Wednesday, March 14, 2018

Metformin shown for the first time to activate Telomere enzyme Telomerase in human cells via AMPK: "Good" stress links Progeria and HIV



Goldsmith Content Providers: CDC/ C. Goldsmith, P. Feorino, E. L. Palmer, W. R. McManus [Public domain], via Wikimedia Commons;The Cell Nucleus and Aging: Tantalizing Clues and Hopeful Promises. Scaffidi P, Gordon L, Misteli T. PLoS Biology Vol. 3/11/2005, e39 
 
 
A recently published study in the journal Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease in 2018 demonstrated for the first time that chronic treatment with the anti-diabetic drug metformin activated human telomerase reverse transcriptase (hTERT) in human aortic endothelial cells (HAECs) and significantly delayed endothelial senescence in an AMPK-dependent manner [1]. AMPK is activated by the induction of cellular stress, mediated by increases in intracellular calcium (Ca2+), reactive oxygen species (ROS), and/or an AMP(ADP)/ATP ratio increase, etc. [41].  Telomeres are specialized regions of repetitive nucleotide sequences located at the ends of eukaryotic chromosomes that protect chromosomal ends from deterioration [2].  However, continuous cell division leads to telomere shortening, impeding the replenishment of tissues and triggering cellular senescence (i.e. cells cease to divide).  Although human telomeres shorten with age, telomeres may be lengthened by the enzyme telomerase, a ribonucleoprotein that consists of the catalytic subunit hTERT, telomerase RNA, and the nucleolar protein dyskerin [2,3]. hTERT, which is considering limiting for telomerase activity, is a protein that exhibits reverse transcriptase activity and synthesizes telomeric DNA from an RNA template [4]. 

The authors of the study initially demonstrated that both metformin and the AMPK activator AICAR significantly increased hTERT levels and enhanced AMPK activation in human aortic endothelial cells (HAECs) [1].  Importantly, inhibition or knockdown of AMPK with compound C or siAMPKα inhibited the metformin-induced increase in hTERT while metformin failed to reverse siAMPKα-induced senescence in HAECs, indicating that hTERT expression is regulated by AMPK activation in endothelial cells.  Indeed, continuous culturing of HAECs in the presence of metformin significantly increased hTERT protein levels and activity, reduced the expression of the senescence markers p53, p21, p27, and p16, and reduced senescence-associated beta-galactosidase (SA-β-gal, a biomarker of cellular senescence) staining in HAECs, again indicating that metformin delays cellular senescence and increases hTERT levels via AMPK activation [1].  Strikingly, using ApoE-/- mice (which spontaneously develop atherosclerosis and age faster compared to normal mice), the authors also showed that chronic low-dose metformin administration for fourteen months in drinking water enhanced the levels of activated AMPK and Pgc-1α observed in the endothelial layer of the aorta [1].  Metformin also increased the transcript and protein levels of Tert, decreased senescence markers (p16, p21, p27, p53) in the total aortic homogenate, and significantly reduced SA-β-gal staining of aorta compared to untreated ApoE-/- mice, demonstrating that metformin-induced AMPK activation delays vascular aging and protects from age-associated atherosclerosis in ApoE-/- mice [1].

Interestingly, telomere length has been shown to be significantly reduced in cells derived from patients with the accelerated aging disorder Hutchinson-Gilford progeria syndrome (HGPS) and telomere shortening in normal cells that occurs during cellular senescence activates progerin production, a toxic protein that leads to an accelerating aging phenotype in children with HGPS via aberrant alternative splicing of the LMNA gene [5].  Normal lamin A plays a critical role in supporting nuclear architecture and morphology.  Lamin A binding to subtelomeric repeats also localizes telomeres to the nuclear periphery and loss of lamin A leads to defects in telomeric heterochromatin, altered nuclear distribution and shortening of telomeres, inefficient processing of dysfunctional telomeres by non-homologus end joining, and increased genomic instability [6]. Telomere length has been found to be significantly reduced in fibroblasts derived from HGPS patients and a recent study also confirmed that in normal human fibroblasts, progressive telomere damage that occurs during cellular senescence activates progerin production and also leads to extensive changes in alternative splicing of many other genes, highlighting a striking similarity between normal aging and accelerated aging in HGPS patients [5,7]. Indeed, transfection of HGPS fibroblasts with human telomerase (hTERT) mRNA restored cell proliferation, reduced cell loss, extended cellular lifespan, increased telomerase activity and telomere length, and reduced SA-β-gal staining compared to HGPS cells expressing catalytically inactive hTERT mRNA [8].  Because metformin also increases hTERT expression and inhibits senescence in human cells, it is likely that cellular stress-induced AMPK activation, mediated by increases in intracellular calcium (Ca2+), reactive oxygen species (ROS), and/or an AMP(ADP)/ATP ratio increase, etc., represents a central node linking structurally diverse compounds and methodologies that alleviate accelerated aging in HGPS cells.              

As the splicing factor SRSF1 has been shown to increase progerin production by promoting the use of a cryptic splice located in the LMNA gene, metformin was recently shown to significantly reduce the expression of SRSF1 and progerin, activate AMPK, and improve nuclear architecture in HGPS cells, indicating that AMPK activation by metformin beneficially alters gene splicing in HGPS cells by modulating SRSF1, a hypothesis that I first proposed and published in 2014 [9-11].  Also, p32, a splicing-associated protein that is an endogenous inhibitor of SRSF1 and is critical for the maintenance of mitochondrial functionality and oxidative phosphorylation, has also been shown to be essential for rapamycin- or starvation-induced autophagy mediated by ULK1 [12,13].  Because rapamycin, also an AMPK activator in vivo,  improves accelerated aging defects in HGPS cells by reducing progerin levels via induction of autophagy, AMPK activation likely also beneficially modulates the activity of p32, leading to inhibition of SRSF1 splicing activity and enhancement of mitochondrial functionality [14,15].  Moreover, PGC-1α, which is activated by AMPK and metformin and promotes telomere transcription, is downregulated in HGPS cells, leading to significant mitochondrial dysfunction [1,16,17].  Methylene blue, which activates AMPK in vivo, was shown to increase PGC-1α levels, induce progerin solubility, and alleviate accelerated aging defects in HGPS cells [16,18].  Additionally, the rapamycin analog temsirolimus alleviated accelerated aging defects in HGPS cells but transiently increased ROS and superoxide anion levels in both HGPS and normal cells within the first hour of treatment, again indicating that cellular stress-induced AMPK activation represents a common mechanism for inhibiting senescence and ameliorating symptoms associated with accelerated aging [19].    

The inhibition of SRSF1 and the promotion of hTERT expression and telomere transcription by metformin via AMPK activation also link HGPS and telomere integrity with HIV-1 latency.  Increased splicing activity of SRSF1 inhibits reactivation of latent HIV-1 residing in infected immune cells, preventing immune system detection and destruction of the virus [20].  Reactivation of latent HIV-1 (i.e. the “shock and kill” approach) leads to a reduction in SRSF1 but an increase in p32 activity and bryostatin-1 (a PKC modulator) has been shown to reactivate latent HIV-1 via AMPK activation [20,21].  Interestingly, p32 modulation via AMPK activation may also enhance and stabilize the splicing activities of hnRNPA1, a heteroribonuclear protein that associates with p32, antagonizes the splicing function of SRSF1, prevents splicing of the HIV-1 genome (promoting viral reactivation), and participates in the maintenance and preservation of telomeres [22-25].  hnRNPA1 is also decreased in senescent human fibroblasts and antagonizes cellular senescence and the senescence-associated secretory phenotype (SASP) via increasing SIRT1 expression [26,27].  Resveratrol, a plant-derived polyphenol that activates AMPK, increases hnRNPA1 protein expression and SIRT1, a histone deacetylase that plays a role in a number of age related diseases and in the extension of lifespan, is also activated by AMPK [28-30].  Also, T cell activation, an efficient method for reactivating latent HIV-1, is dependent on increases in intracellular Ca2+ and ROS, telomerase is transiently increased on T cell activation, and AMPK knockdown leads to T cell death during in vitro activation [31-34].  Furthermore, resveratrol reactivates latent HIV-1 and preliminary data demonstrated that metformin decreased the percentage of CD4+ T cells expressing PD-1, TIGIT, and TIM-3, each markers associated with T cells latently infected with HIV-1, in chronically-infected HIV-1 patients [35-37].  Such evidence strongly suggests that cellular stress-induced AMPK activation, mediated by increases in intracellular calcium (Ca2+), reactive oxygen species (ROS), and/or an AMP (ADP)/ATP ratio increase, etc. links the alleviation of accelerated cellular aging defects in HGPS with the potential eradication of HIV-1, a hypothesis that I first proposed in 2015 [38].        
 
The evidence presented in the Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease publication further substantiates that AMPK activation represents a central node that connects the therapeutic benefits of chemically distinct compounds in diseases as seemingly dissimilar as HGPS and HIV-1 latency.  Indeed, AMPK activators including metformin increase hTERT expression, promote telomere transcription and integrity, decrease the splicing activity of SRSF1 that increases progerin production but prevents latent HIV-1 reactivation, and potentially beneficially modulates the activity of the SRSF1 inhibitor p32 and the ribonucleoprotein hnRNPA1.  As AMPK activators (e.g. ionomycin) induce human oocyte activation (giving rise to normal healthy children) and AMPK is localized throughout the entire acrosome in human sperm (likely promoting the acrosome reaction), AMPK activation links normal human aging, Progeria, and HIV-1 latency with the creation of all human life (i.e. the “shock and live” approach) [11,38-40].  

 
 
References:

  1. Karnewar S, Neeli PK, Panuganti D, et al. Metformin regulates mitochondrial biogenesis and senescence through AMPK mediated H3K79 methylation: Relevance in age-associated vascular dysfunction. Biochim Biophys Acta. 2018 Apr;1864(4 Pt A):1115-1128.
  2. O'Sullivan RJ, Karlseder J. Telomeres: protecting chromosomes against genome instability. Nat Rev Mol Cell Biol. 2010 Mar;11(3):171-81.  
  3. Cohen SB, Graham ME, Lovrecz GO, Bache N, Robinson PJ, Reddel RR. Protein composition of catalytically active human telomerase from immortal cells. Science. 2007 Mar 30;315(5820):1850-3.
  4. Cong YS, Wen J, Bacchetti S. The human telomerase catalytic subunit hTERT: organization of the gene and characterization of the promoter. Hum Mol Genet. 1999 Jan;8(1):137-42.
  5. Cao K, Blair CD, Faddah DA, et al. Progerin and telomere dysfunction collaborate to trigger cellular senescence in normal human fibroblasts. J Clin Invest. 2011 Jul;121(7):2833-44.
  6. Gonzalez-Suarez I, Redwood AB, Perkins SM, et al. Novel roles for A-type lamins in telomere biology and the DNA damage response pathway. EMBO J 2009;28(16):2414–27.
  7. Decker ML, Chavez E, Vulto I, Lansdorp PM. Telomere length in Hutchinson-Gilford progeria syndrome. Mech Ageing Dev. 2009 Jun;130(6):377-83.
  8. Li Y, Zhou G, Bruno IG, Cooke JP. Telomerase mRNA Reverses Senescence in Progeria Cells. J Am Coll Cardiol. 2017 Aug 8;70(6):804-805.
  9. Egesipe AL, Blondel S1, Cicero AL, et al. Metformin decreases progerin expression and alleviates pathological defects of Hutchinson-Gilford progeria syndrome cells. NPJ Aging Mech Dis. 2016 Nov 10;2:16026.
  10. Park SK, Shin OS. Metformin alleviates ageing cellular phenotypes in Hutchinson-Gilford progeria syndrome dermal fibroblasts. Exp Dermatol. 2017 Oct;26(10):889-895.
  11. Finley J. Alteration of splice site selection in the LMNA gene and inhibition of progerin production via AMPK activation. Med Hypotheses. 2014 Nov;83(5):580-7.
  12. P32/gC1qR is indispensable for fetal development and mitochondrial translation: importance of its RNA-binding ability. Nucl Acids Res 2012;40(19):9717–37.
  13. Jiao H, Su GQ, Dong W, et al. Chaperone-like protein p32 regulates ULK1 stability and autophagy. Cell Death Differ. 2015 Nov;22(11):1812-23.
  14. Cao K, Graziotto JJ, Blair CD, et al. Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson-Gilford progeria syndrome cells. Sci Transl Med. 2011 Jun 29;3(89):89ra58.
  15. Chiao YA, Kolwicz SC, Basisty N, et al. Rapamycin transiently induces mitochondrial remodeling to reprogram energy metabolism in old hearts. Aging (Albany NY). 2016 Feb;8(2):314-27.
  16. Xiong ZM, Choi JY, Wang K, et al. Methylene blue alleviates nuclear and mitochondrial abnormalities in progeria. Aging Cell. 2016 Apr;15(2):279-90.
  17. Diman A, Boros J, Poulain F, et al. Nuclear respiratory factor 1 and endurance exercise promote human telomere transcription. Sci Adv. 2016 Jul 27;2(7):e1600031.
  18. Xie L, Li W, Winters A, Yuan F, Jin K, Yang S. Methylene blue induces macroautophagy through 5' adenosine monophosphate-activated protein kinase pathway to protect neurons from serum deprivation. Front Cell Neurosci. 2013 May 3;7:56.
  19. Gabriel D, Gordon LB, Djabali K. Temsirolimus Partially Rescues the Hutchinson-Gilford Progeria Cellular Phenotype. PLoS One. 2016 Dec 29;11(12):e0168988.
  20. Berro R, Kehn K, de la Fuente C, et al. Acetylated Tat regulates human immunodeficiency virus type 1 splicing through its interaction with the splicing regulator p32. J Virol. 2006 Apr;80(7):3189-204.
  21. Mehla R, Bivalkar-Mehla S, Zhang R, et al. Bryostatin modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute infection in a receptor independent manner. PLoS One. 2010 Jun 16;5(6):e11160.
  22. Expert-Bezançon A, Sureau A, Durosay P, et al. HnRNP A1 and the SR proteins ASF/SF2 and SC35 have antagonistic functions in splicing of beta tropomyosin exon 6B. J Biol Chem 2004 Sep 10;279(37):38249–59.
  23. LaBranche H, Dupuis S, Ben-David Y, Bani MR, Wellinger RJ, Chabot B. Telomere elongation by hnRNP A1 and a derivative that interacts with telomeric repeats and telomerase. Nat Genet 1998 Jun;19(2):199–202.
  24. Petersen-Mahrt SK, Estmer C, Ohrmalm C, Matthews DA, Russell WC, Akusjärvi G. The splicing factor-associated protein, p32, regulates RNA splicing by inhibiting ASF/SF2 RNA binding and phosphorylation. EMBO J 1999;18(4):1014–24.
  25. Caputi M, Mayeda A, Krainer AR, Zahler AM. hnRNP A/B proteins are required for inhibition of HIV-1 pre-mRNA splicing. EMBO J. 1999 Jul 15;18(14):4060-7.
  26. Zhu D1, Xu G, Ghandhi S, Hubbard K. Modulation of the expression of p16INK4a and p14ARF by hnRNP A1 and A2 RNA binding proteins: implications for cellular senescence. J Cell Physiol. 2002 Oct;193(1):19-25.
  27. Wang H, Han L, Zhao G. hnRNP A1 antagonizes cellular senescence and senescence-associated secretory phenotype via regulation of SIRT1 mRNA stability. Aging Cell. 2016 Sep 9. doi: 10.1111/acel.12511. [Epub ahead of print].
  28. Cantó C, Gerhart-Hines Z, Feige JN, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009 Apr 23;458(7241):1056-60.
  29. Moshiri A, Puppo M, Rossi M, Gherzi R, Briata P. Resveratrol limits epithelial to mesenchymal transition through modulation of KHSRP/hnRNPA1-dependent alternative splicing in mammary gland cells. Biochim Biophys Acta. 2017 Mar;1860(3):291-298.
  30. Chiang MC, Nicol CJ, Cheng YC. Resveratrol activation of AMPK-dependent pathways is neuroprotective in human neural stem cells against amyloid-beta-induced inflammation and oxidative stress. Neurochem Int. 2017 Oct 5. pii: S0197-0186(17)30362-5.
  31. Dahabieh MS, Battivelli E, Verdin E. Understanding HIV latency: the road to an HIV cure. Annu Rev Med. 2015;66:407-21.
  32. Sena LA, Li S, Jairaman A, et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity. 2013 Feb 21;38(2):225-36.
  33. Weng NP, Levine BL, June CH, Hodes RJ. Regulated expression of telomerase activity in human T lymphocyte development and activation. J Exp Med. 1996 Jun 1;183(6):2471-9.
  34. Rao E, Zhang Y, Zhu G, et al. Deficiency of AMPK in CD8+ T cells suppresses their anti-tumor function by inducing protein phosphatase-mediated cell death. Oncotarget. 2015 Apr 10;6(10):7944-58.
  35. Zeng X, Pan X, Xu X, et al. Resveratrol Reactivates Latent HIV through Increasing Histone Acetylation and Activating Heat Shock Factor 1. J Agric Food Chem. 2017 Jun 7;65(22):4384-4394.
  36. G.M. Chew, D.C. Chow, S.A. Souza, et al. Impact of adjunctive metformin therapy on T cell exhaustion and viral persistence in a clinical trial of HIV-infected adults on suppressive ART.  Journal of Virus Eradication 2017; 3 (Supplement 1): 6–19.
  37. http://viruseradication.com/abstract-details.php?abstract_id=1188, last accessed March 14, 2018.
  38. Finley J. Reactivation of latently infected HIV-1 viral reservoirs and correction of aberrant alternative splicing in the LMNA gene via AMPK activation: Common mechanism of action linking HIV-1 latency and Hutchinson-Gilford progeria syndrome. Med Hypotheses. 2015 Sep;85(3):320-32.
  39. Finley J. Oocyte activation and latent HIV-1 reactivation: AMPK as a common mechanism of action linking the beginnings of life and the potential eradication of HIV-1. Med Hypotheses. 2016 Aug;93:34-47.
  40. Calle-Guisado V, de Llera AH, Martin-Hidalgo D, et al. AMP-activated kinase in human spermatozoa: identification, intracellular localization, and key function in the regulation of sperm motility. Asian J Androl. 2017 Nov-Dec;19(6):707-714.
  41. Auciello FR, Ross FA, Ikematsu N, Hardie DG. Oxidative stress activates AMPK in cultured cells primarily by increasing cellular AMP and/or ADP. FEBS Lett. 2014 Sep 17;588(18):3361-6.