×

The Blog

Building a Future Free of Age-Related Disease

Exercise is Currently the Best Way to Slow Down Aging

We have all heard that exercise is good for our health. However, it can not only keep you healthy, it can also slow down some aspects of aging. Some researchers even think that it might be possible to use this knowledge to develop new therapies against aging. While waiting for that to happen, we need to exercise in order to slow down the effects of aging.

How important is it to keep fit?

So, how beneficial is exercising? Well, one of the best studies conducted on this subject showed that women will live 5.6 years longer and men 6.2 years longer if they exercise between 1 and 2.5 hours per week [1]. This makes exercise a better lifestyle choice than any other, at least as long as you’re not counting avoiding downright dangerous behavior, such as smoking.

The main benefits of physical activity may come from better health for the heart. Exercise lessens the risk for many types of heart disease [2]. It is even more beneficial for people who already suffered age-related conditions, including stroke and coronary heart disease, and it is more effective than any known drug in preventing repeated episodes [3].

The key improvements also include increased muscle strength, stronger bones, better weight control, and improved cognitive function. This means less risk of age-related diseases, such as Alzheimer’s disease, as well as lethal falls, which are a major risk for the elderly.

The conclusion is that exercise helps with a lot of different aspects of your health in several ways, and we can summarize its effects as improving quality of life and increasing healthspan.

How much exercise do you need?

More exercise does not always improve outcomes. Professional athletes exercise more than the rest of us, and they generally live longer than the average person [4]. However, correlation isn’t causation, and robust individuals are perhaps more likely to become athletes, instead of the other way around. There could even be negative effects from too much exercise, although that is far from certain.

Even moderate exercise leads to better health. Half an hour a day seems to be enough to see positive effects, and it is also a common recommendation for the minimum amount of exercise you should get. The biggest difference can be seen between people who hardly move around at all and people who get at least a little exercise a few times a week. Taking the stairs and walking short distances is clearly better than nothing. In fact, some studies show that even light activity, such as housework, can have an effect on mortality risk.

It should also be noted that there are different types of exercise and that these could have different benefits. Jogging increases your aerobic ability, which should, among other things, lead to better heart health. Lifting weights is an anaerobic exercise that improves strength and should bring other benefits, such as stronger bones. A lot of research about this has been done already, but so far, we don’t definitively know the optimal amount and type of training for each particular type of person.

Drugs to mimic exercise

Some of the positive effects have to do with the anti-inflammatory processes that occur when exercising [5]. Other mechanisms appear to be involved, although more research on these mechanisms is needed.

Since the advantages of exercise are clear, the idea has occurred to some researchers that it may be possible to mimic the effects of exercise without doing the hard work and getting sweaty. Research is now being conducted using drugs that target the same mechanisms to try to get the same benefits of exercise.

This typically involves adjusting a part of the human metabolism, which is not an easy matter. However, there have been at least some tentative breakthroughs already, and last year, a team found a drug that boosted the endurance of mice by roughly 70 percent [6]. Where this might lead in the future is not clear, but some positive effects may come from this research.

Will exercise lead to longevity?

Even though exercise is beneficial for your health, there is no guarantee it will keep you alive until you reach 100, although staying fit will almost certainly improve your chances. This is why if we want to remain in good health and live longer, we need to develop rejuvenation biotechnology and therapies that address the aging processes directly. That said, if you want to increase your chances of living long enough to see these therapies arrive, then exercise is the best option you have right now.

Literature

[1] Schnohr, P., Lange, P., Scharling, H., & Jensen, J. S. (2006). Long-term physical activity in leisure time and mortality from coronary heart disease, stroke, respiratory diseases, and cancer. The Copenhagen City Heart Study. European Journal of Cardiovascular Prevention & Rehabilitation, 13(2), 173-179. [2] Jakovljevic, D. G. (2017). Physical activity and cardiovascular aging: Physiological and molecular insights. Experimental Gerontology. [3] Naci, H., & Ioannidis, J. P. (2013). Comparative effectiveness of exercise and drug interventions on mortality outcomes: metaepidemiological study. Bmj, 347, f5577. [4] Lemez, S., & Baker, J. (2015). Do elite athletes live longer? a systematic review of mortality and longevity in elite athletes. Sports medicine-open, 1(1), 16. [5] Fan, W., Waizenegger, W., Lin, C. S., Sorrentino, V., He, M. X., Wall, C. E., … & Auwerx, J. (2017). PPARδ Promotes Running Endurance by Preserving Glucose. Cell Metabolism, 25(5), 1186-1193. [6] Dimitrov, S., Hulteng, E., & Hong, S. (2017). Inflammation and exercise: Inhibition of monocytic intracellular TNF production by acute exercise via β 2-adrenergic activation. Brain, behavior, and immunity, 61, 60-68.

Obesity Found to Cause Lasting Damage to Hematopoietic Stem Cells

You might think that being overweight may cause you health problems only until you finally shed those extra pounds, at which point everything will be fine again. It sounds reasonable, but it might be not so easy. According to a new study published in the Journal of Experimental Medicine on December 27, the negative effects of obesity might last even after that extra weight has been lost.

Yes, it appears that being overweight can have lasting consequences that persist long after the weight comes off. Damage to the hematopoietic stem cells can be caused by being overweight, making it all the more reason to try to maintain a healthy weight as part of your personal health and longevity strategy.

What the study found out

In a nutshell, the study shows that the oxidative stress induced by obesity causes the overexpression of the transcription factor Gfi1 in hematopoietic stem cells (HSCs), pushing them to produce myelocytes that, if you’re out of luck, may well end up becoming preleukemic as well[1].

As if this wasn’t bad enough, the overexpression may persist even if affected HSCs are then transplanted into a normal environment. This means that the damage has a lasting and long-term effect, and even restoring a healthy environment does not fully rectify the damage done.

To explain further, oxidative stress is basically an imbalance between reactive oxygen species (ROS) and the body’s ability to clear them out. ROS are oxygen-containing molecules that, like their name implies, are extremely reactive. Even though ROS have normal, beneficial functions in the body as well, because of their high reactivity, they often end up reacting with molecules better left alone, damaging them in the process or causing them to do things they shouldn’t do.

Obesity tends to increase the body’s oxidative stress, and as the study found out, this causes the production of too much Gf1 in hematopoietic stem cells. As said, Gfi1 is a transcription factor—that is, a protein controlling the rate at which certain genetic information contained in DNA is transcribed into RNA and ultimately translated into specific proteins with a certain job.

Too much Gf1 expression in hematopoietic stem cells—that is, stem cells that “produce” blood and immune cells—will skew the production towards myelocytes, white blood cells that are normally found in bone marrow but appear in circulating blood as a consequence of diseases such as myelogenous leukemia.

Of mice and men

In the mouse model the researchers used in their study, the negative effects of obesity on HSCs lasted even after they were transplanted into a non-obese mouse. This doesn’t necessarily mean that we would observe the exact same results in obese human patients, but it does suggest that using damaged HSCs from obese human donors may present some risk.

The next step will be to determine if human HSCs suffer the same fate as those in mice. It is worth noting that humans have more robust repair systems that allow us to resist and repair ROS damage better than mice. It may be the case that human HSCs could be more resistant to such damage and that you may need to be overweight for a longer time before significant damage occurs.

Conclusion

Naturally, as prevention is always better than cure, you should try your best to maintain a healthy weight and spare yourself—and other people, should you ever be a HSC donor—obesity-induced troubles. Even if human HSCs are more resistant to such long-term damage, there are a myriad of other reasons why maintaining a healthy weight is a good idea for your health and longevity.

Literature

[1] Lee, J. M., Govindarajah, V., Goddard, B., Hinge, A., Muench, D. E., Filippi, M. D., … & Reynaud, D. (2017). Obesity alters the long-term fitness of the hematopoietic stem cell compartment through modulation of Gfi1 expression. Journal of Experimental Medicine, jem-20170690.

The Gut Microbiota Could Contribute to Sarcopenia

We have all seen it, the age-related loss of muscle mass and increasing frailty that generally accompanies advancing age. Recently the World Health Organization classified this age-related muscle wastage as a disease and thus sarcopenia entered official usage to describe it. There are a number of potential causes of sarcopenia and new research suggests that there is a nutritional link between the microbiota and development of the condition.

What is sarcopenia?

Sarcopenia is the condition that causes the familiar, age-related loss of muscle strength and mass in older people, and it leads to ever-increasing frailty. Frailty makes everyday tasks difficult, affects balance, and can lead to falls, which can be very dangerous for older people.

How sarcopenia develops is a more complex matter; there are a number of possible causes for sarcopenia, including reduced protein intake, sedentary lifestyle, chronic inflammation that inhibits cell function and tissue repair, reduced stem cell activity from the same inflammatory signals, mitochondrial dysfunction, and reduced oxygenation resulting from deteriorating blood vessels.

These are only some of the possible factors involved in the onset of sarcopenia, and more research is needed to determine the exact cause, although it currently appears that reduced stem cell activity is a strong contender.

Could the gut influence sarcopenia?

A new study has researchers considering that age-related changes of the gut microbiota may possibly contribute to sarcopenia[1]. In recent years, the role of the gut microbiota in aging has increasingly become the focus of research. In this paper, the researchers focus on age-related changes in these populations of gut microbes in an attempt to explain the link between nutrition and sarcopenia.

There is certainly ample evidence to suggest that changes in the gut microbiota can promote inflammation and contribute to the smoldering chronic inflammation known as “inflammaging”. It may be the case that changes to the microbiota promote inflammation, which inhibits stem cell activity and thus provokes sarcopenia; we know that inflammatory signals do inhibit stem cells, so this is certainly plausible.

Conclusion

The interaction of the gut microbiota with the aging processes and other metabolic systems is intriguing. However, the interplay of the gut microbiome with our biology is highly complex, so it remains to be seen if anything useful will come from this particular line of research in the near future.

As ever, our interest lies in research that focuses on the root causes of aging and has the potential to prevent sarcopenia at its root. A repair-based approach to aging, as proposed in the Hallmarks of Aging and the SENS model, is what we believe could lead to the most significant increases of healthy longevity, making these approaches the ones we consider most worthy of development and funding. That said, research such as this is still intriguing and adds to our scientific understanding, and that is a never a bad thing.

Literature

[1] Ticinesi, A., Lauretani, F., Milani, C., Nouvenne, A., Tana, C., Del Rio, D., … & Meschi, T. (2017). Aging Gut Microbiota at the Cross-Road between Nutrition, Physical Frailty, and Sarcopenia: Is There a Gut–Muscle Axis?. Nutrients, 9(12), 1303.

Stem Cell Trials For Osteoarthritis Repair Cartilage

Mesenchymal stem cells (MSCs) are one of the most thoroughly studied and understood stem cell types. They are used in a wide range of therapies, and the many studies using MSCs have enjoyed varied levels of success, depending on delivery methods, patients, co-therapies and other factors.

Today, we will be taking a look at MSCs and a new human clinical trial focused on treating osteoarthritis, an age-related inflammatory condition that leads to the breakdown of bone and cartilage.

What are mesenchymal stem cells?

Mesenchymal stem cells are multipotent cells, which means that they can differentiate (transform) into a variety of cell types, including osteoblasts (bone cells), chondrocytes (cartilage cells), myocytes (muscle cells) and adipocytes (fat cells)[1]. Sources of MSCs are varied and include bone marrow, which is the original source of MSCs and still the most frequently used; umbilical cord tissue; adipose tissue; amniotic fluid; and molar cells.

MSCs have been used to treat multiple joint and musculoskeletal problems with the goals of reducing inflammation and promoting tissue regeneration. In general, the transplanted MSCs do not tend to survive long in the body; however, they last long enough in most cases to generate anti-inflammatory signals and promote tissue regeneration.

MSCs to treat osteoarthritis in human trials

Today, we wanted to bring your attention to a small open access study that used MSCs to treat age-related osteoarthritis, a common complaint for many older people[2]. This condition is caused by localized inflammation in the joints, leading to the breakdown of bone and cartilage and painful and potentially crippling outcomes for the patient.

The presence of proinflammatory senescent cells are very likely a major factor in how this condition develops, and it will be important to see how senolytic therapies that remove these cells might ameliorate osteoarthritis. It would potentially be even more interesting to see how MSCs and senolytics used as a co-therapy might ameliorate osteoarthritis, given that reducing inflammation enhances tissue regeneration in stem cell therapies.

The study here is also different from the majority of previous studies, as it tracked patient outcomes for two years following MSC therapy, a considerably longer period than other studies. This is important because we need to know more about the long-term effects of these therapies in order to refine and optimize the approaches used. This study was a phase 1 and 2 human clinical trial which essentially means that this approach is both safe and effective at combating osteoarthritis. If you want to you can learn more, check out our topic that explains clinical trial phases.

Conclusion

Mesenchymal stem cell therapies do not address the root cause of osteoarthritis, which is chronic inflammation from senescent cells, cell debris, microbial burden and other sources, but they do locally suppress inflammation long enough to promote tissue repair.

The study results here show that there was a significant improvement in patients with osteoarthritis. Despite the limited number of patients in the study, the therapy was shown to safely increase knee cartilage thickness; this demonstrates its ability to improve the function and structure of joints and shows that it could be an effective therapy for osteoarthritis patients.

Literature

[1] Nardi, N. B., & da Silva Meirelles, L. (2008). Mesenchymal stem cells: isolation, in vitro expansion and characterization. In Stem cells (pp. 249-282). Springer Berlin Heidelberg.

[2] Al-Najar, M., Khalil, H., Al-Ajlouni, J., Al-Antary, E., Hamdan, M., Rahmeh, R., … & Al-jabbari, E. (2017). Intra-articular injection of expanded autologous bone marrow mesenchymal cells in moderate and severe knee osteoarthritis is safe: a phase I/II study. Journal of orthopaedic surgery and research, 12(1), 190.

 

People Support Living Longer If It Means Remaining Healthy

A number of studies in different countries show that when people are asked “how long would you like to live?”, they respond with a figure equal to or slightly higher than the current life expectancy in a given country [1-4]. So, why does the public often lack enthusiasm for longevity?

These studies have shown that, generally, the public is uninterested in living longer than normal because they believe that these extra years will be spent suffering from the illnesses of old age. This is why the public often reacts to words like ‘longevity’ this way; to them, ten extra years likely means a decade spent in a wheelchair or some other decrepit state robbed of independence and health.

Asking the right question is important if you want the right answer

When asking people about living longer, it is essential to frame the question properly so that a considered response with all the facts can be made. When talking about longevity, it is critical to ask the right question and use the right language.

In a recent study, when it was made clear that continued physical and mental health would accompany these extra years, the responses were generally more favorable towards living longer [5]. In this study in particular, people even changed their responses to support longer life when it was made clear that continued mental and physical health would be part of that extra time.

We surveyed 1000 individuals (through “Ask Your Target Market”) about how long they wished to live (to age 85, 120, 150, or indefinitely), under 3 scenarios: (1) sustained mental and physical youthfulness, (2) mental youthfulness only, (3) physical youthfulness only. While responses to the two partial youthfulness conditions recapitulated the results of previous surveys (Cicirelli, 2011; Kogan et al., 2011; Partridge et al., 2011; Duncan, 2012; Pew Research Center, 2013), i.e., most responders (65.3%) wished to live to age 85 only—under scenario (1) the pattern of responses was completely different. When guaranteed mental and physical health, 797 of 1000 people wanted to live to 120 or longer, and 53.1% of the 797 desired unlimited life spans. Furthermore, 70.1% of the people who responded 85 to scenario (2) or (3) changed their answer to 120 or longer in scenario (1).

A new study once again suggests asking the right question is critical

Today, we see yet more vindication of this in a new study by researchers from the University of Kansas[6]. The team asked ninety elders in China, Germany, and the U.S. about their ideal lifespans.

A larger number of respondents did mention they wanted to extend their lives. Yet less than half of that group noted a specific amount of time they desired to live.

The strongest opinion among that group was the desire to live longer only if they maintained their current or what they deemed to be acceptable levels of health.

Source: KU

Conclusion

Once again, we can see that continued health is very much a proviso of continued longevity. This is a perfectly understandable reaction, and it is fortunate that rejuvenation biotechnology has the same goal: to prevent age-related diseases by targeting the aging processes, thus offering people longer and healthier lives.

Ultimately, the right question to ask is “Would you want to live longer if your continued health is guaranteed for those additional years?” People are generally more supportive of the idea if they know that those extra years will be good years, and most of us really want to be healthy enough to enjoy life.

Literature

[1] Lang, F. R., Baltes, P. B., & Wagner, G. G. (2007). Desired lifetime and end-of-life desires across adulthood from 20 to 90: A dual-source information model. The Journals of Gerontology Series B: Psychological Sciences and Social Sciences, 62(5), P268-P276.

[2] Lugo, L., Cooperman, A., & Funk, C. (2013). Living to 120 and Beyond: Americans’ Views on Aging, Medical Advances, and Radical Life Extension. Pew Research Center, August, 6. URL: https://www.pewforum.org/2013/08/06/living-to-120-and-beyond-americans-views-on-aging-medical-advances-and-radical-life-extension/

[3] CARP Radical Life Extension Poll Report (2013). URL: https://www.carp.ca/wp-content/uploads/2013/09/Life-Extension-Poll-Report.pdf

[4] Financial University of the Government of the Russian Federation, Sociology Faculty (2015). Most of Russians want to live up to 80 years only. (Bol’shinstvo rossijan hochet dozhit’ tol’ko do 80 let).

[5] Donner, Y., Fortney, K., Calimport, S. R., Pfleger, K., Shah, M., & Betts-LaCroix, J. (2016). Great desire for extended life and health amongst the American public. Frontiers in genetics, 6, 353.

[6] Ekerdt, D. J., Koss, C. S., Li, A., Münch, A., Lessenich, S., & Fung, H. H. (2017). Is longevity a value for older adults?. Journal of Aging Studies, 43, 46-52.

Ending Aging – A Book Review

The book I’m reviewing today is pretty much a classic by now. Published for the first time in 2007, “Ending Aging” by Dr. Aubrey de Grey is not only “an audacious blueprint for cheating the reaper”—as Janet Cromley of the Los Angeles Times put it—but also a manifesto, a call to arms, if you will, openly denouncing biological aging as public enemy number one and explaining why and how we should and could put an end to it.

Dr. de Grey’s enthusiasm and confidence are often regarded with skepticism by people who are new to the idea of defeating aging, and while initially the scientific community wasn’t persuaded that the SENS “maintenance approach” was even worth discussing, this paradigm has been confirmed by other academics and is shaping the rejuvenation biotechnologies of tomorrow.

Like every rejuvenation advocate eventually learns, a conversation on the defeat of aging with a newbie is composed of roughly three parts: explaining why aging is a problem, explaining what aging is and how we may conquer it, and finally, addressing the inevitable concerns of the audience. “Ending Aging” does a great job of the first two parts, though it dedicates only little to the third one, replacing it with a more pragmatic discussion of what needs to be done, in terms of advocacy and policy-making, in order to get from here to a world free of age-related diseases. Dr. de Grey normally addresses typical concerns about extended longevity with two general answers without going into the finer points of each specific concern, as he understandably believes that eliminating diseases and saving lives is an absolute priority and no excuses can justify delayed action to address it—or worse, failing to address it altogether.

My personal feeling is that the book might have benefited from even a brief section dedicated specifically to the details of the most common concerns; as it is, “Ending Aging” may well persuade readers that defeating aging is feasible and desirable, but unaddressed worries of some people might deter them from wholeheartedly joining or supporting the cause. Then again, I understand why Dr. de Grey might prefer dedicating more attention to the scientific and technical aspects of the issue and leaving the discussion of related but peripheral matters for more appropriate venues.

Although the book presumes little to no knowledge of biology and goes to great lengths in attempting to make the topic accessible to everyone, its tone is never condescending; rather, it’s friendly, engaging, and peppered with humor, making the readers feel as though they were all part of a single team trying to take aging down—which reflects de Grey’s conviction that only with sufficiently widespread support across the globe can the crusade against aging eventually succeed.

Being aimed at non-specialists, “Ending Aging” is rather non-technical, although the second part—which describes SENS’s seven damage categories and the proposed regenerative interventions against them—can be difficult to get through. Simplified as it may be, it is full of details, and there’s a lot going on that the reader will need to keep track of; I can tell from experience that a second readthrough is very helpful. Readers with strong biology backgrounds will probably wish to investigate further, and they may do so via the extensive references provided at the end of the book or by getting in touch with de Grey himself—which he explicitly invites readers to do, should they have any questions.

While “Ending Aging” is not the first book discussing medical advancements that may spell the end of aging as we know it, to my knowledge, it is the only one so overtly endeavoring to change the public’s perception of defeating age-related diseases from a purely scientific quest fueled by vanity into a global, humanitarian cause. Dr. de Grey correctly identifies the current, skewed perception of aging as something necessary and even desirable and enjoyable as the biggest roadblock on the way to a healthier world, and his book is only part of his efforts to remove this roadblock through advocacy.

Personally, I believe that, without Dr. de Grey’s work, the popularization of rejuvenation biotechnologies might well have been slowed down by decades. In particular, it was this very book that pushed me to become an advocate and grow an interest in biology; for this reason, I think it’s a must-read for every supporter of the cause and a recommended read for just about anyone who’d rather be healthy than sick at any age.

The Best of the SENS AMA

Dr. Aubrey de Grey from the SENS Research Foundation (SRF) did an Ask Me Anything on Reddit on December 7th, and there were many great questions and answers; we thought it would be a great time to summarize some of the best ones and offer a little commentary.

What do you think were the biggest wins of the last couple of years in SENS-relevant advocacy, research, and development? What has moved the needle?

There have been lots. On the research, I would highlight our paper in Science two years ago, which shows how to synthesize glucosepane, and our paper in Nucleic Acids Research one year ago, which shows simultaneous allotopic expression of two of the 13 mitochondrial genes. Both of those projects have been greatly accelerated in the meantime as a result of those key enabling breakthroughs; watch this space.

On advocacy, I think the main win has been the arrival of private capital; I would especially highlight Jim Mellon and his Juvenescence initiative because he is not only a successful, energetic and visionary investor, he is also a highly vocal giver of investment advice.

We are pleased to have been involved with the second project mentioned here, as we hosted the MitoSENS project at Lifespan.io, where it raised 153% of its initial fundraising goal. Less than a year later, after raising this money, it went on to publish the groundbreaking study showing that backup copies of mitochondrial genes could indeed be created in the nucleus. Dr. de Grey originally proposed the idea over a decade ago amid much scepticism; it is really good to see that years later he has been vindicated. This is the power of crowdfunding and how we as a community can make big changes in science by working together.

How do you feel about the impact of groups like LEAF advocating and reporting on rejuvenation biotech? Has the advocacy and reporting of these groups made your life any easier?

Massively! A huge thing that I say all the time is that advocacy absolutely relies upon the diversity of its messengers. Different people listen to different forms of words, different styles of messaging, etc. The more, the better.

It’s good to know that our work is appreciated and helping. Working together as a community is essential for progress, so it was nice to see this question and response from someone we respect a great deal.

We have said many times before effective advocacy efforts are just as important as the research itself. Professional advocacy has the potential to increase public support and funding, paving the way for the arrival of rejuvenation biotechnology. In the past decade or so, advocacy has mostly been left to volunteers and people such as Dr. de Grey.

Popular causes attract celebrities, public support, funding and investment; if we want a revolution in medicine and how we treat aging, then we must popularize the movement. There has been a serious shortage of full-time and organized advocacy; therefore, we decided to create LEAF to support groups like the SRF, advocate to popularize the cause, and help to raise much-needed funds for research efforts. We are only able to do this thanks to the support of the community, and we are extremely grateful to our Lifespan Heroes for helping us to do the work we do.

Aside from funding, what do you consider to be a burden or delay for your type of research?

Nothing. Seriously, nothing at all. We have the plan, and we have the people. It’s all about enabling those people by giving them the resources to get on with the job.

Indeed, funding for research is one of the four major bottlenecks slowing down the development of therapies that address the aging processes. The more funding the field gets, the more projects can be launched, the sooner breakthroughs can potentially happen, and the greater the benefits will likely be for all of us.

Is there anything new you are able to say about the breaking of cross-links in the extracellular matrix?

Absolutely. Short story, we now have a bunch of glucosepane-breaking enzymes, and we are within a few months of spinning the work out into a startup.

A suspected cause of degenerative aging is the accumulation of sugary metabolic wastes known as advanced glycation end-products (AGEs). These are wastes that are, in some cases, hard for our metabolism to break down fast enough or even at all. Some types, such as glucosepane, can form cross-links, gumming together important proteins such as those making up the supporting extracellular matrix scaffold.

The properties of elastic tissues (skin and the blood vessel walls) derive from the particular structure of the extracellular matrix, and cross-links degrade that structure, preventing it from functioning correctly. AGEs’ presence contributes to blood vessel stiffening with age, and it is implicated in hypertension and diabetes.

That SRF now has candidate enzymes is very significant because it means that there are now potential ways to remove these crosslinks from our tissues. There are many types of crosslinks, and we already know of compounds and drugs that can break other kind of crosslinks; the major problem is glucosepane, as it lasts a very long time, and, so far, nothing is known to remove it. Given that other types of crosslinks can be removed, Dr. de Grey rightly thought that there must be ways to remove (cleave) glucosepane from tissues; now, it seems that we are a step closer to that potentially becoming a reality.

If the SRF is successful in finding ways to break glucosepane crosslinks, this has huge implications for diabetes, hypertension and aging. It is great to hear that the organization is now reaching the point at which it is almost time to develop this as a therapy by creating a startup company.

It seems likely that artificial intelligence will be a necessary tool in order to reach longevity escape velocity. I was wondering how much of a role does artificial intelligence play in your research? Is this something you devote many resources to?

We don’t, but that is because other major players in this field (and good friends of mine), such as Alex Zhavoronkov and Kristen Fortney, are doing it so well already (with Insilico Med and BioAge, respectively). Check out the BioData West conference that will occur in SF a couple of days before our Undoing Aging conference in Berlin; I will be chairing a session on this.

We believe that the application of AI and, in particular, machine learning will prove to be a very valuable tool for research in the coming years. Such systems are ideally suited for high-throughput, laborious tasks that also require high attention to detail and would take humans a long time to do. Drug discovery, image analysis and many more tasks in the lab could potentially be automated, saving time and freeing up researchers to work on other critical tasks.

We are proud to have hosted the MouseAge project this year, which is an AI-based visual aging biomarker application that helps researchers determine the age of mice without the use of harmful tests. In a few months, researchers will be able to use the MouseAge application in the lab to help speed research progress up. This is just one example of how AI can be used in aging research and how the community helped to make it happen.

Given current funding, how far away from robust mouse rejuvenation do you think you are?

My estimate is 5-7 years, but that’s not quite “given current funding”. My overoptimism in saying “10 years” 13 years ago consisted entirely of overoptimism about funding – the science itself has not thrown up any nasty surprises whatsoever – but, nonetheless, I am quite optimistic as of now about funding, simply because the progress we have made has led to a whole new world of startups (including spinoffs from the SENS Research Foundation) and investors, so it’s not only philanthropy anymore. Plus, the increase in overall credibility of the approach is also helping to nurture the philanthropic side. We are still struggling, that’s for sure, but I’m feeling a lot surer that the funding drought’s days are numbered than I felt even two or three years ago.

Robust mouse rejuvenation (RMR) has long been a goal for the SENS Research Foundation, going back to when the SENS approach was initially proposed. RMR was originally outlined as being able to demonstrate and replicate SENS to double the remaining life expectancy of an already aged mouse. This would not mean the first RMR would be a total implementation of all the SENS approaches or that rejuvenation would need to be absolute; it would be a first pass to demonstrate the viability of multiple SENS approaches combined to produce robust results.

Being able to achieve a first-pass RMR could do much to convince academia that the repair approach to aging is plausible and attract more funding and interest in the approach. While RMR working in mice may not sound that exciting, it has huge implications for the field and potentially the rate of funding and progress.

How confident are you still in your previous prediction that humans will be able to control aging by 2029?

I think we’ve slipped a few years, entirely because of lack of funding. The tipping point will be when results in mice convince a critical mass of my curmudgeonly, reputation-protecting expert colleagues that rejuvenation will eventually work, such that they start to feel able to say so publicly. I think that’s on the order of five years away.

We think that the tipping point could well be if senolytics have the same result in humans as they have in mice. Enhanced tissue repair and regeneration in older people would be a very strong case for the repair approach to aging and almost certain to convince the academics sitting on the fence.

Certainly, if AGE breakers could be demonstrated to work in humans, this would also go a long way towards not only convincing academia but also grabbing public interest. Removing AGEs from the skin may potentially reverse wrinkles, for example, and restore skin elasticity, offering a very visual demonstration of repair being plausible.

There is almost certainly going to be a tipping point at which the bulk of academic and public support swings in favour of a repair approach to aging; the only question is when? Well, the sooner the basic science can be done and moved to translational research, the sooner we can all potentially benefit from these technologies. This makes supporting both the research and advocacy of rejuvenation biotechnology very important for progress.

Boosting Mitochondrial Quality Control to Combat Alzheimers

Scientists at the École polytechnique fédérale de Lausanne (EPFL) have found a way to make mitochondria more resistant to damage, which could potentially be used to halt Alzheimer’s and other, similar, diseases.

Globally, Alzheimer’s disease is the most common form of dementia and cause of neurodegeneration. It causes brain damage and symptoms such as long-term memory loss. It is an amyloid-based disease, with the characteristic hallmark being the formation of toxic plaques in the brain made from the aggregated beta-amyloid inside the neurons.

Improving mitochondrial quality control

Diseases like Alzheimer’s are the result of the loss of proteostasis, which leads to the creation of sticky misfolded proteins, which form plaques by bonding together. There is still no cure for the disease, and it causes tremendous suffering while posing a considerable burden on the public health system. Many treatments have focused on trying to reduce the formation of the plaques, but, as of yet, the results are inconclusive. One group of researchers has tried a different approach and is looking at Alzheimer’s as a metabolic disease.

The new study focused on the mitochondria, the powerhouses of the cell that produce the energy needed for cellular functions central to metabolism[1]. The research team showed that in worms and mice, they could make the mitochondria more robust and resistant to a particular form of protein stress, allowing them to better protect themselves and, more importantly, reduce the accumulation of amyloid plaques.

During the aging process, cells are exposed to increased levels of damage, and that includes damage to their mitochondria. Damaged mitochondria become dysfunctional, and cells normally replace them via a process known as mitophagy, a selective degradation of the mitochondria by autophagy.

Unfortunately, over time, our cells become increasingly poor at removing these problem mitochondria, thus leaving them vulnerable to Alzheimer’s disease.

The researchers identified two key processes that act as a quality control system for the mitochondria. First, there is the process of mitophagy, which recycles damaged mitochondria, and second, there is the mitochondrial unfolded protein response (UPRmt), which shields mitochondria from stress. Taken together, these processes are one possible solution to reducing mitochondrial damage and could help combat diseases like Alzheimer’s.

Better recycling improves cognition

It has been known for some time that the mitochondria are dysfunctional in the brains of Alzheimer’s patients, but this is the first evidence suggesting that the disease might be combated by boosting these quality control systems. These two processes are present in C.elegans flatworms, mice, and humans, so worm and mouse research could translate well into human beings. The researchers decided to try activating these systems using a drug to see if they could boost protection and quality control in the mitochondria.

They used the antibiotic doxycycline and the vitamin nicotinamide riboside (NR), both known to activate the UPRmt and mitophagy systems in worm models of Alzheimer’s. They noticed that the health and lifespan of treated worms increased dramatically compared with untreated controls, and the formation of plaques was also significantly reduced. Intriguingly, they also observed similar improvements when they activated the same two systems in human neuronal cells in culture using the same two drugs.

Finally, the researchers tested NR in a mouse model of Alzheimer’s disease. They observed the same improvement to mitochondrial function and reduction of plaques as seen in the flatworms. Perhaps most importantly, the researchers noted a dramatic improvement in the cognitive function of the mice.

Conclusion

The usual caveats apply; the results are only so far confirmed in flatworms, Alzheimer’s model mice and human cells in culture. However, the two target quality control systems are present in all three species and can be activated in the same way, which leaves some room for optimism. Will it prove to be more effective than the direct removal of amyloids? Or could it even be a co-therapy for those other approaches? Only time and a clinical trial will tell.

Literature

[1] Vincenzo Sorrentino, Mario Romani, Laurent Mouchiroud, John S. Beck, Hongbo Zhang, Davide D’ Amico, Norman Moullan, Francesca Potenza, Adrien W. Schmid, Solène Rietsch, Scott E. Counts, Johan Auwerx.Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature 06 December 2017. DOI: 10.1038/nature25143.

SENS: Progress in the Fight Against Age-Related Diseases

Given that there is to be a Reddit AMA on December 7th with Dr. Aubrey de Grey in the Futurology subreddit, we think it’s a great time to have a look at the progress that the SENS Research Foundation has made in tackling the aging processes. What follows is a brief summary of some of the highlights of their research efforts as well as the details of the AMA, in which you can ask Aubrey anything you like about his work.

Today, there are many drugs and therapies that we take for granted. However, we should not forget that what is common and easily accessible today didn’t just magically appear out of thin air; rather, at some point, it used to be an unclear subject of study on which “more research was needed”, and even earlier, it was just a conjecture in some researcher’s head.

Hopefully, one day not too far into the future, rejuvenation biotechnologies will be as normal and widespread as aspirin is today, but right now, we’re in the R&D phase, so we should be patient and remind ourselves that the fact that we can’t rejuvenate people today doesn’t mean that nothing is being done or has been achieved to that end. On the contrary, we are witnessing exciting progress in basic research—the fundamental building blocks without which rejuvenation, or any new technology at all, would stay a conjecture.

In particular, SENS Research Foundation (SRF), a pioneering organization of the field, is sometimes unjustly accused by skeptics for failing to produce results. But produce results it has, and many at that. Skeptics either decide to ignore them or do not have access to reliable sources. For the benefit of the latter, we’ll discuss below what has been achieved by SRF over the past few years, in relation to the infamous “seven deadly things”, the seven categories of damage that aging causes as described in the SENS repair approach.

Mitochondrial mutations

In a nutshell, a mitochondrion is a cell component that is in charge of converting food nutrients into ATP (adenosine triphosphate), a chemical that powers cellular function. Your DNA is contained within the nucleus of each of your cells, but this isn’t the only DNA in your body; mitochondria have their own DNA (known as mtDNA), likely because, at the dawn of life, they were independent organisms that eventually entered a symbiotic relationship with eukaryotic cells, such as those found in our bodies.

Unfortunately, as mitochondria produce ATP, they also produce so-called free radicals as a byproduct—atoms with unpaired electrons that seek to “pair up” with other electrons, and to do so, they’ll gladly snatch them from other molecules nearby, damaging them. As free radicals are created by mitochondria, they’re very close to mtDNA, which is thus very susceptible to being damaged and undergoing mutations.

Mitochondria with damaged DNA may become unable to produce ATP or even produce large amounts of waste that cells cannot get rid of. To add insult to injury, mutant mitochondria have a tendency to outlive normal ones and take over the cells in which they reside, turning them into waste production facilities that increase oxidative stress—one of the driving factors of aging.

MitoSENS: How to solve this problem, and how far we’ve got

Cell nuclei are far less exposed to free-radical bombardment than mitochondria, which makes nuclear DNA less susceptible to mutations. For this reason, the cell nucleus would be a much better place for mitochondrial genes, and in fact, evolution has driven around 1000 of them there. Through a technique called allotopic expression, we could migrate the remaining genes to the nucleus and solve the problem of mitochondrial mutations.

Human-made allotopic expression was a mere theory until late 2016, when, thanks to the successful MitoSENS crowdfunding campaign on Lifespan.io, a proof of concept was finally completed. Dr. Matthew O’Connor and his team managed to achieve stable allotopic expression of two mitochondrial genes in cell culture, as reported in the open-access paper[1] they published in the journal Nucleic Acids Research. As Aubrey de Grey himself explains in this video, of the 13 genes SRF is focusing on, it’s now managed to migrate almost four. This had never been done before and is a huge step towards addressing this aspect of aging in humans. In the past few months, the MitoSENS team has presented its results around the world and worked on some problems encountered in the project.

A list of SRF-funded papers on the topic of mitochondrial mutations can be found here. A more detailed description of its intramural MitoSENS research can be found here.

Lysosomal dysfunction

Lysosomes are digestive organelles within cells that dispose of intracellular garbage—harmful byproducts that would otherwise harm cells. Enzymes within lysosomes can dispose of most of the waste that normally accumulates within cells, but some types of waste, collectively known as lipofuscin, turn out to be impossible to break down. As a result, this waste accumulates within the lysosomes, eventually making it harder for them to degrade even other types of waste; in a worst-case scenario, overloaded lysosomes can burst open and spread their toxic contents around.

This eventuality is especially problematic for cells that replicate little or not at all, such as heart and nerve cells—they’ve got all the time in the world to become swamped in waste, which eventually leads to age-related pathologies, such as heart disease and age-related macular degeneration.

LysoSENS: How to solve this problem, and how far we’ve got

As normal lysosomal enzymes cannot break down lipofuscin, a possible therapy could equip lysosomes with better enzymes that can do the job. The approach suggested by SRF originates with ERT—enzyme replacement therapy—for lysosomal storage diseases. This involves identifying enzymes capable of breaking down different types of intracellular junk, identifying genes that encode for these enzymes, and finally delivering the enzymes in different ways, depending on the tissues and cell types involved.

SRF funded a preliminary research project on lipofuscin clearance therapeutics at Rice University[2] and another project relating to atherosclerosis and the clearance of 7-ketocholesterol[3] (a lipofuscin subtype), which eventually spun into Human Rejuvenation Biotechnologies, an early-stage private startup funded by Jason Hope.

A LysoSENS-based approach is currently being pursued by Dr. Kelsey Moody, who used to work at SRF. Dr. Moody has been working on an ERT treatment for age-related macular degeneration. The treatment consists in providing cells of the macula (a region of the eye’s retina) with an enzyme capable of breaking down a type of intracellular waste known as A2E. The treatment, called LYSOCLEAR, is being worked on by Moody’s company Ichor Therapeutics, which earlier this year has announced a series A offering to start Phase I clinical trials of its product.

If LYSOCLEAR proves successful, it could pave the way for future LysoSENS-based therapies to treat lysosomal dysfunction in different tissues.

A list of SRF-funded papers on the topic can be found here.

Cellular senescence

As cells divide, their telomeres—the end-parts of chromosomes protecting them from damage—shorten. Once a critical length has been reached, cells stop dividing altogether and enter a state known as senescence. Senescent cells are known to secrete a cocktail of chemicals called SASP (Senescence Associated Secretory Phenotype), which promotes inflammation and is associated with several age-related conditions.

However, senescent cells are a bit of a double-edged sword; as explained by Professor Judy Campisi during RB2016, as long as they’re not too numerous, senescent cells carry out an anti-cancer function and may promote wound healing; however, too many of them have the opposite effect, and on top of that, they induce neighboring cells to undergo senescence themselves, starting a dangerous spiral.

Normally, senescent cells destroy themselves via programmed cell death, known as apoptosis, and are then disposed of by the immune system, but some of them manage to escape destruction, and as the immune system declines with age, this gets worse.

The result is that late in life, senescent cells have accumulated to unhealthy amounts and significantly contribute to the development of age-related diseases. Osteoarthritis, cardiovascular diseases, cancer, metabolic disorders such as diabetes, and obesity are all linked to the chronic age-related inflammation to which senescent cells contribute.

ApoptoSENS: How to solve this problem, and how far we’ve got

The proposed SENS solution is straightforward: if senescent cells become too numerous, then they need to be purged. Since they are useful in small amounts, the optimal solution would be periodically removing excess senescent cells without eradicating them entirely—and more importantly, leaving other cells unharmed.

This could potentially be achieved by either senolytic drugs or gene therapies that selectively target senescent cells and trigger programmed cell death. Indeed, a great deal of recent focus by researchers have been on finding ways to remove senescent cells using senolytic therapies.

Another approach that could complement senolytics is to address why the immune system stops clearing senescent cells effectively in the first place. This approach focuses on macrophages and other immune cells involved in clearing senescent cells, aiming to reduce inflammation so that these cells begin to function properly again. The irony is that as inflammation rises with age, the immune system that is supposed to clear senescent cells and keep inflammation levels down actually starts to create more inflammation and becomes part of the problem by not doing its job properly.

SRF has funded a number of studies on the subject of cellular senescence, and it’s recently begun working on a project in collaboration with the Buck Institute for Research on Aging, which is focusing on the immune system and its role in clearing senescent cells. Another extramural project, again with the Buck Institute, is focussed on SASP inhibition.

Senescent cell clearance has been all the rage for the past two years or so; Lifespan.io has hosted the MMTP project, which focused on testing senolytics in mice, and this was later followed by CellAge’s project to design synthetic biology-based senolytics.

There are other companies that have joined the race to add senescent cell clearance to the standard toolkit of doctors, such as Unity Biotechnology and Oisin Biotechnologies.

Unity’s approach uses a drug-based approach to senolytics and is scheduled to enter human clinical trials in 2018. A number of other research teams are also developing drug-based approaches to removing senescent cells, and the competition looks set to be fierce in this area in the coming years.

Oisin’s approach, which we discussed here, makes use of suicide genes and hopefully will be tested in clinical trials not too far into the future, thanks to venture funding presently being collected. If this system can be made to work, it will allow very selective targeting of senescent cells by destroying only those giving off a target gene or genes. Thus, if a unique gene expression profile for senescent cells is determined, it would mean only those cells were destroyed, with less risk of off-target effects.

Oisin owes its existence to the SENS Research Foundation and the Methuselah Foundation, which provided the necessary seed funding. Kizoo Technology Ventures has also invested in Oisin.

Extracellular crosslinks

The so-called extracellular matrix is a collection of proteins that act as scaffolding for the cells in our body. This scaffolding is rarely if ever replaced, and a really bad consequence of this is that its parts eventually end up being improperly linked to each other through a process called glycation—the reaction of (mainly) blood sugar with the proteins that make up the extracellular matrix itself.

The resulting cross-links impair the function and movement of the linked proteins, ultimately stiffening the extracellular matrix, which makes organs and blood vessels more rigid. Eventually, this leads to hypertension, high blood pressure, loss of skin elasticity, and organ damage, among other problems.

While there are different types of cross-links—known as AGEs, short for advanced glycation end-products—glucosepane is arguably the worst, being the most common and long-lasting of all, and the body is very ill-equipped to break it down.

GlycoSENS: How to solve this problem, and how far we’ve got

In order to eliminate unwanted cross-links, the SENS approach proposes to develop AGE-breaking molecules that may indeed sever the linkages and return tissues to their original flexibility. Of course, in order to do so, crosslink molecules need to be available for research to attempt to combat them with drugs, and especially in the case of glucosepane, this has been a problem for years.

Glucosepane is a very complex molecule, and very little of it can be extracted from human bodies, and not even in its pure form. This has been greatly hampering the progress of research against glucosepane, but thankfully, this problem is now solved thanks to a collaboration between the Spiegel Lab at Yale University and the SENS Research Foundation, which financially supported the study. It is now possible to fully synthesize glucosepane, allowing for researchers to create it on demand and at a cost-effective price.

The Spiegel Lab’s scientists are now developing anti-glucosepane monoclonal antibodies to cleave unwanted cross-links. The collaboration between the Spiegel Lab and SRF dates all the way back to 2011, but it was in 2015 that the Lab announced its success and published a related paper [4] in the journal Science.

Further information on glucosepane cross-link breakers can be found in this interview with Dr. David Spiegel from Yale University on Fight Aging!; a list of studies on the subject funded or otherwise supported by the SRF is available here.

SRF also worked with the Babraham Institute on a cross-link quantification project.

Let’s help SRF move forward

As we have seen, SRF has produced plenty of results; the projects we discussed are either recent or ongoing, and yet others were carried out in the past. However, much more work still needs to be done before the first rejuvenation therapies move from the lab to the clinic.

If you would like to know more about SRF’s work or have questions not covered in this article, you have a great opportunity to ask none other than Dr. Aubrey de Grey himself during his AMA (“ask me anything”) on Reddit. On December 7,  at 14:00 PST (22:00 UTC, 17:00 EST), Aubrey will be on /r/futurology to answer any question at all, so save the date! The Futurology mods are also taking pre-AMA questions here if you cannot wait to ask Aubrey anything or cannot make the event.

Readers who wish to donate to SRF to help the organization in its crusade against the ill health of old age can do so by contributing to its winter fundraiser or even becoming SRF patrons. Have a look at SRF’s donation page to find out more.

Literature

[1] Boominathan, A., Vanhoozer, S., Basisty, N., Powers, K., Crampton, A. L., Wang, X., … & O’Connor, M. S. (2016). Stable nuclear expression of ATP8 and ATP6 genes rescues a mtDNA Complex V null mutant. Nucleic acids research, 44(19), 9342-9357.

[2] Gaspar, J., Mathieu, J., & Alvarez, P. (2016). A rapid platform to generate lipofuscin and screen therapeutic drugs for efficacy in lipofuscin removal. Materials, Methods and Technologies, 10, 1-9.

[3] Mathieu, J. M., Wang, F., Segatori, L., & Alvarez, P. J. (2012). Increased resistance to oxysterol cytotoxicity in fibroblasts transfected with a lysosomally targeted Chromobacterium oxidase. Biotechnology and bioengineering, 109(9), 2409-2415.

[4] Draghici, C., Wang, T., & Spiegel, D. A. (2015). Concise total synthesis of glucosepane. Science, 350(6258), 294-298.

SENS logo white

Dr. Aubrey de Grey: Undoing Aging

Dr. Aubrey de Grey is the Chief Science Officer and founder of the SENS Research Foundation (SRF) and one of the original proponents of a damage repair-based approach to aging and age-related diseases. His work has inspired many others to think about aging differently and entertain the idea that, perhaps, we do not have to accept the suffering that age-related diseases cause.

In this article, Dr. de Grey writes a summary of the strategy of rejuvenation based on a periodic approach to repairing age-related damage. This approach has steadily gained increasing traction as the data supporting it grows; certainly, the clearance of senescent cells, a SENS approach, has enjoyed great public interest recently. Interestingly, while he certainly is not the only researcher to have suggested the idea, he was talking about removing senescent cells as a therapy back in 2002 [1].

The work of Dr. de Grey has also encouraged others to pursue repair based approaches to aging, including the much-cited Hallmarks of Aging, which is almost certainly inspired by SENS [2].

The SRF has actively funded research into senescent cells, and companies like Oisin are a direct result of the work of the foundation and are now developing therapies to bring to market. Imagine what might be achieved if all seven of the SENS damages received the same level of support and interest. As a community, we can help to make this happen and maybe have a shot at healthy and longer lives as a result.

Conclusion

In the face of the increasing amount of research and data in support of SENS, there has been a large shift in academic enthusiasm and support for the repair approach to aging. The stakes are high here, but ultimately, if we can develop the tools and methods we need to address the damage that aging causes, we have a real shot at longer and most importantly healthier lives.

Literature

[1] Grey, A. D., Ames, B. N., Andersen, J. K., Bartke, A., Campisi, J., Heward, C. B., … & Stock, G. (2002). Time to talk SENS: critiquing the immutability of human aging. Annals of the New York Academy of Sciences, 959(1), 452-462.

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

Rejuvenation Roundup November 2017

November has been a busy month in the world of rejuvenation biotechnology and life extension, as there has been both really good news and some rather bad news. Let’s have a look together, starting with the bad news and then moving on to decidedly more uplifting things.

Healthy aging and the WHO agenda

It appears that WHO has forgotten to include priorities related to aging in its 13th draft programme of work 2019-2023. This extremely important issue was popularised thanks to the efforts of Dr. Ilia Stambler, who recently co-authored a paper on the importance of healthy longevity as a topic for the WHO programme. The authors of this jointly developed position include leaders of the International Society on Aging and Disease (ISOAD), American Federation for Aging Research (AFAR), International Federation on Ageing (IFA), International Association of Gerontology and Geriatrics – IAGG (European Region and Asia-Oceania), International Longevity Center – Australia, the Gerontological Society of the Russian Academy of Sciences, and the African Society for Ageing Research and Development (ASARD). LEAF board director Elena Milova raised the same issue here, and LEAF’s official position has also been discussed by the Russian newspaper Medvestnik.

Multiple pro-longevity organizations have invited community members to take part in the open consultation on the programme in order to promote the inclusion of the issues of the aging population. Thanks to a joint effort, WHO has received plenty of feedback (90% out of 400 responses) lamenting the absence of aging in the draft, and now WHO will hopefully consider including its earlier Global Strategy and Action Plan on Aging and Health in the draft. Advocacy efforts are in progress.

SENS Winter Fundraiser

Like every year, the folks at the SENS Research Foundation have launched their end-of-year winter fundraiser, and the goal this year is $250,000. The generous support of donors and patrons is essential in the fight against aging, and while helping the cause is rewarding in and of itself, a little incentive is never unwelcome; this year, SENS is doing something a bit different.

Anyone who contributes $250 or more between October 15th and December 31st will automatically receive a SENS Research Foundation t-shirt just like the ones worn by the team. Anyone who donates $500 or more will get a cool SENS Research Foundation Polo shirt. You can find out more about the winter fundraiser here.

LEAF Conference in Moscow on November 4

LEAF, in collaboration with Singularity University Moscow Chapter and consulting group Deloitte, hosted the conference “6 ways to talk to people about aging” in Moscow. Six brilliant speakers shared their insights with the audience on how best to inform society about the potential and the advancement of gerontology and preventive medicine.

The list of speakers included Jose Luis Cordeiro, fellow of the World Academy of Art and Science (WAAS), director of life extension advocacy organization Humanity Plus; Valerija Udalova (Pride), director of Russian cryocompany KrioRus and methodologist of the Human Aging System Diagram project; Eugeny Kuznetsov, ambassador of Singularity University in Russia; Maxim Kholin, co-founder of the group «Gero», which develops therapies and blockchain solutions for significant lifespan extension; Anastasia Egorova, vice president of the Foundation Science for Life Extension and director of Open Longevity ICO; and Carlos Garcia Hernandez, director of Lolabooks publishing house, which released the book by Aubrey de Grey and Michael Rae, Ending Aging, in Spanish, and translated books by Ray Kurzweil and other great thinkers of our time.

The conference was attended by nearly 200 people! Video recordings of the event will soon be available on our website as well as Facebook and Youtube pages.

Lifespan.io on Kurzgesagt… again!

Kurzgesagt has released the second video on aging created with the help of the Lifespan.io team on November 3. While the first video introduced viewers to the possibility of ending aging within a few decades, discussing why we should do this, this new video discusses some of the medical technologies currently being developed to address the various hallmarks of aging. If you’ve not done so yet, we definitely recommend you watch these videos! It has been a pleasure to work with Kurzgesagt, and we look forward to future collaborations.

Support for rejuvenation biotechnology and LEAF is growing

A most welcome outcome of our collaboration with Kurzgesagt has been a large increase in our followers and supporters. Judging by the positive responses from the majority of the audience to these videos, it seems very clear that the dissenters are very much in the vocal minority.

Fight Aging! Matching Fund and other news

Just as traditional as the SENS Winter Fundraiser, the matching fund put up by Fight Aging! is running since October 15th and will continue until December 31st. Reason, Josh Triplett, and Christophe and Dominique Cornuejols have put together a $36,000 fund with which they’ll match the first full year of donations made to SENS by anyone who signs up as a monthly patron before the end of 2017.

In other news from FA!, Reason reported an open-access paper discussing the connection between mitochondrial dysfunction and cellular senescence in relation to age-related respiratory diseases, such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. The degradation of defective mitochondria declines in senescent cells and the paper suggests that this might contribute to the pro-inflammatory aspects of cellular senescence.

Interview with Dr. Michael Fossel

Our friends at Geroscience have published the second part of their interview with Dr. Michael Fossel, a leading expert on telomere attrition and telomerase therapy.

The first part of the interview, published in October, discusses his views on aging and the reasons why he focuses on telomeres especially; in the second part, Dr. Fossel explains how telomere replenishment could be done and the benefits we can expect to reap. Check it out!

A panel about life extension prospects in the near future

Biogerontology Research Foundation Trustee Dmitry Kaminskiy and Chief Science Officer Dr. Alex Zhavoronkov spoke at the Aging and Longevity Panel at the Financial Times Global Pharmaceutical and Biotechnology Conference in London on November 10th, 2017, alongside Dr. Aubrey de Grey, Chief Science Officer of SENS Research Foundation, and Joseph Antoun, Chairman of the Global Healthspan Policy Institute.

The discussion panel focused on the prospects for geroscience to deliver practical solutions to aging in the near future. This is of particular interest given that the topic of targeting aging to prevent disease has been openly discussed at one of the largest European biotechnology conferences, at which all the major pharmaceutical companies were present. Interest in the field and preventative medicine has grown dramatically in the last few years, and this is yet more indication of its rising popularity.

Conversations with the future

Nikola Danaylov’s new book “Conversations with the future” is trending on Amazon. There is an interview with Michael Fossel, PhD, MD, professor of Clinical Medicine of Michigan State University, a famous researcher of aging. The section discusses the main mechanisms of aging, the possibility to manipulate them and related emerging technologies while touching upon some of the most frequent ethical issues related to getting rid of aging. Readers can also find a few recommendations of currently available methods to extend healthy life.

Coming up in December

SENS AMA

On December 7th, in conjunction with the ongoing winter fundraiser, Dr. Aubrey de Grey, Chief Science Officer of SENS Research Foundation, will be on Reddit for an AMA (“ask me anything”). Dr. Aubrey de Grey is one of the original proponents of a repair based approach to aging and age-related diseases and this is your opportunity to ask him about the progress being made in research, his thoughts about the future or whatever else you might like to know about this fascinating scientist.

Anyone with a question for Dr. de Grey will have a chance to ask him personally in the /r/futurology subreddit, starting at 2 PM PST (5 PM EST, 10 PM UTC). If you want to know how SENS research is progressing, or ask Aubrey anything at all, this is your chance!

More LEAF events

LEAF is hosting two events in Moscow in December. On December 6, LEAF Director Elena Milova is giving a lecture called “How to defeat aging?” at one of the state libraries. The week after, Pam Keefe, member of the advisory board of the Coalition for Radical Life Extension and co-organizer of the RAADFEST Bangkok conference, is visiting Moscow, and we will use this opportunity to organize a friendly discussion of longevity communication strategies entitled “Revolution against aging: principles, methods, examples”.

Project for Awesome

LEAF is taking part in the “Project for Awesome 2017” contest taking place on December 15-17, and we hope that you will come to support us by voting for our videos and making your own videos to say why you support the work we do. Please reference the Lifespan Extension Advocacy Foundation in the video description.

Why do Some People Fear Using Science to Live Longer Healthy Lives?

With the recent increase of enthusiastic support for the idea of defeating aging, I really shouldn’t be complaining about the lingering holdouts opposing the idea of living healthy and longer lives for whatever reason. Nonetheless, I feel compelled to point out the jarring contradiction of a species whose members are constantly on the lookout to avoid danger, yet need to be reassured that at some point they will die and that science is by no means trying to prevent that.

Fear of life extension

This curious phenomenon was pointed out earlier this year by James Goodwin in the journal of the Gerontological Society of America[1]. In his article “Fear of life extension”, he argues that the commandment of geriatrics is “quality, not quantity” because policymakers fear a future in which longer life means overpopulation of nursing homes, and as a consequence, researchers working on interventions on aging need to clearly state that their goal is just making our final years a little better—but making us live longer? God forbid, no! We only want to live in perfect health for about 80 or 90 years, then magically drop dead for no apparent reason.

Quite concisely, Goodwin explains that you can’t have significant longevity without an equally significant improvement of your health, and vice-versa; so did I, in my own small way, in this article. Yet, the idea of living longer than the status quo, or even suggesting that this might be a good thing, is almost blasphemous to some, and I’m not talking just about academics bending the truth a little for the sake of getting their next grant. “Quality, not quantity” has become a mantra, not only because of the largely unjustified concerns about the potential downsides of longer lives but also because it has somehow been incorporated into the set of conventional wisdom that nearly no one dares challenging for the simple reason that nearly no one else does.

Don’t worry—you’ll die anyway

I’ve personally talked to a number of people who think it would make no sense if your life never came to an end; there has to be closure for it to have meaning, they say. I’ve spoken to people who “know” they’ll want to die at some point, and I’m not talking about bitter old men who’re tired of their failing bodies and the many disappointments life has served them; I’m talking about barely adult people in their early twenties, with their health and looks still perfectly in place and pretty much a whole “normal-length” life to live. This, I must ascribe to the power that clichés have on the human mind; a lot of people repeat that accepting, wanting, and cherishing the finitude of life is wise, so it must be true, and so they follow suit. That sort of thing.

And it is no joke because advocates of rejuvenation often need to reassure people that this is not about eliminating death; it’s not about making you unable to die. Granted, that is true—rejuvenation doesn’t make you immortal—but I’ve lost count of articles and videos on the subject of aging that, while discussing advancements that may improve our lives and maybe even make them a little longer, still make it absolutely clear to their audience that they needn’t worry—fear not, you’ll still die at some point! Whew, what a relief! For a second there, I actually thought I’d be able to get out of life alive.

If you think it through, though, this is most strange. Like most if not all creatures on this planet, we have evolved to avoid danger and situations that might cause our own demise. To put it bluntly, we have evolved to fear death, yet we have people who say there’s nothing to fear in it.

If that’s so, why do we have hospitals to fix us up, ambulances rushing to accident sites in the hopes of making it in time to save the most people, safety measures to minimise our risk of death? Why do we look after our young, why do we take medicines and vaccines, why do we have suicide helplines? If there’s nothing to fear in death, why do we go through so much trouble to avoid it? Seems a bit odd to me.

Just to be clear—I don’t fear what lies after death because my personal view is that there’s nothing at all on the other side; and while I do fear a painful death, that’s not what I’m getting at here. Death would put an end to all I like and love, and quite frankly, that would kind of throw a spanner in my works. If something had gone wrong with me and I didn’t have the instinct to fear death anymore, I’d probably have a harder time avoiding death, but I’d still have plenty of non-fear-related reasons to want to avoid it.

Conclusion

What are the roots of this fear of longevity? Why is it that it almost can’t be spoken about? Is it the misplaced concern of longer decrepitude? Is it a fox-and-grapes situation? Is it some sort of Stockholm syndrome that makes us grow fond of our persecutor? Is it the fear of overpopulation and boredom? Maybe a combination of all of these possibilities?

I’m not sure, but I do know that it might be a roadblock on the way to a world without age-related diseases. As I said at the beginning, there certainly are reasons to think that the roadblock, if it is even there, is slowly being dismantled; nonetheless, I suggest not to underestimate this danger and to keep doing our best to explain why healthy longevity, not the finitude of life, needs to be cherished.

Literature

[1] Goodwin, J. S. (2017). Fear of life extension.

Does WHO 5 Year Plan Leave Healthy Aging Out of the Picture?

When it comes to healthcare, WHO remains the global flagship in terms of setting priorities and adjusting policies in the ever-changing landscape of healthcare needs. In the past few years, WHO has been making steps towards preparing our society to handle the “silver tsunami” and the increasing needs of the elderly population, which is expected to reach 22% of the global population by 2050.

The last 12th General Programme of Work, adopted by the World Health Assembly in May 2013, listed population aging and morbidity of age-related diseases as one of the points of great concern. However, in the recent Draft thirteenth general programme of work 2019–2023, the priorities related to aging and age-related disease are… gone.

What is this, a tragic accident or the worst example of ageism at the level of the global policymaking? Can the Open Consultation running until November 15 help bring healthy aging back into the WHO agenda? You will also find The LEAF Position regarding the Draft programme at the end of this article, but first, let’s discuss why this is important to keep healthy aging among the top priorities.

After all the efforts to introduce aging and the problems of the growing share of the elderly into the international agenda, WHO seems to fall back by 30 years and leave this most important question out the discourse for the next 5 years. While the Draft thirteenth general programme of work 2019–2023 claims to be based on the Sustainable Development Goals, it is clear that it is throwing out the problems of the elderly. For instance, the SDG indicated the need to promote the development and implementation of medicines against noncommunicable diseases; this never made it to the new draft, and neither did the four main diseases with the highest mortality rates. Here are the quotes from the SDG indicators:

3.4 

By 2030, reduce by one-third premature mortality from non-communicable diseases through prevention and treatment and promote mental health and well-being

3.4.1 

Mortality rate attributed to cardiovascular disease, cancer, diabetes or chronic respiratory disease

3.B Support the research and development of vaccines and medicines for the communicable and non-communicable diseases that primarily affect developing countries, provide access to affordable essential medicines and vaccines, in accordance with the Doha Declaration on the TRIPS Agreement and Public Health, which affirms the right of developing countries to use to the full the provisions in the Agreement on Trade-Related Aspects of Intellectual Property Rights regarding flexibilities to protect public health, and, in particular, provide access to medicines for all.

The priorities of the new draft moved elsewhere and do not pay much attention to age-related diseases, which account for most deaths among the ones caused by NCDs. Here is how the set of priorities on NCDs is formulated in the draft:

3. Noncommunicable diseases (NCDs), mental health, substance use, road traffic injuries
  • Reduce tobacco use by 25%
  • Reduce harmful alcohol use by 10% (per capita consumption)
  • Keep the levels stable for overweight (including obesity) in children and adolescents
  • Eliminate industrially produced trans fats
  • Reduce prevalence of raised blood pressure by 20%
  • Increase service coverage for severe mental health disorders by 40 percentage points
  • Reduce road traffic fatalities by 20%

This list is barely touching upon age-related health issues (high blood pressure is probably the most relevant point), while the priorities 1, 2 and 4 are completely different. Looking at this document, we are desperately asking ourselves why WHO is suddenly turning its back on the elderly population and what we shall expect to happen to the previous strategic programs, as the draft mentions that it will also influence the Programme budget of 2018−2019 through a mechanism of resource reallocation. Should we treat it as a warning that the resources previously expected to help fulfill the Global strategy and plan of action on ageing and health are going to be redirected to this new “ageist” set of goals?

WHO is currently running an open public consultation on the draft, and we invite you to take part and provide your view regarding the necessity of keeping the issue of age-related diseases in the WHO agenda in the years to come. However, before we provide you with the position that we are going to send to WHO (the example is at the end of this article), we would like to remind you of the key indicators and documents with which you should be familiar in order to make an informed decision regarding the global healthcare agenda.

The age group 60+ is growing faster than any other

The global population is aging rapidly. Here is a quote from the UN Population Prospects 2017 Revision Key Findings concerning this change:

In 2017, there are an estimated 962 million people aged 60 or over in the world, comprising 13 per cent of the global population. The population aged 60 or above is growing at a rate of about 3 per cent per year. Currently, Europe has the greatest percentage of population aged 60 or over (25 per cent). Rapid ageing will occur in other parts of the world as well, so that by 2050 all regions of the world except Africa will have nearly a quarter or more of their populations at ages 60 and above. The number of older persons in the world is projected to be 1.4 billion in 2030 and 2.1 billion in 2050, and could rise to 3.1 billion in 2100. Over the next few decades, a further increase in the population of older persons is almost inevitable, given the size of the cohorts born in recent decades.

NCDs disproportionately affect people in low- and middle-income countries

Worldwide, infectious diseases have been replaced by chronic non-communicable diseases (NCDs) as the leading causes of death; this is called the epidemiological transition. This transition is associated with the most developed countries, but this is a widespread misconception. According to WHO, NCDs disproportionately affect people in low- and middle-income countries, where more than three-quarters of global NCD deaths – 31 million annually – occur. The most frequent NCDs are cardiovascular diseases (heart attacks and stroke), cancers, chronic respiratory diseases (such as COPD), and diabetes. Here is the quote from the WHO fact sheet regarding NCDs:

Cardiovascular diseases account for most NCD deaths, or 17.7 million people annually, followed by cancers (8.8 million), respiratory diseases (3.9 million), and diabetes (1.6 million).

Chronic diseases make most of the top 10 causes of death globally

If we look at the WHO estimates of the main causes of death globally, we can see that age-related chronic diseases account for the vast majority of deaths, with infectious diseases and road injury comprising a much smaller part:

Most recent WHO strategic documents are focused on age-related issues

This data on population aging, combined with the data on morbidity, informed the previous WHO activities related to aging. In 2015, it released the World report on ageing and health, creating an exhaustive overview of population aging, discussing related health, social and economic changes, and describing the necessary actions to make the older population as healthy as possible. The report set the following definition of healthy aging and its mandatory elements:

Healthy Ageing is the process of developing and maintaining the functional ability that enables well-being in older age.

Functional ability comprises the health-related attributes that enable people to be and to do what they have reason to value. It is made up of the intrinsic capacity of the individual, relevant environmental characteristics and the interactions between the individual and these characteristics.

Intrinsic capacity is the composite of all the physical and mental capacities of an individual.

Environments comprise all the factors in the extrinsic world that form the context of an individual’s life. These include – from the micro-level to the macro-level – home, communities and the broader society. Within these environments are a range of factors, including the built environment, people and their relationships, attitudes, and values, health and social policies, the systems that support them, and the services that they implement.

Well-being is considered in the broadest sense and includes domains such as happiness, satisfaction and fulfilment.

Based on the indicators listed in this report, the Global strategy and plan of action on ageing and health was developed and signed into power in 2016, setting the following strategic objectives and goals to prepare for the Decade of Healthy Aging (2020-2030):

VISION A world in which everyone can live a long and healthy life

STRATEGIC OBJECTIVES 1. Commitment to action on Healthy Ageing in every country 2. Developing age-friendly environments 3. Aligning health systems to the needs of older populations 4. Developing sustainable and equitable systems for providing long-term care (home, communities and institutions) 5. Improving measurement, monitoring and research on Healthy Ageing

PLAN OF ACTION 2016–2020

GOALS 1. Five years of evidence-based action to maximize functional ability that reaches every person. 2. By 2020, establish evidence and partnerships necessary to support a Decade of Healthy Ageing from 2020 to 2030.

This document also points out the need to support the development of innovative interventions to address age-related diseases and their underlying causes:

  1. Finally, better clinical research is urgently needed on the etiology of, and treatments for, the key health conditions of older age, including musculoskeletal and sensory impairments, cardiovascular disease and risk factors such as hypertension and diabetes, mental disorders, dementia and cognitive declines, cancer, and geriatric syndromes such as frailty. This must include much better consideration of the specific physiological differences of older men and women and the high likelihood that they will be experiencing multimorbidities. This could also be extended to include possible interventions to modify the underlying physiological and psychological changes associated with ageing.

The LEAF Position regarding the Draft thirteenth general programme of work 2019–2023

Taking this information into account, we suggest the following position regarding the Draft thirteenth general programme of work 2019–2023 that can provide insight and inspiration for your own position. The Open Consultation will be running until November 15. To take part and share your vision with WHO, please visit this WHO page, scroll down, and fill the mandatory fields of the survey.

  1. The global population is aging rapidly, and according to the UN Population Prospects 2017 Revision, the share of people aged 60+ will make up 22% of the global population by 2050[a]. Sadly, higher life expectancy does not necessarily mean an extension of the healthy period of life, and more people around the globe live longer while suffering from chronic age-related diseases, with the developing countries bearing the most burden.
  2. Most countries already experience epidemiological transition, with non-communicable diseases, particularly age-related diseases, becoming leading causes of death. The most frequent NCDs are cardiovascular diseases (heart attacks and stroke), cancers, chronic respiratory diseases (such as COPD) and diabetes. Cardiovascular diseases account for most NCD deaths, or 17.7 million people annually, followed by cancers (8.8 million), respiratory diseases (3.9 million), and diabetes (1.6 million)[b].
  3. The Sustainable Development Goals set the goal to “reduce mortality rate attributed to cardiovascular disease, cancer, diabetes or chronic respiratory disease” (3.4.1), and to “support the research and development of vaccines and medicines for the communicable and noncommunicable diseases that primarily affect developing countries, provide access to affordable essential medicines and vaccines…” (3.B).
  4. The latest WHO World report on ageing and health (2015) and the Global strategy and plan of action on ageing and health (2016) underline the need to promote healthy aging for all, and they recommend the corresponding set of measures, including better clinical research on the etiology of, and treatments for, the key health conditions of older age, including musculoskeletal and sensory impairments, cardiovascular disease and risk factors such as hypertension and diabetes, mental disorders, dementia and cognitive declines, cancer, and geriatric syndromes such as frailty, along with possible interventions to modify the underlying physiological and psychological changes associated with aging (with consideration of the specific physiological differences of older men and women and the high likelihood that they will be experiencing multimorbidities)[c].
  5. Clinical trials of several innovative interventions that can address the underlying mechanisms of aging, postpone and cure age-related diseases, and extend the healthy period of life (senolytics, thymus regeneration in situ, therapies to regulate NAD+ and improve DNA repair, metformin) are currently being performed, and these interventions can enter the market in the next 5 years. WHO supervision and guidance regarding the implementation of these interventions is required to ensure their rapid, worldwide dissemination, affordability and equal access.
  6. While we recognize the need to promote universal health coverage, to keep fighting infectious diseases and mental illnesses, and to promote the health of women, children and adolescents, we find efforts to ensure healthy aging to be of vital importance for our society’s well-being in the decades to come. If this most important topic will be excluded from the global agenda, and the corresponding funding reallocated to solve other health issues, not only it will be an unhealthy example of ageism at the highest level of policymaking, but our society will also face the growing needs of the increasing older population unprepared and disoriented.
  7. The global and local effects of not addressing these issues can be devastating, posing an increasing burden on healthcare and social protection systems (for instance, public health expenditures in the United States are projected to rise from 6.7 percent of GDP in 2010 to 14.9 percent in the year 2050), causing shortages of professional caregivers for aged patients, which will turn a significant part of the working population into compulsory family caregivers and undermine sustainable economic growth.
  8. In order to extend good health and productivity throughout the life course and to ensure that older people preserve their intrinsic ability for as long as possible, we need to keep working in the directions set by the Global strategy and plan of action on ageing and health (2016), including the promotion of clinical research on possible interventions to modify the underlying physiological and psychological changes associated with aging. Hence, we kindly ask you to include promoting healthy aging as a priority of the Draft thirteenth general programme of work 2019–2023.

[a] United Nations, Department of Economic and Social Affairs, Population Division (2017). World Population Prospects: The 2017 Revision, Key Findings and Advance Tables. Working Paper No. ESA/P/WP/248.

[b] WHO. The top 10 causes of death. Fact sheet. Updated January 2017.

[c] WHO Global Strategy and Action Plan on Ageing and Health,” World Health Organization, accessed November 8, 2017.

Thank you very much for finding time to fill out this survey!

Providing policymakers like WHO with our vision regarding the potential of rejuvenation technologies to tackle the problems related to population aging is very important. This way, we help disseminate information about rejuvenation biotechnologies in our society while we encourage more people to support researchers and to start using the interventions that are already available.

We would like to thank Dr. Ilia Stambler for bringing this matter to our attention.

A Demonstration of Rejuvenation in Old Human Cells

Today, we are going to have a look at a new study that shows that senescent, non-dividing aged cells can be rejuvenated using a new technique.

Before we do that, let’s take a look at what senescent cells are and how cellular senescence protects us from cancer and other harmful diseases.

Cellular senescence protects us from damaged cells

Cellular senescence is a safety mechanism that removes aged and potentially damaged cells from circulation. Having mutated and otherwise damaged cells replicating indefinitely would be a recipe for disaster, so this safety system kicks in to shut down these cells and summon the immune system to dispose of them.

Senescent cells are cells that have ceased replicating in response to damage or environmental stimuli or cells that have reached their replicative limit and have critically shortened telomeres that signal the cells to shut down. Senescent cells do not replicate and generally destroy themselves by programmed cell death, which is known as apoptosis; in some cases, they evade this process and remain in situ, causing inflammation and secreting a range of toxic signals that poison the local tissue.

Over time, the number of senescent cells increases in tissue, leading to a rising background of chronic inflammation that causes the immune system to become increasingly dysfunctional, impairs tissue repair and drives the progression of multiple age-related diseases[1].

The most obvious solution to this problem is to remove these senescent cells periodically from the body, and the removal of senescent cells is exactly what Dr. Aubrey de Grey and the SENS Research Foundation have been proposing for over a decade. This has been the focus of much research in recent years, and a new class of therapies known as senolytics aims to do exactly this.

However, some researchers have been tinkering with ways to reverse senescence in cells instead of the more direct approach of removing the problem in the first place.

Reversing cellular senescence

This senescent state was once thought to be an irreversible process, and while this is certainly true during the normal processes of biology, it is not the case when science intervenes. Using cell reprogramming, cell fate can be changed and senescence can be reversed, as demonstrated once again in a new study that shows another technique to achieve this[2].

Senescence is not a one-way street, and a cell can be reprogrammed back to working order from senescence, provided the correct signals are given to it. In fact, a whole range of cell states can be programmed; it is just a case of working out the correct signals and manipulating the genetic machinery to get the result you want. This is no easy task despite the great progress that has been made in the understanding of cell fate in the last decade.

We now understand how to change some cell states quite well, and we have become adept at resetting some cells back to a pluripotent state. Similar to embryonic stem cells, these cells can then be guided down different paths to create new kinds of cells. More recently, we have discovered that cells do not even need to be reverted back to this pluripotent state; instead, they can be directly changed from one type to another without this additional step via the process of transdifferentiation.

Once cell fate is reprogrammed, senescent cells can be made to begin replicating again, and these cells can even operate at a functionally younger level, with a number of aging markers reversed. The researchers of this study have found another technique to restore senescent cells to work.

They exposed aged cells to analogs of the common supplement resveratrol, which caused splicing factors, which are progressively turned off as we age, to be turned back on. Within a few hours, the cells appeared younger and started to rejuvenate, behaving like younger cells and dividing again.

Conclusion

As interesting as this experiment is, is it really a good idea to bring back senescent cells from their arrested replicative state? Considering that senescent cells are generally senescent for a reason, it seems to be a less than wise approach.

Senescent cells may simply be senescent because they have reached their replicative limit, but they might also be damaged and could be harboring any number of mutations that caused them to shut down in the first place. These mutations could potentially cause them to become cancerous, and indeed, cellular senescence is designed to protect us from this.

So, this approach is a double-edged sword. On one hand, the potential for returning damaged cells to circulation will almost certainly increase the risk of cancer, but on the other hand, the harm that senescent cells cause via their inflammatory secretions is also significant.

It seems that there could be short-term benefits to restoring senescent cells to circulation due to removing their toxic secretions, but in the long term, the risks are not trivial. Senescent cells are generally likely to be damaged, and, really, instead of returning them to work, they should be destroyed as the body intended.

This is why the senolytic approach of destroying these cells offers the most robust and lower-risk option in dealing with senescent cells, and why, in our view, the approach here is inferior and a poor second-best choice in the long-term. If we can simply remove these aged, damaged cells, why would we bother to paper over the problem? The answer seems obvious.

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] Latorre, E., Birar, V. C., Sheerin, A. N., Jeynes, J. C. C., Hooper, A., Dawe, H. R., … & Harries, L. W. (2017). Small molecule modulation of splicing factor expression is associated with rescue from cellular senescence. BMC cell biology, 18(1), 31.

Why Bringing Aging Under Medical Control Probably Will Not Create a Gerontocracy

As I discussed in another article, rejuvenation biotechnology could potentially allow older adults to continue working and producing wealth for much longer than they can today, thus benefiting society in many ways.

However, some people are concerned that this might do more harm than good; imagine all those rejuvenated old farts holding onto their jobs forever, preventing the young from getting jobs themselves! Not to mention the risk of a gerontocratic world, where powerful older people get a touch too attached to their chairs, never allowing younger people a chance!

New is not always better

If you’re more concerned that dictators could live for centuries, then you should have a look at this article; here, I’m going to deal with another scenario: old, rejuvenated people who hold on to positions of power—not necessarily as heads of countries—or their jobs for a really long time.

Quite frankly, what’s wrong with that?

Just because someone has been in charge of the same position for long, it doesn’t mean that it’s necessarily a bad thing. If you think otherwise, you might be making the assumption I rebutted here, namely that, rejuvenated or not, older people will always tend to do things in old ways, eventually making them a worse choice than younger people. On the contrary, their long experience might make them more fit than others, especially if we’re talking about chronologically older but open-minded people who keep up to date. Younger people aren’t necessarily the default better option. Think about all those times when a great person of our time died and you found yourself thinking that the world would have been better off if he or she had lived longer.

Personally, I think what matters is that people in certain positions, whether within government or a company, are the right people for the job. If they aren’t, old or young, they should be replaced by other people who are more fit, and, generally, there are more efficient and humane ways to do so than letting them get age-related diseases—for example, voting for someone else or hiring a different person. Granted, if the person to be kicked out is really powerful, this may well be easier said than done; however, when the holder of a position of such power dies of old age, his successor is rarely a nobody with no string-pulling abilities whatsoever. It’s not really a matter of longevity; rather, it’s that power attracts power, and I doubt that creating or not creating rejuvenation will make much of a difference in this respect.

Where does power come from?

It’s easy to hypothesize that a generation of rejuvenated 200-year-olds could end up becoming a gerontocratic elite that maintains power over younger people, but how would this be accomplished, exactly?

Maybe the older generation is rich and powerful, but unless we’re talking about a totalitarian world in which the masses are intentionally kept ignorant and poor, younger generations do have fair chances to make positions for themselves. Power and wealth come from knowledge, and, these days, knowledge is more freely and widely available than ever before.

Learning new professional skills and acquiring knowledge, in general, is possible for virtually everyone, thanks to the pervasiveness of educational media and the open availability of information on the Internet, including free (or reasonably affordable) online education projects, such as Coursera and Edx. The recent European initiative to ensure that all scientific papers are open access by 2020 represents another step towards a world that shares information rather than hides it.

Truth to be told, power and wealth don’t come only from knowledge; they also come from powerful and wealthy ancestors. If we didn’t develop rejuvenation, certainly all the Scrooge McDucks of the world would die sooner than they would otherwise, but their power and wealth would go to their heirs, and so on over the generations, which wouldn’t do much to prevent the creation of an elite. So, no, old age is not an easy way out of the problem of powerful elites ruling the world, and its absence wouldn’t make the problem any worse, really. The only possible way out is giving everyone equal access to knowledge and equal opportunities.

Inevitably, some will end up being more successful and thus more powerful than others anyway; however, if this allows them to become an oppressive force on the rest of us, I think this is a problem with our socio-economic system, not with the existence of lifesaving medical technology. I don’t know about you, but I’m not very keen on waiting until the “perfect” society or “perfect” economic system are built before we decide to cure the diseases of old age.

The fortune teller’s error

We shouldn’t make the mistake of predicting a negative outcome without considering the actual odds of it happening. I think fears of a society where rejuvenated elderly make younger people’s lives more difficult are misplaced in that they assume present-day scenarios will exist in the far future.

Take the concern about jobs, for example, rejuvenated old people would stick to their jobs forever and make it harder for young people to enter the workforce. It sounds bad, but there are a few assumptions behind it that we should question.

First, would rejuvenated old people actually stick to their jobs forever? Why? You hardly hear of a professional who was in the exact same job for forty years these days. More broadly, career change is a thing already. After all, after 40 years in the same line of work, it’s conceivable you might want to try something else, thus making room for others to take your place.

Will rejuvenated old people be allowed to stick to their jobs forever? Not everyone is a manager in charge of decisions, and your boss may well decide to lay you off, rejuvenated or not, and hire someone else.

Even if old rejuvenated people did stick to the same jobs forever, would they never take a break? Even if you’re in the prime of health, after a few decades, you may well wish to “retire” for a few years before going back to work, and your employer is probably not going to wait for you that long.

Will there be so many chronologically younger people in need of jobs in the future? The world population growth rate has been hopelessly corkscrew-diving for over 50 years now, and it is projected to keep going down as larger portions of the world transition from a developing to a developed economy. (In case you’re wondering, the population growth rate is going down not because more people die, but on the contrary, because fewer are born.)

Will people’s living depend on having a job in the future? We can’t expect indefinite life extension to happen very soon; before we can have 200-year-old people in the workforce, it’ll be at least a century. Is the economy going to be the same as today’s by then? Automation already seems on its way to cause the end of work as we know it.

I’d say it’s rather silly to oppose rejuvenation today for the reason that, in a century or two, it might cause an unemployment problem due to too many people being alive. It’s simply too long a time to make any even remotely accurate predictions on what the job market will be like or if there even will be any. In all honesty, I think it makes more sense to worry about a concrete problem that we already have today—the ill health of old age—than worry about a hypothetical one that might or might not happen in a hundred years’ time—massive unemployment. As time goes by, we’ll have a better picture of potential future problems lying ahead, and we’ll be in a better position than we are in today to do something about them.

Conclusion

I’ve said this many times over: the bottom line is always the same. Yes, life extension will most likely bring challenges along with benefits, but none of these challenges are certain, insurmountable, or not more than compensated for by the expected benefits. Let’s not deny ourselves and our descendants the chance for healthier, longer lives.