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










Publication year range
1.
Cell Mol Life Sci ; 79(8): 425, 2022 Jul 16.
Article in English | MEDLINE | ID: mdl-35841484

ABSTRACT

Positive single-strand RNA (+ RNA) viruses can remodel host cell membranes to induce a replication organelle (RO) isolating the replication of their genome from innate immunity mechanisms. Some of these viruses, including severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), induce double-membrane vesicles (DMVs) for this purpose. Viral non-structural proteins are essential for DMV biogenesis, but they cannot form without an original membrane from a host cell organelle and a significant supply of lipids. The endoplasmic reticulum (ER) and the initial mechanisms of autophagic processes have been shown to be essential for the biogenesis of SARS-CoV-2 DMVs. However, by analogy with other DMV-inducing viruses, it seems likely that the Golgi apparatus, mitochondria and lipid droplets are also involved. As for hepatitis C virus (HCV), pores crossing both membranes of SARS-CoV-2-induced DMVs have been identified. These pores presumably allow the supply of metabolites essential for viral replication within the DMV, together with the export of the newly synthesized viral RNA to form the genome of future virions. It remains unknown whether, as for HCV, DMVs with open pores can coexist with the fully sealed DMVs required for the storage of large amounts of viral RNA. Interestingly, recent studies have revealed many similarities in the mechanisms of DMV biogenesis and morphology between these two phylogenetically distant viruses. An understanding of the mechanisms of DMV formation and their role in the infectious cycle of SARS-CoV-2 may be essential for the development of new antiviral approaches against this pathogen or other coronaviruses that may emerge in the future.


Subject(s)
COVID-19 , Hepatitis C , Endoplasmic Reticulum/metabolism , Hepacivirus/genetics , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , SARS-CoV-2 , Viral Nonstructural Proteins/genetics , Virus Replication
2.
J Viral Hepat ; 29(9): 719-726, 2022 09.
Article in English | MEDLINE | ID: mdl-35633087

ABSTRACT

It was recently suggested that the composition of circulating hepatitis B subviral particles (SVPs) could be used to differentiate the various stages in chronic hepatitis B virus (HBV) infection, with significantly lower proportions of L and M proteins in inactive carriers than in individuals with chronic hepatitis. L protein is abundant in virions and filamentous SVPs but almost absent from spherical SVPs. We, therefore, performed a morphometric analysis of SVPs in these two groups of patients, by conducting a retrospective analysis on sera from 15 inactive carriers and 11 patients with chronic hepatitis infected with various HBV genotypes. Subviral particles were concentrated by centrifugation on a sucrose cushion, with monitoring by transmission electron microscopy. The percentage of filamentous SVPs and filament length for 100 SVPs was determined with a digital camera. The L protein PreS1 promoter was sequenced from viral genomes by the Sanger method. No marked differences were found between patients, some of whom had only spherical SVPs, whereas others had variable percentages of filamentous SVPs (up to 28%), of highly variable length. High filament percentages were not associated with a particular sequence of the L protein promoter, HBV genotype or even disease stage. High levels of circulating filamentous SVPs are probably more strongly related to individual host factors than to viral strain characteristics or disease stage.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Genotype , Hepatitis B Surface Antigens , Hepatitis B virus/genetics , Humans , Retrospective Studies
3.
Cell Microbiol ; 22(8): e13205, 2020 08.
Article in English | MEDLINE | ID: mdl-32216005

ABSTRACT

Hepatitis B virus (HBV) is a leading cause of cirrhosis and hepatocellular carcinoma worldwide, with 250 million individuals chronically infected. Many stages of the HBV infectious cycle have been elucidated, but the mechanisms of HBV entry remain poorly understood. The identification of the sodium taurocholate cotransporting polypeptide (NTCP) as an HBV receptor and the establishment of NTCP-overexpressing hepatoma cell lines susceptible to HBV infection opens up new possibilities for investigating these mechanisms. We used HepG2-NTCP cells, and various chemical inhibitors and RNA interference (RNAi) approaches to investigate the host cell factors involved in HBV entry. We found that HBV uptake into these cells was dependent on the actin cytoskeleton and did not involve macropinocytosis or caveolae-mediated endocytosis. Instead, entry occurred via the clathrin-mediated endocytosis pathway. HBV internalisation was inhibited by pitstop-2 treatment and RNA-mediated silencing (siRNA) of the clathrin heavy chain, adaptor protein AP-2 and dynamin-2. We were able to visualise HBV entry in clathrin-coated pits and vesicles by electron microscopy (EM) and cryo-EM with immunogold labelling. These data demonstrating that HBV uses a clathrin-mediated endocytosis pathway to enter HepG2-NTCP cells increase our understanding of the complete HBV life cycle.


Subject(s)
Clathrin/metabolism , Endocytosis , Hepatitis B virus/physiology , Virus Internalization , Clathrin/ultrastructure , Cryoelectron Microscopy , Hep G2 Cells , Hepatitis B virus/ultrastructure , Host Microbial Interactions , Humans , Microscopy, Electron , RNA Interference , Viral Envelope Proteins/metabolism
4.
Sci Rep ; 9(1): 16178, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31700077

ABSTRACT

Hepatitis B virus (HBV) production requires intricate interactions between the envelope and core proteins. Analyses of mutants of these proteins have made it possible to map regions involved in the formation and secretion of virions. Tests of binding between core and envelope peptides have also been performed in cell-free conditions, to study the interactions potentially underlying these mechanisms. We investigated the residues essential for core-envelope interaction in a cellular context in more detail, by transiently producing mutant or wild-type L, S, or core proteins separately or in combination, in Huh7 cells. The colocalization and interaction of these proteins were studied by confocal microscopy and co-immunoprecipitation, respectively. The L protein was shown to constitute a molecular platform for the recruitment of S and core proteins in a perinuclear environment. Several core amino acids were found to be essential for direct interaction with L, including residue Y132, known to be crucial for capsid formation, and residues L60, L95, K96 and I126. Our results confirm the key role of L in the tripartite core-S-L interaction and identify the residues involved in direct core-L interaction. This model may be valuable for studies of the potential of drugs to inhibit HBV core-envelope interaction.


Subject(s)
Capsid/metabolism , Hepatitis B virus/metabolism , Viral Core Proteins/metabolism , Viral Envelope Proteins/metabolism , Cell Line, Tumor , Hepatitis B virus/genetics , Humans , Viral Core Proteins/genetics , Viral Envelope Proteins/genetics
5.
Hepatology ; 57(4): 1303-13, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23150224

ABSTRACT

UNLABELLED: The development of a prophylactic vaccine against hepatitis C virus (HCV) has become an important medical priority, because 3-4 million new HCV infections are thought to occur each year worldwide. Hepatitis B virus (HBV) is another major human pathogen, but infections with this virus can be prevented with a safe, efficient vaccine, based on the remarkable ability of the envelope protein (S) of this virus to self-assemble into highly immunogenic subviral particles. Chimeric HBV-HCV envelope proteins in which the N-terminal transmembrane domain of S was replaced with the transmembrane domain of the HCV envelope proteins (E1 or E2) were efficiently coassembled with the wild-type HBV S protein into subviral particles. These chimeric particles presented the full-length E1 and E2 proteins from a genotype 1a virus in an appropriate conformation for formation of the E1-E2 heterodimer. Produced in stably transduced Chinese hamster ovary cells and used to immunize New Zealand rabbits, these particles induced a strong specific antibody (Ab) response against the HCV and HBV envelope proteins in immunized animals. Sera containing anti-E1 or anti-E2 Abs elicited by these particles neutralized infections with HCV pseudoparticles and cell-cultured viruses derived from different heterologous 1a, 1b, 2a, and 3 strains. Moreover, the anti-hepatitis B surface response induced by these chimeric particles was equivalent to the response induced by a commercial HBV vaccine. CONCLUSIONS: Our results provide support for approaches based on the development of bivalent HBV-HCV prophylactic vaccine candidates potentially able to prevent initial infection with either of these two hepatotropic viruses.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hepacivirus/immunology , Hepatitis B virus/immunology , Viral Envelope Proteins/immunology , Viral Hepatitis Vaccines/immunology , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Female , Hepacivirus/metabolism , Hepatitis B/prevention & control , Hepatitis B virus/metabolism , Hepatitis C/prevention & control , Immunity, Humoral/immunology , Protein Folding , Rabbits , Viral Envelope Proteins/metabolism , Viral Hepatitis Vaccines/therapeutic use
6.
Hepatology ; 54(4): 1135-48, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21674561

ABSTRACT

UNLABELLED: Understanding the immunological correlates associated with protective immunity following hepatitis C virus (HCV) reexposure is a prerequisite for the design of effective HCV vaccines and immunotherapeutics. In this study we performed a comprehensive analysis of innate and adaptive immunity following HCV reexposure of two chimpanzees that had previously recovered from HCV-JFH1 infection. One of the chimpanzees, CH10274, became protected from active viremia by repeated challenges with homologous HCV-JFH1 and developed neutralizing antibodies, but was later infected with high-level viremia by a heterologous challenge with the HCV H77 virus that persisted for more than 1 year. The other chimpanzee, CH10273, was protected from a similar, heterologous H77 challenge without any evidence of neutralizing antibodies. Peripheral HCV-specific T-cell responses were present in both chimpanzees after challenges and, interestingly, the overall magnitude of response was lower in uninfected CH10273, which, however, exhibited a more robust CD8+ T-cell response. CH10273 showed higher hepatic expression of CD8 and CD56 (natural killer) markers than CH10274 did shortly after inoculation with H77. The heightened T-cell response was associated with an enhanced hepatic production of interferons (both type I and II) and interferon-stimulated genes (ISGs) in CH10273. Therefore, protection or clearance of HCV reinfection upon heterologous rechallenge depends on the activation of both intrahepatic innate and cellular immune responses. Furthermore, our results suggest that serum neutralizing antibodies may contribute to early control of viral replication and spread after homologous HCV rechallenges but may not be sufficient for a long-term protective immunity. CONCLUSION: Our study shows that protective immunity against HCV reinfection is orchestrated by a complex network of innate and adaptive immune responses.


Subject(s)
Adaptive Immunity/physiology , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/prevention & control , Immunity, Innate/physiology , Adaptive Immunity/immunology , Animals , Disease Models, Animal , Hepatitis C Antibodies/blood , Hepatitis C Antibodies/immunology , Humans , Immunity, Innate/immunology , Pan troglodytes , Polymerase Chain Reaction , Sensitivity and Specificity , Viral Hepatitis Vaccines/administration & dosage , Viremia/immunology , Virus Replication/immunology
7.
Cell Microbiol ; 11(11): 1561-70, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19673892

ABSTRACT

After cell hijacking and intracellular amplification, non-lytic enveloped viruses are usually released from the infected cell by budding across internal membranes or through the plasma membrane. The enveloped human hepatitis B virus (HBV) is an example of virus using an intracellular compartment to form new virions. Four decades after its discovery, HBV is still the primary cause of death by cancer due to a viral infection worldwide. Despite numerous studies on HBV genome replication little is known about its morphogenesis process. In addition to viral neogenesis, the HBV envelope proteins have the capability without any other viral component to form empty subviral envelope particles (SVPs), which are secreted into the blood of infected patients. A better knowledge of this process may be critical for future antiviral strategies. Previous studies have speculated that the morphogenesis of HBV and its SVPs occur through the same mechanisms. However, recent data clearly suggest that two different processes, including constitutive Golgi pathway or cellular machinery that generates internal vesicles of multivesicular bodies (MVB), independently form these two viral entities.


Subject(s)
Hepatitis B virus/physiology , Virus Assembly , Virus Release , Cell Membrane/virology , Endoplasmic Reticulum/virology , Host-Pathogen Interactions , Humans , Models, Biological , Virion/ultrastructure , Virosomes/biosynthesis
8.
N Biotechnol ; 25(4): 226-34, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19356608

ABSTRACT

The hepatitis B virus (HBV) envelope protein (S) self-assembles into subviral particles used as commercial vaccines against hepatitis B. These particles are excellent carriers for foreign epitopes, which can be inserted into the external hydrophilic loop or at the N- or C-terminal end of the HBV S protein. We show here that the N-terminal transmembrane domain (TMD) of HBV S can be replaced by the TMDs of the hepatitis C virus (HCV) envelope proteins E1 and E2, to generate fusion proteins containing the entire HCV E1 or E2 sequence that are efficiently coassembled with the HBV S into particles. This demonstrates the remarkable tolerance of the HBV S protein to sequence substitutions conserving its subviral particle assembly properties. These findings may have implications for the design of new vaccine strategies based on the use of HBV subviral particles as carriers for various transmembrane proteins and produced using the same industrial procedures that are established for the HBV vaccine.


Subject(s)
Hepacivirus/metabolism , Hepatitis B virus/metabolism , Protein Engineering/methods , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virion/chemistry , Virion/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Design , Hepacivirus/genetics , Hepatitis B virus/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Viral Envelope Proteins/genetics , Viral Vaccines , Virion/genetics
9.
J Virol ; 81(8): 3842-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17267490

ABSTRACT

Hepatitis B virus (HBV) is unusual in that its surface proteins (small [S], medium, and large [L]) are not only incorporated into the virion envelope but they also bud into empty subviral particles in great excess over virions. The morphogenesis of these subviral envelope particles remains unclear, but the S protein is essential and sufficient for budding. We show here that, in contrast to the presumed model, the HBV subviral particle formed by the S protein self-assembles into branched filaments in the lumen of the endoplasmic reticulum (ER). These long filaments are then folded and bridged for packing into crystal-like structures, which are then transported by ER-derived vesicles to the ER-Golgi intermediate compartment (ERGIC). Within the ERGIC, they are unpacked and relaxed, and their size and shape probably limits further progression through the secretory pathway. Such progression requires their conversion into spherical particles, which occurred spontaneously during the purification of these filaments by affinity chromatography. Small branched filaments are also formed by the L protein in the ER lumen, but these filaments are not packed into transport vesicles. They are transported less efficiently to the ERGIC, potentially accounting for the retention of the L protein within cells. These findings shed light on an important step in the HBV infectious cycle, as the intracellular accumulation of HBV subviral filaments may be directly linked to viral pathogenesis.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B virus/ultrastructure , Animals , Cell Line , Cricetinae , Endoplasmic Reticulum/virology , Golgi Apparatus/virology , Hepatitis B Surface Antigens/metabolism , Immunohistochemistry , Microscopy, Confocal , Microscopy, Electron, Transmission , Morphogenesis , Protein Transport , Transport Vesicles/virology , Viral Envelope Proteins/metabolism , Virion/metabolism , Virion/ultrastructure
10.
Cell Microbiol ; 9(4): 1014-27, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17257269

ABSTRACT

Hepatitis C virus (HCV) core protein, expressed with a Semliki forest virus (SFV) replicon, self-assembles into HCV-like particles (HCV-LPs) at the endoplasmic reticulum (ER) membrane, providing an opportunity to study HCV particle morphogenesis by electron microscopy. Various mutated HCV core proteins with engineered internal deletions were expressed with this system, to identify core domains required or dispensable for HCV-LP assembly. The HCV core protein sequence was compared with its counterpart in GB virus B (GBV-B), the virus most closely related to HCV, to identify conserved domains. GBV-B and HCV display similar tropism for liver hepatocytes and their core proteins are organized similarly into three main domains (I, II and III), although GBV-B core is smaller and lacks approximately 35 amino acids (aa) in domain I. The deletion of short hydrophobic domains (aa 133-152 and 153-167 in HCV core) that appear highly conserved in domain II of both GBV-B and HCV core proteins resulted in loss of HCV core ER anchoring and self-assembly into HCV-LPs. The deletion of short domains found within domain I of HCV core protein but not in the corresponding domain of GBV-B core according to sequence alignment had contrasting effects. Amino acids 15-28 and 60-66 were shown to be dispensable for HCV-LP assembly and morphogenesis, whereas aa 88-106 were required for this process. The production of GBV-B core protein from a recombinant SFV vector was associated with specific ER ultrastructural changes, but did not lead to the morphogenesis of GBV-B-LPs, suggesting that different budding mechanisms occur in members of the Flaviviridae family.


Subject(s)
Endoplasmic Reticulum/metabolism , GB virus B/metabolism , Hepacivirus/metabolism , Viral Core Proteins/physiology , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , GB virus B/genetics , GB virus B/growth & development , Hepacivirus/genetics , Hepacivirus/growth & development , Hydrophobic and Hydrophilic Interactions , Microscopy, Confocal , Microscopy, Electron, Transmission , Molecular Sequence Data , Mutation , Sequence Deletion , Sequence Homology, Amino Acid , Viral Core Proteins/chemistry , Viral Core Proteins/genetics
11.
J Gen Virol ; 87(Pt 4): 855-860, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16528035

ABSTRACT

Hepatitis C virus (HCV) core protein, expressed with a Semliki Forest virus replicon, self-assembles into HCV-like particles (HCV-LP) at the endoplasmic reticulum (ER) membrane, providing an opportunity to study HCV assembly and morphogenesis by electron microscopy. This model was used to investigate whether the processing of the HCV core protein by the signal peptide peptidase (SPP) is required for the HCV-LP assembly. Several mutants were designed as there are conflicting reports concerning the cleavage of mutant proteins by SPP. Production of the only core mutant protein that escaped SPP processing led to the formation of multiple layers of electron-dense ER membrane, with no evidence of HCV-LP assembly. These data shed light on the HCV core residues involved in SPP cleavage and suggest that this cleavage is essential for HCV assembly.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Hepacivirus/metabolism , Viral Core Proteins/metabolism , Virion/metabolism , Animals , Cell Line , Cricetinae , Endoplasmic Reticulum/metabolism , Hepacivirus/genetics , Humans , Intracellular Membranes/metabolism , Mutation , Transfection , Viral Core Proteins/chemistry , Viral Core Proteins/genetics , Virus Assembly
SELECTION OF CITATIONS
SEARCH DETAIL
...