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

Photo
* * * * - 6 votes

Alternative methods to extend telomeres

telomeres nad nampt ampk resveratrol allicin methylene blue nmn sirtuins statin

  • Please log in to reply
989 replies to this topic

#781 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 01 August 2022 - 01:44 PM

The clock was created by researchers. It doesn't exist in the cell. And stem cells have very little methylation.

 

I was looking into this, and whilst stem cells may have little CpG methylation, methylation of histones - atleast in ESCs - is of paramount importance. 

 

Here is an extract from an imfomative paper:

 

Methionine metabolism is critical for the maintenance of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) pluripotency. However, little is known about the regulation of the methionine cycle to sustain ESC pluripotency. Here, we show that adenosylhomocysteinase (AHCY), an important enzyme in the

methionine cycle, is critical for the maintenance and differentiation of mouse embryonic stem cells (mESCs). We show that mESCs exhibit high levels of methionine metabolism, whereas decreasing methionine metabolism via depletion of AHCY promotes mESCs to differentiate into the three germ layers. AHCY is posttranslationally modified with an O-linked β-N-acetylglucosamine sugar (O-GlcNAcylation), which is rapidly removed upon differentiation. O-GlcNAcylation of threonine 136 on AHCY increases its activity and is important for the maintenance of trimethylation of histone H3 lysine 4 (H3K4me3) to sustain mESC pluripotency. Blocking glycosylation of AHCY decreases the ratio of S-adenosylmethionine versus S-adenosylhomocysteine (SAM/SAH), reduces the level of H3K4me3, and poises mESC for differentiation. In addition, blocking glycosylation of AHCY reduces somatic cell reprogramming. Thus, our findings reveal a critical role of AHCY and a mechanistic understanding of O-glycosylation in regulating ESC pluripotency

and differentiation. source: https://www.pnas.org.../DCSupplemental.

 

 

In short the paper shows that methylation of H3K4 is vital to keep ESCs pluripotent and prevent differentiation. If the SAM/SAH ratios gets too low, differentiation occurs. Interestingly they didn't focus on SAMe supplementation, but in the clearance of SAH by an enzyme called AHCY - glycosylation by NAG made it go faster, so helped stem cells stay pluripotent.

 

Anyway, it shows why methionine is so important to stem cells. I wonder why methionine is associated with faster aging, if it maintains stem cells?



#782 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 02 August 2022 - 01:22 PM

Hyperfunctional Telomeres Part II:

How do you lengthen the telomeres of the small number of important stem cells, those cells responsible for the health of the whole organ in which they reside, in a way which will later restore lost youth to the body?

 

...Experimental protocol to follow.

 

 

 

Telomerase Activation Experiment Report I

 

After my previous series of posts including that quoted above as well as here regarding synergistic activation and assembly of telomerase, I began a new protocol designed to exploit this understanding mid-April 2022.

 

The basic weekly protocol is as follows:
Day 1+3: epitalon 2mg subQ (AM), TAM818 + Astaxanthin (AM/PM)
Day 2+4: TAM818+Astaxanthin (AM only day 4)
Day 5-7: break

 

Rationale: I reasoned previously that epitalon upregulates telomerase assembly via nucleolar proteins and combining this with a supplement that increases the genetic expression of unassembled telomerase mRNA would be synergistic.

 

I reasoned increased nucleolus activity would last much longer than upregulated mRNA (days for ribosomes Vs minutes for mRNA) so I could dose epitalon less often.

 

A powerful mitochondrial antioxidant like astaxanthin should also minimize the telomerase diverted from the nucleus to deal with ROS.

 

This theoretical plan seemed to bear fruit as I got a far more powerful effect with this protocol than using epitalon alone in the past. For example, several times previously I have done an epitalon 'cycle' of 10mg/day for 10 days with no adverse reaction. This time I found that more than 3mg of epitalon in a single dose caused me to break out. I put this down to an overactivated immune system (see below).

 

Additions: at different times I also tried adding silymarin or gotu kola (high content of asiaticoside and madecassoside) supplements. I noticed no additional effect from silymarin (it is a weak telomerase activator compared to TAM818). Adding gotu kola caused breakouts however, suggesting it is a potent TA (it is not known at what stage of the telomerase process it acts).

 

mTOR inhibition: I also reasoned that decreasing cellular proliferation via mTOR inhibition would increase telomere length for a given quantity of telomerase produced. For more information see here. So after 7 iterations of the basic TA protocol, with various changes to find the right dose, at the end of May I added everolimus to the protocol. I take 2.5 mg on Day 1+3. The only side effects have been some tiredness on Day 3. I have completed 8 iterations of the modified TA protocol.

 

Results

 

There are three significant positive results from this protocol.

 

1.Hair Growth. I've still got a full head of hair (age 43) with zero grey, but it has been looking thinner. I was really noticing that compared to my son's hair - when we both got it cut to the same length - you could see my scalp but not his. This protocol has thickened my hair to the extent that you can't see the scalp anymore, even after a haircut.

 

2. Immunity. My partner got covid-19; my team at work got covid-19. But despite sharing a bed with my partner and a long trip on a train with my team (sitting next to people who shortly afterwards came down with it), I didn't get ill and continued to test negative. I've also had no other illnesses in this period.

 

3. Weight loss. I've lost 3.1kg since I started this protocol. I have started swimming regularly, so it is unclear whether I just have more energy to exercise, or whether it's a direct effect. I have reduced other exercise in this period (weights and martial arts) to make way for swimming.

 

There are three minor changes (mostly improvements) in daily monitored biomarkers.

 

1.HRV has trended gently upwards during this period. (HRV Average 70.46 vs. 70.24 or RMSSD 100.37 vs. 96.6) comparing with equal length period prior to starting protocol)
It is notable that I've had many more readiness 10/10 days during this period (being well rested with balanced sympathetic and parasympathetic activity).

 

2.BP slightly down (123.26/62 Vs 126.13/65.8)

 

3. But heart rate is slightly up (55.67 vs. 53.9).

 

There are two areas where I was hoping for improvements but haven't (yet) obtained them:

 

1.I've had no improvements in my skin appearance. 

 

2.The grey in my stubble/beard (3-5% of total) has not changed.

 

Future plans

The potency of gotu kola surprised me. I may try substituting TAM818 or epitalon and observe the results.

Using an mTOR inhibitor was partially motivated by a fear that I was not generating enough telomerase to compensate for the telomere shortening of cellular proliferation. But if I have a powerful enough TA protocol, I may actually be able to stimulate proliferation and get a better/faster result, because telomerase can only actually elongate telomeres during S-phase (DNA copying).


Edited by QuestforLife, 02 August 2022 - 01:40 PM.

  • Informative x 6
  • WellResearched x 1
  • like x 1

#783 dlewis1453

  • Member
  • 151 posts
  • 45
  • Location:USA
  • NO

Posted 03 August 2022 - 07:27 PM

 

 

 

This theoretical plan seemed to bear fruit as I got a far more powerful effect with this protocol than using epitalon alone in the past. For example, several times previously I have done an epitalon 'cycle' of 10mg/day for 10 days with no adverse reaction. This time I found that more than 3mg of epitalon in a single dose caused me to break out. I put this down to an overactivated immune system (see below).

 

 

 

Great experiment with promising results! The hair growth in particular is a great sign that something powerful is happening. I'm curious about a couple of things: 

 

1. Why do you think acne break out is a sign of an overactivated immune system? Could this be caused by an elevated immune response against P. Acnes? 

 

2. Do you think demethylating agents will need to be paired with this protocol to prevent the development of the "selfish" stem cells you have discussed earlier? 


  • Cheerful x 1

sponsored ad

  • Advert

#784 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 03 August 2022 - 08:55 PM

Great experiment with promising results! The hair growth in particular is a great sign that something powerful is happening.

Hair growth and telomerase is well established with several major papers published (for example this one from Blasco, doi: 10.1083/jcb.200704141)

I'm curious about a couple of things:

1. Why do you think acne break out is a sign of an overactivated immune system? Could this be caused by an elevated immune response against P. Acnes?

A couple of things made me think that. Firstly my hayfever was particularly bad at the time and I wondered if it was activating my immune system (not an unreasonable conclusion if WBCs have more ability to divide into larger numbers). Secondly my complete immunity to catching covid despite many exposures made me think it might be an immune effect.


But it's all just guesswork. And then I read this..

Telomerase reverse transcriptase induces basal and amino acid
starvation-induced autophagy through mTORC


Here, we report that TERT inhibits the kinase activity
of mTOR complex 1 (mTORC1) in multiple cell lines, resulting in the activation of autophagy under both
basal and amino acid-deprived conditions.
http://dx.doi.org/10...brc.2016.08.094

This isn't that surprising, given we already know telomerase upregulates mitophagy. But it blew me away because the only other time I've had acne break outs from my stack is when I used rapamycin with grapefruit juice. This is a well known side effect of rapamycin (see DOI:10.1159/000088511. This is also now reported on Alan Green's website as the MAIN side effect of rapamycin in his patients).

But I had this side effect BEFORE I even added everolimus to the protocol. So maybe it is possible all my telomerase supplements are activating autophagy and this is behind the breakouts.

The paper does seem strange and contradictory as I thought mTOR inhibition would reduce proliferation - the reverse of what telomerase encourages. But maybe that is only an mTOR2 not mTOR1 thing? I.e. telomerase only turns down mTOR1 but leaves mTOR2 (and proliferation) alone.

2. Do you think demethylating agents will need to be paired with this protocol to prevent the development of the "selfish" stem cells you have discussed earlier?

I've discussed numerous times with Turnbuckle that I believe the epigenetic age accelaration from telomerase activators is simply because they permit more cell divisions, so more opportunities for the epigenetic machinery to fall behind. I've then theorised this can lead to selection for selfish (non-differentiating) stem cells.

But as I'm taking everolimus to reduce proliferation (via mTOR2?) to get greater telomere extension, I should be reducing the amount of epigenetic drift.

Nevertheless I have continued to take AKG when I remember (every few days) as this was effective at stopping epigenetic aging when taking TAM818 previously.

Edited by QuestforLife, 03 August 2022 - 09:01 PM.

  • Informative x 2

#785 dlewis1453

  • Member
  • 151 posts
  • 45
  • Location:USA
  • NO

Posted 04 August 2022 - 08:12 PM


But as I'm taking everolimus to reduce proliferation (via mTOR2?) to get greater telomere extension, I should be reducing the amount of epigenetic drift.
 

 

And the everolimus also helps to focus the telomere lengthening towards the stem cell populations rather than the somatic cells? 

 

You wrote earlier that: "we should be aiming to inhibit mTOR whilst boosting telomerase activity. This to me looks like the best bet for age reversal because it targets stem cell renewal whilst providing them with more of what they need to renew, namely telomerase. Recent increases in our understanding of the various stages of telomerase activation [6] also aid this cause."

 

Thinking along those lines, I wonder if the AKG is having a similar effect to the everolimus. Could you benefit by adding stearic acid as well? 



#786 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 04 August 2022 - 09:15 PM

And the everolimus also helps to focus the telomere lengthening towards the stem cell populations rather than the somatic cells?

You wrote earlier that: "we should be aiming to inhibit mTOR whilst boosting telomerase activity. This to me looks like the best bet for age reversal because it targets stem cell renewal whilst providing them with more of what they need to renew, namely telomerase. Recent increases in our understanding of the various stages of telomerase activation [6] also aid this cause."

Thinking along those lines, I wonder if the AKG is having a similar effect to the everolimus. Could you benefit by adding stearic acid as well?


The basic idea is to reduce proliferation (but not stop it entirely) so a given amount of telomerase will give greater lengthening. The reason you don't want to stop proliferation entirely is because telomerase can only add to telomeres during s-phase (when DNA copies). It's a balancing act.

I believe AKG might actually encourage division because it can be converted to glutamine. But it acts as a demethylase so all things being equal will reduce differentiation, which we do want (it is more complicated than this as demethylation maintains pluripotency in adult stem cells but causes differentiation in ESCs). I'll still use it - intermittently. My feeling on AKG is you top it up and you're dome for a while (and my results support long breaks).

Stearic acid is a fasting trigger. May have a place in this protocol. But at the moment I'm steering clear of C60 as that will trigger proliferation.

You could come up with a completely different protocol timing TA with your stem cell release, but I'm playing it cautiously at the moment.
  • Good Point x 1
  • Informative x 1

#787 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 16 August 2022 - 05:39 PM

Paving the way for the 'speeding car': Explaining the benefits of long telomeres in the context of mTOR

I recently wrote a short paper explaining why long telomeres are an evolutionary adaptation to fast growth. I decided to write my ideas up in a paper because I've never read anything like them in the literature.

The explanation given in this paper brings telomere length (as well as ROS defence) under the umbrella of Blagosklonny's Hyperfunction theory, as adaptations that permit faster growth. This also has important implications for Anti-Aging research because it explains how longer telomeres can offset the harm of mTOR.

I would like to get this published in a respected journal, so any comments on the (non-peer reviewed) draft attached here would be most welcome.

Attached Files


  • Informative x 2
  • WellResearched x 1
  • like x 1

#788 johnhemming

  • Member
  • 311 posts
  • 122
  • Location:Birmingham, UK

Posted 16 August 2022 - 06:54 PM

a) I have found journals very anti anyone who does not come from academia so I have given up trying to publish in journals and simply blog things.

 

b) Telomerase is encoded by hTert.  One thing that stimulates hTert is hyperacetylation of the histone. Not sure about others.


Edited by johnhemming, 16 August 2022 - 06:54 PM.

  • Agree x 1

#789 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 16 August 2022 - 09:17 PM

a) I have found journals very anti anyone who does not come from academia


Very true. And yet so often the answers come from outside of expected channels.

No one else in aging research or evolutionary biology in gemeral has made the connection between telomere length and hyperfunction (mTOR). So I'll try and push it out there anyway I can.
  • Good Point x 1
  • like x 1

#790 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 17 August 2022 - 06:13 PM

Here is the draft paper, edited for clarity. I hope it is of interest and sparks some debate.

Paving the way for the 'speeding car': the evolutionary advantage of long telomeres in the context of high mTOR

Draft manuscript (not peer reviewed)
M B Williams
Aug 16 2022 Updated Aug 17

Abstract

Small animals have long telomeres but short lifespans, which is in direct contradiction to what is observed in cell culture, where long telomeres delay senescence. This paper resolves this paradox by explaining longer telomeres (and active telomerase) as a selective adaptation to greater growth signalling, as described by Hyperfunction theory. From an evolutionary perspective longer telomeres are selected to delay senescence (geroconversion), permitting faster development and potentially extended reproduction, rather than longer life. This may also be permissive for cancer, which is why longer telomeres are precluded in larger animals. But from an anti aging perspective, activating telomerase is likely to synergise with post-development mTOR inhibition.

The understanding presented in this paper extends the influence of Blagosklonny's proximal driver of aging, mTOR signalling, to other putative aging mechanisms, primarily the Hayflick limit, previously posited to limit life only after hyperfunction.

*

Telomeres are a paradox in aging. Long telomeres and active telomerase are associated with faster aging species like mice, whereas humans have shorter telomeres and inactive telomerase, but live much longer [1]. This is in contrast to the benefits of longer telomeres in cell culture, where they prolong cellular division and delay the gradual conversion to senescence (geroconversion), as demonstrated in a recent rework of Hayflick's experiments [2], where it was proven that senescence sets in gradually and long before cells reach the Hayflick limit. This presents us with the possibility that longer telomeres could be an advantage during the growth phase of a young animal, and also, indirectly, delay aging.

In the light of hyperfunction theory [3], longer telomeres can be understood as an evolutionary adaptation to offset the geroconversion caused by faster growth.
Longer telomeres permit cells to divide rather than become blocked in the cell cycle and then become senescent. Active telomerase reduces or eliminates telomere shortening, and the consequent slowing of division and increase in geroconversion that occurs with continual growth stimulation. The greater the stimulation, the faster the division, and the greater the telomerase production required to offset it. This is a direct consequence of telomere length itself, where shorter telomeres mean slower cellular division, and greater cell size, as can be seen in cell culture experiments.

Therefore longer telomeres are an adaptation to permit faster growth. But despite their much longer telomeres and active telomerase enzyme, mice still lose telomere repeats faster than humans [4].
This parallels the situation with murine reactive oxygen species (ROS) defence, which regardless of its efficacy relative to humans, cannot compensate for their higher ROS production[5]. Telomere length (and possibly also ROS defence) are likely increased by evolution to protect against the senescence induced by faster growth. Without such adaptation, aging might occur before reproductive maturity in fast growing animals.

On the flip side, it has been postulated that long telomeres are a risk factor for cancer and so are selected against in larger species with a protracted growth phase, despite the deleterious effects of short telomeres in later life [6], which exist in a post reproductive evolutionary 'shadow'. This is as expected, given longer telomeres delay senescence, which is protective against cancer.

Despite the logic of this theory, and indirect support from Mendelian studies in humans [7], it is contradicted by some experiments - where mice created from embryonic stem cells with hyperlong telomeres were healthier and lived longer than controls, with a delay in diseases of aging, including cancer [8]. Longer lifespans were also achieved by providing middle and old aged mice with telomerase gene therapy, without increases in cancer [9].

It is unknown why increases in cancer were not seen in these studies, but mice born with hyperlong telomeres were metabolically healthier than controls[5], suggesting telomere length regulation of growth signalling (more on this below). Further stratification of the data in Mendelian studies[7] may discover that those humans that have longer telomeres and went on to develop cancer, also matured faster because of elevated growth signalling and that this is the real driver of cancer occurrence, with longer telomeres merely being permissive.

Returning to aging, if antioxidant defence is akin to 'catching bullets', then longer telomeres can be considered as 'paving the way' for Blagosklonny's 'Speeding Car without brakes' [10]. Paving more road can delay the crash (aging), which is what is required for reproductive success in fast growing animals.

In further support of this idea, it has been found that telomerase inhibits mTOR1 without inhibiting mTOR2 and proliferation [11]. Inhibition of mTOR1 upregulates autophagy (an adaptation to faster cellular protein production) and can be understood as negative feedback against excessive growth stimulation, allowing continued proliferation with reduced geroconversion (senescence). This is utilised by cancer, but equally it might also be used by other fast proliferating cells like human T lymphocytes (which do have active telomerase). So perhaps mTOR does have brakes, after all. This may explain why mTOR stimulation is so harmful later in life, as telomere length is reduced.

What other evidence do we have for the theory that longer telomeres are an adaptation to faster growth?

1.Similar to the body-size longevity relationship [12], where larger species live longer, but smaller individuals within those species live the longest, telomeres are shorter in large, slow growing creatures but longer in smaller, faster growing creatures. This matches the hypothesis that longer telomeres are required to offset geroconversion in species that grow faster. But within a species, longer telomeres (like smaller size), are associated with longer life. This is because telomere length is largely set in infancy [13] and those with most telomere repeats remaining after development is complete will live the longest.

This is exemplified by African killifish [14]. Killifish evolve their life cycle to match the pools in which they live. In areas with a shorter wet season, killifish must grow, reproduce and lay eggs before the pools dry out, and in this case the species has long telomeres, as is predicted by the requirement for faster growth, relative to the killifish species that live in longer lasting pools. But within a given killifish species, those individuals with longer telomeres will survive the longest, showing longer telomeres are beneficial later in life, even when the body is fully grown.

2. People of African ancestry have longer telomeres than Europeans [15]. People of African ancestry also grow up faster than Europeans, on average, as seen in studies of menarche onset [16]. Therefore even within the human species there is evidence for longer telomeres within faster maturing ethnic groups.

Furthermore, it is also expected that a smaller, slower developing individual, if they should happen to also inherit longer telomeres, would be very long lived indeed, benefiting from both lower growth stimulus, and greater compensating telomere length.

Conclusions and Consequences for Aging Research

Blagosklonny proposed hyperfunction driven aging is proximal [17], with other potential aging mechanisms like the Hayflick limit and ROS limiting life only if hyperfunction is prevented first.

In this short paper I propose that the Hayflick limit (and ROS) are not independent from growth signalling (mTOR), but argue that their regulation has evolved in direct response to the level of mTOR.

Evidence is presented here to show that longer telomeres are a selective adaptation that compensates for faster growth driven by mTOR. This compensation is sufficient to maintain health to reproductive age, and likely permits faster growth than would otherwise be possible.

Future developments in anti aging research should focus on telomerase activation as an adjuvant to mTOR inhibition. Such a combination is likely to be synergistic because whereas telomerase permits more cell division, mTOR inhibition (specifically inhibiting mTOR2) reduces proliferation. This preservation of telomere length will then lead to extended post development lifespan, so long as growth signalling remains repressed.

mTOR inhibition is regarded as deleterious before development is complete. Because of cancer concerns, telomerase activation might likewise also be started later in life, when telomeres have shortened relative to their length during development.

References:

[1] Rodrigo T. Calado et al.,Science Direct, 11 June 2013, Telomere dynamics in mice and humans,
https://doi.org/10.1...tol.2013.03.030

[2] Michelle Chan et al., Elifesciences, Feb 4 2022, Novel insights from a multiomics
dissection of the Hayflick limit
https://doi.org/10.7554/eLife.70283

[3] Mikhail V. Blagosklonny, Cell Cycle, Sept 15 2006, Aging and immortality: quasi-programmed senescence and its pharmacologic inhibition
https://doi.org/10.4161/cc.5.18.3288

[4] Kurt Whittemore et al., PNAS, July 8 2019, Telomere shortening rate predicts species life span https://doi.org/10.1...pnas.1902452116

[5] Gustavo Barja, Science Direct, August 20 2014, The Mitochondrial Free Radical Theory of Aging https://doi.org/10.1...94625-6.00001-5

[6] Peter M Lansdorp, Blood, Feb 10 2022, Telomeres, Aging and Cancer: The Big Picture, https://doi.org/10.1...lood.2021014299

[7] The Telomeres Mendelian Randomization Collaboration, JAMA Oncol, May 2017, Association Between Telomere Length and Risk of Cancer and Non-Neoplastic Diseases https://doi.org/10.1...oncol.2016.5945

[8] Miguel A. Muñoz-Lorente et al., Nature Communications, Oct 17 2019, Mice with hyper-long telomeres show less metabolic aging and longer lifespans
https://doi.org/10.1...467-019-12664-x

[9] Bruno Bernardes de Jesus et al., EMBO Mol Med, May 15 2012, Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer
https://doi.org/10.1002/emmm.201200245

[10] Mikhail V. Blagosklonny, Cell Cycle, Dec 15 2009, TOR-driven aging: speeding car without brakes https://doi.org/10.4161/cc.8.24.10310

[11] Muhammad Ali et al., Science Direct, Sept 23 2016, Telomerase reverse transcriptase induces basal and amino acid starvation-induced autophagy through mTORC1 http://dx.doi.org/10...brc.2016.08.094

[12] Mikhail V. Blagosklonny, Aging, April 20 2013, Big mice die young but large animals live longer https://doi.org/10.18632/aging.100551

[13] Jacob B Hjelmborg et al., Medical genetics, Oct 14 2014, The heritability of leucocyte telomere length dynamics
http://dx.doi.org/10...net-2014-102736

[14] Martin Reichard et al., Wiley Online Library, Nov 26 2021, Lifespan and telomere length variation across populations of wild-derived African killifish https://doi.org/10.1111/mec.16287

[15] Steven C Hunt, Wiley Online Library, July 10 2008, Leukocyte telomeres are longer in African Americans than in whites https://doi.org/10.1...26.2008.00397.x

[16] Maria E. Bleil et al., Springer Link, Aug 7 2017, Race disparities in pubertal timing: Implications for cardiovascular disease risk among African American women https://doi.org/10.1...1113-017-9441-5

[17] Mikhail V. Blagosklonny, Aging, Dec 30 2012, Answering the ultimate question "what is the proximal cause of aging?" https://doi.org/10.18632/aging.100525

Attached Files


Edited by QuestforLife, 17 August 2022 - 06:15 PM.

  • Informative x 3
  • Well Written x 2

#791 dlewis1453

  • Member
  • 151 posts
  • 45
  • Location:USA
  • NO

Posted 20 August 2022 - 07:02 PM

Here is the draft paper, edited for clarity. I hope it is of interest and sparks some debate.
 

 

Thanks so much for compiling your insightful research and ideas concerning Telomeres and mTOR into this paper! I have read through it and found it very interesting but am reading it through again a few more times to properly digest everything. 

 

One initial thought I had was that it might be informative to expand your analysis of the link between Telomeres and mTOR to include epigenetics. You have already put forward convincing theories and research in earlier posts that seek to explain (i) why increasing telomerase leads to an increase in epigenetic age in the short term, and (ii) why increasing telomeres in stem cells will ultimately lead to a decrease in body-wide epigenetic age in the long term. Not only would this discussion be interesting and tie in nicely with mTOR, but it may also increase the impact of your paper, since the epigenetics of aging is a hot topic currently. Although I understand that it may be outside the scope of your current paper. 


  • Good Point x 2

#792 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 21 August 2022 - 04:34 PM

Thanks so much for compiling your insightful research and ideas concerning Telomeres and mTOR into this paper! I have read through it and found it very interesting but am reading it through again a few more times to properly digest everything.

Bill Andrews has been kind enough to give me some pointers and as a result I've made some changes to the paper (attached). I've also uploaded it here: https://independent....MarkWilliams143

I've also written an even shorter paper explaining the core of my idea about long telomeres evolving in response to fast growth conditions. I'll post that here later.

A paper on methylation in long lived cell lines will have to wait for now!

Attached Files


Edited by QuestforLife, 21 August 2022 - 04:35 PM.

  • like x 2

#793 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 23 August 2022 - 03:45 PM

Just how much can we inhibit mTOR?

 

Hyperfunction theory proposes that aging is the arrest and subsequent geroconversion (senescence) of post mitotic cells. The baby grows into an adult, stops growing, but the growth signal keeps going, eventually making that adult old.

 

So let's all turn mTOR off, right?

 

Not so fast. Not all the body is post mitotic. Some pretty important cells continue to divide, think immune and blood cells of all types, intestinal cells in the gut, not to mention buccal cells, skin, nails and hair. The telomere theory of aging argues that it is these cells that age, and cause the rest of the body to become old (astrocytes age and harm neurons, endothelial cells age and harm the heart, etc). Of course the two theories are not mutually exclusive. Slowing mTOR will reduce cellular proliferation and telomere loss. It is just that we don't want to slow it too much.

 

What happened to my hair, dude?

 

So we can't just turn mTOR off, only turn it down. But how much? Let's look at transplant patients. They take (2-20mg) rapamycin everyday. But that is because they want to suppress their immune system. We don't want that. Normal range for WBCs is 4500-11000 cells/uL. Low is under 4000. Just a back of the envelope calculation assuming we start in the middle of the range (7750) and fall to 4000 (but no further), that's a drop of about 50% in number. Assuming WBC loss rates are constant that implies and equal 50% drop in WBC production. What inhibition of mTOR does this relate to? No idea. But it is at least a proxy we can measure. Has anyone measured WBC counts before and after rapamycin or everolimus use?


Edited by QuestforLife, 23 August 2022 - 03:47 PM.

  • Good Point x 2
  • Informative x 1
  • like x 1

#794 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 02 September 2022 - 10:09 AM

Supertelomerase!

 

It is often stated that human cells lack TERT (telomerase reverse transcriptase) but not TERC (telomerase RNA component, sometimes called TR) expression - they generally express the RNA template part that is used to copy the telomere, but lack the catalytic part of the protein that does the copying. So the logical conclusion is that if you want to increase telomere length you need to increase TERT.

 

And yet…humans with mutations in either TERC or TERT suffer from short telomere syndromes (1); I discussed previously using TERC upregulation to increase telomere length in stem cells (2), and discussed here how GDF11 lengthens telomeres in MSCs via TERC upregulation (3). Therefore TERC concentration can be limiting in some situations.

 

I also discussed here how Telomerase is made in the nucleolus (4)(5)(6)(7), and how it may be possible to increase telomerase levels by increasing assembly of TERT and TERC in the nucleolus.

 

Therefore we have a three part strategy:

1. Increase TERT
2. Increase TERC
3. Increase assembly speed/efficiency of TERT and TERC into complete telomerase protein

 

Today I'd like to talk about a paper that shows how effective putting 1. and 2. together could be. Overexpressing TERT and TERC together produces 'Supertelomerase', a name I rather like!

 

Telomere length homeostasis requires that telomerase levels are limiting (8) 

 

 

Concomitant overexpression of TERT and TR was necessary and sufficient to substantially increase telomerase activity. Upon overexpression, more telomerase associated with telomeres and telomeres elongated at a constant rate (up to 0.8 kb/population doubling (PD)) in a length-independent manner. Thus, in less than 50 PDs, the length of telomeres increased 3–8-fold beyond physiological size, while telomere-bound TRF1 and TRF2 increased proportionally to telomere length. Thus, long telomeres do not permanently adopt a structural state that is non-extendible

 

What they show in this paper is that whilst short telomeres may be preferentially lengthened, long telomeres do not become blocked from further elongation, which it was theorized that the telomere binding proteins (TRF1 and TRF2) might do.

 

But more importantly for us, they showed the relative contributions of TERT and TERC to telomere lengthening via production of Supertelomerase.

 

I shall concentrate on the part of the study where they looked at normal, non-cancer, human cells.

 

 

we also determined the effects of super-telomerase expression in primary telomerase-negative human lung fibroblasts (HLF). Also in HLF, co-overexpression of hTERT and hTR most efficiently increased telomerase activity. Telomere length increased at a constant rate (780 bp - per division (ed.)) to 31 kb after 28 PD, which corresponds to at least twice the physiological size of telomeres in embryos and six times the initial size at the time of infection. Thus, also in primary cells telomere overelongation was not prevented upon supertelomerase expression.

 

As you can see in the picture (B) below (8), over expressing TERC had no impact of telomerase levels (as has been found in many human cell line studies previously), over expressing TERT increased telomerase levels 100 times, but wait for it…over expressing TERC and TERT together increased telomerase 350 times!

 

embj7600952-fig-0003-m.jpg

 

So even though TERC does not seem to be rate limiting in the creation of the complete telomerase protein in normal circumstances, it can become rate limiting if there is an excess of TERT. It's just chemistry after all!

 

Therefore, I conclude that searching exclusively for activators of TERT (as Bill Andrews does(9)) may be a mistake. Each cell will have a certain concentration of TERC and TERT and a certain efficiency of their assembly that will in part depend on their relative concentration and co-location in the nucleolus. We should search for ways of increasing 1, 2 and 3. Moreover, the deleterious effect of TERC mutations on telomeropathies, and the low concentration of TERT in human bone marrow stem cells, suggests the point at which TERC concentration becomes limiting is low and therefore accessible to intervention.

 

References

  1. Holohan B, Wright WE, Shay JW. Cell biology of disease: Telomeropathies: an emerging spectrum disorder. J Cell Biol. 2014 May 12;205(3):289-99. doi: 10.1083/jcb.201401012. PMID: 24821837; PMCID: PMC4018777.
  2. Nagpal N, Wang J, Zeng J, Lo E, Moon DH, Luk K, Braun RO, Burroughs LM, Keel SB, Reilly C, Lindsley RC, Wolfe SA, Tai AK, Cahan P, Bauer DE, Fong YW, Agarwal S. Small-Molecule PAPD5 Inhibitors Restore Telomerase Activity in Patient Stem Cells. Cell Stem Cell. 2020 Jun 4;26(6):896-909.e8. doi: 10.1016/j.stem.2020.03.016. Epub 2020 Apr 21. PMID: 32320679; PMCID: PMC7275922.
  3. Mutual regulation between GDF11 and TET2 prevents senescence of mesenchymal stem cells Jiaming Gao, Hao Wang, Junyan Shen, Xiaojing Liu, Xiaoqi Zhu, Enfeng Zhao, Gongchen Li, Yao Sun, Feng Yin, Zhongmin Liu, Yi Eve Sun, Hailiang Liu, doi: https://doi.org/10.1...20.03.30.008722
  4. Coutts, F., Palmos, A.B., Duarte, R.R.R. et al. The polygenic nature of telomere length and the anti-ageing properties of lithium. Neuropsychopharmacol 44, 757–765 (2019). https://doi.org/10.1...1386-018-0289-0
  5. Jessica Nolte.Lrrc34 Interacts with Oct4 and Has an Impact on Telomere Length in Mouse Embryonic Stem Cells.Stem Cells and Development.Nov 2021.1093-1102.http://doi.org/10.1089/scd.2021.0113
  6. Sandra Lührig, Iliana Siamishi, Marieke Tesmer-Wolf, Ulrich Zechner, Wolfgang Engel, and Jessica Nolte.Lrrc34, a Novel Nucleolar Protein, Interacts with Npm1 and Ncl and Has an Impact on Pluripotent Stem Cells.Stem Cells and Development.Dec 2014.2862-2874.http://doi.org/10.1089/scd.2013.0470
  7. Iarovaia OV, Minina EP, Sheval EV, Onichtchouk D, Dokudovskaya S, Razin SV, Vassetzky YS. Nucleolus: A Central Hub for Nuclear Functions. Trends Cell Biol. 2019 Aug;29(8):647-659. doi: 10.1016/j.tcb.2019.04.003. Epub 2019 Jun 5. PMID: 31176528.
  8. Cristofari G, Lingner J. Telomere length homeostasis requires that telomerase levels are limiting. EMBO J. 2006 Feb 8;25(3):565-74. doi: 10.1038/sj.emboj.7600952. Epub 2006 Jan 19. PMID: 16424902; PMCID: PMC1383536.
  9. https://sierrasci.co...strategic-plan/

Edited by QuestforLife, 02 September 2022 - 10:17 AM.

  • WellResearched x 5
  • Good Point x 1
  • Informative x 1

#795 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 11 September 2022 - 01:40 PM

Mitochondrial hyperfusion via metabolic sensing of regulatory amino acids (1)

This is a very interesting paper on mitochondrial hyperfusion triggered by the consumption of certain amino acids (AAs) specifically: glutamine, leucine, arginine.

Firstly, a mild fusion state is triggered by cell AA starvation and then severe hyperfusion is induced by addition of the three AAs mentioned. Maximum effect is from 4-8 hours.

Effect requires absence of other AAs, which block fusion. But glucose only partially blocks fusion.

So a human could overnight fast, then take Glut+leuc+argin to get these effects (if desired). Fasting should continue at least 4 hours, with lunch being carb (not protein) based.

Mechanism of action was examined with the two most effective AAs of the three (glut+leuc) acting through Krebs cycle and downstream, the creation of GTP, which upregulates fusion protein mfn1. One wonders if direct supplementation of the guanosine nucleoside would be easier (if possible)?

Of further note:

1. dGTP concentrations increase speed of the telomerase protein activity, see my discussion link(2), paper ref (3). Therefore upregulation of GTP by this mechanism could benefit (stem) cells that have low telomerase levels by increasing how many base pairs telomerase can add to the telomere during s-phase.

2. Alpha ketoglutarate has been shown to reverse methylation age, and likely overlaps with the AA pathway described above, see my discussion link(4), paper ref (5). So leucine and glutamine consumption would be expected to do the same (my prediction).

3. As a corollary of the above, depletion of glutamine, leucine and arginine would be expected to increase methylation age (again, my prediction), and might explain negative effect of telomerase activators on methylation-age, mediated via increased cellular division (which is known to use up glutamine, see my previous discussion (6)).

4. The advantages of mitochondrial hyperfusion for Turnbuckle's 'stem cell protocol' are obvious (7).

References:

(1)Mitochondrial hyperfusion via metabolic sensing of regulatory amino acids, Abdullah et al., 2022, Cell Reports 40, 111198 August 16, 2022 https://doi.org/10.1...rep.2022.111198

(2) Chen Y, Podlevsky JD, Logeswaran D, Chen JJ. A single nucleotide incorporation step limits human telomerase repeat addition activity. EMBO J. 2018 Mar 15;37(6):e97953.https://doi:10.15252/embj.201797953, Epub 2018 Feb 12. PMID: 29440226; PMCID: PMC5852417.

(3)Nucleotides (specifically guanine) for elongation of telomeres: eat Anchovies and Herring!
https://www.longecit...-15#entry904277

(4)Plan to reduce both telomere and methylation age:
https://www.longecit...e-9#entry895170
No improvement in methylation age from 3 months of AKG:
https://www.longecit...-10#entry896760
Improvement in methylation age from 6 months of AKG:
https://www.longecit...-13#entry899822

(5)Demidenko O, Barardo D, Budovskii V, Finnemore R, Palmer FR, Kennedy BK, Budovskaya YV. Rejuvant®, a potential life-extending compound formulation with alpha-ketoglutarate and vitamins, conferred an average 8 year reduction in biological aging, after an average of 7 months of use, in the TruAge DNA methylation test. Aging (Albany NY). 2021 Nov 30;13(22):24485-24499. https://doi:10.18632/aging
203736 Epub 2021 Nov 30. PMID: 34847066; PMCID: PMC8660611.

(6)Glutamine depletion from telomerase activators? https://www.longecit...-24#entry911858

(7)Turnbuckle's protocol: https://www.longecit...-24#entry911858
  • Informative x 4
  • WellResearched x 2

#796 Castiel

  • Guest
  • 374 posts
  • 86
  • Location:USA

Posted 16 September 2022 - 05:07 PM

I read mention of gotu kola. Would like to remind that while potent Ive heard it has long half life and can accumulate to dangerous levels if used indefinitely. It is preferable to use it for short periods.

Also while checking thread saw mention of rock inhibitors, dont know if previously mentioned but it seems resveratrol may be a rock inhibitor

>This confirms the proposed hypothesis that resveratrol may be a potential inhibitor of Rho kinase in smooth muscle. In the literature, we can find data confirming the inhibition of ROCK by resveratrol already at the concentration of 0.03 mM/l. https://www.ncbi.nlm...les/PMC6996688/

Btw quest, what dose of astaxanthin did you use in your protocol?
  • Good Point x 1
  • Informative x 1

#797 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 17 September 2022 - 08:23 AM

I read mention of gotu kola. Would like to remind that while potent Ive heard it has long half life and can accumulate to dangerous levels if used indefinitely. It is preferable to use it for short periods.

Also while checking thread saw mention of rock inhibitors, dont know if previously mentioned but it seems resveratrol may be a rock inhibitor

>This confirms the proposed hypothesis that resveratrol may be a potential inhibitor of Rho kinase in smooth muscle. In the literature, we can find data confirming the inhibition of ROCK by resveratrol already at the concentration of 0.03 mM/l. https://www.ncbi.nlm...les/PMC6996688/

Btw quest, what dose of astaxanthin did you use in your protocol?


In the TA protocol I do 4 days on, 3 days off. I agree with you about the gotu kola.

Not convinced about the resveratrol; at those doses it will affect all sorts of receptors and therefore have all sorts of unintended side effects.

The astaxanthin soft gels are 4mg/each. I've dropped it now as I suspect it's an mTOR inhibitor and it was adding to the effect of everolimus.
  • Informative x 2
  • Good Point x 1

#798 Learner056

  • Guest
  • 96 posts
  • 3
  • Location:Lake Camelot, WI

Posted 25 September 2022 - 06:09 PM

QuestForLife:  Correct me if I am wrong, and if able to answer, my kind regards: 

 

Fact1: We know that nucleotides as GTP (or its likes UTP, ATP etc) provides chemical energy (and other things).  The concept of 'hyperfusion' relies on GTP etc.  

Fact2: UCPs (besides their beneficial side) waste energy aka take it away from Fusion. 

Fact3: UCPs get in-activated by Nucleotides.  

Fact4: UCPs get in-activated by Polyamines (Agmatine, Spermidine) too. 

 

Question:  Do we know (or can guess) the effect of Nucleotides vs Polyamines on Fusion/stem cell stimulation

 

Mechanism of action was examined with the two most effective AAs of the three (glut+leuc) acting through Krebs cycle and downstream, the creation of GTP, which upregulates fusion protein mfn1. One wonders if direct supplementation of the guanosine nucleoside would be easier (if possible)?

4. The advantages of mitochondrial hyperfusion for Turnbuckle's 'stem cell protocol' are obvious (7).
 

 

Question:  Is there a fear that AKG gets hijacked (by some process) in transit or something that you are trying it in liposomal?.  So far you feel any difference b/w the 2 versions? 

 

You wrote: 

In addition I have begun to supplement (homemade) liposomal AKG.

 

 

Question:  I don't know much about this, my initial searches indicate that GDF would age skeletal muscle?  

You wrote: 

GDF11

 

Question: I am exploring agents (topical/oral - dual use) that could stimulate Melanocyte SCs.  i.e. increase symmetric+assymetric replication.  I understand the constraint here is that to expand the pool you need atleast 1 SC within hair follicle, is that true?  Can't stem cells be mobilized from somewhere else and put near hair follicles?

 

Question: I feel that Polyamines (Agmatine, Spermidine) very potently play a role in Fusion, but I can be wrong?  They give strong energy.  Any indication where they fit in Fusion vs Fission.  Also Alpha Lipoic while it does not give energy, but it feels good in Fusion.  (Fyi: I am not specifically looking for energy, but mention it, as it is a material characteristic of importance in fusion and proliferation as I learnt from you people here)


Edited by Learner056, 25 September 2022 - 06:19 PM.

  • Well Written x 2

#799 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 26 September 2022 - 12:11 PM

QuestForLife:  Correct me if I am wrong, and if able to answer, my kind regards: 

 

Fact1: We know that nucleotides as GTP (or its likes UTP, ATP etc) provides chemical energy (and other things).  The concept of 'hyperfusion' relies on GTP etc.  

Fact2: UCPs (besides their beneficial side) waste energy aka take it away from Fusion. 

Fact3: UCPs get in-activated by Nucleotides.  

Fact4: UCPs get in-activated by Polyamines (Agmatine, Spermidine) too. 

 

Question:  Do we know (or can guess) the effect of Nucleotides vs Polyamines on Fusion/stem cell stimulation

 

 

Question:  Is there a fear that AKG gets hijacked (by some process) in transit or something that you are trying it in liposomal?.  So far you feel any difference b/w the 2 versions? 

 

You wrote: 

In addition I have begun to supplement (homemade) liposomal AKG.

 

 

Question:  I don't know much about this, my initial searches indicate that GDF would age skeletal muscle?  

You wrote: 

GDF11

 

Question: I am exploring agents (topical/oral - dual use) that could stimulate Melanocyte SCs.  i.e. increase symmetric+assymetric replication.  I understand the constraint here is that to expand the pool you need atleast 1 SC within hair follicle, is that true?  Can't stem cells be mobilized from somewhere else and put near hair follicles?

 

Question: I feel that Polyamines (Agmatine, Spermidine) very potently play a role in Fusion, but I can be wrong?  They give strong energy.  Any indication where they fit in Fusion vs Fission.  Also Alpha Lipoic while it does not give energy, but it feels good in Fusion.  (Fyi: I am not specifically looking for energy, but mention it, as it is a material characteristic of importance in fusion and proliferation as I learnt from you people here)

 

I am unsure of the influence of nucleosides on UCPs, please add some references if you have that information. We do know they influence fusion/fission of mitochondria, as I posted on previously*. 

 

I have not found it practical to dose Glutamine (+leucine+arginine) however. My telomerase protocol means that I always have lots of cells wanting to divide and this means I can get glutamine depleted (I've measured this and posted it previously). But adding glutamine leads to exhaustion (in my experience). 

 

Supplementing Arginine-AKG may be an indirect way to solve this problem as the body can exchange ketoglutarate and arginine for glutamate and vice versa, but it does not seem to do this at a high rate. 

 

Supplementing a GTP salt should be beneficial both for mitochondrial fusion (whilst fasting) rather than doing this indirectly though amino acids, AND increase the speed of telomerase (see references above), but it is not available as a supplement, only as a very expensive chemical from bespoke suppliers.

 

* It is not clear to me whether the benefits of C60 fullerenes in oil or with a fat come from mitochondrial fusion or mitochondrial uncoupling or some combination. Turnbuckle posits that C60 can block UCPs, but if that is the case then why is triggering UCPs with olive oil beneficial? I would guess that anything that reduces energy supply (ATP) increases lifespan, and this is where the benefits of keto and fasting come from. Adding a powerful antioxidant into the mix seems to somehow make this more long lasting; I've tackled (but not completely understood) this effect with alpha lipoic acid before (1).

 

(1) https://www.scienced...000961?via=ihub

 

ps I am not going to wade in on the GDF11 good or bad argument here. There are studies on either side of that divide, but I believe GDF11 is highly beneficial for muscle so long as you don't over dose it, when it can then trigger GDF8 receptors. 


Edited by QuestforLife, 26 September 2022 - 12:14 PM.

  • Informative x 2
  • Good Point x 1

#800 Learner056

  • Guest
  • 96 posts
  • 3
  • Location:Lake Camelot, WI

Posted 27 September 2022 - 06:46 PM

Very insightful.  UCPs, low ATP vs high ATP is intriguing area to me, as I have sensed accelerated aging with higher ATP (but I am keeping open-mind on varying viewpoints)  

 

Question:  I read that each cell has same genes across our body.  From a stem cell vs aged cell perspective, stem cell would not have the many mutations later acquired through cell division process, however stem cells will still carry the mutations that are foundational? (for e.g. progeroid mutation where DNA repair is seriously slow)

 

 

 

* It is not clear to me whether the benefits of C60 fullerenes in oil or with a fat come from mitochondrial fusion or mitochondrial uncoupling or some combination. Turnbuckle posits that C60 can block UCPs, but if that is the case then why is triggering UCPs with olive oil beneficial? I would guess that anything that reduces energy supply (ATP) increases lifespan, and this is where the benefits of keto and fasting come from. Adding a powerful antioxidant into the mix seems to somehow make this more long lasting; I've tackled (but not completely understood) this effect with alpha lipoic acid before (1).

 

 



#801 Castiel

  • Guest
  • 374 posts
  • 86
  • Location:USA

Posted 29 September 2022 - 03:40 AM

The astaxanthin soft gels are 4mg/each. I've dropped it now as I suspect it's an mTOR inhibitor and it was adding to the effect of everolimus.

Doesn't preliminary data from Interventions testing program suggest Astaxanthin might have significant lifespan extension on rodents?


  • Informative x 1

#802 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 29 September 2022 - 09:40 AM

Doesn't preliminary data from Interventions testing program suggest Astaxanthin might have significant lifespan extension on rodents?

 

I did not know that, but based on my experiments that would not surprise me - astaxanthin clearly added to the effects of everolimus, and for me this was too much: my hair thickness regrowth from the TA protocol reversed. Inhibiting growth is a tricky business and you don't want to overdo it. 



#803 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 29 September 2022 - 09:44 AM

Very insightful.  UCPs, low ATP vs high ATP is intriguing area to me, as I have sensed accelerated aging with higher ATP (but I am keeping open-mind on varying viewpoints)  

 

Question:  I read that each cell has same genes across our body.  From a stem cell vs aged cell perspective, stem cell would not have the many mutations later acquired through cell division process, however stem cells will still carry the mutations that are foundational? (for e.g. progeroid mutation where DNA repair is seriously slow)

 

Yes of course, any mutations you are born with will be present in every cell. As you grow, mutations crop up randomly across tissues, leading to a mosaic effect, but any harm from this is thought to be limited in importance by the hierarchy of stem cells-progenitors-disposable cells, with the stem cells only occasionally dividing, and by somatic cells that once in place almost never divide (neurons, heart cells, etc).

 

It is an interesting paradox that aging leads to a loss of ATP (we have less energy as an organism), but that most (all?) interventions that extend lifespan seem to reduce ATP generation (in cells). 


  • Needs references x 1
  • Informative x 1

#804 Learner056

  • Guest
  • 96 posts
  • 3
  • Location:Lake Camelot, WI

Posted 01 October 2022 - 08:03 PM

I was reading through your thread, you are very young (younger than my own youngest and most of my grandchildren).  The proficiency you have developed already at this age, give you 5 years you will eclipse today's big name scientists.

 

Not directly related, but I read through certain threads on forum about Lapachol (or Pau D'arco).  On surface, when I use Pau D'arco, it revs up metabolic system very fast, that no matter what I eat it just disappears.  I think it activates UCPs, but then that may not be good for stem cells?.  If you have you looked up this herb any opinion?

 

  

Yes of course, any mutations you are born with will be present in every cell. As you grow, mutations crop up randomly across tissues, leading to a mosaic effect, but any harm from this is thought to be limited in importance by the hierarchy of stem cells-progenitors-disposable cells, with the stem cells only occasionally dividing, and by somatic cells that once in place almost never divide (neurons, heart cells, etc).

 

It is an interesting paradox that aging leads to a loss of ATP (we have less energy as an organism), but that most (all?) interventions that extend lifespan seem to reduce ATP generation (in cells). 

 


  • Cheerful x 1

#805 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 02 October 2022 - 08:59 AM

I was reading through your thread, you are very young (younger than my own youngest and most of my grandchildren). The proficiency you have developed already at this age, give you 5 years you will eclipse today's big name scientists.

Not directly related, but I read through certain threads on forum about Lapachol (or Pau D'arco). On surface, when I use Pau D'arco, it revs up metabolic system very fast, that no matter what I eat it just disappears. I think it activates UCPs, but then that may not be good for stem cells?. If you have you looked up this herb any opinion?


Thanks! I'm 43 - 38 according to methylation-age. I feel right on the cusp of the downward slope and am fighting it with everything I have.

Beta-Lapachone is a component of Pau D'arco with potential as a geroprotective. Indeed it increases energy expenditure, which follows the pattern we have found in lifespan extension. Here are some papers that implicate UCP activation:

Beta-Lapachone, a Modulator of NAD Metabolism, Prevents Health Declines in Aged Mice

https://doi.org/10.1...al.pone.0047122

Here, we demonstrate that the facilitation of NQO1 activity by feeding β-lapachone (βL), an exogenous NQO1 co-substrate, prevented age-dependent decline of motor and cognitive function in aged mice. βL-fed mice did not alter their food-intake or locomotor activity but did increase their energy expenditure as measured by oxygen consumption and heat generation.


Beta-lapachone prevents diet-induced obesity by increasing energy expenditure and stimulating the browning of white adipose tissue via down-regulation of miR-382 expression

http://dx.doi.org/10.2337/db15-1423

Here, we showed that BLC stimulated the browning of white adipose tissue (WAT), increased the expression of brown adipocyte-specific genes (e.g., uncoupling protein 1 [UCP1]), decreased body weight gain, and ameliorated metabolic parameters in high-fat diet-fed mice. Consistently, BLC-treated mice showed significantly higher energy expenditure compared to control mice.


  • Informative x 2

#806 Learner056

  • Guest
  • 96 posts
  • 3
  • Location:Lake Camelot, WI

Posted 04 October 2022 - 11:06 PM

QuestForLife, would you be able to shed insight on the 'art' of deciphering research studies - this would be a big favor to this community.  When reading through papers we most often encounter data/effects of a compound on a certain cancer cell line.  In hindsight, naive would dis-regard that information, arguing our normal cells are not cancer cells.  Though taking that approach is self-defeating (akin to denying the value of even mice studies), depriving such strategic data asset would be a big loss (since most research exists on tumorigenic cells).  Recently learning cell-cycle foundations with you, makes me realize that a cancer cell or stem cell or normal cell is conceptually still a 'cell'.  Keeping maybe cell cycle phases or the regulatory CDK and Telomerases etc as key variables. 

 

Key question being that what specifically can be "reasonably" ascertained from these studies vs "ball-park" ascertainment vs it would be just foolishly wrong to infer anything. I realize this is maverick topic, that MANY would not be open-minded to share with others (they would rather hoard it ...as they say, tricks of the trade) but I have faith that if anyone here can selflessly share knowledge, it is you. 



#807 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 05 October 2022 - 09:03 AM

QuestForLife, would you be able to shed insight on the 'art' of deciphering research studies - this would be a big favor to this community.  When reading through papers we most often encounter data/effects of a compound on a certain cancer cell line.  In hindsight, naive would dis-regard that information, arguing our normal cells are not cancer cells.  Though taking that approach is self-defeating (akin to denying the value of even mice studies), depriving such strategic data asset would be a big loss (since most research exists on tumorigenic cells).  Recently learning cell-cycle foundations with you, makes me realize that a cancer cell or stem cell or normal cell is conceptually still a 'cell'.  Keeping maybe cell cycle phases or the regulatory CDK and Telomerases etc as key variables. 

 

Key question being that what specifically can be "reasonably" ascertained from these studies vs "ball-park" ascertainment vs it would be just foolishly wrong to infer anything. I realize this is maverick topic, that MANY would not be open-minded to share with others (they would rather hoard it ...as they say, tricks of the trade) but I have faith that if anyone here can selflessly share knowledge, it is you. 

 

There are no hard and fast rules, I'm afraid. If you come from a technical background like me, but in an unrelated field, the main barrier is in understanding the terminology and what they are doing with the various bench techniques. Google and just reading more papers will solve this issue.

 

Probably the two main things to bear in mind are that many/most 'aging' papers are not relevant to us because of various shortcomings in design, and that a sizeable minority (?) of papers probably won't replicate.  On the latter point, I knew this was an issue, particularly with the proliferation of Chinese authored papers, but I didn't know how bad it was until I had an email exchange with a scientist and he told me of a major paper in the telomere field that no one else can replicate. But to my knowledge no one has repudiated the paper - academia is based more on reputation than results, I'm afraid.

 

That is why a forum dedicated to research and self-experimentation like Longecity is so important. If you try something and it makes you healthier, then it probably will extend your life as well. If you try something and you feel worse, guess what...  


  • Good Point x 1

#808 johnhemming

  • Member
  • 311 posts
  • 122
  • Location:Birmingham, UK

Posted 05 October 2022 - 10:10 AM

That is why a forum dedicated to research and self-experimentation like Longecity is so important. If you try something and it makes you healthier, then it probably will extend your life as well. If you try something and you feel worse, guess what...  

 

One functional test that interests me is repair to insult.  I do weekly blood tests which means a venipuncture each week.  I have been monitoring how quickly and how well it repairs depending upon inputs.  I think this is a useful test as one aspect of aging deterioration is slowness of repair of injury.


  • unsure x 1
  • Good Point x 1
  • Informative x 1

#809 ambivalent

  • Guest
  • 745 posts
  • 167
  • Location:uk
  • NO

Posted 05 October 2022 - 11:27 AM

 I would guess that anything that reduces energy supply (ATP) increases lifespan, and this is where the benefits of keto and fasting come from. 

 

Could you elaborate a but further Quest, is it not likely the signalling from reduced ATP than the ATP itself which is providing that benefit - fasting mimentics aren't changing ATP?

 

Also we have NAD+ supplementation which only marginally extends life, but does increase ATP - which was rather surprising given its cellular rejuvenating effects. 

 

So might it be that maintaining high levels of ATP accelerate some aspect of aging - say shorten telomere, deplete stem cell reserves?

 

I note here that ATP increases stem cell pluriopotency:

 

https://pubmed.ncbi....h.gov/29222327/

 

Does this presumably mean they are more likely to divide and so eventually reserves deplete more rapidly as high levels of ATP are maintained? 

 

The NAD lifespan studies, and I forget which precursor was used, wasn't exactly the candle burning twice as bright for half as long but does appear to lack an explanation as to how such a regenerative intervention doesn't significantly extend lifespan, but does healthspan.  


Edited by ambivalent, 05 October 2022 - 12:00 PM.

  • Informative x 1

#810 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 05 October 2022 - 12:03 PM

Could you elaborate a but further Quest. Is it not likely the signalling from reduced ATP than the ATP itself which is providing that benefit - fasting mimentics aren't changing ATP.

 

Also we have NAD+ supplementation which only marginally extends life, but does increase ATP - which was rather surprising given its cellular rejuvenating effects. 

 

So might it be that maintaining high levels of ATP accelerate some aspect of aging - say shorten telomere, deplete stem cell reserves?

 

I note here that ATP increases stem cell pluriopotency:

 

https://pubmed.ncbi....h.gov/29222327/

 

Does this presumably mean they are more likely to divide and so eventually reserves deplete more rapidly as high levels of ATP are maintained? 

 

The NAD lifespan studies, and I forget which precursor was used, wasn't exactly the candle burning twice as bright for half as long but does appear to lack an explanation as to how such a regenerative intervention doesn't significantly extend lifespan, but does healthspan.  

 

It is just an observation that successful lifespan studies (all?) impair (directly or indirectly) the cellular energy pathways. 

 

Even the hyperlong telomere, longer lived mice made by Blasco et al. had improved metabolic function (1).

 

It is a paradox (and we should always be interested in paradoxes) that we lose energy as we age, but restricting energy input seems to delay this process. Blagosklonny has come closest to explaining this with hyperfunction theory - growing faster means you die faster (2). But putting mTOR at the head of this river seems a mistake to me, it is more likely to be energy. Physics trumps biology.

 

The following is pure speculation on my part, but I believe that cells don't necessarily want to remain in this 'society' we call a body, and easily revert to individualistic tendencies. This seems to always involve bypassing oxidative  phosphorylation (mitochondria) and splitting glucose directly in the cytoplasm. This is what cells did originally and it was more than enough energy for them, just not enough to build a body with. Maintaining mitochondria seems to be the way that you keep cells co-operative. 

 

NAD+/NADH is just one ratio involved in this process and it doesn't appear to be a very important one.

 

Telomeres, by the way, eventually sabotage even cells where the mitochondria have been kept small and pristine by massive doses of nicotinamide, and this self-destruct mechanism is sent to mitochondria (3).

 

(1) Muñoz-Lorente, M.A., Cano-Martin, A.C. & Blasco, M.A. Mice with hyper-long telomeres show less metabolic aging and longer lifespans. Nat Commun 10, 4723 (2019). https://doi.org/10.1...467-019-12664-x

(2) Blagosklonny MV. Aging and immortality: quasi-programmed senescence and its pharmacologic inhibition. Cell Cycle. 2006 Sep;5(18):2087-102. doi: 10.4161/cc.5.18.3288. Epub 2006 Sep 15. PMID: 17012837.

(3) Kang HT, Lee HI, Hwang ES. Nicotinamide extends replicative lifespan of human cells. Aging Cell. 2006 Oct;5(5):423-36. doi: 10.1111/j.1474-9726.2006.00234.x. Epub 2006 Aug 25. PMID: 16939485.


  • Informative x 2





Also tagged with one or more of these keywords: telomeres, nad, nampt, ampk, resveratrol, allicin, methylene blue, nmn, sirtuins, statin

0 user(s) are reading this topic

0 members, 0 guests, 0 anonymous users