Sunday, May 13, 2018

Metformin & AMPK Link Nobel Prize-winning Telomeres & Jumping Genes with Learning, HIV, & the Creation of Human Life




Nobel Prize winners, from left to right: Elizabeth Blackburn (discovered telomerase), Barbara McClintock (discovered “jumping genes”), and Françoise Barré-Sinoussi (discovered HIV). By Science History Institute, CC BY-SA 3.0, https://commons.wikimedia.org/w/index.php?curid=30112731; By Smithsonian Institution/Science Service; Restored by Adam Cuerden - Flickr: Barbara McClintock (1902-1992), Public Domain, https://commons.wikimedia.org/w/index.php?curid=25629182; By Prolineserver (talk) - Own work, GFDL 1.2, https://commons.wikimedia.org/w/index.php?curid=5395403


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 in human aortic endothelial cells (HAECs) and significantly delayed endothelial senescence in an AMPK-dependent manner [11]. Telomeres are specialized regions of repetitive nucleotide sequences located at the ends of eukaryotic chromosomes that protect chromosomal ends from deterioration [63]. 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 [64].

This study substantiates and confirms several novel proposals in a recently published paper I authored in April of 2018 in which I first proposed that because telomerase is derived from a “jumping gene” (see below for discussion), metformin would activate telomerase via AMPK [6]. My paper also highlights a novel link between hippocampal long-term potentiation (essential for learning and memory), alleviation of accelerated cellular aging in Hutchinson-Gilford progeria syndrome, oocyte activation and the sperm acrosome reaction (prerequisites for human life creation), and transposable element (i.e. “jumping genes”)-mediated promotion of learning, memory, and the creation of human life [1-7]. Indeed, these novel proposals also link several Nobel Prize-winning discoveries, including the discovery of telomerase by Elizabeth Blackburn (photo-left), the discovery of “jumping genes” by Barbara McClintock (photo-middle), and the discovery of HIV by Françoise Barré-Sinoussi (photo-right).
 
The link between such disparate physiological and pathophysiological phenomena is cellular stress-induced modulation of energy metabolism, leading to the activation of the master metabolic regulator AMPK, a kinase that increases lifespan and healthspan in several model organisms [12]. I was the first to propose and publish (2014) that an increase in beneficial levels of cellular stress (e.g. increases in the levels reactive oxygen species [ROS], calcium [Ca2+], and/or an AMP(ADP)/ATP ratio increase, etc.) and activation of AMPK by compounds including metformin would alleviate accelerated cellular aging defects in children diagnosed with the genetic disorder Hutchinson-Gilford progeria syndrome (HGPS), a disease characterized by an accelerated aging phenotype and death at ~14.6 years of age [1]. This hypothesis was substantiated in 2016 and 2017, with metformin activating AMPK in cells taken from HGPS kids and ameliorating accelerated cellular aging defects (e.g. correcting nuclear morphology, decrease in senescence markers, etc.) [8,9]. Additionally, transfection of telomerase been shown to reverse senescence in HGPS cells [65]. As metformin activates telomerase and normal humans make the same toxic protein (called progerin) that leads to accelerated aging in HGPS kids (just at lower amounts that accumulate with age), AMPK activation may also play a significant role in ameliorating diseases associated with physiological aging [10,11].

AMPK also links HGPS with potential virus eradication. I first proposed in 2015 that AMPK activation links alleviation of accelerated aging in HGPS with the potential eradication of HIV-1 via the “shock and kill” approach, a method currently being pursued by HIV cure researchers to possibly eradicate HIV-1 [2,13]. The same gene splicing factor that promotes accelerated aging in HGPS (called SRSF1 or ASF/SF2) also inhibits reactivation of latent HIV-1 (i.e. “shock”), preventing immune system detection and virus destruction (i.e. “kill”) [8,14]. Metformin was shown to slow aging in HGPS cells by decreasing this splicing factor, as I originally predicted in 2014, and several compounds that potently induce latent HIV-1 reactivation in T cells from infected patients, including PMA (a phorbol ester) combined with ionomycin, each activate AMPK [1,8,15-17]. AMPKα1 deletion leads to a decrease in primary T cell responses to bacterial and viral infections in vivo, AMPK knockdown leads to cell death on T cell activation, and metformin has recently been shown to inhibit Zika and Dengue viruses, the malaria parasite, and Legionella pneumophila [18-23]. Intriguingly, early data presented at the International AIDS Conference in 2017 demonstrated that metformin destabilized the latent HIV-1 reservoir in chronically-infected HIV patients and decreased the percentage of CD4+ T cells expressing the immune checkpoint receptors PD-1, TIGIT, and TIM-3, each markers associated with T cells latently infected with HIV-1, indicating that AMPK activation may indeed contribute to a cure for HIV-1 [24,25].

As I first proposed in 2016 and 2017, the induction of cellular stress and AMPK activation also links HGPS and potential HIV-1 eradication with oocyte activation and the sperm acrosome reaction, prerequisites for the creation of human life [3,4]. Increases in both ROS and Ca2+ are critical for T cell activation (and hence latent HIV-1 reactivation) and ROS is transiently increased in HGPS cells when treated with a rapamycin analog to alleviate accelerated aging [26-28]. Stress-induced activation of AMPK by AICAR and other compounds promotes oocyte maturation, which precedes and is essential for efficient oocyte activation [29,30]. Oocyte activation is indispensable for the creation of all human life and PMA and ionomycin, which collectively reactivates latent HIV-1, activates mouse and human oocytes, respectively [31,32]. AMPK is also found in the acrosome of the human sperm head and ionomycin induces the acrosome reaction in human sperm, a process necessary for oocyte penetration and fertilization [33,34]. Ionomycin is also used extensively during fertility procedures to activate human oocytes (i.e. “shock”), creating normal, healthy children (i.e. “live”) [32]. Interestingly, ionomycin is a narrow spectrum antibiotic produced by certain species within the bacterial genus Streptomyces, from which ~70 percent of clinically useful antibiotics are derived [35,36]. Cellular stress, mediated by increases in ROS, Ca2+, and/or an AMP(ADP)/ATP ratio increase, etc. also enhances antibiotic production in many Streptomyces strains, reinforcing the notion that the beneficial effects of cellular stress induction crosses species boundaries [37,38].

Cellular stress induction and AMPK activation also link HGPS, potential HIV-1 eradication, and human life creation with learning and memory, a hypothesis I originally proposed in 2018 [6]. Hippocampal long-term potentiation (LTP) is considered the cellular correlate of learning and memory and AMPK has been found localized in hippocampal CA1 dendrites and is activated in neurons by metformin, AICAR, ionomycin, and glutamate, a neurotransmitter essential for hippocampal LTP induction [39-41]. The glutamate receptors AMPAR and NMDAR are found on and modulate T cell activation, AMPK activation increases synthesis and membrane insertion of AMPARs (critical for LTP expression), PMA enhances hippocampal CA1 LTP, and inhibition of ROS significantly impairs hippocampal CA1 LTP [42-46]. Also, neuronal depolarization decreases the recruitment efficiency of SRSF1 to nascent RNAs and promotes SRSF1 nuclear speckle accumulation [6]. SRSF1, a gene splicing factor that is inhibited by metformin, enhances progerin production in HGPS cells and prevents latent HIV-1 reactivation [2,8]. Metformin also significantly reduces pathology-associated reductions in LTP in animal models in vivo, indicating that learning and memory are linked to HGPS, potential HIV-1 eradication, and human life creation via the induction of beneficial levels of cellular stress [47].

Lastly, cellular stress and AMPK activation also links the activation and mobilization of transposable elements (i.e. “jumping genes”) with telomerase activation, potential HIV-1 eradication, learning and memory, and the creation of human life, a hypothesis I originally proposed in 2018 [6]. Transposable elements (TEs) are DNA sequences first described by Nobel laureate Barbara McClintock that comprise nearly half of the human genome, are able to transpose or move from one genomic location to another, and have played an extensive role in human genome evolution [48-50]. Strikingly, McClintock also described in her Nobel Prize speech that a genome “shock” seemed to promote TE activation and mobilization [50]. As first noted in my recently published paper, this “shock” is the same “shock” that HIV cure researchers are using during the “shock and kill” approach to reactivate latent HIV-1 to potentially effectuate a cure [6]. Indeed, several forms of cellular stress, including heat shock and radiation, have been convincingly shown to activate and enhance TE mobilization in several model organisms and in human cells [51-53]. This same “shock” McClintock referred to, mediated by increases in ROS, Ca2+, and/or an AMP(ADP)/ATP ratio, etc. is also what leads to the creation of human life, as the antibiotic ionomycin activates AMPK, promotes TE activation, and induces human oocyte activation [17,32,54]. LINE-1 (L1), a member of the retrotransposon class of TEs, is active and capable of mobilization in human oocytes, human sperm, and in human neural progenitor cells [55-57]. Inhibition of L1 impairs both oocyte maturation in vitro and long-term memory formation in vivo in mice [58,59]. L1 has also been detected in the human brain and is capable of mobilization in human neurons [57]. As noted above, AMPK is critical for oocyte maturation and metformin promotes hippocampal neurogenesis and spatial memory formation [29,60]. The landmark initial sequencing of the human genome also noted that both telomerase and RAG1 (promotes DNA cleavage and transposition in human cells) are derived from TEs [49]. Because metformin activates both telomerase and RAG1 via AMPK, it is likely that cellular stress-induced AMPK activation facilitates beneficial TE activation and mobilization (i.e. learning and memory associated with L1 mobilization), linking human genome evolution and the creation of human life with hippocampal LTP, HGPS, and potential HIV-1 eradication [61,62].

https://www.linkedin.com/pulse/metformin-ampk-link-nobel-prize-winning-telomeres-jumping-finley/


References 
  1. 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.
  2. 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.
  3. 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.
  4. Finley J. Elimination of cancer stem cells and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking inhibition of tumorigenesis and the potential eradication of HIV-1. Med Hypotheses. 2017 Jul;104:133-146.
  5. Finley J. AMPK activation as a common mechanism of action linking the effects of diverse compounds that ameliorate accelerated cellular aging defects in Hutchinson-Gilford progeria syndrome. Med Hypotheses. Manuscript submitted.
  6. Finley J. Facilitation of hippocampal long-term potentiation and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking learning, memory, and the potential eradication of HIV-1. Med Hypotheses. 2018 Jul;116: 61-73.
  7. Finley J. Transposable elements, placental development, and oocyte activation: Cellular stress and AMPK links jumping genes with the creation of human life. Med Hypotheses. Under Review.  
  8. Egesipe AL, Blondel S, 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.
  9. Park SK, Shin OS. Metformin alleviates ageing cellular phenotypes in Hutchinson-Gilford progeria syndrome dermal fibroblasts. Exp Dermatol. 2017 Oct;26(10):889-895.
  10. McClintock D, Ratner D, Lokuge M, et al. The mutant form of lamin A that causes Hutchinson-Gilford progeria is a biomarker of cellular aging in human skin. PLoS One. 2007 Dec 5;2(12):e1269.
  11. 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.
  12. Salminen A, Kaarniranta K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev. 2012 Apr;11(2):230-41.
  13. Kim Y, Anderson JL, Lewin SR. Getting the "Kill" into "Shock and Kill": Strategies to Eliminate Latent HIV. Cell Host Microbe. 2018 Jan 10;23(1):14-26.
  14. 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;80(7):3189–204.
  15. Spina CA, Anderson J, Archin NM, et al. An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog 2013;9(12):e1003834.
  16. Zogovic N, Tovilovic-Kovacevic G, Misirkic-Marjanovic M, et al. Coordinated activation of AMP-activated protein kinase, extracellular signal-regulated kinase, and autophagy regulates phorbol myristate acetate-induced differentiation of SH-SY5Y neuroblastoma cells. J Neurochem 2015;133 (2):223–32.
  17. Tamás P, Hawley SA, Clarke RG, et al. Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med. 2006 Jul 10;203(7):1665-70.
  18. Blagih J, Coulombe F, Vincent EE, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity. 2015 Jan 20;42(1):41-54.
  19. 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.
  20. Cheng F, Ramos da Silva S, Huang IC, Jung JU, Gao SJ. Suppression of Zika virus infection and replication in endothelial cells and astrocytes by PKA inhibitor PKI 14-22. J Virol. 2017 Dec 6. pii: JVI.02019-17.
  21. Soto-Acosta R, Bautista-Carbajal P, Cervantes-Salazar M, Angel-Ambrocio AH, Del Angel RM. DENV up-regulates the HMG-CoA reductase activity through the impairment of AMPK phosphorylation: A potential antiviral target. PLoS Pathog. 2017 Apr 6;13(4):e1006257.
  22. Ruivo MT, Vera IM, Sales-Dias J, et al. Host AMPK Is a Modulator of Plasmodium Liver Infection. Cell Rep. 2016 Sep 6;16(10):2539-45.
  23. Kajiwara C, Kusaka Y, Kimura S, et al. Metformin Mediates Protection against Legionella Pneumonia through Activation of AMPK and Mitochondrial Reactive Oxygen Species. J Immunol. 2017 Dec 15. pii: ji1700474.
  24. 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.
  25. http://viruseradication.com/abstract-details.php?abstract_id=1188, last accessed March 14, 2018.
  26. Oh-hora M, Rao A. Calcium signaling in lymphocytes. Curr Opin Immunol 2008;20(3):250–8.
  27. 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.
  28. Gabriel D, Gordon LB, Djabali K. Temsirolimus Partially Rescues the Hutchinson-Gilford Progeria Cellular Phenotype. PLoS One. 2016 Dec 29;11(12):e0168988.
  29. Chen J, Hudson E, Chi MM, et al. AMPK regulation of mouse oocyte meiotic resumption in vitro. Dev Biol. 2006 Mar 15;291(2):227-38.
  30. LaRosa C, Downs SM. Stress stimulates AMP-activated protein kinase and meiotic resumption in mouse oocytes. Biol Reprod. 2006 Mar;74(3):585-92.
  31. Uranga JA, Pedersen RA, Arechaga J. Parthenogenetic activation of mouse oocytes using calcium ionophores and protein kinase C stimulators. Int J Dev Biol. 1996 Apr;40(2):515-9.
  32. Deemeh MR, Tavalaee M, Nasr-Esfahani MH. Health of children born through artificial oocyte activation: a pilot study. Reprod Sci. 2015 Mar;22(3):322-8.
  33. 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.
  34. Sánchez-Cárdenas C, Servín-Vences MR, José O, Treviño CL, Hernández-Cruz A, Darszon A. Acrosome reaction and Ca²+ imaging in single human spermatozoa: new regulatory roles of [Ca²+]i. Biol Reprod. 2014 Sep;91(3):67.
  35. Liu WC, Slusarchyk DS, Astle G, Trejo WH, Brown WE, Meyers E. Ionomycin, a new polyether antibiotic. J Antibiot (Tokyo). 1978 Sep;31(9):815-9.
  36. Kitani S, Miyamoto KT, Takamatsu S, et al. Avenolide, a Streptomyces hormone controlling antibiotic production in Streptomyces avermitilis. Proc Natl Acad Sci U S A. 2011 Sep 27;108(39):16410-5.
  37. Meng L, Li M, Yang SH, Kim TJ, Suh JW. Intracellular ATP levels affect secondary metabolite production in Streptomyces spp. Biosci Biotechnol Biochem. 2011;75(8):1576-81.
  38. Wang D, Wei L, Zhang Y, Zhang M, Gu S. Physicochemical and microbial responses of Streptomyces natalensis HW-2 to fungal elicitor. Appl Microbiol Biotechnol. 2017 Sep;101(17):6705-6712.
  39. Potter WB, O'Riordan KJ, Barnett D, et al. Metabolic regulation of neuronal plasticity by the energy sensor AMPK. PLoS One. 2010 Feb 1;5(2):e8996.
  40. Sample V, Ramamurthy S, Gorshkov K, Ronnett GV, Zhang J. Polarized activities of AMPK and BRSK in primary hippocampal neurons. Mol Biol Cell. 2015 May 15;26(10):1935-46
  41. Terunuma M, Vargas KJ, Wilkins ME, et al. Prolonged activation of NMDA receptors promotes dephosphorylation and alters postendocytic sorting of GABAB receptors. Proc Natl Acad Sci U S A. 2010 Aug 3;107(31):13918-23.
  42. Ganor Y, Besser M, Ben-Zakay N, Unger T, Levite M. Human T cells express a functional ionotropic glutamate receptor GluR3, and glutamate by itself triggers integrin-mediated adhesion to laminin and fibronectin and chemotactic migration. J Immunol. 2003 Apr 15;170(8):4362-72.
  43. Miglio G, Varsaldi F, Lombardi G. Human T lymphocytes express N-methyl-D-aspartate receptors functionally active in controlling T cell activation. Biochem Biophys Res Commun. 2005 Dec 30;338(4):1875-83.
  44. Wang G, Amato S, Gilbert J, Man HY. Resveratrol up-regulates AMPA receptor expression via AMP-activated protein kinase-mediated protein translation. Neuropharmacology. 2015 Aug;95:144-53.
  45. Kim EC, Lee MJ, Shin SY, et al. Phorbol 12-Myristate 13-Acetate Enhances Long-Term Potentiation in the Hippocampus through Activation of Protein Kinase Cδ and ε. Korean J Physiol Pharmacol. 2013 Feb;17(1):51-6.
  46. Klann E. Cell-permeable scavengers of superoxide prevent long-term potentiation in hippocampal area CA1. J Neurophysiol. 1998 Jul;80(1):452-7.
  47. Asadbegi M, Yaghmaei P, Salehi I, Ebrahim-Habibi A, Komaki A. Neuroprotective effects of metformin against Aβ-mediated inhibition of long-term potentiation in rats fed a high-fat diet. Brain Res Bull. 2016 Mar;121:178-85.
  48. Muñoz-López M, García-Pérez JL. DNA transposons: nature and applications in genomics. Curr Genomics. 2010 Apr;11(2):115-28.
  49. Lander ES, Linton LM, Birren B, et al. Initial sequencing and analysis of the human genome. Nature. 2001 Feb 15;409(6822):860-921.
  50. McClintock B. The significance of responses of the genome to challenge. Science. 1984 Nov 16;226(4676):792-801.
  51. Stanley D, Fraser S, Stanley GA, Chambers PJ. Retrotransposon expression in ethanol-stressed Saccharomyces cerevisiae. Appl Microbiol Biotechnol. 2010 Jul;87(4):1447-54.
  52. Vasilyeva LA, Bubenshchikova EV, Ratner VA. Heavy heat shock induced retrotransposon transposition in Drosophila. Genet Res. 1999 Oct;74(2):111-9.
  53. Farkash EA, Kao GD, Horman SR, Prak ET. Gamma radiation increases endonuclease-dependent L1 retrotransposition in a cultured cell assay. Nucleic Acids Res. 2006 Feb 28;34(4):1196-204.
  54. Magun BE, Rodland KD. Transient inhibition of protein synthesis induces the immediate early gene VL30: alternative mechanism for thapsigargin-induced gene expression. Cell Growth Differ. 1995 Jul;6(7):891-7.
  55. Georgiou I, Noutsopoulos D, Dimitriadou E, et al. Retrotransposon RNA expression and evidence for retrotransposition events in human oocytes. Hum Mol Genet. 2009 Apr 1;18(7):1221-8.
  56. Lazaros L, Kitsou C, Kostoulas C, et al. Retrotransposon expression and incorporation of cloned human and mouse retroelements in human spermatozoa. Fertil Steril. 2017 Mar;107(3):821-830.
  57. Coufal NG, Garcia-Perez JL, Peng GE, et al. L1 retrotransposition in human neural progenitor cells. Nature. 2009 Aug 27;460(7259):1127-31.
  58. Luo YB, Zhang L, Lin ZL, et al. Distinct subcellular localization and potential role of LINE1-ORF1P in meiotic oocytes. Histochem Cell Biol. 2016 Jan;145(1):93-104.
  59. Bachiller S, Del-Pozo-Martín Y, Carrión ÁM. L1 retrotransposition alters the hippocampal genomic landscape enabling memory formation. Brain Behav Immun. 2017 Aug;64:65-70.
  60. Ahmed S, Mahmood Z, Javed A, et al. Effect of Metformin on Adult Hippocampal Neurogenesis: Comparison with Donepezil and Links to Cognition. J Mol Neurosci. 2017 May;62(1):88-98.
  61. 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.
  62. Um JH, Brown AL, Singh SK, et al. Metabolic sensor AMPK directly phosphorylates RAG1 protein and regulates V(D)J recombination. Proc Natl Acad Sci U S A. 2013 Jun 11;110(24):9873-8.
  63. O'Sullivan RJ, Karlseder J. Telomeres: protecting chromosomes against genome instability. Nat Rev Mol Cell Biol. 2010 Mar;11(3):171-81.  
  64. 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.
  65. 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.





No comments:

Post a Comment