Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 17(5): e1009549, 2021 05.
Article in English | MEDLINE | ID: mdl-33984068

ABSTRACT

The antiviral innate immune response mainly involves type I interferon (IFN) in mammalian cells. The contribution of the RNA silencing machinery remains to be established, but several recent studies indicate that the ribonuclease DICER can generate viral siRNAs in specific conditions. It has also been proposed that type I IFN and RNA silencing could be mutually exclusive antiviral responses. In order to decipher the implication of DICER during infection of human cells with alphaviruses such as the Sindbis virus and Semliki forest virus, we determined its interactome by proteomics analysis. We show that DICER specifically interacts with several double-stranded RNA binding proteins and RNA helicases during viral infection. In particular, proteins such as DHX9, ADAR-1 and the protein kinase RNA-activated (PKR) are enriched with DICER in virus-infected cells. We demonstrate that the helicase domain of DICER is essential for this interaction and that its deletion confers antiviral properties to this protein in an RNAi-independent, PKR-dependent, manner.


Subject(s)
Alphavirus Infections/drug therapy , Antiviral Agents/pharmacology , DEAD-box RNA Helicases/metabolism , Protein Interaction Domains and Motifs/drug effects , Ribonuclease III/metabolism , Semliki forest virus/drug effects , Virus Replication , eIF-2 Kinase/metabolism , Alphavirus Infections/metabolism , Alphavirus Infections/pathology , DEAD-box RNA Helicases/genetics , HEK293 Cells , Humans , Interferon Type I/pharmacology , Ribonuclease III/genetics , eIF-2 Kinase/genetics
2.
Gut ; 66(5): 896-907, 2017 05.
Article in English | MEDLINE | ID: mdl-27609828

ABSTRACT

OBJECTIVE: The HCV life cycle and the lipid metabolism are inextricably intertwined. In the blood, HCV virions are associated with lipoproteins, forming lipoviroparticles (LVPs), which are the most infectious form of the virus. Apolipoprotein E (apoE), a key LVP component, plays an essential role in HCV entry, assembly and egress. ApoE is also a cell host factor involved in lipoprotein homeostasis. Although the majority of apoE is associated with lipoproteins, a lipid-free (LF) form exists in blood. However, the role of LF-apoE in both lipid metabolism and HCV life cycle is poorly understood. DESIGN: In this study, using the cell culture-derived HCV model system in human hepatoma Huh7.5.1 cells and primary human hepatocytes (PHH), we investigated the effect of LF-apoE on the early steps of HCV life cycle and on the lipid metabolism of hepatic cells. RESULTS: A dose-dependent decrease in HCV replication was observed when Huh7.5.1 cells and PHH were treated with increasing amounts of LF-apoE. We showed that LF-apoE acts on HCV replication independently of previously described apoE receptors. We observed that LF-apoE induced a marked hepatic cholesterol efflux via the ATP-binding cassette subfamily G member 1 (ABCG1) protein that in turn inhibits HCV replication. LF-apoE also increases both apolipoprotein AI and high-density lipoprotein production. CONCLUSIONS: Our findings highlight a new mechanism in lipid metabolism regulation and interaction of the lipid metabolism with the HCV life cycle, which may be important for viral pathogenesis and might also be explored for antiviral therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Apolipoproteins E/metabolism , Apolipoproteins E/pharmacology , Cholesterol/metabolism , Hepacivirus/physiology , Virus Replication/drug effects , Apolipoprotein A-I/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Hepacivirus/growth & development , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Life Cycle Stages/drug effects , Lipoproteins, HDL/biosynthesis , Membrane Microdomains , Virus Internalization
3.
Gastroenterology ; 150(1): 206-217.e4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26404951

ABSTRACT

BACKGROUND & AIMS: Efforts to develop an effective vaccine against hepatitis C virus (HCV) have been hindered by the propensity of the virus to evade host immune responses. HCV particles in serum and in cell culture associate with lipoproteins, which contribute to viral entry. Lipoprotein association has also been proposed to mediate viral evasion of the humoral immune response, though the mechanisms are poorly defined. METHODS: We used small interfering RNAs to reduce levels of apolipoprotein E (apoE) in cell culture-derived HCV-producing Huh7.5-derived hepatoma cells and confirmed its depletion by immunoblot analyses of purified viral particles. Before infection of naïve hepatoma cells, we exposed cell culture-derived HCV strains of different genotypes, subtypes, and variants to serum and polyclonal and monoclonal antibodies isolated from patients with chronic HCV infection. We analyzed the interaction of apoE with viral envelope glycoprotein E2 and HCV virions by immunoprecipitation. RESULTS: Through loss-of-function studies on patient-derived HCV variants of several genotypes and subtypes, we found that the HCV particle apoE allows the virus to avoid neutralization by patient-derived antibodies. Functional studies with human monoclonal antiviral antibodies showed that conformational epitopes of envelope glycoprotein E2 domains B and C were exposed after depletion of apoE. The level and conformation of virion-associated apoE affected the ability of the virus to escape neutralization by antibodies. CONCLUSIONS: In cell-infection studies, we found that HCV-associated apoE helps the virus avoid neutralization by antibodies against HCV isolated from chronically infected patients. This method of immune evasion poses a challenge for the development of HCV vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Apolipoproteins E/metabolism , Hepacivirus/immunology , Hepatitis C Antibodies/immunology , Cells, Cultured , Hepacivirus/genetics , Hepatitis C/blood , Hepatocytes/immunology , Humans , Statistics, Nonparametric , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virus Internalization
4.
Sci Rep ; 5: 10693, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26024431

ABSTRACT

In plants and invertebrates RNA silencing is a major defense mechanism against virus infections. The first event in RNA silencing is dicing of long double stranded RNAs into small interfering RNAs (siRNAs). The Dicer proteins involved in this process are phylogenetically conserved and have the same domain organization. Accordingly, the production of viral derived siRNAs has also been observed in the mouse, but only in restricted cell types. To gain insight on this restriction, we compare the dicing activity of human Dicer and fly Dicer-2 in the context of Sindbis virus (SINV) infection. Expression of human Dicer in flies inefficiently rescues the production of viral siRNAs but confers some protection against SINV. Conversely, expression of Dicer-2 in human cells allows the production of viral 21 nt small RNAs. However, this does not confer resistance to viral infection, but on the contrary results in stronger accumulation of viral RNA. We further show that Dicer-2 expression in human cells perturbs interferon (IFN) signaling pathways and antagonizes protein kinase R (PKR)-mediated antiviral immunity. Overall, our data suggest that a functional incompatibility between the Dicer and IFN pathways explains the predominance of the IFN response in mammalian somatic cells.


Subject(s)
Alphavirus Infections/metabolism , Alphavirus Infections/virology , Ribonuclease III/metabolism , Sindbis Virus/genetics , Alphavirus Infections/genetics , Animals , Animals, Genetically Modified , Cell Line , Drosophila , Gene Expression , Gene Silencing , High-Throughput Nucleotide Sequencing , Humans , Interferons/metabolism , RNA Interference , RNA, Small Untranslated/genetics , Ribonuclease III/genetics , Signal Transduction
5.
PLoS One ; 9(4): e95550, 2014.
Article in English | MEDLINE | ID: mdl-24751902

ABSTRACT

Hepatitis C virus (HCV) is a major cause of liver disease worldwide and HCV infection represents a major health problem. HCV associates with host lipoproteins forming host/viral hybrid complexes termed lipoviral particles. Apolipoprotein E (apoE) is a lipoprotein component that interacts with heparan sulfate proteoglycans (HSPG) to mediate hepatic lipoprotein uptake, and may likewise mediate HCV entry. We sought to define the functional regions of apoE with an aim to identify critical apoE binding partners involved in HCV infection. Using adenoviral vectors and siRNA to modulate apoE expression we show a direct correlation of apoE expression and HCV infectivity, whereas no correlation exists with viral protein expression. Mutating the HSPG binding domain (HSPG-BD) of apoE revealed key residues that are critical for mediating HCV infection. Furthermore, a novel synthetic peptide that mimics apoE's HSPG-BD directly and competitively inhibits HCV infection. Genetic knockdown of the HSPG proteins syndecan (SDC) 1 and 4 revealed that SDC4 principally mediates HCV entry. Our data demonstrate that HCV uses apoE-SDC4 interactions to enter hepatoma cells and establish infection. Targeting apoE-SDC interactions could be an alternative strategy for blocking HCV entry, a critical step in maintaining chronic HCV infection.


Subject(s)
Apolipoproteins E/metabolism , Hepacivirus/physiology , Lipids/chemistry , Syndecan-4/metabolism , Virus Internalization , Amino Acid Sequence , Apolipoproteins E/chemistry , Cell Line , Heparan Sulfate Proteoglycans/metabolism , Hepatitis C/metabolism , Hepatitis C/pathology , Hepatitis C/virology , Humans , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary
6.
Viruses ; 5(5): 1292-324, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23698400

ABSTRACT

Hepatitis C virus (HCV) is a leading cause of chronic liver disease, including chronic hepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Hepatitis C infection associates with lipid and lipoprotein metabolism disorders such as hepatic steatosis, hypobetalipoproteinemia, and hypocholesterolemia. Furthermore, virus production is dependent on hepatic very-low-density lipoprotein (VLDL) assembly, and circulating virions are physically associated with lipoproteins in complexes termed lipoviral particles. Evidence has indicated several functional roles for the formation of these complexes, including co-opting of lipoprotein receptors for attachment and entry, concealing epitopes to facilitate immune escape, and hijacking host factors for HCV maturation and secretion. Here, we review the evidence surrounding pathogenesis of the hepatitis C infection regarding lipoprotein engagement, cholesterol and triglyceride regulation, and the molecular mechanisms underlying these effects.


Subject(s)
Cholesterol/metabolism , Hepacivirus/physiology , Hepatitis C/virology , Lipoproteins/metabolism , Virus Assembly , Virus Internalization , Virus Release , Hepacivirus/metabolism , Hepacivirus/pathogenicity , Hepatitis C/pathology , Humans , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...