×

The Blog

Building a Future Free of Age-Related Disease

Mitochondrial dysfunction in aged brain cells

Thanks to a new technique, researchers from the Salk Institute’s Gage laboratory have shown that impaired energy production might be a reason why human brains are susceptible to age-related diseases in the first place [1].

In particular, Salk scientists observed that induced neurons (iNs) obtained from fibroblasts of older individuals had dysfunctional mitochondria and therefore decreased energy levels compared to younger neurons. Out-of-shape mitochondria have previously been implicated in degenerative brain diseases, such as Alzheimer’s and Parkinson’s, and this finding might help reveal more about the connection between these diseases and this particular hallmark of aging.

Mitochondrial dysfunction 101

Our readers are probably familiar with the 2013 study “The Hallmarks of Aging”, a review describing in detail what is known of the typical signs of age-related degeneration at the molecular level [2]. Mitochondrial dysfunction is a hallmark in its own right, and it can be thought of as the meltdown of cellular energy production facilities.

Mitochondria are organelles found in each and every of your cells; they are responsible for harnessing the chemical energy of the food you eat to keep you alive. Mitochondria generate energy but also toxic waste—namely, reactive oxygen species, also known as free radicals. Free radicals are molecules with unpaired electrons desperately looking to pair up, which makes them extremely reactive and prone to stealing electrons from nearby molecules. Molecules they steal electrons from become damaged; mitochondrial DNA, which is different from nuclear DNA and nowhere near as well protected from free radicals, is easily damaged by the free radicals created by mitochondria themselves. These mutations reduce mitochondrial efficiency and tend to spread out, with healthy mitochondria being slowly supplanted by mutated ones, ultimately turning cells into free-radical production facilities.

A novel way to study older neurons

Neuronal tissue isn’t easy to obtain from living humans. Samples have been taken from deceased donors, of course, but being able to study neurons coming from patients who are still alive may provide useful insights. Up until recently, this was achieved by turning different types of cells more easily sampled from living patients, such as skin cells, into pluripotent stem cells first and finally into neurons. However, this procedure has the side effect of largely rejuvenating the converted cells, wiping away the very signs of aging that the researchers wish to study.

While it is possible to simulate the effects of aging on these cells by exposing them to stressors, this procedure doesn’t necessarily lead to the same markers that aging would produce; for this reason, in 2015, the Gage lab researchers developed a method to turn human fibroblasts directly into neurons without having to turn them into pluripotent stem cells first and showed that doing so preserves age-related changes, such as gene expression [3]. Neurons so obtained are the previously mentioned induced neurons.

Results on mitochondria

In 2018, Salk researchers decided to use their 2015 technique to investigate the state of mitochondria in aged neurons. The scientists sampled skin cells from humans between the ages of zero and 89 years, examined their mitochondria, turned the cells into iNs, and finally observed mitochondria again after the conversion.

While they were still fibroblasts, harvested cells from every patient exhibited few age-related changes in general, but this was no longer the case once the cells were turned into neurons. Mitochondria in neurons induced from fibroblasts of older donors were fragmented, less dense, and less efficient than in iNs coming from younger fibroblasts; this hints that neural mitochondria are more sensitive to age-related changes than skin cell mitochondria. Researchers found these neural mitochondria to be defective in virtually every aspect; it is their guess that, since neurons rely more heavily on mitochondria for energy than other cells do, the effects of aging on them are more prominent than on other cell types.

Conclusion

The authors of this paper believe that their creation will be a very useful tool to study neurological aging and age-related diseases in general; indeed, Gage lab researchers believe that their 2015 method to turn fibroblasts directly into neurons may be adapted to create older heart and liver cells, for example, enabling scientists to more easily study the effects of aging in the body.

As always, the road ahead is still very long, but every newly added piece of the puzzle, however tiny, gets us closer to understanding and addressing the causes of age-related diseases.

Literature

[1] Kim, Y., Zheng, X., Ansari, Z., Bunnell, M. C., Herdy, J. R., Traxler, L., … & Schlachetzki, J. C. (2018). Mitochondrial Aging Defects Emerge in Directly Reprogrammed Human Neurons due to Their Metabolic Profile. Cell Reports, 23(9), 2550-2558.

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

[3] Mertens, J., Paquola, A. C., Ku, M., Hatch, E., Böhnke, L., Ladjevardi, S., … & Gonçalves, J. T. (2015). Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell stem cell, 17(6), 705-718.

A New Way to Control High Blood Pressure

Researchers at the National University of Singapore (NUS) have potentially found a new way to combat hypertension by discovering how blood pressure is controlled.

What is hypertension?

High blood pressure, or hypertension, rarely has noticeable symptoms. However, if it is left untreated, your risk of serious problems, such as heart attack and stroke, is increased.

Persistently high blood pressure can increase your risk of a number of serious and potentially life-threatening conditions, such as heart disease, heart attack, stroke, heart failure, peripheral arterial disease, aortic aneurysm, kidney disease, and vascular dementia.

The only way to find out if your blood pressure is too high is to have your blood pressure checked. Blood pressure is recorded using two numbers. The systolic pressure (upper number) is the force at which your heart pumps blood around your body. The diastolic pressure (lower number) is the resistance to blood flow in the blood vessels. They are both measured in millimeters of mercury (mmHg).

You can test your blood pressure at home using a blood pressure monitoring device, and your physician can also test this for you. In general, the guidelines for blood pressure are:

Blood Pressure CategorySystolic mm Hg (upper number)Diastolic mm Hg (lower number)
NORMALLess than 120andLess than 80
ELEVATED120 – 129andLess than 80
HIGH BLOOD PRESSURE Stage 1 Hypertension130 – 139or80 – 89
HIGH BLOOD PRESSURE Stage 2 Hypertension140 or higheror90 or higher
HYPERTENSIVE CRISIS Consult your doctorHigher than 180and/orHigher than 120

Generally, patients with Stage 1 hypertension are mostly recommended to make lifestyle changes to reduce their risks, but those with Stage 2 hypertension or higher normally have to take anti-hypertensive medicines to keep their blood pressure under control. This new research could potentially lead to better ways of managing high blood pressure.Hypertension is a very common condition worldwide. The World Health Organization estimates that high blood pressure causes 7.5 million deaths globally; this is over 12 percent of all deaths.

A new way to combat hypertension

A new study published in the journal Circulation showed that the protein galectin-1 influences the function of another protein known as the L-type (Cav1.2) calcium channel [1]. The L-type Cav1.2 calcium channel is found throughout the animal kingdom and is critical for CNS function, cardiac and smooth muscle contractility, neuroendocrine regulation, and many other processes.

In the arteries, the L-type Cav1.2 calcium channel causes the blood vessels to contract, and by lowering the activity of this calcium channel, the researchers showed that galectin-1 can reduce blood pressure. Given that hypertension leads to the development of other diseases, preventing it has the potential to halt or delay the development of many diseases at once.

Traditionally, calcium channel blockers (CCBs) are used to treat hypertension, but the problem with these is they can have serious side effects. Such medications can increase the risk of heart failure in people with hypertension, as they totally shut down the calcium channel. This makes the discovery here very important, because having a drug that can adjust the activity of the L-type (CaV1.2) calcium channel, not just shutting it down totally and preventing normal function, could prove to be a solution to hypertension.

There is more good news: Galectin-1 only targets the L-type (CaV1.2) calcium channel in blood vessels, meaning that it does not interfere with other calcium channels elsewhere in the other tissues of the body and so does not disrupt their function. The researchers believe that this signifies minimal side effects.

Conclusion

Ways to better control hypertension are certainly welcome, and while this does not address the age-related damage that causes blood pressure to rise in the first place, it is a potentially better solution than CCBs.

Literature

[1] Hu, Z., Li, G., Wang, J. W., Chong, S. Y., Yu, D., Wang, X., … & Colecraft, H. M. (2018). Regulation of Blood Pressure by Targeting CaV1. 2-Galectin-1 Protein Interaction. Circulation, CIRCULATIONAHA-117.

Increased Autophagy Promotes Healthy Longevity in Mice

A new study suggests that autophagy, the recycling, and disposal system that cells use to remove unwanted garbage, can extend the healthy lifespan of mammals.

The study, led by Drs. Salwa Sebti and Alvaro Fernández from the Center for Autophagy Research, has discovered that mice with higher levels of autophagy live longer and healthier than regular mice.

This study builds on decades of research suggesting that autophagy aids longevity and that the body’s gradual, age-related loss of autophagy likely drives an aging process. However, this question has remained unanswered: “Does increasing autophagy throughout the life of a mammal increase healthy lifespan?”

In order to answer this question, the research team created special, genetically engineered mice that were designed to have more autophagy than regular mice. They achieved this by creating a mutation in the already known autophagy-associated protein Beclin 1, a protein that reduces its binding to the Bcl-2 protein, which normally inhibits the function of Beclin 1. This resulted in the mice having a higher level of autophagy from birth.

The mice in the study had around a 10% increase in lifespan and were less likely to develop age-related diseases, such as cancer and age-related, pathological changes to the heart and kidneys. They also demonstrated that increased autophagy protects the mice from the premature death that normally occurs when mice lack the longevity-associated hormone Klotho.

Abstract Autophagy increases the lifespan of model organisms; however, its role in promoting mammalian longevity is less well-established1,2. Here we report lifespan and healthspan extension in a mouse model with increased basal autophagy. To determine the effects of constitutively increased autophagy on mammalian health, we generated targeted mutant mice with a Phe121Ala mutation in beclin 1 (Becn1F121A/F121A) that decreases its interaction with the negative regulator BCL2. We demonstrate that the interaction between beclin 1 and BCL2 is disrupted in several tissues in Becn1F121A/F121A knock-in mice in association with higher levels of basal autophagic flux. Compared to wild-type littermates, the lifespan of both male and female knock-in mice is significantly increased. The healthspan of the knock-in mice also improves, as phenotypes such as age-related renal and cardiac pathological changes and spontaneous tumorigenesis are diminished. Moreover, mice deficient in the anti-ageing protein klotho3 have increased beclin 1 and BCL2 interaction and decreased autophagy. These phenotypes, along with premature lethality and infertility, are rescued by the beclin 1(F121A) mutation. Together, our data demonstrate that disruption of the beclin 1–BCL2 complex is an effective mechanism to increase autophagy, prevent premature ageing, improve healthspan and promote longevity in mammals.

Conclusion

It is increasingly clear that enhancing autophagy is beneficial, and this study suggests that it should be safe to consistently increase autophagy. The results suggest that improved autophagy could help to combat neurodegeneration.

Rejuvenation Roundup May 2018

Summer is coming, and, albeit on a slightly longer timeframe, so is a world free of aging! So, grab an iced drink, sit comfortably on your beach chair, and let’s have a look together at some of the latest rejuvenation news.

The first LEAF conference in NYC is coming!

May saw us announce Ending Age-Related Diseases: Investment Prospects & Advances in Research, a special one-day conference taking place on July 12th in the heart of New York City. Join us for an action-packed day of research and biotech investment talks and panels from industry leaders as we build the longevity research and investment ecosystem!

We have confirmed the following exciting speakers for the event, and we invite you to visit our event page to learn more about them and the packed event program.

Dr. Aubrey de Grey – SENS Research Foundation Keith Comito – Lifespan.io Dr. Oliver Medvedik – Kanbar Center for Biomedical Engineering, Lifespan.io Dr. Vadim Gladyshev – Harvard Medical School Michael West – AgeX,  Biotime Joe Betts Lacroix – Vium Dr. Mark Hammond – Deep Science Ventures Ramphis Castro – ScienceVest Dr. Sudhir Paul – Covalent Bioscience Steven A. Garan – Center for Research and Education on Aging (CREA) Reason – Fightaging.org, Repair Biotechnologies Kelsey Moody – Ichor Therapeutics Bobby Brooke – Intervene Immune Dr. Antonei Csoka – Howard University Stephen Hilbert – Oisin Biotechnologies Dr. Alvaro Macieira-Coelho – INSERM (French Institute of Health and Medical Research) Dr. Vera Gorbunova – University of Rochester Dr. James Peyer – Apollo VC

To book your tickets, visit the Eventbrite ticket page today. We are also offering all Lifespan Heroes a 75% discount off the ticket price of the conference—our way of thanking you for supporting our work! If you would like to take advantage of this special offer, please either contact Javier or apply your discount code, if you know it, during the ticket purchasing process.

More great interviews

Our job here at LEAF is to bring the general public and the rejuvenation world closer together, and a great way to do that is through interviewing the people who are actually part of this world.

During UA2018, we talked to Dr. Nichola Conlon, CEO of Nuchido, a company that is soon to be launched and whose goal is to translate into human therapies some recent breakthroughs in the rejuvenation of aged mammals. Nuchido will investigate both NAD+ and senolytics for their potential as rejuvenative treatments.

We’ve also had the pleasure to have a chat with Dr. Sarah Constantin from the recently founded Longevity Research Institute. The LRI aims to identify, by 2030 at the latest, life-extending therapies that demonstrably work. As she explained in her interview, there are several compounds that are very good candidates for the job, yet not enough follow-up studies on them have been done; this is where the LRI plans to step in to change the situation.

We have also interviewed Dr. Matt Kaeberlein, one of the minds behind the Dog Aging Project. Dr. Kaeberlein and his team are working on canine aging and are hopeful that the treatments they’re testing might add up to five years to the healthy lifespan of dogs and cats.

Last but not least, we had an exclusive interview with Reason, the creator of Fight Aging!—quite likely the original rejuvenation advocacy blog. In occasion of the launch of Repair Biotechnologies, the biotech startup that he co-founded with Bill Cherman, we talked with Reason about his motivation to work in this field, his company’s plans, and his opinions on the current state of the rejuvenation industry and what we can expect from it in the future.

We’re nowhere near being done with interviews, so expect more in the coming months!

FA! digest

The debate over whether human adult neurogenesis occurs is far from settled, with evidence pointing in opposite directions; however, researchers have recently discovered that the flow of cerebrospinal fluid in the brain affects the activity of neural stem cells and thus the rate of neurogenesis. The study was reported and commented on FA! here.

Several compounds appear to have senolytic properties—that is, the ability to selectively prune senescent cells from the body without harming other cells. The mechanisms of some of these compounds are fairly clear to scientists, but others, such as piperlongumine, are much less so. As reported by Reason here, a group of scientists has recently published a paper on this compound.

It’s not at all clear why aging, a nearly universal phenomenon, seems to spare a few, lucky organisms such as the hydra—a tiny freshwater polyp that is functionally immortal, meaning that its risk of death, low to start with, doesn’t increase over time as it does for the rest of us. FA! reports a recent study in which researchers suggest that the lower structural complexity of the nuclear envelopes of the hydra’s cells might be a contributing factor to the tiny animal’s non-senescence.

Engineering better cells

Viruses, pretty much like cancer, have a great ally on their side: the power of evolution. Throwing drugs at them may well succeed at keeping them in check, but at the same time, it triggers an arms race where both we and they constantly strive to outsmart each other—which, when you consider that viruses manage quite well and yet aren’t even consciously trying, can be a bit frustrating. A way to end the war once and for all might be to engineer human cells that are resistant to viruses, and possibly other health threats as well, by recoding the human genome through the elimination of redundant codons. That’s what the Human Genome Project-write is set up to do, as explained in this article by Steve Hill.

More UA2018 videos

The organizers of UA2018, the very first conference of the Undoing Aging series, have uploaded three more videos of the talks that took place during the event last March. The newest entries are talks by AgeCurve Ltd. founder Attila Csordas, Director of the Centre for Healthy Ageing at the National University of Singapore Brian Kennedy, AgeX co-CEO Mike West, and Oisin Biotechnologies CSO John Lewis.

LEAF news

Besides crowdfunding, LEAF embarked on a new adventure last month: the Longevity Investor Network led by LEAF board member Javier Noris. This initiative is meant to bring together young rejuvenation biotech companies and potential investors in order to increase startups’ chances of making it through the initial development phase and bringing their ideas to the clinic. Any rejuvenation company opening an investment round may want to give this a shot!

Speaking of our amazing supporters, another one has recently joined the club and became a Lifespan Hero by pledging the very generous amount of 2,000 dollars per month. We’re immensely grateful to this anonymous contributor for his or her trust in us; this pledge is a great leap forward to the $10,000/month goal, which will allow us to organize an annual, full-scale longevity conference in New York City. Our deepest gratitude to all of our heroes, individuals and organizations who made a single contribution, too!

Two of our board members had their birthdays this month: Elena Milova and Steve Hill. They both celebrated in their own, unique ways. Steve organized an individual fundraising campaign on Facebook in support of LEAF; Elena celebrated in Astana, Kazakhstan at the Global Challenges Summit, where she was invited to moderate the longevity panel “Immortality begins now”, which featured Aubrey de Grey, Anthony Atala and Mikhail Batin. She was also a panelist of “Longevity as the new branch of the economy. Regulatory framework”.

As she explained in her opening speech in the first session, our desire to live a life free of diseases has been with us since the early days of our species, has survived in myths, and has been making its way into the reality of our healthcare system; if we put our minds to it, it may finally come completely true once aging is finally defeated.

A possible new weapon against progeria

Progeria—a syndrome that looks an awful lot like accelerated aging and strikes one out of every several million children at a very young age—is caused by a mutation in a gene called LMNA. In this short article on Long Long Life, Anne Fischer talks about progeria and remodelin, a new molecule that appears to ameliorate progeria in mice.

LE and SciFi

Have you ever noticed that, while even the most improbable—and sometimes straight-up impossible—things happen in science fiction, the defeat of aging is hardly ever presented as a future scenario? When the possibility of rejuvenation is touched upon, it generally only has a very marginal role in the story, and in a worst-case scenario, it ends up being demonized more or less overtly. We discussed a few examples of this interesting phenomenon and the possible reasons behind it in this article.

Speaking of science fiction, in the recent movie Avengers: Infinity War, the very same concern of overpopulation that some bring up in the context of life extension is the core motivation of the movie’s villain, Thanos. Thanos’ way of dealing with overpopulation is a little too drastic, as LEAF President Keith Comito explained in his newest article.

Aubrey de Grey on Chronicle Chats

On May 8, the Herbst Theatre in San Francisco hosted a conversation between Dr. Aubrey de Grey, CSO of SENS Research Foundation, and Prof. William Hurlburt from Stanford Medical School. The conversation, moderated by San Francisco Chronicle Editor in Chief Audrey Cooper, was focused on the topic of life extension and the ethical implications of potentially unlimited lifespans. The event was streamed live, but in case you missed it, you can still watch it here.

News from Ichor Therapeutics

Ichor Therapeutics, a company famous for its early-stage implementation of a LysoSENS-based approach to treating age-related macular degeneration, has recently published a paper about RPtag—an antibody-like scaffold with applications in protein manufacturing, diagnostics, and even clinical antibody therapy. RPtag’s resilience and stability make it easy to administer and preserve, even under extreme conditions.

In other Ichor-related news, on May 11, the rather aptly named stem cell-focused bioengineering company Ship of Theseus announced the initiation of a lifespan study on mice, which will be conducted at Ichor Therapeutics. Particularly, this study will be examining the effects of hematopoietic stem cell treatments on aging. Another lifespan study, this time by the Longevity Research Institute, has recently been announced and will be conducted at Ichor Therapeutics.

WHO Programme of Work: a certified victory!

Many of you will surely remember that, some time back, many organizations in the area of life extension were concerned that WHO had almost completely neglected to mention aging and the problems faced by elderly people in the draft of its Programme of Work. Thanks to the efforts of advocates in the LE community, LEAF included, WHO made a U-turn and included several provisions related to healthy aging in a newer draft of the same document. Today, we’re extremely happy to announce that the document has been approved and signed. Resolutions can be found here. This is no small achievement, but it was obtained with individual efforts: people like us were concerned about WHO’s choices and sent it feedback that asked for healthy aging to be included in its new Programme. This is a testament to how much we can achieve if we work together!

More acceptance of life extension even among religious people

One might think that religious people would all be completely against life extension, for ethical reasons among others, but it is not necessarily so. Early in May, during the Fourth International Vatican Conference, a panel on the morality of life extension was held featuring Reverend Nicanor Pier Giorgio Austriaco, Ph.D., 16th US NIH Director Francis S. Collins, M.D., Rabbi Edward Reichman, Elder Dale G. Renlund, M.D., and XPRIZE  Founder and Chairman Peter H. Diamandis, M.D. It might surprise you to learn that, while religious panelists weren’t eager to live to 250—as they understandably look forward to the afterlife in which they believe—they don’t really think that there’s anything unethical about life extension, and they seem to mostly agree on the benefits that it would bring to everyone.

Art for life extension

Dr. Laura Weston, a medical doctor, artist, and LEAF volunteer, has recently launched her own high-end art gallery featuring transhumanist themes and is supporting LEAF with proceeds from the sale of her work. We’re very grateful to Laura for her support, and if you are passionate about art as well as life extension, you may want to check out her awesome pieces.

In conclusion, we’d like to look back on this time period as the initial stage of an age-free future. Therefore, we’d like to thank researchers for their pioneering intellectual contributions, biotechnology corporations for bringing new, effective therapies into the clinic, fellow LE advocates for helping let the world know about rejuvenation biotechnology, and, of course, our Heroes for their consistent support!

A Health and Longevity Strategy

Who wants to lose weight, feel great, and live a long and healthy life, and what does it take to achieve these goals? Diet and exercise are equally important in long-term health, but let’s look at what recent science is telling us about the healthiest diets.

This article will rely heavily on University of Southern California professor Valter Longo’s work because I consider it to be the gold standard for nutrition research, and his recommendations in his book The Longevity Diet are well-supported with both data and good logic. Longo is the director of the Longevity Institute at USC and the IFOM Program on Longevity and Cancer in Milan.

He comes from an area of Italy known for very long lives, and part of his research focus has been looking at similar areas around the world and why those people live so much longer than normal.

Longo’s Longevity Diet

The short summary of Longo’s “Longevity Diet” is as follows:

  • Eat mostly plant-based foods, with occasional low-mercury fish or other seafood (2-3 times a week) and lots of nuts and olive oil
  • Generally eat modest amounts of protein, whether it’s plant-based or animal-based
  • Very limited or no dairy; goat’s milk and cheese are okay
  • Minimize saturated fats and sugar
  • Eat foods from your ancestral homelands as long as they are otherwise healthy
  • Eat 2-3 meals a day, ideally two solid meals and one snack, in a 10-12 hour window, and don’t eat 3-4 hours before bedtime
  • Take multivitamins every three days
  • A few times a year, if you are under age 65-70 and otherwise healthy, do a five-day water fast or a “fasting-mimicking diet,” which includes food but mimics the benefits of actual fasting.

Longo’s longevity diet is a plant-centric diet, which means that the large majority of what you eat is plants, fruits, nuts and legumes. It’s not going to kill you to eat occasional meat or dairy, but if you are under age 65, Longo recommends that you keep these very low or absent in your diet. However, if you are over age 65, Longo recommends eating higher portions of animal protein along with good sources of plant protein because our bodies are programmed to lose muscle mass as we age.

Longo argues against a strictly vegan diet or cutting out oil, as some researchers have recommended. He argues that consumption of fish, nuts and olive oil is associated with reduced risk of heart disease as well as very long lives in communities in Japan, Greece, and Italy (the famous “blue zones” where people are living to 100 years old far more than the average population).

There are definitely good ethical and environmental reasons to be a strict vegan, and a later piece will examine these arguments, but Longo argues that being strictly vegan, as opposed to mostly vegan, is not optimal for human health.

Longo advises strongly against the various “keto” diets that include high animal protein, high fat, and low carbs. While these diets will help you lose weight in the short term through increased ketogenesis, they are likely to lead to potentially serious health problems down the road.

Longo writes: “If we examine the laboratory studies, we see that both high protein intake and high saturated fat intake are associated with aging and disease, an additional and key vote against a high-protein, high-saturated-fat diet.” If there is any doubt, he adds later in the book that this kind of diet is “the worst of all possible regimens” for overall mortality risk, cancer risk, heart disease and diabetes.

In short, diet and good exercise may be the magic pill for health and longevity that isn’t available (yet) in actual pill form. Longo doesn’t promise immortality, of course, in following his recommendations. But he does suggest that we’ll maximize our chances of living to a hundred or more.

The new science of fasting

Author Michael Pollan got basic eating advice pretty much right in his well-known book The Omnivore’s Dilemma: “Eat food, not too much, mostly plants.” [1].

Unpacking Pollan’s statement a little, his point of view is that we should eat real, whole foods, not overly processed pretend foods; we should eat in moderation; and we should stick mostly to plants, fruits, and nuts, with occasional meat and fish to taste.

This is still great advice, but we are now at the point that, due to the efforts of researchers such as Longo, we can make more specific recommendations based on well-established science.

Longo would probably agree entirely with Pollan’s three-part mantra, though he doesn’t discuss it explicitly in his book. Where Longo goes beyond the mantra is in his recommendations on intermittent fasting and the fasting-mimicking diet.

Longo recommends eating within a 10-12 hour window each day, which is a type of intermittent fasting. He also recommends engaging in either a water (no food) fast or a fasting-mimicking diet, which achieves the same or similar results as actual fasting, two or more times a year.

The benefits of caloric restriction and intermittent fasting are now well-established. We don’t really understand yet why evolution built us this way, but it seems that when we stress our body by mimicking mild starvation, it responds by cleaning things up inside our cells as well as killing off sickly cells, and, more generally – as Josh Mitteldorf argues here – by slowing down many of the programmed aging aspects that are built into our genes. This includes increased telomere length, which I’ll cover in a later article.

Many times in his book, Longo stresses the need to consult with a doctor or other medical professional before engaging in any fast longer than a day.

Can diet really prevent or even cure disease?

This leads to what is perhaps the most groundbreaking area of Longo’s research: the fact that he and other researchers have now developed a body of work that shows remarkable preventive and even curative effects of diet, fasting, and exercise on many serious diseases. As he explains, “Genetic or dietary interventions can not only delay diseases but actually eliminate a major portion of chronic diseases in mice, monkeys, and even humans to extend longevity.” Mice fed various versions of Longo’s longevity diet have lived up to 40 percent longer than average.

Longo presents good data showing that the diet he recommends does not only lead to optimal weight and health but also provides strong protection against the major ailments of old age, including cancer, cardiovascular disease, diabetes, and even Alzheimer’s and other neurodegenerative diseases.

For example, Longo shows (citing a 2007 study [2]) that cardiovascular disease and cancer occur with remarkably lower frequency among Okinawans, and the Japanese more generally, than among Americans. Longo and the study’s authors pin the difference on diet.

I’m embarking on my own adventure using Longo’s longevity diet to improve my health and longevity. I’ll report back with results at a later date.

A key benefit of Longo’s diet recommendations is that they can be eased in and nothing needs to be eliminated entirely from your diet. For people who prefer not to go “cold turkey” on giving up things they like to eat, this can be a real benefit. At the end of his book, Longo includes a two-week meal plan for getting started on his longevity diet.

Eating a plant-centric diet can be very fulfilling and lead to all sorts of tasty new meals. Once you get past the notion that a “good meal” needs to have some kind of meat, a whole new world of tastes and satisfaction awaits.

Literature

[1] Pollan, M., & Andrews, M. (2015). The omnivore’s dilemma: The secrets behind what you eat. Listening Library.

[2] Willcox, B. J., Willcox, D. C., Todoriki, H., Fujiyoshi, A., Yano, K., He, Q., … & Suzuki, M. (2007). Caloric restriction, the traditional Okinawan diet, and healthy aging. Annals of the New York Academy of Sciences, 1114(1), 434-455.

Thanos, Overpopulation, and How to Save the Universe

Now that some time has passed since the release of Avengers: Infinity War, we should probably talk about Thanos, the nigh-omnipotent “Mad Titan” at the heart of Marvel’s latest blockbuster, and how he perfectly embodies one of the most pervasive societal misconceptions circling the topic of life extension. This might, therefore, be the first post here on LEAF that necessitates a spoiler warning, so here it is!

SPOILER ALERT — IF YOU CARE ABOUT HAVING INFINITY WAR, WHICH IS AN EXCELLENT MOVIE THAT YOU SHOULD SEE, SPOILED, PLEASE ABANDON SHIP AND HEREUPON RETURN AFTER WATCHING

Why Thanos Is Wrong

In Infinity War, the character of Thanos, his name itself an analog for the Greek death god Thanatos, is singularly focused on destroying half of all life in the universe as a means to “balance the scales”. Fairly textbook villain stuff, perhaps, but where it gets interesting and relevant is his reasoning behind this goal.

After witnessing the downfall of his home planet, Thanos reckoned that this culling is a necessary panacea to the anticipated side effects of overpopulation, such as scarcity of resources and environmental destruction.

Watching this play out, it is impossible not to think of the 18th-century cleric and demographer Thomas Malthus, whose influential 1798 treatise An Essay on the Principle of Population sounded dire warnings against the ills of a growing population – famine, war, disease, and poverty – as food production fails to keep pace.

While Malthus did not advocate genocide as a solution to this problem, opting instead for preaching abstinence and delayed marriages, his theories went on to inspire many draconian population control tactics throughout history, such as the Poor Law Amendment Act of 1834, denial of food aid during the Irish Potato Famine, and justification for the darker aspects of eugenics.

These ideas have also left a powerful mark on pop culture long before Thanos stepped onto the silver screen: Ebenezer Scrooge in A Christmas Carol initially stating that the poor should die quickly to decrease surplus population, contraceptive-dispensing “Malthusian Belts” in Aldous Huxley’s Brave New World, the villain from Dan Brown’s Inferno seeking to release a plague on the world to keep population at bay, etc.

Of particular note in recent history is the 1968 best-seller The Population Bomb by Paul and Anne Erlich, which predicted impending Malthusian doom for the entire planet and had a powerful impact on public discourse. To quote from the early editions: “The battle to feed all of humanity is over…at this late date, nothing can prevent a substantial increase in the world death rate.” As one specific case, for example, they stated that it would be impossible for the country of India to ever reach food sustainability, and they expressed support for government-mandated sterilization of all Indian males with three or more children as a solution to the problem.

So, what happened?  

It turns out that at the very same time the Ehrlich’s were preaching calamity, the Indian food production problem was already being solved. This was thanks largely to the work of Norman Borlaug, who used genetic crossbreeding techniques to create variations of wheat that were vastly more productive. This was the birth of the “Green Revolution”, and it is innovations like this that doomsayers like Malthus and the Ehrlich’s failed to predict. As this chart shows, the decline of poverty has been rapid thanks to technological and scientific progress.

When it comes to sustainability, the issue is far more complex than only the number of people on the planet: it is the efficiency of the tools that people use to generate resources from the land, how these resources are allocated, and the capacity for human ingenuity to anticipate and solve problems, as Norman Borlaug did. This is, fundamentally, what Thanos got wrong.

That being said, issues like inequity of resources, species extinction, and ecological destruction are legitimate concerns to be discussed and addressed, but their root lies chiefly in matters of policy, not population. A particularly salient example of this is the current situation in Venezuela, where its command and control economic structure has left the majority of the population susceptible to extreme resource shortage and poverty. This is actually an issue that is addressed directly in another recent Marvel blockbuster, Black Panther, with how certain characters critique the policies of Wakanda regarding the sequestration of wealth and resources. To quote William Gibson: “The future is already here — it’s just not evenly distributed”.

History is rich with examples of how improper management of resources and growing economic inequality leads to societal collapse; it is therefore on all of us to work towards policies and goals that overcome these eventualities. Call me optimistic, but I believe that we can do far better than Thanos in coming up with solutions to such challenges.

Why Thanos Is Stupid

In addition to Thanos being incorrect in his assumptions, it is also worth pointing out how his chosen solution is straight-up stupid — even if his overpopulation-as-the-root-of-all-evil assessment was true. Forbes and others have already run pieces explaining a few reasons why, but here’s a few more and some expounding on those already mentioned:

    • If the goal is to create a paradise where everyone’s basic needs are sustainably met, Thanos could simply use the Infinity Gauntlet to directly make this a reality rather than perhaps achieving this indirectly via population control. Indeed, one would think that randomly murdering half the population of every inhabited planet would yield highly unpredictable results. Thanos seems familiar with Earthling media, judging from his knowledge of Tony Stark, but apparently, he’s never watched the Leftovers.
    • Even if the only answer was to limit population growth, why not *snap* to sterilize a certain percentage of the population, or, better yet, make easy and effective contraceptives available to everyone and see how it goes for a bit.
    • Thanos acts as if once he accomplishes the “halvening”, his work will be forever done and he can enjoy the peace of a grateful universe. Due to the fact that birth rates historically have a far greater effect on population growth than death rates, however, Thanos would likely have to repeat his work every generation to keep total population stable over time. While it could, to be fair, turn out that Thanos timed his snap perfectly in terms of making Earth’s population sustainable over the long term, this timing is in no way guaranteed for every planet in the universe.
    • Even if the power of the Infinity Gauntlet is somehow unable to solve the problem of limited resources directly, Thanos could surely use it to create a Dyson Sphere around Earth’s sun and the stars of other inhabited solar systems — supplying effectively infinite energy for everyone in the universe. You might suggest that maybe he did not think of this, but the great forge Nidavellir, where Thor and Peter Dinklage fashion Stormbreaker, is essentially a limited Dyson Sphere, and we know Thanos has been there.

Why Thanos Could Use Some Coffee And Conversation

Despite his moniker as the “Mad Titan”, the version of Thanos we see in Infinity War is actually quite sane, measured, and even empathetic (in his own way). This leads me to believe that he, just as others who may think like him in the real world, can be reasoned with. For Thanos, the chain of his logic is understandable, given the starting point of his assumptions; however, it is in these assumptions themselves where he has gone wrong.

Thanos might be convinced to be reasonable.Perhaps if he were to be engaged on this directly, with earnest conversation rather than violent confrontation, he could come to see things differently.

It is easy to fall in love with the trappings of heroism: Cap’s shield, Iron Man’s lasers, drawing a sword to slay a dragon. Sometimes, this is necessary, but, sometimes, being a hero means seeking to understand the real problem that both you and your “enemy” are trying to solve and finding a way forward together.

Who knows; this could indeed be how the universe is restored in Avengers: Infinity War Part 2. In the meantime, if you see him, please point Thanos my way; I’d love to share some coffee and talk over some graphs.

And if you enjoyed this discussion about why Thanos was wrong, there is also a video on the topic over at Youtube Channel WorldofGeekdom.

Matt Kaeberlein interview

Dr. Matt Kaeberlein – The Dog Aging Project

The Dog Aging Project (DAP) has sparked wide interest, and today, we have the pleasure to bring you an interview with one of the lead scientists behind it, Dr. Matt Kaeberlein.

In much the same way that other projects are aiming to extend healthy human lifespan, the DAP team intends to do the same, targeting the aging processes directly. Given the rate of progress in geroscience over the past years, Dr. Kaeberlein and his team are optimistic that, in the near future, the interventions that have been shown to slow down aging in mice and rats could do the same in our furry companions.

Besides Dr. Kaeberlein—who is a Professor of Pathology and an Adjunct Professor of both Genome Sciences and Oral Health Sciences at the University of Washington in Seattle—the DAP team is comprised of Dr. Daniel Promislow, Professor of the Departments of Pathology and Biology at the University of Washington; Dr. Kate Crevy, Associate Professor of Small Animal Internal Medicine at Texas A&M University’s College of Veterinary Medicine; Dr. Tammi Kaeberlein, a research scientist at the Department of Pathology at the University of Washington; Dr. Silvan Urfer, a veterinarian and Senior Fellow of Washington University’s Department of Pathology; and Kelly Jin, a doctoral student currently pursuing a Ph.D. in Molecular Medicine and Mechanisms of Disease at the University of Washington.

The team believes that extending our pets’ healthy lifespan will have benefits in both their and our lives, which is perfectly understandable. We all would like to spend a longer time with our loved ones—and these rightfully include our pets—in the prime of their and our health. To this end, DAP intends to run a longitudinal study of aging in pet dogs and is running a rapamycin intervention trial in dogs, phase 1 of which has already been completed. Rapamycin is an mTOR pathway inhibitor.

Dr. Kaeberlein was kind enough to answer our questions about the DAP’s story and work; without further ado, here’s what we talked about.

Could you tell us the story of DAP? How did it all start?

About 5 years ago, I was able to recruit Daniel Promislow to the University of Washington to join the UW Healthy Aging and Longevity Institute. At that time, Daniel had recently obtained a small grant from NIH to develop companion dogs as a model to understand the genetic and environmental determinants of aging. After a series of discussions with Daniel, it occurred to me that we had an opportunity not just to study aging in dogs but to potentially develop interventions to delay or even reverse aspects of aging in dogs from those that had already been shown to increase lifespan and healthspan in laboratory rodent models.

I decided to focus on rapamycin first, because it was (and still is) the most validated and effective pharmacological approach for increasing longevity in mice, and it has the added benefit that it is effective even when initiated in middle age. After spending a couple of months convincing myself that we could safely perform a rapamycin veterinary clinical trial in dogs, I organized a conference in Seattle in 2014, where I pitched the idea. Soon after that, we started getting quite a bit of media attention, and we decided that we should officially form the Dog Aging Project, which, at the time, consisted of myself, Daniel, my wife Tammi, and our lead vet, Kate Creevy. Tammi quickly built the website, and I started putting together funds for our Phase I rapamycin intervention trial.

What can you tell us about trials you’ve already run and their results?

So far, we’ve only completed one trial, a 10-week, randomized, double-blind, placebo-controlled study of rapamycin in pet dogs. The results of that study were as positive as we could have hoped.  We saw no evidence for increased side effects in the dogs that received rapamycin and statistically significant improvements in two of the three measures of age-related cardiac function that we looked at.

Are there any trials you’re running right now or are preparing to launch soon?

Yes, the Phase 2 rapamycin intervention trial is currently enrolling dogs. That trial is funded by the Donner Foundation and is a one-year trial to, again, assess effects of rapamycin on cardiac function and to also look at effects on cognitive function and activity.

Depending on the outcome of our submitted NIH grant, we hope to begin officially enrolling dogs into the Longitudinal Study of Aging and Phase 3 of the rapamycin intervention trial toward the end of 2018 or early 2019. We hope to have an official announcement on the outcome of that proposal within the next 3-4 weeks.

Can I volunteer my dog for the program, and how do I do that?

Anyone can nominate their dog to participate in either the Longitudinal Study of Aging or the Rapamycin Intervention Trials through the Dog Aging Project website at www.dogagingproject.com. The Longitudinal Study is currently open to all breeds, ages, and sizes of dogs. The Rapamycin Intervention Trials are restricted to healthy dogs of at least 6 years old and at least 40 lbs in weight.

What are the most promising lines of intervention that you are studying to extend the healthy lifespan of dogs?

Currently, rapamycin is the only intervention we are testing due to budgetary limitations.  I have recently begun pitching the idea of a Companion Animal Intervention Testing Program to take the most promising interventions from basic geroscience research and test them in pet dogs as long as that can be done safely. Obvious candidates would include NAD+ precursors, metformin, acarbose, deprenyl, 17α-estradiol, spermidine, and NDGA.

On your website, you say that the possibility to increase the healthy lifespan of our pets by up to five years is within our reach. Could you give us an estimate of how long it’ll take before these therapies might be available for our pets?

I’ll answer that assuming you want to know how long it will take to obtain scientifically rigorous evidence that an intervention is working. There is nothing stopping an owner from giving NAD+ precursors or rapamycin to their pet if that person can identify a vet who will prescribe it.

Based on our power calculations, we anticipate that it will take about 3 years of treatment, beginning in a middle-aged cohort of appropriate size, to detect significant extension of lifespan. Our Phase 3 study plans to enroll dogs over the first year and a half, so the total timeframe is 5 years, start to finish. Other changes, such as improvements in heart function, can be detected much more rapidly.

Those same power calculations and design features are equally applicable to other interventions. When and if we are able to test them will depend on funding.

What about cats; do you think we might do the same for our pet cats as well as dogs?

In general, I expect that any intervention that slows aging in dogs will have a similar effect in cats, although it is the case that certain compounds are toxic in cats and not in dogs and vice versa. I know that rapamycin has been used in cats for other indications, and there’s nothing about the biology of mTOR that makes me think that rapamycin will be less effective in cats than it is in dogs. From a clinical trial perspective, it makes sense to start in dogs for a couple of reasons. First, big dogs age faster than cats, to the outcome is knowable in a shorter time frame. Second, it’s easier for owners to give a pill to their dogs than to their cats.

How expensive do you imagine the first generation of anti-aging therapies for pets will be? Can we expect them to eventually become affordable?

If rapamycin is shown to be effective at slowing aging in dogs, I think it will become quite affordable. The cost of rapamycin currently is largely driven by relatively low demand, but it is off-patent, and generic versions are available. If there is sufficient demand to drive competition, prices will drop substantially. Even at current prices, it would probably cost an owner $50-200/month, depending on the size of dog and dose given, which is affordable to many people. Some other candidates from the list I suggested are even cheaper than rapamycin.

Recently, Prof. George Church co-founded a new start-up, Rejuvenate Bio, which is poised to reverse aging in dogs using gene therapy in the hopes of eventually doing the same in people. What do you think of this approach?

I think the approach is viable in theory and could turn out to be quite powerful in the future, but it has yet to even be proven in a laboratory mouse model of aging. Show me that you can increase the lifespan and healthspan of a mouse by 25% through this approach without significant detrimental effects, and I’ll be more optimistic. Reproduce it in a dozen labs as has been done with rapamycin in mice, and I’ll jump on board the bandwagon.

I don’t know precisely what they are planning, but I do have major concerns about the safety of applying gene therapy to healthy dogs at this stage. In general, I would urge anyone considering performing intervention studies in companion animals to recognize how important it is to ensure that you don’t harm anyone’s pet.

I suspect that they will start with specific diseases of aging, and I’ve read that they are planning to attempt some sort of gene therapy to treat heart disease in Cavalier King Charles Spaniels. Again, I would like to see proof of principle that such a therapy can work in a laboratory model of valvular regurgitation (the primary problem in that breed of dogs) before moving to testing in people’s pets.

It is clear that in the last decade, there has been a real leap in our understanding of what aging is and the processes and potential points of intervention. While we do indeed have lots more work ahead of us before the medical control of aging is plausible, are you optimistic about progress in the field?

Absolutely. I’m more optimistic than I’ve ever been, both because of the scientific advances and because of the growing recognition and respect that geroscience is receiving among the broader scientific community and the biotech/pharmaceutical world. I do have concerns that the field continues to be hindered and progress is delayed by fringe elements who give us a bad reputation among funders and policy makers, but we are gradually overcoming this as real interventions that have been validated in preclinical studies begin to make their way to the clinic.

What are some of the key studies that convinced you that doing something about aging was plausible?

I never really doubted it once I became knowledgeable about the field.  It seems obvious that longevity is quite malleable – all you have to do is look across the animal kingdom to see that. We’ve known for many years that single-gene mutations can alter aging rates in every laboratory species that we’ve examined, and it’s increasingly becoming clear that it’s not that hard to find them. So, aging is malleable at the species level as well. The big unknown in my mind is how well laboratory interventions will translate into the real world. I think that this is where companion animals can help immensely, since a true longevity clinical trial in people is simply not feasible.

If I have to point to one study though, it is the ITP study showing that rapamycin treatment increases lifespan in mice when started in middle age. Before this, everyone was testing interventions beginning in young age, which is obviously not translationally viable. Now, we know of several interventions that appear to have effects in middle age in mice (NAD+ precursors, senolytics, “young blood”, etc.) and that all of these interventions probably not only slow aging but actually rejuvenate function in at least some organs and tissues in aged animals. In the case of rapamycin at least, there is now initial evidence that mTOR inhibition can rejuvenate immune function in healthy elderly people as well. I would be surprised if other effects of rapamycin in mice don’t translate just as well.

Currently, one of the most highly regarded theories of aging is the 2013 Hallmarks of Aging, which describes the various aging processes with particular emphasis on humans. How relevant to humans is your research in dogs; do they share the same aging processes?

Everything we know indicates that dogs age very much like people, only 7-10 times faster. Dogs get all of the same diseases of aging that people do, although not necessarily at the same frequency. Vascular disease, for example, is a bigger killer in people than in dogs.

Do you think that your work may prove useful to scientists working on therapies against human aging?

For the longitudinal study of aging, undoubtedly.  Any environmental or genetic factor that we can correlate to healthy aging in dogs will be a high-priority candidate for validation in humans.

If we are successful at showing that rapamycin (or another intervention) can increase healthy longevity in dogs, this will demonstrate that the intervention works in a large mammal that shares the human environment. It’s an important next step toward human application. Perhaps equally important is that this should be quite helpful in terms of convincing the FDA and other regulatory bodies that aging is a viable endpoint for treatment. Keep in mind that the FDA also regulates veterinary medicine. While it’s not feasible to do a lifespan study in healthy elderly people, it is completely feasible to do several of them in healthy elderly dogs, so a true clinical trial with longevity and/or healthspan as the endpoint is doable.

In addition, if we are able to convince owners that it is possible to slow aging in their pets, this should accelerate acceptance of and support for geroscience among the general public.

Clearly, the DAP team thinks that we can at the very least slow down aging; do you think we might be able to reverse its effects, potentially postponing “the unpleasantries of extended life”, as Prof. Jay Olshansky once put it, indefinitely?

It’s clear that most of the interventions I’ve mentioned above can restore function to at least a subset of aged organs and tissues. I don’t know if this is really “reversing aging” as a whole, but it is at least functionally rejuvenating some aspects of aging.

Whether this can be done indefinitely is pure speculation at this point. Is it possible? Probably. Is it likely in the near future? I’m doubtful, but I’ve been wrong before and I wouldn’t mind being wrong now. My personal opinion is that it’s irresponsible and damaging to the field for people to talk about immortality being a reality in X number of years from now. These people simply don’t understand the complexity of biological aging if they think they can put a number on how long it will take to achieve immortality when the best we’ve been able to do in a rodent is a 50% extension of lifespan. That’s not much better than Clive McCay managed in 1935.

Some researchers suggest that aging is a disease or, more specifically, a co-morbid syndrome; would you agree with this or not, and why?

Frankly, I think it’s a semantic argument that some people in the field spend way too much time arguing about. Despite what some people think, it doesn’t matter from a regulatory perspective, since aging could be an indication if you had sufficient quantitative endpoints to prove efficacy of an intervention.

I think the more fundamental question here is whether aging is actually a molecular process that can be targeted or a collection of unrelated ways that different organs and tissues break over time to cause different diseases of aging. To me, the answer is obvious. You can’t explain the unrelated processes model given that we know about single gene mutations that not only extend lifespan but also delay most (if not all) of the functional declines of aging.

I think what we still don’t quite have a handle on is whether aging at some level can be boiled down to one molecular process/pathway or if it will turn out to be a collection of processes (i.e. the current Hallmarks framework). In any case, there are key nodes in the network that you can hit, such as mTOR, that appear to affect all of the Hallmarks simultaneously, so, at some level, there is a unifying mechanism.

The idea of human life extension is often met with fierce, irrational opposition. In your experience, do people react in a similar way to canine life extension, or are they more supportive of your work?

I’ve never really experienced fierce opposition to the idea of increasing healthy longevity in pets. Some people do express concern that there are already many pets in need of homes and wonder if this might make it harder to find good homes for those pets. The point I make in the context of targeting aging, for both dogs and people, is that this is really no different in principle than what we already do. If your dog or your spouse has cancer, your goal is to treat the cancer to keep them alive longer. We are just trying to do it in a more efficient and humane way by preventing our dogs and our loved ones from getting sick in the first place. When explained that way, most people get it.

In the past few years, senolytics have made the headlines more than once as the first bona fide rejuvenation therapies that might become clinically available for people. Are you optimistic about senolytics as a potential way to address aging in both dogs and humans?

Definitely. I think there are still some concerns about safety with the current generation of senolytics, but I expect that this will be improved upon. I do wonder if they will be any more effective than rapamycin, however. So far, there’s not much evidence that I’m aware of to suggest that will be the case. The excitement may have gotten a bit ahead of the data in that regard. It will be important to test combination treatments of senolytics and mTOR inhibition in mouse models to address this question.

What do you think is the greatest bottleneck to progress in aging research?

Funding and perception, and these two are related. There is insufficient funding for scientific research in general and insufficient funding for geroscience in particular. That has been the biggest bottleneck to research progress. The field has also suffered from a reputation problem for many years as not being particularly rigorous, and this has hurt the flow of research dollars into the field. This has been changing but is still a problem.

Do you have a take-home message for our readers?

I think we know enough about the biology of aging to increase healthy longevity in pet dogs by 3-5 years today. Almost certainly, one or more of the interventions we could test today would be successful. I know that we all want progress in translational geroscience to be as rapid as possible, and I believe that rigorously demonstrating that we can increase healthspan and lifespan in pet dogs will be a huge step toward gaining the support and credibility that the field needs.  Plus, I want my dogs to live longer ☺.

We thank Dr. Kaeberlein for taking the time to talk to us, and we look forward to seeing the future results and developments of the Dog Aging Project.

Brain synapses

Alzheimer’s Reversed by Editing a Single Gene

 

Researchers at Gladstone Institutes in San Francisco report that a gene variant associated with Alzheimer’s works differently in mice and humans, and they also demonstrate how modifying this gene could potentially prevent the plaques associated with Alzheimer’s from forming and damaging the brain[1].

An ApoE4 gene variant is associated with Alzheimer’s disease

The gene apolipoprotein E3 (ApoE3) has a variant known as ApoE4, which is associated with the development and progress of Alzheimer’s disease. People with just one copy of the ApoE4 gene are at twice the risk as people without this gene variant. Some people even have two copies of the ApoE4 gene, which makes their risk of Alzheimer’s a staggering twelve times greater.

Both the ApoE genes produce their own forms of ApoE protein, which differ in structure. The ApoE4 protein is fragile and fragments because it cannot function in the same way as the regular ApoE3 protein in human nerve cells. These fragmented protein pieces are associated with the increased production of amyloid-β peptides and tau phosphorylation that are typical of Alzheimer’s disease.

The researchers wanted to find out how ApoE4 is linked to Alzheimer’s disease. They considered the possibility that the increased amyloid-β and tau phosphorylation from the fragmentation of ApoE4 drives disease progression. Another possibility involved the negative consequences of a lack of ApoE3 proteins, as they were replaced by the ApoE4 variant. The team also considered a combination of both of these possibilities.

The team investigated these potential answers by examining the effects of ApoE3 and ApoE4 on human nerve cells. Neurons were created by using pluripotent stem cells taken from volunteers who had either two copies of ApoE3 or two copies of the ApoE4 gene, and the researchers programmed these cells to become the desired type of neuronal cell.

The team compared the ApoE3 and ApoE4 neurons against neurons that were unable to produce either type of the ApoE protein. They discovered that neurons that produced no ApoE protein worked the same as those that produced ApoE3. This confirmed that it was not a lack of ApoE3 causing the problem but that ApoE4 protein alone was linked to Alzheimer’s disease.

This finding also sheds light on why treatments for Alzheimer’s that work in mice fail to translate to humans. The production of amyloid-β in mice is not influenced by ApoE4; this means that treatments that prove effective in mice may not work in humans, as the mouse models of the disease do not emulate the ApoE4-related form of Alzheimer’s that humans get. However, therapies that focus on reducing amyloid-β have worked in mice[2], so while ApoE4 functions differently in mice and humans, this is not the full story of Alzheimer’s. This research does, however, clearly show a way in which mouse models differ from humans, helping to guide future research.

Converting ApoE4 to ApoE3

The results of the study suggest that therapies that seek to modify the ApoE4 gene protein before it fragments might be a way to combat Alzheimer’s. This is how traditional medicine would generally approach the problem, treating the symptoms and not the cause.

However, the researchers took this one step further to a far more robust solution. Rather than simply attempting to treat the consequences of having an ApoE4 gene producing sub-par proteins, they completely removed the problem by using gene therapy to edit the genes and convert them from ApoE4 to ApoE3.

The converted genes ceased to produce the unstable ApoE4 protein and produced the stable ApoE3 version of it instead. This served to correct the problem at the root rather than trying to slap a band-aid on the consequences.

Conclusion

With so many failures to combat Alzheimer’s disease, it is easy to become disillusioned. We have seen mice cured of the disease numerous times, but these cures have failed to translate to humans. These new findings help to progress knowledge in the field and offer potential new ways to defeat Alzheimer’s.

What is refreshing about this study is how the researchers have opted to attack the problem at the root cause: the production of misfolded proteins that lead to the progression of the disease. It is becoming ever more clear that if we are going to make progress on ending age-related diseases, we must target the aging processes themselves, which cause these diseases.

Literature

[1] Yuang, Y. et al. (2018) Gain of toxic apolipoprotein E4 effects in human iPSC-derived neurons is ameliorated by a small-molecule structure corrector. Nature Medicine doi:10.1038/s41591-018-0004-z

[2] Hu, X., Das, B., Hou, H., He, W., & Yan, R. (2018). BACE1 deletion in the adult mouse reverses preformed amyloid deposition and improves cognitive functions. Journal of Experimental Medicine, jem-20171831.

NAD+ Regulates the Creation of Fat Cells

Researchers from the University of Texas have discovered how NAD+ creation and consumption in cells is linked to glucose metabolism and their transformation from preadipocytes into adipocytes, or as most people know them, fat cells.

INTRODUCTION Nicotinamide adenine dinucleotide (NAD) is an essential small molecule that is involved in a variety of physiological and pathological processes. The oxidized form, NAD+, serves as a cofactor in metabolic pathways, as well as a substrate for various enzymes that consume it, such as the poly[adenosine diphosphate (ADP)–ribose] polymerases (PARPs) and sirtuins (SIRTs). PARPs and SIRTs cleave NAD+ into nicotinamide and ADP-ribose, resulting in the irreversible breakdown of NAD+. Therefore, the resynthesis of NAD+ is necessary for maintaining normal cellular functions. Increasing evidence has revealed that (i) reduced NAD+ levels result in altered metabolism and increased disease susceptibility and (ii) restoration of NAD+ levels can prevent disease progression. Thus, understanding NAD+ synthesis and catabolism is important for understanding physiological and pathological processes.

NAD+ regulates adipogenesis

Adipogenesis is a process of cell transformation, called differentiation, by which preadipocyte cells become adipocyte (fat) cells. The new study, published in the journal Science, shows how glucose is converted into fat and how NAD+ plays a key role in this process. During the study, the research team looked at why consuming too much glucose makes the body produce fat and results in weight gain.

Previous studies have demonstrated that NAD is a key molecule that plays a role in a myriad of cellular and pathological processes. The oxidized form, NAD+, has been shown to act as a cofactor in metabolic pathways and is consumed by a number of enzymes involved in gene regulation.

After NAD+ is consumed, it is broken down into nicotinamides and ADP-ribose; the researchers concluded that this means that NAD+ must be resynthesized following this in order for normal cellular functions to continue. This converges with previous studies, which suggest that NAD+ decline leads to changes in metabolism and an increased risk factor for some diseases; this typically happens as the result of aging, as NAD+ levels begin to fall.

With this in mind, the research team thought that cellular metabolism and gene regulation were potentially connected to NAD+ synthesis. They discovered evidence suggesting that compartmentalized NAD+ synthesis and the subsequent consumption are integrated with glucose metabolism and adipogenic transcription as part of the adipocyte differentiation process.

NAD+ synthesis acts as a mediator of PARP-1-regulated transcription during the differentiation of adipocytes, linking cellular metabolism and the adipogenic transcription process. During adipogenesis, nuclear NAD+ levels fall, causing the induction of NMNAT-2, the cytoplasmic NAD+ synthase. This increased level of NMNAT-2 then reduces the availability of NMN and leads to a reduction of nuclear NAD+ synthesis via NMNAT-1. The drop in NAD+ levels then results in decreased PARP-1 activity, which then reduces levels of inhibitory ADP-ribosylation of the adipogenic transcription factor C/EBPβ. Reduced ADP-ribosylation of C/EBPβ means that it is able to bind its target genes, thus promoting the differentiation of preadipocytes into adipocytes. In other words, a decline of NAD+ encourages an increase of preadipocytes turning into adipocyte fat cells.

The researchers found that directly decreasing NAD+ synthesis significantly reduced the activity of PARP-1 and increased adipogenesis. They also found that blocking the decline of NAD+ levels reduced adipogenesis and the creation of fat cells. Finally, supplying exogenous NMN to preadipocytes bypassed the competition to consume NMN between NMNAT-1 and NMNAT-2; this led to an increased level of nuclear NAD+ synthesis during the differentiation process, which increased PARP-1 activity and ultimately blocked adipocyte differentiation. In summary, providing higher amounts of NAD+ precursor NMN reduced the creation of fat cells.

Conclusion

This study shows that compartmentalized synthesis of NAD+ regulates the differentiation of adipocytes, a process that is critical for metabolic health, via transcriptional control. It also explains why a decline of NAD+ results in an increasing amount of fat cells and weight gain.

Literature

[1] Ryu, K. W., Nandu, T., Kim, J., Challa, S., DeBerardinis, R. J., & Kraus, W. L. (2018). Metabolic regulation of transcription through compartmentalized NAD+ biosynthesis. Science, 360(6389), eaan5780.

The right to die and the right to live

On May 10 this year, Australian ecologist David Goodall took his own life before aging could. The scientist, aged 104, reportedly said he “regretted” having reached that age, because the quality of his life had significantly deteriorated as a consequence of his declining health. Unhappy with his condition, though not suffering from any terminal disease—except for aging itself—Goodall opted to end his life through assisted suicide. As the practice is currently not allowed in Australia, he flew with friends and family all the way to a clinic in Switzerland, where he flipped a switch and administered his own lethal injection while listening to Beethoven’s Ninth Symphony. Interestingly, the cost of his trip to Switzerland was covered with money collected through a crowdfunding campaign.

A matter of rights

Goodall was a lifelong supporter of euthanasia, and both he and his fellows from Exit International—a non-profit organization advocating the legalization of voluntary euthanasia and assisted suicide—said that people should have the right to die with dignity when they feel the time has come.

As he himself put it, the elderly scientist didn’t have much time left anyway, and, given the circumstances, this is an understandable choice. Given the current state of rejuvenation medicine, there was no chance he could ever benefit from it; the odds are that he might have never heard of it. This was his personal choice, and I doubt I would have made the same one, but there is rather little point to life when it is reduced to prolonged suffering with no hope of improvement and a near certainty of worsening. This suffering is both physical and psychological, as in advanced old age, you have to come to terms with the fact that the person you used to be is really no more and what you loved doing is now beyond your capabilities. If the situation really is so dire and grim, and all hope for a better life is utterly lost, then euthanasia should be an available option. Hopefully, cryonics might one day become a reliable lifeline to make euthanasia unnecessary, but as your life really is your own and nobody else’s, you should have the right to decide to terminate it if you see fit, provided your judgment is not impaired in any way.

On the page of Dr. Goodall’s crowdfunding campaign, there is a striking bolded sentence. It reads “All rational adults deserve a peaceful death at a time of their choosing.” Assuming that a person desires death for whatever reason, there’s no reason why it shouldn’t be peaceful; why make it worse than it has to be? (As a side note, the fact there are organizations campaigning for the right of people to have a “good death”—which is exactly what the Greek word “euthanasia” means—betrays the already obvious fact that death isn’t generally very good.) Making death peaceful is already well within our technical abilities; the only obstructions are some obsolete legal frameworks. However, it’s the “at a time of their choosing” part that’s worrisome—as things stand, you can choose to hasten your death, but you can’t really choose to postpone it. If a rational adult chose age 200 as his or her preferred time of death, this right to choose couldn’t currently be honored. Shouldn’t we fight for this right as well?

More similar than you think

Naturally, the battles for euthanasia and for life extension are fought against two different foes. The former is a battle against inconsiderate laws; the latter is a battle against the limitations that nature didn’t bother removing. However, the two causes share a common goal.

Crowdfunding someone’s death is, intrinsically, somewhat disturbing, and there is a stark contrast between the kind of crowdfunding that Lifespan.io does, which is aimed at giving people longer, better lives, and what Exit International did in this case. However, the goal of the crowdfunding campaign for Goodall’s trip to Switzerland wasn’t that of terminating his life; the goal was terminating his suffering, and, at that point, this goal could only be achieved through death. Because of this, I commend Exit International and the campaign backers for allowing Goodall to free himself of his pain as he wished.

The shared goal of life extension and euthanasia is to end pointless suffering. Life extension, being basically nothing more than next-generation medicine, is meant to eliminate unnecessary suffering the old-fashioned way: eliminating the diseases that cause it. Unlike other types of medicine, though, the diseases in the crosshairs of life extension are age-related diseases, and to prevent them, it is imperative to attack aging itself. On the other hand, euthanasia is an extreme solution, the last resort that may be used when all else has failed and a person is ready to pay the highest price to be relieved from useless, unbearable suffering.

Because of this shared goal, it’s logical to believe that people willing to donate to ending Dr. Goodall’s suffering would also donate towards ending age-related diseases, thus helping to prevent the terrible suffering that results in euthanasia becoming a sensible choice.

The obvious reality of aging

In some pictures taken shortly before his death, Goodall sports a jumper with a tag reading “Ageing disgracefully”. His arthritis was bad enough that, in a somewhat bitter twist of irony, he didn’t even manage to flip the lethal injection switch the first time. This jumper refers to the concept of “aging gracefully”, which is largely a myth. A healthy lifestyle, combined with winning the genetic lottery, may certainly make your aging less worse than others’; however, the chronic diseases of aging don’t all happen because most people have unhealthy lifestyles or got unlucky with their genes. They happen because aging is, by definition, a chronic, progressive process of deterioration and loss of functionality. However “gracefully” you may be deteriorating—whatever that means—it will always, invariably, kill you. Think about it: however gracefully or disgracefully aging is happening in your body, it is always, with absolutely no exception whatsoever, serious enough to eventually cause death.

Sometimes—too often, in fact—aging is disgraceful enough that not everyone is willing to wait until the end. Goodall himself waited quite a while, but he eventually decided to check out before aging could make his life any more miserable. The popular comedian Robin Williams, struck by an unusually aggressive form of Lewy body disease, did so much earlier and in a more tragic fashion—he hung himself at age 63. He couldn’t bear with how his condition was eating away his mental faculties.

Neither Goodall nor Williams were alone. Although suicide rates in general have been slowly but steadily decreasing during the past 30 years or so, suicide rates among the elderly are, by far, the highest in the world, as can be seen from the chart below (courtesy of Our World In Data).

In 2016, 27 out of every 100,000 people aged 70 and older took their own lives. If that doesn’t seem like much, the grand total of people in that age cohort who committed suicide that year was over 110,000. Given that aging kills just about that number of people in one day, it’s a bit as if 2016 had one extra day when everyone over the age of 70 who died did so at their own hand. From the same source, here is the breakdown of suicides by age range since 1990.

Younger people constitute a larger portion of the total suicides each year—somewhat unsurprisingly, since at the moment world population is comprised of far more people below age 60 than above—but suicides from age 50 and up still accounted for nearly 40% of the total in 2016.

Research on suicide among the elderly is still somewhat neglected, though loss, loneliness, and psychological as well as disability and physical disease are all contributing factors, particularly among older white men, who are especially likely to take their own lives [1]. Despite the lack of data, you don’t have to be a clairvoyant to realize that losing your friends and relatives (who are dying of aging just every bit as you are), being disabled, having a chronic illness that can only get worse, and feeling cut out from the world all have extremely good chances of making you depressed and your life less worth living; worse, they can make it unbearable to be alive.

Double standards

The general attitude toward death is interestingly ambivalent. The WHO website speaks of suicide in terms of something that can and should be prevented. We have suicide hotlines that you can call for help if you’re having suicidal thoughts. Euthanasia, as we have seen, is a very controversial topic, and it’s frowned upon and largely prohibited even when patients actively don’t want to live anymore and there’s no hope for their conditions to improve. This might be due to the rather clear fact that, if a person wishes to die, there’s quite likely something very bad going on in his or her life, and as there’s no going back once the plug has been pulled, we feel that it’s advisable to try to solve whatever problem is causing suicidal wishes—especially since people who feel suicidal may easily see reality as being far bleaker than it is, their judgement being clouded by their intense desire to put an end to their pain.

You’d think that, given this premise, we should be a society that strives to prevent death by all possible means, putting more effort into addressing the leading and more widespread causes of death. This is indeed what we do in most cases; aging is the only exception to the norm. If someone was letting himself die by starvation, no matter his age, we’d call that suicide and urge him to seek help; we’d do the same if someone was letting herself die by refusing treatment for a lethal though curable condition. On a grander scale, though, most people don’t consider the acceptance of age-related death to be suicide; they call it the “natural cycle of life”, the “completion of an arc”, “walking into the sunset”, and similar things. Why is that? If we, as a society, insist that people don’t give up on their lives, however hard the times they’re going through may be, why do we demand that everyone should give up on life in old age, rejecting the idea of even trying to prevent all these deaths? If a young person, however sick or desperate, should not let go because he or she still has so much time ahead and there’s so much left to accomplish, why should we consider the life of an 80 year old to be “complete” and ripe for termination by aging? Instead, shouldn’t we attempt to give this person the same health of a youngster and abundant time to live? The idea itself that life could ever be complete, as if it were a sticker album, is laughable—besides, when you complete an album, you can always get a new one if you want to. Maybe, one day, you might feel like you’ve had enough of attaching stickers, but you should have the option to keep going if you so wish.

The third option

Goodall was faced with only two options to end his suffering: waiting for his health to deteriorate enough to cause his death, making whatever was left of his life even more miserable, or choosing a quick, painless death. He chose the latter course of action because his failing body was making him unhappy. However, what if his body hadn’t been failing him? What if he had been in perfect health, completely able-bodied and disease-free? Would he have felt that the time was ripe for him to die just because he was 104? This would be for him, and others in the same situation, to decide, but he didn’t have this chance. None of us do. We can’t currently decide to live for however long we see fit; in a best-case scenario, we can only choose which way to die. Life extension biotechnology could give us a third option: preserving our health, potentially indefinitely, allowing us to choose life rather than the least worse of two deaths.

Simply trying to keep the elderly engaged, suggesting that they attend yoga or computer classes, making resolutions to spend more time with our aged parents or grandparents, or providing them with better, cheaper nursing homes doesn’t cut it. These measures, although better than nothing, won’t give old people their health back and won’t, as a rule, spare them a forced choice between two evils. Forget about wishy-washy solutions; if we really care about our elderly, then it’s time to take life extension seriously.

Literature

[1] Alves, V. D. M., Maia, A. C. C., & Nardi, A. E. (2014). Suicide among elderly: a systematic review. MedicalExpress, 1(1), 9-13.

Reason – Fight Aging! blog and Repair Biotechnologies

Most people interested in rejuvenation and life extension are familiar with Fight Aging!, one of the very first rejuvenation advocacy blogs dating back all the way to the early 2000s; if you’re one of them, then you certainly are familiar with Reason, the man behind FA!.

Over the years, Reason has been a patient yet relentless advocate, acting not only as an information provider for the public but also helping out innumerable organizations and companies in the field of rejuvenation biotechnology in financial and other ways. Back in the day when SRF didn’t exist yet, Reason was a volunteer for Methuselah Foundation; eventually, he helped fund companies such as Oisìn Biotechnologies, CellAge, and LysoCLEAR; and, earlier this month, Reason and Bill Cherman co-founded Repair Biotechnologies, a company focused on gene therapy for rejuvenation, as announced on FA!.

Bill Cherman is an investor in the rejuvenation community who, just like Reason, has contributed to development of many ventures in the field. He is a holder of a gold medal in the Brazilian Mathematics Olympiad, a BA in economics, and a candidate in the Master of Biotechnology Enterprise and Entrepreneurship program at Johns Hopkins. He founded Front Seat Capital, a venture capital firm looking to invest in startups with the potential to change the world.

Repair Biotechnologies, which is presently looking for a Chief Science Officer, will kickstart its activities with a project on thymic regeneration in partnership with Ichor Therapeutics—the creators of LysoCLEAR, Antoxerene, and RecombiPure. The goal of the company, as you can imagine, is to shorten the journey of rejuvenation therapies from the lab to the clinic.

It is extremely heartening to see more and more rejuvenation-focused companies and organizations sprouting and building up to the turning point when this emerging field of science will cease being fringe and become a hot topic not only in the relatively small circle of biogerontology (where it has been one for a while now) but also in business and public discourse. We’re very grateful to Reason and Bill for taking us yet another step closer to the finish line and for answering our questions.

We’d like to ask some details of your story as a rejuvenation advocate. When and under what circumstances did it become clear to you that aging is a problem?

While it would be delightful to claim that I am a rational entity who came to that conclusion through utilitarian thought, in fact, it was more of a bolt from the blue. For no apparent reason, it suddenly came to me one evening that I didn’t want to die – and not in the academic way that most people hold that conviction but a deep, visceral, adrenaline-laden realization of the sort in which one accepts immediately that something important in life has been done and determined, a corner turned. Before that happened, I was no more than passingly interested in aging as a topic, but afterwards… well, I woke up. Of course, that was a long time ago now, far prior to my present understanding of what is plausible and possible, and realization on its own achieves nothing. It took years to learn enough to progress any sort of understanding as to how a non-life-scientist could make a difference.

We have noticed that there has been a sea change in both progress and enthusiasm from the academic community for rejuvenation biotechnology and targeting aging directly to prevent age-related diseases. Have you observed a similar rise in support, and what factors, if any, do you think are driving this?

I think that these things progress in cycles, based on the timescale of human collaboration. It takes a few years to go from desire to setting up an organization, a few years for the organization to get somewhere, and a few years for others to be inspired to their ventures by the organization. Bootstrapping only looks smooth in hindsight. We have been transitioning from one business cycle to another these past few years, which looks like a big leap in enthusiasm as it occurs, but the roots of this were set down five to ten years ago. I would say those roots included the final tipping point studies for senolytics, the spin-off of the SENS Research Foundation from the Methuselah Foundation, the injection of funding for SENS around then, and a number of other, related items.

It we look around today, a bigger community is planting a larger crop of seeds that will come to fruition in the mid-2020s, and today’s seeds include startup biotechnology companies in the SENS space, new advocacy initiatives like LEAF hitting their stride, and so forth.

Thanks to the efforts of many advocates, yours included, public perception of rejuvenation is also shifting. How close do you think we are to widespread acceptance?

I don’t think acceptance matters – that might be the wrong term to focus on here. Acceptance will occur when the therapies are in the clinic. People will use them, and everyone will conveniently forget all the objections voiced. The most important thing is not acceptance but rather material support for development of therapies. The help of only a tiny fraction of the population is needed to fund the necessary research to a point of self-sustained development, and that is the important thing. Create beneficial change, and people will accept it. Yet, you cannot just go and ask a few people. Persuading many people is necessary because that is the path to obtaining the material support of the necessary few: people do not donate their time and funds to unpopular or unknown causes; rather, they tend to follow their social groups.

Last year, you talked about the importance of sustained advocacy being as important as supporting the research itself. You wrote about a number of approaches to advocacy, including ours. Have you noticed an improvement in the quality of advocacy since then, and do you still maintain that professional advocacy is as important to the cause as research is?

Fishing for compliments? I’m very pleased with the progression of LEAF and with advocacy in general in our space. People have come and gone over the years, but this latest group of advocates appears to have set up shop for the long term. That is important and a welcome change. I can’t keep writing Fight Aging! forever, if only because hands and schedules eventually give way under the accumulated burdens of the years. There must be far more voices doing this same sort of work, all in their own varied ways. Diversity and redundancy are both important aspects of advocacy – many people arguing in their own ways for a given point of view are needed in order to persuade the world at large.

Presently, rejuvenation is a relatively unknown topic; people who say they’re against this technology probably don’t think it’s a concrete possibility anyway. However, as more important milestones will be reached—for example, robust mouse rejuvenation—this might change. Do you think that these milestones will result in opponents changing their attitudes or becoming more entrenched?

Opposition to human rejuvenation therapies is almost entirely irrational; either (a) it’s a dismissal of an unfamiliar topic based on the heuristic that 95% of unfamiliar topics turn out to be not worth the effort when investigated further, or (b) it’s a rejection of anything that might result in sizable change in personal opinion, life, and plans, such as the acceptance of aging and death that people have struggled to attain. This sort of opposition isn’t based on an engagement with facts, so I think a sizable proportion of these folk will keep on being irrational in the face of just any scientific advance or other new factual presentation short of their physicians prescribing rejuvenation therapies to treat one or more of their current symptoms of aging.

On the other hand, there will be steady progress in winning people over in the sense of supporting rejuvenation in the same sense as supporting cancer research: they know nothing much about the details, but they know that near everyone supports cancer research, and cancer is generally agreed to be a bad thing, so they go along. Achieving this change is a bootstrapping progress of persuading opinion makers and broadcasters, people who are nodes in the network of society. Here, milestones and facts are much more helpful.

After years of financially supporting other rejuvenation startups, you’re now launching your own company focused on gene therapies relevant to rejuvenation. What drove your decision to do this?

In the course of funding companies, one learns a great deal about the bounds of what might be achieved and the sort of work that is needed: it isn’t uncommon for investors to become entrepreneurs and vice versa. There are large overlaps in the mental toolkits required, and it is a logical evolution seen from either side. Moreover, in the course of investing in startups, one meets people in the community, such as my cofounder Bill, who intend to both fund and run companies, and it turns out that we work together quite well. As in all such things, it has a lot more to do with happenstance leading to the right arrangements of people and much less to do with the technical landscape at the time.

Your company’s first objective is thymic regeneration. Why do you think the thymus is the ideal initial target for your work?

It is a very straightforward goal, with a lot of supporting evidence from the past few decades of research. It think it is important to set forth at the outset with something simple, direct, and focused, insofar as any biotechnology project can be said to have those attributes. This is a part of the SENS rejuvenation research agenda in the sense of cell atrophy: the core problem is loss of active thymic tissue, which leads to loss of T cell production and, consequently, immunodeficiency. However, the immune system is so core to the health of the individual that any form of restoration can beneficially affect a great many other systems. The many facets of the immune system don’t just kill off invading pathogens; they are also responsible for destroying problem cells (cancers, senescent cells), and they participate in tissue maintenance and function in many ways.

You are using gene therapy; why have you chosen this delivery method specifically and not, for example, a small-molecule approach?

If your aim is to raise or lower expression of a specific protein, and you don’t already have a small molecule that does pretty much what you want it to do without horrible side-effects, then you can pay $1-2M for a shot at finding a starting point in the standard drug discovery databases. That frequently doesn’t work, the odds of success are essentially unknown for any specific case, and the starting point then needs to be refined at further cost and odds of failure. This is, for example, the major sticking point for anyone wanting to build a small-molecule glucosepane breaker – the price of even starting to roll the dice is high, much larger than the funding any usual startup crew can obtain.

On the other hand, assuming you are working with a cell population that can be transduced by a gene therapy to a large enough degree to produce material effects, then $1-2M will fairly reliably get you all the way from the stage of two people in a room with an idea to the stage of having animal data sufficient enough to start the FDA approval process.

You are working with SRF spin-off company Ichor Therapeutics; what was the reason for choosing to work with Kelsey and the Ichor team?

Because they are great. Kelsey has achieved considerable success, bootstrapping from nothing but a plan, and has an excellent team. Their philosophy of development dovetails well with ours, both in terms of short-term development of a biotech startup and in the longer term of how we’d like to see this industry develop over the next 10-15 years.

Will your company focus on lab work, or do you plan to run human trials once a sufficiently advanced stage has been reached?

We’re absolutely signed up for the end-to-end path of getting a therapy into the clinic. That is the whole point of the exercise – to bring therapies into general availability. Of course, there will be a great deal of lab work to accomplish between here and there.

The FDA doesn’t recognize aging as a disease, so it won’t approve drugs to target it directly. Is this a problem for your company’s activities?

Remember that when talking to the FDA, one usually starts with just a small patient group with a single age-related condition, a fraction of everyone that might eventually be helped. This is done to control costs and ensure the best possible chance of a successful approval by narrowing the focus to a very clear, simple experiment. After this, one expands to larger patient groups and more expensive trials. As it happens, the effects of immunosenescence on health are so widespread and similar from individual to individual that it wouldn’t be hard to pick a clearly defined condition and patient population that covers near everyone in late life. Unfortunately, one would have to have very deep pockets indeed to pick that as the first option for entering the approval process – one has to work up to it.

What are Repair Biotechnologies’ possible future targets after thymic regeneration?

We’re looking into a couple of interesting options, guided by the SENS philosophy of damage repair, but it is very challenging to say at this stage which of them will prove the most advantageous to attempt. Obviously, at this stage, the primary focus has to be on success in our first venture.

What do you think are currently the most promising research avenues within each rejuvenation therapy subfield?

We have a challenge today in that we have the DNA of a patient advocacy community trying to get work to proceed at all. So, for fifteen years, our measure of success was “are people paying more attention to this?” Now, we have to start thinking like a development community, in which success revolves around “does this implementation actually work in humans, and how well does it work, and how much does it cost?”

In all too many cases, we don’t yet know the answers to these questions: the data isn’t there yet for senolytics, for example. So, you can look at senolytic efforts and know who has the most funding and attention but have no idea which of the therapeutic approaches actually represent the most significant progress at the end of the day. For all we know, dasatinib might turn out to be the most cost-effective of all of the current small-molecule approaches, with everything everyone has done since then coming in a poor second-best, and we won’t find this out for years, as no one has any incentive to run the necessary large-scale trials on an existing drug.

Dr. de Grey is hopeful, but not certain, that immunotherapy might make OncoSENS unnecessary. What do you think?

I have long thought that canonical OncoSENS – whole-body interdiction of lengthening of telomeres – might be rendered unnecessary by sufficiently advanced incremental progress in other areas of cancer research. That said, it should be so cost-effective that it is hard to imagine “sufficiently advanced incremental progress” not incorporating interference with telomeres in some way. People other than SENS-funded groups are working on it, after all.

If you think about it, restoring the immune system to youthful capacity should also help to achieve this goal; there is evidence to suggest that age-related immune dysfunction drives age-related cancer risk and that this correlates well with thymic decline. The world will still need highly effective, low-side-effect cancer therapies even if everyone has the cancer risk profile of a young adult, of course, but far less frequently.

What do you realistically expect might happen, over the next 25 years, in terms of rejuvenation research results, funding, clinical applications, and availability?

Well, that’s an essay in and of itself. I think my views on the technology itself are fairly widely known: I’ve written a few short essays on likely ordering of development. The funding will  continue to grow year-over-year to the degree that any success is achieved in the clinic. However, everything takes a very long time in medicine due to the way in which regulation works, no matter how fast the technology is running in the labs, and the pace of technological progress in biotechnology is accelerating. At some point, the system exemplified by the FDA will break because cheap and effective therapies coming out of the labs will be so far ahead of what is available in the clinic that they will leak out into some other form of commercial development. Who knows what that will look like? Perhaps it will be a network of overseas non-profits that run their own, lighter and faster, validations of trials and presentations of human data gathered from participating clinics. I think that next-generation gene therapies, evolutions of CRISPR, will likely precipitate this sort of reordering of the landscape.

Do you expect that aging might relatively soon be officially considered a disease, or a co-morbid syndrome, by WHO and the FDA?

No. Regulation typically lags behind reality by many years. What will probably take place is some sort of battle of wills and lawyers over widespread off-label use for rejuvenation therapies, most likely senolytics, that have only been narrowly approved for specific age-related conditions. That will go on for a while and, ultimately, generate sufficient critical press attention to induce regulators to back off from trying to suppress that off-label use and, instead, accept aging as an approved indication. This hypothetical scenario could run a decade or more from beginning to end.

The availability of rejuvenation therapies doesn’t depend only on their cost; it also depends on how they’re regulated in each specific country. Do you imagine “rejuvenation tourism” will exist for long, or at all, before these treatments are part of the standard medical toolkit everywhere?

The development of stem cell therapies is the example to look at here. These therapies were available via medical tourism for a decade prior to the first approved treatments in the US, and this continues to be the case even afterwards, as only a narrow slice of therapies have been approved. Medical regulation is slow-moving, and so medical tourism will be long-lasting. I think this will work exactly the same way for other broad classes of therapy, such as gene therapies.

What is, in your view, the biggest bottleneck to progress in aging research?

Either (a) the lack of funding for research and early-stage startup development or (b) the low number of entrepreneurs, one of the two. Probably funding, as money can be used to craft an 80/20 solution to the shortage of entrepreneurs, but entrepreneurs can only reliably solve the lack of funding problem if there are a lot of them. Almost every specific instance of things not moving forward that I’ve seen could be addressed by a well-thought-out application of funds to the situation.

The chasm between academic research and early-stage commercial development is also a sizable issue. The academic side does a terrible job of reaching out to find entrepreneurs and companies that can carry forward their research to benefit patients. The entire biotechnology industry (entrepreneurs, investors, bigger companies and funding entities) collectively does a terrible job of reaching back into the academic community to fund, encourage, and adopt the most promising research. So, projects that should move instead languish for years because no one is taking the obvious steps to improve on the situation.

Right now, there don’t seem to be any unexpected problems with the science that might jeopardize the development of rejuvenation. Do you think that any particular areas of research might run into difficulties down the road?

No. I think all the unexpected problems will be implementation details. It is perfectly possible to have the correct strategy and the wrong tactics, and this happens all the time in complex fields such as biotechnology – it doesn’t take much of an error in interpreting research results to derail the original plan and require a new direction. Most such challenges are short-term and can be worked around with some loss of time and money, but there are certainly past instances in which the company is lost because there is no viable way to salvage a better path.

This is what happened to one of the early AGE-breaker efforts, the development of ALT-711: removing AGEs still seems very much a correct approach to the age-related stiffening of tissues, but a drug that works in rodents will do nothing in people because the physiologically relevant AGEs are completely different. At that time, the researchers didn’t have that critical piece of information. We will no doubt see similar stories occur again in the future.

Caloric restriction and exercise may also potentially convey some small increase in life expectancy. Given that the goal is to reach longevity escape velocity, do you practice a particular diet or exercise program, and would you encourage people to consider such approaches?

I have always suggested that people look into the simple, reliable things they can do for better health. The way to look at this is through the lens of cost-effectiveness. Calorie restriction and exercise are cheap, easy, and highly reliable. They don’t adjust your life expectancy by decades, but since they are cheap, easy, and reliable, you should still look into it. There are many different ways to approach both, so just because an attempt fails or isn’t palatable, that’s no excuse to give up on the whole endeavor. At the end of the day, it is a personal choice, of course. We can always choose to be less healthy; that’s easy to do in the present environment.

You’ve written many articles on the topic of self-experimentation on FA. Can you summarize your views?

The current self-experimentation community – and here I include many disparate groups, only tenuously linked, with interests in nootropics, anti-aging, muscle building, and so forth – is woefully disorganized and ill-educated when it comes to the risks and scientific knowledge of the compounds they try. If one in twenty of the people who have tried dasatinib as a senolytic have (a) read the papers on pharmacokinetics in human volunteers, (b) recalculated likely human doses from the senolytic animal studies and compared them with human chemotherapy studies, or (c) actually tested the compound delivered by a supplier to ensure purity, I would be astoundingly surprised.

The bar for quality and safety in this community needs to be raised, and that is the primary purpose behind my writing articles on self-experimentation. Whatever I say, people are going to be out there trying senolytics – many of these compounds are cheap, easily available, and hyped. What they should be doing instead of rushing in is thinking for themselves and reading widely. If I can do a little to help make that happen, then all to the good.

What is your take-home message for our readers?

There is always a way to help accelerate the development of rejuvenation therapies – there is always something that one can do and feel good enough about doing to do it well. Don’t know what that something might be? Then talk with people in the community. Reach out, go to meetings, post online. Don’t force it. It will come to you in time.

Best of luck to Reason and Bill in their new endeavor. If they can bring thymic rejuvenation to the clinic, they might possibly save millions of people from several devastating, age-related diseases.

New Harvard Startup Wants to Reverse Aging in Dogs

Professor George Church of Harvard Medical School has co-founded a new startup company, Rejuvenate Bio, which has plans to reverse aging in dogs as a way to market anti-aging therapies for our furry friends before bringing them to us.

Dogs first, humans next

The company has already carried some initial tests on beagles and plans to reverse aging by using gene therapy to add new instructions to their DNA. If it works, the goal is ultimately to try the same approach in people, and George Church may be one the first human volunteers.

“Dogs are a market in and of themselves,” Church stated during the recent Radical Wellness event in Boston. “It’s not just a big organism close to humans. It’s something that people will pay for, and the FDA process is much faster. We’ll do dog trials, and that’ll be a product, and that’ll pay for scaling up in human trials.”

Church and the team also understand that developing therapies that address aging in humans and getting them approved would not be so easy. It would take too long to prove something worked. “You don’t want to go to the FDA and say we extend life by 20 years. They’d say, ‘Great, come back in 20 years with the data,'” Church said during the same event.

So, the team has taken a different tack; rather than aiming to increase human lifespan as its main focus, it is instead focusing on the typical age-related diseases common to dogs. The hope is that by targeting the aging processes directly, these diseases could be entirely prevented from developing. If successful, this would lend additional supporting evidence that directly treating aging to prevent age-related diseases could also work in humans.

Working on rejuvenation

In 2015, the Church lab at Harvard began testing rejuvenation therapies on mice by using gene therapy to deliver CRISPR, a new and superior gene editing system. Since then, the lab has been engaged in dozens of studies on aging with a view towards the rejuvenation of aged animals.

The team has used gene therapies to add additional instructions into the DNA of aged mice, using these instructions to regulate and modify the activity of various genes. The researchers increase or inhibit target gene expression, depending on its contribution to aging, in a bid to restore gene expression and the production of secreted molecules to youthful levels.

The lab has been working on a collection of over 60 different gene therapies and has been testing their effects both individually and in combinations. The team intends to publish a report on an approach that extends mouse lifespan by modifying two genes that protect against heart and kidney failure, obesity, and diabetes. Professor Church has commented that the results of this study are “pretty eye-popping”.

The new startup has been contacting dog breeders, veterinarians, and ethicists to discuss its plans for restoring youth and increasing the lifespan of dogs. Its plan is to gain a foothold in the pet market and then use that as the basis for moving therapies to people.

Starting a dog trial

Last year, the lab contacted the owners of Cavalier King Charles Spaniels, a breed of toy dogs, with plans for a trial of gene therapy to combat mitral valve disease, a common heart condition that kills almost 50% of this breed at age ten. The nature of the gene therapy is not yet public knowledge, though it may involve inhibiting the cytokine TGF-beta, which facilitates the scarring and thickening of heart valves, and the team has tested a similar therapy on mice to stop heart damage.

Some readers may recall that inhibiting TGF-beta also produced tissue regeneration in brain and muscle when tested by the Conboy lab in 2015 [1]. The Conboys famously demonstrated this and the fact that balancing blood factors to youthful levels encourages tissue rejuvenation. It may be that by reducing levels of this cytokine to those seen in youth, this heart condition may be prevented in these spaniels.

Earlier this year, company owners Davidsohn and Oliver traveled to Chicago to the breed’s national show, where several thousand dollars were raised to begin the trial. The president of the American Cavalier King Charles Spaniel Club, Patty Kanan, said that this research is “seriously meaningful to the American Cavalier King Charles Spaniel Club.”

Conclusion

There have, of course, been the usual detractors of this initiative, such as skeptics and fortune tellers predicting that it will not work before it is even tested. Thankfully, the people behind this new company, which is backed by the expertise and vision of George Church, plan to proceed and see if the data can prove the detractors wrong.

Fortune favors the bold, and these are the people in the field getting the important therapies tested. If this pays off, the potential is huge, because if a therapy works in dogs, you can be sure that the pressure to move it to humans will soon pile up.

Literature

[1] Yousef, H., Conboy, M. J., Morgenthaler, A., Schlesinger, C., Bugaj, L., Paliwal, P., … & Schaffer, D. (2015). Systemic attenuation of the TGF-β pathway by a single drug simultaneously rejuvenates hippocampal neurogenesis and myogenesis in the same old mammal. Oncotarget, 6(14), 11959.

Sarah Constantin – The Longevity Research Institute

Today, we have an interview with the Longevity Research Institute, a new group set to launch in April 2018 of this year. The goal of the Institute is to identify therapies that can demonstrably extend healthy human lifespan by 2030 at the latest.

Searching for longevity

There are dozens of compounds and therapies that have been demonstrated to increase the lifespan of mammals. Recently, there have been some impressive examples of rejuvenation in animals using a variety of approaches, including partial cellular reprogramming, stem cell therapy, and senescent cell removal. More importantly, in many of these studies, age-related diseases have been delayed or even reversed.

Unfortunately, very few of these studies have had independent follow-ups or replication, and that is slowing down progress. The Longevity Research Institute is aiming to bridge the gap between basic science and commercial drug development.

It has chosen the field of aging research as its area of focus for one simple reason: age-related diseases are the leading cause of death globally. Heart disease, stroke, cancer, diabetes, Parkinson’s, Alzheimer’s and many more diseases are all caused by the various processes of aging.

The data from hundreds of animal studies tell us that aging is not a one-way process and that the rate of aging is something we can slow down or even reverse. Experimental results show that we can increase the healthy lifespan of animals significantly and delay the onset of age-related diseases in doing so. If we could translate those findings to humans, we could potentially increase the healthy period of life, known as healthspan, or even increase our lifespan beyond current norms while remaining healthy.

The majority of aging research consists of basic science that focuses on the mechanisms of aging, studies involving invertebrates like worms or fruit flies, and experiments that examine the effect of therapies on biomarkers of aging. However, the Longevity Research Institute believes that the way to find effective treatments that could translate to humans is by testing interventions on mammals to see if they increase lifespan or if they delay or reverse symptoms of aging, such as frailty, cognitive decline, and the prevalence of age-related diseases. Robust mammalian lifespan studies are quite rare in aging research due to their long duration and thus cost; the Institute believes they are worth doing despite this challenge.

Its philosophy is to be skeptical of results that depend on too many uncertain assumptions, such as particular mechanisms of aging or analogies between invertebrate and human biology. It believes that the closest way to measure the health and lifespan of a human is to measure the same things in mammals.

Replicating and Extending Lifespan Results

The majority of studies that have been shown to increase lifespan are rarely independently replicated to confirm the findings. There are therapies that, decades later, still have had no follow-up, and the Longevity Research Institute would like to change this situation.

To that end, it will be engaged in grant writing to obtain funds so that researchers studying aging will be able to conduct lifespan studies in mice and rats. The Longevity Research Institute also plans to commission its own studies and contract research organizations to carry them out.

It has a long list of promising interventions and is considering becoming involved with carboxyfullerenes, epithalamin, and stem cell transplants, for example. It is also interested in testing combinations of therapies to see if they have synergistic effects.

As translational research on aging is really a new, uncharted territory, the Institute is working with the Interventions Testing Program and METRICS to design reproducible animal studies. As part of that process, it will be testing genetically heterogeneous animals and using blind, randomized studies to reduce bias. A blind experiment is an experiment in which information about the test is hidden from participants, to reduce or eliminate bias, until after a trial outcome is known.

Best practices and transparency

Establishing best practices and protocol for translational aging research is a top priority here, and its work could help set the stage for future translational efforts. If superbly designed research protocols can be designed and made accessible to everyone, then they could be a real help in standardizing aging research and ensuring that the quality of results is the best it can be.

As part of its commitment to transparency and knowledge sharing, a condition of funding projects is that all experimental data will be made freely available to the public, as will pre-registration of experimental designs. The Institute will further protect this open science initiative by using blockchain technology to make immutable, publicly accessible records of everything it does.

We had the opportunity to talk with Sarah Constantin, Ph.D. and one of the key figures at the Longevity Research Institute, about their work. Sarah is a data scientist specializing in machine learning.

Your group believes that we need to conduct lifespan studies in mice in order to confirm that something might translate. However, some researchers believe that using multiple biomarkers of aging allows them to project, within a reasonable margin of error, changes to potential lifespan. This is becoming more relevant as the accuracy of biomarkers, and the use of comprehensive biomarker panels, becomes more commonplace. How do you respond to this?

There’s some very interesting stuff going on with biomarkers of aging.  We’re able to predict mortality with AUCs of 0.8-0.9, which is quite good, with aging biomarkers, including things like blood panels of inflammatory and metabolic markers, DNA methylation, and phenotypic markers such as BMI and frailty. Some of these biomarkers are things we’re planning to measure in our animal studies, and they should give us interim results on whether the interventions we’re testing affect the predictors of aging. I still believe that we can be most confident in whether a treatment promotes longevity when we’ve tracked its effects throughout an organism’s lifespan. We do know of examples (such as calorie restriction in primates) in which it’s equivocal whether the treatment extends lifespan but it clearly improves age-related biomarkers, and you have to do a lifespan study to distinguish those cases.

Advances in deep learning and systems pharmacology are allowing us to project interactions and potential therapies far more efficiently than ever before. What are your thoughts on these approaches, and will you be looking to use them in your work?

The deep learning and systems pharmacology approaches are actually where I started in biotech; I did machine learning at Recursion Pharmaceuticals, which is taking those approaches for doing phenotypic screens for genetic disease treatments. I think they’re really useful for drug discovery, at the beginning of the pipeline, where they can enable you to search a wider space of drug candidates. At LRI, we’re starting all the way at the other end of the pipeline, with drugs that have already been tested and shown promise in vivo. However, once we make some progress on those, then yes, it could make sense to start doing some of these machine learning-enabled approaches.

What is the ideal mouse strain for aging research, particularly lifespan studies, in your view?

Well, the Interventions Testing Program at the National Institute of Aging is using three-way heterozygous mouse crosses, which I think is the ideal. A single inbred strain of mouse doesn’t have much genetic diversity, so often what you’re testing is the effect of a treatment on that particular strain of mouse, and the results won’t transfer to another strain.

The use of progeria mice is common in aging research due to the shorter study time, but these models are often criticized as not being representative of true aging; what are your thoughts on the prevalence of progeria mice in aging research, and are they a relevant model for what we are trying to achieve?

I think progeria mice are an imperfect proxy. There are a lot of different kinds of progeria, and they exhibit some but not all of the typical symptoms of natural aging.  I’d have more confidence in studies done on aged mice than progeric mice.

We see that you have a strong commitment to ensuring public access to scientific knowledge. What inspired you to make such a wonderful and strong commitment to open science?

Well, coming from a data science background, I’m hyper-aware of how easy it is to fool yourself with data.  You can massage anything into a spurious result if you test enough hypotheses and pick your subgroups artfully. Really, the best way to guard against that is to share the raw data so that people can run their own analyses. Making science more open is how you make it more trustworthy.

Is there a publically viewable list of the targets that you are interested in testing?

The list is still evolving, but some of the first things we’re looking into testing are carboxyfullerenes, which seem to have neuroprotective and life-extending effects, and epithalamin, which is a pineal gland-derived peptide that’s been reported to extend lifespan and even reduce human mortality. Both of these are sort of in the sweet spot of not being the subject of that much research to date, but what there is is very promising, so the value of information is high.

What is likely to be your first target for studies, and what is the rationale behind your choice?

I think people should know that there’s a lot of low-hanging fruit in aging research — treatments that we have reason to believe might work but that we’d still have to test. The misperceptions are either that life extension is so speculative that we’ll never get there or that we already know how to do it and you just have to take the right supplements to live forever. I think the reality is that we’ll have to do a lot of experimental work, but it’s highly possible that, in time, we might find something that extends healthy lifespan in humans.

We would like to thank Sarah for taking the time to do this interview with us, and we look forward to seeing her team’s progress in the near future.

Life Extension Technology in Science Fiction

Today, we take an amusing look at how science fiction is often portrayed in a jarring way especially when dealing with the topic of life extension.

Those of us who fancy science fiction stories are used to all sorts of technological miracles taking place in them; some are plausible and might become reality at some point in the future, while others are mere fantasies, artistic liberties that are taken to tell a better story and will likely never translate into real-life technologies—or, if they will, they will do so at the cost of rethinking fundamental principles that we’ve thus far considered to be fully established.

In science fiction, we’ve seen faster-than-light travel, teleportation, portals, energy weapons, strong AI, telepathic powers, and radiation-induced superpowers of all kinds; unfortunately, the only “superpower” known to be actually induced by radiation thus far is cancer. Entire imaginary worlds have revolved around the existence of one or more of these marvels, and series and shows have assumed that they’re possible and imagined what our society would be like with them, but one particular possibility has been neglected or relegated to one or two episodes and then forgotten, as if it was of no importance whatsoever: the defeat of aging.

There is no elephant; but if there is, it’s okay

Star Trek, a popular space opera I have loved since my early teens, is famous for its optimistic depiction of a future where space travel is as common as trains are today. Space travel is not, per se, the topic of Star Trek; episodes don’t generally discuss whether space travel is feasible or desirable, nor do they praise or demonize it; quite simply, space travel is taken for granted as an everyday reality that functions as a plot device. Star Trek episodes answer the very general question, “What might life be like if we were a space-faring species?“

Now, just how many series can you recall, from the top of your head, doing the same thing with the defeat of aging in place of space travel? How many shows are there where people don’t die of aging anymore and this is not the topic being discussed but is rather just a background element? In other words, how many shows are there where aging is no longer a thing and the show doesn’t revolve around all the alleged problems and drawbacks of it? How many shows assume that aging has been medically conquered and try to answer the question, “What might life be like in a society where age-related diseases no longer exist?” Not many, I’m willing to bet.

However, many science fiction shows have an episode or two where the possibility to reverse aging is hinted at, sometimes very clearly, but none of the characters seem to realize the bleedingly obvious potential for an unprecedented medical revolution staring them right in the face. If anyone bothers reacting in any way at all, it’s normally just to dish out stale clichés and moral lessons about how the finitude of life is a blessing or how tampering with nature is unnatural and hence bad (which is a rather bizarre thing to say for someone standing on a spaceship traveling faster than the speed of light).

This is not so strange when you think about it. This is nothing but the umpteenth manifestation of the pro-aging trance, which blinds us to the tragedy of aging and forces us to do all manner of embarrassing mental gymnastics to pretend it’s some sort of blessing in disguise. Our acceptance of aging is so strong and pervasive that it’s slipped unnoticed into every layer of our society and culture, especially the arts.

Sometimes, this acceptance doesn’t necessarily lead us to praise aging rather than rage against it; some of us would gladly do away with it if they only knew that it might be possible, but they’ve grown so accustomed to its alleged inevitability that the possibility doesn’t even cross their minds. Some of the clearest examples of the different manifestations of the pro-aging trance in science fiction come mainly, but not exclusively, from Star Trek.

To not-so-boldly go

Dr. Beverly Crusher on Captain Picard’s Enterprise and the holographic doctor on Captain Janeway’s Voyager have performed nearly all the medical wonders you can imagine. They’ve triumphed over all sort of diseases, brought in people who were transmuted into aliens or assimilated by the Borg into cybernetic organisms and turned them back into humans, the list goes on. Yet, 300 years into the future, no one has yet figured out what to do about aging. Humans keep aging more or less at the same rate, or perhaps a bit slower than that, since most people seem to live to around 140 years old, although they still grow sick and decrepit.

However, every now and again, some characters in Star Trek do happen to stumble upon a cure for aging or a phenomenon that might hold the potential to become one. You’d be amazed to see how many different ways there are to undo aging in the 24th century and how, apparently, no one could care less about them.

In The Next Generation’s episode “Too short a season”, a very elderly and disabled Starfleet Admiral manages to get his hands on an alien concoction that is said to cause rejuvenation (even in humans, apparently, despite its alien origin). In an attempt to speed up the rejuvenation process in time to face an old enemy, the Admiral takes twice the recommended dose, obtaining the desired effect at the cost of his own life: the extremely accelerated process causes too much stress on his body, which gives in when he has been rejuvenated to about the age of 20. Not only did none of the characters seem even slightly interested in the potential of this miraculous cure—which, with some fine-tuning, could end the diseases of aging for good—the authors also managed to shoehorn in yet another lesson about the perils of tampering with nature.

In another, more fanciful, TNG episode, the Enterprise’s counselor Deanna Troi falls prey to a sly, manipulative negotiator who, through a strange ritual, manages to turn her into a receptacle of all his negative emotions. The burden quickly becomes too heavy and Deanna, just like other women that the negotiator exploited before her, experiences accelerated aging that almost kills her. Thankfully, the crafty Dr. Crusher manages to break the bond between the attacker and his victim, instantly rejuvenating Deanna and killing the negotiator, who turned into an old man in the blink of an eye. Perhaps the technique used by Dr. Crusher here could have been of some medical interest to treat the diseases of aging, but the idea didn’t seem to cross her mind.

Of particular interest in this same episode is a bit of small talk casually thrown around by Commander Riker. When the negotiator first boards the Enterprise, he is accompanied by an elderly, demented woman whom he passed for his mother. (She’s actually a chronologically young woman used as his current receptacle.) The “mother” eventually dies and the negotiator replaces her with Deanna, but before this happens, Deanna talks with Riker about how the sight of the elderly, mentally ill woman upset her. As they talk about it, the Commander nonchalantly comments that maybe, one day, we will all be like that elderly woman—as if the prospect of losing our health and wits is no cause for concern whatsoever.

In Star Trek IX, “Insurrection”, the crew of the Enterprise finds out that the particular location of a planet within a nebula exposes it to a type of radiation which has rejuvenative and regenerative effects, which are experienced by the crew themselves. Busy as they are foiling the villain’s plan to deport the local population of the planet, none of our heroes think that this beneficial radiation should be studied for its therapeutic potential. (Unsurprisingly, only the villain is interested in the properties of this radiation, and, naturally, he can’t think of a better way to reap its benefits than wreaking havoc on the planet. Once again, the moral of the story seems to be that only the bad guys would ever want to extend their lives, and they always do so at others’ expense. Sigh.)

In a Star Trek Voyager episode, the stuffy and stern Lieutenant Tuvok is stranded on an unknown planet in the company of a handful of local, alien kids who are terrified that a terrible monster will come and kill them if they get too close to a cave where their own people left them to die. Initially bewildered that a species might purposely try to kill its own young—an idea which he, as any good Vulcan would do, deems “illogical”—Tuvok eventually discovers that the children are actually elderly, and their species’ growth process works the other way around, almost a la Benjamin Button; they’re born as elderly grown-ups, and they rejuvenate and de-develop from adults into children as they approach the end of their lifespan; the monster is just a story.

Forget that this makes no biological or evolutionary sense whatsoever; forget the unique opportunity to study a spontaneous process of multicellular rejuvenation and its potential applications; forget that this would imply death in this species occurs only for the sake of ending each individual’s life, with no apparent cause. The really grave offense here is Tuvok’s parting words to the last of the terrified children after the rest have disappeared (read: died): there is nothing to fear at the “natural” end of her life cycle (why not? And why, then, is there anything to fear at the “unnatural” end of life?), which he can’t and doesn’t want to interfere with. Apparently, in this particular instance, the otherwise inflexibly logical Tuvok was glad to throw logic out the airlock. Though to be fair, under the circumstances, Tuvok couldn’t really do much else, and what is most despicable aren’t his actions as much as the message the authors tried to convey.

There are more examples of how Star Trek glosses over the problem of aging and how some characters have an ambivalent attitude towards death, but I’d rather stop here. Don’t get me wrong; I still love Star Trek, but in some respects, it didn’t go nearly as boldly as it could have.

Dr. Hypocrite

Dr. Who’s episodes are often just as scientifically cringeworthy as they are brilliantly hilarious—so much so that one is willing to turn a blind eye to the numerous examples of downright nonsense. However, I couldn’t bring myself to do so in the case of the episode “The Lazarus Experiment.”

An ingenious yet obviously unscrupulous elderly scientist invents a machine to rejuvenate people. In a public demonstration of his invention, the man steps into the fancy-looking piece of machinery, and after some light show, he leaves the contraption some fifty years younger. The Doctor and his trusty companion are nearby and visibly worried about what he’s just witnessed, he confronts the scientist about it. The Doctor knows how the machine works and tries to persuade the rejuvenated scientist that each man has his own time and that “It’s not the time [that you live] that matters, it’s the person [you are].” Forgetting clichés and non sequiturs, it is important to keep in mind that the Doctor belongs to a species whose members, despite their entirely human appearance, are able to regenerate whenever they’re close to death; at this point, the Doctor had regenerated ten times already (and his incarnations aren’t always very faithful to the original), and ironically, he was about to do it again in the very next episode, when his life was at risk. Each man might have his own time, but the Doctor has been postponing his meeting with the Reaper for a good nine hundred years (and counting).

To complete this rather hypocritical collection of clichés, the rejuvenated scientist eventually transforms into a monster—yet another “lesson” that trying to extend your life is a bad idea.

The example of “Passengers”

The movie “Passengers” is set on a fully automated starship in which people who want to start over on a new world are placed in stasis capsules and sent off to a faraway planet in a journey that will take 120 years. The unlucky main character’s capsule malfunctions and he wakes up, all alone, 90 years in advance. There’s no reason for me to spoil the plot further. There’s nothing in it that I criticize per se, and the whole story hinges on the fact that he, in his 30s or so, will be long gone before the ship may reach its destination; without aging, this plot wouldn’t make any sense. Yet, it is another example of a story where, apparently, in the future, we’ve made no progress against aging. I couldn’t help thinking about it the whole time; in the future portrayed by this movie, we managed to build a huge, fusion-powered starship that is full of stasis capsules that bring metabolism to a complete halt (for the very purpose of preventing death by aging during the trip) and then restart it, but we’re still unable to cure aging.

Conclusion

Superluminal starships, teleporters, and holodecks, despite the fact that they don’t exist yet and we’re not even sure that they can, are something we’re perfectly used to and can easily imagine, because many people have dared to describe what life with them might be like; however, very few, if any, have dared to envision and show a world without aging, even though the science that might take us there, early-stage as it may be, is already here. Furthermore, rather than showing curiosity towards this brave new world that might await us, many people show disinterest or even contempt.

Of course, stories in which humanity manages to overcome aging or death do exist, but generally, it’s a power that only a few have and is generally depicted as a burden to carry, even though there is no evidence that this would be the case.

It’s a glaring demonstration of foxes disdaining grapes, of an extremely long-term Stockholm syndrome, and of the pervasiveness of the pro-aging trance. It’s high time that we woke up from it.

Recoding Human Cells to Make them Virus Proof

On May 1, around 200 scientists from the Genome Project-write (GP-write) met in Boston and announced the first target of their project: the creation of cells that cannot be infected by viruses.

What is the Genome Project-write?

GP-write includes sub-projects like the Human Genome Project-write (HGP-write), which was formally announced on June 2, 2016, and is an extension of the Genome Projects, which were launched in 1984. These projects were created to develop ways to read DNA in microbes, plants and multiple animal species, including humans.

GP-write was initially met by some level of panic and alarm, as some of the media misrepresented the project and people’s imaginations ran amok. Suggestions of “secret meetings” and stories of scientists creating designer babies and superhumans were doing the rounds, regardless of the actual scientific reality of the project. However, stories of mutants, superhumans, and manbearpigs are greatly exaggerated.

The newly created GP-write project will be managed by the Center of Excellence for Engineering Biology, a new nonprofit organization. With a firm commitment to cost reduction, safety, and ethical conduct, there could be considerable scientific advances as a result of this project.

The Genome Project-write (GP-write) is an open, international research project led by a multidisciplinary group of scientific leaders who will oversee a reduction in the costs of engineering and testing large genomes in cell lines more than 1,000-fold within ten years.

GP-write will include whole genome engineering of human cell lines and other organisms of agricultural and public health significance. Thus, the Human Genome Project-write (HGP-write) will be a critical core activity within GP-write focused on synthesizing human genomes in whole or in part. It will also be explicitly limited to work in cells, and organoids derived from them only. Because of the special challenges surrounding human genomes, this activity will include an expanded examination of the ethical, legal and social implications of the project.

Engineering virus-resistant cells

Jef Boeke, one of the four leaders of GP-write, said, “There is very strong reason to believe that we can produce cells that would be completely resistant to all known viruses.” He continued further: “It should also be possible to engineer other traits, including resistance to prions and cancer.”

The project takes the original Human Genome Project and builds upon it. The GP-write project could see rapid progress, given the sheer number of collaborating labs comparing data, verifying the results, and conducting safety analysis. This could be a game changer for stem cell therapies, as these robust, virus-proof cells could be used to treat a variety of diseases.

The team plans to engineer resistance to viruses using a technique known as recoding; this depends on the fact that much of the genetic code is redundant. Each three-letter sequence of DNA letters is known as a codon, and these codes are specific to the production of a particular amino acid. The chart below shows the codons for each amino acid.

Amino AcidDNA codons
IsoleucineATT, ATC, ATA
LeucineCTT, CTC, CTA, CTG, TTA, TTG
ValineGTT, GTC, GTA, GTG
PhenylalanineTTT, TTC
MethionineATG
CysteineTGT, TGC
AlanineGCT, GCC, GCA, GCG
GlycineGGT, GGC, GGA, GGG
ProlineCCT, CCC, CCA, CCG
ThreonineACT, ACC, ACA, ACG
SerineTCT, TCC, TCA, TCG, AGT, AGC
TyrosineTAT, TAC
TryptophanTGG
GlutamineCAA, CAG
AsparagineAAT, AAC
HistidineCAT, CAC
Glutamic acidGAA, GAG
Aspartic acidGAT, GAC
LysineAAA, AAG
ArginineCGT, CGC, CGA, CGG, AGA, AGG
Stop codonsTAA, TAG, TGA

Hundreds or even thousands of amino acids are linked together to create proteins; these signal our cells to perform certain functions, and they include insulin, collagen, myosin, and many thousands of others.

Despite this deeper complexity, there are just 20 amino acids and a “stop” signal, and there are 64 combinations of the four DNA nucleotides. However, life can continue with only one codon for each amino acid. For example, during recoding, you might choose the GTT in, say, valine, and whenever the three redundant ones (GTC, GTA, and GTG) appear in the genome, they are replaced by GTT. This essentially means that every time one codon is replaced by another, you are removing one codon from the genetic codebook and reducing the complexity.

But why would we bother doing this?

Here is where the viral resistance comes in. Because the genes of a virus contain these redundant codons, the virus can enter the cell and take control of the cell’s genetic machinery. In the case of a recoded cell, the virus would not be able to take over the control system and use it to create more viruses; the cell would lack the ability to create the viral proteins it needs to proliferate, and so the cell would be immune to viral infection.

George Church, one of the GP-write project leaders, has already tested the approach in E.coli. His team replaced all 321 instances of one redundant codon in its genome, which rendered the bacteria resistant to the bacteriophage T7 virus. Church took this a step further in 2016 and replaced seven redundant codons in around half of the same bacteria’s genome [1]. This required the replacement of 62,214 redundant codons with synonymous alternatives across all protein-coding genes.

The project aims to make virus-proof human cells within 10 years, but to do that, the number of codon replacements would be around 400,000 in order to affect around 20,000 protein-coding genes. This is a monumental task, and it is why the GP-write leaders suggest that writing genomes from scratch, not editing them with CRISPR, is a better approach.

Jef Boeke said, “We have nothing against CRISPR. We love it and use it all the time. But we’re talking about changes that are just massive. It’s like if you’re editing a short story: if you’re changing so much of it, you might as well just rewrite the whole damn thing.”

In order to build a genome from scratch, it would mean synthesizing DNA one nucleotide at a time and joining thousands of these together. While it is possible to do this with today’s technology, it is a laborious and slow process. To give you an idea, it costs around a dollar to synthesize 10 DNA letters, meaning that it would cost $300 million to do the human genome.

The GP-write project team aims to drive down synthesis costs by a factor of 1,000, thus making the idea a much more practical proposition. Church has already indicated that he wants to synthesize all human genes through joining 200-letter-long strands, which we can do now with currently expensive technology. Those familiar with Church will almost certainly know that he has been instrumental in driving down the costs of gene editing in the past, and the members of this project are confident that they can do so again here.

Conclusion

There is huge potential here for not only reducing costs and contamination but also for creating in vivo therapies that potentially introduce replacement cells that are impervious to viruses, such as HIV, resistant to other diseases, such as cancer, and even resistant to damage from aging and radiation. A look at the goals of the project published last year makes it quite clear that the ultimate goal is to develop this for in vivo therapies, such as stem cell transplants.

There is an unmet need for an “Ultrasafe human cell line” designed to serve as a platform for many biomedical applications, from production of biologics, to modeling cell and tissue behaviors, to actual ex vivo and ultimately in vivo therapeutic applications.

As we improve our ability to edit and create genomes from scratch, a world of options opens up for combating diseases, and we look forward to seeing more progress from the GP-write team.

Literature

[1] Ostrov, N., Landon, M., Guell, M., Kuznetsov, G., Teramoto, J., Cervantes, N., … & Shrock, E. (2016). Design, synthesis, and testing toward a 57-codon genome. Science, 353(6301), 819-822.