• 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
1028 replies to this topic

#601 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 24 June 2021 - 11:30 AM

From reading and agreeing with "Turbuckle's" approach to live extension as well as my personal experiences, I am coming to the conclusion that we do not want to take things like NAD+ boosters, AKG, and the like on a semi-permanent basis. I think we want to take these things only as a part of a short-term or periodic protocol, and not take them the rest of the time. I think this is especially true for senolytics and NAD+. My own experience with NAD+ over a three year period is that I developed a persistent cough that was annoying to my wife and other people around me. It turned out that a lot of the cause of it was elevated histamine levels that was caused, in part, by NAD+ compounds as well as Zinc. I stopped taking both of these and my cough largely went away. I also think senolytics can be too much of a good thing. Your body needs a certain amount of inflammation signaling for both wound healing as well as immune defense system. I've used senolytics in the past (Fisetin and Quercetin), but only for short-term protocols. My experience with Curcumin also reinforced my beliefs about this.

 

Zinc also makes my hayfever worse, although it is great for the immune system in general, which may be a priority in covid times.

 

Zinc is also good for telomeres, see post #488



#602 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 25 June 2021 - 08:17 AM

Summary of ‘Alternative Methods to Extend Telomeres’ Sept 2018 to June 2021

 

April-June 2021 new entries in red italics.

 

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 and other important telomere papers
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
Discussion of Blasco paper on hyperlong telomere mice
https://www.longecit...ndpost&p=901986
Discussion of actual in vivo rate of telomere attrition
https://www.longecit...-14#entry902137
GDF11 lengthens telomeres in MSCs via TERC upregulation
https://www.longecit...-15#entry903694
Possible benefit of Klotho to telomeres
https://www.longecit...-15#entry903694
Nucleotides (specifically guanine) for elongation of telomeres: eat Anchovies and Herring!
https://www.longecit...-15#entry904277
Blasco and short telomeres in kidney disease plus possible connection of short telomeres and the cancer causing epithelial to mesenchymal transition
https://www.longecit...-15#entry904567
What is the most powerful telomerase activator and a comparison of methods of measurement
https://www.longecit...-15#entry905188
Melatonin is the best antioxidant for telomeres?
https://www.longecit...-16#entry905284
More on melatonin
https://www.longecit...-18#entry906399
AKG and telomere length (in mice)
https://www.longecit...-16#entry905690
Discussion of a cell permeable, oxidation resistant form of Vit C and telomeres plus follow on discussion of ROS hormesis in some cell types
https://www.longecit...-16#entry905240
Various discussions on the bioavailability of Asiatic acid/asiaticoside (a purported telomerase activator) and why you may only want a very small dose
https://www.longecit...-14#entry903398
Should we be taking Zinc for our telomeres?
https://www.longecit...-17#entry906002

Clear benefits to life expectancy, CVD and Cancer Risk Factors with longer telomeres: a study with 500k people
https://www.longecit...-17#entry906042
Ability of endothelial cells to make new lining is telomere length dependent
https://www.longecit...-17#entry906088
Caffeine promotes telomerase expression
https://www.longecit...-17#entry906174
Dark chocolate for telomeres
https://www.longecit...-17#entry906249
Alternatives to a telomere test: NLR and CRP
https://www.longecit...-19#entry906563
Hyperfunctional telomerase: do you want more cell division or longer telomeres?
https://www.longecit...-20#entry907024
We should be aiming for mouse levels of telomerase, not HELA levels
https://www.longecit...-20#entry907165

 

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
More on Selfish Cell theory of aging (2021)
https://www.longecit...-14#entry902349
Age related methylation and the connection with the Selfish Cell Theory of Aging
https://www.longecit...-16#entry905284
Plus why aging is cancer
https://www.longecit...-16#entry905627
Putting together telomere and hyperfunction theories of aging
https://www.longecit...-20#entry907460

 

 

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
Further improvement in epigenetic age (-6.6 years)
https://www.longecit...-14#entry903105
Summary of GDF11 experience with biomarkers
https://www.longecit...-15#entry905149
May 2021 Methylation age results
https://www.longecit...-20#entry907470

 

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
Possible use of pioglitazone with telomerase activators to increase subcutaneous fat without bladder cancer risk
https://www.longecit...-14#entry902929
Resveratrol is weird.
https://www.longecit...-16#entry905283
Demethylating the klotho promoter with hydrogen sulphide
https://www.longecit...-17#entry905925


Edited by QuestforLife, 25 June 2021 - 08:20 AM.

  • WellResearched x 3
  • Informative x 1

#603 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 30 June 2021 - 08:32 AM

New intranasal and injectable gene therapy for healthy life extension

 

 
Previous studies showed that adenovirus-associated virus (AAV) vector induced overexpression of certain proteins can suppress or reverse the effects of aging in animal models. Here, we sought to determine whether the high-capacity cytomegalovirus vector can be an effective and safe gene delivery method for two such-protective factors: telomerase reverse transcriptase (TERT) and follistatin (FST). We found that the mouse cytomegalovirus (MCMV) carrying exogenous TERT or FST (MCMVTERT or MCMVFST) extended median lifespan by 41.4% and 32.5%, respectively. This is the first report of CMV being used successfully as both an intranasal and injectable gene therapy system to extend longevity. Treatment significantly improved glucose tolerance, physical performance, and prevented loss of body mass and alopecia. Telomere shortening seen with aging was ameliorated by TERT, and mitochondrial structure deterioration was halted in both treatments. Intranasal and injectable preparations performed equally well in safely and efficiently delivering gene therapy to multiple organs, with long-lasting benefits and without carcinogenicity or unwanted side effects. Translating this research to humans could have significant benefits associated with increased health span.
https://www.biorxiv.....06.26.449305v1

 

This work has been sponsored by Bioviva (Liz Parrish). Independent researchers ran the study however, and George Church advised, so I’m not too worried about bias or rigour.

 

It is different to the work done by Blasco in 2012 (DOI 10.1002/emmm.201200245) because instead of just one viral telomerase treatment at either 1 or 2 yo, here the mice had a dose every month from 18mo (equivalent 56yo human) to 29mo (equiv age ~80, when the last control mouse died), and then restarted from 32mo till death. The current paper also used two routes of administration: intra-peritoneal (injected into the tail) and intranasal (using a nose spray). One of the discoveries of this paper is that the nose spray was just as effective at extending lifespan as the injection. The other main difference in this paper is that instead of using the usual AAV (adeno-associated) viral vector that carries the telomerase (or follistatin gene), they use a CMV (cytomegalovirus) carrier. Although this isn’t done in the paper, CMV could carry much more payload than AAV – for example both the genes that in this paper were given to different groups, or maybe other anti-aging payload as well (klotho, GDF11, etc.). Not doing that in this paper allowed them to separate out the effects of telomerase, follistatin, and the two different methods of administration (plus 3 control groups: a genuine control and empty vectors for each gene therapy). The downside of this approach is with so many groups they only had 9 mice per group, and could only sacrifice 1 per group to determine tissue telomere lengths at a single time point. It is also possible that they could have achieved more life extension if they combined telomerase and follistatin in one group, (though this is speculation at this point).

 

 

Because the mouse groups were small, they needed a lot of life extension to find a significant effect, but nevertheless did so with median lifespan extended 32.5% in the follistatin groups and 41.4% in the telomerase groups (see Fig A below). Max lifespan was also considerably extended here, though I’d urge some caution in over interpretation, as the control groups were dead faster than I’d expect. Because median lifespan was about right however, I’d just put this down to the small sample size and nothing fishy.

 

F2.large.jpg?width=800&height=600&carous

 

From a telomere point of view the most interesting finding was the  effect of monthly telomerase treatment (from 18-24mo) on the telomere lengths in various tissues (as measured in sacrificed mice@24mo). Compared to a young 6mo mouse, telomere lengths shortened tremendously in the brain, heart, kidney, liver, lung  and muscle in the control (intranasal empty vector) and follistatin (intranasal) groups. By contrast the telomerase (intranasal) group had only very slightly (~8%) shorter telomeres than the young mouse (see Fig F, bottom right). This is pretty remarkable and is also reflected in far better appearance, speed over a narrow beam, mitochondrial health markers, climbing attempts*, and the fact all the control mice were dead by 29mo, but the first telomerase mouse did not die until ~33mo and the last didn’t die until 40mo. That would be the human equivalent of the control group starting to die at about 60yo and all being dead by 80, and the telomerase group starting to die in their mid 80s and the last not dying till about 110 yo.

 

*follistatin gene therapy also improved appearance, beam test speed and mitochondrial health markers (but not climbing as they were too big).

 

If telomeres were mostly stopped from shortening in the telomerase group, why didn’t the mice live even longer?  Given telomere and mitochondrial markers were well preserved, is there another cause of death? There are a couple of shortcomings in the study before we can conclude that. Firstly, they only sacrificed mice once at 24mo to check telomere length. At that stage telomere length was well preserved in the telomerase therapy sacrifice (but severely reduced in control, See Fig F), and this explains why 5 months later all the controls were dead, but none of the telomerase groups were anywhere near dying. But telomeres were not measured again. The paper showed that one treatment boosted telomerase considerably, falling gradually back to baseline in about 25 days. But we don’t know the effect of multiple administrations. We don’t know if CMV continues to evade the immune system after many, many treatments. My guess would be that its effectiveness declines and telomeres do shorten even with repeated treatments. But we’ll have to wait for another study to find out if I'm right.


Edited by QuestforLife, 30 June 2021 - 08:37 AM.

  • like x 3
  • Informative x 2

sponsored ad

  • Advert

#604 OlderThanThou2

  • Registrant
  • 120 posts
  • 28
  • Location:France

Posted 02 July 2021 - 10:20 AM

I'd be curious to know if cells that became senescent through telomere shortening became functional again. Perhaps the other types of senescent cells continued to accumulate and prevented even better results.


  • like x 2
  • Enjoying the show x 1

#605 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 02 July 2021 - 11:47 AM



I'd be curious to know if cells that became senescent through telomere shortening became functional again. Perhaps the other types of senescent cells continued to accumulate and prevented even better results.

 

There are improved ways of measuring of senescent cells becoming available to researchers now, for example see here:

 

 

Senescence-associated β-galactosidase reveals the abundance of senescent CD8+ T cells in aging humans

Using a second-generation fluorogenic substrate for β-galactosidase and multi-parameter flow cytometry, we demonstrate here that peripheral blood mononuclear cells (PBMCs) isolated from healthy humans

increasingly display cells with high senescence-associated β-galactosidase (SA-βGal) activity with advancing donor age. The greatest age-associated increases were observed in CD8+ T-cell populations, in which the fraction of cells with high SA-βGal

activity reached average levels of 64% in donors in their 60s.

Source: DOI: 10.1111/acel.13344

 

 

But it is far from clear to me how separate the different forms of senescence are. For example in a high ROS cell culture, cells will be constantly becoming senescent, but so long as telomeres are sufficiently long in other cells, then those cells can be replaced and as the culture is passaged they will continue to proliferate. Of course a body is not the same as cells being divided on petri dishes, but you get my point. I liken this to the analogy of the leaky dingy. Mice have lots of air in their inflatable, but lots of holes. Humans' have a dingy that sits lower in the water, but loses air very slowly by comparison. 

 

I talked about this previously in contrasting mouse cells in culture@20% oxygen that senescence quickly VS human cells@20% oxygen that senesce more slowly, and then mouse cells@3% oxygen that never senesce(*) VS humans cells@3% oxygen that do eventually senesce. This tells us that mouse cells are set up differently to human cells; in low Oxygen conditions ROS and growth signalling are low enough that they can divide perpetually, whereas human cells are limited by their short (non-renewed) telomeres. But in higher oxygen conditions, even with their longer telomeres (with active telomerase) mouse cells fare worse than human cells, which much maintain lower ROS and/or growth signalling than mouse cells. This makes total sense when you look at the different life strategies; grow up quick and breed before you die (mice) vs grow up slowly and then live and breed a long time (humans). 

 

(*)Here I am talking about senescence of the culture (completely stopping dividing); obviously some individual cells will always senesce.

 

Therefore you'd expect a much slower build up of senescent cells in humans than mice, but with humans you'd expect a bias of senescent cell accumulation later in life when the telomeres of the body start to get short. The third type of senescence we haven't mentioned is oncogene induced senescence. I'd expect that would be proportional to growth signal levels, and again would kick at a much greater rate as telomeres get short (and genomic stability plummets) in humans.

 

In terms of the study in question, it is interesting that the follistatin group also had pretty short telomeres at 24months, but didn't start dying for another 6 months, with controls all dead after 5. I'd attribute this to better mitochondrial health with consequent reduced ROS. One of the ways short telomeres cause senescence is by jacking up ROS. For example you can see from the following Figure(B) how nicotinamide held mitochondrial ROS down for longer than in control cells (compare Green and Red traces), but near telomere exhaustion it still suddenly shoots up and causes the culture to senesce. Do you see what I mean about ROS induced and telomere induced senescence being difficult to separate out?

 

acel_234_f4.gif

Sourcehttps://onlinelibrar...26.2006.00234.x

 

 

The one thing I forgot to mention in reviewing Liz Parrish's new study, is that it finally puts to bed the false argument that mice do not suffer from short telomeres with age. They clearly do, though as I've expounded here, those short telomeres are largely due to ROS and growth signalling. 


Edited by QuestforLife, 02 July 2021 - 11:53 AM.

  • WellResearched x 2

#606 OlderThanThou2

  • Registrant
  • 120 posts
  • 28
  • Location:France

Posted 02 July 2021 - 01:36 PM

Regarding the oxygen cencentration affecting longevity, there is this study that shows that centenarians are slightly anemic probably due to genetic factors, and that their longevity may be related to less ROS being produced because of that:

 

0354-46641600043H.pdf (nb.rs)

 

 

Abstract: Hemoglobin (HGB) in the blood carries oxygen from the lungs to other organs to produce energy. Calorie restriction has been shown to slow aging and extend lifespan. Thus, we hypothesized that HGB may be associated with human longevity as a link to energy metabolism. To test this hypothesis, HGB levels in the blood of 60 centenarian (CEN) families were measured and its association with age (20-80 and 20-100 years) was studied, as well as the associations of CEN HGB with levels in first generation offspring (F1) and their spouses (F1SP). The results showed no association of HGB with age between 20 and 80 years (r=-0.097, p=0.160); however a strikingly inverse relationship with age between 20 and 100 years (r=-0.526, p<0.001) was revealed. After dividing the samples into four age groups (20-39, 40-59, 60-80 and ≥100 years), the HGB in CEN were significantly lower than that of F1SP (p<0.001). Interestingly, the HGB levels of CEN were significantly associated with that of F1 (r=0.379, p=0.015) but not with F1SP (r=0.022, p=0.451), suggesting that HGB could be a heritable phenotype. Furthermore, the genes methylenetetrahydrofolate reductase (MTHFR), nuclear receptor subfamily 2, group C, member 1 (NR2C1) and NR2C2 were differentially expressed in CEN when compared to F1SP, which may likely be responsible for the changes in HGB levels. In conclusion, our results suggest that HGB is a heritable phenotype which associates with familial longevity.

 

People on calorie restriction often become slightly anemic, that may at least partially explain why CR improves longevity. Me for instance, I'm doing CR, and my hemoglobin is low ( even if my iron level is normal ), so that might help my cells produce less ROS.

 

Also I hope I am not wrong on that, nicotimamide seems to be a sirt6 inhibitor. See the figure at the bottom here:

Figure: Inhibition of SIRT6 Enzyme activity by Nicotinamide using kit protocol.

Microsoft Word - PK-CA577-K323.doc (promocell.com)

 

Here they use nicotinamide's ability to inhibit the SIRTs, including SIRT6, in cancer cells.

JAKO202007963128005.pdf (koreascience.or.kr)

 

So in vivo, even if nicotinamide has a beneficial effect on NAD+, perhaps it is compensated by its ability to inhibit SIRT6?

 

 

 


  • Informative x 1

#607 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 03 July 2021 - 11:36 AM

Regarding the oxygen cencentration affecting longevity, there is this study that shows that centenarians are slightly anemic probably due to genetic factors, and that their longevity may be related to less ROS being produced because of that:

Thank you, very interesting study. As a cross sectional you can never be sure whether the haemoglobin falls with age or is associated with genetic longevity (at any age). Given they found haemoglobin was also lower in the children of centenarians, we know it is atleast partially inherited.

This is exactly as we would expect based on my observations that slower growth factors (oxygen) lead to slower telomere attrition (both via direct ROS induced senescence and telomere exhaustion via faster division of cells).

This quote from your paper is particularly interesting.

Haslam et al. [22], who reported that anemia in centenarians
was only associated with lower hand-grip and leg strength, but not with physical
function in everyday activities.

Again,this is what we'd expect from (potential) centenarians being an 'economy model' human, built to last but not for performance.

Indeed 'the master' Blagoskonny has said exactly this.

Why human lifespan is rapidly increasing: solving "longevity riddle" with "revealed-slow-aging" hypothesis

Healthy life span is rapidly increasing and human aging seems to be postponed. As recently exclaimed in Nature, these findings are so perplexing that they can be dubbed the 'longevity riddle'. To explain current increase in longevity, I discuss that certain genetic variants such as hyper-active mTOR (mTarget of Rapamycin) may increase survival early in life at the expense of accelerated aging. In other words, robustness and fast aging may be associated and slow-aging individuals died prematurely in the past. Therefore, until recently, mostly fast-aging individuals managed to survive into old age. The progress of civilization (especially 60 years ago) allowed slow-aging individuals to survive until old age, emerging as healthy centenarians now. I discuss why slow aging is manifested as postponed (healthy) aging, why the rate of deterioration is independent from aging and also entertain hypothetical use of rapamycin in different eras as well as the future of human longevity.
Source: https://pubmed.ncbi....h.gov/20404395/


Edited by QuestforLife, 03 July 2021 - 11:37 AM.

  • Informative x 4

#608 OlderThanThou2

  • Registrant
  • 120 posts
  • 28
  • Location:France

Posted 05 July 2021 - 01:05 PM

It seems, as your data seems to show, that mice are more resistant to ROS than humans. In this study they showed that reducing GPX4 increases lifespan in mice

 

Reduction in Glutathione Peroxidase 4 Increases Life Span Through Increased Sensitivity to Apoptosis | The Journals of Gerontology: Series A | Oxford Academic (oup.com)

 

 

Glutathione peroxidase 4 (Gpx4) is an antioxidant defense enzyme that plays an important role in detoxification of oxidative damage to membrane lipids. Because oxidative stress is proposed to play a causal role in aging, we compared the life spans of Gpx4 heterozygous knockout mice (Gpx4+/− mice) and wild-type mice (WT mice). To our surprise, the median life span of Gpx4+/− mice (1029 days) was significantly longer than that of WT mice (963 days) even though the expression of Gpx4 was reduced approximately 50% in all tissues of Gpx4+/− mice. Pathological analysis revealed that Gpx4+/− mice showed a delayed occurrence of fatal tumor lymphoma and a reduced severity of glomerulonephritis. Compared to WT mice, Gpx4+/− mice showed significantly increased sensitivity to oxidative stress-induced apoptosis. Our data indicate that lifelong reduction in Gpx4 increased life span and reduced/retarded age-related pathology most likely through alterations in sensitivity of tissues to apoptosis.

 

The same is true for SOD2, lower SOD2 increases lifespan:

 

 

 

The life span of Gpx4+/− mice appears to be inconsistent with the Oxidative Stress Theory of Aging, i.e., increased oxidative stress should reduce life span, not extend life span. Previously, our group showed that Sod2+/− mice, which have reduced expression of MnSOD in all tissues and increased levels of oxidative damage to DNA, had an identical life span as control, WT mice (52). The life-span data of Gpx4+/− and Sod2+/− mice show that a deficiency in one component of the antioxidant defense does not directly lead to accelerated aging in mice.

 

 

The normal mice die quickly from cancer, so it high ROS helps increase lifespan by killing cancer ( lymphoma in that case ). It seems to me like it's a tug of war between resistance to ROS which lengthens the telomeres and cancer. Both high and very low ROS are bad with regard to cancer. The former causes genetic mutation leading to cancer, while the latter causes lack of apoptosis in cancer cells.

 

 

Why would an anti-oxidant like C60 extend the lifespan of mice then? Perhaps it both extends the telomeres by reducing very much the ROS, but  also kills cancer cells, as this study shows:

C 60 fullerene and its nanocomplexes with anticancer drugs modulate circulating phagocyte functions and dramatically increase ROS generation in transformed monocytes | Cancer Nanotechnology | Full Text (biomedcentral.com)

 

 

Moreover, spontaneous induction of ROS generation by C60 fullerenes plays a pivotal role in their toxic effect on eukaryotic normal and malignant cells. C60 fullerenes and their derivatives stimulate oxidative metabolism in erythrocytes and fibroblasts, malignant lymphocytes and epithelial cells, and endothelial cells and macrophages (Trpkovic et al. 2012). 

 

 

Perhaps evolution tweeks cancer resistance and telomere lengthening via ROS to pick the right lifespan for a particular type of species. Species with a high breeding rate reproduction strategy like mice don't live much longer after they've reproduced because they were made by evolution not resistant to cancer. But Humans ( who rarely die from cancer in primitive populations ) live longer after they've reproduced because they are more resistant to cancer, but in the end, they die from telomere shortening

 

Blagoskonny's point may be confirmed by that study on centanarians. Those who have the genetic mutation than made them rather anemic  probably were not favored in the past when maximum resistance to stress was needed to survive. Same thing for mTOR, those that expressed it less had less ability to repair from injuries and such.

 

 



#609 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 05 July 2021 - 04:27 PM

It seems, as your data seems to show, that mice are more resistant to ROS than humans. In this study they showed that reducing GPX4 increases lifespan in mice

Reduction in Glutathione Peroxidase 4 Increases Life Span Through Increased Sensitivity to Apoptosis | The Journals of Gerontology: Series A | Oxford Academic (oup.com)



I was actually saying the OPPOSITE, that mouse cells are MORE sensitive to ROS than human cells. This is the only way to explain why mouse cells don't senesce@3% O2, but much more quickly than human cells@20% O2.

The study on Gpx4+/− mice you reference, is messing with a subset of the mouse anti-oxidant system particular to protection from lipid peroxidation, and it had the unexpected effect of increasing rather than decreasing lifespan, which they attribute to cancer suppression due to increased apoptosis. It is anybody's guess if this is the real reason. No one is arguing that fully disabling the Gpx4 (or SOD) gene would be harmful (even lethal). I expect these mice would be very sensitive to oxidative stress and this would translate to reduced survival in the wild. There is also the argument that mice have telomeres to spare, so can afford to replace extra cells lost to apoptosis.

I've read studies in the past where partially decreasing one part of the antioxidant system upregulated another part. Conversely, C60 (purported to be a powerful antioxidant) in the Baati study resulted in downregulation of glutathione in the dosed rats, presumably because they no longer needed as much.

In the GDip group i.p. pre-treated with C60-oil, the GSSG/TGSH
was even significantly lower than in the control group. Source: https://doi.org/10.1...als.2012.03.036


Overall ROS seems to be finely balanced in each species, which is why I’ve settled on telomeres and MTOR as a more viable strategy for life extension.
  • Informative x 1

#610 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 07 July 2021 - 02:59 PM

New GDF11 telomerase paper in Nature: 

Growth differentiation factor 11 attenuates cardiac ischemia reperfusion injury via enhancing mitochondrial biogenesis and telomerase activity

It has been reported that growth differentiation factor 11 (GDF11) protects against myocardial ischemia/reperfusion (IR) injury, but the underlying mechanisms have not been fully clarified. Considering that GDF11 plays a role in the aging/rejuvenation process and that aging is associated with telomere shortening and cardiac dysfunction, we hypothesized that GDF11 might protect against IR injury by activating telomerase. Human plasma GDF11 levels were significantly lower in acute coronary syndrome patients than in chronic coronary syndrome patients. IR mice with myocardial overexpression GDF11 (oe-GDF11) exhibited a significantly smaller myocardial infarct size, less cardiac remodeling and dysfunction, fewer apoptotic cardiomyocytes, higher telomerase activity, longer telomeres, and higher ATP generation than IR mice treated with an adenovirus carrying a negative control plasmid. Furthermore, mitochondrial biogenesis-related proteins and some antiapoptotic proteins were significantly upregulated by oe-GDF11. These cardioprotective effects of oe-GDF11 were significantly antagonized by BIBR1532, a specific telomerase inhibitor. Similar effects of oe-GDF11 on apoptosis and mitochondrial energy biogenesis were observed in cultured neonatal rat cardiomyocytes, whereas GDF11 silencing elicited the opposite effects to oe-GDF11 in mice. We concluded that telomerase activation by GDF11 contributes to the alleviation of myocardial IR injury through enhancing mitochondrial biogenesis and suppressing cardiomyocyte apoptosis.
Source: https://www.nature.c...19-021-03954-8 

Here is a nice visual abstract if you're in a hurry:

41419_2021_3954_Fig8_HTML.png?as=webp

 

 

 

As we saw with the recent Liz Parrish paper I reviewed very recently here, telomerase treatment not only benefited telomere length, but also mitochondrial health.

After taking TAM818 orally for a month, I noticed my energy levels were considerably enhanced and my recovery from heavy weights is at least three times quicker (3 days to 1 day). It seems unlikely this effect could be mediated by an increase in telomere length in such a short space of time. Mitochondrial health is a possible explanation.

 

Back to the paper, they used genetic techniques to overexpress GDF11, called oe-GDF11, in cardiomyocytes (heart muscle cells)  to see how this protected the cells from a simulated form of heart attack.

 

The following Figure C is interesting, as it is the first time I have seen plasma GDF11 concentration vs age (in a healthy human control group). Fig A also shows that the healthy controls on average have higher plasma GDF11 than chronic coronary patients, who in turn have higher levels than acute coronary patients. 

41419_2021_3954_Fig1_HTML.png?as=webp

 

 

The following Figure shows the telomere length of cultured neonatal rat cardiomyocytes - both control (blue bars) and subject to AR= Anoxia/reoxygenation (a way of simulating the ischemia/reperfusion that occurs to heart cells in a heart attack) (red bars), as well as cells subject to AR that had GDF11 overexpressed (green bars), as well as if a telomerase inhibitor was added (grey bars). 

 

Telomere fluorescence (Fig B) indicated GDF11 overexpression not not only restored the Telomere length of cells subjected to AR but made them 3x longer than controls!. PCR measurements indicated telomere length was only ~ doubled (Fig C). The telomere length increases were only partially blocked by the telomerase inhibitor indicating that GDF11 is upstream of telomerase activation and also, may act through telomerase independent pathways - my supposition. Either way, Fig D and E proved that the telomerase protein and gene expression (mRNA) respectively were both upregulated by GDF11 genetic over-expression. Fig F also shows other parts of the telomere gene machinery also had altered expression. For some reason they didn't look at TERC (the RNA templated used by telomerase to elongate the telomere).

 

41419_2021_3954_Fig6_HTML.png?as=webp

 

Note that I previously reviewed a paper where GDF11 lengthened telomeres in human MSCs via TERC upregulation, see post 424 here. This may be a difference between the human cells used previously and the mice cells used in this current paper. The other main difference is that human MSCs had recombinant GDF11 added, rather than using genetic manipulation, which is less representative of what we could actually do. 

 

Conclusion: Here is some more evidence that GDF11 is beneficial to telomeres, and hence protective against heart attack risk.

 

 


Edited by QuestforLife, 07 July 2021 - 03:05 PM.

  • Informative x 4

#611 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 08 July 2021 - 06:47 AM

Do you have an opinion on what is GDF11 half life is? Steve Perry and the enthusiast community are set on treating it like it has a half life similar to Vitamin D, with heavy loading phase and small maintaining dose.

I find it strange.

 



#612 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 08 July 2021 - 07:39 AM

Do you have an opinion on what is GDF11 half life is? Steve Perry and the enthusiast community are set on treating it like it has a half life similar to Vitamin D, with heavy loading phase and small maintaining dose.

I find it strange.

 

I think Steve Perry is right - I shared in post 424 that exogenous GDF11 upregulates some of the demethylases (TETs), and they demethylate the GDF11 promoter. So in theory, taking GDF11 can increase your endogenous production of GDF11. That is also what I experienced - to begin with I was taking GDF11 more than once a week, but now I am heading towards once a month. I measure the requirement to take another dose based on reaction time trending. This is by far the biggest change for me with GDF11 use; BP and HRV also improved, but it took longer and was much less pronounced, plus those biomarkers can be affected more easily by other things. One other thing, I also found that using AKG made me much more sensitive to GDF11 - which is what you'd expect as AKG is a TET co-factor. If I took them together, or if I took too much I felt like my BP was dropping too much (it didn't drop much, but it felt very strange). So it is likely that other things I was taking like AKG, plus my relative youth (42) meant I needed much less GDF11 than others have reported. Looking at the graph in post #610, serum GDF11 varies widely in youth but funnels in and drops significantly after 60 (;look at all the bunching below the trend line by that age). 


Edited by QuestforLife, 08 July 2021 - 07:42 AM.

  • Good Point x 2
  • Informative x 2

#613 marcobjj

  • Guest
  • 313 posts
  • 29
  • Location:California

Posted 13 July 2021 - 02:28 AM

New intranasal and injectable gene therapy for healthy life extension

 

 

This work has been sponsored by Bioviva (Liz Parrish). Independent researchers ran the study however, and George Church advised, so I’m not too worried about bias or rigour.

 

It is different to the work done by Blasco in 2012 (DOI 10.1002/emmm.201200245) because instead of just one viral telomerase treatment at either 1 or 2 yo, here the mice had a dose every month from 18mo (equivalent 56yo human) to 29mo (equiv age ~80, when the last control mouse died), and then restarted from 32mo till death. The current paper also used two routes of administration: intra-peritoneal (injected into the tail) and intranasal (using a nose spray). One of the discoveries of this paper is that the nose spray was just as effective at extending lifespan as the injection. The other main difference in this paper is that instead of using the usual AAV (adeno-associated) viral vector that carries the telomerase (or follistatin gene), they use a CMV (cytomegalovirus) carrier. Although this isn’t done in the paper, CMV could carry much more payload than AAV – for example both the genes that in this paper were given to different groups, or maybe other anti-aging payload as well (klotho, GDF11, etc.). Not doing that in this paper allowed them to separate out the effects of telomerase, follistatin, and the two different methods of administration (plus 3 control groups: a genuine control and empty vectors for each gene therapy). The downside of this approach is with so many groups they only had 9 mice per group, and could only sacrifice 1 per group to determine tissue telomere lengths at a single time point. It is also possible that they could have achieved more life extension if they combined telomerase and follistatin in one group, (though this is speculation at this point).

 

 

Because the mouse groups were small, they needed a lot of life extension to find a significant effect, but nevertheless did so with median lifespan extended 32.5% in the follistatin groups and 41.4% in the telomerase groups (see Fig A below). Max lifespan was also considerably extended here, though I’d urge some caution in over interpretation, as the control groups were dead faster than I’d expect. Because median lifespan was about right however, I’d just put this down to the small sample size and nothing fishy.

 

F2.large.jpg?width=800&height=600&carous

 

From a telomere point of view the most interesting finding was the  effect of monthly telomerase treatment (from 18-24mo) on the telomere lengths in various tissues (as measured in sacrificed mice@24mo). Compared to a young 6mo mouse, telomere lengths shortened tremendously in the brain, heart, kidney, liver, lung  and muscle in the control (intranasal empty vector) and follistatin (intranasal) groups. By contrast the telomerase (intranasal) group had only very slightly (~8%) shorter telomeres than the young mouse (see Fig F, bottom right). This is pretty remarkable and is also reflected in far better appearance, speed over a narrow beam, mitochondrial health markers, climbing attempts*, and the fact all the control mice were dead by 29mo, but the first telomerase mouse did not die until ~33mo and the last didn’t die until 40mo. That would be the human equivalent of the control group starting to die at about 60yo and all being dead by 80, and the telomerase group starting to die in their mid 80s and the last not dying till about 110 yo.

 

*follistatin gene therapy also improved appearance, beam test speed and mitochondrial health markers (but not climbing as they were too big).

 

If telomeres were mostly stopped from shortening in the telomerase group, why didn’t the mice live even longer?  Given telomere and mitochondrial markers were well preserved, is there another cause of death? There are a couple of shortcomings in the study before we can conclude that. Firstly, they only sacrificed mice once at 24mo to check telomere length. At that stage telomere length was well preserved in the telomerase therapy sacrifice (but severely reduced in control, See Fig F), and this explains why 5 months later all the controls were dead, but none of the telomerase groups were anywhere near dying. But telomeres were not measured again. The paper showed that one treatment boosted telomerase considerably, falling gradually back to baseline in about 25 days. But we don’t know the effect of multiple administrations. We don’t know if CMV continues to evade the immune system after many, many treatments. My guess would be that its effectiveness declines and telomeres do shorten even with repeated treatments. But we’ll have to wait for another study to find out if I'm right.

 

 

Very interesting stuff. QuestForLife, Thanks for keeping this thread going over the years.


  • Cheerful x 1

#614 dlewis1453

  • Member
  • 173 posts
  • 56
  • Location:USA
  • NO

Posted 13 July 2021 - 02:36 PM

Yes, thank you for sharing all of your meticulously compiled research and useful insights with us! I always look forward to updates from this thread.


  • Agree x 3

#615 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 14 July 2021 - 10:30 AM

Thank you for all the encouragement, it does make a difference.

 

I have a very big theory post coming soon. 

 

I think it explains most of aging: it will tie together telomeres, ROS, 'damage', MTOR, methylation, stem cells and cancer.


  • like x 5

#616 dlewis1453

  • Member
  • 173 posts
  • 56
  • Location:USA
  • NO

Posted 14 July 2021 - 01:54 PM

Thank you for all the encouragement, it does make a difference.

 

I have a very big theory post coming soon. 

 

I think it explains most of aging: it will tie together telomeres, ROS, 'damage', MTOR, methylation, stem cells and cancer.

 

Sounds great! Looking forward to the insights in this comprehensive theory post. 



#617 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 14 July 2021 - 02:51 PM

This post is going to serve as a kind of preface to the large theory of aging post I’ve promised soon.

 

I’ve written numerous times about how AKG is a cofactor in the TET demethylases and hence how taking AKG as a supplement can reduce methylation (and hence methylation age).

 

A bit of background: although methylation both increases (at CpG ‘islands’) and decreases ('globally') during aging, Horvath’s latest pan-species clock shows that the bulk of methylation changes common to all species with aging is methylation, not demethylation (https://doi.org/10.1...21.01.18.426733) Hence the benefit of increasing demethylation via more AKG.

 

Here is another piece of the puzzle: ROS decreases the activity of demethylases – at least those involved in histone demethylation – which has important consequences in terms of chromatin accessibility. It may also explain the benefit of Vitamin C in reprogramming.
The ROS theory of aging is not dead yet! 

 

 

Oxidative stress alters global histone modification and DNA methylation

 

The JmjC-domain-containing histone demethylases (JHDMs) can remove histone lysine methylation and thereby regulate gene expression. The JmjC-domain uses iron Fe (II) and α-ketoglutarate (αKG) as cofactors in an oxidative demethylation reaction via hydroxymethyl-lysine. We hypothesize that reactive oxygen species will oxidize Fe (II) to Fe (III), thereby attenuating the activity of JmjC-domain-containing histone demethylases. To minimize secondary responses from cells, extremely short periods of oxidative stress (3 hours) were used to investigate this question. Cells that were exposed to hydrogen peroxide (H2O2) for 3 hours, exhibited increases in several histone methylation marks including H3K4me3 and decreases of histone acetylation marks including H3K9ac and H4K8ac; pre-incubation with ascorbate attenuated these changes. The oxidative stress level was measured by generation of 2′, 7′-dichlorofluorescein (DCF), GSH/GSSG ratio and protein carbonyl content. A cell free system indicated H2O2 inhibited histone demethylase activity where increased Fe (II) rescued this inhibition. TET protein also showed a decreased activity under oxidative stress. Cells exposed to a low dose and long term (3 weeks) oxidative stress also showed increased global levels of H3K4me3 and H3K27me3. However, these global methylation changes did not persist after washout. The cells exposed to short term oxidative stress also appeared to have higher activity of class I/II histone deacetylase HDAC) but not class III HDAC. In conclusion, we have found that oxidative stress transiently alters epigenetic program process through modulating the activity of enzymes responsible for demethylation and deacetylation of histones.

Source: doi:10.1016/j.freeradbiomed.2015.01.028

 

 

I interpret this as meaning oxidative stress leads to DNA being coiled up defensively. I believe that this will not only apply to methylation of histones but also CpG islands; my results have shown AKG is effectively in reducing epigenetic age as measured on the islands.

 

Though they found the changes in methylation were temporary once the oxidative stress was removed, we know oxidative stress increases with age and hence this will be accompanied with greater methylation of the genome in the sites found by Horvath in his pan species clock. We know from the same Horvath paper  that these methylated sites include the promoters of many developmental genes, so are likely related to differentiation. I’ve written before about how aging can be considered a failure of differentiation, and I’ll be saying a lot more on this soon.


Edited by QuestforLife, 14 July 2021 - 02:54 PM.

  • Informative x 3
  • Good Point x 1
  • WellResearched x 1
  • like x 1

#618 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 15 July 2021 - 10:40 AM

I really wanted this post to be comprehensive and conclusive. I can feel the shape of the answer in my head, but there are so many puzzle pieces I can’t yet fully complete it. But it is still pretty good. In the places where I’ve indicated I am missing something, if you can help, please do. I really think that aging is close to being understood. I have never been more optimistic that aging will be solved, and soon. Now I need to get this out before I burst,.....

Please stick with me whilst I try and lay out all the puzzle pieces.

 

Let me summarise quickly:

 

  • Oxidant stress decreases TET enzymes, post #617 (above)
  • TET enzymes keep GDF11 promoter ‘clean’ of methylation so it can be expressed, post #424
  • Methylation (in women’s skin) of the oxytocin receptor contributes to skin aging, post #473
  • High ROS environment decreases TET activity and methylates klotho promoters, leading to kidney disease, post #486
  • Part jigsaw piece - Vital mitochondrial energy production is turned down via age related methylation changes! Remember that paper back in 2015 that showed simple glycine could reverse age related mitochondrial defects? Well the relevant nuclear gene in humans, SMHT2, is important for methylation (via making glycine for SAM) but also in making new mitochondrial DNA (via making N-formylmethionine, which is the starting amino acid in mitos and bacteria, not just plain methionine like in human DNA). Hence the SHMT2 gene is vital in every cell’s energy production and it is downregulated with age. I have not yet found a link to exactly how this happens, but we do know that Yamanaka factors reverse SHMT2 silencing and we know cell reprogramming doesn’t work without the TETs (as shown by David Sinclair). Much of what I know is from the following paper https://www.nature.c...cles/srep10434 

The above posts and references all show that age related methylation (which we know from the Horvath reference in the last post is much more prevalent in aging than demethylation) is causing the inactivation of important genes. Most interesting is GDF11, which is pro-differentiation. On that subject I have always been worried that molecules that cause increased differentiation, like retinol in skin cells or stem cell stimulants like AFA or C60, or GDF11 (even though it also increases telomerase), could cause stem cell exhaustion. But there has been growing evidence that much of aging is actually the reverse problem; stem cells are still there but they aren’t differentiating.

 

In a recent experiment they found that Loss of Dnmt3a (a de novo methyltransferase) Immortalizes Hematopoietic Stem Cells In Vivo, but that they would no longer differentiate! https://www.scienced...211124718303541

These HSCs could proliferate forever and never lost telomeres

Dnmt3aKO HSCs showed no erosion of telomere length

, but never produced any blood cells for the poor mice hosting them

 

 

Also from https://www.nature.c...rticles/ncb1386 , Blasco and friends showed,

Mouse embryonic stem (ES) cells genetically deficient for DNMT1, or both DNMT3a and DNMT3b have dramatically elongated telomeres compared with wild-type controls. Mammalian telomere repeats (TTAGGG) lack the canonical CpG methylation site. However, we demonstrate that mouse subtelomeric regions are heavily methylated, and that this modification is decreased in DNMT-deficient cells.

So telomerase is under methylation control.

 

 

We also see an increase in de novo methyltransferases with age, see; https://pubmed.ncbi....h.gov/17929180/. I don’t know if the demethylators (TETs) failing with age is related to the de novo methylators increasing with age., but I’m throwing it in here. Note cocoa decreases DNMTs (https://pubmed.ncbi.nlm.nih.gov/23840361/).

Moving back to telomeres, I spent a lot of time recently looking at how in both human and mice cells, differentiation downregulates telomerase production, see post #588. It seems likely that this is accomplished via DNMTs. But more importantly this might be the ‘selection pressure’ causing the whole of aging. My Selfish Cell theory of aging says that stem cells that differentiate die, whilst those that stay as stem cells live; see Post #122 and Post #130. You can see that even back then, I was on the right path.

Stress causes differentiation of progenitor cells or harder work in somatic cells . This is an intentional response to support tissues. But it will over time select for cells that are resistant to stress AND resistant to responding to that stress.  With age ever greater stress is required to renew tissues. Although this gives us some confidence that exercise, CR, IF, autophagy inducers, etc. will slow aging this has a practical limit. Other treatment options are required.

 

 

Allow me a metaphor: The body survives only because of the sacrifice of cells being willing to differentiate and potentially die, rather like a soldier being willing to die for his country. Once the country only has lazy, selfish people who put their needs above that of the country, it's days are surely numbered. But in times of bodily stress (like War for a country), the best tragically die first. Life is constant stress on the body that requires differentiation of cells. But differentiation turns down telomerase. We are literally training the cells of our body to do what those selfish HSCs cells with no DNMT3a did by human design - proliferate forever and refuse to differentiate. We are aging because we are slowly turning back into stem cells.

 

In post #476, I talked about how ‘aging is cancer’. This was a pretty ballsy statement. But the referenced study ‘DNA Methylation Patterns Separate Senescence from Transformation Potential and Indicate Cancer Risk’ https://www.scienced...535610818300084 showed that the cells of aging humans are more commonly like cancer cells than senescent cells, i.e. they bore methylation patterns that looked more like stem cells than cellular senescence. Going back to GDF11 for a moment, bear in mind that Steve Perry has shown GDF11 is robustly anti-cancer for some cancer types in dogs. I wouldn’t be surprised if GDF11 is generally anti cancer because it encourages proper differentiation of pre-cancerous or actual cancerous cells.

 

I speculate that the body takes more extreme measures to force stem cells to differentiate and replenish tissues as we age, and this might be behind the rise in the destructive hormones Jeff Bowles talks about so much (LH, FSH, etc). But even though this might be successful in causing continued differentiation, the cells produced are not phenotypically normal, and bear a pre-cancerous phenotype as shown in the above study. A skin cell is not fully a skin cell, a liver cell is not exactly a liver cell. I think of elevating growth signalling to replenish tissues from an unwilling stem cell pool to be like driving with both the accelerator and brake pedal pressed down (or driving with the handbrake on).

 

There is also the possibility that pro-oxidant like hormones (LH, FSH) actually cause a downregulation in the TETs via ROS, and this would fit with growth signals like MTOR being necessarily for growth and development from a child but later causing unwanted methylation of important differentiation genes, as we’ve already discussed. This is an area where I don’t have all the jigsaw pieces yet. It does link nicely with anti-oxidant hormones like melatonin being anti aging (see Post #472), whilst also downregulating sex drive, potentially delaying menopause etc (more Jeff Bowles stuff). At the moment my stance is the hormone elevation happens too late to be the initiator of aging, but it might deliver the coup de grace. Or it might be a gradually increasing feedback loop as the stem cell pool becomes dominated with ‘selfish cells’; it starts rising gradually and this accelerates until you literally destroy the tissues producing the hormones. 

 

An interesting consequence of growth signals forcing reticent stem cells to differentiate, is that this might cause the preservation of telomere length in the very old. As partially differentiated cells predominate, some telomerase expression may be preserved. See: https://www.ncbi.nlm...les/PMC4634197/
In this kind of study you see telomere length falling with age (it is only a cross sectional, so they aren’t the same people) and then rise in the centenarians that survive the longest. They then say telomeres are not important for aging. I always put this down to survivor bias, with those with long telomeres being those that survive to very old age, but maybe there is more to it and the very old have longer telomeres because telomerase is no longer being shut down in their partially-differentiated cells. Typically long telomeres and active telomerase are blamed for cancer by lazy researchers. But my work here shows another possibility, that cells are becoming more cancerous because of selection pressure on cells to be selfish, and this naturally preserves telomere function because of the lack of methylation-mediated downregulation that comes with proper differentiation.

 

That’s been a long post!

 

Conclusions:

 

  • The body needs constant replenishment and this is accomplished by differentiation from the stem cell pool into vital specialist roles (skin, liver, blood, vessel lining, etc.)
  • Differentiation can only occur if important developmental genes are kept demethylated. Elevated ROS can impede the demethylases (TETs) and cause these genes to be methylated, blocking differentiation and causing the cells that can be made to differentiate to have a stemlike/pre-cancerous phenotype.
  • Differentiation also causes downregulation or complete abolishment of telomerase, therefore eventual death via ROS/telomere attrition/both for that cell line (this downregulation of telomerase is accomplished by de novo methyltransferases DNMTs. Blocking DNMT3a caused blood stem cells to become immortal but never produce blood cells). DNMTs go up with age, possibly as a defence mechanism against rising pre-cancerous cell numbers. Longer telomeres in the very old might be evidence of telomerase not being properly turned off in spite of this.
  • The source of the rising ROS that disabled the TETs and stopped differentiation in the first place might be the pro-oxidant hormones (LH, FSH) required for growth from a child to an adult. (There are numerous other possible triggers that could start this off, which is why aging is so robust).
  • Because differentiation is basically a telomerase-blocking death sentence for cells (but a lifesaver for us) as time goes on ‘Selfish’ stem cells will come to dominate the reserve pool - these cells require stronger and stronger growth signalling to differentiate, and this might be behind destructive sex hormones rising in late middle age.

 

Personal Epilogue

 

I haven’t got time to write a whole lot on ‘what should we do about this’ given everything I’ve said. For now know I haven’t changed my view on telomerase activators (I use TAM818, also Vit D (+K2)in the winter, I also take B6/9 with it), anti-oxidants (telomerase protects mitochondria and lowers ROS, melatonin/epitalon is probably the best anti-oxidant; but I also like Vit C for demethylation and ROS, and beta-carotene plus Zinc for multiple reasons) or AKG (surely a godsend discovery for upregulating TETs, but requires cycling ), GDF11 (in small amounts is very useful for noticeable de-aging, particularly with AKG) and mTOR inhibitors to temporarily decrease proliferation and give telomeres a chance to lengthen (I mainly use coffee, occasionally rapamycin/everolimus - but I get side effects really easily); There are lots of other possible interventions like klotho, oxytocin, (good) sex hormones (that fall with age), stem cell stimulants like C60 and AFA. I’ve tried oxytocin and it didn’t seem additive with other things I’m doing. AFA works but can cause fatigue. I think C60 could be a winner, but I need to find a manufacturer I trust. Klotho is definitely high on my list. I’ve haven’t tried pregnenolone/DHEA etc. 

A personal note: mentally I feel as good, or better, than I ever have.  Maybe the closest I can recall to this is being a rather acerbic 16 year old with a ruthless intellect intent on criticising the world. Luckily the return of my mental acuity has not turned me back into a teenager. Physically I can work out every night if I want to now. My skin still looks older than I’d like, though considerably younger than many of my peers. My hair is full and dark. I have a little grey in my beard. I wonder whether the ‘damage’ theories of aging will still be relevant at some point - even if damage is due to aging and not the cause of aging, it is still damage and will not necessarily all disappear once full youthful homeostasis in the body is restored. I expect this will mainly be a problem for those who are already very old and consequently have a lot of unrepaired damage.


Edited by QuestforLife, 15 July 2021 - 10:52 AM.

  • Informative x 5
  • like x 2

#619 dlewis1453

  • Member
  • 173 posts
  • 56
  • Location:USA
  • NO

Posted 15 July 2021 - 02:21 PM

Great post! An excellent linking of multiple pieces of research and how they all interact with each other. Wouldn't it be funny if the first accurate comprehensive outline of the aging process was posted in a discussion thread on Longecity rather than a research paper? 

 

I will be reading through your post multiple times to try to understand everything. One initial question I have relates to the following: "Part jigsaw piece - Vital mitochondrial energy production is turned down via age related methylation changes!"

 

My question is whether Turnbuckle's mitochondrial protocol, which allegedly repairs mtDNA and reverses mtDNA methylation, would be of any benefit to this aspect of aging that you have described? 



#620 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 15 July 2021 - 03:29 PM

One initial question I have relates to the following: "Part jigsaw piece - Vital mitochondrial energy production is turned down via age related methylation changes!"

My question is whether Turnbuckle's mitochondrial protocol, which allegedly repairs mtDNA and reverses mtDNA methylation, would be of any benefit to this aspect of aging that you have described?

I didn't go into detail on the glycine deficit mediated mito decay talked about in the paper. But it is regulated by a NUCLEAR encoded gene (SHMT2). So demethylating mitochondrial DNA whilst you fission it (Turnbuckles idea, I believe) wouldn't work. However AKG will over time demethylate nuclear gene promoters too, so AKG should benefit mitochondria in the long run.

Also the problem they found in the paper was the inability to make new mitochondria, not its quality control (which involves breaking it down=mitophagy). To me it looks like mitochondrial decline is linked to methylation changes in the nucleus, the same as all other parts of aging. Even if we couldn't solve that bit yet,we can just take glycine. I've had some startling reports from folks in their 50-60s taking high dose glycine.

From my experience, the best energy boost I've ever had is not from mitochondrial supplements, but telomerase activators. My best guess is telomerase locates to mitochondria when needed.

...evidence has accumulated that telomere-independent functions of telomerase also exist and that the protein component of telomerase TERT shuttles between the nucleus and mitochondria under oxidative stress. source: https://www.frontier...2019.00274/full


Edited by QuestforLife, 15 July 2021 - 04:25 PM.

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

#621 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 15 July 2021 - 05:28 PM

From my experience, the best energy boost I've ever had is not from mitochondrial supplements, but telomerase activators. My best guess is telomerase locates to mitochondria when needed.


I looked into this a little more and found a mind blowing 2020 paper.

We determined that hTERT negatively regulates the cleavage and cytosolic processing of PINK1 and enhances its mitochondrial localization by inhibiting mitochondrial processing peptidase β (MPPβ). Consequently, hTERT promotes mitophagy...
source: https://www.nature.c...1419-020-2641-7


So my subjective experience of better energy and rapid recovery from exercise on telomerase activators (TAM818 or Epitalon) is probably due to enhanced mitophagy because telomerase both decreases PINK1 cleavage and helps locate it to depolarised mitochondria.
  • WellResearched x 3
  • Good Point x 2

#622 kurt9

  • Guest
  • 269 posts
  • 28

Posted 15 July 2021 - 05:58 PM

My question is whether Turnbuckle's mitochondrial protocol, which allegedly repairs mtDNA and reverses mtDNA methylation, would be of any benefit to this aspect of aging that you have described? 

 

I'm doing this right now. I'm on cycle 6. My gym workouts are a bit slow on fission days. But I can tell you I'm full of piss and vinegar on fusion days. I actually have to stay away from reading the news on fusion days. So, yeah. I think it works.
 


  • Informative x 1

#623 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 16 July 2021 - 08:19 AM

1. What is the reaction time you are aiming at with GDF11 dosing?

After a plateau with a 180-190s as a minimum time I started to get 140-170 (1 or 2 out of 5, and max can be around 230 so SD is rather big)

 

2. Do you have an opinion on how supplementing with B vitamins can influence methylation status? I am a homozygous for MTHFR and take Swansons Activated Bs everyday to keep my homocysteine low(as a marker of methylation pathway not going haywire).

Literature, for example A higher degree of LINE-1 methylation in peripheral blood mononuclear cells, a one-carbon nutrient related epigenetic alteration, is associated with a lower risk of developing cervical intraepithelial neoplasia (nih.gov) shows that at least peripheral blood cells are significantly more methylated when more folate+b12 present. Do you think its counterproductive? 

 



#624 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 16 July 2021 - 08:56 AM

1. What is the reaction time you are aiming at with GDF11 dosing?

After a plateau with a 180-190s as a minimum time I started to get 140-170 (1 or 2 out of 5, and max can be around 230 so SD is rather big)

 

2. Do you have an opinion on how supplementing with B vitamins can influence methylation status? I am a homozygous for MTHFR and take Swansons Activated Bs everyday to keep my homocysteine low(as a marker of methylation pathway not going haywire).

Literature, for example A higher degree of LINE-1 methylation in peripheral blood mononuclear cells, a one-carbon nutrient related epigenetic alteration, is associated with a lower risk of developing cervical intraepithelial neoplasia (nih.gov) shows that at least peripheral blood cells are significantly more methylated when more folate+b12 present. Do you think its counterproductive? 

 

I try and stay in the 200-220ms range (average of 5). Generally a good result for me is 200-210 but 210-220 is acceptable. If I trend towards the upper end for a while, I'll dose. I very rarely seem to average above 220ms now, but if I did for more than a day I would dose.  I tend to get occasional 170,180 or 190ms individual results, and occasional 230ms+ individual results, but my average is fairly stable now. I don't think a consistent average below 200ms is realistic; this is in accordance with the literature, which states 200ms is about as low as simple reaction time (with no options to choose between, which makes it slower) can get. If I do occasionally release early and get something ridiculously fast, I just restart. Morning is faster than evening, but can be more erratic, I find. After a big meal can also be slower. I don't have lots of attempts, one is ideal, unless it goes wrong for some reason. Before GDF11 I was much slower. I have attached a picture of my simple reaction times since last December. This is all for me, a relatively fit 42yo. People reading this might be slower or faster; the point is to honesty track your progress to look for changes. As you can see, mine dropped quite a lot and is now highly stable; I now dose GDF11 only once or twice a month at a very low dose.

 

I feel that B vitamins don't influence the methylation that comes with aging. I don't have evidence for this, but it appears that the demethylation required to differentiate, and the methylation required to differentiate (for example, to suppress telomerase), appear to be largely independent. If you have any other information on this I'd be interested. It might be something to do with the TETs and the DNMTs working independently on different targets. Decline in TETs definitely contributes to aging; I'm not sure if the rise in DNMTs does - it might be a consequence of aging (like suppressing pre-cancerous cells, as I said in my big Post # above). The short answer is if you need B vitamins to stay healthy, take them. It is like MTOR inhibition. You can suppress it for anti-aging, but you don't want to do it so much you live covered in bacterial colonies and infested with viruses. You can't live forever if you're ill.

Attached Thumbnails

  • Reaction times.png

Edited by QuestforLife, 16 July 2021 - 09:02 AM.

  • Informative x 2

#625 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 16 July 2021 - 09:18 AM

 

 

  Thank you!

  I have same observations that morning on a fasted state is optimal for getting low reaction times. TBH I ve probably trained myself to get to low numbers as it also includes some eye-hand coordination and sometimes I notice a moment of contemplation - ok its green, what to do now? (super fast but its still there) If I concentrate on making this decision beforehand and 'prime' my trigger finger I can get those 150s but its not guaranteed.

 

  I will try to read into the literature about B vitamins. at least its clear that low folate+B12 is highly detrimental with cancer and CVD risk going up so it doesnt look like I have much choice. )



#626 QuestforLife

  • Topic Starter
  • Member
  • 1,595 posts
  • 1,177
  • Location:UK
  • NO

Posted 16 July 2021 - 09:35 AM

More on melatonin

 

See Post #472 and #525 for my previous comments on this anti-oxidant hormone.

 

In this study they looked at if melatonin is made by a developing embryo (they found it was) and if and how that benefited that embryo (they found blocking production impeded embryonic development). As to the how, they found melatonin was made in the mitochondria and that melatonin reduced ROS and mutations under oxidative stress, so they speculate melatonin might protect the mitochondria and maintain ATP production.

 

The fact that the rate limiting step for melatonin production Aanat (aralkylamine N-acetyltransferase), was found to mostly localize in the mitochondria, suggests to me that mitochondrial dysfunction might impair melatonin production as we age.

 

Even more interestingly, they found that:

Aanat knockdown reduced tet methylcytosine dioxygenase 2 (Tet2) expression and DNA demethylation in blastocyst and melatonin supplementation rescued these processes

 

TET2 is one of the demethylases we've discussed before whose loss is central to the loss of cell differentiation potential and (probably) aging. In Post #424 we saw TET2 was involved in GDF11 expression via demethylating its promoter.

We don’t yet know exactly why melatonin production falls with age, but this paper hints that it might be damage to mitochondria affecting its rate limiting step of production.  In any case, melatonin supplementation should increase TET2 and benefit GDF11 levels. . 

 

Aanat Knockdown and Melatonin Supplementation in Embryo Development: Involvement of Mitochondrial Function and DNA Methylation

Aims: In addition to pineal gland, many cells, tissues, and organs also synthesize melatonin (N-acetyl-5-methoxytryptamine). Embryos are a group of special cells and whether they can synthesize melatonin is still an open question. However, melatonin application promoted embryo development in many species in in vitro condition. The purpose of this study was to investigate whether embryos can synthesize melatonin; if it is so, what are the impacts of the endogenously produced melatonin on embryo development and the associated molecular mechanisms. These have never been reported previously. Results: Melatonin synthesis was observed at different stages of embryonic development. Aanat (aralkylamine N-acetyltransferase), a rate-limiting enzyme for melatonin production, was found to mostly localize in the mitochondria. Aanat knockdown significantly impeded embryonic development, and melatonin supplementation rescued it. The potential mechanisms might be that melatonin preserved mitochondrial intact and its function, thus providing sufficient adenosine 5'-triphosphate for the embryo development. In addition, melatonin scavenged intracellular reactive oxygen species (ROS) and reduced the DNA mutation induced by oxidative stress. In the molecular level, Aanat knockdown reduced tet methylcytosine dioxygenase 2 (Tet2) expression and DNA demethylation in blastocyst and melatonin supplementation rescued these processes. Innovation: This is the first report to show that embryos synthesize melatonin, and its synthetic enzyme Aanat was located in the mitochondria of embryos. An effect of melatonin is to maintain Tet2 expression and normal methylation status, and thereby promote embryonic development. Conclusion: Embryos can produce melatonin that reduces ROS production, preserves mitochondrial function, and maintains Tet2 expression and the normal DNA methylation.
Source: https://pubmed.ncbi....h.gov/30343588/

 


Edited by QuestforLife, 16 July 2021 - 09:43 AM.

  • Informative x 3
  • like x 1

#627 Castiel

  • Guest
  • 374 posts
  • 86
  • Location:USA

Posted 17 July 2021 - 04:20 PM

 

 

We also see an increase in de novo methyltransferases with age, see; https://pubmed.ncbi....h.gov/17929180/. I don’t know if the demethylators (TETs) failing with age is related to the de novo methylators increasing with age., but I’m throwing it in here. Note cocoa decreases DNMTs (https://pubmed.ncbi.nlm.nih.gov/23840361/).
 

  • The body needs constant replenishment and this is accomplished by differentiation from the stem cell pool into vital specialist roles (skin, liver, blood, vessel lining, etc.)
  • Differentiation can only occur if important developmental genes are kept demethylated. Elevated ROS can impede the demethylases (TETs) and cause these genes to be methylated, blocking differentiation and causing the cells that can be made to differentiate to have a stemlike/pre-cancerous phenotype.
  • Differentiation also causes downregulation or complete abolishment of telomerase, therefore eventual death via ROS/telomere attrition/both for that cell line (this downregulation of telomerase is accomplished by de novo methyltransferases DNMTs. Blocking DNMT3a caused blood stem cells to become immortal but never produce blood cells). DNMTs go up with age, possibly as a defence mechanism against rising pre-cancerous cell numbers. Longer telomeres in the very old might be evidence of telomerase not being properly turned off in spite of this.
  •  

 

Was researching the DNMTs and resveratrol and found the following

 

 

Resveratrol Restores LINE-1 Methylation Levels by Modulating SIRT1 and DNMTs Functions in Cellular Models of Age-Related Macular Degeneration

Resveratrol Restores LINE-1 Methylation Levels by Modulating SIRT1 and DNMTs Functions in Cellular Models of Age-Related Macular Degeneration[v1] | Preprints

 

 

 

 

Further, we evaluate the literature supporting the potentiation of HDAC inhibitors and the inhibition of DNMTs by resveratrol in different human cancers. 

Differential Methylation and Acetylation as the Epigenetic Basis of Resveratrol’s Anticancer Activity (nih.gov)

 

resveratrol activities decrease with age due to NAD+ depletion I hypothesize.(for example telomerase activation far greater in young, than old.).    What would happen if resveratrol was given along with NAD+ boosters like cd38 inhibitors or NMN?


Edited by Castiel, 17 July 2021 - 04:28 PM.

  • like x 1

#628 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 17 July 2021 - 07:19 PM

Was researching the DNMTs and resveratrol and found the following

 

 

Differential Methylation and Acetylation as the Epigenetic Basis of Resveratrol’s Anticancer Activity (nih.gov)

 

resveratrol activities decrease with age due to NAD+ depletion I hypothesize.(for example telomerase activation far greater in young, than old.).    What would happen if resveratrol was given along with NAD+ boosters like cd38 inhibitors or NMN?

 

 IDK, to me resveratrol looks like a very "dirty" drug, it influences a lot of things at different concentrations and not all of them beneficial.

https://www.google.c...jc0IfN3xZlA6_Lm

 

If majority of the beneficial effects are due to SIRT1 activation than oleic acid should be a first choice 

David A. Sinclair on Twitter: "@dday247 Yep! Oleic acid is a potent activator of Sirt1 and found in olive oil and avocados (and in the blood of people who are fasting.)" / Twitter


  • Informative x 1
  • WellResearched x 1
  • Agree x 1

#629 Castiel

  • Guest
  • 374 posts
  • 86
  • Location:USA

Posted 17 July 2021 - 09:46 PM

 IDK, to me resveratrol looks like a very "dirty" drug, it influences a lot of things at different concentrations and not all of them beneficial.

https://www.google.c...jc0IfN3xZlA6_Lm

 

If majority of the beneficial effects are due to SIRT1 activation than oleic acid should be a first choice 

David A. Sinclair on Twitter: "@dday247 Yep! Oleic acid is a potent activator of Sirt1 and found in olive oil and avocados (and in the blood of people who are fasting.)" / Twitter

resveratrol also activates other sirtuins(including sirt4, through which it appears lengthens telomeres significantly, another way it lengthens telomeres appears to be through activation of splicing factors rejuvenating old human cells), and foxo3, and master regulator gene nrf-2, which activates hundreds of antioxidant and detox genes protecting against all manner of issues

 

The Master Protector Gene: How to Trigger It - YouTube

 

Not only that, but it has lengthened lifespan of yeast by 70%, of fish, of mitochondrial mutant mice, of senescent accelerated mice, of normal aging but short lived mice, and various other species.

 

So far toxic doses, appear to exceed those available in any supplement, and appear to be more a scare tactic from big pharma.   As resveratrol is up there with melatonin, vitamin d, and vitamin c.

 

Perhaps oleic acid also activates everything resveratrol does, after all Sinclair said resveratrol appeared to be mimicking another naturally occuring molecule.   I've already heard rumors oleic acid activates multiple sirtuins, olive oil also has hydroxytyrosol, that some say may protect vitamin c levels in the body, but according to Bill Sardi may be able to act as a molecular aid to allow transcription of final vitamin C synthesis.   Resveratrol itself has apparently similar function for another gene able to correct an actual mutation, iirc.   But it remains to be seen if Sardi's claims hold up to scrutiny.

 

Vitamin C defect is major issue in aging humans, many negligible senescent animals have vitamin c synthesis, low vs high vitamin c intake in mutant mice similar to humans leads to vast lifespan increase in high c case compared to low c.

 

You'd likely need to take c every 4-6 hours at least to deal with C mutation defect of humans.   Or take extra virgin olive oil or a hydroxytorosol supplement and that might correct mutation defect if Bill Sardi's correct.

 

Even as early as 7 years of age humans already showing arterial lesions, vit c is critical, without adequate c guinea pigs with similar mutation get clogged arteries.

 

Heart Disease Starts at Age 7 - YouTube

 

edit:

With that said there is debate about optimal resveratrol dose.   Some say 25mg, others 100mg, others like Sinclair appear to espouse 1000mg.   There's also the issue that consuming with fat allows like 5x higher absorption.  And black pepper, iirc boost time in body by like 5-10~x iirc.


Edited by Castiel, 17 July 2021 - 09:51 PM.

  • Good Point x 1
  • like x 1

#630 rodentman

  • Guest
  • 208 posts
  • 44

Posted 17 July 2021 - 11:39 PM

With that said there is debate about optimal resveratrol dose.   Some say 25mg, others 100mg, others like Sinclair appear to espouse 1000mg.   There's also the issue that consuming with fat allows like 5x higher absorption.  And black pepper, iirc boost time in body by like 5-10~x iirc.

 

I wonder why he takes it in the morning with yogurt, and 'breaks' his fast.  Than never made sense to me.


  • Good Point x 1





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

66 user(s) are reading this topic

0 members, 66 guests, 0 anonymous users