Wednesday, September 21, 2016

Does Beta Hydroxybutyrate Increase FOXO1 and Telomerase?

I have rewritten the bolded paragraph below to reveal the central idea.

The present studies suggest that FoxO1 plays beneficial roles by inducing genes involved in telomerase activity, as well as anti-oxidant, autophagic, and anti-apoptotic genes under conditions of increased beta hydroxybutyrate promotion of starvation gene set, and suggest that FoxO1 signaling may be an important mediator of metabolic equilibrium during conditions of increased beta hydroxybutyrate promotion of starvation gene set.

Calore restriction is equal to conditions of increased beta hydroxybutyrate promotion of starvation gene set.

FOXO1 is elevated when insulin and IGF1 is low or when fasting.  These studies are performed best in single celled organism that do not exercise.  One could prove this by adding a PPAR alpha agonist such as Fenofibrate or Ursolic acid to cell culture without calorie restriction.

One usually has ejaculation a sympathetic action usually with erections a parasympathetic activity.  Only in ED do you find ejaculation separately.  In nature fasting creates beta hydroxybutyrate but so does exercise.  Would exercise or its chemical mimetic BHB induce FOXO1 effect on telomerase just as fasting.  My conjecture is yes?  Both pathways can be expressed simultaneously but perhaps with or without synergy.

FoxO1 signaling plays a pivotal role in the cardiac telomere biology responses to calorie restriction

This study examined whether the forkhead transcription factors of O group 1 (FoxO1) might be involved in telomere biology during calorie restriction (CR). We used FoxO1-knockout heterozygous mice (FoxO1+/−) and wild-type mice (WT) as a control. Both WT and FoxO1+/− were subjected to ad libitum (AL) feeding or 30 % CR compared to AL for 20 weeks from 15 weeks of age. The heart-to-body weight ratio, blood glucose, and serum lipid profiles were not different among all groups of mice at the end of the study. Telomere size was significantly lower in the FoxO1+/−-AL than the WT-AL, and telomere attrition was not observed in either WT-CR or FoxO1+/−-CR. Telomerase activity was elevated in the heart and liver of WT-CR, but not in those of FoxO1+/−-CR. The phosphorylation of Akt was inhibited and Sirt 1 was activated in heart tissues of WT-CR and FoxO1+/−-CR. However, the ratio of conjugated to cytosolic light chain 3 increased and the level of p62 decreased in WT-CR, but not in FoxO1+/−-CR. A marker of oxidative DNA damage, 8-OhdG, was significantly lower in WT-CR only. The level of MnSOD and eNOS increased, and the level of cleaved caspase-3 decreased in WT-CR, but not FoxO1+/−-CR. Echocardiography showed that the left ventricular end-diastolic and systolic dimensions were significantly lower in WT-CR or FoxO1+/−-CR than WT-AL or FoxO1+/−-AL, respectively. The present studies suggest that FoxO1 plays beneficial roles by inducing genes involved in telomerase activity, as well as anti-oxidant, autophagic, and anti-apoptotic genes under conditions of CR, and suggest that FoxO1 signaling may be an important mediator of metabolic equilibrium during CR.

Thursday, September 1, 2016

Rejuvenation of Metabolic Health Reverses Biological Aging

The sum of the words below means that activating the 44 starvation gene sets with BHB promotes mtDNA repair, metabolic Health and lengthening and stabilization of telomeres leading to reversing cell aging.

Also note that Ursolic acid activates Sirt-1 and peroxisome proliferator-activated receptor gamma co-activator 1α/β (PGC-1α/β).

Mitochondrial metabolic failure in telomere attrition-provoked aging of bone marrow mesenchymal stem cells

The proliferation and differentiation potential of bone marrow mesenchymal stem cells (BMMSCs) declines with age and with in vitro passages. However, the underlying mechanisms and putative approaches to maintain their function are not fully understood. Recent studies have revealed telomere attrition as the core initiator determining functional decline in aging of BMMSCs. Telomere attrition activates downstream p53 signaling and compromises mitochondrial metabolism via the peroxisome proliferator-activated receptor gamma co-activator 1α/β (PGC-1α/β), a key process possesses peculiarities in BMMSCs distinct from other stem cells and their mature derivatives. Despite of the shortened telomere, the mitochondrial failure could be overcome through metabolic regulation by caloric restriction (CR) and its mediator Sirtuin 1 (SIRT1). Researches have shown that mitochondrial metabolic reprogramming by CR and SIRT1 alleviates functional decline of BMMSCs in aging. In this review, we intend to summarize our understanding about how telomere attrition initiates and induces mitochondrial compromise in functional decline of BMMSCs in aging, and the potential therapeutic strategies based on metabolic reprogramming.