason we age and get sick, and remove them from the body.

Senescent cells and aging

As we age, increasing numbers of our cells become dysfunctional, entering into a state known as senescence. Senescent cells no longer divide or support the tissues and organs of which they are part; instead, they secrete a range of harmful inflammatory chemical signals, which are collectively known as the senescence-associated secretory phenotype (SASP).

 

Dr. Judith Campisi from the Buck Institute for Research on Aging, along with her research team, identified that senescent cells secreted the various harmful chemicals that characterize the SASP in 2008, which was when interest in senescent cells really began [1]. In 2010, building on this initial research, Dr. Campisi went on to show the link between the SASP and cancer [2].

The SASP increases inflammation, harms tissue repair and function, causes the immune system to malfunction, and raises the risk of developing age-related diseases such as cancer. It can also encourage other nearby healthy cells to become senescent via the so-called bystander effect. Therefore, a small number of these cells can cause a great deal of harm.

Normally, senescent cells destroy themselves by a self-destruct process known as apoptosis before being cleared away by the immune system. Unfortunately, as we age, the immune system becomes weaker, and senescent cells start to build up in the body. The accumulation of senescent cells is considered to be one of the reasons why we age and develop age-related diseases.

It has been suggested that the clearance of senescent cells might help address a number of age-related diseases at once, as senescent cells are thought to be one of the fundamental reasons that we age. Drugs that can remove these unwanted, damaged cells are known as senolytics.

Human trial results for senolytics

This new publication by researchers at the Mayo Clinic, including James Kirkland, one of the pioneers of senolytic drugs, shows the results of a pilot study that uses dasatinib and quercetin to treat idiopathic pulmonary fibrosis [3].

Pulmonary fibrosis causes scarring of the lung tissue, which leads to the progressive loss of lung function over time. When the disease’s origin is unknown, it is called idiopathic pulmonary fibrosis, or IPF. The treatment options for this disease are extremely limited with no currently known cure.

The researchers in this new study tested a combination of dasatinib and quercetin, one of the earliest senolytic drug combinations that was tested in mice and shown to have beneficial results, particularly for the cardiovascular system [4-5]. It was also shown in a previous study that clearing senescent cells using dasatinib plus quercetin was able to alleviate idiopathic pulmonary fibrosis (IPF)-related dysfunction in a mouse model of the disease.

Fourteen patients with IPF were recruited for this pilot study, and the initial results, while leaving room for improvement, are promising.

Physical function evaluated as 6-min walk distance, 4-m gait speed, and chair-stands time was significantly and clinically-meaningfully improved (p < .05). Pulmonary function, clinical chemistries, frailty index (FI-LAB), and reported health were unchanged. DQ effects on circulating SASP factors were inconclusive, but correlations were observed between change in function and change in SASP-related matrix-remodeling proteins, microRNAs, and pro-inflammatory cytokines (23/48 markers r ≥ 0.50).

It should be noted that this was only a small pilot study and that the optimal human dosage and frequency is yet to be established. Typically, the next step is to launch a larger-scale study to establish this dosage.

The researchers also note that these results warrant evaluation of dasatinib plus quercetin in larger, randomized, and controlled trials for senescence-related diseases. In other words, they would like to test senolytics in larger studies for various age-related diseases, and the results certainly support doing exactly that.

Conclusion

These initial results are positive, despite there being plenty of room for improvement. The combination of these two drugs also appears to favor particular cell and tissue types over others, much like other senolytic drugs, which were discovered after dasatinib and quercetin were originally shown to clear senescent cells. It may be that a combination of different senolytics will be needed as a “cocktail” of sorts to fully clear out all the unwanted senescent cells, as different senescent cells appear to use various survival pathways to evade apoptosis, and no single drug can target them all.

We greet these early results positively and look forward to the beginning of larger-scale studies for multiple age-related diseases. Given how senescent cells appear to be implicated in most if not all age-related diseases, there are some exciting possibilities ahead.

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

Literature

[1] Coppé, J. P., Patil, C. K., Rodier, F., Sun, Y., Muñoz, D. P., Goldstein, J., … & Campisi, J. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS biology, 6(12), e301.

[2] Coppé, J. P., Desprez, P. Y., Krtolica, A., & Campisi, J. (2010). The senescence-associated secretory phenotype: the dark side of tumor suppression. Annual Review of Pathological Mechanical Disease, 5, 99-118.

[3] Nambiar, A., Justice, J., Pascual, R., Tchkonia, T., Lebrasseur, N., Kirkland, J., … & Kritchevsky, S. (2018). Targeting pro-inflammatory cells in idiopathic pulmonary fibrosis: an open-label pilot study of dasatinib and quercitin. Chest, 154(4), 395A-396A.

[4] Zhu, Y., Tchkonia, T., Pirtskhalava, T., Gower, A. C., Ding, H., Giorgadze, N., … & O’hara, S. P. (2015). The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging cell, 14(4), 644-658.

[5] Roos, C. M., Zhang, B., Palmer, A. K., Ogrodnik, M. B., Pirtskhalava, T., Thalji, N. M., … & Zhu, Y. (2016). Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging cell.

[/column]

An Interview with Dr. Kris Verburgh, M.D.

At the Fourth Eurosymposium on Healthy Ageing, which was held in Brussels last November, Elena and I met Dr. Kris Verburgh, a medical doctor who is especially interested in biogerontology and the potential of this field of study to turn medicine on its head.

Dr. Verburgh is only about 33 years old and has already written several science books—one of which, written when he was only 16, made him the youngest science author in Europe. Another prominent interest of his is nutrition, which he believes is one of the best, if not the best, ways we currently have to slow down the march of aging and buy ourselves more time to live until the rejuvenation age; his latest book, The Longevity Code, is centered around this topic.

Dr. Verburgh is also a strong supporter of the idea that AI will play a more and more important role in research, leading the way to a not-too-far age of personalized medicine—this was one of the theses he touched upon during the panel in which he participated at EHA.

Also a researcher at the Free University of Brussels, Dr. Verburgh agreed to this interview with us, where he shares his thoughts about aging, rejuvenation, and how both are perceived by the medical establishment.

As a medical doctor, what is your view on the idea that aging is not a one-way street and may be amenable to intervention?

Fascinating recent research shows that aging can be partially reversed in lab animals. These studies are important: they show that aging doesn’t have to be a one-way inexorable decline into frailty and decrepitude but that aging is a plastic process that is amenable to reversal. The ultimate goal of aging research isn’t simply just trying to slow down aging anymore but actually trying to partially reverse the aging process. Addressing aging itself is the most powerful method to address many aging-related diseases at the same time and keep people healthy for as long as possible.

What evidence has convinced you that this view is correct?

In various recent studies, old mice were rejuvenated. Old, inactive mice with grey fur get turned into active, younger-looking mice with a shiny black fur, while their cells and organs work and regenerate better. Such rejuvenation can be accomplished in various ways. One way is by epigenetic reprogramming. The epigenome is the complex molecular machinery that surrounds the DNA that determines which genes are active or switched off. When we get older, the epigenome gets dysregulated; some genes that shouldn’t be active become activated, like cancer-promoting genes, and vice versa. By using Yamanaka factors in a cyclical way, researchers could rejuvenate mice. Another way is by clearing away senescent cells. The older we get, the more that senescent cells accumulate everywhere in our tissues. These cells secrete substances that damage healthy neighboring cells. Senescent cells in the skin damage healthy skin cells, which plays a role in the formation of wrinkles. Senescent cells in cartilage damage the cartilage, contributing to osteoarthritis. Studies show that when you eradicate senescent cells, you partially reverse aging symptoms. Scientists are working on much other interesting technologies, not just to slow down aging but to also partially reverse it. Examples of such aging therapies are cross-link breakers, lysosomal enzyme therapy, aging-vaccines and autophagy-inducers to clear up aggregated protein, next generation stem cell therapies, telomere elongation, rejuvenating components in young blood, etc. We are living in very interesting times because scientists never before have had access to so many new biotechnology tools that finally enable them to impact the aging process.

What first got you interested in addressing aging?

As an MD, I realized that the best way to keep people healthy for as long as possible is by addressing the root cause of the majority diseases that afflict them, which is aging itself. Aging is the driving factor behind heart disease, Alzheimer’s disease, osteoporosis, sarcopenia, eye diseases such as macular degeneration, and so on. Addressing aging is the most powerful method to address all these diseases at the same time and is much more effective than focusing on treating only the individual diseases.

Do you have a particular model of aging that you favor, such as the Hallmarks of Aging, SENS, or the deleteriome?

Aging is very complex. Scientists have tried to boil it down into a few important aging mechanisms, or reasons why we age, such as epigenetic dysregulation, mitochondrial dysfunction, cross-linking, DNA damage, telomere attrition, or protein accumulation. However, it’s very unlikely that all of these aging mechanisms are equally important. Personally, I believe that epigenetic dysregulation is one of the more important causes of aging. When the epigenome is dysregulated, it can lead to more DNA damage, protein accumulation, cross-linking, and so on. In nature, a 30-year-old egg cell in a 30-year-old mother can be reprogrammed to zero years after fertilization so that a baby is born young, and it is composed of cells that are zero years old, not 30 years old like the mother. That is mainly accomplished by the epigenetic reprogramming of the fertilized egg cell. Also, during the cloning process, in which an old somatic nucleus from an old cell is transferred into a egg cell, the old nucleus gets reprogrammed into a much younger state, and this is also mainly through epigenetic changes. One of the most impressive studies in which aging was partially reversed is a study in which scientists epigenetically reprogrammed cells.

Do you agree with the idea that targeting the aging processes directly has the potential to delay, prevent, or even reverse multiple age-related diseases at once?

Yes. Aging is the root cause of aging-related diseases like cardiovascular disease or Alzheimer’s. Animals in which aging is retarded not only live longer but also get these diseases much later. They stay healthy for a much longer time. Long-lived species get aging-related diseases much later. Bowhead whales, which can live up to 220 years, do not get heart disease, cancer or Alzheimer’s disease when they are 90 years old, like we do. They get these diseases when they are 200 years old. Postponing aging is the most powerful way to keep people healthier for a much longer time.

Lots of people are focusing on curing single diseases, such as heart disease, cancer, and so on. However, even if we could cure one of these, would it really make much difference to our lifespans?

Addressing individual aging-related diseases, like heart disease, is not enough to really make an impact on the health of people. Even if we suddenly have a magical cure that could fix all heart disease, which is the most important cause of death in most developed countries, people would live only about 2.8 years longer. That is not much. People wouldn’t die of heart attacks anymore, but they would still die a few years later of another aging-related disease, like Alzheimer’s or cancer. So, therefore, it’s paramount to address aging itself, instead of treating individual aging diseases.

Current medical practice often tries to use an infectious disease approach to age-related diseases; in other words, it treats symptoms rather than causes. Why has medicine been slow to embrace the concept of treating age-related diseases by treating the aging processes directly?

Aging is often not a very well-taught subject at medical school. In medicine, most emphasis is put on treating aging symptoms and not on addressing the underlying cause, namely aging itself. Many MDs still think that aging is mainly caused by free radicals and DNA damage. Also, in medicine, aging is often considered to be a natural process, so there is no imperative to treat it. While most MDs consider aging to be a natural process, many aging researchers consider aging to be a 100 percent heritable, 100 percent fatal multisystemic disease caused by evolutionary negligence. In medicine, aging of individual organs is called a disease: aging of the heart is called heart disease and aging of the brain is called Alzheimer’s disease, but aging of the whole body is not considered a disease. It would be much more interesting for medical students to approach diseases from an aging viewpoint and to also deepen their understanding of diseases. Take, for example, heart disease: many MDs will say that it’s not really caused by aging, but brought about by white blood cells that accumulate in the blood vessel wall. However, if you look deeper, you see that that this happens because of typical aging processes, like lysosomal dysfunction in the white blood cells,  making them stack up oxidized cholesterol and accumulate in the blood vessel walls, telomere attrition of the endothelial cells in the blood vessel wall so the endothelial cells let through more white blood cells, senescence and epigenetic changes in the stem cells in the blood vessel walls, and so on. We urgently need to train MDs more about aging, the process that causes most of the diseases that they will treat on an everyday basis.

Why have many medical doctors not embraced the idea that aging can be addressed directly?

Sometimes, this is because they haven’t seen the recent studies that show that aging can be partially reversed, or it’s because they have not sufficiently been educated in aging and how it causes so many aging-related diseases. All too often, aging is considered as too complex a process to interfere with. However, you do not have to understand all the intricacies of aging to do something about it. For example, four simple Yamanaka factors, cyclically applied, can rejuvenate cells. We don’t have to exactly know all the specific aging pathways that are impacted by these Yamanaka factors. We just see that it works.

What can we do to encourage more medical doctors to consider ways of treating the aging processes directly in order to treat multiple age-related diseases at once?

We should explain more that aging causes most diseases that MDs deal with, such as heart disease, cancer and Alzheimer’s. We should explain more how aging originates and the fascinating new insights and studies regarding reversing aging. We should emphasize more that slowing down or reversing aging is the most powerful method to keep people healthier for a much longer time. We should avoid the term “anti-aging”, because this term is loaded with unproven therapies, but instead use the term biogerontology, which is the scientific field that studies aging.

Is the field of medicine prepared to address the silver tsunami, and how might biogerontology help with this problem?

Medicine is very ill-prepared to address the silver tsunami. Medicine is badly prepared to even do something about the many chronic diseases that impact people today. Medicine mainly focuses on treating diseases when it’s too late, such as when people already have had a heart attack or when people already have cancer. Too little emphasis goes onto preventing diseases through healthy nutrition and lifestyle in general. We know that, in many cases, type 2 diabetes and heart disease can be reversed. Unfortunately, this doesn’t happen for most people, because they get very ineffective, watered-down health and lifestyle advice in the best case and only medication in the worst case. It’s unfortunate to see how the root cause of their problems is not addressed. With the aging population, and the many aging-related and metabolic diseases that come along, this problem will only compound and could lead to the bankruptcy of health care systems and insurance industries of whole countries if we are not going to do something drastic about it. Giving people an app or a walking stick with a sensor is not going to really address the issue.

What approaches and technologies that may address the aging processes in the near future are you most optimistic about?

I am enthusiastic about modulation of the epigenome to really impact the aging process. Clearing senescent cells is also promising. It’s a low-hanging-fruit approach to aging, given that it is quite easy to clear senescent cells despite the fact that it would mainly extend median lifespan and not maximum lifespan. New technologies like CRISPR proteins, which allow scientists to much more accurately reprogram genes, and transcriptomic drugs, like mRNA, siRNA and microRNA, are also promising to tackle aging and aging-related diseases. I estimate that in the next 10 to 15 years, the first therapies to treat aging will come about.

What can we do now to slow aging down and stay healthy while we wait for these approaches and technologies to arrive?

The method we have to live longer now is our food. Next come exercise, psychology, sleep, relationships, and so on. However, only nutrition can impact maximum lifespan, while exercise can at best extend median lifespan. Eating lots of animal protein, especially red meat, sugar and starchy foods, and trans fats accelerates aging. We should eat less red meat, fried foods, sugary foods, potatoes, pasta, bread, and rice and should consume more vegetables, mushrooms, fruit, nuts, white meat, fish, green tea, and other nutrients that can slow down aging.

Do you currently engage in any practices to increase your longevity?

Of course. I eat healthy. Now and then, I fast. I try to get enough sleep and exercise. I do meditation and practice positive psychology to improve happiness. I take supplements, such as vitamin D, iodine, magnesium, B vitamins, nicotinamide riboside, and others. Some people go even a step further and take metformin or low-dose aspirin. However, healthy nutrition is more impactful on extending lifespan than supplements or drugs.

What is the biggest barrier to progress in the field?

One big barrier is lack of money. Many scientists cannot pursue great ideas or develop promising technologies because they don’t have enough funding. Biotech research is extremely expensive. Governments should provide much more funding to study and address aging, which is the main cause of the silver tsunami that could bankrupt them and that causes 86 percent of healthcare expenses. Many problems can be solved if enough money is thrown at them. Another important barrier is a lack of knowledge. If you are not aware of the recent progress made in the aging field, and of what new companies and start-ups are working on, you tend to think that tackling aging is impossibly difficult to do. However, you could be pleasantly surprised. Scientists and investors are now working on fixing aging; they consider death to be a technical problem that can and must be solved, and they look at the human body as a complex, walking and talking amalgam of 3D algorithms that can be reprogrammed into a more youthful state. Recent studies show that they might not be far off.

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

An Interview with Dr. Leonid Peshkin

Determined but not complacent, grounded but hopeful, Dr. Leonid Peshkin is one of the scientists working on understanding aging so that it may one day be treated like we treat any other ailment.

As he revealed in an interview with the Boston Globe in mid-2018, the idea of having to lose oneself and one’s loved ones to aging never made any sense to him, and ever since he was a child, he has been preoccupied with aging and the fear that it might take away his father, who was almost 60 when Leon was 10 and, sadly, passed away in July 2018 at the age of 96.

Dr. Peshkin, a 48-year-old from Moscow, Russia, possesses a master’s degree in applied mathematics and a Ph.D. in machine learning. He currently works at the Systems Biology Department at Harvard Medical School; his primary interests are embryology, evolution, and aging, which he has studied for over a decade.

In this interview with Lifespan.io, he makes no mystery of his wish for aging to be defeated as soon as possible, but unlike other scientists in the field, who tend to prefer specific theories of aging, he thinks that our understanding isn’t yet deep enough to take sides, and in this, he is somewhat reminiscent of João Pedro de Magalhães.

As he makes clear, Leon is not particularly fond of the overly optimistic attitude of some people in the longevity community and some singularitarians, as he finds that complacency may well hinder progress in a battle that, at this point, is all but won. He began as follows:

Thank you for the opportunity to share my views. I must mention that my Ph.D. dissertation was done in the field of artificial intelligence, and for the past 12 years, I have worked at the Systems Biology department at Harvard Medical School, yet I am a relative novice in the field of aging biology. As a way of introduction, I’d like to offer a caricature of the currently popular sensationalist view in the field of aging:

“We are the chosen generation. Singularity is near. Rejuvenation therapy is almost here. Not one, several a-la-carte: stem cells, factors from young blood, senolytics, Skulachev’s ions, NAD, etc. Companies backed up by luminaries from business and science are already sorting out the remaining details, helped by the formidable force of AI technology called ‘deep learning’.”

This fairy tale is beautiful, and deep in my heart, I hope I am mistaken, but I think that at the moment, this positive mysticism is not justified and is rather counterproductive. The excessive optimism is, unfortunately, standing in the way of progress, as I will try to explain.

There are many proposed models of aging, such as the Hallmarks of Aging, SENS, and the deleteriome. Which, if any, of these models do you believe reflects the reality of aging?

I would not want to take part in religious wars. People get very passionate and clash about often vaguely defined terms. Which of the observed hallmarks of aging, from the molecular to the organism levels, are correlates and which are causes of aging is hard to say. Biology has not yet matured to become an exact science. Perhaps owing to my training in quantitative science (M.Sc. in Applied Math, Ph.D. in Machine Learning) I take a “model” to mean a level of quantitative understanding that allows for “modeling”; that is, forecasting and answering “what if” questions. Such a model might not be ultimately expressed by a set of crisp human-readable mathematical formulae but rather a large set of tuned parameters in an artificial neural net or some other representation that has not yet been invented.

It must, however, provide a way to assess the current state of an organism and predict its lifespan and healthspan in a stable environment, outside of a major perturbation, and then go further to allow for perturbations and adjust the predictions. Today, I can’t even say that there is an agreement in the field of what is a useful definition of “aging”. I like “increase of hazard rate (i.e. the probability of dying) with time”, which is admittedly a very mathematical notion — precise and not terribly useful. Inverting this formula, we get a curious metaphor – a life without aging can be imagined as a life where, say, once a year, you undergo a treatment that rejuvenates you a year in biological age, or, with some small but non-negligible probability, kills you. Life is a game of chance.

Do you believe that aging is a one-way process or something that is flexible and amenable to intervention?

It is both. I think that it is a one-way process but also flexible and amenable to intervention. Imagine one dramatic intervention: one day, we invent a way to cryo-protect a warm-blooded organism like ours so that it can undergo a freeze-thaw cycle without damage. Now, you are faced with a challenge to design a schedule that determines when, and in what size fractions, you’d like to use up your lifespan. While you are frozen, time stops. While you are alive, you age: the “deleteriome” kicks in, ionizing radiation wrecks your DNA, your defrosted friends and family du jour stress you out, etc. That’s what things would look like ad absurdum, illustrating the tradeoffs. Now, back to the interventions: I imagine a process not unlike a beauty salon, in which you do your nails and hair and get an occasional facelift; all of these are tradeoffs, even if people do not recognize it. Beauty treatments make you look younger at the moment, but cosmetics products may poison your skin and accelerate actual aging.

There is evidence of such tradeoffs across organisms in nature; extending lifespan in many species can be accomplished at the expense of reproduction, and in cold-blooded organisms, you can multiply the lifespan several-fold by just cooling the environment down or slowing down metabolic processes in other ways. I believe that the first results will be not so much in giving people free tickets to longer lives but in making the tradeoffs more explicit, educating people and putting them in control of decision making. These are never easy choices, not unlike the one that a cancer patient faces when given a choice between quick and painless death or taking a chance at the expense of painful agony. Life is a game of chance.

What are some of the studies that have convinced you that this is the case?

I did not have to be convinced; it follows from well-known observations. There are closely related species with drastically different lifespans, the germ-line reset that we discuss below, the dependency of lifespan on diet, and other obvious environmental conditions, such as ambient temperature for cold-blooded animals. I was tangentially involved in the effort to obtain the complete genome of the naked mole rat, which lives ten times longer than closely related species.

I currently study aging in daphnia – a small aquatic organism that goes from infancy to frailty in one month. We all know about aging, but this familiarity becomes understanding as you closely watch one organism being created from a single cell, form organs, grow, mature, procreate by itself, then exist for a while in a perfect environment with the right temperature, water quality, nutrients and light, just to decay and fall dead, all in one month. This organism is not hard to relate to, as it has very recognizable parts of common anatomy with us and lots of recognizable cell types. Yes, it is an invertebrate, but the muscle, heart, blood, primitive immune system, gut and auxiliary digestive system, eyes, and other sensory organs made of sensory and control neurons are easily observed using an ordinary dissection microscope. We are looking for signs of aging that are similar to those known in people, such as formation of cataracts, changes in bone density, hardening of blood vessels, etc. With such a short lifespan, we are able to manipulate the conditions with a variety of perturbations in order to hopefully build a detailed understanding and a causal picture of aging.

Eventually, we will even build a “model” in a rigorous sense of the word. We and others already see that calorie intake has a strong effect on the lifespan of daphnia; yet, even here, we do not have a clear, causal picture. Our approach to teasing out causality builds upon machine learning and was developed and published with a focus on cancer and metastasis, specifically cell motility. Roughly, the idea is to use broad-specificity drugs (polypharmacology) with well-characterized molecular targets as “twenty questions” in order to systematically explore which pathways do and do not affect a phenotype, which is lifespan in our case. Unlike with a drug screen, we do not expect to find a magic pill out of thousands or hundreds of thousands of candidates; rather, we use dozens of specifically selected drugs and respective doses to get a complex system to reveal its modularity to us by targeting modules in a disjointed fashion.

What is the epigenome, and how does it relate to the genome?

I like to think of the genome as an encyclopedic-scale cookbook, whereas the epigenome is a collection of bookmarks and handwritten notes on the margins as well as sauce stains and syrup spills. The genome is often likened to a book, but I insist on having a functional aspect for this metaphor to really work. We must use that book to create a product according to a recipe. It adds a lot to the biological metaphor; branches of the same fast-food chain use the same pages out of this book in a consistent, perhaps ever-slightly different, way. These are akin to cells of the same type. Worn-out pages lead to omitted or incorrectly interpreted steps, et cetera. Trying to implement a recipe that is already somewhat damaged might lead to a cook spending more time with it; in turn, this leads to more damage to this and adjacent pages, a process that greatly amplifies initial small random differences in a chaotic way, leading to naively inexplicable dramatic differences in the lifespan of originally twin-identical systems in near-identical environments.

Apparently, the epigenome can be used to accurately “age” the specimen, that is, to tell how old an organism is from a sample. This will be very useful in aging research, as to this day, we know how to age only a few selected species that keep track of their age in a visible way, like the annual circles on a tree stump, the similar layers on an otolith, and the scales of some fish. Having access to an organism’s age allows us to correlate its age to other markers, such as gene and protein expression in various tissues, behavior changes and physical wear of structures.

It has been proposed that epigenetic alterations are a primary cause of aging and that both genetic and epigenetic interventions are keys to potentially controlling aging. Do you consider epigenetic alterations as a cause of aging or a downstream consequence?

Neither cause nor downstream. There is no linear causal chain with the two links of “aging” and “epigenetic alterations”; instead, there are loops and amplifiers in the circuits of aging.  Epigenetic alterations have to be caused by something else; these, in turn, control many things. On the other hand, DNA damage is clearly pretty early in the causal network but is hard to undo. There is more hope to proofread and fix “epigenetic alterations”. Going back to my fast-food chain metaphor, you would imagine a quality inspector, examining a cookbook, reducing the stains and shaking off crumbs and accidental bookmarks while completely shutting down the restaurants with hopelessly damaged cookbooks.

While I am not sure about the arrow of causality, I am very much interested in this direction of research, so much so that we are planning an experiment around it in daphnia, looking at changes in the distributions of cell types in cell populations that make up young and old individuals. The expectation is that epigenetic alterations lead to de-differentiation and mis-differentiation of cells in old organisms, which could be characterized and further used as end-points for aging interventions. We would need to find a reliable and affordable way to profile the epigenetic state at a single-cell level for tens of thousands of cells, which is a big challenge.

In December 2016, researcher Juan Carlos Izpisua Belmonte reset epigenetic aging markers in living animals using partial cellular reprogramming, similar to how we make induced pluripotent stem cells and how embryos “reset” their aging. This appeared to reverse multiple aspects of aging and increased healthy lifespan in mice. Are you optimistic that resetting the epigenetic biomarkers of aging in aged adults is a promising direction of research, and why?

I do think that it is a very promising direction of research, but I wouldn’t rush to inject myself with Yamanaka factors just yet. There is way too much hype surrounding research in the field of aging nowadays, which is simply harmful to the field. Biology in general is suffering from the reproducibility problem. Very few results translate from mice to humans. “Normal” laboratory mice are far from “wild type”; they are inbred and live in unhealthy conditions. It would be great to create a natural, healthy, predator-free environment for mice and many other species and get good lifespan statistics someday, both natural and under perturbations.

For now, we have very scarce data on “natural” aging of “natural” animals and have to rely on statistics for lab strains of animals that are cheaper to keep in large numbers, such as worms and flies. Even for these “unnatural” mice, I do not think that we yet have the lifespan data on “Yamanaka infusion”; instead, we saw encouraging results on some overall health markers in progeric mice. In order to rationalize the hypothetical rejuvenation reset, one has to imagine that Yamanaka factors work differently in different cell types, re-aligning each cell to its original profile. That sounds like too much to hope for as a solution to rejuvenation.

Speaking of resetting epigenetics and thus aging, there is already solid evidence that this happens every day when children are born. If aging was a one-way process, then our children would be born old, but they are not. There is a strong connection with aging and embryology, so why is it that an old oocyte cell grows into a young organism rather than an already aged one?

I love this “germ line reset” question. I see two and a half possibilities here:

1: The germline needs no reset. The germline is created early; very few cell cycles happen between generations. If some damage does occur, it is selected against at the cellular level or, later, at the organism level.

2: The germline does age, but

(a): it rejuvenates in an intense cleansing event somewhere during germ cell maturation and fertilization. The dream is to identify this mechanism and get it to work elsewhere. One of my projects has to do with precisely this magic, as it looks at the progesterone-induced maturation of frog oocytes. In this project, we use quantitative mass spectrometry to analyze the dynamics of protein abundances and phosphorylation events in order to look for the hints of a proverbial reset.

(b): age-related damage is not a problem. It is ignored and then gets diluted in the massive expansion from a single cell to an organism.

My own established line of research on protein expression in early embryogenesis made a strong case against this. I established that up to a point in vertebrate development, when hundreds of thousands of cells are there and many complex tissues have been germinated, the vast majority of protein molecules in use were deposited by the mother into the oocyte rather than synthesized de novo in the embryo. Simply keeping “bad” old protein until it gets diluted and not using it in development would be a tricky proposition, so the “reset” (abrupt degradation of the protein aggregates and carbonylated proteins) is a very reasonable theory.

The use of AI, specifically deep learning, in research has been in the news a great deal lately; how are you using it as part of your research?

I am not. Even though my Ph.D. is in AI, or precisely because it is, I am not of a mind to “unleash AI onto the problem and let it figure out aging”. There are some encouraging methods being developed, such as “deep learning”, but these are just that – tools, technologies that are only powerful when correctly applied in specific circumstances. Yes, they are useful for board games, image analysis, and self driving cars, but not yet biology nor aging.

Some of the aging-related projects that I am involved with produce image data and therefore might use machine learning someday. One example is developing “healthspan assays” for the short-lived organism daphnia, which involves collecting a lot of image data. We would like to develop a standardized platform for testing many perturbations’ effects on lifespan and healthspan. These could be drugs, changes to the environment such as light cycle and ambient temperature, diet, etc. Even though there are related initiatives, I am surprised that such a platform has not been developed yet; it’s badly needed and could be crowdsourced. Some machine learning methods will be useful there.

Also, I already mentioned one example of where machine learning tools are used in my work: a method we developed called “KIR – Kinome Regularization”, which uses drugs as perturbations and regularized regression as a way to uncover which pathways are responsible for a particular phenotype.

I am a lead co-author of a, fair to say, very highly cited paper that uses AI in biology. The paper is about classifying variants in protein-coding genes as deleterious or neutral. It’s definitely the most known and used method in this field and is licensed by many groups in academia and industry. The approach in that pipeline is the most basic AI method, which has been a workhorse of AI for decades: Naive Bayes. What’s important is that it is robust to outliers and that the results are understandable and interpretable by human experts.  Every step is important for machine learning to work: the quality of the data, correct annotation of the training set, informative representation of the features relevant to that particular domain, and a good algorithm. Machine learning has gone a long way towards producing methods that are robust to noise in feature discovery and selection, but if you start with poor, inconsistent, and mislabeled data, it’s hard to accomplish anything. Biology is indeed at the point of having produced Big Data, which generates a lot of excitement for applying machine learning to it; the issue is that unfortunately a substantial component of low-quality infusion turns the whole treasure trove into Big Bad Data.

AI in aging research is still in its early stages; how do you see it developing in the next decade, and what needs to happen for it to become an optimal tool in the fight against aging and age-related diseases?

In order for AI to start being applicable to aging and biology in general, the data standards in biology must catch up to the data standards and quality of traditional AI domains. So far, AI, whether deep or other, has impressively fit within a very constrained and homogeneous setting, such as board games. The number and types of objects are very well defined, the data is curated, all situations are mostly at the same scale, and so forth. Even in voice recognition, self-driving cars, and robotics, the environments are extremely consistent and curated. Biology is very different in this respect; or, rather, our perception of biology at this point is different. We have not found invariant ways to describe phenomena, so any situation is unique.

Let’s look at one simple question: lifespan. For most species, we only have anecdotal, unreliable data. We would want to know how long species live in a setting where there is no predation and death comes “of old age”. It only happens at the zoo, and zoo data is a pretty good source, but making these records consistent across various countries is tricky. It’s shocking perhaps that even the answer to the question of how long people live has a lot of room for improvement. What’s the maximum registered lifespan? Jeanne Calment’s famous record of 122 years has recently come into question. What is the median? This depends on the country and many other things. The American life expectancy has been slightly falling in recent years. Is at least the overall distribution a Gompertz distribution? It’s not clear. There is a discussion of a flattening of the hazard rate in old age; this is seen in other species such as flies, but is there no flattening in humans?

I recently lost my father to old age; he passed away one week short of 97 after many months at hospitals and rehabs. I personally witnessed what I’d call an “assisted homicide” phenomenon – there is strong pressure not to provide elderly patients with the level of medical care that is standard for younger patients with the same conditions. Doctors avoid procedures when their perceived risk is high, as it reflects negatively on the practice (spoils statistics); they like to call it “useless care”. Social workers push relatives to “pull the plug” to “avoid needless suffering”, exaggerating risks and downplaying chances of recovery; nurses and unskilled helpers quietly withdraw medical care and assistance, projecting their own religious beliefs of the afterlife and other superstitions onto what’s good for the patient. Such norms are likely country-specific and naturally skew lifespan statistics; we ignore this component of mortality risk in old age, which has to do with the medical system’s bias against elderly patients.

Another example involves data on the perturbations of drugs on lifespan. There is vast literature and databases that attempt to agglomerate the results. However, not all the parameters are reflected, and often you find the same drug and dose having different effects in the same species. Drugs themselves are surprisingly hard to study because there is no consistent information widely available on which drug is called what or has which chemical structure and CAS identifier; there are rampant inconsistencies and misannotations. Getting the “same drug” from two different vendors leads to different results because drugs have different purity, etc. Overall, the vision that we can fund large optimized data factories for producing Big Data in biology and then unleash AI on it to discover how biology works has not paid off, probably for very fundamental reasons.

We do not know where to look for invariants in biology, so measuring e.g. gene and protein expression in a cancer cell line and assuming that your measurement captures the “normal” state and reaction to perturbation by drugs is naive; so many factors influence what things look like, and time of day, cell density, and microenvironment are just a few, so tomorrow, you can remeasure and get very different results. So, getting the results of multiple studies from these giant data repositories and hoping to find things in common could be very disappointing.

I mentioned that I was involved with a project of sequencing the naked mole rat, helping the Gladyshev laboratory at Harvard. The idea was to look for genes explaining its exceptional longevity and health. Such analysis has to be done by comparison to other species, so, naturally, it relies on having accurate and complete information on several other genomes. However, the quality of most so-called published complete genomes is such that you can never be sure if a certain gene is missing because it is really not there in these species or because it was missed by the genome sequencing and annotation pipeline.

One of my projects has to do with the quality control and attributes of complete genomes, so such analyses can be informed by the overall quality of the genome of each species, and gene sequences that are likely incomplete, bad, or represent the fusion of several real genes are labeled as such. I do not mean to downplay the work of the other teams that produced the complete genomes; there are hard challenges that are organism-specific, and we should be grateful to have as much information as we do, yet an objective way to assign confidence and take it into account has been missing.

You can probably see that the data quality in biology is a personal crusade for me.

AI is part of broader advances in research; automated microfluidics, high-throughput assays, and other automation are also improving research quality and speed. What other ways do you see automation in the lab helping to speed up research?

I am fortunate to have been involved with method development and to teach people about some of the cutting-edge technologies for high-throughput measurements, such as quantitative mass-spec proteomics and single cell transcriptomics. These are very promising technologies, but here, again, one has to be very careful with the interpretation of the data and particularly careful when expecting to merge data from multiple, disjointed studies. In single-cell studies, everything depends on which tissue you work with, how successfully you managed to rapidly dissociate the sample into single cells without killing a lot of cells, particularly in some biased way that masks some cell types entirely, whether (or rather to what extent) cells lyse in the device, and with what parameters you run the device and make and sequence libraries.

The same goes for proteomics: it’s easy to miss whole classes of proteins, such as low-abundance transcription factors or lipid-soluble proteins.  In both cases, splitting the same exact sample in half and running it through a pipeline would often give you dramatically different outcomes. So while there is indeed some degree of automation, and automation is helpful, it is only practical to a very small extent, and even where it is used, it has to be used with extreme care.

I already mentioned that the data quality in biology is a personal crusade for me. I myself work on integrating AI into these methods. One project which is just now being submitted for publication is about assigning confidence to quantitative mass spectrometry measurements of relative protein abundance. Comparing protein expression across samples is currently done by reporting the most likely value, which does not allow us to notice significant but small changes or to rank candidate genes for follow-up research in a statistically sound way. With colleagues from Princeton, we developed a rigorous statistical model [15], which allows us to confidently judge a shift of protein expression down to 1%. This is an application of a Bayesian statistical approach borrowed from machine learning, and it will empower many new studies and allow us to re-analyze already published data to get to new findings. We will immediately plug this into our aging project, which involves life-long profiling of protein levels in daphnia.

Progress in aging research seems to have been speeding up in the last few years; however, there are still a number of bottlenecks holding things back. As a researcher, what is the biggest bottleneck holding us back from making rapid progress in the field?

While I, very humanly, would love to believe that we are the fortunate generation to witness and benefit from the awesome moment of the maturation of science and technology known as the “singularity”, if I try to soberly consider this “speed up”, I find it hard to believe. What I mean is that I am not sure if our time will seem like the singularity in retrospect. I also suspect that 10, 100 and 1000 years ago, there were people who lived through the same breathtaking experience of an imminent “singularity”; new tools tend to give us exaggerated sense of control over nature.

What is holding us back falls into two broad categories: things that are not in our power to change and things that are. The current system of incentives in science, funding and publication puts a lot of pressure on scientists to compete, not to share their best data and code, and to obfuscate even when they do. Competition can be extremely useful at the stages when cures need to be brought to the market, but the current state of aging research badly needs cooperation. We need platforms for the uniform profiling of perturbations. We need consistent, clean information on drugs and lifespan in species.

Perhaps most importantly, we need a platform for crowdsourcing efforts and doing citizen science. There is enormous enthusiasm, which largely is wasted in social media groups that discuss food supplements and cheer. I know many bright, generous, and resourceful non-specialists who would be happy to contribute their time in curating data, coding up useful snippets of software, and following instructions to collect samples. Organizing this force of nature will take a Task Rabbit or Mechanical Turk kind of platform. Also, it amazes me that, to this day, in the United States, someone can not get their own blood work done “out of curiosity”. People are already getting armed with technology to run some tests at home. If we trust people to spit into a tube to get their DNA sequenced, we can surely develop instructions to run what would essentially amount to a “citizen-run clinical trial” in which interventions are consistently tested.

All in all, the distinction between an atmosphere of competition or cooperation in the field really depends on the perception of whether the field is very young and far from having mature results and “products” or is very close to getting the first drugs and therapies to the market. That’s mainly why the false sense of accomplishment is very harmful to progress. The field is in its infancy, and it’s way too early to worry about marketing, patents and profits and to build walls; we have to find ways to be very open and collaborative.

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

Fisetin May be a Low-Hanging Fruit for Aging

There has been considerable interest in fisetin recently, especially for its potential as a senolytic, which clears away dysfunctional senescent cells that accumulate with aging. Researchers believe that fisetin may be useful in increasing the healthy period of life known as healthspan.

What is Fisetin?

Fisetin is a naturally occurring flavonol and part of the flavonoid family of polyphenols. Fisetin also acts as a pigment and influences the color of various fruits and vegetables. It can be found in many common fruits and vegetables, although the amounts greatly vary.

Fruit/VegetableAmount µg/g
Strawberry160
Apple26.9
Persimmon10.6
Lotus Root5.8
Onion4.8
Grape3.9
Kiwi2.0
Peach0.6
Cucumber0.1

As you can see, strawberries have a much higher concentration of this flavonol than other fruits and vegetables which may be why people associate it with this fruit in particular. At 160 µg/g, a pound of freeze dried strawberries would yield 72 mg of fisetin which is a considerable amount of fruit plus you would need to seriously like strawberries to eat that much every day! Typical supplement pills are in the 100 mg range, though without proper clinical trials there is no way to know if this dose is beneficial or harmful in humans.

Senescent cells and senolytics

As we grow older, we accumulate more and more damaged cells which cease dividing and enter a state known as senescence. Once they enter this state, they begin to secrete a cocktail of inflammatory signals known as cytokines, which summon the immune system to remove them.

When we are young, this system works perfectly, and the damaged cells are promptly removed; however, as we grow older, our immune systems slow down and senescent cell clearance begins to falter. Eventually, clearance slows down to the point that senescent cells begin to build up in the tissues contributing to the chronic inflammation known as inflammaging, harming nearby healthy cells and making them also senescent, preventing tissue repair, and facilitating the development of various age-related diseases.

It was proposed that finding ways to help the body clear away senescent cells may be the key to delaying or preventing various age-related diseases, and indeed in 2011, that idea was put to the test and seemed to bear fruit. Jan van Deursen and his team at Mayo Clinic engineered a mouse that was able to destroy its senescent cells when given a certain chemical [1].

This photograph shows this study’s impressive results. These two mice are littermates; the only difference is that the mouse on the left has aged normally while the mouse on the right has had its senescent cells removed and appears considerably more healthy.

A follow-up study in 2016 using regular mice again produced similar results, which confirmed the validity of removing senescent cells as an approach to maintaining health during aging [2]. This is when compounds that could remove senescent cells were dubbed senolytics, and they have since become an area of intense research.

Fisetin appears to be a senolytic

A number of plant flavonoids, such as quercetin, apigenin, luteolin, and curcumin, have been examined recently for their senolytic activity, and fisetin is the latest of these to enter the spotlight.

University of Minnesota Medical School faculty member Paul D. Robbins and Laura J. Niedernhofer and Mayo Clinic investigators James L. Kirkland and Tamara Tchkonia recently published “Fisetin is a senotherapeutic that extends health and lifespan” in the journal EBioMedicine [3]. During their study, they discovered that fisetin was able to remove senescent cells from aged mice, which improved their health and lifespan.

This flavonoid is a natural compound present in many fruits and vegetables such as apples, persimmon, grapes, onions, cucumbers, and strawberries, suggesting that it is imminently translatable. Importantly, no adverse effects of fisetin have been reported, even when given at high doses. Thus, our results suggest that supplementation or even intermittent treatment with this safe, natural product could improve healthy aging, even in elderly individuals.

Given that we have known about fisetin for some time and there have been various studies showing that it has anti-inflammatory and neuroprotective properties in animals, you might be wondering why are we only just now hearing so many things about it.

Unfortunately, despite having an excellent safety profile, there is a lack of long-term human data for fisetin. Simply put, there are a number of questions that are still to be answered, such as the correct dosage and frequency, and, until now, how it worked was not properly understood.

However, the researchers of this study have confirmed that fisetin is targeting and destroying senescent cells rather than simply blocking their signals or altering them in some beneficial way but not removing them. They were able to confirm senescent cell destruction was happening by using mass cytometry, or CyTOF, a technology pioneered by the University of Minnesota. This allowed them to go into detail, demonstrating the effects of fisetin on specific subsets of senescent cells in specific tissues.

The researchers also compared fisetin against other compounds, including resveratrol, luteolin, rutin, epigallocatechin gallate, curcumin, pirfenidone, myricetin, apigenin, and catechin. This study showed that fisetin was the most effective of these compounds.

Further studies in support of fisetin

The lack of long-term data for human use is currently a problem; however, we can gain some insight from the various animal studies that have been conducted with fisetin.

One of the common mechanisms by which it appears to work is via the reduction of inflammation, which could either be via removing senescent cells and their inflammatory secretions or by mediating immune response and blocking inappropriate inflammatory signaling. While not all of these studies directly relate to aging, many are linked to inflammation, which is critical to aging, so the data could be valuable in this respect.

In another recent study, a team of researchers at the Salk Institute tested fisetin, curcumin, and three variants of these compounds and found that they reduced various biomarkers of aging, increased the median lifespan of mice and flies, and reduced the signs of dementia [4]. The modified versions of fisetin and curcumin were shown to work, but so were the naturally occurring ones.

There have been other studies which found that fisetin has a potent effect on inflammation via blocking the activity of lipoxygenases, thus reducing the level of pro-inflammatory factors [5-6].

Another study demonstrated that treatment with fisetin was able to suppress mast cell activity induced by interaction with activated T cell membranes [7]. Interactions between populations of mast cells and activated T cells are implicated in allergic and non-allergic inflammatory disorders. Fisetin was able to prevent the activation of human mast cells and the resulting inflammatory response.

Fisetin also appears to have a potential application in combating high blood sugar in diabetics, which can cause inflammation of blood vessels and tissue damage. Researchers found that fisetin was able to block this inflammatory response in human cell lines and in mice [8].

Researchers in a 2003 study showed that fisetin reduced allergic reactions in mice when they examined it and a number of other related flavonoids, including astragalin, fisetin, kaempferol, myricetin, quercetin, and rutin [9]. In the study, the related flavonoids kaempferol and quercetin also showed substantial inhibitory activities in cytokine expression; however, fisetin was significantly more potent in preventing inflammatory cytokine production by basophils, which are a type of bone marrow-derived circulating leukocyte.

A 2014 study found that fisetin was an effective treatment for eczema in mice, as it was able to reduce the presence of immune cells such as T-cells, mast cells, and eosinophils, which are commonly encountered in the skin lesions that eczema causes [10].

There are too many studies to comment on here in this short review, but here are a few more that may be of interest:

ConditionPMIDReference
DiabetesPMID: 21738623[11]
DiabetesPMID: 24939606[12]
DiabetesPMID: 23791753[13]
DiabetesPMID: 21816145[14]
DiabetesPMID: 25064342[15]
HypertensionPMID: 26741654[16]
HypertensionPMID: 26759702[17]
ObesityPMID: 23517912[18]

Conclusion

Fisetin is a cheap and readily available supplement and has potential, but the various anecdotal reports from the biohacking community while interesting lack the consistency and scale of clinical trials. In order to properly confirm any benefits against aging and age-related diseases that fisetin may or may not confer, things need to move into larger scale human studies. We suggest people at home wait for the results of clinical trials and for the correct dosage and frequency to be determined before considering taking fisetin.

Thankfully these initial results have already prompted the launch of a clinical trial at the Mayo Clinic to evaluate the benefits of periodic fisetin treatment on certain aspects of aging, such as frailty and inflammation. This is good news because fisetin could potentially be low-hanging fruit for aging and age-related disease. We look forward to seeing the results published in the near future once the researchers have concluded their clinical study.

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

Literature

[1] Baker, D. J., Wijshake, T., Tchkonia, T., LeBrasseur, N. K., Childs, B. G., Van De Sluis, B., … & Van Deursen, J. M. (2011). Clearance of p16 Ink4a-positive senescent cells delays ageing-associated disorders. Nature, 479(7372), 232.

[2] Baker, D. J., Childs, B. G., Durik, M., Wijers, M. E., Sieben, C. J., Zhong, J., … & Khazaie, K. (2016). Naturally occurring p16 Ink4a-positive cells shorten healthy lifespan. Nature, 530(7589), 184

[3] Yousefzadeh, M. J., Zhu, Y., McGowan, S. J., Angelini, L., Fuhrmann-Stroissnigg, H., Xu, M., … & McGuckian, C. (2018). Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine, 36, 18-28.

[4] Schubert, D., Currais, A., Goldberg, J., Finley, K., Petrascheck, M., & Maher, P. (2018). Geroneuroprotectors: Effective Geroprotectors for the Brain. Trends in Pharmacological Sciences.

[5] Sadik, C. D., Sies, H., & Schewe, T. (2003). Inhibition of 15-lipoxygenases by flavonoids: structure–activity relations and mode of action. Biochemical pharmacology, 65(5), 773-781.

[6] Maher, P. (2015). Fisetin Acts on Multiple Pathways to Reduce the Impact of Age and Disease on CNS Function. Frontiers in bioscience (Scholar edition), 7, 58.

[7] Nagai, K., Takahashi, Y., Mikami, I., Fukusima, T., Oike, H., & Kobori, M. (2009). The hydroxyflavone, fisetin, suppresses mast cell activation induced by interaction with activated T cell membranes. British journal of pharmacology, 158(3), 907-919.

[8] Kwak, S., Ku, S. K., & Bae, J. S. (2014). Fisetin inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflammation Research, 63(9), 779-787.

[9] Higa, S., Hirano, T., Kotani, M., Matsumoto, M., Fujita, A., Suemura, M., … & Tanaka, T. (2003). Fisetin, a flavonol, inhibits TH2-type cytokine production by activated human basophils. Journal of Allergy and Clinical Immunology, 111(6), 1299-1306.

[10] Kim, G. D., Lee, S. E., Park, Y. S., Shin, D. H., Park, G. G., & Park, C. S. (2014). Immunosuppressive effects of fisetin against dinitrofluorobenzene-induced atopic dermatitis-like symptoms in NC/Nga mice. Food and Chemical Toxicology, 66, 341-349.

[11] Maher, P., Dargusch, R., Ehren, J. L., Okada, S., Sharma, K., & Schubert, D. (2011). Fisetin lowers methylglyoxal dependent protein glycation and limits the complications of diabetes. PLoS One, 6(6), e21226.

[12] Prasath, G. S., & Subramanian, S. P. (2014). Antihyperlipidemic Effect of Fisetin, a Bioflavonoid of Strawberries, Studied in Streptozotocin‐Induced Diabetic Rats. Journal of biochemical and molecular toxicology, 28(10), 442-449.

[13] Prasath, G. S., & Subramanian, S. P. (2013). Fisetin, a tetra hydroxy flavone recuperates antioxidant status and protects hepatocellular ultrastructure from hyperglycemia mediated oxidative stress in streptozotocin induced experimental diabetes in rats. Food and chemical toxicology, 59, 249-255.

[14] Modulatory effects of fisetin, a bioflavonoid, on hyperglycemia by attenuating the key enzymes of carbohydrate metabolism in hepatic and renal tissues in streptozotocin-induced diabetic rats.

[15] Prasath, G. S., & Subramanian, S. P. (2013). Fisetin, a tetra hydroxy flavone recuperates antioxidant status and protects hepatocellular ultrastructure from hyperglycemia mediated oxidative stress in streptozotocin induced experimental diabetes in rats. Food and chemical toxicology, 59, 249-255.

[16] Im, W. K., Park, H. J., Lee, K. S., Lee, J. H., Kim, Y. D., Kim, K. H., … & Jeon, S. H. (2016). Fisetin-Rich Extracts of Rhus verniciflua Stokes Improve Blood Flow Rates in Mice Fed Both Normal and High-Fat Diets. Journal of medicinal food, 19(2), 120-126.

[17] Je, H. D., Sohn, U. D., & La, H. O. (2016). Endothelium-Independent Effect of Fisetin on the Agonist-Induced Regulation of Vascular Contractility. Biomolecules & therapeutics, 24(1), 57.

[18] Jung, C. H., Kim, H., Ahn, J., Jeon, T. I., Lee, D. H., & Ha, T. Y. (2013). Fisetin regulates obesity by targeting mTORC1 signaling. The Journal of nutritional biochemistry, 24(8), 1547-1554.

LEAF Festive Fireside Chat 2018

Join us for the LEAF Festive Fireside chat, where we talk about this year’s coolest science, our favorite moments, and our future goals. We can only do what we do thanks to the support of the community and our monthly patrons – The Lifespan Heroes. Please consider supporting us by joining the Heroes and help us along with the drive to end age-related diseases.
We would like to ask you a small favor. We are a non-profit foundation, and unlike some other organizations, we have no shareholders and no products to sell you. All our news and educational content is free for everyone to read, but it does mean that we rely on the help of people like you. Every contribution, no matter if it’s big or small, supports independent journalism and sustains our future.

A Biohacker’s Letter to Santa

Dear Santa, My name is Elena Milova, and I am from Moscow, Russia. I am a science popularizer, biohacker, and public health advocate in the field of aging and longevity. I am 39, single, and without children, but if you think that I am reaching out to you to ask for a CRISPR-designed baby, I am not. I believe that this type of wish is rather in the pile of letters from China.

I am not asking you for a particular health improvement, as one could not wish for a better HOMA-IR (mine is 0.40, because I greatly reduced fast carbs) or total cholesterol level (below 4 mmol/L). I am fine without a new smartphone, too.

The thing that I am going to ask you for is much more tricky to get. I want everyone on Earth to realize that biological aging is amenable to medical intervention and that treatments targeting various mechanisms of aging are already in human clinical trials. 7.6 billion minds, one idea. That is my only wish.

Why this is so important to me

You have probably noticed yourself that your clientele is changing over time. There are more and more people over 60 in the world, and I assume that the number of wishes for recovery from this or that age-related disease are spiking higher every year. This must be a problem for you, as for many diseases of old age, there is still no effective treatment that would actually help to cure people. It must be frustrating to not be able to fulfill a sincere wish of a good person, especially when a child asks for her grandparents to recover so that they can walk and throw snowballs together.

By 2050, the elderly will be a quarter of the global population, and these people will likely be suffering from several chronic diseases at once, gradually losing their health, independence and dignity. For so many people, being a burden on their families because of their deteriorating health is unacceptable, which is why the number of suicides in this age group is so high.

Is aging an invisible problem?

The numbers of these voluntary deaths are very upsetting, but what is even more upsetting is that diseases of old age are the major cause of death worldwide and aging kills around 100,000 people every day. This is the population of a small city. Imagine what would happen if everyone in a city like Cambridge, Massachusetts were to die in one day. I bet that there would be a lot of media attention and that thousands of experts would be on television discussing the potential causes of death and ways of preventing this tragedy in the future. Let’s say that the next day, another city becomes deadly peaceful. Take the Russian city of Domodedovo, which has its own airport. Everyone dead. People in neighboring cities would probably be frightened, and some charismatic politicians would be trying to calm down the public and promising to do something about all these deaths. The next day, this happens to yet another city, maybe in India. Then another one in Australia. It would not take long before G20 would set up an urgent conference call to set up an international commission and allocate money and scientists to investigate and solve the problem.

Guess what? This type of thing never happens in relation to aging, because people dying from it are spread around the globe, so the disaster does not make the headlines. The public only notices the problem when an actor, scientist, or other significant public figure dies from an age-related disease – most often heart disease, stroke, or cancer. Do you want an example? “Santa Claus, age 90, dies from a heart attack: a critical blow to the industry of giftmaking.” Sorry, sorry. I didn’t mean to scare you, but you get the point, right? From looking at your pictures, I could suspect that you might have some minor problems with glucose metabolism, but your extensive physical activity during gift delivery should be compensating for that, so you should be fine. For other people aged 60 and older, aging is an ever-increasing problem. Here, we come to the other important issue.

What is aging? How it can be addressed?

You see, aging is the accumulation of damage that happens due to normal bodily functions. This damage builds up over time, normal cell functions erode, and, at some point, this leads to the manifestation of age-related diseases. Normal operations, damage accumulation, disease, more damage, aggravation of disease, death. Simple.

It turns out that at the beginning of this century, British scientist Aubrey de Grey published an article in which he described several types of damage done by aging. He suggested the heretical idea of targeting these damages with medical interventions instead of trying to cure the symptoms of each age-related disease. He argued that age-related diseases are only a consequence of damage accumulation and that it would be much more effective to address the root causes.

The seed that Dr. de Grey dropped into the fertile soil of scholarship produced nice fruit in 2013, which is when a group of famous researchers of aging published The Hallmarks of Aging, a paper in which they described nine types of damage that accumulate with age and could be made into new therapeutic targets.

There were other fruits as well: animal studies have definitively proven that even only addressing one type of damage can extend the healthy period of life, postpone age-related diseases, keep animals more active, and, as a positive side effect, extend lifespan. It is worms that hold the best record so far, as tweaking some of their longevity-related genes has allowed them to live 10 times longer. The results in mice are also impressive – the researchers can extend both their healthy period of life and lifespan by 30-35%. Honestly, I find myself jealous of these mice, sometimes. I would not mind adding another 30% of youthful and healthy years to my life, even if I would have to take some pills or get some regular injections.

Can we control aging in humans?

You see, Santa, where I am going with this. I am sure that you sometimes leave gifts under the trees of people who work for the FDA. Accumulating a critical mass of knowledge about interventions against murine aging made it possible to develop the same type of interventions for people. Now, drugs and therapies addressing some of the root mechanisms of aging are in official human clinical trials. At some point, some of these trials will be successful, and drugs and therapies targeting aging itself will come to market. If you don’t believe me, here is the short list of people whose chimneys’ stacks are the best source of additional information on the topic: George Church, Anthony Atala, Judy Campisi, Vadim Gladyshev, Maria Blasco, Michael West, Vera Gorbunova, Irina Conboy, Kelsey Moody, Brian Kennedy, Linda Partridge, Alexey Moskalev, Cynthia Kenyon, Claudio Franceschi, Alex Zhavoronkov, Nir Barzilai, and, of course, Aubrey de Grey. He wears a great beard, so you have more in common with gerontologists than you would think.

Listen to these people tell their families about their research, and you will get my point. We are on the edge of a revolution in rejuvenation biotechnology. Yet, most people don’t know about it and don’t realize what kind of potential benefit this advancement holds for them and for our aging society as a whole. Most importantly, as they know nothing, they have no say in decision making. How can people possibly speed up the pace of aging research if they don’t realize that aging is amenable to intervention? How can they foster technology transfer and local production of the cures for aging, such as senolytics, in their countries? How can they control prices and make future distribution and access equal? How can they ensure that old people in their families, who need these new treatments the most, would get them sooner? Knowledge is power. We hear this in almost every interview, and you should be hearing it every Christmas from the researchers of aging, too. They have golden brains; the only thing they need is an appropriate amount of funding to solve the problem of aging more quickly. A strong public movement for aging research could be a game changer and could act as leverage to allocate government funding towards researching and developing treatments that target the underlying mechanisms of aging.

vEnding aging and age-related diseases is possible

It is obvious that you are a kind person, Santa. You are perceptive and generous; you know what people want, and you try to give them what they want. However, if you don’t help me with my information campaign, in a couple of decades from now, you will be delivering billions of adult diapers and wheelchairs all over the globe. Wouldn’t it be nicer if you were to pile these up in your warehouse to be covered in dust while you give people therapies and drugs that prevent aging and wipe age-related diseases out of human lives? Just imagine how much happier people would be if they could remain healthy and independent, enjoy full and productive lives, achieve more, and stay with their families and friends for longer.

I was a good girl the whole year, attending scientific conferences, interviewing researchers, speaking at public events, and supporting our partners and colleagues in every way I could, even if that much socializing makes me suffer from an introvert’s hangover. I was eating healthy food and promoting evidence-based means to slow down aging among my relatives and friends. I deserve a nice Christmas gift. All you have to do is to let everyone on the planet know that aging is amenable to intervention and that treatments addressing the root causes of aging are currently being created. For real. That would make me the happiest creature on the planet. Thank you in advance!

Sincerely, Elena

Instead of a conclusion

I am 39 years old, and I am an agnostic. There is not much evidence that Santa Claus exists. However, I do believe that miracles happen: the miracles that we create with our own hands. You who are reading these words (thanks for getting this far, by the way!) possess this special power, too. Use it! Let people around you know that science is close to bringing aging under medical control, and let’s build a world where healthy longevity for everyone is a reality.

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

An Interview With Daniel Muñoz-Espín

During the Fourth Eurosymposium on Healthy Ageing (EHA), which was held in Brussels, Belgium last November, we had the opportunity to meet Dr. Daniel Muñoz-Espín from the Oncology Department of the University of Cambridge.

Dr. Muñoz-Espín received his PhD from the Autonomous University of Madrid, Spain, within the viral DNA replication group at the Centre of Molecular Biology Severo Ochoa, where he worked under the supervision of one of the most famous Spanish scientists, Dr. Margarita Salas. Dr. Muñoz-Espín’s postdoctoral research resulted in several published papers and a 2013 patent focused on DNA replication; he then joined the Centro Nacional de Investigaciones Oncológicas, or CNIO, the Spanish National Centre for Cancer Research, specifically the team of Dr. Manuel Serrano, co-author of The Hallmarks of Aging.

The research that Dr. Muñoz-Espín conducted during this time demonstrated how cellular senescence doesn’t play a role just in aging and cancer but also in normal embryonic development, where it contributes to the shaping of our bodies—a process that was termed “developmentally-programmed senescence”, whose concept was very favorably received by the scientific community.

Currently, Dr. Muñoz-Espín serves as Principal Investigator of the Cancer Early Detection Programme at the Department of Oncology of Cambridge University; with his current team, Dr. Muñoz-Espín developed a novel method to target senescent cells, which was reported in EMBO Molecular Medicine. This topic was the subject of Dr. Muñoz-Espín’s talk at EHA2018 and one of the many fascinating others that he discussed in this interview.

There are many proposed animal models of aging; do you have a particular favorite that you believe is close to the reality of what aging is?

In the particular case of murine models, I definitely think that the direct use of naturally-aged mice is by far the most accurate approach. Of course, keeping and maintaining mice alive for 1.5-3 years is very expensive and time-consuming, and this possibility is not available for many laboratories. Because the process of ageing is very complex, the use of naturally-aged mice entails multiple pathological manifestations and represents more rigorously the genetic variability of humans, although it makes some studies unachievable because of the number of mice that would be required for delivering solid conclusions.

Genetically engineered mouse models of accelerated ageing or progeria are useful to recapitulate some characteristics of normal ageing, such as osteoporosis, osteoarthritis, intervertebral disc degeneration, fat loss, sarcopenia, alopecia, cataracts, etc., but they also present other features not seen in the elderly. Depending on their physiological alterations, some models of accelerated ageing are more suitable than others for a particular investigation.

Why did you choose cellular senescence as one of your main research focuses?

In 2011, I joined the laboratory of Dr. Manuel Serrano at the Spanish National Cancer Research Centre as a postdoc, where he proposed that I work in the field of cellular senescence. Historically, senescence was a process related to ageing and cancer, and numerous studies at that time began to associate senescence with multiple age-related pathologies, including cardiovascular diseases, fibrosis, sarcopenia, obesity, osteoarthritis, type 2 diabetes and neurological disorders. This is the reason that senescent cells are commonly known as “zombie” cells. I was immediately fascinated by such an interesting cellular process.

Currently, there is a huge excitement in the fields of biomedicine and gerontology because of the recent realisation that, beyond a circumstantial association with these pathologies, senescence can play a causative role. In fact, in the last few years, it has been demonstrated that the eradication of accumulated senescent cells in mouse models ameliorates and even reverts the pathological manifestations and, importantly, substantially extends the lifespan of naturally aged mice.

We’ve learned that the growing burden of senescent cells is likely to be a key driver of age-related pathologies; however, the work you published in 2013 showed that cell senescence also plays a role during the course of normal embryonic development [1]. Can you tell us more about this?

I think the word “senescence” is quite unfortunate to describe this process, because we tend to think immediately in ageing. The main role of cellular senescence is, however, to remove unwanted cells from our bodies. This is a mechanism of defense against multiple types of stress, such as oncogenic stress. When a cell is damaged or stressed often implements the senescent programme, which implies a permanent cell cycle arrest to prevent the multiplication of cells that are damaged, and also the secretion of a complex cocktail of proteins and inflammatory factors aimed at instructing nearby cells and recruiting the immune system to eliminate senescent (dysfunctional) cells. Therefore, clearance of senescent cells facilitates, in some contexts, tissue regeneration (an example is wound healing), and it is the basis for tumour suppression.

Our groundbreaking discovery was to find that cellular senescence is a programmed process that occurs during normal embryonic development, and it plays a fundamental role in tissue remodelling and morphogenesis. In this case, cellular senescence is not triggered by stress or damage but by developmental programmes, and it is intimately coordinated with other processes, such as programmed cell death, in order to “sculpture” our tissues and organs. The detrimental roles of senescence occur when these cells accumulate in tissues and are not eliminated by the immune system. This happens when there is persistent damage or stress, in chronic disorders, and also during ageing. In these scenarios, the process is not efficiently resolved and senescent cells are not cleared, and because they are dysfunctional cells and persistently secrete proinflammatory factors, they can accelerate ageing and contribute to disease.

In summary, depending on the context, both pro-senescent and anti-senescent therapies can be beneficial.

At EHA2018, you presented a drug delivery system that was developed by your research team to target senescent cells. In a process called gal‐encapsulation, you encapsulated drugs using galacto‐oligosaccharides, which are released into cells after digestion with lysosomal β‐galactosidase, which happens more readily in senescent cells. Can you explain how the system works for our readers?

Basically, we developed and validated tiny capsules or beads (nanocapsules) containing drugs. These nanocapsules are coated with sugars, particularly galacto-oligosaccharides. Senescent cells are characterised by increased lysosomal activity, and one of the more active proteins in these organelles is β‐galactosidase, which digests the coat and preferentially releases the drug cargo in senescent cells. Our work has been recently published in the journal EMBO Molecular Medicine, where we present proof of principle of the therapeutic use of nanocapsules targeting senescent cells in two experimental mouse models, namely pulmonary fibrosis and cancer chemotherapy. These diseases are characterized by the presence of damaged areas, and the eradication of senescent cells resulted in the restoration of pulmonary function and the elimination of tumours, respectively.

Why do senescent cells take in the drug more readily than healthy cells?

The uptake of encapsulated drugs does not occur more readily in senescent cells when compared to healthy cells. However, because healthy cells do not efficiently digest the nanocapsules, then the drug is not released, and the beads are eventually eliminated from the cells in a process of exocytosis. It is important to mention that our encapsulated drugs appear to accumulate more efficiently in tumours enriched in senescent cells, presumably by a process of extravasation that is known as the enhanced permeability and retention (EPR) effect.

What inspired you and your team to take this approach?

When I was doing research as a postdoc in Dr Manuel Serrano’s laboratory, we were interested in developing a therapeutic tool to target cellular senescence. At that time, there were no available drugs to ablate senescent cells in preclinical studies, and I found that this was a very attractive field for my future independent research as a group leader. Then, Dr Manuel Serrano was aware that Prof Ramón Martínez-Mañez (Polytechnic University of Valencia) was exploring the possibility of developing cargo-delivery systems based on nanocapsules to manipulate senescent cells, and we immediately established a formal collaboration with his group.

What are the advantages of gal‐encapsulation compared to traditional systemic drug delivery?

One of the main advantages is that, by encapsulating drugs or senolytics, we can prevent the unwanted side effects of these drugs. As an example, we found that cardiotoxicity associated with doxorubicin (a commonly used chemotherapy) was prevented in our experimental mouse models as well as thrombocytopenia related to navitoclax (a senolytic drug) administration. It is remarkable that gal-encapsulation can be used not only for therapeutic interventions but also for diagnosis. We have validated this approach by the encapsulation of fluorophores, which were preferentially released in damaged areas of tissues accumulating senescent cells and detected by bioimaging techniques.

Our next step, in a potential human setting, will be the encapsulation of contrast agents (to be detected by Nuclear Magnetic Resonance, NMR) or radionuclides (to be detected in positron emission tomography scannings, PET). This approach could be used to determine the senescent burden after treatments with senescence-inducing chemotherapies or in a variety of age-related pathologies.

There is accumulating evidence that different kinds of senescent cells use different pro-survival pathways to remain alive, which is likely to be why there has not yet been a single drug capable of killing all senescent cells. Could your system be used to deliver a senolytic “cocktail” of drugs?

Yes, absolutely. Not only this, our encapsulation system can be used to encapsulate inhibitors, DNAs, siRNAs, proteins, and multiple macromolecules. We know now that the secretory phenotype of senescent cells (known as SASP) is crucial to impact nearby cells and the tissue microenvironment, and these paracrine effects can be either immunoregulatory or immunosuppressive, depending on the context. Persistent proinflammatory effects are related to ageing and age-related disorders, whereas immunosuppressive effects can promote tumour progression. This opens up the possibility of, instead of killing senescent cells, manipulating this process in vivo. For instance, reducing the senescence-associated inflammatory response in chronic diseases. Not only that, we can also target active signalling pathways in senescent cells to have a better understanding of the fundamental biology of this process. The possibility to “reprogramme” senescent cells is very exciting as a potential therapeutic strategy.

Could this system also mean that we would not need to use senolytic drugs and that we could simply deliver a generally cytotoxic payload to the target cells to kill them?

Yes, and the best example is our preclinical study, where we encapsulated doxorubicin, a chemotherapeutic drug capable of eradicating both normal and senescent cells, in a similar manner. By releasing doxorubicin preferentially in senescent cells, our nanocapsules reduced the fibrotic scar in a model of pulmonary fibrosis and also eradicated tumours undergoing therapy-induced senescence.

Some people are calling your system a “smart bomb” for senescent cells, but is there a risk of the system targeting non-senescent cells that express β‐galactosidase, such as macrophages and stem cells?

This risk exists; there is no perfect system or therapeutic tool devoid of concerning side effects. Some cell types are known to have increased β‐galactosidase activity, such as osteoclasts and macrophages. However, by treating mice daily for three weeks with encapsulated doxorubicin, we did not observe any alterations in their serum profiles when compared to healthy mice. In addition, their tissues (particularly the liver, a sensitive organ to drugs) did not exhibit abnormal architecture, as per histology analyses. The versatility of our approach is exemplified by the fact that we can increase the size of the sugar coating with the aim of making more restrictive or stringent nanocapsules, if required.

In addition, there is the possibility of using direct routes of administration, for example by oral gavage, if the target is the digestive system; by aerosolized inhalation, if the target is the lungs; or applied topically, if the target is the skin. An additional improvement for second-generation nanocapsules would be extra funcionalisation with antibodies against senescence biomarkers. We (and other laboratories) are working in the screening of surface proteins overexpressed or specific for senescent cells.

Senolytics have worked wonders in mice, and there’s growing enthusiasm for the prospect that they might be very beneficial for humans, even though some researchers are more skeptical that this might be the case. Are you optimistic that we may see similar results in people as we have in mice, such as improved tissue repair and function?

I am very optimistic for the potential use of senolytics in humans. First, because cellular senescence, similarly to mice, is a defining feature of multiple precancerous lesions and age-related disorders in humans. We have solid evidence that senescent cells accumulate in multiple tissues during ageing across vertebrates, particularly in mice, primates and humans. Second, the preclinical validation of a collection of first-generation senolytics has been performed in a number of mouse models of different disorders. A substantial number of these models efficiently recapitulate the corresponding human diseases (i.e. at the level of molecular pathways involved, genetic features,  epigenetic changes, histopathology, etc.).

Finally, it is worth highlighting that the beneficial effects of senolytics are remarkably relevant; they can prevent or even revert chronic pathological manifestations and extend the lifespan up to 30% in murine models. Importantly, some senolytics are already in early-phase clinical trials and second-generation senolytics are “on the road”. I foresee a strong potential for senolytics in being used in combination with other drugs in precision medicine, depending on the patient and the disease.

There are a number of proposed aging hypotheses, such as Hallmarks, SENS, and the deleteriome. Do you have a particular favorite which you believe reflects closely what aging is?

This is a million-dollar question. We do not have yet a unified theory of ageing. Over the years, many theories of ageing have been proposed; however, all of them have weaknesses or are incomplete. Ageing can be defined as a progressive decline of our tissues, which finally results in dysfunction, and the dysfunction (isolated or in combination) of different tissues can drive numerous pathological manifestations that we know as age-related disorders. Ageing is a complex multifactorial process, but it is very difficult to define or to specify a particular number of categories or hallmarks of ageing. The main reason is because many of these hallmarks are intrinsically interconnected and work in a “cause-effect” fashion. For instance, DNA damage, telomere attrition and mitochondrial dysfunction are intimately ligated to cellular senescence.

Personally, I do not believe in programmed theories of ageing. I think that the cellular programmes that we have are, in any case, aimed at living but not at dying. The fact that our life expectancy or, more specifically, our biological maximum lifespan is inevitably related to the robustness and imperfectness of our genome and our epigenome is a different matter, but the existence of a programme for ageing has not been demonstrated in a similar manner that there is a programme for embryonic development.

Finally, as a researcher, what is the greatest barrier to progress in developing therapies that target the processes of aging in order to prevent age-related diseases?

The main barriers are the (still) limited funds and resources for research. Our discoveries and the development of efficient therapeutic approaches to target ageing, and by extension to prevent age-related disorders, will be a direct function of capital investment. Due to the complexity of ageing, I think that therapeutic interventions to delay this process, or to promote the rejuvenation of our tissues, will require combined approaches of precision and personalised medicine. Researchers and clinicians are, for the first time, positioned to dramatically extend our lifespan and healthspan in the coming decades.

Dr. Muñoz‐Espín’s enthusiasm for the possibility of undoing aging is heartening and contagious; we’re very grateful to him for the work he’s personally doing to push this most important cause forward and for the time he dedicated to our interview.

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

[1] Programmed cell senescence during mammalian embryonic development. Muñoz-Espín D, Cañamero M, Maraver A, Gómez-López G, Contreras J, Murillo-Cuesta S, Rodríguez-Baeza A, Varela-Nieto I, Ruberte J, Collado M, Serrano M. Cell. 2013 Nov 21;155(5):1104-18.

[2] Muñoz‐Espín, D., Rovira, M., Galiana, I., Giménez, C., Lozano‐Torres, B., Paez‐Ribes, M., … & Garaulet, G. (2018). A versatile drug delivery system targeting senescent cells. EMBO molecular medicine, 10(9), e9355.

We Might Be Able to Eradicate Cytomegalovirus

Cytomegalovirus (CMV) is a β-herpesvirus that infects the majority of people in the world. It lies dormant in the body, waiting for an opportunity to strike when the immune system is weakened. This persistent virus infects people for their entire lives, and now researchers have discovered how the virus spreads, opening the door to ways to destroy it.

What is cytomegalovirus?

CMV is part of the β-subfamily of herpesviruses, which are believed to have been co-evolving with their hosts for around 180 million years [1]. CMV infection is asymptomatic; this means it causes no symptoms and is a latent infection; in other words, it lies dormant in the cell, awaiting activation under certain conditions [2].

It is thought that CMV periodically reactivates and attempts to spread, so in order to prevent this, the immune system must keep infected cells under constant, lifelong immune surveillance in order to keep it under control. In this way, it has been proposed that CMV and other similar persistent viruses contribute to infectious burden and aging, as our dwindling supplies of immune cells are increasingly tied up in keeping it under control. Indeed, patients who have compromised immune systems are at greater risk for CMV reactivation, which can lead to an increase of morbidity and mortality [3].

CMV is a highly successful virus that has managed to infect most humans on the planet. This is because CMV is spread readily via exposure to infected secretions and subsequent mucosal contact, which then goes on to infect multiple cell types in the body. After this initial infection, the virus spreads and then enters a dormant (latent) period.

In order to reactivate and awaken from this dormant state, CMV, like all herpesviruses, progresses through an ordered cascade that starts with the expression of intermediate-early genes, which serve as a trigger for further (early and late) viral gene expression [4]. Studies show that CMV periodically expresses these intermediate-early genes at random during its dormant period, meaning that there is a constant background level of CMV reactivation [5-6].

Once these initial genes are expressed, the cycle moves onto expressing genes for host manipulation, DNA replication, and viral packaging. The expression of the intermediate-early genes is closely linked to proinflammatory transcription factors, including NF-κB, TNFα and interleukin-1β. These proinflammatory signals can then reactivate CMV as a result of this association [7-8].

In this way, CMV reacts to its environment, and its activity is largely regulated by the presence of localized inflammation. This means that injuries and other sources of inflammation, such as those that are part of the aging processes, might reactivate CMV from dormancy and help it spread. Also, as we age and our immune systems decline, this leaves us with fewer immune cells to maintain surveillance on infected cells, which further facilitates its spread.

A new way to target CMV

A research team led by Dr. Leor S. Weinberger, William and Ute Bowes Distinguished Professor and director of the Gladstone–UCSF Center for Cell Circuitry, has discovered how the CMV virus replicates [9]. The study was recently published in the journal PNAS, and it opens the door for therapies that could target the persistent virus and remove it from the body.

Under normal circumstances, when a virus invades the cell, the cell responds by inhibiting the viral DNA and preventing it from working. In order to succeed, the virus must somehow compromise this defense system. CMV, it turns out, does exactly that and, thanks to this new study, we now know how it bypasses our cellular defenses. CMV invades the cell, carrying its viral DNA with it, but it also takes with it PP71 proteins, which it then uses to replicate and spread the virus to more healthy cells.

Interestingly, the PP71 proteins that the virus uses to replicate and spread only last a few hours before they break apart, so the researchers questioned how the virus could spread when the PP71 was gone. The answer was that while the PP71 is in the cell, it interacts with another protein called IE1, which occurs not long after the virus invades the cell. Once PP71 is gone, the IE1 facilitates the creation of new viruses, allowing it to spread.

During the study, the research team developed a synthetic version of the virus that allowed them to calibrate the amount of IE1 protein. This allowed them to observe the virus invading cells while controlling the amount of IE1 protein available and how long it lasted. They observed that when IE1 is present at normal levels and it degrades slowly, the virus has plenty of time to replicate and spread to other cells.

Conclusion

This confirms that CMV requires the presence of the IE1 protein and for it to be around for a long-enough period for the virus to use it to replicate. Therefore, IE1 is an attractive target for therapies aimed at destroying CMV and other related herpesviruses, such as Epstein-Barr, and herpes simplex virus type 1 and 2, which cause most cold sores and genital herpes.

The effects of CMV infection on the immune system and its role in aging may be quite significant, given how common and widespread it is in the human population; therefore, understanding how this virus is able to replicate is an important development in finding ways to remove it from the body. Therapies that target IE1 could effectively shut down CMV and potentially allow us to eradicate it forever.

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

Literature

[1] McGeoch, D. J., Cook, S., Dolan, A., Jamieson, F. E., & Telford, E. A. (1995). Molecular phylogeny and evolutionary timescale for the family of mammalian herpesviruses. Journal of molecular biology, 247(3), 443-458.

[2] Fields, B. N., Knipe, D. M., Howley, P. M., & Griffin, D. E. (2007). Fields virology. 5th.

[3] Crough, T., & Khanna, R. (2009). Immunobiology of human cytomegalovirus: from bench to bedside. Clinical microbiology reviews, 22(1), 76-98.

[4] Hermiston, T. W., Malone, C. L., Witte, P. R., & Stinski, M. F. (1987). Identification and characterization of the human cytomegalovirus immediate-early region 2 gene that stimulates gene expression from an inducible promoter. Journal of Virology, 61(10), 3214-3221.

[5] Grzimek, N. K., Dreis, D., Schmalz, S., & Reddehase, M. J. (2001). Random, Asynchronous, and Asymmetric Transcriptional Activity of Enhancer-Flanking Major Immediate-Early Genes ie1/3 andie2 during Murine Cytomegalovirus Latency in the Lungs. Journal of Virology, 75(6), 2692-2705.

[6] Henry, S. C., & Hamilton, J. D. (1993). Detection of murine cytomegalovirus immediate early 1 transcripts in the spleens of latently infected mice. Journal of Infectious Diseases, 167(4), 950-954.

[7] Hummel, M., Zhang, Z., Yan, S., DePlaen, I., Golia, P., Varghese, T., … & Abecassis, M. I. (2001). Allogeneic transplantation induces expression of cytomegalovirus immediate-early genes in vivo: a model for reactivation from latency. Journal of Virology, 75(10), 4814-4822.

[8] Cook, C. H., Trgovcich, J., Zimmerman, P. D., Zhang, Y., & Sedmak, D. D. (2006). Lipopolysaccharide, tumor necrosis factor alpha, or interleukin-1β triggers reactivation of latent cytomegalovirus in immunocompetent mice. Journal of virology, 80(18), 9151-9158.

[9] Vardi, N., Chaturvedi, S., & Weinberger, L. S. (2018). Feedback-mediated signal conversion promotes viral fitness. Proceedings of the National Academy of Sciences, 115(37), E8803-E8810.

Therapies Targeting Aging are in Human Trials Now

A number of therapies that directly target the aging processes are currently in human trials and could change medicine significantly in the next decade if the results are positive.

What is aging?

Aging is basically the accumulation of damage and errors caused by a collection of varied processes that harm the functions of the body through the accumulation of waste, imperfect repair, the deregulation of cellular processes, the dysfunction of the immune system, chronic inflammation, and other disorders.

The body attempts to maintain an equilibrium between the damage that aging causes and the repairs it makes to try to offset this; this balance is known as homeostasis. Unfortunately, as time passes, the repair mechanisms cannot keep pace with the damage; it begins to build up and eventually reaches pathological levels, leading to the familiar diseases of old age that we all recognize. Quite simply, the aging processes cause damage, which eventually leads to age-related diseases.

So, now we understand what aging is, what can we do about it in order to delay, prevent, or even reverse age-related diseases?

Rejuvenation biotechnology targets the aging processes directly

The current approach used in medicine tends to focus on treating symptoms rather than the root causes of diseases; this is the infectious disease model of treatment, and it works fine when dealing with infectious diseases, but when it comes to the treatment of age-related diseases, it is a losing proposition. It becomes a downward spiral of treatment attempting to suppress the symptoms; over time, more age-related diseases appear, and treatment becomes increasingly futile and expensive.

An increasing number of researchers are starting to shift towards the idea that age-related diseases could be halted by directly targeting the aging processes. This makes sense because if these processes can be targeted effectively, then the damage or errors they cause could potentially be kept below the level at which they cause age-related diseases to develop. In other words, by periodically repairing the damage, age-related diseases could be prevented, and people could enjoy healthy and longer lives. This is the approach being taken by a new form of medicine: rejuvenation biotechnology.

There are nine proposed reasons that we age as described in the Hallmarks of Aging, and each is a target for rejuvenation biotechnology therapies.

  • Genomic instability: Damage to nuclear and mitochondrial DNA by free radicals, radiation, and mutagens.
  • Epigenetic alteration: Modifications in gene expression, turning on pro-aging genes and shutting down youthful ones, leading to system-wide loss of function.
  • Telomere attrition: Wearing down of the protective caps on chromosomes.
  • Loss of proteostasis: Deregulation of the mechanisms responsible for protein folding and recycling, leading to the accumulation of harmful by-products.
  • Deregulated nutrient sensing: Deterioration of the cell’s nutrient level response, leading to impairments in energy production, cell growth, and other essential functions.
  • Mitochondrial dysfunction: Damage to the DNA stored in the mitochondria, resulting in reduced efficiency in energy (ATP) production, an increase in oxidative stress, and the contamination of other mitochondria in a chain reaction.
  • Cellular senescence: Accumulation of senescent (non-dividing) cells in the body, impairing tissue function and increasing inflammation.
  • Stem cell exhaustion: Depletion of stem cell reserves, leading to a weaker immune system and inadequate tissue repair.
  • Altered intercellular communication: Deregulation of the communication channels between cells, causing chronic inflammation and tissue damage.

In addition to this, many age-related diseases share these same common hallmarks at their root, so any therapy that can target one or more of these processes has the potential to halt multiple age-related diseases at once. This would be a vast improvement to the single-disease approach used by current medicine and has the potential to revolutionize healthcare.

The first therapies that target the aging processes are already in human trials

In the last decade or so, we have come a long way in our understanding of how aging works and how it relates to age-related diseases. There are now a number of rejuvenation biotechnology-based therapies already in human trials right now.

Stem cell therapies

Stem cell therapies are probably one of the best-known forms of rejuvenation biotechnology, having been around for over a decade, and their use is becoming increasingly common in medical practice. Stem cell exhaustion is a reason we age, as our pool of replacement cells runs out or is prevented from working by chronic age-related inflammation, and the repair and regeneration of our tissues and organs grind to a halt.

Mesenchymal stem cells (MSCs) have been in the spotlight recently and are currently in human trials to test their effectiveness against age-related frailty as part of the CRATUS program [1]. Named after the Greek God of strength and power, CRATUS clinical trials aim to reduce frailty in the elderly via MSC therapy.

Initial safety and efficacy studies have shown that MSCs can improve the physical performance of frail elderly people. This is good news, as frailty has a serious impact on independence and the quality of life in older people and can lead to falls, which often prove fatal.

These positive results are no doubt linked to the beneficial secretions produced by MSCs, which reduce the chronic, age-related inflammation of elderly people, thus spurring tissue repair and regeneration. The ongoing human trials showed that transplanting MSCs significantly reduced systemic inflammation, meaning that this treatment holds potential for addressing age-related frailty.

This is just one of many examples of where stem cells are currently being tested to combat age-related diseases. There are other trials investigating the use of stem cells of various kinds against diseases such as Parkinson’s and Alzheimer’s and for replacing cartilage in joints and even regenerating the thymus to boost the aging immune system. While there are challenges and hurdles to overcome, the field of stem cell research holds great potential in the battle against age-related diseases.

Senolytics

Senolytic drugs that address cellular senescence, which is one of the hallmarks of aging, have enjoyed considerable press exposure in the last few years.

As we grow older, more and more of our cells enter a state known as senescence: they cease dividing and supporting the tissues of which they are a part; instead, they secrete a range of harmful signals that prevent tissue repair and cause healthy nearby cells to also become senescent.

These signals contribute to the smoldering background of chronic inflammation that most older people experience and are thought to facilitate a variety of age-related diseases [2-8]. Under normal conditions, senescent cells destroy themselves via the process of apoptosis, a kind of self-destruct mechanism, and are then disposed of by the immune system. However, as we age, more of these cells evade apoptosis as our immune systems decline, and more of them begin to build up in our bodies.

It has been proposed that removing these senescent cells could prove beneficial for health and help prevent age-related diseases. So, in 2011, researchers put this idea to the test. Here are two mice from a Mayo Clinic study; the two mice are littermates and are the same age [9]. The mouse on the left has been left to age normally, the mouse on the right has had the senescent cells purged from its body. You can see a striking difference in physical appearance, and the treated mouse was healthier, lived for longer, and remained free from age-related diseases for a longer time. The caveat is that these mice were engineered to react to a compound to purge the cells and their senescent cells were not accumulated via natural aging, so this is not really representative of true aging.

This is where a new class of drugs called senolytics comes in. These drugs selectively target senescent cells and cause them to enter apoptosis, which they should have done naturally. In 2016, a follow-up to the 2011 experiment used senolytic drugs in normally aging mice to see if the results would be similar [10]. Once again, the researchers discovered that the removal of senescent cells improved the health of mice and seemed to delay various aspects of aging in tissues and organs.

This was the experiment that really got the interest in senolytics moving, and, as of today, they are now in human clinical trials with a company called UNITY Biotechnology, and there are various other companies hot on their heels to develop effective senescent cell removal therapies. Senolytics have huge potential in delaying, preventing, and even reversing multiple age-related diseases.

NAD+ therapy

As we grow older, our body has increasing difficulty detecting nutrients and energy levels and responding to them in an appropriate way; this is deregulated nutrient sensing, which is another hallmark of aging. In the last few years, interest in a molecule central to energy metabolism and nutrient sensing has grown considerably in relation to aging.

The molecule in question is nicotinamide adenine dinucleotide (NAD+), which is an important metabolic signaling molecule found in the cells of all species. NAD+ helps facilitate cellular functions, DNA repair, growth, and many more things. Quite simply, without NAD+, life would be impossible.

Unfortunately, as we age, inflammation and other factors reduce the amount of NAD+ available to our cells, and our metabolism becomes dysfunctional as a result, leading to metabolic disorders such as T2 diabetes and obesity [11-12], vascular aging [13], and potentially heart disease [14].

Researchers are now seeing if restoring NAD+ levels in aged people might help them to remain healthier and delay or even prevent age-related diseases. They are doing this by using a compound called nicotinamide mononucleotide (NMN), which our bodies turn into NAD+ through a number of chemical steps and which has been shown in animals and human cells to increase the level of NAD+ available.

Earlier this year, researchers led by Dr. David Sinclair at Harvard Medical School showed that NAD+ was also able to restore blood flow in aged mice [15]. They gave the mice NMN, which, in turn, increased the level of NAD+ and encouraged the formation of new blood vessels supplying muscle tissue and organs, thus improving blood flow, much like what happens when you exercise.

During the study, the aged mice saw a 60% improvement to their treadmill exercise time compared to untreated mice of the same age. The mice also doubled their exercise endurance to a level similar to or better than younger mice. This suggests that NMN is able to fool the body into reacting as if it had been exercising.

Essentially, if this could be developed into a therapy, a pill might be created that emulates the effects of running 10 miles a day without having to do so, and it could help us to remain healthy and free from age-related diseases for longer. It has potential not only for age-related diseases but also for treating patients who are unable to move or who have mobility issues caused by age-related frailty.

NMN is currently in human clinical trials at Brigham and Women’s Hospital near the Harvard Medical School facility, and it is hoped that initial results will be available sometime in 2019.

Conclusion

These are just three examples of a new generation of medicine that is being developed and could dramatically change the way we think about and treat age-related diseases. There are other therapies currently being developed that address the various reasons we age, and these therapies are in different preclinical and clinical stages.

The idea of treating the aging processes directly to deal with age-related diseases is gaining traction, and, hopefully, it will replace the outdated idea that such diseases can be addressed using an infectious disease model of treatment. The results of these early human trials will be critical in shaping how the future of medicine pans out.

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

Literature

[1] Schulman, I. H., Balkan, W., & Hare, J. M. (2018). Mesenchymal Stem Cell Therapy for Aging Frailty. Frontiers in Nutrition, 5.

[2] Roos, C. M., Zhang, B., Palmer, A. K., Ogrodnik, M. B., Pirtskhalava, T., Thalji, N. M., … & Zhu, Y. (2016). Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging cell.

[3] Childs, B. G., Baker, D. J., Wijshake, T., Conover, C. A., Campisi, J., & van Deursen, J. M. (2016). Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science, 354(6311), 472-477.

[4] Palmer, A. K., Tchkonia, T., LeBrasseur, N. K., Chini, E. N., Xu, M., & Kirkland, J. L. (2015). Cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes, 64(7), 2289-2298.

[5] Velarde, M. C., & Demaria, M. (2016). Targeting Senescent Cells: Possible Implications for Delaying Skin Aging: A Mini-Review. Gerontology.

[6] Xu, M., Bradley, E. W., Weivoda, M. M., Hwang, S. M., Pirtskhalava, T., Decklever, T., … & Lowe, V. (2016). Transplanted Senescent Cells Induce an Osteoarthritis-Like Condition in Mice. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, glw154.

[7] Bussian, T. J., Aziz, A., Meyer, C. F., Swenson, B. L., van Deursen, J. M., & Baker, D. J. (2018). Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature.

[8] Coppé, J. P., Desprez, P. Y., Krtolica, A., & Campisi, J. (2010). The senescence-associated secretory phenotype: the dark side of tumor suppression. Annual review of pathology, 5, 99.

[9] Baker, D. J., Wijshake, T., Tchkonia, T., LeBrasseur, N. K., Childs, B. G., Van De Sluis, B., … & Van Deursen, J. M. (2011). Clearance of p16 Ink4a-positive senescent cells delays ageing-associated disorders. Nature, 479(7372), 232.

[10] Baker, D. J., Childs, B. G., Durik, M., Wijers, M. E., Sieben, C. J., Zhong, J., … & Khazaie, K. (2016). Naturally occurring p16 Ink4a-positive cells shorten healthy lifespan. Nature, 530(7589), 184

[11] Elhassan, Y. S., Philp, A. A., & Lavery, G. G. (2017). Targeting NAD+ in metabolic disease: new insights into an old molecule. Journal of the Endocrine Society, 1(7), 816-835.

[12] Mouchiroud, L., Houtkooper, R. H., & Auwerx, J. (2013). NAD+ metabolism: a therapeutic target for age-related metabolic disease. Critical reviews in biochemistry and molecular biology, 48(4), 397-408.

[13] Sinclair D. Bonkowski, M. Impairment of an Endothelial NAD+-H2S Signaling Network Is a Reversible Cause of Vascular Aging (2018) doi.org/10.1016/j.cell.2018.02.008

[14] Nencioni, A., Da Silva, R. F., Fraga-Silva, R. A., Steffens, S., Fabre, M., Bauer, I., … & Pelli, G. (2014). Nicotinamide phosphoribosyltransferase inhibition reduces intraplaque CXCL1 production and associated neutrophil infiltration in atherosclerotic mice. Thrombosis and haemostasis, 112(02), 308-322.

[15] Sinclair D. Bonkowski, M. Impairment of an Endothelial NAD+-H2S Signaling Network Is a Reversible Cause of Vascular Aging (2018) doi.org/10.1016/j.cell.2018.02.008

Human Stem Cell Trial Successful Against Age-Related Frailty

Today, we want to highlight results from human trials in which stem cell transplants have been shown to reduce age-related frailty.

Age-related frailty and stem cell transplants

Currently, there are no specific approved therapies to address age-related frailty, which can cause elderly people to suffer potentially fatal falls and injuries. There has been considerable interest in stem cell therapies to combat frailty in recent years, and the results we will discuss today are from one of the more advanced human clinical trials exploring mesenchymal stem cell (MSC) transplants [1].

Named after the Greek god of strength and power, the CRATUS series of stem cell studies involves transplanting mesenchymal stem cells into patients to try to address age-related frailty. There have been numerous studies showing that MSCs have potential in improving tissue repair, so this application is the logical conclusion of years of testing and study.

These health improvements are almost certainly linked to the reduction of chronic inflammation, which is known to impair tissue repair and regeneration, by inhibiting the activity of tissue resident stem cells. The presence of MSCs appears to reduce the background of chronic age-related inflammation and thus facilitates healing and tissue repair due to the beneficial secretions that these cells produce.

Perhaps most interestingly, MSCs rarely join with the tissues to which they are transplanted; however, they do remain active for a considerable time, often for months, and help reduce inflammation, giving damaged tissue time to recover before the inflammation returns to continue its relentless assault.

Chronic diseases and degenerative conditions are strongly linked with the geriatric syndrome of frailty and account for a disproportionate percentage of the health care budget. Frailty increases the risk of falls, hospitalization, institutionalization, disability, and death. By definition, frailty syndrome is characterized by declines in lean body mass, strength, endurance, balance, gait speed, activity and energy levels, and organ physiologic reserve. Collectively, these changes lead to the loss of homeostasis and capability to withstand stressors and resulting vulnerabilities. There is a strong link between frailty, inflammation, and the impaired ability to repair tissue injury due to decreases in endogenous stem cell production. Although exercise and nutritional supplementation provide benefit to frail patients, there are currently no specific therapies for frailty. Bone marrow-derived allogeneic mesenchymal stem cells (MSCs) provide therapeutic benefits in heart failure patients irrespective of age. MSCs contribute to cellular repair and tissue regeneration through their multilineage differentiation capacity, immunomodulatory, and anti-inflammatory effects, homing and migratory capacity to injury sites, and a stimulatory effect on endogenous tissue progenitors. The advantages of using MSCs as a therapeutic strategy include standardization of isolation and culture expansion techniques and safety in allogeneic transplantation. Based on this evidence, we performed a randomized, double-blinded, dose-finding study in elderly, frail individuals and showed that intravenously delivered allogeneic MSCs are safe and produce significant improvements in physical performance measures and inflammatory biomarkers. We thus propose that frailty can be treated and the link between frailty and chronic inflammation offers a potential therapeutic target, addressable by cell therapy.

Results of phase 1 and 2 human clinical trials

The researchers conducted phase 1 and 2 clinical trials under the CRATUS program using mesenchymal stem cell transplants to reduce frailty in aged patients [2-4]. The ultimate goal of the CRATUS program is to restore or maintain cognitive and physical function in elderly patients and lengthen their healthy lifespan. These clinical trials were designed to assess the safety and efficacy of transplanting allogeneic bone marrow-derived MSCs to patients with mild to moderate frailty, and their success was based on an improvement of physical performance, quality of life, and reduction of chronic systemic inflammation.

The phase 1 results showed that MSC therapy is well tolerated, and those results led to a phase 2 efficacy trial using both 100-million and 200-million dosages. At the 100-million dose, patients were observed to have improved physical abilities, including performance in a 6-minute walking test, a physical performance exam, and an improvement of forced expiratory volume, suggesting improved lung function. The higher 200-million dose was less effective.

Blood levels of inflammatory TNF-α and activated T-cells were reduced significantly in both dosage groups. This suggests that the MSCs reduced systemic inflammation and excessive activation of the immune system, which are typical in old age.

Conclusion

These are promising and positive results indeed, and the next step will be a 2b study with a larger test group to see if the results remain consistent. Should this prove successful, the therapy will most likely pass through the approval process, and, at last, patients will have an available solution for combating frailty.

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

Literature

[1] Schulman, I. H., Balkan, W., & Hare, J. M. (2018). Mesenchymal Stem Cell Therapy for Aging Frailty. Frontiers in Nutrition, 5.

[2] Tompkins, B. A., DiFede, D. L., Khan, A., Landin, A. M., Schulman, I. H., Pujol, M. V., … & Mushtaq, M. (2017). Allogeneic Mesenchymal Stem Cells Ameliorate Aging Frailty: A Phase II Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences, 72(11), 1513-1522.

[3] Golpanian S, DiFede DL, Khan A, Schulman IH, Landin AM, Tompkins BA, et al. Allogeneic human mesenchymal stem cell infusions for aging frailty. J Gerontol Ser A Biolog Sci Med Sci. (2017) 72:1505–12. doi: 10.1093/gerona/glx056

[4] Golpanian, S., DiFede, D. L., Pujol, M. V., Lowery, M. H., Levis-Dusseau, S., Goldstein, B. J., … & Heldman, A. W. (2016). Rationale and design of the allogeneiC human mesenchymal stem cells (hMSC) in patients with aging fRAilTy via intravenoUS delivery (CRATUS) study: a phase I/II, randomized, blinded and placebo controlled trial to evaluate the safety and potential efficacy of allogeneic human mesenchymal stem cell infusion in patients with aging frailty. Oncotarget, 7(11), 11899.

Investigating Jeanne Calment’s Longevity Record

Jeanne Calment has the claim of being the longest lived human, but two researchers believe this claim may be false.

Is the oldest recorded human being potentially a hoax?

If you open an article dedicated to supercentenarians, it is very likely that at its very beginning, you will see the name of Jeanne Calment, the oldest known person in the world, who is believed to have lived for up to 122 years. Jeanne is not merely a unique phenomenon from the point of view of statistics; over the years, she became a symbol of extraordinary human capacities.

For a person who sticks to a healthy lifestyle or even engages in biohacking in order to live longer, Jeanne’s record is a teasing goal to achieve and surpass;however, to the researchers of aging, this extremely rare event is rather a reason for curiosity-and skepticism.

A couple of weeks ago at the joint meeting of the Gerontological Society of RAS and the Gerontology Department of Moscow Society of Naturalists (Moscow State University), there was a report by Nikolay Zak, who holds a PhD in physico-mathematical sciences, that shed new light on the case of Jeanne Calment. The main hypothesis of this independent investigation is that the person who we know as Jeanne Calment is actually her daughter, Yvonne, who took the place of her mother after her death in 1934 in order to help her family avoid heavy financial losses related to inheritance.

The initiator of this independent investigation, Valery Novoselov, assistant professor of the Department of Gerontology and Geriatrics of RUDN University(Moscow), is convinced that Calment’s case has to be revalidated.

We need practical solutions-and this is why any data concerning aging should be robust

In Russia, Valery Novoselov is mostly known for his studies of medical documents of historic persons such as Vladimir Lenin. His review of medical archives dedicated to shedding light on the real causes of death of this great Russian politician is ready but, so far, remains unpublished; there are too many stakeholders interested in this issue, including people who would probably prefer to see politically motivated malice behind Lenin’s death.

While Valery discusses the hurdles involved in gathering the relevant data, and the warnings of his lawyer, I find myself thinking about a simple yet shocking truth:the quality of medical data and its public availability can literally make history. It makes a great introduction to the main topic of our conversation.

E. (Elena Milova) -Valery, you are currently involved in revalidating longevity records. What was your motivation to engage in these investigations in the first place?

V. (Valery Novoselov) -You know, my main focus of interest is people. I don’t like to deal with animals, because I believe that due to evolutionary mechanisms, the processes of aging in different species are not homologous. So, I am only interested in analyzing human data with some practical application of the results. Back in 2016, I was curious how many centenarians there were in the Moscow region. I live here, so it could be feasible to gather the data and even meet these people to conduct a survey and collect information about the factors that could have promoted their longevity. You see, there are Blue Zones where there are many centenarians, and the data about their lifestyle can probably explain their longevity. However, most of Russia is located in a totally different climate zone, and what is known about the life-extending lifestyles of, say, Mediterranean countries cannot be applied to our latitude.

V. -So, I was curious about the centenarians in the Moscow region, and I have sent requests to two different agencies:the Department of Labor and Social Security and the Federal Agency of Statistics. They provided me with two absolutely different sets of data. The one from the agency gave me 4135 people aged 100 and older, and the Department of Labor gave me 735 people. 6-fold difference. In my conversations with people working in these organizations, there were many surprises. First of all, they themselves were not sure about the numbers. Second, there were hallmarks of unreliability, such as a huge prevalence of centenarians only in the regions of Moscow and Saint-Petersburg, and a steep growth in this age group in one year-up to 30%. We requested personal information on these people, as this is the only way to check the real numbers. However, the main idea here is this:too much variance of data is likely an indicator of errors. In centenarians, the possibility of error is the highest.

The case of Jeanne does not look like it fits into the survival curve models developed by the leading demographers of aging

E. -So, what was the starting point in the investigation of Jeanne Calment’s case? What was the first thing that caused the initial skepticism?

V. -In the last few years, there were many interesting articles on the survival curve of centenarians and supercentenarians. Some of them were written by Leonid and Natalia Gavrilovs and some by Elisabetta Barbi and her colleagues, and despite their differing views on the survival plateaus of marginal age groups, the case of Jeanne Calment didn’t fit into any of the refined math models behind their studies. If we imagine the curves of survival in these studies, Jeanne is a dot away from the main trend that they describe. One more reason for suspicion is how far from other longevity records her age is. There are only two cases of this kind:Jeanne and Sarah Knauss, whose record is 119 years. All other supercentenarians are several years apart from them. Several years of difference in my or your age is nothing surprising compared to the same difference in very old age. Most longevity records are very close to one another. Whenever a new record is set, the person dies several days or several weeks later, very rarely several months later. However, we are never speaking about years apart, definitely not several years. Furthermore, we looked at the French database of supercentenarians of the Max Planck Institute for Demographic Research (there are 49 such French people), and we have found that Jeanne was the first(by birth date)validated supercentenarian in France and that she is also the most long-lived person in the world. It is a very unlikely combination of events from the point of view of statistics.

E. -Yeah, this sounds somewhat counterintuitive. It would be more logical if people who were born later than Jeanne would preserve their health for longer and live longer, as they would have access to more advanced medical technologies.

V. -That too. We know from history that there were many cases when people had exaggerated their ages, but it was much easier to reveal because it was most often in men, and as you know, men tend to live shorter lives than women, so in old age, the number of women is much higher. But Jeanne is a woman, so her longevity is by default less surprising and suspicious. Nevertheless, there was a big commission that was formed to validate supercentenarians. In France, such an organization is funded by the French pharmaceutical company Ipsen.

From suspicion to revalidation of the case

E. -So, you started to check the data from this validation group?

V. -I had many ideas at once. I am a geriatrician, and in my work, I rely on visual assessment a lot. My eyes were telling me that Jeanne didn’t have the hallmarks of frailty that would correspond to her official age, such as the fact that unlike other supercentenarians, she was able to sit straight in her chair without others’ help. I didn’t see enough signs of dermal atrophy nor atrophy of subcutaneous tissue. Most of all, it was my clinical experience that made me question Jeanne’s age and believe that we will find something interesting.

V. -As a first step, I decided to run a survey to see how people assessed Jeanne’s age by comparing her photos and videos to the photos and videos of other supercentenarians. The participants (233 random people) were massively reducing her age by around 20-25 years compared to her official age on the date when this picture was taken. There was one more person with the same perception, the Major of Arles. He is responsible for congratulating people who have reached 100, and, in accordance with tradition, he should have visited Jeanne. However, she said that instead she would prefer to come along. This willingness to take a walk across the city is already surprising in a 100-year-old, but apart from that, the Mayor, who was used to dealing with centenarians, didn’t recognise his hero of the occasion in a spry old lady sitting in his lobby.

E. -What was the next stage of the investigation?

V. -I invited a young mathematician, Nikolay Zak, to help me analyze the existing datasets on supercentenarians and see if Jeanne could fit in. He has found that she could not. The models developed by him claimed that if we rely on the laws of statistics, Jeanne as a phenomenon should not exist. It was such a big surprise to Nikolay that he decided to personally revalidate this case. His French is fairly good, so he reached out to the holders of the archives in Arles, found some volunteers there, and started to check every small detail.

V. -You know, on most of the occasions when I raised the issue with my colleagues, their first counterargument was “How could an error or even a deception take place if Jeanne lived in a small city where people knew one another very well?” It turns out that this was a misconception, as Arles was one of the biggest French communes at that time with 38 thousand people(even now, not every Moscow satellite city has that many people), and apart from that, Jeanne didn’t live in the city all the time; she and her daughter often spent their time in a homestead 16 kilometers away from Arles. The more that Nikolay checked, the more that small inconsistencies, errors, and even signs of intentional fraud were revealed. After looking at all the data that Nikolay has managed to collect, including the known intentional destruction of the family archive on Jeanne’s orders, we developed a hypothesis that is now being checked. In 1934, there was a death in the Calment family. The official story is that in 1934, Jeanne had lost her only daughter, Yvonne. We think that in reality it was Jeanne who had died, aged almost 59, and her daughter took her name and personality.

E. – Detective work teaches us that a person who is suspected to have broken the law should have had some sort of motive for that.

V. – Indeed, and there was a motive. The 1930s were dire years for the family. Her mother in law and her father both died in 1931, and the family had to pay huge inheritance taxes in each case. Unlike their levels at the beginning of the century, these taxes were up to 35% of the property’s cost, as the government was likely preparing for the next world war. We could expect the family to be in quite a miserable financial situation. If Jeanne had died, her daughter Yvonne and her husband would have to pay a lot of money. However, if it were Yvonne who died, the family wouldn’t have to pay any taxes, as she didn’t own the homestead.

Dealing with the archive… or its absence

E. – Is there more precise proof that Yvonne has replaced Jeanne?

V. – We suspect that the passport of Jeanne, which was issued by the French gendarmerie in the 1930s, can be considered to be proof of replacement. The features described in the passport, such as eye color and height, do not correspond to the features of Jeanne when she was old. There is another interesting fact, too. In case of death, the usual formal procedures require a witness to look at the body and sign the statement of death. How you would do it normally, if one of your relatives would suddenly die? You would spare yourself the effort and would call some neighbors, I guess. This was not the case, as an old stranger living far away was invited instead. When I say old, I mean around 70 years old, and, at that time, this could have meant a lot of health issues, including sight problems. Why invite total strangers from far away, and why should they be old?

E. – Yes, this sounds weird. Right, these are suspicious documents, but if we are talking about a family conspiracy, there should be some unconscious leaks in their behavior or speech.

V. – There are, of course. For instance, Yvonne’s husband Joseph Charles Frédéric Billot never got remarried, despite the fact that he was only 42 at the moment of her “death”. There were many mentions that he was getting along very well with Jeanne and they were raising Yvonne’s son Frédéric Jean Paul together. You would expect a husband to treat his own wife well, wouldn’t you? The kid, by the way, was calling Jeanne “mamzanne”, that is, Mom Jeanne. It also looks unusual. Next, Jeanne used to mention the maid that took her to school. Nikolay has found the date of birth of this maid, Marthe Fousson, in the birth certificate, and year of birth in the census of 1911, and it turns out that she was 10 years younger than Jeanne, which means that she could only be taking her daughter Yvonne to school. Hunting was an important part of her lifestyle, but the age of her first hunt jumped around impressively in the span of 20 years.

E. -Documents, behavior, anything else?

V. -The last test was based on the photos. As you know, even if people age, most proportions of the face, such as the distance between the eyes, the nose shape, and the level of the hairline in women don’t change. Some of them don’t change at all, as they are defined by the skull shape. Nikolay suggested placing the pictures of young and old Jeanne together and seeing if the proportions match. It turns out that the features of the young Yvonne match the features of the old Jeanne. However, when you compare the features of the young Jeanne to the old Jeanne, they don’t match. Surprising, isn’t it?

E. -A finding of this scale should have shaken the academic community. How did your fellow researchers of aging respond to the preliminary results of the investigation?

V. -There was some initial skepticism, but after a proper exchange of data with my Russian and foreign colleagues, there is a growing interest in seeing the results of this investigation. The president of the Gerontological Society of RAS, Vladimir Anisimov, encourages us to keep investigating until we find the truth. As you probably know, the main professional organization that is performing validation is the Gerontology Research Group. It is currently led by Robert D. Young, who is also a Senior Consultant for Gerontology at Guinness World Records. I was very pleased with his interest and support. Working together, we will hopefully come to a definitive conclusion sooner.

E. -It is nice to learn that the community is open to the idea of revalidation.

V. -Indeed. However, I am asking myself why the revalidation was not initiated earlier, as the more you dig, the more questions arise. I have found a hint to a possible explanation in the book “L’assurance et ses secrets” (Insurance and its secrets) by Jean-Pierre Daniel that was published in 2007. Here it is:

V. -“Chacun se souvient de Jeanne Calment officiellement morte à 122 ans, le 4 août 1997, Il avait été dit à l’époque que cette dame bénéficiait d’une rente viagère, ce qui etait vrai. Celle-ci etait versée par une grande société française que cette longévité exceptionelle ne réjouissait pas. La société était d’autant plus marrie qu’elle savait pertinemment qu’elle ne payait pas Jeanne Calment, mais sa fille. En effect, au décès de la vraie Jeanne Calment, sa fille qui évidemment n’était plus une gamine, avait endossé l’identité de sa mère pour continuer à toucher la rente. La société d’assurance avait découvert l’usurpation d’identité, mais en accord – ou à la demande ? – des pouvoirs publics, elle n’avait pas souhaité la dénoncer tant le personnage de la “doyenne des Français” était devenu mythique.”

V. -“Everyone remembers Jeanne Calment, who has officially died at age 122 on August 4, 1997. It was said at the time that this lady had benefited from having a life annuity, which was true. This was paid by a large French company that was not happy at all with this exceptional longevity. The company was even more upset as it knew that it had been paying not Jeanne Calment, but her daughter. In reality, after the death of the real Jeanne Calment, her daughter who obviously was no longer a child, had taken her mother’s identity to keep receiving the annuity. The insurance company had discovered identity theft, but in agreement with – or on the demand of? – the public authorities, it had not wished to reveal the truth, given how much the character of the “grandmother of the French” had become legendary.”

E. -So, there were some players involved who would apparently prefer to protect the image of a national hero, even if it meant sacrificing the accuracy of data used for scientific studies?

V. -It could be. However, I would also consider another prerequisite for this particular situation. It is a lack of focusing on the most important goals and lack of a spirit of rebellion in the scientific community. I’ll explain why I think so. What is the main goal of gerontology as a science?It is to help people remain healthy and live longer. If you are setting this as a goal, it is obvious that you could not stand it if the data on your hands were questionable. If you want science to progress and to bear fruit such as reliable lifestyle recommendations or a new drug to slow down aging, even a slight suspicion should be enough to spark further investigation in order to make the data as robust as possible. A spirit of rebellion is absolutely necessary to be able to follow your scientific intuition despite the accepted views.

E. -Rebellion, I like that!Well, what will be the next step of your investigation?Do you plan to contact Guinness World Records to let them know that the case of Jeanne Calment likely requires revalidation?

V. -We already sent them our materials and are waiting for a reply. Right now, we need to exchange more information with our colleagues at the GRG, as they are the most experienced group in the world when it comes to longevity validation. Ideally, the next step is to prepare and publish scientific peer-reviewed articles with all the information that we have been able to collect.

E. -In your opinion, what is the main lesson of this story?

V. -Well, to be completely honest, the main lesson is still to be learned. You see, the current buzz around longevity records can be easily distracting us from the goals that are truly important. I’d really want this story to be reduced to a revalidation by a qualified group of researchers and to an update of all corresponding books. In my view, it just does not deserve the hype. There was a mistake, we will correct it, and that is it. We will be seeing new longevity records again and again; it will never stop, because there is no proven limit of human healthspan and lifespan.

V. -There are many signs, however, that both healthspan and lifespan can largely depend on the medical technologies that we have. The population is aging very fast, and I believe that we need to focus our efforts on developing and testing the interventions that would effectively bring aging under medical control in humans. This is priority number one for Russian gerontology. If we let things of relatively small practical value, such as the discussion of longevity records and the personal stories behind them, become a talk show and distract us beyond measure, we may come to 2050 completely unprepared.

There will be 2 billion people who are 60 years old and older by that time, which is 1/4 of the global population. How are we supposed to cope with the overload of our healthcare system without powerful therapies that can address the underlying mechanisms of aging and thus prevent and cure age-related diseases? We need to undertake preemptive steps. We need honesty, courage, openness and the ability to act fast in creating these innovative treatments. We need the flexibility to find new ways and bypass obstacles. We need cool heads. This is what we truly need, and we need it right now.

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

Rejuvenation Roundup November 2018

‘Tis the season to be jolly, friends of healthy longevity! Time to dim the lights, gather around a crackling fireplace, cuddle up into a warm blanket, and recap the rejuvenation news of last November.

LEAF News

Highlight: Keith at The Young Turks

LEAF President Keith Comito will have a lot of sleep to catch on once rejuvenation therapies finally reach the clinic—right now, he’s too busy building bridges to a world free of age-related diseases to get a little shuteye, and getting the word out through as many outlets as possible is part of the job description. Among his many outreach efforts, he was recently featured on The Young Turks, a popular American news and commentary YouTube channel with a following of over four million people; during his chat with host Cenk Uygur, Keith discussed the science of healthy life extension and Lifespan.io’s mission to crowdfund the cure for aging. It was great to see that Cenk was 100% onboard with the cause, and if you are too, a super simple, effective, and zero-cost way to help the cause is to share the video below on all your social media!

Team and activities

NAD+ Mouse Project wrap-up. The NAD+ Mouse Project came to a conclusion in early November, and thanks to our generous supporters, it was an amazing success! The donation meter reached far beyond the initial goal of $30,000, smashing the third stretch goal and hitting an incredible $75,285! Our deepest gratitude goes out to everyone who helped make this possible! Thank you also from Dr. David Sinclair, who commented: “We are both humbled and so very grateful for the generous support of the NAD mouse project. As the excitement and potential of the NAD longevity field continues to grow, we can’t wait to see what we learn and report to our supporters about what NMN can do.”

Stay tuned for our next campaign!

Podcast: Ryan O’Shea of the Future Grind podcast returns to bring you the November edition of the Rejuvenation Roundup podcast.

LEAF at EHA. The Fourth Eurosymposium on Healthy Ageing, a biannual event organized in Brussels by the Healthy Life Extension Society, took place from November 8 to November 10. The conference featured a number of interesting talks by leading scientists on the topics of cell senescence, proteostasis, and DNA repair as well as panels and talks about advocacy as well as social and financial issues related to rejuvenation. 

Journal Club. November’s Journal Club saw the discussion of a paper on the link between cellular senescence and tau protein aggregation in the brain, one of the key factors in Alzheimer’s disease.

Lifespan.io interviews

Leonid Gavrilov and Natalia Gavrilova. The two well known biodemographers of aging had a chat with us about the possible effects of vastly extended lifespans on the population as well as other, broader questions related to aging and life extension.

Events

Undoing Aging 2019. The second instalment of the Undoing Aging conference series is getting closer and closer, and the list of amazing speakers that it will feature was recently updated last month and just keeps growing! Next year in Berlin, you’ll have a chance to meet Dr. Julie Andersen and Dr. Judy Campisi from the Buck Institute for Research on Aging, Dr. Jerry Shay from the University of Texas Southwestern Medical Center, Dr. Ruby Yanru Chen-Tsai from Applied StemCell, Inc., and many more! If you plan on offering a presentation of your own, the deadline to send in an abstract is January 31.

SRF winter fundraiser. SENS Research Foundation, the engine room of multiple rejuvenation biotechnology initiatives, is running its traditional winter fundraiser, which is due to terminate on December 31. The fundraiser’s goal is $500,000, and the meter currently stands at just over $183,000 in no small part thanks to Jim Mellon’s generous donation of $100,000. In addition, Josh Triplett, Christophe and Dominique Cornuejols, and Fight Aging! have also put together a matching fund of $54,000 to match the next year of donations of any new SRF patrons who pledge a monthly donation. If you want to help SRF in its quest to bring aging under full medical control, here’s your chance!

The Longevity Forum. The first Longevity Forum, an initiative by investor Jim Mellon, was held in London on November 5th; the event brought together illustrious figures of the rejuvenation research and investment landscape, such as President of the Buck Institute for Research on Aging Eric Verdin, AgeX Therapeutics CEO Mike West, and Jim Mellon himself. As stated on its website, the Forum “aims to explore and establish new ways of addressing opportunities/issues related to longevity by displaying proof of concept”. You can read a firsthand report by Fight Aging!‘s editor Reason, who attended the event.

Longevity Leaders. The Longevity Leaders Conference will take place next February in London and will see our own Steve Hill as a panel moderator. As always, you can expect interviews and event coverage, so keep an eye on the blog!

Longevity Therapeutics. Yet another event of interest that we discussed last month was the Longevity Therapeutics Summit, which will be held in San Francisco at the end of January. Speakers of the caliber of UNITY‘s Dr. Nathaniel David and Buck Institute Professor Judith Campisi will be present, so you might want to attend as well.

Research roundup

Cellular senescence

Two known antibiotics as potential senolytics. A paper by scientists from the University of Salford suggests that the known antibiotic drugs azithromycin and roxithromycin may have senolytic effects, as they appear to target senescent human fibroblasts. The study was conducted in cell cultures, and as Fight Aging! points out in a short commentary, it is too early to say whether the same effects observed in vitro will translate to animals—or people.

Smart bombs against senescent cells. Senolytic Therapeutics is one of the many biotech startups working on a particular approach to effectively remove excess senescent cells from the body. Its approach has been likened to “smart bombs” by Michael Rae on SRF’s blog in that, while the chemotherapeutic load of the drug is capable of killing any cell to which it is delivered, it is encapsulated within a nanotube that can be unlocked only by senescent cells. Further commentary can be found in this article by Fight Aging!

Loss of proteostasis

AMBAR to slow down Alzheimer’s. A new therapy known as AMBAR has been shown to significantly slow down the progression of Alzheimer’s disease during a phase III clinical trial; even though AMBAR is not a cure for the disease, it is still a significant step forward, and it should become commercially available following FDA registration within the next few years.

Geroneuroprotectors against Alzheimer’s disease. Geroneuroprotectors are a subset of geroprotectors that have shown promise in terms of their ability to slow down aging in mice, particularly the effects of Alzheimer’s disease. In a study by the Salk Institute, researchers focused their efforts on three compounds that they synthesized from fisetin and curcumin, and they are now hoping to get two of these compounds into human clinical trials.

Deregulated nutrient sensing

mTORC1, type 2 diabetes, and aging. A recent paper, commented on here by Fight Aging!, discusses the hyperactivation of the mTORC1 pathway that is observed in both type 2 diabetes and aging; aging increases the likelihood of developing type 2 diabetes, while type 2 diabetes can exacerbate other aspects of aging.

Other

Longevity isn’t all about genetics. A paper published in early November suggests that the genetic contribution to human longevity, traditionally estimated at up to 30%, is even lower than that—under 10%. 

Dysfunctional immune system connects gut bacteria and aging. An experiment on fruit flies showed that disrupting the insects’ immune system by knocking out the PGRP-SD gene, which is responsible for the recognition of foreign bacteria and the consequent immune response, resulted in higher levels of Lactobacillus plantarum in the flies’ guts. These bacteria produce lactic acid, and without the immune system effectively purging them, the production of this acid increased substantially, causing tissue aging and reduced lifespans. Increasing the production of PGRP-SD had the opposite effect.

Measuring aging biomarkers with a lab on a chip. Keck Graduate Institute scientists have developed a system called Click-A+Chip, which is a palm-sized testing system capable of measuring changes in blood proteins associated with aging, thereby assessing the decline in tissue health and functionality. This new system promises to be much faster and cheaper than traditional methods.

Klotho mitigates brain inflammaging. Gladstone Institutes scientists found out that the natural decrease in the expression of the klotho gene in the choroid plexi of mice allows inflammatory molecules to more easily reach the brain, increasing brain inflammation and impairing brain functions. The researchers think that klotho depletion might also be to blame for brain inflammaging in humans, and they plan to study whether increasing klotho in the choroid plexus of the brain might help suppress the cognitive decline associated with aging.

The decline of NAD+ with age. A small-scale study recently showed that NAD+ in human plasma tends to decrease with age, though it remains to be seen whether this happens because of increased consumption or decreased production of the molecule.

Boosting A. muciniphila gut bacteria reduces insulin resistance. A study in the journal Science Translational Medicine shows that increasing levels of the bacterium Akkermansia muciniphila helps in reducing insulin resistance in both mice and monkeys. Mounting evidence suggests that these bacteria play a role in the prevention of age-related conditions such as cancer and type 2 diabetes. According to the study, older monkeys showed decreased levels of the bacteria, resulting in lower levels of a protective molecule known as butyrate, thereby contributing to the thinning of the animals’ intestinal walls. The resulting leaky gut eventually leads to higher levels of chronic inflammation and insulin resistance. The scientists think that increasing this bacterium in the human gut might be a viable therapeutic avenue for the prevention of several age-related conditions.

News nuggets

Epigenome editing on LLL. The epigenome series on LLL continues, with part 4 having been published on November 9; the full series can be found here.

Sequencing the genome of your pet. In this article, Neo.Life‘s Grace Rubenstein explains the potential benefits of sequencing the genome of pets—not just for pets themselves, but also for us humans.

CRISPR babies. Recently, Chinese researchers have announced the first CRISPR-edited babies with engineered resilience against HIV. As expected, the announcement has stirred controversy, but it was also a good opportunity to discuss why designer babies might be a good thing after all.

Coming up in December

Project4Awesome 2018. An initiative of the Foundation to Decrease World Suck, Project4Awesome is a contest in which charities from all over the world can win large monetary prizes to carry out their charitable activities. Everyone is invited to produce YouTube videos of any budget to endorse their favorite charities, and viewers’ votes decide the winners—yes, there’s more than one. Lifespan.io will participate as well, so we encourage all our fantastic supporters to muster their artistic talents and make videos to show everyone why you believe in us!

P4A will livestream from December 7 to December 9, and if your video is particularly interesting or well-made, it might be featured on the show! You can submit your creations starting on December 5 at midnight EST until the very end of the livestream at 11:59 AM EST on December 9. 

That was all for November, and a very busy month it was! We wish you and your loved ones a happy holiday season; the roundup will be back for December as well as our yearly roundup for 2018.

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

Would I Want a Designer Baby? CRISPR, Gene-Editing, and You

A couple of days ago, news about the first designer babies has shaken the world. A Chinese researcher, Jiankui He, and his team at the Southern University of Science and Technology of China have been recruiting couples in order to create the world’s first babies with artificially increased resilience to HIV. The embryos were modified using the CRISPR/Cas9 gene editing tool before being implanted in their mother.

According to the research lead, the twins were born healthy a few weeks ago, and genetic testing performed after they were born confirmed that the editing had actually taken place. While the academic community discusses whether it is acceptable or not to modify human genes, taking into account that the changes made will be inherited by these babies’ offspring and are now added to mankind’s genetic pool, I want to share my own views on designer babies.

Are these the first designer babies? Hardly.

First, and most importantly: If by “design”, we mean that the parents have consciously chosen the qualities of their children, then these are not the first designer babies. When cryobanks with human reproductive cells first opened, they offered the parents-to-be the opportunity to choose their donors and thus their future babies’ qualities, including eye color, height, and blood group. As the donors must also have met the requirements of the bank regarding health and age, one can say that the parent-to-be purposefully improved his or her bloodline. In my opinion, babies that were born as a result of in vitro fertilization (IVF) using chosen reproductive material from a cryobank are designer babies.

We should also keep in mind that IVF was developed in 1978, and, since then, couples who could not have children naturally (because the woman is past her reproductive age or the male partner cannot have children), have been messing with nature to allow their genes to stay in the genetic pool. Over 8 million children have, so far, been born thanks to IVF.

Over time, information about the donors became more and more detailed, and, several years ago, potential parents became more able to design their babies’ features thanks to the possibility of sequencing the parents’ genomes, which provides a fairly sharp estimate of what qualities their babies could possibly have. Obviously, parents with severe health problems were the first people to test this innovation.

This technique, called preimplantation genetic diagnosis (PGD), was developed back in the 1990s. The idea is fairly simple: if both parents have hereditary diseases, and there is a high risk that their babies are going to inherit them or an aggravated form of them, then, after IVF, the doctors test the embryos’ DNA and only implant the healthy ones into the prospective mother’s uterus. At this early stage, when the embryo only has 8-16 cells, it is safe to take out one cell to test its DNA; it was proven that this loss does not lead to any deformities or illnesses in the baby.

Right now, most places that provide in vitro fertilization also offer IVF+PGD services, which means that we are already surrounded by designer babies whose features remain in the genetic pool. These little miracles, the completely healthy offspring of two ill parents, don’t make the headlines as often as deserved. The IVF+PGD approach is what I would call a very soft and positive version of eugenics: the improvement of our common genetic pool without any sort of violence.

What is more interesting is that IVF+PGD has begun to be used even by healthy women over 35, who would prefer to exclude the possibility of having a child with chromosome abnormalities. However, as genetic testing allows potential parents to know the child’s features in advance, there is a growing demand for choosing the sex and looks of the offspring. We may expect more and more countries to submit to the invisible hand of the market and allow the parents to decide what their new family member should look like.

One more recent example of designer babies involves three parents. Sometimes, reproductive cells are not good enough to be used for IVF, yet the parents still want the child to be their own with minimal genetic influence from other people. A couple of years ago, a boy was born as a result of using a revolutionary “pronuclear transfer” technique. This method, which is now approved in the UK to provide hope to women suffering from Leigh syndrome, involves a fertilized donor egg with healthy mitochondria (the power plants of the cell, which have their own DNA). As Leigh syndrome lies in mitochondrial DNA defects, the nucleus of the mother’s fertilized egg is put into the donor egg after its nucleus has been removed. As a result, the child has inherited his features from three parents: healthy nuclear DNA from mom and dad and healthy mitochondrial DNA from the donor. If that is not a designer baby, I don’t know what is.

If designer babies are already here, then why is there so much discussion of what Jiankui He has done using CRISPR/Cas9?

One answer is novelty. Novelty is frightening, and this is a perfectly natural reaction, as, indeed, CRISPR is a new technique; we don’t know much about the long-term effects of its use yet, we know that its accuracy is still in development, and we are right to be concerned about the CRISPR-edited babies’ health. However, every new reproductive technique has gone through this stage before becoming publicly approved and massively implemented. The current situation with CRISPR/Cas9 is exactly the same as it was with IVF, IVF+embryo cryopreservation, IVF with donor cells, and IVF+PGD. No one knew the long-term consequences when these techniques were first used, and one could accurately refer to them as “immature” back then.

However, in all of these cases, the researchers had the ultimate goal of improving the lives of the parents and the future child by removing the (harmful) limitations imposed by the natural bloodline. In my view, this goal is perfectly ethical.

An ordinary person’s view on designer babies

To be honest, I am not a big fan of reproduction. I find other things in life more appealing than producing and raising a child. It was this way from the very beginning of my conscious life, and it will hardly change in the foreseeable future, despite the fact that I am almost 40 and know that I am getting closer to the end of my reproductive age. If science finds a way to slow down aging and bring it under medical control, my chances will increase, but there is a little problem.

My genetic layout is far from the best, according to the information I already have about my genome. If I give birth naturally, my children will likely inherit a high risk of developing glaucoma and macular degeneration early in life, and they would be likely to develop gout and a vast variety of connective tissue disorders, including problems with heart valves and rhythm. Do I want for my children the life that I have, which involves constantly monitoring my sight, joints, vessels, and heart, avoiding factors that can let my genetic predisposition manifest itself? I am pretty good at that, which is why I have managed to avoid lots of health problems so far, but I would still want a perfectly healthy baby who would be able to just enjoy life!

Can IVF+PGD be enough to have a healthy baby? Likely not, as my problems involve too many genes at once. Sure, I could seduce a perfectly healthy young man whose genes would compensate for some of the problems, but it still would mean consciously putting my child through my problems, even if their effects were reduced. It also would mean that I would consciously decrease the health of the bloodline of my man, which, to be frank, I find unethical.

My only solutions would be germ-line gene therapies that would affect my reproductive cells and clear my bloodline from heritable disorders or gene-edited embryos controlled by PGD before implantation.

So, would I vote for applying CRISPR/Cas9 editing techniques to human genes? I would. My reasons would include the work that is now done by George Church and other scientific teams around the globe to define the genes that we could modify to achieve healthy life extension. I totally would not mind becoming a GMO or, say, a “designer adult” with improved health and an extended lifespan or my children being born with perfect health and 200-year lifespans (even in the absence of further rejuvenation biotechnologies) either. The colossal efforts I put into engineering a lifestyle that would protect me from the harmful parts of my genetics are not as handy as a few injections of a viral vector with new genes.

It seems that the members of our community think the same way. In a poll on Facebook, I asked if people would prefer a designer baby or an ordinary one, and I received the results that I expected: around 85% of people voted for a designer baby. A few people also noted in the comments that they would like to see more adults genetically edited – starting with themselves – to improve health, lifespan, and mental capacities. The main reason that people voted against the technique was that they did not trust the safety of the current gene editing procedures.

Three reasons for supporting gene editing and even designer babies:

1. WHO’s main goal is to help everyone in all nations in achieving the highest possible standard of health, and it defines health as a state of complete physical, mental and social well-being and not merely the absence of disease or infirmity. Think of that. Perfect health. Can perfect health be compatible with any genetic weaknesses like mine? It can’t. WHO’s bylaws obligate this UN agency to support the development of diagnostic techniques and therapies that can make people healthier, so it is only a matter of time and research before the agency promotes gene editing tools such as CRISPR/Cas9. After all, gene editing is just another tool of evidence-based preventative medicine, which fits into its mission.

2. It looks like it is far cheaper to begin with a healthy embryo from IVF+PGD, or to create one through IVF+CRISPR+PGD, than to provide lifelong treatment to a person born with certain genetic problems or weaknesses. Jiankui He and his team offered $40,000 in medical services for the children born through this procedure. The other costs are not revealed, but we know that an initial cycle of IVF+PGD usually costs around $20k, while the application of CRISPR/Cas9 to target a specific gene can cost another $10k. So, it is highly possible that the overall cost of the procedure was around $70k, which sounds expensive. However, according to the Centers for Disease Control and Prevention, the lifetime treatment cost of an HIV infection is estimated at $379,668 (in 2010 dollars), which can be considered the amount of money saved by preventing an HIV infection. We are not only discussing money; we are talking about avoiding unnecessary human suffering on a large scale. According to WHO, there were around 37 million people with HIV in the world in 2017, and around 60% of them were receiving treatment for it.

3. Nature makes life forms adapt to the environment. The epidemic of AIDS we see now is likely not the first one in human history. This is why the European population already has some share of people resistant to HIV, which inspired the Chinese researchers to edit the corresponding CCR5 gene. Therefore, nature would do the same thing that the researchers are doing now but with a greater number of mistakes and failures (because nature cannot pick one gene and edit it – it must wait for a mutation to accidentally occur and then wait for the carrier of this gene to survive to prove its usefulness), and this process takes millions of years. Does it really make any sense to wait if there is a way to speed up progress?

Conclusion

This procedure still has risks and potential negative consequences for children, and there are still perfectly valid concerns, such as increasing the inequality that can currently be linked to health and intellectual capacity. However, the remedy for these concerns is not to stop medical studies or to close regenerative medicine clinics; instead, it is to work on resolving these issues more actively.

I hope that we have learned our lesson from a setback in gene therapy after the Gelsinger case, in which a young man died during clinical trials of a gene therapy aimed at curing his OTCD, a disease that prevents the body from processing nitrogen in the blood. Years and years of progress in gene therapy development were lost due to the public overreaction that followed. Individual adverse reactions happen, and yes, they are often unpredictable, so no one can make clinical trials absolutely safe. It would not be wise to repeat this mistake by condemning a pioneer just because he had the luxury – or the burden – of actually becoming the first to perform a study.

A Review of Fasting and Calorie Restriction

Today, we want to discuss a review published in the journal Science, as it provides a whistle-stop tour of caloric restriction, intermittent fasting, and time-restricted feeding [1].

There is a considerable amount of data supporting the effects of caloric restriction and similar dietary approaches on both health and lifespan in multiple species. In general, the more simple the organism, the greater the observed effect tends to be, although the effect is less so in longer-lived organisms. For example, caloric restriction has a significant effect on mouse lifespan, but it appears to do little, if anything, to the lifespan of humans.

Why is this? One explanation could be that, as humans, we have already evolved efficient repair systems that more thoroughly address the damages of aging than the repair systems of mice and other short-lived species. In other words, there is little improvement to be made to human repair systems compared to those of mice.

However, that is not to say that caloric restriction is useless for humans, and there is plenty of evidence to support its various benefits to our health. Caloric restriction appears to promote autophagy, a regulated form of cellular garbage disposal that breaks down and recycles unnecessary or dysfunctional cellular components.

It promotes the expression and activity of NRF2, which activates a number of antioxidative and carcinogen-detoxifying enzymes. Periodic cycles of fasting have systemic anti-inflammatory effects and increase progenitor stem cells. Caloric restriction also appears to reduce the activity of the insulin–IGF-1 signaling (IIS) and mTOR pathways, which are metabolic pathways whose excessive activity is associated with aging. There are a myriad of other positive effects, which the review describes in further detail.

The downside is that practicing caloric restriction is challenging, as it requires precision to do correctly while avoiding malnutrition and takes considerable willpower to stick with. Fortunately, the review discusses dietary approaches that may mimic the beneficial effects of caloric restriction.

Abstract

Nutrient composition and caloric intake have traditionally been used to devise optimized diets for various phases of life. Adjustment of meal size and frequency have emerged as powerful tools to ameliorate and postpone the onset of disease and delay aging, whereas periods of fasting, with or without reduced energy intake, can have profound health benefits. The underlying physiological processes involve periodic shifts of metabolic fuel sources, promotion of repair mechanisms, and the optimization of energy utilization for cellular and organismal health. Future research endeavors should be directed to the integration of a balanced nutritious diet with controlled meal size and patterns and periods of fasting to develop better strategies to prevent, postpone, and treat the socioeconomic burden of chronic diseases associated with aging.

Conclusion

The bottom line is while such dietary approaches may do little, if anything, for human lifespan, they can almost certainly influence healthspan. Alongside exercise, caloric restriction and similar mimetics may represent cost-effective ways to remain healthy for as long as possible, making them worth considering.

Although the first versions of some technologies that can effectively manage the aging processes are already in human trials, most of them are still a decade or two away; therefore, if you are serious about maximizing your chances of living long enough to enjoy a full suite of rejuvenation therapies, it might be wise to consider dietary practices that offer the benefits of caloric restriction.

You can learn more by checking out our topic on fasting and how it works.

Literature

[1] Di Francesco, A., Di Germanio, C., Bernier, M., & De Cabo, R. (2018). A time to fast. Science362(6416), 770-775.

Too much mTOR is Linked to Diabetes and Aging

A new study takes a look at the relationship between metabolism, aging, and type 2 diabetes and in particular the mTORC1 protein complex, part of the mTOR pathway.

The mTOR pathway

The mechanistic target of rapamycin (mTOR) pathway is a major part of metabolism and is one of four major pathways that control it; collectively, the four pathways are part of deregulated nutrient sensing, which is one of the aging processes.

The mTOR pathway includes two distinct protein complexes: mTORC1 and mTORC2. The pathway senses amino acids and is associated with nutrient abundance. It is a kinase, which means that it adds phosphates to molecules. mTOR is a master regulator of anabolic metabolism, the process of creating new proteins and tissues.

Studies show that less mTOR activity increases lifespan in various species, including mice, yeast, worms, and flies. Think of high mTOR activity being an analog of the phrase “Live fast, die young”, because too much activity is good for growth but bad for lifespan. However, too little mTOR activity is not beneficial either and can disrupt healing and insulin sensitivity and can cause cataracts in mouse models [1].

mTOR has been well studied in the last few years, particularly for its role in caloric restriction. Inhibiting mTOR has been tried as an approach to emulate some of the positives of caloric restriction, particularly the mTORC1 pathway, which appears to be the better of the two complexes to target. Targeting mTORC1 increases healthy lifespan in mice due to the activation of stress response mechanisms and enhanced autophagy (the recycling of unwanted cellular components).

The study sees researchers looking at mTORC1 and its role as a regulator of metabolism and its relationship to aging and type 2 diabetes [2].

Abstract Type 2 Diabetes Mellitus (T2DM), a worldwide epidemics, is a progressive disease initially developing an insulin resistant state, with manifest pancreatic beta islet overwork and hyperinsulinemia. As the disease progresses, pancreatic β cells are overwhelmed and fails in their capacity to compensate insulin resistance. In addition, it is usually associated with other metabolic diseases such as hyperlipidemia, obesity and the metabolic syndrome. During the progression to T2DM there is a chronic activation of mTORC1 signaling pathway, which induces aging and acts as an endogenous inhibitor of autophagy. The complex 1 of mTOR (mTORC1) controls cell proliferation, cell growth as well as metabolism in a variety of cell types through a complex signaling network. Autophagy is involved in the recycling of cellular components for energy generation under nutrient deprivation, and serves as a complementary degradation system to the ubiquitin-proteasome pathway. Autophagy represents a protective mechanism for different cell types, including pancreatic β cells, and potentiates β cell survival across the progression to T2DM. Here, we focus our attention on the chronic overactivation of mTORC1 signaling pathway in β islets from pre-diabetics patients, making these cells more prone to trigger apoptosis upon several cellular stressors and allowing the progression from prediabetes to type 2 diabetes status.

Conclusion

In both aging and type 2 diabetes, the level of mTORC1 activity is elevated and drives both the disease and one of the aging processes. This suggests that approaches that reduce excessive mTORC1 activity may be beneficial for healthy longevity and for combating metabolic diseases such as type 2 diabetes.

Literature

[1] López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M., & Kroemer, G. (2013). The hallmarks of aging. Cell, 153(6), 1194-1217.

[2] Guillén, C., & Benito, M. (2018). mTORC1 overactivation as a key aging factor in the progression to type 2 Diabetes Mellitus. Frontiers in endocrinology, 9, 621.