Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Emerg Microbes Infect ; 13(1): 2348508, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38661085

RESUMO

The Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne bunyavirus that causes high mortality in humans. This enveloped virus harbors two surface glycoproteins (GP), Gn and Gc, that are released by processing of a glycoprotein precursor complex whose maturation takes place in the ER and is completed through the secretion pathway. Here, we characterized the trafficking network exploited by CCHFV GPs during viral assembly, envelopment, and/or egress. We identified membrane trafficking motifs in the cytoplasmic domains (CD) of CCHFV GPs and addressed how they impact these late stages of the viral life cycle using infection and biochemical assays, and confocal microscopy in virus-producing cells. We found that several of the identified CD motifs modulate GP transport through the retrograde trafficking network, impacting envelopment and secretion of infectious particles. Finally, we identified PACS-2 as a crucial host factor contributing to CCHFV GPs trafficking required for assembly and release of viral particles.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Transporte Proteico , Montagem de Vírus , Humanos , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Animais , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Domínios Proteicos , Motivos de Aminoácidos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Chlorocebus aethiops , Células HEK293 , Células Vero
2.
mBio ; 13(2): e0292321, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35258330

RESUMO

Nup98, an essential component of the nuclear pore that also participates in annulate lamella pore structures localized in the cytosol, is involved in hepatitis C virus (HCV) assembly. Here, we combined confocal microscopy and biochemical assays to study the interplay between Nup98, core (i.e., the HCV capsid protein), and viral genomes. Our results show that in HCV-infected cells, core protein is necessary and sufficient to induce relocalization of Nup98 from annulate lamellae to lipid droplet-apposed areas in which core/NS5A and HCV genomic RNA [(+)RNA] are clustered to promote viral assembly. Furthermore, we found that Nup98 interacts with HCV RNA and that upon Nup98 downregulation, the viral (+)RNA genome was specifically excluded from areas that contain active translating ribosomes and the core and NS5A proteins. Altogether, these results indicate that Nup98 is recruited by HCV core from annulate lamellae to viral assembly sites to locally increase the concentration of (+)RNA genome, which may favor its encapsidation into nascent virions. IMPORTANCE Nup98 is an essential component of the nuclear pore that also participates in annulate lamella pore structures localized in the cytosol. Nup98 is involved in HCV assembly, though its role remains elusive. Here, we show that Nup98 is retrieved from annulate lamellae during HCV infection. We demonstrate that Nup98 interacts with viral genome within infected cells and that these interactions are essential to maintain viral (+)RNAs in subcellular regions promoting viral replication, assembly, and translation. Importantly, we also show that HCV core nucleocapsid protein is the viral component responsible for the retrieval of Nup98 protein from annulate lamellae, hence allowing an enrichment of Nup98 complexed with viral (+)RNAs in core protein-containing areas. Altogether, our results indicate that Nup98 is recruited from annulate lamellae to viral assembly sites by HCV core protein to promote viral assembly, which highlights a novel virus-induced subversion mechanism of nuclear pore complex components.


Assuntos
Hepatite C , Proteínas do Core Viral , Hepacivirus/genética , Humanos , Complexo de Proteínas Formadoras de Poros Nucleares/genética , RNA Viral/genética , RNA Viral/metabolismo , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo , Montagem de Vírus/fisiologia
3.
Traffic ; 23(1): 63-80, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34729868

RESUMO

Lipid droplets (LDs) are involved in viral infections, but exactly how remains unclear. Here, we study the hepatitis C virus (HCV) whose core capsid protein binds to LDs but is also involved in the assembly of virions at the endoplasmic reticulum (ER) bilayer. We found that the amphipathic helix-containing domain of core, D2, senses triglycerides (TGs) rather than LDs per se. In the absence of LDs, D2 can bind to the ER membrane but only if TG molecules are present in the bilayer. Accordingly, the pharmacological inhibition of the diacylglycerol O-acyltransferase enzymes, mediating TG synthesis in the ER, inhibits D2 association with the bilayer. We found that TG molecules enable D2 to fold into alpha helices. Sequence analysis reveals that D2 resembles the apoE lipid-binding region. Our data support that TG in LDs promotes the folding of core, which subsequently relocalizes to contiguous ER regions. During this motion, core may carry TG molecules to these regions where HCV lipoviroparticles likely assemble. Consistent with this model, the inhibition of Arf1/COPI, which decreases LD surface accessibility to proteins and ER-LD material exchange, severely impedes the assembly of virions. Altogether, our data uncover a critical function of TG in the folding of core and HCV replication and reveals, more broadly, how TG accumulation in the ER may provoke the binding of soluble amphipathic helix-containing proteins to the ER bilayer.


Assuntos
Retículo Endoplasmático , Hepatite C , Retículo Endoplasmático/metabolismo , Hepacivirus/fisiologia , Hepatite C/metabolismo , Humanos , Gotículas Lipídicas/metabolismo , Triglicerídeos/metabolismo , Proteínas do Core Viral/metabolismo
4.
Elife ; 102021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34190687

RESUMO

Cell entry of enveloped viruses relies on the fusion between the viral and plasma or endosomal membranes, through a mechanism that is triggered by a cellular signal. Here we used a combination of computational and experimental approaches to unravel the main determinants of hepatitis B virus (HBV) membrane fusion process. We discovered that ERp57 is a host factor critically involved in triggering HBV fusion and infection. Then, through modeling approaches, we uncovered a putative allosteric cross-strand disulfide (CSD) bond in the HBV S glycoprotein and we demonstrate that its stabilization could prevent membrane fusion. Finally, we identified and characterized a potential fusion peptide in the preS1 domain of the HBV L glycoprotein. These results underscore a membrane fusion mechanism that could be triggered by ERp57, allowing a thiol/disulfide exchange reaction to occur and regulate isomerization of a critical CSD, which ultimately leads to the exposition of the fusion peptide.


Assuntos
Antígenos de Superfície da Hepatite B/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Precursores de Proteínas/metabolismo , Proteínas do Envelope Viral/metabolismo , Ligação Viral , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Regulação Viral da Expressão Gênica , Vírus da Hepatite B , Hepatócitos , Humanos , Masculino , Fusão de Membrana , Camundongos , Isomerases de Dissulfetos de Proteínas/genética , Proteínas do Envelope Viral/genética
5.
J Biol Chem ; 296: 100111, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33229438

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a ß-coronavirus, is the causative agent of the COVID-19 pandemic. Like for other coronaviruses, its particles are composed of four structural proteins: spike (S), envelope (E), membrane (M), and nucleoprotein (N) proteins. The involvement of each of these proteins and their interactions are critical for assembly and production of ß-coronavirus particles. Here, we sought to characterize the interplay of SARS-CoV-2 structural proteins during the viral assembly process. By combining biochemical and imaging assays in infected versus transfected cells, we show that E and M regulate intracellular trafficking of S as well as its intracellular processing. Indeed, the imaging data reveal that S is relocalized at endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) or Golgi compartments upon coexpression of E or M, as observed in SARS-CoV-2-infected cells, which prevents syncytia formation. We show that a C-terminal retrieval motif in the cytoplasmic tail of S is required for its M-mediated retention in the ERGIC, whereas E induces S retention by modulating the cell secretory pathway. We also highlight that E and M induce a specific maturation of N-glycosylation of S, independently of the regulation of its localization, with a profile that is observed both in infected cells and in purified viral particles. Finally, we show that E, M, and N are required for optimal production of virus-like-particles. Altogether, these results highlight how E and M proteins may influence the properties of S proteins and promote the assembly of SARS-CoV-2 viral particles.


Assuntos
Proteínas do Envelope de Coronavírus/genética , Proteínas do Nucleocapsídeo/genética , SARS-CoV-2/crescimento & desenvolvimento , Glicoproteína da Espícula de Coronavírus/genética , Proteínas da Matriz Viral/genética , Vírion/crescimento & desenvolvimento , Montagem de Vírus/fisiologia , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteínas do Envelope de Coronavírus/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Retículo Endoplasmático/virologia , Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Complexo de Golgi/virologia , Células HEK293 , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas do Nucleocapsídeo/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Proteínas da Matriz Viral/metabolismo , Vírion/genética , Vírion/metabolismo , Internalização do Vírus , Liberação de Vírus/fisiologia
6.
Nat Commun ; 10(1): 2098, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31068585

RESUMO

Hepatitis D virus (HDV) doesn't encode envelope proteins for packaging of its ribonucleoprotein (RNP) and typically relies on the surface glycoproteins (GPs) from hepatitis B virus (HBV) for virion assembly, envelopment and cellular transmission. HDV RNA genome can efficiently replicate in different tissues and species, raising the possibility that it evolved, and/or is still able to transmit, independently of HBV. Here we show that alternative, HBV-unrelated viruses can act as helper viruses for HDV. In vitro, envelope GPs from several virus genera, including vesiculovirus, flavivirus and hepacivirus, can package HDV RNPs, allowing efficient egress of HDV particles in the extracellular milieu of co-infected cells and subsequent entry into cells expressing the relevant receptors. Furthermore, HCV can propagate HDV infection in the liver of co-infected humanized mice for several months. Further work is necessary to evaluate whether HDV is currently transmitted by HBV-unrelated viruses in humans.


Assuntos
Coinfecção/transmissão , Hepatite D/transmissão , Vírus Delta da Hepatite/fisiologia , Montagem de Vírus , Animais , Linhagem Celular Tumoral , Coinfecção/virologia , Flavivirus/metabolismo , Hepacivirus/metabolismo , Hepacivirus/patogenicidade , Hepatite D/virologia , Vírus Delta da Hepatite/isolamento & purificação , Vírus Delta da Hepatite/patogenicidade , Hepatócitos/transplante , Hepatócitos/virologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Cultura Primária de Células , RNA Viral/isolamento & purificação , Ribonucleoproteínas/metabolismo , Vesiculovirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
7.
PLoS Pathog ; 13(12): e1006774, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29253880

RESUMO

Viroporins are small transmembrane proteins with ion channel activities modulating properties of intracellular membranes that have diverse proviral functions. Hepatitis C virus (HCV) encodes a viroporin, p7, acting during assembly, envelopment and secretion of viral particles (VP). HCV p7 is released from the viral polyprotein through cleavage at E2-p7 and p7-NS2 junctions by signal peptidase, but also exists as an E2p7 precursor, of poorly defined properties. Here, we found that ectopic p7 expression in HCVcc-infected cells reduced secretion of particle-associated E2 glycoproteins. Using biochemical assays, we show that p7 dose-dependently slows down the ER-to-Golgi traffic, leading to intracellular retention of E2, which suggested that timely E2p7 cleavage and p7 liberation are critical events to control E2 levels. By studying HCV mutants with accelerated E2p7 processing, we demonstrate that E2p7 cleavage controls E2 intracellular expression and secretion levels of nucleocapsid-free subviral particles and infectious virions. In addition, our imaging data reveal that, following p7 liberation, the amino-terminus of p7 is exposed towards the cytosol and coordinates the encounter between NS5A and NS2-based assembly sites loaded with E1E2 glycoproteins, which subsequently leads to nucleocapsid envelopment. We identify punctual mutants at p7 membrane interface that, by abrogating NS2/NS5A interaction, are defective for transmission of infectivity owing to decreased secretion of core and RNA and to increased secretion of non/partially-enveloped particles. Altogether, our results indicate that the retarded E2p7 precursor cleavage is essential to regulate the intracellular and secreted levels of E2 through p7-mediated modulation of the cell secretory pathway and to unmask critical novel assembly functions located at p7 amino-terminus.


Assuntos
Hepacivirus/fisiologia , Hepacivirus/patogenicidade , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Células HEK293 , Hepacivirus/genética , Hepatite C/etiologia , Hepatite C/virologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Modelos Biológicos , Mutação , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/fisiologia , Proteínas Virais/química , Proteínas Virais/genética , Virulência/genética , Virulência/fisiologia , Montagem de Vírus/genética , Montagem de Vírus/fisiologia
8.
J Virol ; 91(6)2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28053108

RESUMO

The assembly of hepatitis C virus (HCV), a complicated process in which many viral and cellular factors are involved, has not been thoroughly deciphered. NS3 is a multifunctional protein that contains an N-terminal amphipathic α helix (designated helix α0), which is crucial for the membrane association and stability of NS3 protein, followed by a serine protease domain and a C-terminal helicase/NTPase domain. NS3 participates in HCV assembly likely through its C-terminal helicase domain, in which all reported adaptive mutations enhancing virion assembly reside. In this study, we determined that the N-terminal helix α0 of NS3 may contribute to HCV assembly. We identified a single mutation from methionine to threonine at amino acid position 21 (M21T) in helix α0, which significantly promoted viral production while having no apparent effect on the membrane association and protease activity of NS3. Subsequent analyses demonstrated that the M21T mutation did not affect HCV genome replication but rather promoted virion assembly. Further study revealed a shift in the subcellular localization of core protein from lipid droplets (LD) to the endoplasmic reticulum (ER). Finally, we showed that the M21T mutation increased the colocalization of core proteins and viral envelope proteins, leading to a more efficient envelopment of viral nucleocapsids. Collectively, the results of our study revealed a new function of NS3 helix α0 and aid understanding of the role of NS3 in HCV virion morphogenesis.IMPORTANCE HCV NS3 protein possesses the protease activity in its N-terminal domain and the helicase activity in its C-terminal domain. The role of NS3 in virus assembly has been mainly attributed to its helicase domain, because all adaptive mutations enhancing progeny virus production are found to be within this domain. Our study identified, for the first time to our knowledge, an adaptive mutation within the N-terminal helix α0 domain of NS3 that significantly enhanced virus assembly while having no effect on viral genome replication. The mechanistic studies suggested that this mutation promoted the relocation of core proteins from LD to the ER, leading to a more efficient envelopment of viral nucleocapsids. Our results revealed a possible new function of helix α0 in the HCV life cycle and provided new clues to understanding the molecular mechanisms for the action of NS3 in HCV assembly.


Assuntos
Retículo Endoplasmático/virologia , Hepacivirus/fisiologia , Mutação Puntual , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus , Liberação de Vírus , Linhagem Celular , Hepacivirus/genética , Hepatócitos/virologia , Humanos , Metionina/genética , Metionina/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Treonina/genética , Treonina/metabolismo , Proteínas não Estruturais Virais/genética
9.
Gastroenterology ; 152(4): 895-907.e14, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27932311

RESUMO

BACKGROUND & AIMS: Daclatasvir is a direct-acting antiviral agent and potent inhibitor of NS5A, which is involved in replication of the hepatitis C virus (HCV) genome, presumably via membranous web shaping, and assembly of new virions, likely via transfer of the HCV RNA genome to viral particle assembly sites. Daclatasvir inhibits the formation of new membranous web structures and, ultimately, of replication complex vesicles, but also inhibits an early assembly step. We investigated the relationship between daclatasvir-induced clustering of HCV proteins, intracellular localization of viral RNAs, and inhibition of viral particle assembly. METHODS: Cell-culture-derived HCV particles were produced from Huh7.5 hepatocarcinoma cells in presence of daclatasvir for short time periods. Infectivity and production of physical particles were quantified and producer cells were subjected to subcellular fractionation. Intracellular colocalization between core, E2, NS5A, NS4B proteins, and viral RNAs was quantitatively analyzed by confocal microscopy and by structured illumination microscopy. RESULTS: Short exposure of HCV-infected cells to daclatasvir reduced viral assembly and induced clustering of structural proteins with non-structural HCV proteins, including core, E2, NS4B, and NS5A. These clustered structures appeared to be inactive assembly platforms, likely owing to loss of functional connection with replication complexes. Daclatasvir greatly reduced delivery of viral genomes to these core clusters without altering HCV RNA colocalization with NS5A. In contrast, daclatasvir neither induced clustered structures nor inhibited HCV assembly in cells infected with a daclatasvir-resistant mutant (NS5A-Y93H), indicating that daclatasvir targets a mutual, specific function of NS5A inhibiting both processes. CONCLUSIONS: In addition to inhibiting replication complex biogenesis, daclatasvir prevents viral assembly by blocking transfer of the viral genome to assembly sites. This leads to clustering of HCV proteins because viral particles and replication complex vesicles cannot form or egress. This dual mode of action of daclatasvir could explain its efficacy in blocking HCV replication in cultured cells and in treatment of patients with HCV infection.


Assuntos
Antivirais/farmacologia , Genoma Viral , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Imidazóis/farmacologia , Transporte de RNA/efeitos dos fármacos , RNA Viral/metabolismo , Carbamatos , Linhagem Celular Tumoral , Hepacivirus/fisiologia , Humanos , Transporte Proteico/efeitos dos fármacos , Pirrolidinas , Valina/análogos & derivados , Proteínas do Core Viral/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos
10.
J Biol Chem ; 282(44): 32357-69, 2007 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17761674

RESUMO

Cell entry of hepatitis C virus (HCV) is strikingly linked to lipoproteins and their receptors. Particularly, high density lipoprotein (HDL) enhances infectivity of HCV by involving the lipid-transfer function of the scavenger receptor BI, a receptor for both HDL and HCV. Here, we demonstrate that apoC-I, an exchangeable apolipoprotein that predominantly resides in HDL, specifically enhances HCVcc and HCVpp infectivity and increases the fusion rates between viral and target membranes via a direct interaction with the HCV surface. We identify the hypervariable region 1, located at the N terminus of the HCV E2 glycoprotein, as an essential viral component that modulates apoC-I-mediated enhancement of HCV fusion properties. The affinity of apoC-I for the HCV membrane may predispose it for fusion with a target membrane via alterations of its outer phospholipid layer. Conversely, we found that excess apoC-I provided as lipoprotein-free protein induces the disruption of the HCV membrane and subsequent loss of infectivity. Furthermore, our data indicate that HDL neither interacts nor spontaneously exchanges apoC-I with HCV virions. Because apoC-I is not present in serum as a lipoprotein-free form, our results suggest that HDL-embedded apoC-I could be released from the lipoprotein particle through a fine-tuned mechanism regulated via a triple interplay between hypervariable region 1, HDL, and scavenger receptor BI, resulting in optimal apoC-I recruitment on the viral membrane. These results provide the first description of a host serum factor helping the fusion process of an enveloped virus.


Assuntos
Apolipoproteína C-I/metabolismo , Hepacivirus/fisiologia , Internalização do Vírus , Linhagem Celular Tumoral , Humanos , Lipoproteínas HDL/metabolismo , Receptores Depuradores Classe B/metabolismo , Proteínas Virais/metabolismo
11.
Traffic ; 8(7): 835-47, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17547695

RESUMO

Retroviral core proteins, Gag and envelope (Env) glycoproteins are expressed from distinct cellular areas and therefore need to encounter to assemble infectious particles. The intrinsic cell localisation properties of either viral component or their capacity to mutually interact determines the assembly of infectious particles. Here, we address how Env determinants and cellular sorting proteins allow the Env derived from gamma retroviruses, murine leukemia virus (MLV) and RD114, to travel to or from late endosomes (LE), which may represent the Env assembly site of retroviruses in some cells. The individual expression of MLV Env resulted in its accumulation in LE in contrast to RD114 Env that required the presence of gamma retroviral Gag proteins. To discriminate between intrinsic intracellular Env localisation and gamma retroviral Gag/Env interactions in influencing Env viral incorporation, we studied Env assembly on heterologous lentiviral particles on which they are passively recruited. We found that an acidic cluster present at the C-terminus of the RD114 Env cytoplasmic tail determines its sub-cellular localisation and retrograde transport. Mutation of this motif induced late endosomal concentration of the RD114 Env, correlating with increased viral incorporation and infectivity. Reciprocally, the reinforcement of a poorly functional acidic motif in the MLV Env resulted in a marked decrease of its late endosomal localisation, leading to weakly infectious lentiviral particles with low Env densities. Finally, through upregulation versus downregulation of its cellular expression, we show that phosphofurin acidic-cluster-sorting protein 1 (PACS-1) controls the function of the RD114 Env acidic cluster, assigning to this cellular effector a crucial role in modulation of Env assembly of some retroviruses.


Assuntos
Citoplasma/metabolismo , Retroviridae/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Regulação para Baixo , Endossomos/metabolismo , Glicoproteínas/metabolismo , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas de Transporte Vesicular/metabolismo
12.
J Virol ; 80(21): 10579-90, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16928753

RESUMO

Cellular binding and entry of hepatitis C virus (HCV) are the first steps of viral infection and represent a major target for antiviral antibodies and novel therapeutic strategies. We have recently demonstrated that heparan sulfate (HS) plays a key role in the binding of HCV envelope glycoprotein E2 to target cells (Barth et al., J. Biol. Chem. 278:41003-41012, 2003). In this study, we characterized the HCV-HS interaction and analyzed its inhibition by antiviral host immune responses. Using recombinant envelope glycoproteins, virus-like particles, and HCV pseudoparticles as model systems for the early steps of viral infection, we mapped viral and cellular determinants of HCV-HS interaction. HCV-HS binding required a specific HS structure that included N-sulfo groups and a minimum of 10 to 14 saccharide subunits. HCV envelope binding to HS was mediated by four viral epitopes overlapping the E2 hypervariable region 1 and E2-CD81 binding domains. In functional studies using HCV pseudoparticles, we demonstrate that HCV binding and entry are specifically inhibited by highly sulfated HS. Finally, HCV-HS binding was markedly inhibited by antiviral antibodies derived from HCV-infected individuals. In conclusion, our results demonstrate that binding of the viral envelope to a specific HS configuration represents an important step for the initiation of viral infection and is a target of antiviral host immune responses in vivo. Mapping of viral and cellular determinants of HCV-HS interaction sets the stage for the development of novel HS-based antiviral strategies targeting viral attachment and entry.


Assuntos
Hepacivirus/fisiologia , Hepacivirus/patogenicidade , Heparitina Sulfato/metabolismo , Proteínas do Envelope Viral/fisiologia , Animais , Anticorpos Antivirais , Antígenos Virais , Sítios de Ligação , Linhagem Celular , Mapeamento de Epitopos , Células HeLa , Hepacivirus/genética , Hepacivirus/imunologia , Heparitina Sulfato/química , Hepatite C Crônica/etiologia , Hepatite C Crônica/imunologia , Hepatite C Crônica/virologia , Humanos , Camundongos , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Virulência
13.
Mol Ther ; 14(5): 735-44, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16784893

RESUMO

We report here a targeting method that exploits the expression pattern of cell surface proteases to induce gene delivery to specific tissues. We describe retroviral vectors harboring modified surface glycoproteins derived from an avian influenza virus hemagglutinin (HA) for which the cell entry properties, dependent on HA cleavage by producer cells, were conditionally blocked at a postbinding step by insertion of matrix metalloproteinase (MMP) substrates. We demonstrate that such vectors induce gene transfer, both in vitro and in mice harboring human tumor xenografts, only through contact with target cells expressing MMPs that cleave the substrate introduced into the recombinant HA. This selective gene transfer in MMP-rich cells was specifically inhibited by 1,10-phenanthroline, a broad-range MMP inhibitor. Importantly, such MMP-activatable vectors selectively transduced MMP-rich cells in heterogeneous populations containing MMP-rich and MMP-poor cells. These vectors will allow useful gene transfer applications into target cells exhibiting specific protease activities.


Assuntos
Vetores Genéticos/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Leucemia Murina/genética , Metaloendopeptidases/metabolismo , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Ativação Enzimática , Fator Xa/química , Fator Xa/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Metaloproteinases da Matriz/química , Metaloproteinases da Matriz/metabolismo , Dados de Sequência Molecular , Neoplasias/enzimologia , Neoplasias/genética , Ligação Proteica , Especificidade por Substrato
14.
J Biol Chem ; 281(14): 9200-4, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16418166

RESUMO

The capacity of the surface glycoproteins of enveloped viruses to mediate virus/cell binding and membrane fusion requires a proper thiol/disulfide balance. Chemical manipulation of their redox state using reducing agents or free sulfhydryl reagents affects virus/cell interaction. Conversely, natural thiol/disulfide rearrangements often occur during the cell interaction to trigger fusogenicity, hence the virus entry. We examined the relationship between the redox state of the 20 cysteine residues of the SARS-CoV (severe acute respiratory syndrome coronavirus) Spike glycoprotein S1 subdomain and its functional properties. Mature S1 exhibited approximately 4 unpaired cysteines, and chemically reduced S1 displaying up to approximately 6 additional unpaired cysteines still bound ACE2 and enabled fusion. In addition, virus/cell membrane fusion occurred in the presence of sulfhydryl-blocking reagents and oxidoreductase inhibitors. Thus, in contrast to various viruses including HIV (human immunodeficiency virus) examined in parallel, the functions of the SARS-CoV Spike glycoprotein exhibit a significant and surprising independence of redox state, which may contribute to the wide host range of the virus. These data suggest clues for molecularly engineering vaccine immunogens.


Assuntos
Antígenos HIV/química , Glicoproteínas de Membrana/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Proteínas do Envelope Viral/química , Antígenos Virais , Técnicas de Cultura de Células , Cisteína/química , Células Epiteliais , Humanos , Fusão de Membrana/fisiologia , Oxirredução , Dobramento de Proteína , Glicoproteína da Espícula de Coronavírus , Compostos de Sulfidrila/análise , Vacinas Virais
15.
J Virol ; 80(3): 1302-10, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16415007

RESUMO

The spike protein (S) of severe acute respiratory syndrome coronavirus (SARS-CoV) is responsible for receptor binding and membrane fusion. It contains a highly conserved transmembrane domain that consists of three parts: an N-terminal tryptophan-rich domain, a central domain, and a cysteine-rich C-terminal domain. The cytoplasmic tail of S has previously been shown to be required for assembly. Here, the roles of the transmembrane and cytoplasmic domains of S in the infectivity and membrane fusion activity of SARS-CoV have been studied. SARS-CoV S-pseudotyped retrovirus (SARSpp) was used to measure S-mediated infectivity. In addition, the cell-cell fusion activity of S was monitored by a Renilla luciferase-based cell-cell fusion assay. S(VSV-Cyt), an S chimera with a cytoplasmic tail derived from vesicular stomatitis virus G protein (VSV-G), and S(MHV-TMDCyt), an S chimera with the cytoplasmic and transmembrane domains of mouse hepatitis virus, displayed wild-type-like activity in both assays. S(VSV-TMDCyt), a chimera with the cytoplasmic and transmembrane domains of VSV-G, was impaired in the SARSpp and cell-cell fusion assays, showing 3 to 25% activity compared to the wild type, depending on the assay and the cells used. Examination of the oligomeric state of the chimeric S proteins in SARSpp revealed that S(VSV-TMDCyt) trimers were less stable than wild-type S trimers, possibly explaining the lowered fusogenicity and infectivity.


Assuntos
Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , DNA Viral/genética , Humanos , Luciferases de Renilla/genética , Fusão de Membrana/fisiologia , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Receptores Virais/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/genética
16.
Retrovirology ; 1: 29, 2004 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-15447786

RESUMO

BACKGROUND: The aim of this study was to evaluate gene therapy for AIDS based on the transduction of circulating lymphocytes with a retroviral vector giving low levels of constitutive macaque interferon beta production in macaques chronically infected with a pathogenic isolate of SIVmac251. RESULTS: Two groups of three animals infected for more than one year with a pathogenic primary isolate of SIVmac251 were included in this study. The macaques received three infusions of their own lymphocytes transduced ex vivo with the construct encoding macaque IFN-beta (MaIFN-beta or with a vector carrying a version of the MaIFN-beta gene with a deletion preventing translation of the mRNA. Cellular or plasma viremia increased transiently following injection in most cases, regardless of the retroviral construct used. Transduced cells were detected only transiently after each infusion, among the peripheral blood mononuclear cells of all the animals, with copy numbers of 10 to 1000 per 106 peripheral mononuclear cells. CONCLUSION: Long-term follow-up indicated that the transitory presence of such a small number of cells producing such small amounts of MaIFN-beta did not prevent animals from the progressive decrease in CD4+ cell count typical of infection with simian immunodeficiency virus. These results reveal potential pitfalls for future developments of gene therapy strategies of HIV infection.


Assuntos
Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Terapia Genética/métodos , Interferon beta/genética , Interferon beta/uso terapêutico , Transfusão de Linfócitos , Linfócitos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/imunologia , Animais , Sequência de Bases , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/imunologia , Primers do DNA , DNA Viral/genética , DNA Viral/isolamento & purificação , Vetores Genéticos , Interferon beta/administração & dosagem , Macaca fascicularis , Masculino , Regiões Promotoras Genéticas , RNA Viral/sangue , RNA Viral/genética , RNA Viral/isolamento & purificação , Retroviridae , Deleção de Sequência , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/genética , Carga Viral
17.
J Virol ; 76(19): 9673-85, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12208946

RESUMO

Envelope glycoproteins (Env) of retroviruses are trimers of SU (surface) and TM (transmembrane) heterodimers and are expressed on virions in fusion-competent forms that are likely to be metastable. Activation of the viral receptor-binding domain (RBD) via its interaction with a cell surface receptor is thought to initiate a cascade of events that lead to refolding of the Env glycoprotein into its stable fusion-active conformation. While the fusion-active conformation of the TM subunit has been described in detail for several retroviruses, little is known about the fusion-competent structure of the retroviral glycoproteins or the molecular events that mediate the transition between the two conformations. By characterizing Env chimeras between the ecotropic and amphotropic murine leukemia virus (MLV) SUs as well as a set of point mutants, we show that alterations of the conformation of the SU glycoprotein strongly elevate Env fusogenicity by disrupting the stability of the Env complex. Compensatory mutations that restored both Env stability and fusion control were also identified, allowing definition of interactions within the Env complex that maintain the stability of the native Env complex. We show that, in the receptor-unbound form, structural interactions between the N terminus of the viral RBD (NTR domain), the proline-rich region (PRR), and the distal part of the C-terminal domain of the SU subunit maintain a conformation of the glycoprotein that is fusion inhibitory. Additionally, we identified mutations that disrupt this fusion-inhibitory conformation and allow fusion activation in the absence of viral receptors, provided that receptor-activated RBD fragments are added in trans during infection. Other mutations were identified that allow fusion activation in the absence of receptors for both the viral glycoprotein and the trans-acting RBD. Finally, we found mutations of the SU that bypass in cis the requirement for the NTR domain in fusion activation. All these different mutations call for a critical role of the PRR in mediating conformational changes of the Env glycoprotein during fusion activation. Our results suggest a model of MLV Env fusion activation in which unlocking of the fusion-inhibitory conformation is initiated by receptor binding of the viral RBD, which, upon disruption of the PRR, allows the NTR domain to promote further events in Env fusion activation. This involves a second type of interaction, in cis or in trans, between the receptor-activated RBD and a median segment of the freed C-terminal domain.


Assuntos
Vírus da Leucemia Murina/química , Fusão de Membrana/fisiologia , Receptores Virais/fisiologia , Proteínas do Envelope Viral/química , Sítios de Ligação , Humanos , Conformação Proteica , Células Tumorais Cultivadas , Proteínas do Envelope Viral/fisiologia
18.
Blood ; 100(3): 823-32, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12130492

RESUMO

Generating lentiviral vectors pseudotyped with different viral glycoproteins (GPs) may modulate the physicochemical properties of the vectors, their interaction with the host immune system, and their host range. We have investigated the capacity of a panel of GPs of both retroviral (amphotropic murine leukemia virus [MLV-A]; gibbon ape leukemia virus [GALV]; RD114, feline endogenous virus) and nonretroviral (fowl plague virus [FPV]; Ebola virus [EboV]; vesicular stomatitis virus [VSV]; lymphocytic choriomeningitis virus [LCMV]) origins to pseudotype lentiviral vectors derived from simian immunodeficiency virus (SIVmac251). SIV vectors were efficiently pseudotyped with the FPV hemagglutinin, VSV-G, LCMV, and MLV-A GPs. In contrast, the GALV and RD114 GPs conferred much lower infectivity to the vectors. Capitalizing on the conservation of some structural features in the transmembrane domains and cytoplasmic tails of the incorporation-competent MLV-A GP and in RD114 and GALV GPs, we generated chimeric GPs encoding the extracellular and transmembrane domains of GALV or RD114 GPs fused to the cytoplasmic tail (designated TR) of MLV-A GP. Importantly, SIV-derived vectors pseudotyped with these GALV/TR and RD114/TR GP chimeras had significantly higher titers than vectors coated with the parental GPs. Additionally, RD114/TR-pseudotyped vectors were efficiently concentrated and were resistant to inactivation induced by the complement of both human and macaque sera, indicating that modified RD114 GP-pseudotyped lentiviral vectors may be of particular interest for in vivo gene transfer applications. Furthermore, as compared to vectors pseudotyped with other retroviral GPs or with VSV-G, RD114/TR-pseudotyped vectors showed augmented transduction of human and macaque primary blood lymphocytes and CD34+ cells.


Assuntos
Antígenos CD34/sangue , Células Sanguíneas/metabolismo , Vetores Genéticos/genética , Lentivirus/genética , Vírus da Imunodeficiência Símia/genética , Transdução Genética/métodos , Proteínas do Envelope Viral/genética , Animais , Estabilidade de Medicamentos , Vetores Genéticos/normas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Linfócitos/imunologia , Linfócitos/metabolismo , Macaca
19.
J Gen Virol ; 81(Pt 11): 2741-2750, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11038387

RESUMO

To test the in vivo anti-simian immunodeficiency virus (SIV) efficacy of interferon (IFN)-beta-engineered lymphocytes, peripheral blood lymphocytes harvested from two uninfected macaques were transduced with a retroviral vector carrying a constitutively expressed IFN-beta gene and reinfused, resulting in approximately 1 IFN-beta-transduced cell out of 1000 circulating cells. The gene-modified cells were well tolerated and could be detected for at least 74 days without causing any apparent side effects. These two animals together with three untreated control macaques were then infected with SIVmac251. The two IFN-beta-infused macaques are in good health, 478 days after infection, with a reduced plasma virus load and sustained numbers of CD4(+) and CD8(+) cells. Throughout the study, the proportion of IFN-beta-transduced cells has been maintained. Of the three control macaques, two were characterized by a high plasma virus load and a decrease in CD4(+) cells. One was moribund and was sacrificed 350 days after infection and the other now has fewer than 100 circulating CD4(+) cells/ml. Unexpectedly, the third control macaque, which, like the two IFN-beta-infused animals, had a low plasma virus load and a maintenance of CD4(+) and CD8(+) cell number, was characterized by a permanent level of serum IFN-beta, of unknown origin, already present before SIV infection. Although no definite conclusion can be made in view of the limited number of animals, these data indicate that further exploration is warranted of an IFN-beta-based anti-human immunodeficiency virus gene therapy.


Assuntos
Imunoterapia , Interferon beta/genética , Interferon beta/imunologia , Linfócitos/imunologia , Macaca/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia , Animais , Vetores Genéticos , Macaca/virologia , Retroviridae , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA