• 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

#361 Turnbuckle

  • Location:USA
  • NO

Posted 29 October 2020 - 10:51 AM

Here's a much simpler explanation. It is the stochastic damage of metabolism that makes epigenetic clocks tick. Old cells (without ready replacements) acquire dysfunctional mitochondria through stochastic metabolic processes and the passage of time The cell then adapts to a bad situation by making the best of it and down and upregulating various genes in the nucleus. Then (naive) scientists come along and decide the problem (and aging itself) is those adaptations.

Consider this the null hypothesis. Disprove it if you can.

 

There is crosstalk between mitochondria and the epigenome, and low levels of ATP will cause epigenetic modifications that are partially reversible. However, that is surely a minor effect in the overall landscape of steadily increasing epimutations that underlie aging. 

 

 

Mitochondria to Nucleus Retrograde Response

 

Mitochondrial dysfunctions invoke mitochondria-to-nucleus retrograde responses in human cells. Perturbed DNA methylation profiles of certain loci within the human nuclear genome have recently been associated with depleted mitochondrial DNA. Smiraglia et al. developed and studied cells that were void of functional mitochondria (rho0 cells), enabling them to gain an insight into the possible role mitochondria might have in regulating or being associated with epigenetic alterations of the nuclear genome in a gene specific or genome-wide manner, particularly at the level of DNA methylation. Depletion of the mitochondrial genome in both chemically generated MCF12A rho0 or genetically modified 143B rho0 cell lines resulted in the aberrant methylation of promoter CpG islands (high CG-rich regions)84 that were previously unmethylated ... Repletion of wild-type mitochondria back into these 143B rho0 (mtDNA deficient cells) resulted in the partial re-establishment of some methylation profiles back to their original parental state.

https://www.ncbi.nlm...les/PMC3368816/

 



#362 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 29 October 2020 - 04:59 PM

There is crosstalk between mitochondria and the epigenome, and low levels of ATP will cause epigenetic modifications that are partially reversible. However, that is surely a minor effect in the overall landscape of steadily increasing epimutations that underlie aging.


The point is that this process could be driven by metabolism and hence be fundamentally stochastic, even if the downstream consequences are not at all random. It may not even be due to dysfunctional mitochondria. Maybe even normal mitochondria drive this process in long lived cell lines.

What I'd like to see is a study looking at gene expression in cell lines that are rejuvenated from replicative senescence by restoring telomeres. What gene expression reverts to that of younger cells and is there any gene expression that doesn't revert due to errant methylation? I don't think the Horvath clock is adequate for this purpose for reasons already discussed. Also his immortalisation of cells was done in a way I'd expect to affect gene expression independent of telomere elongation.

#363 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 30 October 2020 - 11:02 AM

I don't think so.

 

It is said neural metabolic rate is similar between various species of mammal, presumably so within bowhead whales too.   The brain is 2% of body yet consumes 20% of oxygen, iirc.   10x energy of most other organs.  Most of the energy goes towards information transmission function of neurons.   If energy production was significantly compromised the neuron would stop functioning.   Yet not only have unmodified mice neurons managed to live twice as long when transplanted to rats, and human neurons have lasted over 115 years in humans, but bowhead neurons seem to last for over 200 years.   AFAIK bowhead neurons do not seem to have additional protection mechanisms, though maybe they do.

 

 

 

The brain is a very expensive organ, for which humans pay a heavy price in terms of childbirth deaths. The brain also takes all the available glucose in times of starvation. And yet for humans it is the primary reason, through intelligence, that we escaped predation and evolved a lifespan longer than that of other mammals. But we have not reached negligible senescence. Obviously high losses in the population do regularly occur in our recent history, necessitating genetically mixing every 20 years rather than letting individuals continue indefinitely. Hopefully you are right and it will not be too difficult a task to get humans into a non aging state.

 

Your comments on murine neurons lasting longer in rats is interesting. I once read a paper showing human DNA acquired epimutations at a murine rate when the (human) nucleus was transferred into a mouse cell. But I now can't find the paper anywhere. If true this would link dysregulation of methylation to metabolism and mitochondria.


  • Needs references x 1

sponsored ad

  • Advert

#364 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 30 October 2020 - 11:39 AM

I got back the results of my June TruMe test.

 

Chronological Age: 41.6

Biological Age 41.9

Intervention: 3 1/2 months of AKG. I also did a few rounds of TB's Stem Cell protocol (with no senolytics).

This is disappointing as it means there has been no change since my March 2020 methylation age test result from Zymo (42 yo).

Possible explanations:

TruMe uses a saliva test, which is less reliable than the blood or urine results from Zymo, and might have been affected by my bad hayfever during May and June.

I also took a Vit D and K2 supplment as a precaution against all the calcium in the AKG supplement. Vit D is a telomerase activator.

In my case having reversed my telomere age by 3 years recently through epitalon (and increased my methylation age by the same amount), I might need significantly longer to reverse my methylation age.

 

Future plans: i have another TruMe test ready to go. My hayfever has relented and i've now replaced the VitD/K2 with just K2. I'll continue the AKG with occasional TB Stem Cell cycles (note I'm omitting any senolytics at this point) and report back when I done the next test (Sept or Oct).

 

I finally got the results back for my Sept TruMe test.

 

Chronological Age: 41.8

Biological Age:  36.7

 

There has been an improvement. It is not clear whether this is simply because I just took AKG for longer, or because of anything else. After considering adding berberine, ALA and carnitine at the half way stage, I decided not to give my body any help with fatty acid processing and instead just fed it more long chain fatty acids. I did this by eating a lot of cheddar and butter. The butter I clarified by gently heating it, and drained off the fat leaving the milk proteins in the pan. I then added stearic acid to the butter fat to make it more saturated. I used this to butter bread, butter pasta, cook pancakes, roast potatoes, bake cookies, etc. I found it quite palatable and it had the advantage of allowing me to eat copious amounts of carbs without putting on weight. Perhaps this did help my stem cell populations as I discussed in post 275#

 

 

We know AKG results in a reduction of triglycerides, LDL cholesterol, presumably because of increased demand on the Krebs cycle resulting in a requirement for beta oxidation (https://www.longevit...sults-in-humans). Other supplements like resveratrol that activate SIRT1 also increase beta oxidation. AMPK activation increases fat burning. So does carnitine, which increases import of fats into the mitochondria (https://pubmed.ncbi....h.gov/12404185/)

Alpha lipoic acid also seems to have some benefit as it's also important in the Krebs pathway and in AKG oxidation. Forskolin increases cAMP, which increases fat burning (https://www.ncbi.nlm...one.0029735.pdf) and incidentally also leads to symmetrical division at least in egg cells.

Stearic acid is a trigger fat that signals the body to burn fat (https://www.nature.c...467-018-05614-6). This might be the best way of all - eating highly saturated fat and not relying on any over stimulated supplement pathways.

It turns out that children do more beta oxidation than adults (https://nutritionj.b.../1475-2891-6-19). Elderly patients were able to increase beta oxidation by supplementing glutathione precursors (https://onlinelibrar...1111/acel.12073). Cells burning fats produce more ROS and use fused rather than fissioned mitochondria. Note this is also a signal for cells to become insulin resistant, which helps with weight loss.

I'm not posting all references here - there are many other interesting avenues for research and self experimentation (another time i'll post about using AKG, berberine and ALA). But I want to focus this post in a specific direction - stem cells.

Beta oxidation is very important in maintaining stemness. I found it easy to find supporting evidence in pluripotent stem cells (:text=Recent%20reports%20suggest%20that%20the,cells%20%5B9%2C%2032%5D.&text=Our%20study%20showed%20that%20Cpt1,for%20promoting%20the%20reprogramming%20process.' class='bbc_url' title='External link' rel='nofollow external'>https://stemcellres....amming process.)
Quote

‘Recent reports suggest that the beta-oxidation of fatty acids plays an important role in the maintenance and growth of pluripotent stem cells

Also check out this excellent paper on self renewal in intestinal stem cells (https://www.scienced...934590918301632)
Quote

Diet has a profound effect on tissue regeneration in diverse organisms, and low caloric states such as intermittent fasting have beneficial effects on organismal health and age-associated loss of tissue function. The role of adult stem and progenitor cells in responding to short-term fasting and whether such responses improve regeneration are not well studied. Here we show that a 24 hr fast augments intestinal stem cell (ISC) function in young and aged mice by inducing a fatty acid oxidation (FAO) program and that pharmacological activation of this program mimics many effects of fasting. Acute genetic disruption of Cpt1a, the rate-limiting enzyme in FAO, abrogates ISC-enhancing effects of fasting, but long-term Cpt1a deletion decreases ISC numbers and function, implicating a role for FAO in ISC maintenance. These findings highlight a role for FAO in mediating pro-regenerative effects of fasting in intestinal biology, and they may represent a viable strategy for enhancing intestinal regeneration

Neural stem cells (https://www.ncbi.nlm...les/PMC5583518/)
Quote

Quiescent NSPCs show high levels of carnitine palmitoyltransferase 1a (Cpt1a)-dependent FAO, which is downregulated in proliferating NSPCs.

and hematopoietic stem cells (https://pubmed.ncbi....h.gov/22902876/)
Quote

inhibition of mitochondrial FAO induces loss of HSC maintenance, whereas treatment with PPAR-δ agonists improved HSC maintenance.

As a result I've decided to change my diet to incorporate much more saturated fat with an emphasis on stearic acid.

It will be interesting to see if it has any effect on my epigenetic age. I have already experienced a loss of fat and gain in muscle whilst eating as many carbs as I want. There are also some potential downsides to this diet, which I will cover in a future post.

 

I experienced increasing fatigue from the 4 1/2 month point. I also retrospectively noticed a considerable reduction in weight lifting strength between months 4 and 6. Again, I do not know if this was due to the AKG or the increased saturated fat intake. I quit AKG when I submitted my second test at the 6 month point. Since then I have continued eating the fatty diet. I have also upped my protein intake and my strength has started to recover. I have experienced fatigue at times from the diet that is ameliorated by carnitine and milk thistle. 

 

It would be helpful to do further testing but TruMe testing has slowed to a crawl due to the lockdown.

 

My original plan was to add back telomerase activators in addition to an effective epigenetic age reversal strategy to see if I could have the best of both worlds. Again that will depend on TruMe being able to accept and process samples relatively quickly. 


  • Informative x 4
  • like x 1

#365 dlewis1453

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

Posted 30 October 2020 - 01:51 PM

I finally got the results back for my Sept TruMe test.

 

Chronological Age: 41.8

Biological Age:  36.7

 

 

 

Wow this is a very interesting and impressive result! Thanks for sharing! Hopefully you can maintain and continue to build on this age differential. 



#366 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 30 October 2020 - 02:42 PM

From the article:

Loss of Dnmt3a Immortalizes Hematopoietic Stem Cells In Vivo

Here we show that loss of Dnmt3a endows HSCs with immortality in vivo. The self-renewal potential of Dnmt3aKO HSCs far exceeds that of normal HSCs and the lifespan of the mice from which they were derived. Our data establish that HSCs do not have an inherently finite lifespan but that loss of Dnmt3a augments epigenetic features that enforce self-renewal and enable HSCs to be propagated indefinitely. Further examination of the mechanisms perpetuating immortality in Dnmt3aKO HSCs may provide a window for artificially extending the lifespan ofHSCs, an important biomedical application in the context of the aging human population.

 

Very intriguing article Iporuru.

 

It is a counter intuitive argument that it is the self renewal of stem cells rather than their depletion that leads to deterioration.

 

At first glance it appears to support my idea of the 'selfish cell' surviving longer, see post #130 (https://www.longecity.org/forum/topic/102169-alternative-methods-to-extend-telomeres/page-5#entry880339). 

 

 

 




The selfish cell lives longer



Arguments for the evolution of aging depend on a non aging (selfish) individual being bad for the group.



I think I see a parallel in the cells that survive in the aged body. Whenever I look into an aspect of aging, I always see the same thing. The cells that survive are the ones that are the worst for the body. Everyone seems to assume that a mysterious process called aging is causing these cells to fail with age. But what if it is a selection process that leaves the most selfish cells alive?


 

 

 

It is a great pity that bone marrow stem cell dynamics have not been better studied. It is still not understood how stem cells retain the original DNA strand, whilst letting the cell with both new DNA strands differentiate. (See: https://en.wikipedia.org/wiki/Immortal_DNA_strand_hypothesis). For me this is absolutely central to understanding what is going wrong in aging and whether telomeres and telomerase are central or not to the process.



#367 JamesPaul

  • Guest
  • 85 posts
  • 39
  • Location:Florida
  • NO

Posted 30 October 2020 - 03:56 PM

"Recent reports suggest that the beta-oxidation of fatty acids plays an important role in the maintenance and growth of pluripotent stem cells"

 

I cannot resist quoting a paper about fatty acid β-oxidation and an article about JMJD3 histone demethylation and possible reversal of aging:

https://www.jci.org/...cles/view/97736,

"Fasting-induced JMJD3 histone demethylase epigenetically activates mitochondrial fatty acid β-oxidation"

 

There is a connection to an article that Vince Giuliano and Steve Buss recently posted on their site:

 

https://www.anti-agi...ersal-strategy/

 

"Initiation of adult aging around 24 years of age appears to be an evolutionarily conserved process triggered by histone methylation by the double and triple histone methylases H3K27me2/3. That is, di- and trimethyl-lysine 27 on histone H3.  This results in silencing of numerous genes, including many anti-inflammatory and anti-aging genes that are largely active in youth.  The results of this methylation is down-regulation or inactivation of protective genes related to this particular histone position.  These include genes that encode for protective heat shock proteins.  This may be the essence of adult aging as we know it.  This evolutionarily-conserved shift has been well-studied in nematodes where it all happens in only 4 hours...There are many seemingly practical approaches to demethylating H3K27me2/3 and thus to initiate the YOUNGING.  The main substance that specifically demethylates H3K27me2/3 is JMJD3.  The Jumonji domain-containing protein D3 (JMJD3), specifically demethylates di- and trimethyl-lysine 27 on histone H3 (H3K27me2/3).  So we can ask 'what substances can we practically use to activate JMJD3 and what other substances can be used to demethylate H3K27me2/3?'  It turns out the list of familiar substances that can do that is quite long, including:

  • Certain familiar herbal substances, including Curcumin, and High AkBA Boswellia
  • DHEA
  • Alpha keto-glutarate
  • Vitamin D"

 

I fixed two typos  in the quote.

I know this is off-topic relative to the subject of the thread, but it is related to recent posts.

 


  • Informative x 1

#368 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 30 October 2020 - 05:33 PM

I cannot resist quoting a paper about fatty acid β-oxidation and an article about JMJD3 histone demethylation and possible reversal of aging:
https://www.jci.org/...cles/view/97736,
"Fasting-induced JMJD3 histone demethylase epigenetically activates mitochondrial fatty acid β-oxidation"

It seems clear that fatty acid oxidation - either eating fats or burning fats stored from previous carb or protein consumption - activates some sort of stem cell renewal. This makes sense from an evolutionary point of view as clearly during starvation one needs to reduce outgoings on differentiating cells whilst marshalling stem cell reserves. The exact mechanism is not clear yet however, other than involving the repressive histone lysine position you mention.

Here's another relevant paper, but in planaria, where starvation causes the worm to 'eat itself" reducing in size, whilst increasing telomere length in its pluripotent stem cells.

https://www.jci.org/...cles/view/97736

Clearly planaria are different to people but maybe some of the same mechanisms apply in terms of rejuvenation of stem cell niches.

It's still not clear to me to what extent human stem cell niches suffer from telomere loss however.

Attached Thumbnails

  • 1-s2.0-S2213671119302310-fx1.jpg

Edited by QuestforLife, 30 October 2020 - 05:38 PM.

  • Informative x 2

#369 Castiel

  • Guest
  • 374 posts
  • 86
  • Location:USA

Posted 31 October 2020 - 01:24 AM

It seems clear that fatty acid oxidation - either eating fats or burning fats stored from previous carb or protein consumption - activates some sort of stem cell renewal. This makes sense from an evolutionary point of view as clearly during starvation one needs to reduce outgoings on differentiating cells whilst marshalling stem cell reserves. The exact mechanism is not clear yet however, other than involving the repressive histone lysine position you mention.

Here's another relevant paper, but in planaria, where starvation causes the worm to 'eat itself" reducing in size, whilst increasing telomere length in its pluripotent stem cells.

https://www.jci.org/...cles/view/97736

Clearly planaria are different to people but maybe some of the same mechanisms apply in terms of rejuvenation of stem cell niches.

It's still not clear to me to what extent human stem cell niches suffer from telomere loss however.

 

Might be telomere loss or might be other causes.   But the centenarian blood cell mutation paper suggested she only had two cell lines remaining producing her blood.    So presumably the other stem cell lines were depleted.

 

https://www.scienced...40423132608.htm



#370 Iporuru

  • Guest
  • 216 posts
  • 221
  • Location:Europe

Posted 31 October 2020 - 12:24 PM

It's still not clear to me to what extent human stem cell niches suffer from telomere loss however.

 

Do these two papers (at least partly) answer your question? Haven't read them, just skimmed
 


  • like x 1

#371 Iporuru

  • Guest
  • 216 posts
  • 221
  • Location:Europe

Posted 01 November 2020 - 11:55 AM

Vittorio Sebastiano at Undoing Aging 2019

 

starting at about 4:45 mark:

"We have proven this at the methylation clock level (…) we have shown that all the physiological markers of aging, and we have seen that for those markers that go down with aging they actually increase with our treatments; the ones that go up with aging they decrease with our treatments. The only thing that doesn’t change is telomere length. Now, many people look at his as a very bad result and actually I’m very happy about this result because remember, the claim is that we’re not changing cell identity here; if we were to see telomere elongation that would speak for third activation which means change in cell identity and potential tumorigenesis."


Edited by Iporuru, 01 November 2020 - 11:56 AM.

  • Informative x 1

#372 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 01 November 2020 - 02:04 PM

Vittorio Sebastiano at Undoing Aging 2019

starting at about 4:45 mark:
" The only thing that doesn’t change is telomere length. Now, many people look at his as a very bad result and actually I’m very happy about this result because remember, the claim is that we’re not changing cell identity here; if we were to see telomere elongation that would speak for third activation which means change in cell identity and potential tumorigenesis."


The point of TurnBio (and Yuri's Youtherium) is to de-differentiate cells enough to improve their biomarkers WITHOUT going back to a pluripotent stem cell, which in vitro is partly distinguished by high telomerase activity (I.e. can be cultured indefinitely) and in Vivo can form a terratoma. This is in contrast to adult stem cells and somatic cells that can't proliferate indefinitely in vitro and can't form a terratoma in vivo.

Don't take his statement to be a commentary of the efficacy or otherwise of telomerase as a treatment for aging.

It is my view that any treatment for aging MUST involve telomerase in some way. For example Turnbuckle's ideas revolve around expanding a primitive stem cell population that have active telomerase (i.e. VSELs).
  • Informative x 1
  • like x 1

#373 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 05 November 2020 - 12:40 PM

Do these two papers (at least partly) answer your question? Haven't read them, just skimmed
 

 

I have read the first of your references [1] and skimmed the other [2], as well as reading a further paper [3] that references these studies. I wanted to talk about why whether the bone marrow suffers from shortening telomeres or not is an absolutely critical issue.

I can certainly think of ways that BMSCs could limit their loss of telomeres, even without telomerase, so it is interesting to read the arguments in [1]-[3] for why loss of telomeres in the bone marrow is thought to occur. There is also some indirect evidence this is happening that is mentioned in the papers, despite the primary focus being on simulations. I have also previously mentioned a paper [4], where they sacrificed mice and actually directly measured their BMSC telomeres using FISH, in an effort to validate an anti-oxidant treatment. In that study they did find a severe shortening of telomeres in the BM with age.

Papers [1] and [2] posit that mammals (mice, cats, baboons and humans) have the same number of HSCs, but they divide at a different rate in each species, depending on the demands of the soma, i.e. the rate of division (and loss) of somatic cells. This also touches on an important point about active telomerase in mice cells as opposed to inactive telomerase in human cells: as mentioned in [3] with respect to cancer cells, sometimes active telomerase does not lead to longer telomeres, but shorter ones – if the cells use telomerase for greater proliferation. This may be the purpose of telomerase in mice cells - not to elongate telomeres but to enable faster division. This is also what happens in leukocytes in humans, when an infectious challenge requires rapid division that cannot wait for reinforcements from the bone marrow.

Whether mouse or man a certain number of red and white blood cells, as well as skin, endothelial and fat cells (for example), all need to be replaced at a certain rate to keep the particular tissue functioning. If BMSCs lose telomeres with age, the tissues of the body will either have to reduce their cellular replacement rate (i.e. slow skin turnover), or in the case of white or red blood cells when cell numbers must be maintained, shorter telomeres in each cell will cause non-optimal gene expression (i.e. immune senescence) in those tissues.

This is where the critical issue comes in. It is perfectly possible by various means to increase stem cell release from the BM – see AFA [5], Sea Buckthorn Extract [6], a blend [7] – but if BMSCs can be exhausted, does this extra mobilisation reduce lifespan? Does the body permit this?
If as [1] claims, there are 11,000 (Hematopoietic) stem cells in the bone marrow, and each one divides about once/year in humans, this equates to 30 BMSCs dividing/day or about 1/hour. AFA is claimed in [5] to raise stem cell release by ~20% for a couple of hours. Therefore instead of 2 BMSCs undergoing asymmetric division and supplying the body in a 2 hour window, AFA might cause 3 BM cells to do this. This could then result in many extra downstream cells being available to the body. And indeed AFA seems to make my skin look better, my hair to grow faster, my cut and bruises to heal more quickly. But quite possibly, this boost could deplete my BM sooner! A 20% increase in BM mobility for 2 hours equates to a 1.7% increase (or expenditure) per day. Extrapolate this out to every day for an 80 year lifespan results in losing almost a year and a half of life. It is certainly something to think about.

[1] https://pubmed.ncbi....h.gov/15539081/
[2] https://www.research...nhuman_primates
[3] https://www.nature.c...rticles/2404339
[4] https://www.mdpi.com/2076-3921/9/2/144
[5] https://pubmed.ncbi....h.gov/17765649/
[6] https://pubmed.ncbi....h.gov/30787601/
[7] https://translationa.../1479-5876-8-34


  • WellResearched x 3
  • Informative x 1

#374 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 05 November 2020 - 04:53 PM

https://www.embopres...embj.2019103420

Re‐equilibration of imbalanced NAD metabolism ameliorates the impact of telomere dysfunction

Our findings indicate that critically short telomeres and telomere dysfunction leads to loss of NAD+ homeostasis and establish a new linkage between dysfunctional telomeres and mitochondria that contributes to the deleterious consequences of telomere dysfunction

Attached Thumbnails

  • image-2-768x544.png


#375 JamesPaul

  • Guest
  • 85 posts
  • 39
  • Location:Florida
  • NO

Posted 06 November 2020 - 03:15 PM

https://www.pensumme...cell-treatments

 

states “V-cells (VSEL cells) seem to turn on telomerase in other cells.”

 

The same things is stated here:

https://www.pensumme...ells-or-v-cells

 

which states "Another very interesting and important aspect about these cells [VSELs] is that they trigger the secretion of Telomerase, the enzyme that restores the DNA telomere ends...Trials have shown that V-Cells properly extracted, concentrated, activated and enriched with the correct growth factors have yielded cells with increased telomere length, measured on white blood cell [WBC] leukocytes for up to six months after treatment. This is a repeatable procedure."

 

The results experienced by this clinic's patients may not be as remarkable as the results of those who have done Turnbuckle's stem cell renewal procedure, though.  There is not sufficient data on that site to tell.  That site reports some good results for some serious conditions, though.

 

On that page, further text becomes visible if one clicks the link “Click further reading”.  Another excerpt is as follows:

There is a 25-30% increase in the number of circulating V-Cells following intense physical stress such as 1hr. of running or following the ingestion of a nutraceutical (1 hr. post-ingestion). This is a transient effect in that:

  • Cell numbers returns to the baseline 2-3 hrs. after exercising.

  • Cell numbers returns to the baseline 4-6 hrs. after ingestion of a nutraceutical.”


Edited by JamesPaul, 06 November 2020 - 03:25 PM.


#376 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 06 November 2020 - 05:17 PM

“V-cells (VSEL cells) seem to turn on telomerase in other cells.”

The same things is stated here:

"Another very interesting and important aspect about these cells [VSELs] is that they trigger the secretion of Telomerase, the enzyme that restores the DNA telomere ends...Trials have shown that V-Cells properly extracted, concentrated, activated and enriched with the correct growth factors have yielded cells with increased telomere length, measured on white blood cell [WBC] leukocytes for up to six months after treatment. This is a repeatable procedure."

This website does not seem to reference their claims, so I assumethey are based on their own in clinic work.

I can believe VSELs can increase telomere length in other cells - this could be achieved via two mechanisms:
1. Embryonic cells are often used as 'feeder cells' in vitro, with their telomerase benefiting the cells being cultured. Something similar may be happening in Vivo.
2. More likely however, the VSELs may be differentiating and replacing old somatic cells with fresh cells with longer telomeres.

TB claims this is what is happening in his protocol, though he's never measured telomeres. I couldn't comment on how effective the company's treatment is compared to TB's protocol, I've never noticed any benefit from the latter.

Edited by QuestforLife, 06 November 2020 - 05:23 PM.

  • Informative x 1

#377 Iporuru

  • Guest
  • 216 posts
  • 221
  • Location:Europe

Posted 09 November 2020 - 09:58 AM

A new paper by Tony Wyss-Coray:

Molecular hallmarks of heterochronic parabiosis at single cell resolution

"We performed single-cell RNA-sequencing on 13 organs to reveal cell type specific responses to young or aged blood in heterochronic parabiosis. Adipose mesenchymal stromal cells, hematopoietic stem cells, hepatocytes, and endothelial cells from multiple tissues appear especially responsive. On the pathway level, young blood invokes novel gene sets in addition to reversing established ageing patterns, with the global rescue of genes encoding electron transport chain subunits pinpointing a prominent role of mitochondrial function in parabiosis-mediated rejuvenation. Intriguingly, we observed an almost universal loss of gene expression with age that is largely mimicked by parabiosis: aged blood reduces global gene expression, and young blood restores it. Altogether, these data lay the groundwork for a systemic understanding of the interplay between blood-borne factors and cellular integrity."


  • Informative x 3
  • Off-Topic x 1

#378 granth998

  • Guest
  • 18 posts
  • 9
  • Location:Dallas, TX
  • NO

Posted 09 November 2020 - 10:07 PM

I've been using Teloyears for the past 3 years to check my telomere length.  They are becoming very slow in getting results.  Wondering if there are other more accurate companies that do this type of testing?



#379 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 10 November 2020 - 01:10 PM

I've been using Teloyears for the past 3 years to check my telomere length.  They are becoming very slow in getting results.  Wondering if there are other more accurate companies that do this type of testing?

 

I've never been able to use Teloyears because of being based outside the US. I expect they've slowed down because of the lock-down and supplying covid-19 tests as a priority.

 

Lifelength based in Spain are still up and running. But you need to find a participating clinic as it requires a venous blood draw. It's also considerably more expensive than Teloyears. 



#380 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 17 November 2020 - 09:04 AM

A new paper by Tony Wyss-Coray:

Molecular hallmarks of heterochronic parabiosis at single cell resolution

"We performed single-cell RNA-sequencing on 13 organs to reveal cell type specific responses to young or aged blood in heterochronic parabiosis. Adipose mesenchymal stromal cells, hematopoietic stem cells, hepatocytes, and endothelial cells from multiple tissues appear especially responsive. On the pathway level, young blood invokes novel gene sets in addition to reversing established ageing patterns, with the global rescue of genes encoding electron transport chain subunits pinpointing a prominent role of mitochondrial function in parabiosis-mediated rejuvenation. Intriguingly, we observed an almost universal loss of gene expression with age that is largely mimicked by parabiosis: aged blood reduces global gene expression, and young blood restores it. Altogether, these data lay the groundwork for a systemic understanding of the interplay between blood-borne factors and cellular integrity."

 

Comments on Hallmarks of heterochronic parabiosis

 

Aging (or old blood) affects the gene expression levels of different cells differently (they have different ‘Differentially Expressed Genes’ (DAG)), but there is a lot of agreement between the cell-specific DEGs for both aging and old blood. For some cells, the agreement is very high indeed (~0.6-0.84 for Endothelial Cells, Hematopoietic Stem Cells, Mesenchymal Stem Cells in Adipose Tissue) suggesting that these cells in particular primarily age extrinsically (i.e. via factors in the blood).

Note this does not mean other cells do not age intrinsically, of course.

 

Later on in the paper they talk about how some cells age in co-ordination with the tissue in which they reside, whereas other cell types ignore the tissue they’re in and instead age more in step with the tissue they come from (i.e. some bone marrow cells).

By contrast to the cell-type specific changes with age (or old blood), the rejuvenation provided by young blood seems to act on different cells in mostly the same way, via the mitochondrial pathways of the electron transport chain*.

 

*Weird exception that may prove the rule: brain endothelial cells (BECs) have been observed to undergo increased expression of electron transport chain genes with age. This effect is replicated by exposure to aged mouse plasma and reversed by exposure to young mouse plasma in vivo (ref: https://www.cell.com...81?showall=true Brain Endothelial Cells Are Exquisite Sensors of Age-Related Circulatory Cues).
My note: this suggests the solution to the general downregulation of the electron transport chain is not merely ETC upregulation, as this would be counterproductive to BECs, but instead a single or multiple corrections in the blood that either up- or down- regulates ETC function towards a youthful state as required through some indirect mechanism.

 

Some further thoughts on the problem: given we know the alteration of ETC function is via extrinsic factors in the blood, it would probably be a mistake to think mitochondria can be fixed via some mitochondrial specific molecule such as ALA or Q10. It makes more sense to assume there is some sort of problem in circulation such that mitochondria are not getting enough of something they need or are getting too much or something they do not need. So for example increasing circulation might give the mitochondria more of whatever it is they need that is present in the blood, or breaking down some sort of blood borne toxin might be the key to avoiding overloading the mitochondria with that toxic compound. This marks a significant change in the approach to reversing aging in that we are treating the blood and the circulation first and the cell second. It bears some similarity to the approach of Janic et al. in their short term statin-sartan treatment for pre-clinical atherosclerosis, discussed at length in this thread (for those that haven’t read it already it is one of the keystones of this thread: https://pubmed.ncbi.....gov/26214555/) .

 

One other thing that occurs to me from the paper when the Conboys replaced half the blood plasma with saline plus albumin and produced significant rejuvenation (https://www.ncbi.nlm...les/PMC7288913/), is that we mustn’t dismiss the importance of those blood proteins. For example, albumin, quite apart from its liver functions is an antioxidant buffer in the blood (https://pubmed.ncbi.nlm.nih.gov/21113488/), which should help mitochondria, particularly when in the procedure you are replacing partly oxidised with fully reduced albumin. Fibrinogen is an acknowledged 'damage associated molecular pattern protein' (DAMP) (https://journals.lww.com/shockjournal/Fulltext/2016/10000/Role_of_Elevated_Fibrinogen_in_Burn_Induced.7.aspx) and could also impede rejuvenation via decreased blood flow when it causes blockages in capillaries (that are not severe enough for a stroke).  Transferrin moves iron, of which the main intracellular recipient is mitochondria, and intracellular transferrin is known to increase in Parkinson’s Disease (https://www.sciencedirect.com/science/article/pii/S1357272519300433), which also implicates it in mitochondrial dysfunction. Finally, immunoglobulins – the immune system is known to react to pieces of mitochondrial DNA in the blood (https://pubmed.ncbi.nlm.nih.gov/29125070/), leading to the rise in age related ‘sterile’ inflammation. At the very least these arguments suggest blood protein removal should be eliminated as a cause of aging in these parabiosis studies.

 

So where does this leave telomeres as a valid point of intervention in the aging process? This paper is particularly interesting as it shows how complex the age-related changes in genes are. Different genes are up or down regulated depending on tissue and cell-type. Much like the real situation with telomeres. At any given point there are cells with nice long telomeres and cells with short telomeres, sometimes living right alongside each other. Some tissues seem to maintain telomeres better than others. Even in a very old person there are cells with long telomeres that you can extract and passage in the lab. Sometimes a cell has long telomeres but is rendered functionally arrested because of mitochondrial damage. All these arguments have been used to dismiss telomeres as an important cause of aging. But the real picture is much more complex.

 

Now that we can see the body has a kind of aging imposed on much of it by systemic factors, it is possible to imagine that the machine just needs an oil change to make it as good as new. That would be wonderful. And indeed, this would not contradict the telomere story. How many times have I posted about ‘conditional reprogramming’ whereby old somatic cells with the right culture conditions would revert to a progenitor state and proliferate endlessly so long as the culture conditions were maintained? (for recap: https://linkinghub.e...2944013005944).Note that these culture conditions included telomerase. Perhaps young blood does something similar? Indeed, it is a central tenet of this thread that the statin-sartan treatment of Janic et al is doing just that. This is a throwback to the pre-Hayflick days when scientists though cells were immortal; it was just the body that aged. Perhaps they were not so wrong after all. With the right culture conditions.


Edited by QuestforLife, 17 November 2020 - 09:11 AM.

  • Well Written x 3
  • Informative x 1
  • WellResearched x 1
  • like x 1

#381 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 18 November 2020 - 08:24 AM

In the previous post I meant to write: 'At the very least these arguments suggest blood protein removal should be eliminated as the cause of REJUVENATION in these parabiosis studies' (before looking to other causes). 


Edited by QuestforLife, 18 November 2020 - 08:25 AM.

  • Good Point x 1

#382 Iporuru

  • Guest
  • 216 posts
  • 221
  • Location:Europe

Posted 19 November 2020 - 05:48 AM

A brand new paper:

Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial

Repeated intermittent hyperoxic exposures, using certain hyperbaric oxygen therapy (HBOT) protocols, can induce regenerative effects which normally occur during hypoxia. The aim of the current study was to evaluate whether HBOT affects TL and senescent cell concentrations in a normal, non-pathological, aging adult population.

 

Results: Telomeres length of T helper, T cytotoxic, natural killer and B cells increased significantly by over 20% following HBOT. The most significant change was noticed in B cells which increased at the 30th session, 60th session and post HBOT by 25.68%±40.42 (p=0.007), 29.39%±23.39 (p=0.0001) and 37.63%±52.73 (p=0.007), respectively.

There was a significant decrease in the number of senescent T helpers by -37.30%±33.04 post-HBOT (P<0.0001). T-cytotoxic senescent cell percentages decreased significantly by -10.96%±12.59 (p=0.0004) post-HBOT.

In conclusion, the study indicates that HBOT may induce significant senolytic effects including significantly increasing telomere length and clearance of senescent cells in the aging populations.


  • Informative x 1

#383 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 19 November 2020 - 09:42 AM

A brand new paper:

Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial

Repeated intermittent hyperoxic exposures, using certain hyperbaric oxygen therapy (HBOT) protocols, can induce regenerative effects which normally occur during hypoxia. The aim of the current study was to evaluate whether HBOT affects TL and senescent cell concentrations in a normal, non-pathological, aging adult population.

 

Results: Telomeres length of T helper, T cytotoxic, natural killer and B cells increased significantly by over 20% following HBOT. The most significant change was noticed in B cells which increased at the 30th session, 60th session and post HBOT by 25.68%±40.42 (p=0.007), 29.39%±23.39 (p=0.0001) and 37.63%±52.73 (p=0.007), respectively.

There was a significant decrease in the number of senescent T helpers by -37.30%±33.04 post-HBOT (P<0.0001). T-cytotoxic senescent cell percentages decreased significantly by -10.96%±12.59 (p=0.0004) post-HBOT.

In conclusion, the study indicates that HBOT may induce significant senolytic effects including significantly increasing telomere length and clearance of senescent cells in the aging populations.

 

It concerns me that the real mechanism of action of this therapy may be: 

  • massive oxidative stress pushed throughout the body by high pressure
  • destruction of vulnerable cells (including, admittedly, senescent cells) via ROS
  • forced differentiation of the hematopoietic stem cell compartment to replace the lost cells with those with longer telomeres

Hence we have a situation where a naïve interpretation of two biomarkers of aging (telomere length of peripheral blood cells and senescent cell burden) indicates rejuvenation but the real effect is early exhaustion of the stem cell compartment. If my interpretation is correct then we should expect improved short term health span but reduced lifespan from such a therapy. This is analogous to a senolytic therapy. 

 

Admittedly this interpretation assumes telomere attrition is the primary reason for the failure of stem cells to renew the body with increasing age. If the failure of stem cells to replace lost tissues in the body is due to some other factor, such as the increased chance of survival of stem cells that self renew rather than differentiate (see my 'selfish cell lives longer' idea) then this conclusion may change, although I'd still expect telomeres to play an important role because longer telomeres would allow asymmetrically dividing stem cells to persist even in the face of the competition from more selfish cells.

 

Here is another relevant paper: 

 

Telomere elongation followed by telomere length reduction, in leukocytes from divers exposed to intense oxidative stress--implications for tissue and organismal aging 

 

 

Among the divers following the oxidative stress, significant telomere elongation was observed in granulocytes and naïve T cells, but not in memory T cells and B cells. Telomere length in granulocytes was mildly elongated in the control group as well, a finding that may relate to the extreme physical activity to which they were exposed. While telomere elongation in naïve T cells may be attributed to telomerase activation, we suggest that in granulocytes the elongation results from undifferentiated hematopoietic cells carrying longer telomeres that repopulate the peripheral hematopoietic compartment. This event might be accompanied by enhanced cell division within the repopulating pool. Since the aging of mammalian tissues can be attributed in part to the reduction in the replicative potential of self renewing cells, enhanced cell turnover under conditions of hyperbaric oxidative stress might be directly relevant to tissue and organismal aging.

 

 

Source: https://pubmed.ncbi.....gov/21320523/ 


  • Good Point x 2

#384 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 19 November 2020 - 01:04 PM

Correction to the hyperlink

 

https://pubmed.ncbi....h.gov/21320523/

 


  • Informative x 1

#385 aribadabar

  • Guest
  • 860 posts
  • 267
  • Location:Canada
  • NO

Posted 21 November 2020 - 10:18 PM

It concerns me that the real mechanism of action of this therapy may be: 

  • massive oxidative stress pushed throughout the body by high pressure
  • destruction of vulnerable cells (including, admittedly, senescent cells) via ROS
  • forced differentiation of the hematopoietic stem cell compartment to replace the lost cells with those with longer telomeres

 

From what I read there is significant proliferation/asymmetrical division, not just differentiation during HBOT so it seems net positive, even with the concerns above.

You take C60oo about ~2h prior to hopping into HBOT capsule and you should be fine, if ROS is an issue :)

Or you are saying taking a strong antioxidant will stall the stem cell process as it requires the ROS storm to ensue?

 

 

(see my 'selfish cell lives longer' idea)

 

Cell lives matter! :-D



#386 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 22 November 2020 - 09:53 AM

From what I read there is significant proliferation/asymmetrical division, not just differentiation during HBOT so it seems net positive, even with the concerns above.
You take C60oo about ~2h prior to hopping into HBOT capsule and you should be fine, if ROS is an issue :)
Or you are saying taking a strong antioxidant will stall the stem cell process as it requires the ROS storm to ensue?
.

Asymmetric division involves one cell remaining a stem cell and one differentiating. Symmetrical division means both cells have the same fate, which in the stem cell niche should mean both remain stem cells. There are advantages and disadvantages to both. When I said differentiation I meant asymmetric division, although there might also be some symmetrical division, I suppose.

No one knows for sure why the body does not replace cells that are getting old and damaged. Treatments like senolytics or hyperbaric O2 therapy assume the reason is not that there aren't enough replacement cells.

You see where I'm going with this?

Edited by QuestforLife, 22 November 2020 - 10:17 AM.


#387 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 23 November 2020 - 11:28 AM

It concerns me that the real mechanism of action of this therapy may be: 

  • massive oxidative stress pushed throughout the body by high pressure
  • destruction of vulnerable cells (including, admittedly, senescent cells) via ROS
  • forced differentiation of the hematopoietic stem cell compartment to replace the lost cells with those with longer telomeres

Hence we have a situation where a naïve interpretation of two biomarkers of aging (telomere length of peripheral blood cells and senescent cell burden) indicates rejuvenation but the real effect is early exhaustion of the stem cell compartment. If my interpretation is correct then we should expect improved short term health span but reduced lifespan from such a therapy. This is analogous to a senolytic therapy. 

 

 

 

The full paper is now available and it is interesting that the authors propose a different mechanism for the benefits of HBOT:

 

 

 

These intermittent hyperoxic exposures induce an adaptive response which includes increased upregulation of antioxidants genes [32] and production of antioxidants/scavengers that adjust to the increased ROS generation causing the ROS/scavenger ratio to gradually becomes similar to the ratio under a normal oxygen environment. However, because the scavenger elimination half-life (T1/2) is significantly

longer than the T1/2 of ROS, upon return to normoxia, following repeated hyperoxic exposures, there are significantly higher levels of scavengers and increased antioxidant activity [13, 18]. Thus, similar to physical exercise and caloric restriction, a daily repeated HBOT protocol can induce the hormesis phenomenon. Single exposures increase ROS generation acutely, triggering the antioxidant response, and with repeated exposures, the response becomes protective [13, 18].

 

source: https://www.aging-us...cle/202188/text

 

Essentially they are claiming the oxidative stress triggers a hormetic response that more than makes up for the harm.

 

I remain to be convinced. Feel free to make up your own mind/s. 


Edited by QuestforLife, 23 November 2020 - 11:29 AM.


#388 QuestforLife

  • Topic Starter
  • Guest
  • 1,521 posts
  • 1,133
  • Location:UK
  • NO

Posted 15 December 2020 - 11:21 AM

Summary of ‘Alternative Methods to Extend Telomeres’ Sept 2018 to Dec 2020
Early work on NAD+
https://www.longecit...es/#entry857309
https://www.longecit...e-2#entry868202
SIRT4
https://www.longecit...e-3#entry870174
Loss of NAD+ because of telomere shortening
https://www.longecit...-13#entry900015
Work on Statin-Sartan protocol
https://www.longecit...es/#entry862269
link between ROCK inhibitors and telomerase
https://www.longecit...es/#entry864097
possible link with senolytics
https://www.longecit...es/#entry864534
using ROCK and mTOR inhibitors to reprogram brain cancer cells into normal neurons
https://www.longecit...es/#entry865160
How ROCK inhibitors block differentiation
https://www.longecit...e-4#entry878635
Feedback on protocol
https://www.longecit...e-5#entry881808
Summary of ROCK inhibition action on cells
https://www.longecit...e-6#entry883118
Attempts to come up with alternatives to statin and sartans
https://www.longecit...e-7#entry884915
Diagram of interventions
https://www.longecit...e-8#entry885663
Paper linking up ROCK and ECM
https://www.longecit...e-8#entry885731
ROCK and tgf-b
https://www.longecit...e-8#entry886244
Mean and Max lifespan extension with a ROCK inhibitor
https://www.longecit...-10#entry896988
Work on telomerase activators
Royal Jelly
https://www.longecit...e-2#entry866228
Review of various activators
https://www.longecit...e-4#entry875566
Asiaticoside
https://www.longecit...e-5#entry880274
Some other telomeres studies
https://www.longecit...e-7#entry884556
Effect of antioxidant on telomere shortening in the bone marrow
https://www.longecit...e-8#entry885539
More on the same, later
https://www.longecit...-10#entry896907
Telomere activators and CV diseases
https://www.longecit...e-8#entry885582
Telomere shortening predicts species life span
https://www.longecit...e-9#entry893160
using TERC upregulation to increase telomere length in stem cell
https://www.longecit...-10#entry896804
Telomerase and Splicing Factor regulators
https://www.longecit...-11#entry899109
T cells taking telomere length from other cells
https://www.longecit...-11#entry899161
Do stem cell stimulants deplete the bone marrow pool?
https://www.longecit...-13#entry900006
Hyperbaric oxygen therapy
https://www.longecit...-13#entry900378
View of Aging
Importance of cell size
https://www.longecit...e-4#entry877909
The Selfish Cell lives longer
https://www.longecit...e-5#entry880039
https://www.longecit...e-5#entry880339
Telomeres are NOT passive in aging
https://www.longecit...e-6#entry883065
Discussion of telomeres and cancer
https://www.longecit...e-9#entry892745
Senescence and Cancer, again
https://www.longecit...-10#entry897658
Are methylation changes with age evidence of a program?
https://www.longecit...-12#entry899778
Comments on heterochronic parabiosis
https://www.longecit...-13#entry900319
Skin aging
Stem cell competition – can you have too much symmetrical division?
https://www.longecit...e-4#entry879560
Results
Methylation results from Statin-Sartan protocol
https://www.longecit...e-3#entry873678
Telomere length improvements via Lifelength
https://www.longecit...e-6#entry883063
PhenoAge improvements
https://www.longecit...e-6#entry883130
Epitalon increases methylation age and discussion
https://www.longecit...e-8#entry892505
Further discussion
https://www.longecit...-11#entry899397
https://www.longecit...-11#entry899496
https://www.longecit...-12#entry899538
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
Sundry
Fatty Acid Oxidation
https://www.longecit...-10#entry896447
Starvation and stem cell renewal
https://www.longecit...-13#entry899839
See other thread:
Feeding stem cells: the strange case of dietary restriction and alpha lipoic acid
https://www.longecit...id/#entry885897


  • Informative x 6
  • WellResearched x 4
  • like x 1

#389 dlewis1453

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

Posted 15 December 2020 - 04:55 PM

This is great! Thanks for compiling all of these informative posts into one place. You should consider adding a link to this post to your profile page for ease of reference going forward. 



#390 Castiel

  • Guest
  • 374 posts
  • 86
  • Location:USA

Posted 03 January 2021 - 02:01 AM

  But telomeres too MAY be designed to trade the death of the old for the (breeding) success of the young. Nature is happy for short telomeres to kill the old with cancer or any of the other diseases of aging, in order to avoid the small chance (excessively) long telomeres in the young could lead to more of the much rarer, but deadly, childhood cancers.

Most of all programmed aging lacks a clock. This is ironic given the many methylation clocks quoted by Yuri in its support. But what is the signal that makes them tick? It still hasn't been found. Yuri suggests the germ cell signalling on sexual maturity that leads to heat shock response being downregulated in nematodes. But it's clear any kind of heat is bad for germ cells. Even saunas that make you live longer will damage your sperm count.
 

 

The problem is IIRC Bill Andrews suggested the average telomere length when you're under 20 is already enough to produce a cancer that can grow big enough to kill you even without telomerase.

 

 


Here's a much simpler explanation. It is the stochastic damage of metabolism that makes epigenetic clocks tick. Old cells (without ready replacements) acquire dysfunctional mitochondria through stochastic metabolic processes and the passage of time The cell then adapts to a bad situation by making the best of it and down and upregulating various genes in the nucleus. Then (naive) scientists come along and decide the problem (and aging itself) is those adaptations.

Consider this the null hypothesis. Disprove it if you can.

According to several sources, one of the things that's downregulated is the rate of protein turnover with aging.   Reduced protein turnover leads in turn to the accumulation of damaged proteins and cellular dysfunction.   This reduction in protein replacement rate is reversed with restored telomere length.

 

The cells are not adapting to a bad situation, they're just purposely making everything worse, they're downregulating mitochondrial maintenance, protein maintenance, and in the case of senescent cells releasing protumor formation and proinflammatory toxic chemicals to their surrounding.

 

 

A more recent study out of the Rabinovitch group examined protein turnover among ubiqui-tinated proteins in aging, and found that a striking proportion of ubiquitinated proteins were not turning over in aged animals.[]This effect was not present in young animals and was allevi-ated or reversed in calorie restricted or rapamycin-treated mice 

(PDF) Protein Turnover in Aging and Longevity (researchgate.net)

 

The cell is actively trying to worsen the health of the organism and lead to it being eaten or killed by disease 

 

It's like some companies and their product's designed with specific lifetime, designed to wear out within a specific period, to be replaced by newer models.   The rate of evolution is increased by killing animals and replacing them with a new wave of gene mixed babies.   But signal that food is scarce, the babies might be dying, and the organism will postpone the end of its shelf life, calorie restriction.


  • Needs references x 1





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