Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 12: 673509, 2021.
Article in English | MEDLINE | ID: mdl-34248884

ABSTRACT

Viruses are obligate parasites that depend on the host cell machinery for their replication and dissemination. Cellular lipids play a central role in multiple stages of the viral life cycle such as entry, replication, morphogenesis, and egress. Most viruses reorganize the host cell membranes for the establishment of viral replication complex. These specialized structures allow the segregation of replicating viral RNA from ribosomes and protect it from host nucleases. They also facilitate localized enrichment of cellular components required for viral replication and assembly. The specific composition of the lipid membrane governs its ability to form negative or positive curvature and possess a rigid or flexible form, which is crucial for membrane rearrangement and establishment of viral replication complexes. In this review, we highlight how different viruses manipulate host lipid transfer proteins and harness their functions to enrich different membrane compartments with specific lipids in order to facilitate multiple aspects of the viral life cycle.

2.
Mitochondrion ; 41: 21-27, 2018 07.
Article in English | MEDLINE | ID: mdl-29246869

ABSTRACT

Viruses alter cellular physiology and function to establish cellular environment conducive for viral proliferation. Viral immune evasion is an essential aspect of viral persistence and proliferation. The multifaceted mitochondria play a central role in many cellular events such as metabolism, bioenergetics, cell death, and innate immune signaling. Recent findings accentuate that viruses regulate mitochondrial function and dynamics to facilitate viral proliferation. In this review, we will discuss how viruses exploit mitochondrial dynamics to modulate mitochondria-mediated antiviral innate immune response during infection. This review will provide new insight to understanding the virus-mediated alteration of mitochondrial dynamics and functions to perturb host antiviral immune signaling.


Subject(s)
Host-Pathogen Interactions/immunology , Immune Evasion/immunology , Immunity, Innate/immunology , Mitochondria/immunology , Mitochondrial Dynamics , Viruses/immunology , Animals , Antiviral Agents/pharmacology , Humans , Immune Evasion/drug effects , Mitochondria/pathology , Mitochondria/virology , Signal Transduction
3.
J Virol ; 91(15)2017 08 01.
Article in English | MEDLINE | ID: mdl-28515296

ABSTRACT

Hepatitis C virus (HCV) exists as a lipoprotein-virus hybrid lipoviroparticle (LVP). In vitro studies have demonstrated the importance of apolipoproteins in HCV secretion and infectivity, leading to the notion that HCV coopts the secretion of very-low-density lipoprotein (VLDL) for its egress. However, the mechanisms involved in virus particle assembly and egress are still elusive. The biogenesis of VLDL particles occurs in the endoplasmic reticulum (ER), followed by subsequent lipidation in the ER and Golgi compartment. The secretion of mature VLDL particles occurs through the Golgi secretory pathway. HCV virions are believed to latch onto or fuse with the nascent VLDL particle in either the ER or the Golgi compartment, resulting in the generation of LVPs. In our attempt to unravel the collaboration between HCV and VLDL secretion, we studied HCV particles budding from the ER en route to the Golgi compartment in COPII vesicles. Biophysical characterization of COPII vesicles fractionated on an iodixanol gradient revealed that HCV RNA is enriched in the highly buoyant COPII vesicle fractions and cofractionates with apolipoprotein B (ApoB), ApoE, and the HCV core and envelope proteins. Electron microscopy of immunogold-labeled microsections revealed that the HCV envelope and core proteins colocalize with apolipoproteins and HCV RNA in Sec31-coated COPII vesicles. Ultrastructural analysis also revealed the presence of HCV structural proteins, RNA, and apolipoproteins in the Golgi stacks. These findings support the hypothesis that HCV LVPs assemble in the ER and are transported to the Golgi compartment in COPII vesicles to embark on the Golgi secretory route.IMPORTANCE HCV assembly and release accompany the formation of LVPs that circulate in the sera of HCV patients and are also produced in an in vitro culture system. The pathway of HCV morphogenesis and secretion has not been fully understood. This study investigates the exact site where the association of HCV virions with host lipoproteins occurs. Using immunoprecipitation of COPII vesicles and immunogold electron microscopy (EM), we characterize the existence of LVPs that cofractionate with lipoproteins, viral proteins, RNA, and vesicular components. Our results show that this assembly occurs in the ER, and LVPs thus formed are carried through the Golgi network by vesicular transport. This work provides a unique insight into the HCV LVP assembly process within infected cells and offers opportunities for designing antiviral therapeutic cellular targets.


Subject(s)
COP-Coated Vesicles/virology , Endoplasmic Reticulum/virology , Golgi Apparatus/virology , Hepacivirus/physiology , Lipoproteins, VLDL/metabolism , Virus Assembly , Virus Release , Biological Transport , Hepacivirus/ultrastructure , Humans , Microscopy, Immunoelectron , Virion/metabolism , Virion/ultrastructure
4.
Diabetologia ; 60(4): 729-739, 2017 04.
Article in English | MEDLINE | ID: mdl-28188334

ABSTRACT

AIMS/HYPOTHESIS: In this study, we aimed to evaluate the therapeutic potential of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase, for ameliorating high-fat diet (HFD)-induced pathophysiology in mice. We also aimed to determine whether the beneficial effects of AICAR were dependent on adiponectin. Furthermore, human adipose tissue was used to examine the effect of AICAR ex vivo. METHODS: Six-week-old male C57BL/6J wild-type and Adipoq -/- mice were fed a standard-fat diet (10% fat) or an HFD (60% fat) for 12 weeks and given vehicle or AICAR (500 µg/g) three times/week from weeks 4-12. Diet-induced pathophysiology was examined in mice after 11 weeks by IPGTT and after 12 weeks by flow cytometry and western blotting. Human adipose tissue biopsies from obese (BMI 35-50 kg/m2) individuals were incubated with vehicle or AICAR (1 mmol/l) for 6 h at 37°C, after which inflammation was characterised by ELISA (TNF-α) and flow cytometry. RESULTS: AICAR attenuated adipose inflammation in mice fed an HFD, promoting an M1-to-M2 macrophage phenotype switch, while reducing infiltration of CD8+ T cells. AICAR treatment of mice fed an HFD partially restored glucose tolerance and attenuated hepatic steatosis and kidney disease, as evidenced by reduced albuminuria (p < 0.05), urinary H2O2 (p < 0.05) and renal superoxide levels (p < 0.01) in both wild-type and Adipoq -/- mice. AICAR-mediated protection occurred independently of adiponectin, as similar protection was observed in wild-type and Adipoq -/- mice. In addition, AICAR promoted an M1-to-M2 macrophage phenotype switch and reduced TNF-α production in tissue explants from obese human patients. CONCLUSIONS/INTERPRETATION: AICAR may promote metabolic health and protect against obesity-induced systemic diseases in an adiponectin-independent manner. Furthermore, AICAR reduced inflammation in human adipose tissue explants, suggesting by proof-of-principle that the drug may reduce obesity-induced complications in humans. TRIAL REGISTRATION: ClinicalTrials.gov NCT02322073.


Subject(s)
Adiponectin/metabolism , Diet, High-Fat/adverse effects , Adiponectin/genetics , Animals , Inflammation/immunology , Inflammation/metabolism , Kidney Diseases/immunology , Kidney Diseases/metabolism , Liver Diseases/immunology , Liver Diseases/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , Obesity/metabolism
5.
Antiviral Res ; 109: 132-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25017471

ABSTRACT

Dengue is the most common mosquito borne viral disease in humans. The infection with any of the 4 dengue virus serotypes (DENV) can either be asymptomatic or manifest in two clinical forms, the mild dengue fever or the more severe dengue hemorrhagic fever that may progress into dengue shock syndrome. A DENV replicative cycle relies on host lipid metabolism; specifically, DENV infection modulates cholesterol and fatty acid synthesis, generating a lipid-enriched cellular environment necessary for viral replication. Thus, the aim of this work was to evaluate the anti-DENV effect of the Nordihydroguaiaretic acid (NDGA), a hypolipidemic agent with antioxidant and anti-inflammatory properties. A dose-dependent inhibition in viral yield and NS1 secretion was observed in supernatants of infected cells treated for 24 and 48 h with different concentrations of NDGA. To evaluate the effect of NDGA in DENV replication, a DENV4 replicon transfected Vero cells were treated with different concentrations of NDGA. NDGA treatment significantly reduced DENV replication, reiterating the importance of lipids in viral replication. NDGA treatment also led to reduction in number of lipid droplets (LDs), the neutral lipid storage organelles involved in DENV morphogenesis that are known to increase in number during DENV infection. Furthermore, NDGA treatment resulted in dissociation of the C protein from LDs. Overall our results suggest that NDGA inhibits DENV infection by targeting genome replication and viral assembly.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/virology , Masoprocol/pharmacology , Virus Replication/drug effects , Animals , Cell Line , DNA Replication/drug effects , Dengue/drug therapy , Dengue Virus/genetics , Dengue Virus/growth & development , Dengue Virus/physiology , Genome, Viral/drug effects , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly/drug effects
6.
Proc Natl Acad Sci U S A ; 111(17): 6413-8, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733894

ABSTRACT

Mitochondrial dynamics is crucial for the regulation of cell homeostasis. Our recent findings suggest that hepatitis C virus (HCV) promotes Parkin-mediated elimination of damaged mitochondria (mitophagy). Here we show that HCV perturbs mitochondrial dynamics by promoting mitochondrial fission followed by mitophagy, which attenuates HCV-induced apoptosis. HCV infection stimulated expression of dynamin-related protein 1 (Drp1) and its mitochondrial receptor, mitochondrial fission factor. HCV further induced the phosphorylation of Drp1 (Ser616) and caused its subsequent translocation to the mitochondria, followed by mitophagy. Interference of HCV-induced mitochondrial fission and mitophagy by Drp1 silencing suppressed HCV secretion, with a concomitant decrease in cellular glycolysis and ATP levels, as well as enhanced innate immune signaling. More importantly, silencing Drp1 or Parkin caused significant increase in apoptotic signaling, evidenced by increased cytochrome C release from mitochondria, caspase 3 activity, and cleavage of poly(ADP-ribose) polymerase. These results suggest that HCV-induced mitochondrial fission and mitophagy serve to attenuate apoptosis and may contribute to persistent HCV infection.


Subject(s)
Apoptosis , Hepacivirus/physiology , Mitochondrial Dynamics , Autophagy , Cell Line, Tumor , Dynamins/metabolism , Humans , Immune Evasion , Immunity, Innate , Membrane Proteins/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Mitophagy , Phosphorylation , Phosphoserine/metabolism , Protein Transport
7.
PLoS Pathog ; 9(3): e1003285, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555273

ABSTRACT

Hepatitis C Virus (HCV) induces intracellular events that trigger mitochondrial dysfunction and promote host metabolic alterations. Here, we investigated selective autophagic degradation of mitochondria (mitophagy) in HCV-infected cells. HCV infection stimulated Parkin and PINK1 gene expression, induced perinuclear clustering of mitochondria, and promoted mitochondrial translocation of Parkin, an initial event in mitophagy. Liver tissues from chronic HCV patients also exhibited notable levels of Parkin induction. Using multiple strategies involving confocal and electron microscopy, we demonstrated that HCV-infected cells display greater number of mitophagosomes and mitophagolysosomes compared to uninfected cells. HCV-induced mitophagy was evidenced by the colocalization of LC3 puncta with Parkin-associated mitochondria and lysosomes. Ultrastructural analysis by electron microscopy and immunoelectron microscopy also displayed engulfment of damaged mitochondria in double membrane vesicles in HCV-infected cells. The HCV-induced mitophagy occurred irrespective of genotypic differences. Silencing Parkin and PINK1 hindered HCV replication suggesting the functional relevance of mitophagy in HCV propagation. HCV-mediated decline of mitochondrial complex I enzyme activity was rescued by chemical inhibition of mitophagy or by Parkin silencing. Overall our results suggest that HCV induces Parkin-dependent mitophagy, which may have significant contribution in mitochondrial liver injury associated with chronic hepatitis C.


Subject(s)
Hepacivirus/physiology , Mitochondria, Liver/metabolism , Mitophagy/physiology , Ubiquitin-Protein Ligases/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Gene Expression Regulation , Gene Silencing , Hepacivirus/pathogenicity , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/virology , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Mitochondria, Liver/virology , Phagosomes/metabolism , Phagosomes/ultrastructure , Phagosomes/virology , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Transport , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics , Virus Replication
8.
J Biol Chem ; 287(33): 27637-47, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22745132

ABSTRACT

Hepatitis C virus (HCV) RNA replicates within the ribonucleoprotein complex, assembled on the endoplasmic reticulum (ER)-derived membranous structures closely juxtaposed to the lipid droplets that facilitate the post-replicative events of virion assembly and maturation. It is widely believed that the assembled virions piggy-back onto the very low density lipoprotein particles for secretion. Lipid phosphoinositides are important modulators of intracellular trafficking. Golgi-localized phosphatidylinositol 4-phosphate (PI4P) recruits proteins involved in Golgi trafficking to the Golgi membrane and promotes anterograde transport of secretory proteins. Here, we sought to investigate the role of Golgi-localized PI4P in the HCV secretion process. Depletion of the Golgi-specific PI4P pool by Golgi-targeted PI4P phosphatase hSac1 K2A led to significant reduction in HCV secretion without any effect on replication. We then examined the functional role of a newly identified PI4P binding protein GOLPH3 in the viral secretion process. GOLPH3 is shown to maintain a tensile force on the Golgi, required for vesicle budding via its interaction with an unconventional myosin, MYO18A. Silencing GOLPH3 led to a dramatic reduction in HCV virion secretion, as did the silencing of MYO18A. The reduction in virion secretion was accompanied by a concomitant accumulation of intracellular virions, suggesting a stall in virion egress. HCV-infected cells displayed a fragmented and dispersed Golgi pattern, implicating involvement in virion morphogenesis. These studies establish the role of PI4P and its interacting protein GOLPH3 in HCV secretion and strengthen the significance of the Golgi secretory pathway in this process.


Subject(s)
Golgi Apparatus/metabolism , Hepacivirus/metabolism , Hepatitis C/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Virus Release/physiology , Biological Transport, Active/genetics , Cell Line, Tumor , Golgi Apparatus/genetics , Golgi Apparatus/virology , Hepacivirus/genetics , Hepatitis C/genetics , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/virology , Membrane Proteins/genetics , Myosins/genetics , Myosins/metabolism , Phosphatidylinositol Phosphates/genetics , Virion/genetics , Virion/metabolism
9.
Hepatology ; 54(6): 1936-46, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21858850

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) relies on host lipid metabolic pathways for its replication, assembly, secretion, and entry. HCV induces de novo lipogenesis, inhibits ß-oxidation, and lipoprotein export resulting in a lipid-enriched cellular environment critical for its proliferation. We investigated the effects of a hypolipidemic agent, nordihydroguaiaretic acid (NDGA), on host lipid/fatty acid synthesis and HCV life cycle. NDGA negated the HCV-induced alteration of host lipid homeostasis. NDGA decreased sterol regulatory element binding protein (SREBP) activation and enhanced expression of genes involved in ß-oxidation. NDGA inhibited very low-density lipoprotein (VLDL) secretion by affecting mediators of VLDL biosynthesis. Lipid droplets (LDs), the neutral lipid storage organelles, play a key role in HCV morphogenesis. HCV induces accumulation and perinuclear distribution of LDs, whereas NDGA most notably reduced the overall number and increased the average size of LDs. The antiviral effects of NDGA resulted in reduced HCV replication and secretion. CONCLUSION: NDGA-mediated alterations of host lipid metabolism, LD morphology, and VLDL transport appear to negatively influence HCV proliferation.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hypolipidemic Agents/pharmacology , Lipid Metabolism/drug effects , Masoprocol/pharmacology , Organelles/drug effects , Cell Line, Tumor , Humans , Lipogenesis/drug effects , Lipoproteins, VLDL/drug effects , Lipoproteins, VLDL/metabolism , Virus Replication/drug effects
10.
J Biol Chem ; 286(13): 11265-74, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21285358

ABSTRACT

Hepatitis C virus (HCV) RNA replicates its genome on specialized endoplasmic reticulum modified membranes termed membranous web and utilizes lipid droplets for initiating the viral nucleocapsid assembly. HCV maturation and/or the egress pathway requires host sphingolipid synthesis, which occur in the Golgi. Ceramide transfer protein (CERT) and oxysterol-binding protein (OSBP) play a crucial role in sphingolipid biosynthesis. Protein kinase D (PKD), a serine/threonine kinase, is recruited to the trans-Golgi network where it influences vesicular trafficking to the plasma membrane by regulation of several important mediators via phosphorylation. PKD attenuates the function of both CERT and OSBP by phosphorylation at their respective Ser(132) and Ser(240) residues (phosphorylation inhibition). Here, we investigated the functional role of PKD in HCV secretion. Our studies show that HCV gene expression down-regulated PKD activation. PKD depletion by shRNA or inhibition by pharmacological inhibitor Gö6976 enhanced HCV secretion. Overexpression of a constitutively active form of PKD suppressed HCV secretion. The suppression by PKD was subverted by the ectopic expression of nonphosphorylatable serine mutant CERT S132A or OSBP S240A. These observations imply that PKD negatively regulates HCV secretion/release by attenuating OSBP and CERT functions by phosphorylation inhibition. This study identifies the key role of the Golgi components in the HCV maturation process.


Subject(s)
Gene Expression Regulation, Viral/physiology , Hepacivirus/physiology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Steroid/metabolism , Virus Release/physiology , Amino Acid Substitution , Carbazoles/pharmacology , Cell Membrane/genetics , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Viral/drug effects , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Mutation, Missense , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Steroid/genetics , Sphingolipids/biosynthesis , Sphingolipids/genetics , Virus Release/drug effects
11.
Trends Endocrinol Metab ; 21(1): 33-40, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19854061

ABSTRACT

Hepatitis C virus (HCV) enhances its replication by modulating host cell lipid metabolism. HCV circulates in the blood in association with lipoproteins. HCV infection is associated with enhanced lipogenesis, reduced secretion, and beta-oxidation of lipids. HCV-induced imbalance in lipid homeostasis leads to steatosis. Many lipids are crucial for the virus life cycle, and inhibitors of cholesterol/fatty acid biosynthetic pathways inhibit virus replication, maturation and secretion. HCV negatively modulates the synthesis and secretion of very low-density lipoproteins (VLDL). Components involved in VLDL assembly are also required for HCV morphogenesis/secretion, suggesting that HCV co-opts the VLDL secretory pathway for its own secretion. This review highlights HCV-altered lipid metabolic events that aid the virus life cycle and ultimately promote liver disease.


Subject(s)
Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Host-Pathogen Interactions , Lipid Metabolism , Animals , Fatty Liver/etiology , Fatty Liver/physiopathology , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Lipoproteins, VLDL/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL