Thursday, April 13, 2017

Metformin shown for the first time to restore mitochondria in human fetal cells with Down syndrome: AMPK links Progeria, Down syndrome, & Dengue virus

By Vanellus Foto (Own work) [GFDL (http://www.gnu.org/copyleft/fdl.html) or CC BY-SA 3.0 (http://creativecommons.org/licenses/by-sa/3.0)], via Wikimedia Commons; "Hutchinson-Gilford Progeria Syndrome" by The Cell Nucleus and Aging: Tantalizing Clues and Hopeful Promises. Scaffidi P, Gordon L, Misteli T

In line with recent evidence demonstrating for the first time that metformin improved accelerated aging defects, activated AMPK, and beneficially altered gene splicing in cells derived from Hutchinson-Gilford progeria syndrome (HGPS) patients and exerted significant antiviral effects in dengue virus-infected human liver cells in an AMPK-dependent manner, a study published in the journal Human Molecular Genetics in January of 2017 showed for the first time that metformin corrected mitochondrial dysfunction in human fetal cells taken from aborted fetuses that had been diagnosed with Down syndrome (DS) [1-4]. Metformin induced the expression and activity of PGC-1α (a master regulator of mitochondrial biogenesis) and the PGC-1α target genes NRF-1 and TFAM, enhanced oxygen consumption and ATP production, reversed mitochondrial fragmentation, and increased the expression of the mitochondrial fusion genes OPA1 and MFN2, leading to enhanced mitochondrial activity and strongly promoting mitochondrial biogenesis. Recent studies have also shown that the polyphenols resveratrol and epigallocatechin-3-gallate (EGCG) restored mitochondrial functionality and improved hippocampal neural progenitor cell proliferation in a mouse model of DS in an AMPK-dependent manner, while EGCG significantly reversed cognitive deficits in human DS patients [5,6].

Interestingly, as metformin inhibits dengue virus replication in human liver cells in an AMPK-dependent manner, both EGCG and resveratrol have been shown to increase the lifespan of the mosquito Aedes aegypti (a vector for dengue and Zika viruses), with resveratrol also boosting the immune response and activating AMPK in Aedes aegypti [7]. Recent evidence also demonstrated that resveratrol inhibits dengue virus replication in human liver cells, inhibits emtricitabine-resistant HIV-1, and reactivates latent HIV-1, facilitating immune system detection and destruction [8-10]. EGCG has also shown anti-HIV-1 activity [11]. Although structurally dissimilar, metformin, EGCG, and resveratrol each activate AMPK and AMPK activation links reversal of accelerated aging defects in HGPS with rescue of mitochondrial function in Down syndrome, inhibition of dengue virus and HIV-1 replication, and reactivation of latent HIV-1. Such an interconnectedness, as further explained below, provides substantial support for my initial proposal and publications that AMPK activation links HGPS, latent HIV-1 reactivation, Down syndrome (manuscript in press) and the beginnings of all human life (via oocyte activation and the acrosome reaction) [12-14].

Down syndrome (DS) is caused by either a partial or full trisomy of chromosome 21 and is associated with impairments in cognition, learning and memory, and disorders of the immune system [1]. Human and animal studies strongly suggest that mitochondrial dysfunction is associated with pathogenic features of DS and mitochondrial abnormalities have been found in all DS cells analyzed in culture to date, including neurons, astrocytes, and lymphocytes [1]. Indeed, in DS human fetal fibroblasts, PGC-1α, a master regulator of mitochondrial biogenesis, is down regulated at both the mRNA and protein levels and AMPK phosphorylation of the PGC-1α protein is essential for PGC-1α-dependent induction of the PGC-1α promoter [1].

In the Human Molecular Genetics study, Izzo et al. initially observed that DS human fetal fibroblasts (DS-HFFs) displayed a 40%-50% reduction in PGC-1α mRNA and protein levels compared to non-trisomic counterparts (N-HFFs). DS-HFFs treated with metformin however led to a significant increase in PGC-1α at both the mRNA and protein levels compared to untreated control cells, an increase in the mRNA levels of NRF-1 and TFAM (PGC-1α target genes critical in promoting mitochondrial biogenesis), and an increase in mtDNA content [1]. Additionally, compared to N-HFFs, DS-HFFs were characterized by a decrease in basal oxygen consumption rate (OCR, ~55% inhibition), a decrease in OCR related to ATP production (~58% inhibition), and a reduction in maximal respiratory capacity (~58% inhibition). DS-HFFs treated with metformin increased basal OCR, OCR related to ATP production, ATP concentration, mitochondrial membrane potential, and maximal respiratory capacity, thus restoring mitochondrial respiration in DS-HFFs [1].

Furthermore, compared to N-HFFs, DS-HFFs exhibited extensive mitochondrial damage, as evidenced by swollen cristae (~60%-90% of DS-HFFs vs. ~15% of N-HFFs), mitochondria with intra-oedema (~40% of DS-HFFs vs. ~6% of N-HFFs), and an increase in the number of damaged mitochondria (~90% in DS-HFFs vs. ~25% in N-HFFs). Metformin-treated DS-HFFs, compared to untreated cells, displayed narrower cristae with widths that were comparable to N-HFFs cells, fewer damaged mitochondria (~40%), and fewer mitochondria with intra-oedema (~5%) [1]. Interestingly, as mitochondrial fusion also plays an important role in mitochondrial functionality, the mRNA and protein expression of OPA1 and MFN2 (genes that regulate mitochondrial fusion) were significantly down-regulated in DS-HFFs vs. N-HFFs and correlated with extensive mitochondrial fragmentation in DS-HFFs. Treatment of DS-HFFs with metformin significantly increased the mRNA and protein levels of OPA1 and MFN2 and also reduced fragmentation of the mitochondrial network, indicating that metformin induces correction of mitochondrial phenotype by also restoring mitochondrial fusion machinery [1].        

As noted above, the AMPK activator EGCG reverses cognitive defects in DS patients, promotes oxidative phosphorylation and mitochondrial biogenesis in lymphoblasts and fibroblasts from DS patients (i.e. increased levels and activity of PGC-1α, NRF-1 and TFAM), and restores oxidative phosphorylation, mitochondrial biogenesis, and improves hippocampal neural progenitor cell proliferation in a DS mouse model (Ts65Dn) in combination with the polyphenol resveratrol (i.e. increase in PGC-1α) in an AMPK-dependent manner [5,6,15]. Mitochondrial dysfunction is also a prominent feature associated with the accelerated aging disorder Hutchison-Gilford progeria syndrome (HGPS) and metformin has recently been shown to alleviate accelerated aging defects, activate AMPK, and beneficially alter gene splicing in HGPS patient cells, as I first hypothesized and published in 2014 and 2015 [12,13].

Egesipe et al. initially demonstrated, using mesenchymal stem cells (MSCs) derived from HGPS induced pluripotent stem cells (i.e. HGPS MSCs), a significant dose-dependent decrease in SRSF1 mRNA levels after metformin treatment and up to a 40% decrease in SRSF1 protein levels after treatment with 5 mmol/l of metformin (SRSF1 is a gene splicing factor that is upregulated in HGPS cells and causes faulty gene splicing of the LMNA gene leading to increased production of the toxic protein progerin) [2]. A significant decrease was also observed in both lamin A and progerin mRNA expression in HGPS MSCs treated with 5 mmol/l metformin, with progerin mRNA expression and protein levels reduced to levels lower than that of lamin A mRNA expression and protein levels, indicating that metformin-induced inhibition of SRSF1 led to an increase in the lamin A/progerin ratio and thus beneficially altered gene splicing [2].      

The results obtained using HGPS MSCs were also replicated in additional in vitro cell models, with 5 mmol/l of metformin decreasing progerin mRNA expression up to 50% in LmnaG609G/G609G mouse primary fibroblasts (HGPS mouse model) and decreasing both lamin A and progerin mRNA expression in primary HGPS fibroblasts [2]. Interestingly, 5 mmol/l of metformin also decreased progerin mRNA expression in wild-type/normal MSCs that had been incubated with a compound that induces progerin expression, indicating that metformin may also prove beneficial in reducing progerin levels in normal humans [2].

Most importantly, however, treatment of HGPS MSCs with 5 mmol/l of metformin reduced the percentage of abnormal nuclei from 60% pre-treatment to less than 40% after treatment (wild-type MSCs presented less than 20% of abnormal nuclei). The metformin-induced reduction in abnormal nuclei was comparable to the reference treatment tipifarnib (1 μmol/1), a farnesyl-transferase inhibitor [2]. Additionally, as HGPS MSCs are characterized by premature osteogenic differentiation (indicated by increased alkaline phosphatase activity compared to wild-type osteogenic progenitor cells), 5 mmol/l of metformin led to a significant rescue of alkaline phosphatase activity in HGPS osteogenic progenitor cells, comparable to levels found in tipifarnib-treated cells [2].

Park et al. also characterized the cellular phenotypes of primary dermal fibroblasts derived from HGPS patients of different ages and showed increased staining of senescence-associated beta-galactosidase (SA-β-gal, an indicator of cellular senescence) and increased levels of mitochondrial superoxide in HG8 cells (from an 8 year old patient) compared to HG3 (3 year old) and HG5 (5 year old) cells [3]. All cells expressed the toxic protein progerin, superoxide dismutase 2 (SOD2, a mitochondrial antioxidant enzyme) was highest in normal fibroblasts and lowest in HG8 cells, and cellular proliferation rate slowed at an earlier time in HGPS cells compared to normal fibroblasts [3].

Utilizing HG8 cells (which demonstrated the highest levels of senescence), the authors also elucidated the effects of metformin (2mM), rapamycin (200nM), or a combination of both drugs on the nuclear phenotype of HGPS cells. Rapamycin significantly decreased the number of nuclei with abnormal morphology and metformin treatment also led to a significant increase in the number of cells with normal nuclei compared to control-treated cells [3]. Metformin also reduced senescence in HGPS cells (i.e. reduction in SA-β-gal staining) and co-treatment with rapamycin and metformin led to an approximately 34.2 % inhibition of senescence, with similar results observed in HG3 and HG8 cells [3]. Metformin, rapamycin, or co-treatment with both compounds led to a significant reduction in the number of cells containing more than 20 γ-H2AX foci (a marker of DNA damage) in HG8, HG3, and HG5 cells, indicating that metformin increases the efficiency of DNA repair in HGPS cells [3].

Metformin also exerted antioxidant effects in HGPS cells, as evidenced by a significant decrease in ROS production and mitochondrial superoxide formation compared to control cells as well as an upregulation of SOD2 mRNA expression in aged BALB/c mice (>18 months old) [3]. Most importantly, metformin treatment at 2 and 20mM reduced progerin protein expression by approximately 20 % and 60 %, respectively, compared to mock-treated cells and increased the presence of normal nuclear phenotypes in HGPS cells [3]. Metformin treatment also significantly increased the phosphorylation and activation of AMPK in HGPS cells. Furthermore, western blot analysis indicated that rapamycin increased AMPK activation as well.

As noted previously, metformin-induced AMPK activation also exerts potent antiviral effects. With respect to dengue virus replication, Soto-Acosta et al. first showed that AMPK activation was reduced in DENV-infected Huh7 (human liver) cells (serotype 2 and 4, 2/4) at 12 and 24 hpi compared to mock-treated cells [4]. Importantly, metformin enhanced AMPK activation in DENV 2/4-infected cells compared to mock/vehicle-treated cells, reduced NS3 levels compared to vehicle-treated cells, and reduced the levels of the viral structural proteins E and prM. DENV 2/4 infection also increased the activity of HMGCR (a rate-controlling enzyme of the cholesterol biosynthetic pathway) in vehicle-treated cells compared to mock-infected cells, whereas metformin disturbed the co-localization between HMGCR and NS4A or NS3, disrupted replicative complex integrity, and decreased the levels of the viral proteins NS4A and NS3. Furthermore, metformin also led to a reduction in the levels of DENV dsRNA, indicating that metformin exerted a strong antiviral effect against DENV [4].

Indeed, 24 hour treatment of DENV2/4-infected Huh7 cells with metformin led to a reduction in the amount of infected cells, decreased the viral yield up to one logarithm, and reduced NS1 secretion up to 90%. Metformin also led to a dose-dependent reduction in viral genome copies (up to 0.7 logarithm for DENV2 and 1.5 logarithm for DENV4) compared to non-treated cells [4]. Additionally, treatment of DENV-infected cells with the AMPK inhibitor compound C (CC) increased viral infection compared to non-treated cells and CC-induced AMPK inhibition increased viral genome copies up to a half logarithm in DENV 2- and up to 0.7 logarithm in DENV 4-infected cells, indicating that metformin’s potent antiviral effects against DENV infection and replication is dependent on AMPK activation [4].    

In conclusion, data from recent evidence strongly suggests a compelling and novel assertion that the activation of AMPK via the induction of cellular stress (e.g. intracellular calcium increase, increased reactive oxygen species [ROS] production, and/or AMP/ATP ratio increase, etc.) represents a central node linking the therapeutic effects of structurally diverse compounds (e.g. metformin) with physiological and patho-physiological states, including Down syndrome, HGPS, HIV-1 latency and replication, dengue virus replication, oocyte activation, and sperm acrosome reaction induction. Indeed, AMPK activation is critical for oocyte meiotic resumption and maturation and AMPK has recently been found localized in human spermatozoa across the entire acrosome, indicating that induction of the acrosome reaction likely involves AMPK activation. Because oocyte activation is indispensable for the creation of all human life and AMPK activators including the calcium ionophores ionomycin and A23187 are extensively used to activate human oocytes, creating normal healthy children, AMPK likely connects the amelioration of normal and pathological aging defects with potential viral eradication, Down syndrome, and the creation of all human life.

https://www.linkedin.com/pulse/metformin-shown-first-time-restore-mitochondria-human-finley



References:
  1. Izzo A, Nitti M, Mollo N, et al. Metformin restores the mitochondrial network and reverses mitochondrial dysfunction in Down syndrome cells. Hum Mol Genet. 2017 Jan 13. pii: ddx016. doi: 10.1093/hmg/ddx016. [Epub ahead of print].
  2. Egesipe, Blondel, Cicero, et al. Metformin decreases progerin expression and alleviates pathological defects of Hutchinson–Gilford progeria syndrome cells. npj Aging and Mechanisms of Disease 2, Article number: 16026 (2016); http://www.nature.com/articles/npjamd201626?WT.feed_name=subjects_drug-discovery
  3. Park SK, Shin OS. Metformin Alleviates Aging Cellular Phenotypes in Hutchinson-Gilford Progeria Syndrome Dermal Fibroblasts. Exp Dermatol. 2017 Feb 13. doi: 10.1111/exd.13323. [Epub ahead of print].
  4. 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.
  5. De la Torre R, De Sola S, Pons M, et al. Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits in Down syndrome mouse models and in humans. Mol Nutr Food Res. 2014 Feb;58(2):278-88.
  6. Valenti D, de Bari L, de Rasmo D, et al. The polyphenols resveratrol and epigallocatechin-3-gallate restore the severe impairment of mitochondria in hippocampal progenitor cells from a Down syndrome mouse model. Biochim Biophys Acta. 2016 Jun;1862(6):1093-104.
  7. 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.
  8. Zainal N, Chang CP, Cheng YL, et al. Resveratrol treatment reveals a novel role for HMGB1 in regulation of the type 1 interferon response in dengue virus infection. Sci Rep. 2017 Feb 20;7:42998.
  9. Heredia A, Davis C, Amin MN, et al. Targeting host nucleotide biosynthesis with resveratrol inhibits emtricitabine-resistant HIV-1. AIDS. 2014 Jan 28;28(3):317-23.
  10. Pan XY, Zhao W, Zeng XY, et al. Heat Shock Factor 1 Mediates Latent HIV Reactivation. Sci Rep. 2016 May 18;6:26294.
  11. 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.
  12. 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.
  13. 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.
  14. 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.
  15. Valenti D, De Rasmo D, Signorile A, et al. Epigallocatechin-3-gallate prevents oxidative phosphorylation deficit and promotes mitochondrial biogenesis in human cells from subjects with Down's syndrome. Biochim Biophys Acta. 2013 Apr;1832(4):542-52.

Monday, April 10, 2017

Metformin shown for the first time to inhibit Dengue virus in human liver cells via AMPK: AMPK links Viral Eradication with Oocytes, Sperm, & Progeria


A study recently published in the journal PLoS Pathogens in April of 2017 provided startling evidence demonstrating for the first time that the commonly-prescribed anti-diabetic drug metformin exerted significant antiviral effects in dengue virus-infected human liver cells that was dependent on activation of the master metabolic regulator AMPK [59]. The authors showed that an in increase HMG-CoA reductase (HMGCR) activity, a target of AMPK, was associated with dengue virus (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 [59]. Interestingly, as discussed below, 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 [1]. 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 dengue virus replication [30]. 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 effects of AMPK activation likely crosses species boundaries [31]. Additionally, metformin has recently been shown to beneficially alter gene splicing, activate AMPK, and ameliorate accelerated aging defects in cells derived from Hutchison-Gilford progeria syndrome patients (HGPS), as I first hypothesized and published in 2014 [60-62]. AMPK activation also promotes oocyte meiotic induction and maturation (in preparation for oocyte activation) and AMPK has recently been found localized across the entire acrosome in human spermatozoa [53,55]. The induction of cellular stress (e.g. ROS, intracellular Ca2+, and/or AMP/ATP ratio increase) also promotes oocyte meiotic induction/maturation, oocyte activation, and the acrosome reaction in human sperm, processes critical for the creation of all human life [53,56,63]. As further discussed below, such interconnectedness implicates AMPK as a central mediator in the promotion of lifespan and healthspan, amelioration of pathological aging, the mounting of an effective immune response, and the creation of all human life.   

Transmission of DENV by mosquito vectors including Ae. aegypti may lead to febrile illness (known as dengue fever) in humans characterized by fluid retention, respiratory distress, and/or organ impairment [59]. There are four DENV serotypes (DENV 1-4) and several structural (capsid (C), membrane (M), and envelope (E)) and nonstructural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) are encoded by the DENV genome. Interestingly, lipids and the formation of replication complexes (i.e. membranous compartments) have been shown to play important roles in the DENV replication cycle whereas the viral protein NS4A is directly associated with DENV-associated membrane rearrangement [59]. The authors first showed that AMPK activation was reduced in DENV-infected Huh7 cells (serotype 2 and 4, 2/4) at 12 and 24 hpi compared to mock-treated cells. Importantly, metformin enhanced AMPK activation in DENV 2/4-infected cells compared to mock/vehicle-treated cells, reduced NS3 levels compared to vehicle-treated cells, and reduced the levels of the viral structural proteins E and prM. DENV 2/4 infection also increased the activity of HMGCR (a rate-controlling enzyme of the cholesterol biosynthetic pathway) in vehicle-treated cells compared to mock-infected cells, whereas metformin disturbed the co-localization between HMGCR and NS4A or NS3, disrupted replicative complex integrity, and decreased the levels of the viral proteins NS4A and NS3. Furthermore, metformin also led to a reduction in the levels of DENV dsRNA, indicating that metformin exerted a strong antiviral effect against DENV [59].

Indeed, 24 hour treatment of DENV2/4-infected Huh7 cells with metformin led to a reduction in the amount of infected cells, decreased the viral yield up to one logarithm, and reduced NS1 secretion up to 90%. Metformin also led to a dose-dependent reduction in viral genome copies (up to 0.7 logarithm for DENV2 and 1.5 logarithm for DENV4) compared to non-treated cells [59]. Additionally, treatment of DENV-infected cells with the AMPK inhibitor compound C (CC) increased viral infection compared to non-treated cells and CC-induced AMPK inhibition increased viral genome copies up to a half logarithm in DENV 2- and up to 0.7 logarithm in DENV 4-infected cells, indicating that metformin’s potent antiviral effects against DENV infection and replication is dependent on AMPK activation [59].     

Metformin’s AMPK-dependent antiviral effects against DENV in human liver cells likely represent a common mechanism that crosses species boundaries to effectuate viral eradication. As noted above, current efforts by organizations including the Bill and Melinda Gates foundation and the Google health spin-off Verily has focused on reducing transmission of dengue and Zika viruses by infecting the mosquito Ae. aegypti with the bacterium Wolbachia, leading to inhibition of viral replication. As explained below, inhibition of DENV replication in both mosquitoes and in human cells is likely orchestrated by stress-induced AMPK activation.

Although the method through which Wolbachia is transmitted to mosquito offspring has been well studied, how Wolbachia infection leads to a decrease in dengue and Zika virus replication in, and transmission by, mosquitoes is heavily debated. Interestingly, although Wolbachia does not naturally occur in Ae. aegypti, Wolbachia appears to impart beneficial immunomodulatory effects in these mosquitoes not unlike that of certain beneficial bacteria that normally colonize the human gut. Such bacteria, also known as “probiotics”, are typified by the genera Lactobacillus and Bifidobacterium and have been shown to significantly enhance natural and acquired immunity, induce reactive oxygen species (ROS) generation and the upregulation of Nrf2-dependent cytoprotective genes, produce short chain fatty acids (SCFAs) that promote T cell differentiation, and produce antimicrobial peptides that exhibit bactericidal activity [4-7]. Interestingly, the induction of cellular stress via ROS generation has been shown to activate the master metabolic regulator AMPK and AMPK activation has been shown to be critical for T cell activation and the mounting of an effective immune response in vivo to viral and bacterial infections [8-10]. Indeed, acetate, propionate, and butyrate, three SCFAs abundantly produced by probiotic bacteria, each activate AMPK, indicating that the immunomodulatory effects of probiotic bacteria are dependant on cellular stress-induced activation of AMPK [11]. Furthermore, AMPK has recently been discovered in Ae. aegypti and activation of AMPK in Ae. aegypti by compounds that induce cellular stress leads to an enhanced immune response and increased longevity in Ae. aegypti (see below), implicating the provocative assertion that inhibition of dengue and Zika virus replication via Wolbachia infection occurs as a result of cellular stress-induced activation of AMPK [12].

Wolbachia, which are found in reproductive tissues of arthropods, often engage in a mutualistic relationship with its host. Indeed, Wolbachia has been shown to induce resistance to RNA viral infections in the model organism Drosophila melanogaster [13]. D. melanogaster (also known as the fruit fly) has been studied extensively in biological research and activation of AMPK has been shown to extend lifespan as well as slow aging in D. melanogaster through autophagy induction via upregulation of Atg1 (ULK1 in mammals), reduction of insulin-like peptide levels in the brain, and an increase in 4E-BP [14,15].

Interestingly, insulin-like peptides and hyperphosphorylation of 4E-BP (i.e. inactivation) is positively associated with target of rapamycin (TOR) activation and egg development in Ae. aegypti [16]. Mammalian target of rapamycin (mTORC1/TORC1) is a protein kinase that plays a critical role in promoting mRNA translation and is the primary target of rapamycin, a macrolide drug that extends lifespan in several model organisms including normal genetically heterogeneous mice and D. melanogaster [17,18]. Rapamycin was shown to delay yolk deposition in control Ae. aegypti eggs and inhibit insulin-mediated 4E-BP (a negative regulator of translation) phosphorylation. Silencing of 4E-BP also led to a reduction in lifespan of adult female mosquitoes, mirroring results obtained via AMPK activation in D. melanogaster [15,16,19]. Strikingly, in addition to delaying egg maturation in Ae. aegypti, rapamycin has also been shown to dramatically increase the number of Wolbachia in D. melanogaster oocytes via TORC1 inhibition, whereas somatic hyperactivation of TORC1 decreases Wolbachia titer in oocytes [20]. Interestingly, ablation of insulin-producing cells in the D. melanogaster brain abolished the yeast-induced reduction of Wolbachia in oocytes, mirroring previous results showing that AMPK-induced lifespan extension in D. melanogaster is associated with a decrease in insulin-like peptides in the brain [15,20]. 

Because rapamycin, similar to AMPK activation by diverse compounds, induces autophagy, inhibits mTOR/TORC1, promotes mitochondrial biogenesis, and extends lifespan in several model organisms, it would expected that rapamycin would also induce activation of AMPK, likely via the induction of cellular stress (i.e. increase in the levels of ROS, intracellular Ca2+ increase, increase in the AMP:ADP/ATP ratio, etc). Indeed, two recent publications clearly demonstrated that rapamycin potently induced the activation of AMPK in vivo in normal elderly mice, along with an upregulation of ULK1 (mammalian orthologue of Atg1 critical for the induction of autophagy) and PGC-1a (a transcription factor essential for promoting mitochondrial biogenesis) [21,22]. As rapamycin has been shown to increase the number of Wolbachia in D. melanogaster oocytes, delay egg maturation and inhibit insulin-mediated 4E-BP phosphorylation in Ae. aegypti, and increase lifespan in D. melanogaster, the recent finding that activation of AMPK in Ae. aegypti also increases lifespan and improves the immune response (see below) further bolsters the notion that activation of AMPK via the induction of cellular stress likely promotes lifespan extension, immune system enhancement, and Wolbachia  propagation in oocytes in both D. melanogaster and Ae. aegypti. Moreover, because certain bacteria (e.g. Lactobacillus) that colonize the human gut induces beneficial immune responses by inducing cellular stress (e.g. increased ROS levels) and also produce compounds that activate AMPK, the inhibition of dengue and Zika virus replication in, and transmission by, Ae. aegypti is likely the result of a Wolbachia-induced cellular stress response, leading to the up regulation of anti-viral mechanisms that are likely modulated by AMPK.

Indeed, a recent study by Wong et al. demonstrated that ROS/oxidative stress is positively correlated with Wolbachia-mediated antiviral protection in D. melanogaster [23]. The authors of the study observed that H2O2 (hydrogen peroxide) was increased 1.25- to 2- fold in flies that harbored protective strains of Wolbachia (e.g. wMelCS, wRi, wAu) compared to Wolbachia-free controls. Interestingly, flies with a null mutation in Cu/Zn SOD (an antioxidant enzyme) exhibited elevated endogenous levels of oxidative stress that mimicked Wolbachia-induced oxidative stress. In these Wolbachia-free Cu/Zn SOD mutant flies, a 70% survival rate was observed at 4 days post-infection after Drosophila C virus infection compared to a less than 10% survival rate for non-mutant flies, indicating that elevated levels of oxidative stress induced by protective Wolbachia strains confers a survival advantage by decreasing susceptibility to viral infection via activation of signaling pathways that potentiate anti-viral immune responses [23].

As noted above, ROS/oxidative stress has been shown to activate AMPK and AMPK activation in both D. melanogaster and Ae. Aegypti leads to an increase in lifespan [8,12,15]. Interestingly, Sykiotis et al. showed that oxidants/oxidative stress promote lifespan extension in D. melanogaster males by activating the Nrf2 (CncC) pathway, a master antioxidant transcription factor that induces the expression of several antioxidant genes including NAD(P)H quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1) [24]. Curiously, Sykiotis et al. also observed that the synthetic dithiolthione oltipraz, which has been shown to activate AMPK, inhibit HIV-1 replication, and reverse accelerating aging defects in Hutchinson-Gilford progeria syndrome (see below), also induced Nrf2 signaling in flies [24-27]. AMPK has also been shown to increase the transcriptional activity of Nrf2 and increase Nrf2 nuclear retention via phosphorylation, suggesting that AMPK may represent a central node in ROS/oxidative stress-induced immune and anti-viral responses [28,29].

A recent study by Pan et al. showed that infection of Ae. Aegypti with Wolbachia (wAlbB strain) led to a ROS/oxidative stress-induced upregulation of genes associated with immunity and reduction-oxidation, thus enhancing inhibition of dengue virus replication [30]. Several gene transcripts related to the immunity and redox/stress/mitochondrion group were upregulated in Wolbachia-infected Ae. aegypti females before blood feeding and after infection with DENV serotype 2, including the antimicrobial peptides cecropin D (CECD) and defensin C (DEFC) as well as the antioxidants glutathione peroxidase, CuZnSOD, MnSOD, and glutathione peroxidase [30]. A significant increase in the levels of H2O2 and in the transcript abundance of both NADPH oxidase M (NOXM) and dual oxidase 2 (DUOX2), two enzymes that generate ROS, were also observed in Wolbachia-infected mosquitoes compared to control mosquitoes. Interestingly, the expression of several Toll pathway (a pathway that mediates the production of antioxidants and antimicrobial peptides) marker genes as well as the antimicrobial peptides CECD and DEFC were increased by the addition of H2O2 in a sugar solution given to female control/uninfected mosquitoes [30]. This effect was also mirrored in Wolbachia-infected female mosquitoes, wherein silencing of NOXM and DUOX2 deactivated the Toll pathway and suppressed the expression of CECD and DEFC, indicating that Wolbachia-induced ROS is required for activation of the Toll pathway and induction of antimicrobial peptides. Importantly, individual or double RNAi-induced knockdown of DEFC and CECD significantly increased viral titers of DENV serotype 2 in Wolbachia-infected mosquitoes compared to controls, providing further evidence that Wolbachia-induced ROS leads to a beneficial cellular response characterized by antimicrobial-mediated inhibition of viral replication [30].

Because Wolbachia-induced ROS/oxidative stress leads to beneficial anti-viral cellular responses in both D. melanogaster and Ae. aegypti and because AMPK activation, which increases Nrf2 transcriptional activity, leads to increased lifespan in D. melanogaster, it would be expected that cellular stress-induced AMPK activation in Ae. aegypti would also lead to an increase in lifespan as well as an enhanced immune response. Indeed, Nunes et al. showed that autophagy, immune system activation, and the average lifespan of Ae. Aegypti (a vector for dengue as well as chikungunya and Zika virus) is increased by feeding mosquitoes several plant-based polyphenols [31]. Interestingly, administration of resveratrol (derived from grapes), quercetin (found in many fruits and vegetables), epigallocathechin-3-gallate (EGCG, found in green tea), and genistein (found in soybeans) each increased average lifespan for male and female mosquitoes compared to controls. Insects fed resveratrol also displayed higher levels of AMPK phosphorylation/activation compared to controls and compound C, a pharmacological inhibitor of AMPK, blocked the suppression of triglyceride (TG) content induced by resveratrol [31]. Resveratrol treatment also led to immunomodulatory effects, with reduced bacterial populations for female mosquitoes compared to controls, an effect that was mimicked by AICAR (a prototypical AMPK activator) but inhibited by compound C. Interestingly, resveratrol only slightly decreased bacterial populations in vitro, indicating that the effects of resveratrol are indirect and likely associated with activation of the immune response in mosquitoes. Autophagy was also stimulated in resveratrol- and AICAR-fed mosquitoes which was abolished by silencing of AMPK [31].

Interestingly, each of the polyphenols used in that study (resveratrol, quercetin, epigallocathechin-3-gallate, and genistein) have each been shown to induce cellular stress (e.g. ROS generation), similar to Wolbachia-induced ROS generation, in a number of mammalian cells in addition to activating AMPK [32-35]. As AMPK is activated by a number of different compounds (polyphenols, bacterial metabolites, metformin etc.) and methodologies (e.g. electrical stimulation) that induce cellular stress, it is likely that Wolbachia-induced inhibition of dengue and Zika virus replication in and transmission by Ae. Aegypti is also modulated by AMPK, a master metabolic regulator that increases lifespan and improves immune and antioxidant responses in another Wolbachia-infected insect, D. melanogaster [36,37]. Although Nunes et al. did not determine if the polyphenols tested inhibited dengue virus replication in or transmission by Ae. Aegypti, recent studies have shown that in mammalian cells quercetin and analogs of resveratrol exhibit significant inhibitory activities against dengue virus type 2 [38,39]. EGCG has also recently been shown to inhibit Zika virus entry in Vero E6 cells (albeit at higher concentrations), indicating that cellular stress-induced activation of AMPK may represent a common mechanism of action that spans species boundaries to prevent the replication and/or transmission of dengue and Zika viruses [40]. Additionally, salidroside (derived from the plant Rhodiola rosea) and curcumin (derived from the plant Curcuma longa) have both been shown to exhibit anti-dengue virus activity in vitro and induce AMPK activation in vivo, providing further evidence that many structurally diverse compounds likely share a common mechanism of stress-induced AMPK activation to effectuate antiviral responses [41-44].

Remarkably, cellular stress-induced activation of AMPK (i.e. “Shock”) appears to link the antiviral effects mediated by a Wolbachia-induced increase in ROS production in A. aegypti with the amelioration of accelerated cellular aging defects in the genetic disorder Hutchinson-Gilford progeria syndrome (HGPS), the reactivation of latent HIV-1 in infected T cells (to facilitate immune system detection and virus destruction), oocyte meiotic resumption, and the acrosome reaction in sperm (i.e. “Live”).

Indeed, the proteasome inhibitor MG132 has recently been shown to inhibit progerin (the toxic protein that causes accelerated cellular aging defects) in fibroblasts derived from HGPS patients, inhibit dengue, West Nile, and Yellow fever viruses, and activate AMPK [45-47]. 1α,25-dihydroxyvitamin D3, the most potent metabolite of vitamin D, has also been shown to delay premature senescence and significantly improve accelerated aging in patient-derived HGPS cells, reduce dengue virus infection in human myelomonocyte and hepatic cell lines, and activate AMPK [48-50]. Also, as discussed above, metformin has recently been shown to beneficially alter gene splicing, activate AMPK, and ameliorate accelerated aging defects in cells derived from Hutchison-Gilford progeria syndrome patients (HGPS) and significantly inhibit dengue virus infection and replication in human liver cells [59,60-62]. Interestingly, nuclear aging defects in HGPS patient-derived cells were also reversed via activation of the antioxidant transcription factor Nrf2 by oltipraz. Oltipraz has also been shown to activate AMPK, inhibit HIV-1 replication, and induce Nrf2 signaling in D. melanogaster [24-27].

Furthermore, just as Wolbachia-induced ROS/hydrogen peroxide (H2O2) generation (i.e. “Shock”) leads to a compensatory upregulation of antimicrobial peptides and an enhanced antiviral immune response in Ae. Aegypti (i.e. “Live”), the exposure of HIV-1 latently infected monocyte or lymphocyte cell lines to H2O2 has also been shown to reactivate HIV-1 [51]. Additionally, ROS induces activation of AMPK and AMPK plays a critical role in T cell activation (and thus reactivation of latent HIV-1 that resides in T cell cells) [8,52,64].

AMPK activation has also been shown to play a critical role in the initiation of oocyte meiotic resumption and maturation (in preparation for oocyte activation) and the free radical-generating agent menadione (i.e. “Shock”) has been shown to induce AMPK-dependent meiotic resumption in cumulus-enclosed and denuded mouse oocytes (i.e. “Live”) via promotion of oxidative stress [53]. Lastly, ROS also plays a critical role in the induction of the acrosome reaction in sperm, a process that facilitates oocyte penetration through release of hydrolytic enzymes and is indispensable, along with oocyte activation, for the creation of all human life outside of a clinical setting [54]. Strikingly, AMPK has recently been found for the first time to be localized across the entire acrosome in human sperm and both H2O2/ROS and vitamin D (i.e. “Shock) have been shown to induce the acrosome reaction in human sperm (i.e. “Live”), providing compelling evidence that cellular stress-induced AMPK activation is critical for the induction of the acrosome reaction in human sperm and the creation of human life [55-57].

The antiviral effects mediated by Wolbachia-induced ROS generation paints a clear yet provocative portrait that cellular-stress mediated activation of AMPK represents a common mechanism of action that spans species boundaries, beneficially modulating cellular processes that are involved in the immune response, fertilization, and aging itself. Indeed, structurally distinct compounds including metformin, MG132, and vitamin D have been shown to exert potent antiviral effects against dengue virus and significantly improve accelerated aging defects in HGPS. More tellingly, H2O2/ROS/oxidative stress has been shown to reactivate latent HIV-1, promote oocyte meiotic resumption, and induce the acrosome reaction in human sperm. Because AMPK is critical for oocyte meiotic resumption (and hence oocyte activation) and AMPK has recently been found for the first time to be localized across the entire acrosome in human sperm, AMPK activation may indeed be critical for the creation of all human life, as originally proposed in my recent publication [58]. Perhaps beyond perplexing on first glance, Wolbachia, mosquitoes, and the creation of all human life are likely connected.

https://www.linkedin.com/pulse/metformin-shown-first-time-inhibit-dengue-virus-human-finley



References 
  1. http://www.reuters.com/article/us-health-dengue-mosquitoes-idUSKCN12Q1PE
  2. https://www.bloomberg.com/news/articles/2016-10-06/alphabet-s-verily-joins-zika-fight-with-sterile-mosquito-lab
  3. Werren JH. Biology of Wolbachia. Annu Rev Entomol. 1997;42:587-609.
  4. Castillo NA, Perdigón G, de Moreno de Leblanc A. Oral administration of a probiotic Lactobacillus modulates cytokine production and TLR expression improving the immune response against Salmonella enterica serovar Typhimurium infection in mice. BMC Microbiol. 2011 Aug 3;11:177.
  5.  Gill HS, Rutherfurd KJ, Prasad J, Gopal PK. Enhancement of natural and acquired immunity by Lactobacillus rhamnosus (HN001), Lactobacillus acidophilus (HN017) and Bifidobacterium lactis (HN019). Br J Nutr. 2000 Feb;83(2):167-76.
  6. Jones RM, Desai C, Darby TM, et al. Lactobacilli Modulate Epithelial Cytoprotection through the Nrf2 Pathway. Cell Rep. 2015 Aug 25;12(8):1217-25.
  7. Park J, Kim M, Kang SG, et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol. 2015 Jan;8(1):80-93.
  8. Mungai PT, Waypa GB, Jairaman A, et al. Hypoxia triggers AMPK activation through reactive oxygen species-mediated activation of calcium release-activated calcium channels. Mol Cell Biol. 2011 Sep;31(17):3531-45.
  9. 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;203(7):1665–70.
  10. 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.
  11. Elamin EE, Masclee AA, Dekker J, Pieters HJ, Jonkers DM. Short-chain fatty acids activate AMP-activated protein kinase and ameliorate ethanol-induced intestinal barrier dysfunction in Caco-2 cell monolayers. J Nutr. 2013 Dec;143(12):1872-81.
  12. 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.
  13. Teixeira L, Ferreira A, Ashburner M. The bacterial symbiont Wolbachia induces resistance to RNA viral infections in Drosophila melanogaster. PLoS Biol. 2008 Dec 23;6(12):e2.
  14. Yang S, Long LH, Li D, et al. β-Guanidinopropionic acid extends the lifespan of Drosophila melanogaster via an AMP-activated protein kinase-dependent increase in autophagy. Aging Cell. 2015 Dec;14(6):1024-33.
  15. Ulgherait M, Rana A, Rera M, Graniel J, Walker DW. AMPK modulates tissue and organismal aging in a non-cell-autonomous manner. Cell Rep. 2014 Sep 25;8(6):1767-80.
  16. Gulia-Nuss M, Robertson AE, Brown MR, Strand MR. Insulin-like peptides and the target of rapamycin pathway coordinately regulate blood digestion and egg maturation in the mosquito Aedes aegypti. PLoS One. 2011;6(5):e20401.
  17. Harrison DE, Strong R, Sharp ZD, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009 Jul 16;460(7253):392-5.
  18. Sun X, Wheeler CT, Yolitz J, et al. A mitochondrial ATP synthase subunit interacts with TOR signaling to modulate protein homeostasis and lifespan in Drosophila. Cell Rep. 2014 Sep 25;8(6):1781-92.
  19. Roy SG, Raikhel AS. Nutritional and hormonal regulation of the TOR effector 4E-binding protein (4E-BP) in the mosquito Aedes aegypti. FASEB J. 2012 Mar;26(3):1334-42.
  20. Serbus LR, White PM, Silva JP, et al. The impact of host diet on Wolbachia titer in Drosophila. PLoS Pathog. 2015 Mar 31;11(3):e1004777.
  21. 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.
  22. Lesniewski LA, Seals DR, Walker AE, et al. Dietary rapamycin supplementation reverses age-related vascular dysfunction and oxidative stress, while modulating nutrient-sensing, cell cycle, and senescence pathways. Aging Cell. 2016 Sep 22. doi: 10.1111/acel.12524. [Epub ahead of print].
  23. Wong ZS, Brownlie JC, Johnson KN. Oxidative stress correlates with Wolbachia-mediated antiviral protection in Wolbachia-Drosophila associations. Appl Environ Microbiol. 2015 May 1;81(9):3001-5.
  24. Sykiotis GP, Bohmann D. Keap1/Nrf2 signaling regulates oxidative stress tolerance and lifespan in Drosophila. Dev Cell. 2008 Jan;14(1):76-85.
  25. Prochaska HJ, Fernandes CL, Pantoja RM, Chavan SJ. Inhibition of human immunodeficiency virus type 1 long terminal repeat-driven transcription by an in vivo metabolite of oltipraz: implications for antiretroviral therapy. Biochem Biophys Res Commun. 1996 Apr 25;221(3):548-53.
  26. Kubben N, Zhang W2, Wang L, et al. Repression of the Antioxidant NRF2 Pathway in Premature Aging. Cell. 2016 Jun 2;165(6):1361-74.
  27. Kim TH, Eom JS, Lee CG, Yang YM, Lee YS, Kim SG. An active metabolite of oltipraz (M2) increases mitochondrial fuel oxidation and inhibits lipogenesis in the liver by dually activating AMPK. Br J Pharmacol. 2013 Apr;168(7):1647-61. doi: 10.1111/bph.12057.
  28. Mo C, Wang L, Zhang J, et al. The crosstalk between Nrf2 and AMPK signal pathways is important for the anti-inflammatory effect of berberine in LPS-stimulated macrophages and endotoxin-shocked mice. Antioxid Redox Signal. 2014 Feb 1;20(4):574-88.
  29. Joo MS, Kim WD, Lee KY, Kim JH, Koo JH, Kim SG. AMPK Facilitates Nuclear Accumulation of Nrf2 by Phosphorylating at Serine 550. Mol Cell Biol. 2016 Jun 29;36(14):1931-42.
  30. 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.
  31. 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.
  32. Li GX, Chen YK, Hou Z, et al. Pro-oxidative activities and dose-response relationship of (-)-epigallocatechin-3-gallate in the inhibition of lung cancer cell growth: a comparative study in vivo and in vitro. Carcinogenesis. 2010 May;31(5):902-10.
  33. Chang YF, Chi CW, Wang JJ. Reactive oxygen species production is involved in quercetin-induced apoptosis in human hepatoma cells. Nutr Cancer. 2006;55(2):201-9.
  34. Miki H, Uehara N, Kimura A, et al. Resveratrol induces apoptosis via ROS-triggered autophagy in human colon cancer cells. Int J Oncol. 2012 Apr;40(4):1020-8.
  35. Ullah MF, Ahmad A, Zubair H, et al. Soy isoflavone genistein induces cell death in breast cancer cells through mobilization of endogenous copper ions and generation of reactive oxygen species. Mol Nutr Food Res. 2011 Apr;55(4):553-9.
  36. Matsuyama S, Moriuchi M1, Suico MA, et al. Mild electrical stimulation increases stress resistance and suppresses fat accumulation via activation of LKB1-AMPK signaling pathway in C. elegans. PLoS One. 2014 Dec 9;9(12):e114690.
  37. Dutra HL, Rocha MN, Dias FB, Mansur SB, Caragata EP, Moreira LA. Wolbachia Blocks Currently Circulating Zika Virus Isolates in Brazilian Aedes aegypti Mosquitoes. Cell Host Microbe. 2016 Jun 8;19(6):771-4.
  38. Zandi K, Teoh BT, Sam SS, Wong PF, Mustafa MR, Abubakar S. Antiviral activity of four types of bioflavonoid against dengue virus type-2. Virol J. 2011 Dec 28;8:560.
  39. Han YS, Penthala NR2, Oliveira M, et al. Identification of resveratrol analogs as potent anti-dengue agents using a cell-based assay. J Med Virol. 2016 Aug 10. doi: 10.1002/jmv.24660. [Epub ahead of print].
  40. Carneiro BM, Batista MN, Braga AC, Nogueira ML, Rahal P. The green tea molecule EGCG inhibits Zika virus entry. Virology. 2016 Sep;496:215-8.
  41. Padilla-S L, Rodríguez A, Gonzales MM, Gallego-G JC, Castaño-O JC. Inhibitory effects of curcumin on dengue virus type 2-infected cells in vitro. Arch Virol. 2014 Mar;159(3):573-9.
  42. Sharma N, Mishra KP, Ganju L. Salidroside exhibits anti-dengue virus activity by upregulating host innate immune factors. Arch Virol. 2016 Dec;161(12):3331-3344.
  43. Chang X, Zhang K, Zhou R, et al. Cardioprotective effects of salidroside on myocardial ischemia-reperfusion injury in coronary artery occlusion-induced rats and Langendorff-perfused rat hearts. Int J Cardiol. 2016 Jul 15;215:532-44.
  44. Ejaz A, Wu D, Kwan P, Meydani M. Curcumin inhibits adipogenesis in 3T3-L1 adipocytes and angiogenesis and obesity in C57/BL mice.
  45. Pellegrini C, Columbaro M, Capanni C, et al. All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget. 2015 Oct 6;6(30):29914-28.
  46. Fernandez-Garcia MD, Meertens L, Bonazzi M, Cossart P, Arenzana-Seisdedos F, Amara A. Appraising the roles of CBLL1 and the ubiquitin/proteasome system for flavivirus entry and replication. J Virol. 2011 Mar;85(6):2980-9.
  47. Jiang S, Park DW, Gao Y, et al. Participation of proteasome-ubiquitin protein degradation in autophagy and the activation of AMP-activated protein kinase. Cell Signal. 2015 Jun;27(6):1186-97.
  48. Kreienkamp R, Croke M, Neumann MA, et al. Vitamin D receptor signaling improves Hutchinson-Gilford progeria syndrome cellular phenotypes. Oncotarget. 2016 Apr 27. doi: 10.18632/oncotarget.9065.
  49. Puerta-Guardo H, Medina F, De la Cruz Hernández SI, Rosales VH, Ludert JE, del Angel RM. The 1α,25-dihydroxy-vitamin D3 reduces dengue virus infection in human myelomonocyte (U937) and hepatic (Huh-7) cell lines and cytokine production in the infected monocytes. Antiviral Res. 2012 Apr;94(1):57-61.
  50. Swami S, Krishnan AV, Williams J, et al.. Vitamin D mitigates the adverse effects of obesity on breast cancer in mice. Endocr Relat Cancer. 2016 Apr;23(4):251-64.
  51. Piette J, Legrand-Poels S. HIV-1 reactivation after an oxidative stress mediated by different reactive oxygen species. Chem Biol Interact. 1994 Jun;91(2-3):79-89.
  52. 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.
  53. LaRosa C, Downs SM. Stress stimulates AMP-activated protein kinase and meiotic resumption in mouse oocytes. Biol Reprod. 2006 Mar;74(3):585-92.
  54. Patrat C, Serres C, Jouannet P. The acrosome reaction in human spermatozoa. Biol Cell. 2000 Jul;92(3-4):255-66.
  55. 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].
  56. 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.
  57. Blomberg Jensen M, Bjerrum PJ, Jessen TE, et al. Vitamin D is positively associated with sperm motility and increases intracellular calcium in human spermatozoa. Hum Reprod. 2011 Jun;26(6):1307-17.
  58. 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.
  59. 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.
  60. Park SK, Shin OS. Metformin Alleviates Aging Cellular Phenotypes in Hutchinson-Gilford Progeria Syndrome Dermal Fibroblasts. Exp Dermatol. 2017 Feb 13. doi: 10.1111/exd.13323. [Epub ahead of print].
  61. Egesipe, Blondel, Cicero, et al. Metformin decreases progerin expression and alleviates pathological defects of Hutchinson–Gilford progeria syndrome cells. npj Aging and Mechanisms of Disease 2, Article number: 16026 (2016); http://www.nature.com/articles/npjamd201626?WT.feed_name=subjects_drug-discovery
  62. 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.
  63. 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.
  64. 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.