Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 341
Filter
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34035171

ABSTRACT

Immunoevasins are viral proteins that prevent antigen presentation on major histocompatibility complex (MHC) class I, thus evading host immune recognition. Hepatitis C virus (HCV) evades immune surveillance to induce chronic infection; however, how HCV-infected hepatocytes affect immune cells and evade immune recognition remains unclear. Herein, we demonstrate that HCV core protein functions as an immunoevasin. Its expression interfered with the maturation of MHC class I molecules catalyzed by the signal peptide peptidase (SPP) and induced their degradation via HMG-CoA reductase degradation 1 homolog, thereby impairing antigen presentation to CD8+ T cells. The expression of MHC class I in the livers of HCV core transgenic mice and chronic hepatitis C patients was impaired but was restored in patients achieving sustained virological response. Finally, we show that the human cytomegalovirus US2 protein, possessing a transmembrane region structurally similar to the HCV core protein, targets SPP to impair MHC class I molecule expression. Thus, SPP represents a potential target for the impairment of MHC class I molecules by DNA and RNA viruses.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Hepacivirus/physiology , Immune Evasion/physiology , Animals , Antigen Presentation/immunology , Cell Line , Down-Regulation , Hepacivirus/immunology , Histocompatibility Antigens Class I/immunology , Humans , Mice , Viral Core Proteins/physiology
2.
Virology ; 541: 85-100, 2020 02.
Article in English | MEDLINE | ID: mdl-32056718

ABSTRACT

The endosomal sorting complex required for transport (ESCRT) pathway is required for efficient egress of Autographa californica multiple nucleopolyhedrovirus (AcMNPV). In this study, we found that Ac93, a baculovirus core protein, contains a conserved MIM1-like motif. Alanine substitutions for six leucine residues in MIM1-like motif revealed that L142, L145, L146, and L149 are required for association of Ac93 with the MIT domain of Vps4. Mutations of these residues also blocked self-association and the association of Ac93 with ESCRT-III proteins or other viral core proteins Ac76 and Ac103, and resulted in a substantial reduction of infectious virus production, less efficient nuclear egress of progeny nucleocapsids, and the defect of intranuclear microvesicles formation. Combined with the localization of the association of Ac93 with ESCRT-III/Vps4 and other viral proteins at the nuclear membrane, we propose that the coordinated action of these viral proteins and ESCRT-III/Vps4 may be involved in remodeling the nuclear membrane.


Subject(s)
ATPases Associated with Diverse Cellular Activities/physiology , Cell Nucleus/metabolism , Endosomal Sorting Complexes Required for Transport/physiology , Nucleocapsid/physiology , Nucleopolyhedroviruses/physiology , Vacuolar Proton-Translocating ATPases/physiology , Viral Core Proteins/physiology , ATPases Associated with Diverse Cellular Activities/chemistry , Amino Acid Motifs , Animals , Endosomal Sorting Complexes Required for Transport/chemistry , Host Microbial Interactions , Nucleocapsid/chemistry , Protein Domains , Spodoptera , Vacuolar Proton-Translocating ATPases/chemistry
3.
Microsc Res Tech ; 83(5): 499-506, 2020 May.
Article in English | MEDLINE | ID: mdl-31926041

ABSTRACT

The aim of this study was to assess nucleo-cytoplasmic protein localization to better understand the exact intracellular localization of viral proteins involved with infections. Having determined the general protein localization of hepatitis B virus P22 precore protein, the aim was to more specifically resolve its intracellular organization. This was done using both laser scanning microscopy and Airyscan techniques. Using a 63× objective, the resolution obtained with Airyscan was increased by 1.5-fold as compared to confocal microscopy (p value <.00001).


Subject(s)
Cytoplasm/virology , Hepatitis B Core Antigens/physiology , Hepatitis B virus/chemistry , Image Processing, Computer-Assisted/methods , Viral Core Proteins/physiology , Cell Line , Humans , Microscopy, Confocal , Protein Precursors , Protein Transport
4.
Nucleic Acids Res ; 47(11): 5837-5851, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31066445

ABSTRACT

Ebola virus (EBOV) is a non-segmented, negative-sense RNA virus (NNSV) in the family Filoviridae, and is recognized as one of the most lethal pathogens in the planet. For RNA viruses, cellular or virus-encoded RNA helicases play pivotal roles in viral life cycles by remodelling viral RNA structures and/or unwinding viral dsRNA produced during replication. However, no helicase or helicase-like activity has ever been found to associate with any NNSV-encoded proteins, and it is unknown whether the replication of NNSVs requires the participation of any viral or cellular helicase. Here, we show that despite of containing no conserved NTPase/helicase motifs, EBOV VP35 possesses the NTPase and helicase-like activities that can hydrolyse all types of NTPs and unwind RNA helices in an NTP-dependent manner, respectively. Moreover, guanidine hydrochloride, an FDA-approved compound and inhibitor of certain viral helicases, inhibited the NTPase and helicase-like activities of VP35 as well as the replication/transcription of an EBOV minigenome replicon in cells, highlighting the importance of VP35 helicase-like activity during EBOV life cycle. Together, our findings provide the first demonstration of the NTPase/helicase-like activity encoded by EBOV, and would foster our understanding of EBOV and NNSVs.


Subject(s)
Ebolavirus/genetics , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/virology , Nucleoproteins/physiology , RNA, Double-Stranded/chemistry , Viral Core Proteins/physiology , Adenosine Triphosphate/chemistry , Amino Acid Motifs , Cells, Cultured , DNA Helicases/metabolism , Humans , Nucleocapsid Proteins , Nucleoproteins/genetics , Nucleoproteins/metabolism , Nucleoside-Triphosphatase/genetics , Protein Binding , RNA Helicases/metabolism , RNA, Viral/metabolism , Viral Core Proteins/genetics , Viral Core Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication
5.
Liver Int ; 39(7): 1226-1236, 2019 07.
Article in English | MEDLINE | ID: mdl-30938910

ABSTRACT

BACKGROUND & AIMS: miR-21-5p is a potent oncogenic microRNA targeting many key tumour suppressors including phosphatase and tensin homolog (PTEN). We recently identified PTEN as a key factor modulated by hepatitis C virus (HCV) to promote virion egress. In hepatocytes, expression of HCV-3a core protein was sufficient to downregulate PTEN and to trigger lipid droplet accumulation. Here, we investigated whether HCV controls PTEN expression through miR-21-5p-dependent mechanisms to trigger steatosis in hepatocytes and to promote HCV life cycle. METHODS: MiR-21-5p expression in HCV-infected patients was evaluated by transcriptome meta-analysis. HCV replication and viral particle production were investigated in Jc1-infected Huh-7 cells after miR-21-5p inhibition. PTEN expression and steatosis were assessed in HCV-3a core protein-expressing Huh-7 cells and in mouse primary hepatocytes having miR-21-5p inhibited or genetically deleted respectively. HCV-3a core-induced steatosis was assessed in vivo in Mir21a knockout mice. RESULTS: MiR-21-5p expression was significantly increased in hepatic tissues from HCV-infected patients. Infection by HCV-Jc1, or transduction with HCV-3a core, upregulated miR-21-5p expression and/or activity in Huh-7 cells. miR-21-5p inhibition decreased HCV replication and release of infectious virions by Huh-7 cells. HCV-3a core-induced PTEN downregulation and steatosis were further prevented in Huh-7 cells following miR-21-5p inhibition or in Mir21a knockout mouse primary hepatocytes. Finally, steatosis induction by AAV8-mediated HCV-3a core expression was reduced in vivo in Mir21a knockout mice. CONCLUSION: MiR-21-5p activation by HCV is a key molecular step, promoting both HCV life cycle and HCV-3a core-induced steatosis and may be among the molecular changes induced by HCV-3a to promote carcinogenesis.


Subject(s)
Hepacivirus/physiology , Hepatocytes/metabolism , Hepatocytes/virology , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Viral Core Proteins/physiology , Animals , Carcinogenesis , Cell Line, Tumor , Down-Regulation , Fatty Liver/metabolism , Fatty Liver/pathology , Hepacivirus/genetics , Hepatocytes/pathology , Humans , Lipid Metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Up-Regulation , Virus Replication
6.
PLoS Pathog ; 15(2): e1007593, 2019 02.
Article in English | MEDLINE | ID: mdl-30811485

ABSTRACT

Hepatitis C virus (HCV) infection remains a major cause of hepatic inflammation and liver disease. HCV triggers NLRP3 inflammasome activation and interleukin-1ß (IL-1ß) production from hepatic macrophages, or Kupffer cells, to drive the hepatic inflammatory response. Here we examined HCV activation of the NLRP3 inflammasome signaling cascade in primary human monocyte derived macrophages and THP-1 cell models of hepatic macrophages to define the HCV-specific agonist and cellular processes of inflammasome activation. We identified the HCV core protein as a virion-specific factor of inflammasome activation. The core protein was both necessary and sufficient for IL-1ß production from macrophages exposed to HCV or soluble core protein alone. NLRP3 inflammasome activation by the HCV core protein required calcium mobilization linked with phospholipase-C activation. Our findings reveal a molecular basis of hepatic inflammasome activation and IL-1ß release triggered by HCV core protein.


Subject(s)
Calcium Signaling/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Viral Core Proteins/physiology , Calcium/metabolism , Carrier Proteins , Hepacivirus/metabolism , Hepacivirus/pathogenicity , Hepatitis C , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/physiology , Kupffer Cells , Liver , Liver Diseases , Macrophages/metabolism , NF-kappa B , Primary Cell Culture , Reactive Oxygen Species , THP-1 Cells/metabolism , Type C Phospholipases/metabolism , Viral Core Proteins/metabolism
7.
Med Sci (Paris) ; 34(8-9): 693-700, 2018.
Article in French | MEDLINE | ID: mdl-30230454

ABSTRACT

Chronic infection by hepatitis B virus (HBV) is a major public health problem with more than 250 millions of people chronically infected worldwide who have a high risk to develop cirrhosis and hepatocellular carcinoma. Available treatments reduce viremia but do not eradicate the virus from hepatocytes. Therefore, there is an urgent need to develop new classes of antiviral molecules and the viral capsid protein, Core, constitutes a new favored target. Core protein Allosteric Modulators (CAMs) targeting its assembly functions are in clinical development. In addition, investigation of Core regulatory functions may lead to the development of compounds targeting cellular factors (HTA) that could be used in combined therapies aiming to achieve a better control of HBV replication.


Subject(s)
Biomedical Research/trends , Drug Discovery , Hepatitis B virus/chemistry , Hepatitis B/drug therapy , Viral Core Proteins/physiology , Antiviral Agents/isolation & purification , Antiviral Agents/therapeutic use , Drug Discovery/standards , Drug Discovery/trends , Humans , Molecular Targeted Therapy/standards , Molecular Targeted Therapy/trends
8.
Virus Genes ; 53(3): 400-409, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28190135

ABSTRACT

The Orf virus 050 (ORFV050) gene is located in the core region of the ORFV genome. It is similar to Vaccinia virus (VV) Copenhagen L4R, and encodes the DNA-binding virion core protein VP8, which has structures similar to the VV P25K core protein and may undergo similar proteolytic processing during virus assembly. Three conserved Ala-Gly-X motifs at putative cleavage sites were identified in ORFV050. To investigate the proteolysis of ORFV050 and its participation in viral assembly, full-length and site-directed mutant ORFV050 recombinant proteins were constructed and expressed. Two distinct protein bands of 28.5 and 25 kDa were detected in the infected cells using anti-ORFV050 polyclonal antiserum. A potential cleavage site was identified at amino acids 30-32 of ORFV050. Mutation of AG/A to (R) in ORFV050 abolished the process of proteolysis. ORFV050 is a late gene synthesized during viral replication in the host cytoplasm. According to these results, we conclude that ORFV050 undergoes proteolysis and plays an important role in viral assembly.


Subject(s)
Genes, Viral/genetics , Orf virus/enzymology , Orf virus/genetics , Proteolysis , Viral Proteins/genetics , Viral Proteins/isolation & purification , Amino Acid Sequence , Animals , Antibodies, Viral , Cell Line , Cytoplasm/virology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dipeptides , Ecthyma, Contagious/virology , Gene Expression Regulation, Viral , Molecular Weight , Mutation , Orf virus/drug effects , Orf virus/physiology , Recombinant Fusion Proteins/genetics , Rifampin/pharmacology , Sequence Alignment , Sequence Analysis , Sheep , Vaccinia virus/genetics , Viral Core Proteins/genetics , Viral Core Proteins/physiology , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virion/metabolism , Virus Assembly/physiology , Virus Replication
9.
Biochem Biophys Res Commun ; 483(2): 904-909, 2017 02 05.
Article in English | MEDLINE | ID: mdl-28082202

ABSTRACT

Chronic infection with hepatitis C virus (HCV) induces liver fibrosis and cancer. In particular metabolic alterations and associated oxidative stress induced by the virus play a key role in disease progression. Albeit the pivotal role of biogenic polyamines spermine and spermidine in the regulation of liver metabolism and function and cellular control of redox homeostasis, their role in the viral life cycle has not been studied so far. Here we show that in cell lines expressing two viral proteins, capsid and the non-structural protein 5A, expression of the two key enzymes of polyamine biosynthesis and degradation, respectively, ornithine decarboxylase (ODC) and spermidine/spermine-N1-acetyl transferase (SSAT), increases transiently. In addition, both HCV core and NS5A induce sustained expression of spermine oxidase (SMO), an enzyme that catalyzes conversion of spermine into spermidine. Human hepatoma Huh7 cells harboring a full-length HCV replicon exhibited suppressed ODC and SSAT levels and elevated levels of SMO leading to decreased intracellular concentrations of spermine and spermidine. Thus, role of HCV-driven alterations of polyamine metabolism in virus replication and development of HCV-associated liver pathologies should be explored in future.


Subject(s)
Biogenic Polyamines/metabolism , Hepacivirus/physiology , Hepacivirus/pathogenicity , Acetyltransferases/genetics , Acetyltransferases/metabolism , Cell Line , Gene Expression Regulation, Enzymologic , Hepacivirus/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Spermidine/metabolism , Spermine/metabolism , Viral Core Proteins/physiology , Viral Nonstructural Proteins/physiology , Virus Replication/physiology , Polyamine Oxidase
10.
Microsc Microanal ; 23(1): 56-68, 2017 02.
Article in English | MEDLINE | ID: mdl-28112080

ABSTRACT

The bulk of the major core protein VP7 in African horse sickness virus (AHSV) self-assembles into flat, hexagonal crystalline particles in a process appearing unrelated to viral replication. Why this unique characteristic of AHSV VP7 is genetically conserved, and whether VP7 aggregation and particle formation have an effect on cellular biology or the viral life cycle, is unknown. Here we investigated how different small peptide and enhanced green fluorescent protein (eGFP) insertions into the VP7 top domain affected VP7 localization, aggregation, and particle formation. This was done using a dual laser scanning confocal and transmission electron microscopy approach in conjunction with analyses of the solubility, aggregation, and fluorescence profiles of the proteins. VP7 top domain modifications did not prevent trimerization, or intracellular trafficking, to one or two discrete sites in the cell. However, modifications that resulted in a misfolded and insoluble VP7-eGFP component blocked trafficking, and precluded protein accumulation at a single cellular site, perhaps by interfering with normal trimer-trimer interactions. Furthermore, the modifications disrupted the stable layering of the trimers into characteristic AHSV VP7 crystalline particles. It was concluded that VP7 trafficking is driven by a balance between VP7 solubility, trimer forming ability, and trimer-trimer interactions.


Subject(s)
African Horse Sickness Virus/metabolism , Microscopy, Confocal/methods , Microscopy, Electron, Transmission/methods , Viral Core Proteins/physiology , Viral Core Proteins/ultrastructure , African Horse Sickness Virus/genetics , Animals , Baculoviridae/genetics , Gene Expression Regulation, Viral , Genetic Vectors , Green Fluorescent Proteins , Life Cycle Stages , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/ultrastructure , Sf9 Cells , Viral Core Proteins/chemistry , Viral Core Proteins/genetics , Viral Fusion Proteins/physiology , Viral Fusion Proteins/ultrastructure , Virus Replication
11.
Cell Microbiol ; 19(4)2017 04.
Article in English | MEDLINE | ID: mdl-27665576

ABSTRACT

Lipoprotein lipase (LPL) has been identified as an anti-hepatitis C virus (HCV) host factor, but the cellular mechanism remains elusive. Here, we investigated the cellular mechanism of LPL involving in anti-HCV. The functional activation of peroxisome proliferator-activated receptor (PPAR) α signal by LPL transducing into hepatocytes was investigated in HCV-infected cells, primary human hepatocytes, and in HCV-core transgenic mice. The result showed that the levels of transcriptional transactivity and nuclear translocation of PPARα in Huh7 cells and primary human hepatocytes were elevated by physiologically ranged LPL treatment of either very-low density lipoprotein or HCV particles. The LPL-induced hepatic PPARα activation was weakened by blocking the LPL enzymatic activity, and by preventing the cellular uptake of free unsaturated fatty acids with either albumin chelator or silencing of CD36 translocase. The knockdowns of PPARα and CD36 reversed the LPL-mediated suppression of HCV infection. Furthermore, treatment with LPL, like the direct activation of PPARα, not only reduced the levels of apolipoproteins B, E, and J, which are involved in assembly and release of HCV virions, but also alleviated hepatic lipid accumulation induced by core protein. HCV-core transgenic mice exhibited more hepatic miR-27b, which negatively regulates PPARα expression, than did the wild-type controls. The induction of LPL activity by fasting in the core transgenic mice activated PPARα downstream target genes that are involved in fatty acid ß-oxidation. Taken together, our study reveals dual beneficial outcomes of LPL in anti-HCV and anti-steatosis and shed light on the control of chronic hepatitis C in relation to LPL modulators.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Lipoprotein Lipase/physiology , Liver/enzymology , Animals , CD36 Antigens/metabolism , Cell Line, Tumor , Gene Expression , Hepatitis C/virology , Hepatocytes/enzymology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Immunity, Innate , Lipolysis , Lipoproteins, VLDL/metabolism , Liver/virology , Male , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , PPAR alpha/metabolism , Viral Core Proteins/physiology
12.
World J Gastroenterol ; 22(17): 4354-61, 2016 May 07.
Article in English | MEDLINE | ID: mdl-27158204

ABSTRACT

AIM: To investigate the role of miR-125b in regulating monocyte immune responses induced by hepatitis C virus (HCV) core protein. METHODS: Monocytic THP-1 cells were treated with various concentrations of recombinant HCV core protein, and cytokines and miR-125b expression in these cells were analyzed. The requirement of Toll-like receptor 2 (TLR2) or MyD88 gene for HCV core protein-induced immune responses was determined by the transfection of THP-1 cells with gene knockdown vectors expressing either TLR2 siRNA or MyD88 siRNA. The effect of miR-125b overexpression on TLR2/MyD88 signaling was examined by transfecting THP-1 cells with miR-125b mimic RNA oligos. RESULTS: In response to HCV core protein stimulation, cytokine production was up-regulated and miR-125b expression was down-regulated in THP-1 cells. The modulatory effect of HCV core protein on cellular events was dose-dependent and required functional TLR2 or MyD88 gene. Forced miR-125b expression abolished the HCV core protein-induced enhancement of tumor necrosis factor-α, interleukin (IL)-6, and IL-10 expression by 66%, 54%, and 66%, respectively (P < 0.001), by inhibiting MyD88-mediated signaling, including phosphorylation of NF-κBp65, ERK, and P38. CONCLUSION: The inverse correlation between miR-125b and cytokine expression after HCV core challenge suggests that miR-125b may negatively regulate HCV-induced immune responses by targeting TLR2/MyD88 signaling in monocytes.


Subject(s)
Hepacivirus/physiology , MicroRNAs/physiology , Monocytes/immunology , Myeloid Differentiation Factor 88/physiology , Signal Transduction/physiology , Toll-Like Receptor 2/physiology , Viral Core Proteins/physiology , Cell Line, Tumor , Host-Pathogen Interactions , Humans , Interleukin-10/genetics , Interleukin-6/genetics , Tumor Necrosis Factor-alpha/genetics
13.
J Virol ; 90(10): 5108-5118, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962215

ABSTRACT

UNLABELLED: Dendritic cells (DCs) are major targets of filovirus infection in vivo Previous studies have shown that the filoviruses Ebola virus (EBOV) and Marburg virus (MARV) suppress DC maturation in vitro Both viruses also encode innate immune evasion functions. The EBOV VP35 (eVP35) and the MARV VP35 (mVP35) proteins each can block RIG-I-like receptor signaling and alpha/beta interferon (IFN-α/ß) production. The EBOV VP24 (eVP24) and MARV VP40 (mVP40) proteins each inhibit the production of IFN-stimulated genes (ISGs) by blocking Jak-STAT signaling; however, this occurs by different mechanisms, with eVP24 blocking nuclear import of tyrosine-phosphorylated STAT1 and mVP40 blocking Jak1 function. MARV VP24 (mVP24) has been demonstrated to modulate host cell antioxidant responses. Previous studies demonstrated that eVP35 is sufficient to strongly impair primary human monocyte-derived DC (MDDC) responses upon stimulation induced through the RIG-I-like receptor pathways. We demonstrate that mVP35, like eVP35, suppresses not only IFN-α/ß production but also proinflammatory responses after stimulation of MDDCs with RIG-I activators. In contrast, eVP24 and mVP40, despite suppressing ISG production upon RIG-I activation, failed to block upregulation of maturation markers or T cell activation. mVP24, although able to stimulate expression of antioxidant response genes, had no measurable impact of DC function. These data are consistent with a model where filoviral VP35 proteins are the major suppressors of DC maturation during filovirus infection, whereas the filoviral VP24 proteins and mVP40 are insufficient to prevent DC maturation. IMPORTANCE: The ability to suppress the function of dendritic cells (DCs) likely contributes to the pathogenesis of disease caused by the filoviruses Ebola virus and Marburg virus. To clarify the basis for this DC suppression, we assessed the effect of filovirus proteins known to antagonize innate immune signaling pathways, including Ebola virus VP35 and VP24 and Marburg virus VP35, VP40, and VP24, on DC maturation and function. The data demonstrate that the VP35s from Ebola virus and Marburg virus are the major suppressors of DC maturation and that the effects on DCs of the remaining innate immune inhibitors are minor.


Subject(s)
Dendritic Cells/physiology , Dendritic Cells/virology , Ebolavirus/chemistry , Marburgvirus/chemistry , RNA Viruses/physiology , Viral Proteins/physiology , Viral Regulatory and Accessory Proteins/physiology , Cell Differentiation , Encephalomyocarditis virus/physiology , Host-Pathogen Interactions , Humans , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-gamma/metabolism , Nucleocapsid Proteins , Nucleoproteins/genetics , Nucleoproteins/physiology , Sendai virus/physiology , Transduction, Genetic , Viral Core Proteins/genetics , Viral Core Proteins/physiology , Viral Proteins/genetics
14.
Biochem Biophys Res Commun ; 466(3): 592-8, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26392314

ABSTRACT

This study investigated the effect of HCV core protein on the proliferation of hepatocytes and hepatocellular carcinoma cells (HCC), the influence of HCV core protein on HCC apoptosis induced by the chemotherapeutic agent cisplatin, and the mechanism through which HCV core protein acts as a potential oncoprotein in HCV-related HCC by measuring the levels of NR4A1 and Runt-related transcription factor 3 (RUNX3), which are associated with tumor suppression and chemotherapy resistance. In the present study, PcDNA3.1-core and RUNX3 siRNA were transfected into LO2 and HepG2 cells using Lipofectamine 2000. LO2-core, HepG2-core, LO2-RUNX3 (low) and control cells were treated with different concentrations of cisplatin for 72 h, and cell proliferation and apoptosis were assayed using the CellTiter 96(®)Aqueous Non-Radioactive Cell Proliferation Assay Kit. Western blot and real time PCR analyses were used to detect NR4A1, RUNX3, smad7, Cyclin D1 and BAX. Confocal microscopy was used to determine the levels of NR4A1 in HepG2 and HepG2-core cells. The growth rate of HepG2-core cells was considerably greater than that of HepG2 cells. HCV core protein increased the expression of cyclin D1 and decreased the expressions of NR4A1 and RUNX3. In LO2 - RUNX3 (low), the rate of cell proliferation and the level of cisplatin resistance were the same as in the LO2 -core. These results suggest that HCV core protein decreases the sensitivity of hepatocytes to cisplatin by inhibiting the expression of NR4A1 and promoting the expression of smad7, which negatively regulates the TGF-ß pathway. This effect results in down regulation of RUNX3, a target of the TGF-ß pathway. Taken together, these findings indicate that in hepatocytes, HCV core protein increases drug resistance and inhibits cell apoptosis by inhibiting the expressions of NR4A1 and RUNX3.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Hepatocytes/pathology , Hepatocytes/virology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Viral Core Proteins/physiology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line , Cell Proliferation/drug effects , Cisplatin/pharmacology , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Cyclin D1/metabolism , Drug Resistance, Viral , Hep G2 Cells , Hepatocytes/metabolism , Host-Pathogen Interactions , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Smad7 Protein/metabolism
15.
Inflammation ; 38(5): 1823-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25894282

ABSTRACT

Hepatitis C virus (HCV) is a major cause of chronic liver disease and has led to cirrhosis or hepatocellular carcinoma in a majority of infected individuals. We have previously demonstrated that the HCV alternate reading frame protein (F protein) is related to Th1/Th2 bias in chronic hepatitis C (CHC) patients, and we aimed to explore the relative molecular mechanisms here. A total of 104 cases including CHC patients and healthy donors were enrolled. T-bet and GATA-3 expression levels were analyzed in peripheral blood mononuclear cells (PBMCs). The levels of signal transducer and activator of transcription-1/-6(STAT1/6) and phosphorylated STAT1/6(pSTAT1/6) in PBMCs were measured by Western blotting. Our results showed that the levels of T-bet in PBMCs, as well as the levels of gamma interferon (IFN-γ) in sera, were decreased in anti-F protein antibody seropositive patients compared with anti-F protein antibody seronegative patients, whereas the levels of GATA-3 did not show difference between the two groups. Moreover, the decreased pSTAT1 and increased pSTAT6 were observed in PBMCs by HCV core/F protein stimulation with constant STAT1/6 expression. Taken together, it suggested that T-bet may be involved in Th1/Th2 bias induced by HCV F protein, and the disruption of STAT phosphorylation may participate in this mediation.


Subject(s)
Hepacivirus/physiology , Reading Frames/physiology , T-Box Domain Proteins/biosynthesis , Viral Core Proteins/physiology , Adult , Female , Gene Expression Regulation , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/genetics , Humans , Jurkat Cells , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/virology , Male , Middle Aged , T-Box Domain Proteins/genetics
16.
J Virol ; 89(10): 5462-77, 2015 May.
Article in English | MEDLINE | ID: mdl-25740981

ABSTRACT

The ubiquitous ATP-dependent RNA helicase DDX3X is involved in many cellular functions, including innate immunity, and is a pivotal host factor for hepatitis C virus (HCV) infection. Recently, we showed that DDX3X specifically recognizes the HCV 3' untranslated region (UTR), leading to the activation of IKK-α and a cascade of lipogenic signaling to facilitate lipid droplet biogenesis and viral assembly (Q. Li, V. Pene, S. Krishnamurthy, H. Cha, and T. J. Liang, Nat Med 19:722-729, 2013, http://dx.doi.org/10.1038/nm.3190). The interaction of DDX3X with HCV core protein seems to be dispensable for its proviral role. In this study, through systematic imaging and biochemical and virologic approaches, we identified a dynamic association between DDX3X and various cellular compartments and viral elements mediating multiple functions of DDX3X in productive HCV infection. Upon HCV infection, the HCV 3'UTR interacts with DDX3X and IKK-α, which redistribute to speckle-like cytoplasmic structures shown to be stress granules (SGs). As viral proteins accumulate in infected cells, DDX3X granules together with SG-associated proteins redistribute and colocalize with HCV core protein around lipid droplets (LDs). IKK-α, however, does not relocate to the LD but translocates to the nucleus. In HCV-infected cells, various HCV nonstructural proteins also interact or colocalize with DDX3X in close proximity to SGs and LDs, consistent with the tight juxtaposition of the replication complex and the assembly site at the surface of LDs. Short interfering RNA (siRNA)-mediated silencing of DDX3X and multiple SG components markedly inhibits HCV infection. Our data suggest that DDX3X initiates a multifaceted cellular program involving dynamic associations with HCV RNA and proteins, IKK-α, SG, and LD surfaces for its crucial role in the HCV life cycle. IMPORTANCE DDX3X is a proviral host factor for HCV infection. Recently, we showed that DDX3X binds to the HCV 3'UTR, activating IKK-α and cellular lipogenesis to facilitate viral assembly (Q. Li et al., Nat Med 19:722-729, 2013, http://dx.doi.org/10.1038/nm.3190). Here, we report associations of DDX3X with various cellular compartments and viral elements that mediate its multiple functions in the HCV life cycle. Upon infection, the HCV 3'UTR redistributes DDX3X and IKK-α to speckle-like cytoplasmic structures shown to be SGs. Subsequently, interactions between DDX3X, SG, and HCV proteins facilitate the translocation of DDX3X-SG complexes to the LD surface. HCV nonstructural proteins are shown to colocalize with DDX3X in close proximity to SGs and LDs, consistent with the tight juxtaposition of the HCV replication complex and assembly site at the LD surface. Our data demonstrate that DDX3X initiates a multifaceted cellular program involving dynamic associations with HCV elements, IKK-α, SGs, and LDs for its critical role in HCV infection.


Subject(s)
DEAD-box RNA Helicases/physiology , Hepatitis C, Chronic/etiology , Host-Pathogen Interactions/physiology , I-kappa B Kinase/physiology , 3' Untranslated Regions , Cell Line , Cytoplasmic Granules/physiology , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepacivirus/physiology , Hepatitis C, Chronic/physiopathology , Hepatitis C, Chronic/virology , Humans , Lipid Metabolism , Models, Biological , Viral Core Proteins/physiology , Viral Nonstructural Proteins/physiology , Virus Replication
17.
J Biol Chem ; 289(52): 35770-80, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25381252

ABSTRACT

Liver steatosis is a common health problem associated with hepatitis C virus (HCV) and an important risk factor for the development of liver fibrosis and cancer. Steatosis is caused by triglycerides (TG) accumulating in lipid droplets (LDs), cellular organelles composed of neutral lipids surrounded by a monolayer of phospholipids. The HCV nucleocapsid core localizes to the surface of LDs and induces steatosis in cultured cells and mouse livers by decreasing intracellular TG degradation (lipolysis). Here we report that core at the surface of LDs interferes with the activity of adipose triglyceride lipase (ATGL), the key lipolytic enzyme in the first step of TG breakdown. Expressing core in livers or mouse embryonic fibroblasts of ATGL(-/-) mice no longer decreases TG degradation as observed in LDs from wild-type mice, supporting the model that core reduces lipolysis by engaging ATGL. Core must localize at LDs to inhibit lipolysis, as ex vivo TG hydrolysis is impaired in purified LDs coated with core but not when free core is added to LDs. Coimmunoprecipitation experiments revealed that core does not directly interact with the ATGL complex but, unexpectedly, increased the interaction between ATGL and its activator CGI-58 as well as the recruitment of both proteins to LDs. These data link the anti-lipolytic activity of the HCV core protein with altered ATGL binding to CGI-58 and the enhanced association of both proteins with LDs.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Lipase/metabolism , Lipid Droplets/enzymology , Viral Core Proteins/physiology , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , Humans , Hydrolysis , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Triglycerides/metabolism
18.
Res Vet Sci ; 97(2): 449-54, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25218811

ABSTRACT

Recent studies show that classical swine fever virus (CSFV) NS5A is an essential replicase component, but it is not known how NS5A participates in viral particle production. In this study, deletion and substitution mutations were introduced into the C-terminus of CSFV NS5A. The efficiency of Core protein release and extracellular and intracellular infectivity levels were assessed and NS5A-Core interaction was investigated. These results suggested that CSFV NS5A was a key factor for the assembly of infectious CSFV particles. The C-terminal sequence from amino acids 478 to 487 and amino acids S481 and T482 were necessary for CSFV assembly and production. The effect of NS5A on CSFV assembly and production might be related to NS5A-Core interaction. T482 was found to be conserved in the C-terminus of NS5A proteins of pestiviruses and hepatitis C virus (HCV), therefore suggesting that it might be important for these virus assembly and production.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/physiology , DNA, Viral/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virion/physiology , Virus Assembly/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cells, Cultured , Genes, Essential/genetics , In Vitro Techniques , Kidney/cytology , Molecular Sequence Data , Protein Biosynthesis/genetics , RNA-Dependent RNA Polymerase/genetics , Swine , Transfection/methods , Transfection/veterinary , Viral Core Proteins/analysis , Viral Core Proteins/genetics , Viral Core Proteins/physiology , Viral Nonstructural Proteins/analysis , Virion/genetics , Virus Assembly/genetics
19.
Protein Sci ; 23(11): 1519-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25159197

ABSTRACT

Filoviruses are the causative agents of a severe and often fatal hemorrhagic fever with repeated outbreaks in Africa. They are negative sense single stranded enveloped viruses that can cross species barriers from its natural host bats to primates including humans. The small size of the genome poses limits to viral adaption, which may be partially overcome by conformational plasticity. Here we review the different conformational states of the Ebola virus (EBOV) matrix protein VP40 that range from monomers, to dimers, hexamers, and RNA-bound octamers. This conformational plasticity that is required for the viral life cycle poses a unique opportunity for development of VP40 specific drugs. Furthermore, we compare the structure to homologous matrix protein structures from Paramyxoviruses and Bornaviruses and we predict that they do not only share the fold but also the conformational flexibility of EBOV VP40.


Subject(s)
Nucleoproteins , Viral Core Proteins , Ebolavirus/metabolism , Ebolavirus/physiology , Models, Molecular , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Nucleoproteins/physiology , Protein Conformation , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , Viral Core Proteins/physiology , Virus Assembly , Virus Release
20.
FEBS Lett ; 588(18): 3501-10, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25131930

ABSTRACT

Despite their differential cell tropisms, HIV-1 and HCV dramatically influence disease progression in coinfected patients. Macrophages are important target cells of HIV-1. We hypothesized that secreted HCV core protein might modulate HIV-1 replication. We demonstrate that HCV core significantly enhances HIV-1 replication in human macrophages by upregulating TNF-α and IL-6 via TLR2-, JNK-, and MEK1/2-dependent pathways. Furthermore, we show that TNF-α and IL-6 secreted from HCV core-treated macrophages reactivates monocytic U1 cells latently infected with HIV-1. Our studies reveal a previously unrecognized role of HCV core by enhancing HIV-1 infection in macrophages.


Subject(s)
HIV-1/physiology , Interleukin-6/genetics , Macrophages/virology , Tumor Necrosis Factor-alpha/genetics , Viral Core Proteins/physiology , Virus Replication , Coinfection/virology , HEK293 Cells , HIV Infections/virology , Hepatitis C/virology , Humans , Interleukin-6/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...