Wednesday, January 3, 2018

Metformin shown for the first time to inhibit Legionella infection via AMPK : AMPK links pathogen destruction with Progeria & Human Life Creation

CC-BY-SA-3.0 http://creativecommons.org/licenses/by-sa/3.0/)], via Wikimedia Commons; By CDC/ Dr. Barry S. Fields [Public domain], via Wikimedia Commons

In line with recent evidence demonstrating that the AMPK activator metformin inhibits Zika virus, Dengue virus, and malaria parasite replication (see below), a study published in The Journal of Immunology in December of 2017 showed for the first time that the anti-diabetic drug metformin significantly suppressed the growth of the bacterium Legionella pneumophila (L. pneumophila) in immune cells derived from both mice and humans by activating AMPK [1]. L. pneumophila is a Gram-negative bacterium that is typically found in water-associated environments and may contaminate hot water tanks and air-conditioning units for large buildings and is the causative agent of Legionnaires' disease [1,2]. Legionnaires' disease is a form of atypical pneumonia characterized by fever, cough, and shortness of breath and is usually acquired by inhalation of small air-borne water droplets [2]. According to the Centers for Disease Control and Prevention (CDC), “the bacterium is named after a 1976 outbreak, during which some people who went to a Philadelphia convention of the American Legion suffered from a new type of pneumonia (lung infection) that became known as Legionnaires’ disease.” [3]. Interestingly, the authors of the study observed that metformin increased the production of mitochondrial reactive oxygen species (ROS) in L. pneumophila-infected immune cells and that inhibition of both AMPK activation and ROS production negated metformin-mediated growth suppression of L. pneumophila [1]. Most importantly, metformin significantly reduced bacterial number, activated AMPK, and increased ROS in the lungs of infected mice, thus improving survival and indicating that metformin inhibits L. pneumophila replication in vivo in an AMPK-dependent manner. Metformin also increased mitochondrial ROS in uninfected immune cells, suggesting that cellular stress induced AMPK activation (via increases in ROS, calcium[Ca2+], and/or an AMP/ATP ratio increase, etc) is critical for mounting an effective immune response to bacteria, viruses, and other pathogens [1].

Strikingly, metformin activates AMPK and alleviates accelerating aging defects in cells from children with the genetic disorder Hutchinson-Gilford progeria syndrome (HGPS), promotes the differentiation and/or apoptosis of cancer stem cells in an AMPK-dependent manner, and destabilizes the latent HIV-1 reservoir in chronically-infected HIV-1 patients (facilitating virus elimination and potentially contributing to an HIV-1 cure). Also, because AMPK is critical for oocyte maturation and bacteria-derived antibiotics (e.g. ionomycin, A23187) that activate AMPK are used extensively to activate human oocytes to create normal, healthy babies, it is likely that stress-induced AMPK activation (e.g. via ROS, intracellular Ca2+, and/or AMP/ATP ratio increase, etc.) represents a common mechanism linking pathogen elimination with HGPS, caner stem cell elimination, HIV-1 eradication, and the creation of all human life, as I originally proposed in several recent publications (see below) [4-7].

As noted above, in addition to inhibition of L. pneumophila replication, metformin also potently inhibited ZIKV replication in HUVECs and AMPK activation has recently been found to exert significant antiviral effects against Rift Valley Fever virus as well as multiple arbovirus family members including the Flavivirus Kunjin virus, the Togavirus Sindbis virus, and the Rhabdovirus Vesicular stomatitis virus [8,9]. Interestingly, as both ZIKV and dengue virus (DENV) are transmitted by the same mosquito vector, a study recently published in the journal PLoS Pathogens in April of 2017 demonstrated for the first time that metformin exerted significant antiviral effects in DENV-infected human liver cells that was dependent on activation of AMPK [10]. The authors showed that an increase in HMG-CoA reductase (HMGCR) activity, a target of AMPK, was associated with DENV-infected cells, AMPK activation was reduced in DENV-infected cells at 12 and 24 hours post infection (hpi), and metformin significantly decreased the number of infected cells, viral yield, and viral genome copies, leading the authors to conclude that metformin-induced AMPK activation generates a strong antiviral effect against DENV [10].

Recent efforts funded by the U.S. and British governments, the Bill & Melinda Gates Foundation, and the Google health spin-off Verily have sought to decrease the spread of dengue and Zika viruses through the coordinated release of female and/or male mosquitoes (called Aedes aegypti) that were purposely infected with a bacterium that inhibits the mosquito’s ability to transmit the two viruses to humans [11,12]. Studies have shown that this bacterium, called Wolbachia, enhances the mosquito’s immune response by increasing the levels of reactive oxygen species (ROS), thus enhancing inhibition of DENV replication [13]. Because AMPK is activated by cellular stress (e.g. ROS increase, intracellular calcium [Ca2+] increase, AMP/ATP ratio increase, etc.), has been found in Aedes aegypti (Ae. aegypti), and AMPK activation by stress-inducing compounds (e.g. resveratrol) increased average life span and enhanced the immune response in Ae. aegypti in an AMPK-dependent manner, the recent finding that metformin also inhibits DENV replication in human cells in an AMPK-dependent manner provides compelling evidence that the anti-viral and antimicrobial effects of AMPK activation likely crosses species boundaries [14]. 

Moreover, a study published in the journal Cell Reports in September of 2016 by researchers from the Massachusetts Institute of Technology (MIT) and the University of Lisbon also showed for the first time that metformin and other AMPK activators significantly reduced parasite load in human liver cells of different species of Plasmodium, a protozoan parasite that is the etiological agent of malaria [15]. Importantly, the authors also showed that AMPK activation inhibits growth and replication of different Plasmodium spp. (species) and AMPK activators as well as dietary restriction, which activates AMPK, reduces Plasmodium berghei (malaria-causing species in rodents often used as a model for the study of human malaria) infection in mice [15]. The AMPK-activating compounds salicylate and A769662 also reduced P. berghei and P. falciparum (malaria parasite that infects humans) merozoite formation (infectious parasites generated through replication in erythrocytes) in vitro while salicylate decreased parasitaemia in mice in vivo [16].

Cellular stress-induced AMPK activation has also been shown to exert antiviral effects against HIV-1. AMPK activation and several AMPK-activating compounds, including EGCG, curcumin, tanshinone II A (derived from the plant Salvia miltiorrhiza), bryostatin-1 (isolated from the marine organism Bugula neritina), and resveratrol (found in grapes and in the plant Polygonum cuspidatum) have been shown to exhibit antiviral activity in vitro against HIV-1 [17-21].

An active area among HIV-1 cure researchers, known as the “shock and kill” approach, involves reactivating (i.e. “shock”) a T cell (or another immune cell) that harbors dormant HIV-1, hence reactivating the virus itself and thus inducing destruction of the T cell along with the virus or enhancing recognition and destruction of the virus-infected T cell by the immune system (i.e. “kill”) [5]. Strikingly, AMPK is also critical for the activation of T cells and the mounting of an effective immune response to eliminate viruses, bacteria, and cancer cells [6,22,23]. A recent study demonstrated that metformin, when combined with the protein kinase C modulator bryostatin, induced reactivation of latent HIV-1 in a monocytic cell line in an AMPK-dependent manner. Bryostatin was also shown to induce phosphorylation and activation of AMPK in that study, implying that bryostatin is an indirect AMPK activator as well [20]. Furthermore, the calcium ionophores ionomycin and A23187, both of which activate AMPK and induce human oocyte activation, are often combined with phorbol 12-myristate 13-acetate (PMA) and are extremely efficient in promoting T cell activation-induced latent HIV-1 reactivation [24-26].

Perhaps most convincingly, at the International AIDS Society’s (IAS) HIV Cure and Cancer Forum held in Paris, France in July of 2017, researchers from the University of Hawaii demonstrated for the first time that metformin decreased the percentage of CD4+ T cells expressing the immune checkpoint receptors PD-1, TIGIT, and TIM-3 in HIV-1 patients, receptors that are positively associated with T cells that harbor latent HIV-1. Metformin also destabilized the latent viral reservoir in chronically-infected HIV-1 patients, indicating that metformin may indeed contribute to HIV-1 eradication by inducing an AMPK-mediated reactivation of latent HIV-1, as I initially proposed in 2015 and 2016 [5,6,27-31].

Also, stress-induced AMPK activation likely links latent HIV-1 reactivation with alleviation of accelerated aging defects in cells derived from children with the genetic disorder Hutchinson-Gilford progeria syndrome (HGPS). Studies have shown that efficient reactivation of latent HIV-1 involves a reduction in the splicing of the HIV-1 genome by the gene splicing factor SRSF1 [32-34]. Accelerated cellular aging-like phenotypes in HGPS are primarily linked to aberrant splicing of the LMNA gene, leading to the over production of a toxic protein called progerin [4]. Evidence has also shown that inhibition of the splicing factor SRSF1 leads to a reduction in progerin at both the mRNA and protein levels [5,35].

A recent study published online in the Journal npj Aging and Mechanisms of Disease in November of 2016 provided startling evidence that metformin decreased the expression of progerin and SRSF1 and alleviated pathological defects in cells derived from HGPS patients [36]. Another study published online in the Journal Experimental Dermatology in February of 2017 confirmed that metformin alleviated nuclear defects and premature aging phenotypes and activated AMPK in fibroblasts derived from HGPS patients, substantiating my original hypotheses from 2014 and 2015 proposing that AMPK activators including metformin would improve accelerated aging defects in HGPS cells by inhibiting SRSF1 and activating AMPK [4,5,37]. Temsirolimus, an analog of the macrolide rapamycin (which activates AMPK in vivo), also partially rescued the HGPS cellular phenotype but significantly increased the levels of ROS and superoxide within the first hour of treatment, providing further indication that the induction of cellular stress and subsequent AMPK activation links virus and pathogen elimination with alleviation of accelerated cellular aging defects in HGPS [38,39].

Furthermore, ROS and calcium are well-studied mediators of cellular stress-induced differentiation of embryonic and adult stem cells, AMPK has recently been shown to be essential for mouse embryonic stem cell differentiation, and metformin targets and promotes differentiation and/or apoptosis of cancer stem cells in the deadliest of cancers in an AMPK-dependent manner, including glioblastoma and pancreatic cancer [7]. Such evidence strongly suggests that cellular stress-induced AMPK activation by compounds including metformin links pathogen and virus elimination with HGPS and cancer stem cell differentiation and/or apoptosis, a hypothesis that I first proposed in 2017 [7].

Lastly, AMPK activation also promotes oocyte meiotic induction and maturation (processes that are critical for efficient oocyte activation) and AMPK has recently been found localized across the entire acrosome in human spermatozoa [6,40,41].  The induction of cellular stress (e.g. increases in ROS, intracellular calcium, and/or AMP/ATP ratio increase), which activates AMPK, also promotes oocyte meiotic induction/maturation, oocyte activation, and the acrosome reaction in human sperm, processes critical for the creation of all human life [40,42,43]. Indeed, oocyte activation is indispensable for the creation of all human life and the bacteria-derived calcium ionophore ionomycin, which activates AMPK, is commonly used to promote latent HIV-1 reactivation and is extensively used to activate human oocytes, creating normal healthy children (i.e. the “shock and live” approach) [43-47]. Such evidence indicates and further substantiates the novel and provocative assertion that AMPK activation links L. pneumophila inhibition and pathogen elimination with the amelioration of accelerated aging defects in HGPS cells, HIV-1 latency and replication, adult and cancer stem cells, and the creation of all human life [1,4-7].

https://www.linkedin.com/pulse/metformin-shown-first-time-inhibit-legionella-infection-finley/



References:

  1. 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. doi: 10.4049/jimmunol.1700474. [Epub ahead of print].
  2. https://www.cdc.gov/legionella/about/causes-transmission.html
  3. https://www.cdc.gov/legionella/index.html
  4. 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.
  5. 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.
  6. 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.
  7. 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.
  8. 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. doi: 10.1128/JVI.02019-17. [Epub ahead of print].
  9. Moser TS, Schieffer D, Cherry S. AMP-activated kinase restricts Rift Valley fever virus infection by inhibiting fatty acid synthesis. PLoS Pathog. 2012;8(4):e1002661.
  10. 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.
  11. http://www.reuters.com/article/us-health-dengue-mosquitoes-idUSKCN12Q1PE
  12. https://www.bloomberg.com/news/articles/2016-10-06/alphabet-s-verily-joins-zika-fight-with-sterile-mosquito-lab
  13. Pan X, Zhou G, Wu J, et al. Wolbachia induces reactive oxygen species (ROS)-dependent activation of the Toll pathway to control dengue virus in the mosquito Aedes aegypti. Proc Natl Acad Sci U S A. 2012 Jan 3;109(1):E23-31.
  14. Nunes RD, Ventura-Martins G, Moretti DM, et al. Polyphenol-Rich Diets Exacerbate AMPK-Mediated Autophagy, Decreasing Proliferation of Mosquito Midgut Microbiota, and Extending Vector Lifespan. PLoS Negl Trop Dis. 2016 Oct 12;10(10):e0005034.
  15. 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.
  16. Mancio-Silva L, Slavic K, Grilo Ruivo MT, et al. Nutrient sensing modulates malaria parasite virulence. Nature. 2017 Jul 13;547(7662):213-216.
  17. Zhang HS, Wu TC, Sang WW, Ruan Z. EGCG inhibits Tat-induced LTR transactivation: role of Nrf2, AKT, AMPK signaling pathway. Life Sci. 2012 May 22;90(19-20):747-54.
  18. Zhang HS, Ruan Z, Sang WW. HDAC1/NFκB pathway is involved in curcumin inhibiting of Tat-mediated long terminal repeat transactivation. J Cell Physiol. 2011 Dec;226(12):3385-91.
  19. Zhang HS, Chen XY, Wu TC, Zhang FJ. Tanshinone II A inhibits tat-induced HIV-1 transactivation through redox-regulated AMPK/Nampt pathway. J Cell Physiol. 2014 Sep;229(9):1193-201.
  20. 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.
  21. Zhang HS, Wu MR. SIRT1 regulates Tat-induced HIV-1 transactivation through activating AMP-activated protein kinase. Virus Res. 2009 Dec;146(1-2):51-7.
  22. Pearce EL, Walsh MC, Cejas PJ, et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009 Jul 2;460(7251):103-7.
  23. 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.
  24. Gómez-Gonzalo M, Carretero M, Rullas J et al. The hepatitis B virus X protein induces HIV-1 replication and transcription in synergy with T-cell activation signals: functional roles of NF-kappaB/NF-AT and SP1-binding sites in the HIV-1 long terminal repeat promoter. J Biol Chem. 2001 Sep 21;276(38):35435-43.
  25. 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.
  26. Fogarty S, Hawley SA, Green KA, Saner N, Mustard KJ, Hardie DG. Calmodulin-dependent protein kinase kinase-beta activates AMPK without forming a stable complex: synergistic effects of Ca2+ and AMP. Biochem J. 2010 Jan 27;426(1):109-18.
  27. 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.
  28. http://viruseradication.com/supplement-details/Abstracts_of_the_IAS_HIV_Cure_and_Cancer_Forum_2017/
  29. http://www.iasociety.org/HIV-Programmes/Towards-an-HIV-Cure/Events/HIV-Cure-Cancer-Forum
  30. Fromentin R, Bakeman W, Lawani MB, et al. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART. PLoS Pathog. 2016 Jul 14;12(7):e1005761.
  31. Chew GM, Fujita T, Webb GM, et al. TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection. PLoS Pathog. 2016 Jan 7;12(1):e1005349.
  32. 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.
  33. 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.
  34. Hu M, Crawford SA, Henstridge DC, et al. p32 protein levels are integral to mitochondrial and endoplasmic reticulum morphology, cell metabolism and survival. Biochem J. 2013 Aug 1;453(3):381-91.
  35. Lopez-Mejia IC, Vautrot V, De Toledo M, et al. A conserved splicing mechanism of the LMNA gene controls premature aging. Hum Mol Genet. 2011 Dec 1;20(23):4540-55.
  36. 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.
  37. Park SK, Shin OS. Metformin Alleviates Ageing Cellular Phenotypes in Hutchinson-Gilford Progeria Syndrome Dermal Fibroblasts. Exp Dermatol. 2017 Oct;26(10):889-895.
  38. Gabriel D, Gordon LB, Djabali K. Temsirolimus Partially Rescues the Hutchinson-Gilford Progeria Cellular Phenotype. PLoS One. 2016 Dec 29;11(12):e0168988.
  39. 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.
  40. LaRosa C, Downs SM. Stress stimulates AMP-activated protein kinase and meiotic resumption in mouse oocytes. Biol Reprod. 2006 Mar;74(3):585-92.
  41. 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. 2016 Sep 27. doi: 10.4103/1008-682X.185848. [Epub ahead of print].
  42. de Lamirande E, Tsai C, Harakat A, Gagnon C. Involvement of reactive oxygen species in human sperm arcosome reaction induced by A23187, lysophosphatidylcholine, and biological fluid ultrafiltrates. J Androl. 1998 Sep-Oct;19(5):585-94.
  43. Tesarik J, Sousa M, Testart J. Human oocyte activation after intracytoplasmic sperm injection. Hum Reprod. 1994 Mar;9(3):511-8.
  44. 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.
  45. 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.
  46. 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.
  47. 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.