DENV up-regulates the HMG-CoA reductase activity through the impairment of AMPK phosphorylation: A potential antiviral target

Rubén Soto-Acosta, Patricia Bautista-Carbajal, Margot Cervantes-Salazar, Antonio H Angel-Ambrocio, Rosa M Del Angel, Rubén Soto-Acosta, Patricia Bautista-Carbajal, Margot Cervantes-Salazar, Antonio H Angel-Ambrocio, Rosa M Del Angel

Abstract

Dengue is the most common mosquito-borne viral disease in humans. Changes of lipid-related metabolites in endoplasmic reticulum of dengue virus (DENV) infected cells have been associated with replicative complexes formation. Previously, we reported that DENV infection inhibits HMGCR phosphorylation generating a cholesterol-enriched cellular environment in order to favor viral replication. In this work, using enzymatic assays, ELISA, and WB we found a significant higher activity of HMGCR in DENV infected cells, associated with the inactivation of AMPK. AMPK activation by metformin declined the HMGCR activity suggesting that AMPK inactivation mediates the enhanced activity of HMGCR. A reduction on AMPK phosphorylation activity was observed in DENV infected cells at 12 and 24 hpi. HMGCR and cholesterol co-localized with viral proteins NS3, NS4A and E, suggesting a role for HMGCR and AMPK activity in the formation of DENV replicative complexes. Furthermore, metformin and lovastatin (HMGCR inhibitor) altered this co-localization as well as replicative complexes formation supporting that active HMGCR is required for replicative complexes formation. In agreement, metformin prompted a significant dose-dependent antiviral effect in DENV infected cells, while compound C (AMPK inhibitor) augmented the viral genome copies and the percentage of infected cells. The PP2A activity, the main modulating phosphatase of HMGCR, was not affected by DENV infection. These data demonstrate that the elevated activity of HMGCR observed in DENV infected cells is mediated through AMPK inhibition and not by increase in PP2A activity. Interestingly, the inhibition of this phosphatase showed an antiviral effect in an HMGCR-independent manner. These results suggest that DENV infection increases HMGCR activity through AMPK inactivation leading to higher cholesterol levels in endoplasmic reticulum necessary for replicative complexes formation. This work provides new information about the mechanisms involved in host lipid metabolism during DENV replicative cycle and identifies new potential antiviral targets for DENV replication.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. DENV infection down-regulates AMPK activity.
Fig 1. DENV infection down-regulates AMPK activity.
In A, The AMPK activity, depicted as phosphorylation levels at Thr-172, was evaluated in Huh7 cells infected with DENV 2/4 (MOI 3) at 1, 12, and 24 hpi by ELISA, and NS3 viral protein levels (A, lower panel) were determined as infection test. AMPK activity was expressed as U/mL. *p<0.05 compared to mock infected cells (0 hpi). Data are means ± standard error (S.E) of n = 3 independent experiments realized by duplicate. (B) The levels of AMPK phosphorylated, AMPK total, and NS3 viral protein were analyzed by western blot in whole cell lysates obtained from Huh7 cells infected with DENV2 (MOI 0.1, 1 and 3) for 24 h. Graph represents the relative quantification of pAMPK respect to AMPK total protein. The pAMPK and total AMPK densitometry values were normalized with β-actin and pAMPK/AMPK ratios were calculated, Ratios are represented with respect to the indicated control. *p<0.05 compared to mock infected cells. Data are means ± standard error (S.E) of n = 4 independent experiments. (C) The AMPK activity and NS3 viral protein levels (C, lower panel) were determined in Mock or DENV 2/4 infected Huh7 cells treated with DMSO 0.5% (vehicle, VEH), 10 mM Metformin (MET, AMPK activator) or 10 μM Compound C (CC, AMPK inhibitor) for 24 h. *p<0.05 compared to mock VEH-treated cells, abp<0.05 compared to mock MET-treated cells. Data are means ± standard error (S.E) of n = 3 independent experiments realized by duplicate. (D) The levels of AMPK phosphorylated, AMPK total, and prM viral protein were analyzed by western blot in whole cell lysates obtained from Mock or DENV2 Huh7 infected cells (MOI 1 and 3) in the presence or absence of 10 mM metformin (MET) for 24h. Graph represents pAMPK/AMPK ratios normalized with respect to Mock infected cells with no MET treatment. pAMPK/AMPK ratios were obtained adjusting each protein with β-actin.
Fig 2. HMGCR activity is increased during…
Fig 2. HMGCR activity is increased during DENV infection through down-regulation of AMPK.
In A, The enzymatic activity of HMGCR was evaluated in Huh7 cells infected with Mock or DENV 2/4 (serotype 2 or 4, MOI 3) at 24 hours post-infection (hpi). HMGCR activity was expressed as U/mg protein. *p<0.05 compared to mock infected cells. From same cell lysates, levels of NS3 viral protein (A, lower panel) were determined by WB as infection test. (B) DENV4 infected Huh7 cells treated with DMSO 0.5% (vehicle, VEH), 10 mM metformin (MET, AMPK activator), 50 μM lovastatin or 10 nM Okadaic acid (O.A, PP2A inhibitor) for 24 hpi were assayed for HMGCR activity and NS3 viral protein levels (B, lower panel). *p<0.05 compared to mock infected cells, ap<0.05 compared to DENV4 VEH-treated cells. Relative quantification of NS3 levels (numbers in italics) was normalized to β-actin and represented with respect to the indicated control. (C) The levels of NS3, E (envelope), and prM viral proteins were analyzed by western blot in whole cell lysates obtained from Mock or DENV2 Huh7 infected cells (MOI 3) treated with DMSO 0.5% (vehicle, VEH), 10 mM metformin (MET, AMPK activator), 10 nM Okadaic acid (O.A, PP2A inhibitor), and 50 μM Lovastatin (LOV, HMGCR inhibitor) for 24 h. Graph represents the relative quantification each protein normalized to β-actin and represented with respect to the indicated control (VEH). All data are means ± standard error (S.E) of n = 3 independent experiments.
Fig 3. Intracellular distribution of HMGCR and…
Fig 3. Intracellular distribution of HMGCR and NS4A viral protein during DENV infection.
(A)The distribution of the NS4A (green), a viral protein present at DENV-replication complexes, and the HMGCR (red), a cellular ER-resident protein, was evaluated by confocal microscopy in Huh7 cells infected (MOI 3) with DENV2 and DENV4 at 24 hpi. Nuclei were stained with Hoechst (blue). Scale bar 10 μm. White dashed boxes are depicting the zoom area. (B) Histograms represent the fluorescence intensity for NS4A and HMGCR in determined area (white continuous line) demonstrating the correlation between two signals. In all infected cells, HMGCR colocalized with NS4A, however, the optical cut does not allow us to clearly observed this colocalization. (C)The table indicates HMGCR/NS4A colocalization values for region of interest (ROI, white dashed boxes) and colocalization per infected cell expressed as mean ± S.E. of 52 DENV2 infected cells and 47 DENV4 infected cells from three independent images.
Fig 4. Activated HMGCR is required for…
Fig 4. Activated HMGCR is required for the formation of DENV-replication complexes and the maintenance of its architecture.
The distribution of HMGCR and components of viral replication complexes (NS4A and E viral proteins) was evaluated by confocal microscopy in Huh7 cells infected with DENV2 (MOI 3) and treated with DMSO 0.5% (vehicle), 10 mM Metformin or 50 μM lovastatin (HMGCR inhibitor) for 24h. The integrity of replication complexes is depicted as the co-localization between NS4A and E proteins. In A is indicated the distribution of HMGCR (red), NS4A (light blue), and E protein (green) as well as the colocalization per infected cell of NS4A/HMGCR (B) and NS4A/E (C) represented by mean ± S.E of the colocalization of 60 infected cells per condition. D and E represent the mean fluorescence intensity of NS4A protein (D) and HMGCR (E) analyzed by flow cytometry. Graphs represent the mean fluorescence intensity ± S.E of three independent experiments, the histograms indicate the fluorescence intensity of a representative experiment.
Fig 5. DENV infection stimulates the intracellular…
Fig 5. DENV infection stimulates the intracellular cholesterol accumulation at replicative complexes through the activation of HMGCR.
The distribution of intracellular cholesterol levels stained with filipin III complex (blue), and its co-localization with the viral protein NS4A (green) were evaluated by confocal microscopy in Huh7 cells non-infected, infected with DENV4 and treated with DMSO 0.5% (vehicle), 10 mM metformin or 50 μM lovastatin (HMGCR inhibitor) for 24 h. DENV4 infected cells are marked with (+), and non-infected cells marked with (-). Nuclei were stained with propidium iodide (red). Numbers inserted in images indicate the co-localization index between cholesterol and NS4A for that specific infected cell. Graph represents NS4A and cholesterol colocalization values as mean ± S.E of 50 infected cells analyzed from 3 independent experiments. Scale bar 10 μm. Images correspond to one representative experiment.
Fig 6. Metformin induces an antiviral effect…
Fig 6. Metformin induces an antiviral effect in DENV infected cells.
In A and B, The antiviral effect of metformin-treatment (0, 1 and 10 mM) against DENV infection was evaluated in supernatants from Huh7 cells infected (MOI 3) with DENV2 (A) and DENV4 (B) at 24 hpi through determination of viral yield by foci assay, and NS1 secretion by ELISA. Viral yield is expressed as Foci Forming Units (FFU) / mL. NS1 secretion was normalized respect to infected non-treated cells and expressed as fold change vs 0 mM. (C) The percentage of infected cells after 10 mM metformin-treatment was determined by flow cytometry using a mouse anti-E monoclonal antibody-4G2 to detect the E viral protein in mock or DENV 2/4 infected cells. Upper histograms show the fluorescence of infected cells at 24h (gray filled histograms) or 48h (dark filled histograms) respect to mock infected cells (non-filled histograms). Lower histograms display the fluorescence of DENV infected cells treated with metformin (gray filled histograms) respect to vehicle-treated infected cells (dark filled histograms). (D) The Mean Fluorescence intensity (MFI) is presented on Graphs. (E) DENV 2/4 infected cells treated with 10mM metformin (MET) were visualized at 24 hpi by confocal microscopy using a mouse anti-E monoclonal antibody-4G2 (green). Nuclei were stained with Hoechst (blue). Scale bar 50 μm. Images correspond to one experiment representative of n = 3. (F) The number of viral genome copies of DENV 2/4 infected cells treated with metformin (0, 1, 10 mM) for 24h was examined by qRT-PCR, and expressed as Log of No. Copies. DMSO 0.5% was used as vehicle for all cases (0 mM). Data are means ± S.E of n = 3 independent experiments realized by duplicated. * p<0.05 compared to non-treated cells.
Fig 7. PP2A activity is not altered…
Fig 7. PP2A activity is not altered by DENV, but its inhibition by Okadaic acid has an antiviral effect.
The PP2A activity was analyzed in Huh7 cells infected (MOI 3) with DENV 2/4 at 1, 12 and 24 hpi (A), and in Mock or DENV 2/4 infected cells treated with DMSO 0.05% (vehicle) or 10 nM Okadaic acid (O. A) for 24h (B). Activity is expressed as picomoles of phosphate (phosphates pmoles). From the same cell lysates, the levels of NS3 viral protein (lower panels) were determined by WB as infection test. * p<0.05 compared to mock vehicle-treated cells. The antiviral effect of O. A (0, 1 and 10 nM) against DENV infection was evaluated in supernatants from Huh7 cells infected (MOI 3) with DENV2 (C) and DENV4 (D) by viral yield and NS1 secretion at 24 hpi. Viral yield is expressed as Foci Forming Units (FFU) / mL. NS1 secretion was normalized respect to infected non-treated cells and expressed as fold change vs 0 mM. (E) The number of viral genome copies of DENV 2/4 infected cells treated with O. A (0, 5, 10 nM) for 24h was examined by qRT-PCR, and expressed as Log of No. Copies. * p<0.05 compared to non-treated cells. DMSO 0.05% was used as vehicle for all cases (0 nM). Data are means ± S.E of n = 3 independent experiments realized by duplicated.

References

    1. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, et al. (2013) The global distribution and burden of dengue. Nature 496: 504–507. 10.1038/nature12060
    1. WHO (2016) Dengue vaccine research. Immunization, Vaccines and Biologicals .
    1. Screaton G, Mongkolsapaya J, Yacoub S, Roberts C (2015) New insights into the immunopathology and control of dengue virus infection. Nat Rev Immunol 15: 745–759. 10.1038/nri3916
    1. Kuhn RJ, Zhang W, Rossmann MG, Pletnev SV, Corver J, et al. (2002) Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell 108: 717–725.
    1. Yu IM, Zhang W, Holdaway HA, Li L, Kostyuchenko VA, et al. (2008) Structure of the immature dengue virus at low pH primes proteolytic maturation. Science 319: 1834–1837. 10.1126/science.1153264
    1. Chen Y, Maguire T, Hileman RE, Fromm JR, Esko JD, et al. (1997) Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nat Med 3: 866–871.
    1. Jindadamrongwech S, Thepparit C, Smith DR (2004) Identification of GRP 78 (BiP) as a liver cell expressed receptor element for dengue virus serotype 2. Arch Virol 149: 915–927. 10.1007/s00705-003-0263-x
    1. Lozach PY, Burleigh L, Staropoli I, Navarro-Sanchez E, Harriague J, et al. (2005) Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals. J Biol Chem 280: 23698–23708. 10.1074/jbc.M504337200
    1. Chen ST, Lin YL, Huang MT, Wu MF, Cheng SC, et al. (2008) CLEC5A is critical for dengue-virus-induced lethal disease. Nature 453: 672–676. 10.1038/nature07013
    1. Reyes-Del Valle J, Chavez-Salinas S, Medina F, Del Angel RM (2005) Heat shock protein 90 and heat shock protein 70 are components of dengue virus receptor complex in human cells. J Virol 79: 4557–4567. 10.1128/JVI.79.8.4557-4567.2005
    1. Thepparit C, Smith DR (2004) Serotype-specific entry of dengue virus into liver cells: identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor as a dengue virus serotype 1 receptor. J Virol 78: 12647–12656. 10.1128/JVI.78.22.12647-12656.2004
    1. Wan SW, Lin CF, Lu YT, Lei HY, Anderson R, et al. (2012) Endothelial cell surface expression of protein disulfide isomerase activates beta1 and beta3 integrins and facilitates dengue virus infection. J Cell Biochem 113: 1681–1691. 10.1002/jcb.24037
    1. Cambi A, de Lange F, van Maarseveen NM, Nijhuis M, Joosten B, et al. (2004) Microdomains of the C-type lectin DC-SIGN are portals for virus entry into dendritic cells. J Cell Biol 164: 145–155. 10.1083/jcb.200306112
    1. Puerta-Guardo H, Mosso C, Medina F, Liprandi F, Ludert JE, et al. (2010) Antibody-dependent enhancement of dengue virus infection in U937 cells requires cholesterol-rich membrane microdomains. J Gen Virol 91: 394–403. 10.1099/vir.0.015420-0
    1. Garcia-Garcia E, Brown EJ, Rosales C (2007) Transmembrane mutations to FcgammaRIIA alter its association with lipid rafts: implications for receptor signaling. J Immunol 178: 3048–3058.
    1. Zaitseva E, Yang ST, Melikov K, Pourmal S, Chernomordik LV (2010) Dengue virus ensures its fusion in late endosomes using compartment-specific lipids. PLoS Pathog 6: e1001131 10.1371/journal.ppat.1001131
    1. Gushchin VA, Solovyev AG, Erokhina TN, Morozov SY, Agranovsky AA (2013) Beet yellows virus replicase and replicative compartments: parallels with other RNA viruses. Front Microbiol 4: 38 10.3389/fmicb.2013.00038
    1. Lee CM, Xie X, Zou J, Li SH, Lee MY, et al. (2015) Determinants of Dengue Virus NS4A Protein Oligomerization. J Virol 89: 6171–6183. 10.1128/JVI.00546-15
    1. Teo CS, Chu JJ (2014) Cellular vimentin regulates construction of dengue virus replication complexes through interaction with NS4A protein. J Virol 88: 1897–1913. 10.1128/JVI.01249-13
    1. Hung YF, Schwarten M, Hoffmann S, Willbold D, Sklan EH, et al. (2015) Amino Terminal Region of Dengue Virus NS4A Cytosolic Domain Binds to Highly Curved Liposomes. Viruses 7: 4119–4130. 10.3390/v7072812
    1. Mukhopadhyay S, Kuhn RJ, Rossmann MG (2005) A structural perspective of the flavivirus life cycle. Nat Rev Microbiol 3: 13–22. 10.1038/nrmicro1067
    1. Perera R, Riley C, Isaac G, Hopf-Jannasch AS, Moore RJ, et al. (2012) Dengue virus infection perturbs lipid homeostasis in infected mosquito cells. PLoS Pathog 8: e1002584 10.1371/journal.ppat.1002584
    1. Apte-Sengupta S, Sirohi D, Kuhn RJ (2014) Coupling of replication and assembly in flaviviruses. Curr Opin Virol 9: 134–142. 10.1016/j.coviro.2014.09.020
    1. Stiasny K, Heinz FX (2006) Flavivirus membrane fusion. J Gen Virol 87: 2755–2766. 10.1099/vir.0.82210-0
    1. Perera R, Khaliq M, Kuhn RJ (2008) Closing the door on flaviviruses: entry as a target for antiviral drug design. Antiviral Res 80: 11–22. 10.1016/j.antiviral.2008.05.004
    1. Lee CJ, Lin HR, Liao CL, Lin YL (2008) Cholesterol effectively blocks entry of flavivirus. J Virol 82: 6470–6480. 10.1128/JVI.00117-08
    1. Martinez-Gutierrez M, Castellanos JE, Gallego-Gomez JC (2011) Statins reduce dengue virus production via decreased virion assembly. Intervirology 54: 202–216. 10.1159/000321892
    1. Rothwell C, Lebreton A, Young Ng C, Lim JY, Liu W, et al. (2009) Cholesterol biosynthesis modulation regulates dengue viral replication. Virology 389: 8–19. 10.1016/j.virol.2009.03.025
    1. Tang WC, Lin RJ, Liao CL, Lin YL (2014) Rab18 facilitates dengue virus infection by targeting fatty acid synthase to sites of viral replication. J Virol 88: 6793–6804. 10.1128/JVI.00045-14
    1. Heaton NS, Perera R, Berger KL, Khadka S, Lacount DJ, et al. (2010) Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral replication and increases cellular fatty acid synthesis. Proc Natl Acad Sci U S A 107: 17345–17350. 10.1073/pnas.1010811107
    1. Samsa MM, Mondotte JA, Iglesias NG, Assuncao-Miranda I, Barbosa-Lima G, et al. (2009) Dengue virus capsid protein usurps lipid droplets for viral particle formation. PLoS Pathog 5: e1000632 10.1371/journal.ppat.1000632
    1. Soto-Acosta R, Bautista-Carbajal P, Syed GH, Siddiqui A, Del Angel RM (2014) Nordihydroguaiaretic acid (NDGA) inhibits replication and viral morphogenesis of dengue virus. Antiviral Res.
    1. Soto-Acosta R, Mosso C, Cervantes-Salazar M, Puerta-Guardo H, Medina F, et al. (2013) The increase in cholesterol levels at early stages after dengue virus infection correlates with an augment in LDL particle uptake and HMG-CoA reductase activity. Virology 442: 132–147. 10.1016/j.virol.2013.04.003
    1. Ching YP, Kobayashi T, Tamura S, Hardie DG (1997) Specificity of different isoforms of protein phosphatase-2A and protein phosphatase-2C studied using site-directed mutagenesis of HMG-CoA reductase. FEBS Lett 411: 265–268.
    1. Friesen JA, Rodwell VW (2004) The 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductases. Genome Biol 5: 248 10.1186/gb-2004-5-11-248
    1. Deng X, Dong Q, Bridges D, Raghow R, Park EA, et al. (2015) Docosahexaenoic acid inhibits proteolytic processing of sterol regulatory element-binding protein-1c (SREBP-1c) via activation of AMP-activated kinase. Biochim Biophys Acta 1851: 1521–1529. 10.1016/j.bbalip.2015.08.007
    1. Chukkapalli V, Heaton NS, Randall G (2012) Lipids at the interface of virus-host interactions. Curr Opin Microbiol 15: 512–518. 10.1016/j.mib.2012.05.013
    1. Omkumar RV, Darnay BG, Rodwell VW (1994) Modulation of Syrian hamster 3-hydroxy-3-methylglutaryl-CoA reductase activity by phosphorylation. Role of serine 871. J Biol Chem 269: 6810–6814.
    1. Gaussin V, Skarlas P, Ching YP, Hardie DG, Hue L (1997) Distinct type-2A protein phosphatases activate HMGCoA reductase and acetyl-CoA carboxylase in liver. FEBS Lett 413: 115–118.
    1. Martinez-Gutierrez M, Correa-Londono LA, Castellanos JE, Gallego-Gomez JC, Osorio JE (2014) Lovastatin delays infection and increases survival rates in AG129 mice infected with dengue virus serotype 2. PLoS One 9: e87412 10.1371/journal.pone.0087412
    1. Miller S, Kastner S, Krijnse-Locker J, Buhler S, Bartenschlager R (2007) The non-structural protein 4A of dengue virus is an integral membrane protein inducing membrane alterations in a 2K-regulated manner. J Biol Chem 282: 8873–8882. 10.1074/jbc.M609919200
    1. Lin MH, Hsu HJ, Bartenschlager R, Fischer WB (2014) Membrane undulation induced by NS4A of Dengue virus: a molecular dynamics simulation study. J Biomol Struct Dyn 32: 1552–1562. 10.1080/07391102.2013.826599
    1. Madsen A, Bozickovic O, Bjune JI, Mellgren G, Sagen JV (2015) Metformin inhibits hepatocellular glucose, lipid and cholesterol biosynthetic pathways by transcriptionally suppressing steroid receptor coactivator 2 (SRC-2). Sci Rep 5: 16430 10.1038/srep16430
    1. Mendoza-Oliva A, Ferrera P, Fragoso-Medina J, Arias C (2015) Lovastatin Differentially Affects Neuronal Cholesterol and Amyloid-beta Production in vivo and in vitro. CNS Neurosci Ther 21: 631–641. 10.1111/cns.12420
    1. Stormo C, Kringen MK, Grimholt RM, Berg JP, Piehler AP (2012) A novel 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) splice variant with an alternative exon 1 potentially encoding an extended N-terminus. BMC Mol Biol 13: 29 10.1186/1471-2199-13-29
    1. Zou J, Xie X, Wang QY, Dong H, Lee MY, et al. (2015) Characterization of dengue virus NS4A and NS4B protein interaction. J Virol 89: 3455–3470. 10.1128/JVI.03453-14
    1. Zhan YY, Chen Y, Zhang Q, Zhuang JJ, Tian M, et al. (2012) The orphan nuclear receptor Nur77 regulates LKB1 localization and activates AMPK. Nat Chem Biol 8: 897–904. 10.1038/nchembio.1069
    1. Goransson O, McBride A, Hawley SA, Ross FA, Shpiro N, et al. (2007) Mechanism of action of A-769662, a valuable tool for activation of AMP-activated protein kinase. J Biol Chem 282: 32549–32560. 10.1074/jbc.M706536200
    1. Villareal VA, Rodgers MA, Costello DA, Yang PL (2015) Targeting host lipid synthesis and metabolism to inhibit dengue and hepatitis C viruses. Antiviral Res 124: 110–121. 10.1016/j.antiviral.2015.10.013
    1. Acosta EG, Kumar A, Bartenschlager R (2014) Revisiting dengue virus-host cell interaction: new insights into molecular and cellular virology. Adv Virus Res 88: 1–109. 10.1016/B978-0-12-800098-4.00001-5
    1. Jeon SM (2016) Regulation and function of AMPK in physiology and diseases. Exp Mol Med 48: e245 10.1038/emm.2016.81
    1. Mankouri J, Tedbury PR, Gretton S, Hughes ME, Griffin SD, et al. (2010) Enhanced hepatitis C virus genome replication and lipid accumulation mediated by inhibition of AMP-activated protein kinase. Proc Natl Acad Sci U S A 107: 11549–11554. 10.1073/pnas.0912426107
    1. Huang H, Kang R, Wang J, Luo G, Yang W, et al. (2013) Hepatitis C virus inhibits AKT-tuberous sclerosis complex (TSC), the mechanistic target of rapamycin (MTOR) pathway, through endoplasmic reticulum stress to induce autophagy. Autophagy 9: 175–195. 10.4161/auto.22791
    1. Yu JW, Sun LJ, Liu W, Zhao YH, Kang P, et al. (2013) Hepatitis C virus core protein induces hepatic metabolism disorders through down-regulation of the SIRT1-AMPK signaling pathway. Int J Infect Dis 17: e539–545. 10.1016/j.ijid.2013.01.027
    1. Lo AK, Lo KW, Ko CW, Young LS, Dawson CW (2013) Inhibition of the LKB1-AMPK pathway by the Epstein-Barr virus-encoded LMP1 promotes proliferation and transformation of human nasopharyngeal epithelial cells. J Pathol 230: 336–346. 10.1002/path.4201
    1. Lu J, Wu DM, Zheng YL, Hu B, Zhang ZF, et al. (2010) Quercetin activates AMP-activated protein kinase by reducing PP2C expression protecting old mouse brain against high cholesterol-induced neurotoxicity. J Pathol 222: 199–212. 10.1002/path.2754
    1. Valentine RJ, Coughlan KA, Ruderman NB, Saha AK (2014) Insulin inhibits AMPK activity and phosphorylates AMPK Ser(4)(8)(5)/(4)(9)(1) through Akt in hepatocytes, myotubes and incubated rat skeletal muscle. Arch Biochem Biophys 562: 62–69. 10.1016/j.abb.2014.08.013
    1. Lee CJ, Liao CL, Lin YL (2005) Flavivirus activates phosphatidylinositol 3-kinase signaling to block caspase-dependent apoptotic cell death at the early stage of virus infection. J Virol 79: 8388–8399. 10.1128/JVI.79.13.8388-8399.2005
    1. McArdle J, Moorman NJ, Munger J (2012) HCMV targets the metabolic stress response through activation of AMPK whose activity is important for viral replication. PLoS Pathog 8: e1002502 10.1371/journal.ppat.1002502
    1. Hardie DG (2008) AMPK: a key regulator of energy balance in the single cell and the whole organism. Int J Obes (Lond) 32 Suppl 4: S7–12.
    1. Petti C, Vegetti C, Molla A, Bersani I, Cleris L, et al. (2012) AMPK activators inhibit the proliferation of human melanomas bearing the activated MAPK pathway. Melanoma Res 22: 341–350. 10.1097/CMR.0b013e3283544929
    1. Smith JL, Stein DA, Shum D, Fischer MA, Radu C, et al. (2014) Inhibition of dengue virus replication by a class of small-molecule compounds that antagonize dopamine receptor d4 and downstream mitogen-activated protein kinase signaling. J Virol 88: 5533–5542. 10.1128/JVI.00365-14
    1. Menzel N, Fischl W, Hueging K, Bankwitz D, Frentzen A, et al. (2012) MAP-kinase regulated cytosolic phospholipase A2 activity is essential for production of infectious hepatitis C virus particles. PLoS Pathog 8: e1002829 10.1371/journal.ppat.1002829
    1. Scherbik SV, Brinton MA (2010) Virus-induced Ca2+ influx extends survival of west nile virus-infected cells. J Virol 84: 8721–8731. 10.1128/JVI.00144-10
    1. Gupta N, Bhaskar AS, Lakshmana Rao PV (2011) Transcriptional regulation and activation of the mitogen-activated protein kinase pathway after Japanese encephalitis virus infection in neuroblastoma cells. FEMS Immunol Med Microbiol 62: 110–121. 10.1111/j.1574-695X.2011.00792.x
    1. Albarnaz JD, De Oliveira LC, Torres AA, Palhares RM, Casteluber MC, et al. (2014) MEK/ERK activation plays a decisive role in yellow fever virus replication: implication as an antiviral therapeutic target. Antiviral Res 111: 82–92. 10.1016/j.antiviral.2014.09.004
    1. Noisakran S, Dechtawewat T, Avirutnan P, Kinoshita T, Siripanyaphinyo U, et al. (2008) Association of dengue virus NS1 protein with lipid rafts. J Gen Virol 89: 2492–2500. 10.1099/vir.0.83620-0
    1. Vazquez-Martinez R, Cruz-Garcia D, Duran-Prado M, Peinado JR, Castano JP, et al. (2007) Rab18 inhibits secretory activity in neuroendocrine cells by interacting with secretory granules. Traffic 8: 867–882. 10.1111/j.1600-0854.2007.00570.x
    1. Cardaci S, Filomeni G, Ciriolo MR (2012) Redox implications of AMPK-mediated signal transduction beyond energetic clues. J Cell Sci 125: 2115–2125. 10.1242/jcs.095216
    1. Hardie DG, Schaffer BE, Brunet A (2016) AMPK: An Energy-Sensing Pathway with Multiple Inputs and Outputs. Trends Cell Biol 26: 190–201. 10.1016/j.tcb.2015.10.013
    1. Yuan HX, Xiong Y, Guan KL (2013) Nutrient sensing, metabolism, and cell growth control. Mol Cell 49: 379–387. 10.1016/j.molcel.2013.01.019
    1. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, et al. (2012) Cellular and molecular mechanisms of metformin: an overview. Clin Sci (Lond) 122: 253–270.
    1. Zhang H, Shen WJ, Cortez Y, Kraemer FB, Azhar S (2013) Nordihydroguaiaretic acid improves metabolic dysregulation and aberrant hepatic lipid metabolism in mice by both PPARalpha-dependent and -independent pathways. Am J Physiol Gastrointest Liver Physiol 304: G72–86. 10.1152/ajpgi.00328.2012
    1. Arteaga S, Andrade-Cetto A, Cardenas R (2005) Larrea tridentata (Creosote bush), an abundant plant of Mexican and US-American deserts and its metabolite nordihydroguaiaretic acid. J Ethnopharmacol 98: 231–239. 10.1016/j.jep.2005.02.002
    1. Moser TS, Schieffer D, Cherry S (2012) AMP-activated kinase restricts Rift Valley fever virus infection by inhibiting fatty acid synthesis. PLoS Pathog 8: e1002661 10.1371/journal.ppat.1002661
    1. Nakashima K, Takeuchi K, Chihara K, Hotta H, Sada K (2011) Inhibition of hepatitis C virus replication through adenosine monophosphate-activated protein kinase-dependent and -independent pathways. Microbiol Immunol 55: 774–782. 10.1111/j.1348-0421.2011.00382.x
    1. Simanovsky N, Ofek-Shlomai N, Rozovsky K, Ergaz-Shaltiel Z, Hiller N, et al. (2011) Umbilical venous catheter position: evaluation by ultrasound. Eur Radiol 21: 1882–1886. 10.1007/s00330-011-2129-z
    1. York AG, Williams KJ, Argus JP, Zhou QD, Brar G, et al. (2015) Limiting Cholesterol Biosynthetic Flux Spontaneously Engages Type I IFN Signaling. Cell 163: 1716–1729. 10.1016/j.cell.2015.11.045
    1. Prantner D, Perkins DJ, Vogel SN (2017) AMP-activated Kinase (AMPK) Promotes Innate Immunity and Antiviral Defense through Modulation of Stimulator of Interferon Genes (STING) Signaling. J Biol Chem 292: 292–304. 10.1074/jbc.M116.763268
    1. Aguirre S, Maestre AM, Pagni S, Patel JR, Savage T, et al. (2012) DENV inhibits type I IFN production in infected cells by cleaving human STING. PLoS Pathog 8: e1002934 10.1371/journal.ppat.1002934
    1. Munoz-Jordan JL, Sanchez-Burgos GG, Laurent-Rolle M, Garcia-Sastre A (2003) Inhibition of interferon signaling by dengue virus. Proc Natl Acad Sci U S A 100: 14333–14338. 10.1073/pnas.2335168100
    1. Munoz-Jordan JL, Fredericksen BL (2010) How flaviviruses activate and suppress the interferon response. Viruses 2: 676–691. 10.3390/v2020676
    1. Morrison J, Laurent-Rolle M, Maestre AM, Rajsbaum R, Pisanelli G, et al. (2013) Dengue virus co-opts UBR4 to degrade STAT2 and antagonize type I interferon signaling. PLoS Pathog 9: e1003265 10.1371/journal.ppat.1003265
    1. Pagni S, Fernandez-Sesma A (2012) Evasion of the human innate immune system by dengue virus. Immunol Res 54: 152–159. 10.1007/s12026-012-8334-2
    1. Rodriguez-Madoz JR, Belicha-Villanueva A, Bernal-Rubio D, Ashour J, Ayllon J, et al. (2010) Inhibition of the type I interferon response in human dendritic cells by dengue virus infection requires a catalytically active NS2B3 complex. J Virol 84: 9760–9774. 10.1128/JVI.01051-10
    1. Christen V, Treves S, Duong FH, Heim MH (2007) Activation of endoplasmic reticulum stress response by hepatitis viruses up-regulates protein phosphatase 2A. Hepatology 46: 558–565. 10.1002/hep.21611
    1. Guergnon J, Godet AN, Galioot A, Falanga PB, Colle JH, et al. (2011) PP2A targeting by viral proteins: a widespread biological strategy from DNA/RNA tumor viruses to HIV-1. Biochim Biophys Acta 1812: 1498–1507. 10.1016/j.bbadis.2011.07.001
    1. Duong FH, Christen V, Berke JM, Penna SH, Moradpour D, et al. (2005) Upregulation of protein phosphatase 2Ac by hepatitis C virus modulates NS3 helicase activity through inhibition of protein arginine methyltransferase 1. J Virol 79: 15342–15350. 10.1128/JVI.79.24.15342-15350.2005
    1. Haneji T, Hirashima K, Teramachi J, Morimoto H (2013) Okadaic acid activates the PKR pathway and induces apoptosis through PKR stimulation in MG63 osteoblast-like cells. Int J Oncol 42: 1904–1910. 10.3892/ijo.2013.1911
    1. Reineke LC, Lloyd RE (2015) The stress granule protein G3BP1 recruits protein kinase R to promote multiple innate immune antiviral responses. J Virol 89: 2575–2589. 10.1128/JVI.02791-14
    1. Pang J, Hsu JP, Yeo TW, Leo YS, Lye DC (2017) Diabetes, cardiac disorders and asthma as risk factors for severe organ involvement among adult dengue patients: A matched case-control study. Sci Rep 7: 39872 10.1038/srep39872
    1. Pearson-Stuttard J, Blundell S, Harris T, Cook DG, Critchley J (2016) Diabetes and infection: assessing the association with glycaemic control in population-based studies. Lancet Diabetes Endocrinol 4: 148–158. 10.1016/S2213-8587(15)00379-4
    1. Shi Y, Vanhoutte PM (2017) Macro- and Microvascular Endothelial Dysfunction in Diabetes. J Diabetes.
    1. Whitehorn J, Nguyen CV, Khanh LP, Kien DT, Quyen NT, et al. (2016) Lovastatin for the Treatment of Adult Patients With Dengue: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin Infect Dis 62: 468–476. 10.1093/cid/civ949
    1. Mosso C, Galvan-Mendoza IJ, Ludert JE, del Angel RM (2008) Endocytic pathway followed by dengue virus to infect the mosquito cell line C6/36 HT. Virology 378: 193–199. 10.1016/j.virol.2008.05.012
    1. Ludert JE, Mosso C, Ceballos-Olvera I, del Angel RM (2008) Use of a commercial enzyme immunoassay to monitor dengue virus replication in cultured cells. Virol J 5: 51 10.1186/1743-422X-5-51

Source: PubMed

3
S'abonner