• 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

#691 QuestforLife

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

Posted 27 September 2021 - 10:32 AM



Couldn't taking something like the Piperlongumine during HBOT help by eliminating cells which have weaker anti-oxidative capability?

 

Wouldn't that potentially enhance the hormetic response?

 

Assuming that the benefits of HBOT are in fact due to hormesis in response to oxidative stress, piperlongumine would tip the redox balance even further into oxidative territory. This might prove beneficial (again assuming the theories on the benefits of HBOT are correct,) unless of course the oxidative stress then goes beyond the hormetic limit, tipping over into outright harm.

 

I am conscious that we are being herded into a position whereby clearing or killing cells is 'good'. This might be the case in some circumstances, or it might be bad. Piperlongumine  has been noted for potential senolytic effects (Link: https://doi.org/10.1111/acel.12780)

It has also been touted as a cancer treatment.

 

But we should be cautious. Piperlongumine may have immunosuppressive effects, atleast according to this paper (I do not know whether this would occur in vivo, but it seems quite likely). I suspect that those who are playing with senolytics are careful. But I mention it anyway.

 

 

 Piperlongumine (PL), a natural small molecule derived from the Piper longum Linn plant, has received growing interest as a prooxidative drug with promising anticancer properties. Yet, the influence of PL on primary human T cells remained elusive. Knowledge of this is of crucial importance, however, since T cells in particular play a critical role in tumor control. Therefore, we investigated the effects of PL on the survival and function of primary human peripheral blood T cells (PBTs). While PL was not cytotoxic to PBTs, it interfered with several stages of T cell activation as it inhibited T cell/APC immune synapse formation, co-stimulation-induced upregulation of CD69 and CD25, T cell proliferation and the secretion of proinflammatory cytokines. PL-induced immune suppression was prevented in the presence of thiol-containing antioxidants. In line with this finding, PL increased the levels of intracellular reactive oxygen species and decreased glutathione in PBTs. Diminished intracellular glutathione was accompanied by a decrease in S-glutathionylation on actin suggesting a global alteration of the antioxidant response. Gene expression analysis demonstrated that TH17-related genes were predominantly inhibited by PL. Consistently, the polarization of primary human naïve CD4+ T cells into TH17 subsets was significantly diminished while differentiation into Treg cells was substantially increased upon PL treatment. This opposed consequence for TH17 and Treg cells was again abolished by thiol-containing antioxidants. Taken together, PL may act as a promising agent for therapeutic immunosuppression by exerting prooxidative effects in human T cells resulting in a diminished TH17 but enhanced Treg cell differentiation. source: https://www.frontier...2020.01172/full


  • Good Point x 1
  • like x 1

#692 QuestforLife

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

Posted 30 September 2021 - 07:37 AM

Why long telomeres won’t make you live forever, but short telomeres mean you’ll die young

 

Longer telomeres permit longer survival of precursor cell lines, which permits the accumulation of random methylation on CpG islands in the vicinity of gene promoters[1]. The methylation state of such islands in blood, urine and saliva samples have been used for some years to very accurately predict human age[2]. This random methylation then forms the basis of selection, with a bias emerging for cells that do not differentiate (from a stem cell), but self-renew instead. Evidence supporting this has been found by Horvath et al using his most recent pan-species clocks[3]. This means that with age stem cells and other long lived progenitors are selecting for their own immortality, which ironically increases the mortality of the organism through a reduction in somatic tissue maintenance. An example of this occurring through human design, is the lab engineered downregulation of DNMT3a in murine hematopoietic cells; such cells maintained long telomeres, were functionally immortal permitting repeated transplantation, but produced no new blood cells for their host mice[4]. Therefore attempts to forestall organism aging through the reactivation of telomerase in the adult may increase tissue renewal for a time, but ultimately will fall prey to the same survival of the ‘selfish cell’ that does not help the body.

 

If this random methylation could be prevented, or rolled back through an upregulation of demethylation with respect to methylation (increasing TETs and/or decreasing DNMTs, for example), lack of telomerase activity and short telomeres in human stem cell lines[5] would then lead to the exhaustion of stem cell pools and failure of tissue renewal by a different mechanism.

 

A third option might be that increasing stress signals from the aging body (via inflammation, for example), might persuade self-renewing stem cells to partially differentiate. Unfortunately, this could (and possibly does[1]) populate the body with cells of a pre-cancerous phenotype. Hence aging is not merely tissue atrophy, but is also priming tissue for transformation.

 

Is there a solution to these twin dooms? Possibly re-activation of telomerase coupled with an upregulation of demethylation might both increase stem cell proliferative ability, and maintain proper differentiation. Objections have been raised to the reactivation of telomerase as being oncogenic. But the above argument suggests that demethylation would prevent the emergence of cells primed for oncogenic transformation, even in the presence of telomerase. In addition, active telomerase would prevent the erosion of telomeres that results in aneuploidy, a key step in cancer progression.

 

References
[1] DNA methylation patterns separate senescence from
transformation potential and indicate cancer risk, doi:10.1016/j.ccell.2018.01.008
[2] DNA methylation age of human tissues and cell types,  https://doi.org/10.1...2013-14-10-r115
[3] Universal DNA methylation age across mammalian tissues, doi: https://doi.org/10.1...21.01.18.426733
[4] Loss of Dnmt3a Immortalizes Hematopoietic Stem Cells In Vivo, doi:10.1016/j.celrep.2018.03.025.
[5] Differential cis-regulation of human versus mouse TERT gene expression in vivo: Identification of a human-specific repressive element, www.pnas.orgcgidoi10.1073pnas.0508964102


  • Informative x 1
  • WellResearched x 1

#693 QuestforLife

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

Posted 30 September 2021 - 08:10 AM

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

 

So many posts to add to the contents page! Posts since the last update highlighted in red.

 

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 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
New intranasal and injectable gene therapy for healthy life extension
https://www.longecit...-21#entry907730
New GDF11 telomerase paper in Nature:
Growth differentiation factor 11 attenuates cardiac ischemia reperfusion injury via enhancing mitochondrial biogenesis and telomerase activity

https://www.longecit...-21#entry907939
Telomerase increases mitophagy through PINK1 - explanations for my increased exercise tolerance
https://www.longecit...-21#entry908174
Polymorphic tandem DNA repeats activate the human telomerase reverse transcriptase gene
https://www.longecit...-22#entry908442
Telomere length and telomerase activity in T cells are biomarkers of high performing centenarians
https://www.longecit...-23#entry909206
Caffeine promotes the expression of telomerase reverse transcriptase to regulate cellular senescence and aging
https://www.longecit...-23#entry909654
Are the oncogenic effects of telomerase mediated by methyl transferases?
https://www.longecit...-23#entry910058

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
Oxidative stress alters global histone modification and DNA methylation
https://www.longecit...-21#entry908138
How non-differentiating Selfish (stem) cells come to dominate the stem cell pool; links between methylation, telomerase and ROS
https://www.longecit...-21#entry908160
Finding the Culprit: the hormones required for sexual maturity may be the trigger that starts aging via downregulation of TET2
https://www.longecit...-22#entry908266
Discussion over whether methylation of gene promoters is protective against stem cell loss and the counter evidence: immortalised cells accumulate such methylation
https://www.longecit...-22#entry908322
Discussion of the combined use of telomerase activators, GDF11, AKG, vit A and C
https://www.longecit...-22#entry908398
Summing up the Twin Evils of aging
https://www.longecit...-22#entry908458
The Evolution of the Selfish Cell
https://www.longecit...-23#entry909231
Cancer and the Selfish Cell
https://www.longecit...-23#entry909457
Does the Selfish Cell imply programmed or accidental aging?
https://www.longecit...-23#entry909533
Finding the Culprit II: Species' cellular ROS level sets aging rate via down regulation of demethylases and failure of Circadian Rhythm
https://www.longecit...-23#entry909696
How does the Selfish Cell affect post-mitotic cells?
https://www.longecit...-23#entry909914
Why long telomeres won’t make you live forever, but short telomeres mean you’ll die young
https://www.longecit...-24#entry910244

 

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
Discussion of reaction times on GDF11
https://www.longecit...-21#entry908189

 

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
Melatonin is linked to mitochondrial function and increases TET2 production
https://www.longecit...-21#entry908191
Discussion starting here on reversing thymic involution
https://www.longecit...-23#entry909243


Edited by QuestforLife, 30 September 2021 - 08:12 AM.

  • Informative x 2
  • like x 2
  • Well Written x 1
  • WellResearched x 1

sponsored ad

  • Advert

#694 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 11 October 2021 - 06:31 PM

Update on the effect of AKG on epigenetic age 

Continuing epigenetic self-tests posted here, I took 500 mg arginine alpha-ketoglutarate (AAKG) 2x/day for 22 days, followed by 1000 mg AAKG + 15000 IU vitamin A + 400 mg liposomal vitamin C 2x/day for 12 days. The resulting decrease in epigenetic age appears roughly consistent with Questforlife’s experience (post #594). 

 

AAKG made me feel tired and depressed at a 1000 mg/dose. Depressive feelings may occur with some people probably due to elevated glutamate, as reported here and in multiple publications. In the future, I plan to limit AAKG to 500 mg/day.

 

Test Date         (Epigenetic age - Chronological age), years

12/17/2020.... -10.45      Baseline before epigenetic treatments

02/17/2021.... -10.32      Turnbuckle stem cell protocol every 10 days (6 times total) with 120 mg gotu kola. During this period I did 1 fasting mimicking diet.

04/13/2021.... -12.58      Turnbuckle stem cell protocol every 10 days (6 times total).

06/08/2021.... -13.05      Autophagy (1 time) - for 5 days took 2 tsp/day of liposomal trehalose + 250 mg centrophenoxine 2x/day + 20 mg astaxanthin 2x/day.  Mitophagy (1 time) - for 7 days took 500 mg Urolithin A 1x/day + 20 mg astaxanthin 2x/day. Separately I did 1 fasting mimicking diet.

09/22/2021.... -15.3      500 mg AAKG 2x/day for 22 days, followed by 1000 mg AAKG + 15000 IU vitamin A  + 400 mg liposomal vitamin C 2x/day for 12 days.


  • Informative x 3
  • WellResearched x 1
  • like x 1

#695 QuestforLife

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

Posted 12 October 2021 - 10:25 AM

 

AAKG made me feel tired and depressed at a 1000 mg/dose. Depressive feelings may occur with some people probably due to elevated glutamate, as reported here and in multiple publications. In the future, I plan to limit AAKG to 500 mg/day.

 

 

Very interesting. Since I made my own liposomal Vitamin C and started taking that with my AKG the depressive symptoms have been significantly reduced. I now discover Vitamin C can protect against excess glutamate; it is proposed that Vit C can reduce the responsiveness of the NMDA receptor to glutamate [1]. 

 

When I finished my liposomal Vit C I decided for the next batch to also add glycine, in the hope this would increase glutathione production and further elevate my Vit C levels (in order to increase TET activity - see my previous posts). Whether or not this worked I can't say for sure, but it further reduced my AKG symptoms. There are other mechanism besides increasing Vit C that could be in play here. For example, glycine acts like GABA as an inhibitory neurotransmitter (as others have suggested for Taurine), which would counteract the effects of more glutamate [2].

 

But as this glutamate problem doesn't affect everybody there must be a further, possibly genetic, explanation. I supplement B6 because a 23andme test showed both the copies of the gene I inherited from my parents for synthesising B6 endogenously are mutated and function poorly. I previously found this is responsible for slow histamine processing and contributes to my hay fever (another story). I now find that B6 is an essential cofactor in the conversion of the (excitation inducing) glutamate to (the inhibitory) GABA [3]. Thus I may need to increase my B6 supplementation.

 

[1] https://www.ncbi.nlm...les/PMC2649700/

 

[2] https://www.frontier...2018.00317/full

 

[3] https://journals.plo...al.pone.0157466



#696 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 12 October 2021 - 12:55 PM

When I finished my liposomal Vit C I decided for the next batch to also add glycine, in the hope this would increase glutathione production and further elevate my Vit C levels (in order to increase TET activity - see my previous posts). Whether or not this worked I can't say for sure, but it further reduced my AKG symptoms. 

 

What amount of glycine might counteract  depressive symptoms caused by 1000 mg AKG?

What amounts did you try and to what extent were your symptoms relieved?



#697 QuestforLife

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

Posted 12 October 2021 - 01:52 PM

What amount of glycine might counteract  depressive symptoms caused by 1000 mg AKG?

What amounts did you try and to what extent were your symptoms relieved?

 

Short answer: Glycine 2.5g x2/day, relief of symptoms 75%+.

 

There are some caveats however. AKG seems to wear me down over time, and this latest experiment is not yet 2 weeks in. The glycine benefit is also compounded on top of the benefits I got from liposomal Vit C.

 

I'll give you the complete set of interventions so you can draw your own conclusions.

 

Previously I used only AKG, with only occasional use of Vitamin C and no Vitamin A. The last AKG dosing period terminated at the start of June with the usual symptoms (but favourable methylation results as posted here).

 

I restarted AKG at the start of August this time with Vitamin C and Vitamin A and continued until mid Sept. During this period I took breaks from all supplements at weekends. Since the halfway mark I used started liposomal Vitamin C (approx. 7g x2 day) and the first (AM) of those doses was concurrent with my AKG dose. Lower energy/sex drive was ameliorated but I still terminated the experiment in mid Sept due to an dip in energy levels.

 

I continued taking liposomal vitamin C and felt better and better, and began to experiment with occasional larger doses (~2g) of AKG without side effects.

 

When my liposomal Vit C ran out I made a new mix, this time time including glycine, 2.5g x 2day (the same dose of Vitamin C as before). I restarted AKG at 500mg x2 day along with this new mix a couple of weeks ago, without any breaks, and have not had any symptoms so far. We will see how this holds up. 

 

I don't believe Glycine needs to be part of the liposomal formulation, as it is easily dissolved in hot water, tea or coffee. When I took much higher doses in the past as part of a completely different experiment unrelated to AKG (30g+/day) it did eventually cause digestive distress however. 

 

I have another methylation age result to come in soon. I have previously found you can take big breaks (2 months off, 1 month on) from AKG and still get improved results. But I'd like to know if I can improve my results still further, so will be experimenting in the coming months with larger doses of AKG to discover what the optimum strategy is.

 

You seem to be pushing the envelope with -15 years methylation age Fafner55. Have you experienced any tangible benefits like improvements in appearance or exercise tolerance?



#698 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 12 October 2021 - 03:02 PM

 

You seem to be pushing the envelope with -15 years methylation age Fafner55. Have you experienced any tangible benefits like improvements in appearance or exercise tolerance?

 

I have a blood panel scheduled for later this week and will report any significant results.

 

A couple of years regression in epi-age shouldn't make much difference.  Anecdotally, exercise tolerance is the same and my skin seems marginally better (smoother, less crepiness). 

As for the 15 year difference between my chronological and biological ages, there might be an association between that and my overall good health, but I can't offer anything in the way of proof.



#699 dlewis1453

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

Posted 13 October 2021 - 04:09 PM

 

As for the 15 year difference between my chronological and biological ages, there might be an association between that and my overall good health, but I can't offer anything in the way of proof.

 

Hi Fafner, 

 

If I am reading your earlier post correctly, it appears that your  epi-age baseline before beginning anti-aging interventions was -10 years?! That is a very impressive result. I'm curious about your lifestyle and if you are very healthy compared to your peers. 



#700 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 13 October 2021 - 07:15 PM

Hi Fafner, 

 

If I am reading your earlier post correctly, it appears that your  epi-age baseline before beginning anti-aging interventions was -10 years?! That is a very impressive result. I'm curious about your lifestyle and if you are very healthy compared to your peers. 

 

There is nothing special about my lifestyle. When the weather permits, I might walk 6k to 8k steps / day for exercise. I don't pay much attention to my diet except for avoiding sugary drinks.

 

As for the -10 years, I attribute most of that to clearance of senescent cells, some to the Turnbuckle protocol and some to occasional fasting mimicking diets. Of the many interventions I tried over the years, those gave the most benefit. You can find a summary of my self-experiments in the "About Me" section of my profile.

 

Genetics might work in my favor, not in the sense that there is anything special but rather that there is nothing particularly wrong. My parents are in their 90's and my grandparents lived into that decade of life.


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

#701 QuestforLife

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

Posted 15 October 2021 - 08:48 AM

Defining the steps that lead to cancer - Reinhard Stindl

 

I highlight this genius paper as the next chapter in my cancer series. Stindl’s underappreciated paper explains the vast majority (>80%) of age-related cancer (carcinomas) as being caused by aneuploidy, which is a change in the number or structure of chromosomes. This is caused by telomere exhaustion, which explains cancer’s prevalence with age or repeated specific tissue injury (smoking, alcoholism, etc). In his model, cancer is driven by the forced proliferation of telomere exhausted stem cells, leading to aneuploidy. Aneuploidy then means differentiation signals mediated via methylation (or other mechanisms) cannot work (genes are not in the right place anymore), so stem cells get arrested at an immature (non-differentiated) state. Growth signals can then drive cancer, which is the replication and spread of non-differentiated cells (into various tissues, as stem cells do).

 

This paper has a lot in common with my Selfish Cell model. The stem cells that have depleted telomeres either enter senescence or are forced to proliferate by growth signals (via mTOR, etc.) and can cause cancer (Stindl’s model). Those stem cells that still have sufficient telomeres acquire random methylation and the methylation that increases the likelihood of the cell staying at an immature (undifferentiated) stage are those selected for (my model). This too will increase cancer likelihood, if growth signals drive selfish cells to replicate, but the difference is that selfish stem cells have a normal karyotype; their genes are still in the right place. This means that demethylation (or other mechanisms) can still force them to differentiate. Hence my approach – AKG, GDF11, etc.

 

But if the cancer cells have arisen via telomere exhaustion, they will not have a normal karyotype and demethylation will do nothing to make them differentiate. Stindl speaks of finding ways of making aneuploid cells differentiate. But I don't know of any method to do this at this point. Hence the importance of the telomere elongation part of my strategy. It is incredibly important stem cells don’t get short telomeres.

 

Once the cancer is aneuploid however, it requires a completely different strategy, one based around suppressing growth signals and killing cancer cells. But we must be cautious here that killing cancer cells does not cause more tissue exhaustion, otherwise cancer is bound to arise once more by the mechanism Stindl lays out. There is perhaps a lesson here for senolytics.

 

 

Recently, an influential sequencing study found that more than 1700 genes had non-silent mutations in either a breast or colorectal cancer, out of just 11 breast and 11 colorectal tumor samples. This is not surprising given the fact

that genomic instability is the hallmark of cancer cells. The plethora of genomic alterations found in every carcinoma does not obey the ‘law of genotype–phenotype correlation’, since the same histological subtype of cancer harbors different
gene mutations and chromosomal aberrations in every patient. In an attempt to make sense out of the observed genetic and chromosomal chaos in cancer, I propose a cascade model. According to this model, tissue regeneration depends on the
proliferation and serial activation of stem cells. Replicative telomere erosion limits the proliferative life span of adult stem cells and results in the Hayflick limit (M1). However, local tissue exhaustion or old age might promote the activation
of M1-deficient tissue stem cells. Extended proliferation of these cells leads to telomere-driven chromosomal instability and aneuploidy (abnormal balance of chromosomes and/or chromosome material). Several of the aforementioned steps
have been already described in the literature. However, in contrast to common theories, it is proposed here that the genomic damage blocks the epigenetic differentiation switch. As a result of aneuploidy, differentiation-specific genes
cannot be activated by modification of methylation patterns. Consequently, the phenotype of cancer tissue is largely determined by the epigenetic maturation arrest of tissue stem cells, which in addition enables a fraction of cancer cells to
proliferate, invade and metastasize, as normal adult stem cells do. The new model combines genetic and epigenetic alterations of cancer cells in one causative cascade and offers an explanation for why identical histologic cancer types
harbor a confusing variety of chromosomal and gene aberrations. The Viennese Cascade, as presented here, may end the debate on if and how ‘tumor-unspecific’ aneuploidy leads to cancer. Source: doi:10.1016/j.mehy.2008.01.010 

 

 

1-s2.0-S0306987708000200-gr1.jpg

 

 

 

 


Edited by QuestforLife, 15 October 2021 - 08:52 AM.

  • WellResearched x 3
  • Informative x 1
  • like x 1

#702 QuestforLife

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

Posted 21 October 2021 - 09:35 AM

My most recent Trume epigenetic age results are disappointing. I have regressed from -6.4 to -3.9 years relative to my chronological age. You can see a plot of all my results below. In addition to the latest result, I have also gone back and added the four tests I did from 2017-2020 using MyDNAge (a more expensive but more accurate test, which gives results comparable to Trume Age, compare Feb-20 to June-20). 

 

The main additions during this recent period were vitamin A supplementation and regular (previously it was occasional,) Vitamin C supplementation (liposomal for part of the period). Based on my research both these vitamins should have increased the effectiveness of AKG at reversing epigenetic age. But testing has revealed they haven’t helped.

 

The accuracy of the Trume Age test is stated as being better than 5 years, not accurate enough for me to draw the conclusion that these additions were harmful, but I can be reasonably certain that any benefit, if any, was minor. For now, I must conclude my normal physiological levels of Vitamin A and C were sufficient and increasing them further had no benefit.

 

Taking a step back and looking at my results in the round, along with my notes of the changes I made, I have a wealth of information giving me some hints about what might be effective and what has no effect or is counterproductive.

 

• Rapamycin taken weekly at 2-3mg has no effect on epigenetic age when taken for a year (2017-2018)
• A low dose intermittent cycle (1 month) of a statin and a sartan repeated twice a year, reduces epigenetic age by a few years (2018-2019).
• Epitalon (a telomerase activator) increases epigenetic age (but decreases telomere age) when taken cyclically (for a week, twice or thrice yearly) (2019-2020).
• AKG at 900mg/day is effective at decreasing epigenetic age, when taken for 6 months (Feb- Sept 2020); the lack of benefit at 3 months may be due to the limited accuracy of the test.
• Cycling AKG (2 months off, 1 month on) is still effective (Jan-21).
• Berberine appears to decrease epigenetic age by a further couple of years in combination with AKG (despite reports to the contrary); every time I remove it I get a worse result, bringing it back in my results improve (compare Sept-20 OFF, Jan-21 ON, Mar-21 OFF, May-21 ON, Sept-21 OFF).
• GDF11 didn’t decrease epigenetic age taken approximately weekly for 3 months (Mar-21), although it did improve various biomarkers (BP, HRV, reaction time).
• TAM818 (a telomerase activator), didn’t increase my epigenetic age like epitalon did – in fact my results after 3 months of use improved (May-21), although it is possible continued use for another 3 months did contribute to the deterioration in my recent result (Sept-21). TAM818 did improve my exercise tolerance.

 

Future plans

 

I am acutely aware I shouldn’t draw conclusions based on 3 months interventions with a test only accurate to about 5 years. I am tempted to change very little for the next 3 months. But even using B6/Taurine/Glycine (as recently discussed) I still don’t enjoy being on AKG for long periods. So, I will likely follow the 2 months off, 1 month on protocol. I may experiment with higher occasional doses, rather than daily ones. I will certainly re-introduce Berberine and see if I am correct about its benefits. In the absence of an available telomere test it is difficult for me to decide whether I need a break from telomerase activators so I will likely continue TAM818 until the bottle is finished.

Attached Thumbnails

  • trume Sept-21.png

Edited by QuestforLife, 21 October 2021 - 09:51 AM.

  • Informative x 1
  • like x 1

#703 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 21 October 2021 - 11:09 AM

QuestforLife,

Glycine is described as the main inhibitory neurotransmitter for the brainstem and spinal cord, while GABA is the main inhibitory neurotransmitter for the brain. One can hypothesize that GABA might be more effective than glycine to counter the excitatory effects of glutamate downstream of AKG, but I don't know the dose.


  • Informative x 1

#704 dlewis1453

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

Posted 21 October 2021 - 03:59 PM

My most recent Trume epigenetic age results are disappointing. I have regressed from -6.4 to -3.9 years relative to my chronological age. 

 

I understand why this is disappointing, but when I look at the chart you attached, it doesn't seem that bad.  I imagine that epigenetic age fluctuates normally, and then when you add the measurement error from the Tru-me test on top of that, it is hard to know where you really stand at any given point of time. I think the trend over several measurements is the most important thing. Still, its natural for us to want good results from every test. 

 

Perhaps your liposomal vitamin C consumption has increased telomerase more than you expect, with a resulting slight increase in epigenetic age (similar to your experience with Epitalon). Liposomal vitamin c is potent stuff, and you have been consuming I believe 4 grams of it per day. Assuming this is the case, then shouldn't continued cycles of AKG and GDF11 eventually bring the epigenetic age back down? 


  • Agree x 1

#705 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 21 October 2021 - 05:30 PM

The accuracy of the Trume Age test is stated as being better than 5 years, not accurate enough for me to draw the conclusion that these additions were harmful, but I can be reasonably certain that any benefit, if any, was minor. 

 

Methods of determining methylation patterns, such as bisulfite sequencing, are generally repeatable. My interpretation is that the 5 year error refers to the statistics of a population, not test-to-test differences for a single individual. It is likely that the increase in epigenetic age from your previous test is real.



#706 QuestforLife

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

Posted 26 October 2021 - 11:38 AM

Loss of Growth Differentiation Factor 11 Shortens Telomere Length by Downregulating Telomerase Activity

 

 

Maintenance of telomere length is essential to delay replicative cellular senescence. It is controversial on whether growth differentiation factor 11 (GDF11) can reverse cellular senescence, and this work aims to establish the causality between GDF11 and the telomere maintenance unequivocally. Using CRISPR/Cas9 technique and a long-term in vitro culture model of cellular senescence, we show here that in vitro genetic deletion of GDF11 causes shortening of telomere length, downregulation of telomeric reverse transcriptase (TERT) and telomeric RNA component (TERC), the key enzyme and the RNA component for extension of the telomere, and reduction of telomerase activity. doi: 10.3389/fphys.2021.726345

 

 

In this study they attempt to investigate the purported rejuvenating effects of GDF11 by examining its effect on the senescence of cultured murine neural cells (Neuro 2a type) by genetically deleting GDF11. Before I talk about their results, I want to mention a couple of shortcomings in this study.

 

Firstly, mice neural cells are a strange choice of cell.  They're not human. Their telomerase activity is likely higher than human cells. They claim they are ‘highly proliferative’. I think they mean in culture. I would have preferred a cell type known to proliferate through life in vivo. Also, despite claiming they are studying cellular senescence, and culturing cells for a a couple of months, they didn't really look at cellular senescence at all: no study of passage rates or their decline with time, no culturing until replicative arrest, they didn't even look at markers of cellular senescence like beta galactosidase.

 

Having said all that, they have further elucidated the link between GDF11 and telomeres, so let us look at that.

 

In normal neuro 2a cell culture, between early (<10) passages and later (65+) passages, telomeres shortened - not much on average - but very noticeably in the percentage of short telomeres, i.e. over time there remained a range of telomere lengths from long to short, but a noticeable bunching occurred at the shortest (20%) end with greater passages.

 

In the cells with no functional GDF11 gene there was a large shortening of telomeres (in early and late passage cells the shortening was similar; it didn't seem to be mediated by further cellular division) to about half of their normal length. This was also reflected in ~ a halving in the expression of telomerase protein, the RNA template component (TERC), and other telomere related proteins (RPA, Dclre1b).

 

Exogenous GDF11 added to the culture or genetic overexpression of GDF11 by other means restored TERT RNA expression, but not Telomerase protein expression or TERC. This is strange but probably due to the genetic deletion of GDF11, which as we've seen on this thread in a previous study is involved in mutual upregulation with the demethylase TET2 - without the GDF11 gene demethylation or other beneficial feedback loops are not going to happen [1].

 

The most interesting part of the study comes last. They found that deleting GDF11 increased SMAD2 association with the telomerase gene and decreased it's expression. I've investigated the SMADs before, and found that they are the intra cell equivalent of TGF-B. Inflammatory signalling outside the cell triggers the SMADs within the cell [2], with consequent changes to things like making the cytoskeleton more rigid, which I've discussed here before when looking at ROCK inhibitors (which dissolve the cytoskeleton and allow cells to adopt a more primitive morphology). I’ve also looked at the mechanism of action of asiaticoside before re: telomerase activation, deciding that it most likely acts through the negative SMAD regulator, SMAD7 [3], [4].

 

In the past I've worried that as GDF11 acted through TGF-B it would be contraindicated with asiaticoside, or other things like fish oils that act in an anti-inflammatory way. This study allays this fear, showing that asiaticoside would likely be synergistic with GDF11, as the former will upregulate SMAD7, which down regulates all the other SMADs, whilst GDF11 decreases the association of SMAD2 with the telomerase gene. This should increase telomerase activity more than either would do alone.

 

[1] https://www.longecit...-15#entry903694

[2] https://en.m.wikiped...gnaling_pathway

[3] DOI 10.1007/s00403-010-1114-8

[4] doi:10.1038/sj.cr.7310023

 


Edited by QuestforLife, 26 October 2021 - 11:43 AM.

  • Informative x 2
  • Well Written x 1
  • WellResearched x 1

#707 QuestforLife

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

Posted 27 October 2021 - 10:42 AM

Does Berberine shorten telomeres?

 

With the exception of AKG, berberine has given me the strongest signal for a reduction of epigenetic age over the years I have been testing (2017-2021). But its mechanism is obscure. In the case of AKG we know it is an important cofactor for the demethylases, with methylation of differentiation gene promoters implicated in aging as explained by the ‘selfish cell’.

 

Could it be that berberine shortens telomeres and hence increases cell turnover from the underlying stem cell pool? This would give the appearance of reducing epigenetic age, but probably wouldn’t be something we’d want to do long term.

 

This 2021 paper is pertinent.

 

Berberine Inhibits Telomerase Activity and Induces Cell Cycle Arrest and Telomere Erosion in Colorectal Cancer Cell Line, HCT 116

...in this study, the aspects of telomerase inhibitors on a CRC cell line (HCT 116) were investigated. Screening on HCT 116 at 48 h showed that berberine (10.30 ± 0.89 µg/mL) is the most effective (lowest IC50 value) telomerase inhibitor compared to boldine (37.87 ± 3.12 µg/mL) and silymarin (>200 µg/mL). Further analyses exhibited that berberine treatment caused G0/G1 phase arrest at 48 h due to high cyclin D1 (CCND1) and low cyclindependent kinase 4 (CDK4) protein and mRNA levels, simultaneous downregulation of human telomerase reverse transcriptase (TERT) mRNA and human telomerase RNA component (TERC) levels, as well as a decrease in the TERT protein level and telomerase activity. The effect of berberine treatment on the cell cycle was time dependent as it resulted in a delayed cell cycle and doubling time by 2.18-fold. Telomerase activity and level was significantly decreased, and telomere erosion followed suit. In summary, our findings suggested that berberine could decrease telomerase activity and level of HCT 116, which in turn inhibits the proliferative ability of the cells. Source: https://doi.org/10.3...lecules26020376

 

Note this study is done in cancer cells; in this case it is appropriate as these cells have significant telomerase activity. Remember that both telomere length tests and epigenetic (methylation) aging tests examine leukocytes, which have telomerase activity (when stimulated).

 

A couple of key findings from the paper:

 

10ug/ml of berberine hugely (108 fold) reduced telomerase RNA production in HCT 116 cells.

molecules-26-00376-g006-550.jpg

It wasn’t just RNA, it also reduced the telomerase protein (by 73%) made by these cells.

molecules-26-00376-g007-550.jpg

 

And the reduced telomerase protein wasn’t just some artifact of HCT 116 cells reducing their division due to the growth inhibitory effects of berberine (which also happens). Berberine actually reduced telomere length in these cells (by a fifth).

molecules-26-00376-g008-550.jpg

This is pretty damning. 10ug/ml is a ridiculous concentration of berberine however, nothing like this could ever occur in a human consuming it orally (more realistic serum levels are 0.1 ng/ml!).

 

The 10ug/ml concentration (denoting berberine as moderately cytotoxic) figure quoted is the IC50 level . IC50 is the concentration at which half the maximum inhibition (of telomerase,) is achieved. It is useful to know the IC50 figure because there are often diminishing returns to adding more and more of the substance beyond this point. But it would have been very nice to know what the shape of the inhibition curve looked like. Unfortunately this study does not provide it. Most importantly, what is the minimum concentration of berberine at which telomerase activity was measurably reduced? It is possible that even nanogram levels would produce a meaningful effect over months of taking berberine everyday. I have asked the study authors for clarification.

 

For now it remains an interesting hypothesis.


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

#708 dlewis1453

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

Posted 29 October 2021 - 05:09 PM

 

Could it be that berberine shortens telomeres and hence increases cell turnover from the underlying stem cell pool? This would give the appearance of reducing epigenetic age, but probably wouldn’t be something we’d want to do long term.

 

 

As always, an interesting, insightful, and well-researched post. In my view, this post serves as a reminder that a substance that appears to offer anti-aging benefits (in form of epigenetic age reduction) may in fact be detrimental towards another aspect of aging (telomeres). Berberine may essentially be robbing Peter to pay Paul. Ideally, we should know how any anti-aging substance impacts both epigenetics and telomeres. 


  • Good Point x 2

#709 QuestforLife

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

Posted 09 November 2021 - 02:02 PM

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


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.

 

More in the same vein. 

 

 

Mitochondrial Telomerase Reverse Transcriptase Protects from Myocardial Ischemia/reperfusion Injury by Improving Complex I Composition and Function

 

Background: The catalytic subunit of telomerase, Telomerase Reverse Transcriptase (TERT) has protective functions in the cardiovascular system. TERT is not only present in the nucleus, but also in mitochondria. However, it is unclear whether nuclear or mitochondrial TERT is responsible for the observed protection and appropriate tools are missing to dissect this.

 

Methods: We generated new mouse models containing TERT exclusively in the mitochondria (mitoTERT mice) or the nucleus (nucTERT mice) to finally distinguish between the functions of nuclear and mitochondrial TERT. Outcome after ischemia/reperfusion, mitochondrial respiration in the heart as well as cellular functions of cardiomyocytes, fibroblasts, and endothelial cells were determined.

 

Results: All mice were phenotypically normal. While respiration was reduced in cardiac mitochondria from TERT-deficient and nucTERT mice, it was increased in mitoTERT animals. The latter also had smaller infarcts than wildtype mice, whereas nucTERT animals had larger infarcts. The decrease in ejection fraction after one, two and four weeks of reperfusion was attenuated in mitoTERT mice. Scar size was also reduced and vascularization increased. Mitochondrial TERT protected a cardiomyocyte cell line from apoptosis. Myofibroblast differentiation, which depends on complex I activity, was abrogated in TERT-deficient and nucTERT cardiac fibroblasts and completely restored in mitoTERT cells. In endothelial cells, mitochondrial TERT enhanced migratory capacity and activation of endothelial NO synthase. Mechanistically, mitochondrial TERT improved the ratio between complex I matrix arm and membrane subunits explaining the enhanced complex I activity. In human right atrial appendages, TERT was localized in mitochondria and there increased by remote ischemic preconditioning. The Telomerase activator, TA-65 evoked a similar effect in endothelial cells, thereby increasing their migratory capacity, and enhanced myofibroblast differentiation.

 

Conclusions: Mitochondrial, but not nuclear TERT, is critical for mitochondrial respiration and during ischemia/reperfusion injury. Mitochondrial TERT improves complex I subunit composition. TERT is present in human heart mitochondria, and remote ischemic preconditioning increases its level in those organelles. TA-65 has comparable effects ex vivo and improves migratory capacity of endothelial cells and myofibroblast differentiation. We conclude that mitochondrial TERT is responsible for cardioprotection and its increase could serve as a therapeutic strategy.

 

Source: https://www.ahajourn...NAHA.120.051923

 

 

In this paper they demonstrated the benefits of telomerase for mitochondria as opposed to its known telomere elongating effects in the nucleus.

 

In the previous paper I reviewed it was shown telomerase was able to improve mitophagy. In this paper they demonstrated the benefits of mitochondrially located telomerase for recovery from reperfusion injury (when an organ or tissue in a mouse is starved of oxygen and then has it restored suddenly).

 

They also did some in vitro work showing that the benefits of TA-65 on human endothelial cells (in terms of migration ability and NO production) were dependent on mitochondrially rather than nuclear located increases in telomerase . 


  • Informative x 1

#710 QuestforLife

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

Posted 12 November 2021 - 03:49 PM

Naringenin: super supplement?

 

1.Naringenin lengthens telomeres in mouse neural stem cells, reduces senescent cell markers and increases their proliferation (in vitro). It also enhances the learning and memory of aged mice (in vivo; injected).

 


Anti-aging effects of Ribes meyeri anthocyanins on neural stem cells and aging mice

 

Aging is associated with neurological impairment and cognitive decline. Flavonoids are very promising in anti-aging research in mouse models. Ribes meyeri anthocyanins are rich in abundant flavonoids, but their anti-aging biological activities remain unknown. In this study, we prepared an R. meyeri anthocyanin extract and analyzed its effects on neural stem cell (NSC) senescence in vivo and in vitro. We isolated mouse NSCs and used cell counting kit-8 (CCK-8), cell cycle, reactive oxygen species (ROS), and immunofluorescence methods to analyze the anti-aging effects of R. meyeri anthocyanins as well as naringenin (Nar), which metabolic analysis revealed as an important flavonoid in R. meyeri anthocyanins. RNA-sequencing (RNA-seq) and enzyme-linked immuno sorbent assay (ELISA) methods were also used to investigate Nar-specific mechanisms of anti-aging. After R. meyeri anthocyanin treatment, NSC proliferation accelerated, and NSCs had decreased senescence markers, and reduced P16ink4a expression. R. meyeri anthocyanin treatment also reversed age-dependent neuronal loss in vivo and in vitro. Nar blocked mNSC aging in vitro and improved spatial memory and cognitive abilities in aging mice through downregulation of plasma TNF-α protein. These findings suggest that R. meyeri anthocyanins increase NSC proliferation and improve neurogenesis with aging via Nar-induced reductions in TNF-α protein levels in vivo.

 

DOI: 10.18632/aging.103955

 

2. Naringenin (in doses of 1-50uM) acts through suppressing SMAD3, which is activated by TGF-B.

 


Smad3 Specific Inhibitor, Naringenin, Decreases the Expression of Extracellular Matrix Induced by TGF-"1 in Cultured Rat Hepatic Stellate Cells

 

Results. Naringenin reduced not only the accumulation of ECM, including collagen I!1 (Col I!1), fibronectin (FN), and plasminogen activator inhibitor-1 (PAI-1), but also the production of Smad3 induced by TGF-"1 in both mRNA and protein levels in a dose-dependent manner. Moreover,naringenin selectively inhibited the transcription of Smad3, but not other Smads involved in TGF-1 signaling pathways. Conclusion. Our data demonstrate that naringenin can exert antifibrogenic effects by directly or indirectly down-regulating Smad3 protein expression and phosphorylation through TGF-" signaling.

 

DOI: 10.1007/s11095-005-9043-5

 

3.SMAD3 is a negative regulator of telomerase activity. This is likely relevant to humans with already suppressed levels of telomerase, because the SMAD3 mediated suppression further reduced telomerase, even when its activity in rat cells had been impaired by a promoter mutation.

 

Role of Smad3 in the regulation of rat telomerase reverse transcriptase byTGFb

Telomerase is induced in certain pathological conditions such as cancer and tissue injuryand repair. This induction in fibroblasts from injured lung is repressed bytransforming growth factor b (TGFb) via yet unknown mechanisms. In this study, the role of Smad3 in the inhibition of telomerase reverse transcriptase (TERT) gene transcription byTGFb was investigated. The rat TERT (rTERT) gene promoter was cloned by PCR amplification and fused with a luciferase reporter gene. This construct was used to analyse regulation of promoter activity in fibroblasts isolated from bleomycin-injured lung with induced telomerase activity. The results showed that TGFb inhibited rTERT transcription while stimulating Smad3 expression. Interestingly, TGFb also inhibited the expression of c-myc. Cotransfection with a Smad3 expressing plasmid further repressed rTERT transcription and c-myc expression, while cotransfection with the corresponding antisense Smad3 construct had the opposite effect. Mutation of an E-box in the rTERT promoter suppressed its activity, which could be further reduced by TGFb treatment. In contrast, mutation at a Smad binding element enhanced promoter activity whose inhibition was impaired by TGFb treatment. Thus TGFb inhibition of rTERT gene expression was directly mediated by Smad3 via the Smad binding element, while c-myc appears to primarily regulate its constitutive or induced expression.

 

doi:10.1038/sj.onc.1209140

 

What is this I hear you cry? Those uM doses are quite unobtainable in a human! Normally with oral supplements I would agree, but…

 

4. Tens of uM can easily be obtained in human serum with doses of a few hundred mg. And it doesn’t build up either; even at the highest dose of 900mg Naringenin was completely cleared in 24 hours.

 

 

Safety and Pharmacokinetics of Naringenin: A Randomized, Controlled, Single Ascending Dose, Clinical Trial

 

Aims—This study evaluated the safety and pharmacokinetics of naringenin in healthy adults, consuming a whole orange (Citrus Sinensis) extract.

Methods—In a single ascending dose randomized crossover trial, 18 adults ingested 150mg (NAR150), 300mg (NAR300), 600mg (NAR600), and 900mg (NAR900) doses of naringenin or placebo. Each dose or placebo was followed by a wash-out period of at least one week. Blood safety markers were evaluated pre-dose and 24 hours post-dose. Adverse events were recorded. Serum naringenin concentrations were measured before and over 24 hours following ingestion of placebo, NAR150, and NAR600. Four and 24-hour serum measurements were obtained after

placebo, NAR300, and NAR900 ingestion. Data were analyzed using a mixed effects linear model.

 

Results—There were no relevant adverse events or changes in blood safety markers following ingestion of all naringenin doses. The pharmacokinetic parameters were: Maximal concentration:
15.76±7.88μM (NAR150) and 48.45±7.88μM (NAR600); Time to peak: 3.17±0.74h (NAR150) and 2.41±0.74h (NAR600); Area under the 24-hour concentration-time curve: 67.61±24.36μM×h (NAR150) and 199.06±24.36μM×h (NAR600); and Apparent oral clearance: 10.21±2.34L/h (NAR150) and 13.70±2.34L/h (NAR 600). Naringenin half-life was 3.0h (NAR150) and 2.65h (NAR600). After NAR300 ingestion, serum concentrations were 10.67±5.74μM (4h) and 0.35±0.30μM (24h). After NAR900 ingestion, serum concentrations were 43.11±5.26μM (4h) and 0.24±0.30μM (24h).

 

Conclusions—Ingestion of 150 to 900mg doses of naringenin is safe in healthy adults, and serum concentrations are proportional to the dose administered. Since naringenin (8μM) is
effective in primary human adipocytes, ingestion of 300mg naringenin twice/day will likely elicit a physiologic effect.

 

doi:10.1111/dom.13868.

 

I’ve attached a figure showing serum concentration.

 

What other benefits might we obtain from narigenin?

 

5. You might lose weight.

 

 

Naringenin Promotes Thermogenic Gene Expression in Human White Adipose Tissue

 

Objective—Naringenin, a citrus flavonoid, prevents diet-induced weight gain and improves glucose and lipid metabolism in rodents. There is evidence that naringenin activates brown fat and increases energy expenditure in mice, but little is known about its effects in humans. Our goal was to examine the effects of naringenin on energy expenditure in adipose tissue.

 

Methods—Human white adipocyte cultures (hADSC), and subcutaneous abdominal adipose tissue (pWAT) were treated with naringenin for 7–14 days. Expression (qRT-PCR, immunoblotting) of candidate genes involved in thermogenesis and glucose metabolism was measured. Oxygen consumption rate (OCR) was measured in hADSC using a Seahorse Flux analyzer.

 

Results—In hADSC, naringenin increased expression of the genes associated with thermogenesis and fat oxidation including uncoupling protein 1, adipose triglyceride lipase, and key factors associated with insulin sensitivity including glucose transporter 4, adiponectin, and carbohydrate response element binding protein (p<0.01). Similar responses were observed in pWAT. Basal, ATP-linked, maximal, and reserve OCR increased in the naringenin-treated hADSC (p<0.01).

 

Conclusions—Naringenin increases energy expenditure in hADSC and stimulates expression of key enzymes involved in thermogenesis and insulin sensitivity in hADSC and pWAT. Naringenin may promote conversion of human white adipose tissue to a brown/beige phenotype.

 

doi:10.1002/oby.22352.

 

 

6.Conclusions: Narigenin looks like an easy win for telomeres and health in general; it is super bioavailable, is cleared quickly enough not to worry about taking breaks. How do we supplement? I attach a figure showing you can get a very decent dose of naringin from grapefruit juice. You can also buy near 100% naringin capsules if you desire. Note this is naringin not naringenin, but the former is rapidly converted into the later in the human gut by naringinase. Is there any downside? Grapefruit juice is said to inhibit the CYP3a4 enzyme, responsible for breaking down numerous pharmaceuticals (like rapamycin) and coffee, for example. So take grapefruit juice with care if you are on medication. But it is controversial what component of grapefruit is responsible for this, as a study showed quite weak inhibition of CYP3a4 by naringenin or naringin.

Attached Thumbnails

  • naringin content of grapefruit juice.png
  • Naringenin super supplement.png

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

#711 fauxstradamus

  • Guest
  • 8 posts
  • 2
  • Location:Michigan USA
  • NO

Posted 13 November 2021 - 07:45 PM

Well according to Janic et al, a low dose statin is best, see https://www.ncbi.nlm...pubmed/28951255

Also they found an intermittent rather than chronic dosing is superior with peak benefit in rats at ~6weeks, see: https://www.ncbi.nlm...pubmed/27010807

When I tried out atorvastatin and losartan I found I had better results dosing for 1 month (5mg atorvastatin, 25mg losartan) than I did when dosing for 2 weeks (10mg atorvastatin, 25mg losartan, 30mg pioglitazone). On both occasions I was on Keto, although the latter time was Jan (3 + months keto), the former time I had only just started keto (November).

I'm unsure exactly about the utility of statins + keto, but I still think the ROCK inhibition effects of statins make them a worthwhile addition to use a few times a year. I might have more to say when I've done more research (story of my life!).

 
You might be right, although I have a suspicion the link goes both ways. Even if it doesn't, insulin is a powerful growth hormone in its own right; i'd argue stronger even than mTOR - not pound for pound of course, but over the course of 24 hours - compare the state of someone constantly snacking on carbs (normal western diet) to someone who occasionally eats a meat heavy meal (keto), and I think we'd agree who is anabolic and who is catabolic!

 

QuestforLife, I've been doing a low-dose statin-sartan protocol for a few weeks now and reading through your posts on this topic, and I have a question for you if you would be so kind . . . .   I just registered on the site, and I don't have any scientific training per se.   My reason for doing the protocol is to try to slow or reverse existing atherosclerosis and to try to perhaps improve any of my other health issues via anti-aging effects.  I don't have the luxury of focusing on life extension or longevity because my "healthspan" is already hitting the wall so to speak at age 56 despite many years of great diet, exercise, fasting, "informed" supplement taking, etc.   Autoimmunity and "bad genes" seems to trump lifestyle and active intervention, at least in my experience, for me individually, but I'm not giving up without a fight, and I'm hoping that at some point, systemic rejuvenation in some way, shape or form will take hold and help me combat some of these issues like worsening OA and joint and spine problems.  

 

That said, I'm doing the protocol and my question is about the Statin-Sartan protocol based on the Janic study you have referenced.   I'm taking GG (geranylgeraniol) to mitigate any mito-toxic or muscle-related side effects of the statin (fluvastatin), despite the low dose.   GG is a precursor of CQ10 as you may know, and some scientists, including the discoverer of GG, Dr. Barrie Tan, believe that GG provides better, more direct (upstream) protection against the muscle-toxic or mitotoxic effects of statins.  But I'm wondering if the Rho kinase (ROCK) inhibiting effect of low-dose statins is perhaps negated by taking GG and if the toxic effects of statins and the Rho inhibition effect are inextricably linked?  Or, alternatively, if one gains the benefits of Rho inhibition and avoids most of the toxic effects by taking a low dose for a short period of time??   So I'm wondering if you think that taking GG confers no added benefit in this protocol or if it actually undermines the beneficial, Rho-inhibition effects at low dose?   I don't know if the quote below is actually applicable or not.

 

 

Results from experimental and clinical research indicate that the pleiotropic effects of statins treatment are predominantly derived from its potent effects on inhibiting Rho‐GTPase isoprenylation through reducing geranyl‐geranylpyrophosphate (GGPP) generation during cholesterol biosynthesis.7, 8  GTPase and Atherosclerosis: Pleiotropic Effects of Statins
2015 - Journal of the American Heart Association; P e002113;R doi:10.1161/JAHA.115.002113;U https://www.ahajourn...JAHA.115.002113

 

 

Also, it looks like there is some evidence in the literature that statin withdrawal can upregulate Rho gene transcription and, via a rebound inflammation effect, reverse the beneficial, anti-inflammatory effects of statin treatment;  I wonder if this mainly pertains to higher-dose treatment for a longer period of time (i.e., standard, continuous, as opposed to low-dose, intermittent, statin therapy)?   Not being a scientist, I have not read the study I've quoted below in its entirety.   I'm hoping that the withdrawal rebound effect does not apply to a low-dose, intermittent protocol.   This finding would seem to run 100% counter to the notion that taking a low-dose statin with a low-dose sartan for a month or so only confers a lingering beneficial effect on blood vessels and endothelial function and health that slowly diminshes over several months after discontinuation.   Thanks, and thank you for all your effort and persistence in working out your personal protocols and posting about them on this site!

 

 

Experimental evidence suggests that statin withdrawal rapidly abrogates the beneficial effects of statins and induces rebound inflammation. In mice, statin withdrawal results in a negative feedback up-regulation of Rho gene transcription, increasing Rho activity and suppressing endothelial NO production [129]. In hypercholesterolemic patients, atorvastatin withdrawal rapidly increases pro-inflammatory and pro-thrombotic pathways [130], while simvastatin withdrawal during an acute MI event has been associated with a rebound increase in CRP levels [131]. Retrospective data from clinical registries have suggested that statins discontinuation in non-ST segment elevation MI patients is associated with worse clinical outcomes [19]. Even though this rebound inflammatory response after statins withdrawal does not alter cardiovascular risk in stable coronary patients [132], it may induce plaque instability and subsequent adverse events in ACS.   Antonopoulos AS, Margaritis M, Lee R, Channon K, Antoniades C. Statins as anti-inflammatory agents in atherogenesis: molecular mechanisms and lessons from the recent clinical trials. Curr Pharm Des. 2012;18(11):1519-1530. doi:10.2174/138161212799504803

 

 

 

 

 

 

 

 

 

 

 

 


Edited by fauxstradamus, 13 November 2021 - 07:47 PM.

  • like x 1

#712 fauxstradamus

  • Guest
  • 8 posts
  • 2
  • Location:Michigan USA
  • NO

Posted 13 November 2021 - 10:48 PM


 

 

Experimental evidence suggests that statin withdrawal rapidly abrogates the beneficial effects of statins and induces rebound inflammation.

 

Thinking about this a bit more, maybe tapering off for a few weeks, instead of just stopping the statin cold at the end of the one-month protocol, would prevent any potential inflammation rebound effect, if indeed that would even be a concern with a low-dose regimen followed for such a short time period.  
 



#713 Andey

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

Posted 14 November 2021 - 08:22 AM

  

 

  Not an answer to your question but I think its a big ask to fix autoimmune issues with supplements. 

There is no one way as we are all different and have different issues but things that I have some progress with were

1. Figuring out that I have HLA-DQ8 so my MHC II picks up gliadin molecule and that leads to unnecessary inflammation. It was very subclinical, def not to the level of a celiac disease, yet I got rid of 90% unspecific GI issues.

2. Monitoring homocysteine status and adjusting B vitamins according to that 

3. Keto diet 

4. Just recently started to figure out what fat comp is better for me - looks like I dont process SFA properly and need to rely on PUFA and MUFA more (prob because of PEMT polymorphism)

5  Helping lymph nodes with self massage (they got inflamed sometimes and can get stuck in this state for a very long time, inflaming the tissue around it and making it a vicious cycle, massage can give a quick resolution to it)

6 Prolonged fasting is a help too, but I think keto gives 80%  of benefit.


Edited by Andey, 14 November 2021 - 08:22 AM.


#714 QuestforLife

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

Posted 14 November 2021 - 09:43 AM

Thinking about this a bit more, maybe tapering off for a few weeks, instead of just stopping the statin cold at the end of the one-month protocol, would prevent any potential inflammation rebound effect, if indeed that would even be a concern with a low-dose regimen followed for such a short time period.


There is no evidence of a 'bounce back' in the Janic studies I have read.

See Figure 2 from the following publication, where flow mediated dilation, pulse wave velocity and carotid artery stiffness values all improve dramatically from 30 days of using low dose fluvastatin and valsartan, and then decline gradually to baseline over 6 months.

https://www.spandido...2/etm.2015.2622

I also do not think taking ubiquinone or precursors would abrogate the benefits, as I think the benefits are due to ROCK inhibition, which is only tangentially related to the pathway you discuss. But I don't have all the evidence to hand to prove it to you right now. I found that 30 days of atorvastatin (I didn't have the milder fluvastatin available) gave me mild muscle cramps, so you might want to take precautions against this.
  • Good Point x 1
  • like x 1

#715 dlewis1453

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

Posted 19 November 2021 - 06:48 PM

This new article, which I found linked on the longecity forum front page, states that parabiosis resulted in a widespread epigenetic shift to a younger profile, including an increase in Tert. Since we have discussed GDF11's effect on Tert multiple times in this thread, I thought it would be helpful to share this article. I wonder to what extent GDF11 was responsible for the downregulation of Dmnt3b? 

 

"In all, the researchers ascertained that this intervention caused somewhat similar gene expression changes as other interventions known to extend healthspan and lifespan, such as caloric restriction (CR). The negative association with aging signatures was even stronger with parabiosis than with CR. Three months of parabiosis was found to be much more effective than five weeks in creating lasting changes to gene expression."

 

"Among the genes found to be upregulated with parabiosis were Sirt3, which improves regeneration and decreases reactive oxygen species, along with Tert, which encodes for telomerase reverse transcriptase, a compound that lengthens telomeres and has been shown to have other positive effectsDmnt3b, a gene that produces an enzyme associated with methylation, was downregulated, as were genes that encode the harmful senescence-associated secretory phenotype (SASP)."

 

https://www.longecit...helps-old-mice/


Edited by dlewis1453, 19 November 2021 - 06:48 PM.


#716 QuestforLife

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

Posted 19 November 2021 - 09:35 PM

This new article, which I found linked on the longecity forum front page, states that parabiosis resulted in a widespread epigenetic shift to a younger profile, including an increase in Tert. Since we have discussed GDF11's effect on Tert multiple times in this thread, I thought it would be helpful to share this article. I wonder to what extent GDF11 was responsible for the downregulation of Dmnt3b?


The de Novo methyl transferases are tricky things. Turn them down and telomerase goes up, but then telomerase turns de Novo methyl transferases back up. It's some sort of control mechanism. Totally disable one or more DNMTs and you can immortalise cells, but then methylation doesn't work properly and stem cells can't differentiate.

But there is evidence DNMTs go up with age and TETs go down. And this can also select for selfish stem cells that don't differentiate because of too much methylation (at certain gene promoters, including GDF11 - another feedback mechanism).

I discussed DNMTs before here:

https://www.longecit...-23#entry910058

So far I've only seen evidence GDF11 upregulates TET2/3. I haven't seen anything to show it downregulates any of the DNMTs.

#717 QuestforLife

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

Posted 09 December 2021 - 09:45 AM

Just a quick note from me today.

 

I got an amino acid blood test done. To my astonishment a group of interrelated amino acid were at low or very low levels. They were as follows: Glutamine and Arginine: very low; Glutamic acid and asparagine: low. And this is with me taking AKG (which can be converted to glutamine) on and off since March 2020!

 

Given I’ve been taking telomerase activators and other stem cell stimulants like GDF11 for some time – I wonder whether glutamine has been depleted by cell proliferation. I mean, my levels are normally associated with immune exhaustion (though I have none of the symptoms, and have healthy markers of immune competence like low NLR). 

 

 

 Glutamine is utilised at a high rate by cells of the immune system in culture and is required to support optimal lymphocyte proliferation and production of cytokines by lymphocytes and macrophages. source: https://link.springe...1007/BF01366922

 

 

From this a hypothesis sprung to mind. In the past I’ve discussed at length how long telomeres enable continued proliferation but potentially select for more selfish stem cells; the mechanism proposed was via selection of random epimutations favouring those cells that didn’t differentiate. I still think this may be a mechanism of aging.

 

But here is a simpler explanation, which explains why telomerase activators can very quickly increase methylation age: additional telomerase enables faster cell division in – for example – the immune system; but depletes glutamine. The pool of AKG is then drained in order to replenish glutamine, which leads to increased methylation and reduced stem cell differentiation. 

 

In any case I decided to supplement glutamine. In addition I have begun to supplement (homemade) liposomal AKG.

 

We will have to see what effect it has on my next methylation age result.


Edited by QuestforLife, 09 December 2021 - 09:45 AM.

  • Informative x 5

#718 Andey

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

Posted 11 December 2021 - 07:38 AM

Actually I think AKG has been slowly damaging my liver. My ALT levels were gradually rising from normal to just above normal by April, and then rocketed to double that value in June. Since that time I stopped AKG and it's fallen back towards the normal range. I don't absolutely know 100% it was the AKG, but it is my prime suspect. AKG is cleared rapidly by the liver (according to the video clip above). I wonder if given the effect of demethylases on reducing differentiation, and the highly proliferative nature of the liver, if AKG might also be impeding liver regeneration this way also.

 

 

   Same thing. I havent checked my liver enzymes in a while (became lazy with all this covid stuff), but recently rechecked and ALT was above 100 (AST was higher than NR too).

I discontinued AKG and during 3 weeks ALT returned to the middle of the normal range. 

  I also noticed that my CRP level was way higher on the day I injected GDF11, but need more datapoints to make something out of it. 



#719 QuestforLife

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

Posted 11 December 2021 - 09:20 AM

Same thing. I havent checked my liver enzymes in a while (became lazy with all this covid stuff), but recently rechecked and ALT was above 100 (AST was higher than NR too).
I discontinued AKG and during 3 weeks ALT returned to the middle of the normal range.
I also noticed that my CRP level was way higher on the day I injected GDF11, but need more datapoints to make something out of it.


I was never 100% sure what it was. But after a few months my ALT was totally normal again, even though I went back on AKG.

I think that a lot of things can cause fibrosis in the liver. For example anything that encourages cell proliferation, like GDF11, Oxytocin, etc if that cell division is accompanied with raised inflammation. Like forcing a wound to heal too rapidly causes scarring. So perhaps a solution is to accompany a mitotic stimulus with an anti inflammatory. It's all guesswork at this stage.

I'll look at my liver enzymes again in the Spring.

#720 aribadabar

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

Posted 12 December 2021 - 01:43 AM

I cannot report significant liver issues caused by (Ca)AKG at 1000mg/d for many months but I was concurrently on 400mg/d SAMe, often boosted by 1000 mg/d TMG so they may have neutralized any potential deleterious action.

ALT was 10.







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