Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
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
2.
Antiviral Res ; 170: 104578, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31394119

RESUMO

Zika virus (ZIKV) infection is a serious public health concern due to its ability to induce neurological defects and its potential for rapid transmission at a global scale. However, no vaccine is currently available to prevent ZIKV infection. Here, we report the development of a yeast-derived subunit protein vaccine for ZIKV. The envelope protein domain III (EDIII) of ZIKV was produced as a secretory protein in the yeast Pichia pastoris. The yeast-derived EDIII could inhibit ZIKV infection in vitro in a dose-dependent manner, suggesting that it had acquired an appropriate conformation to bind to cellular receptors of ZIKV. Immunization with recombinant EDIII protein effectively induced antigen-specific binding antibodies and cellular immune responses. The resulting anti-EDIII sera could efficiently neutralize ZIKV representative strains from both Asian and African lineages. Passive transfer with the anti-EDIII neutralizing sera could confer protection against lethal ZIKV challenge in mice. Importantly, we found that purified anti-EDIII antibodies did not cross-react with closely related dengue virus (DENV) and therefore did not enhance DENV infection. Collectively, our results demonstrate that yeast-produced EDIII is a safe and effective ZIKV vaccine candidate.


Assuntos
Anticorpos Antivirais/sangue , Anticorpos Amplamente Neutralizantes/imunologia , Imunização Passiva , Pichia/genética , Vacinas Virais/genética , Infecção por Zika virus/prevenção & controle , Animais , Anticorpos Antivirais/imunologia , Anticorpos Amplamente Neutralizantes/sangue , Reações Cruzadas , Vírus da Dengue/imunologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Zika virus
3.
PLoS Pathog ; 14(3): e1006908, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29505618

RESUMO

Amino-acid coevolution can be referred to mutational compensatory patterns preserving the function of a protein. Viral envelope glycoproteins, which mediate entry of enveloped viruses into their host cells, are shaped by coevolution signals that confer to viruses the plasticity to evade neutralizing antibodies without altering viral entry mechanisms. The functions and structures of the two envelope glycoproteins of the Hepatitis C Virus (HCV), E1 and E2, are poorly described. Especially, how these two proteins mediate the HCV fusion process between the viral and the cell membrane remains elusive. Here, as a proof of concept, we aimed to take advantage of an original coevolution method recently developed to shed light on the HCV fusion mechanism. When first applied to the well-characterized Dengue Virus (DENV) envelope glycoproteins, coevolution analysis was able to predict important structural features and rearrangements of these viral protein complexes. When applied to HCV E1E2, computational coevolution analysis predicted that E1 and E2 refold interdependently during fusion through rearrangements of the E2 Back Layer (BL). Consistently, a soluble BL-derived polypeptide inhibited HCV infection of hepatoma cell lines, primary human hepatocytes and humanized liver mice. We showed that this polypeptide specifically inhibited HCV fusogenic rearrangements, hence supporting the critical role of this domain during HCV fusion. By combining coevolution analysis and in vitro assays, we also uncovered functionally-significant coevolving signals between E1 and E2 BL/Stem regions that govern HCV fusion, demonstrating the accuracy of our coevolution predictions. Altogether, our work shed light on important structural features of the HCV fusion mechanism and contributes to advance our functional understanding of this process. This study also provides an important proof of concept that coevolution can be employed to explore viral protein mediated-processes, and can guide the development of innovative translational strategies against challenging human-tropic viruses.


Assuntos
Evolução Molecular , Hepacivirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Hepatite C/metabolismo , Hepatite C/patologia , Hepatite C/virologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Células Tumorais Cultivadas , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Replicação Viral
4.
J Virol ; 90(2): 992-1008, 2016 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-26537674

RESUMO

UNLABELLED: Hepatitis C virus (HCV) productively infects hepatocytes. Virion surface glycoproteins E1 and E2 play a major role in this restricted cell tropism by mediating virus entry into particular cell types. However, several pieces of evidence have suggested the ability of patient-derived HCV particles to infect peripheral blood mononuclear cells. The viral determinants and mechanisms mediating such events remain poorly understood. Here, we aimed at isolating viral determinants of HCV entry into B lymphocytes. For this purpose, we constructed a library of full E1E2 sequences isolated from serum and B lymphocytes of four chronically infected patients. We observed a strong phylogenetic compartmentalization of E1E2 sequences isolated from B lymphocytes in one patient, indicating that E1E2 glycoproteins can represent important mediators of the strong segregation of two specialized populations in some patients. Most of the E1E2 envelope glycoproteins were functional and allowed transduction of hepatocyte cell lines using HCV-derived pseudoparticles. Strikingly, introduction of envelope glycoproteins isolated from B lymphocytes into the HCV JFH-1 replicating virus switched the entry tropism of this nonlymphotropic virus from hepatotropism to lymphotropism. Significant detection of viral RNA and viral proteins within B cells was restricted to infections with JFH-1 harboring E1E2 from lymphocytes and depended on an endocytic, pH-dependent entry pathway. Here, we achieved for the first time the isolation of HCV viral proteins carrying entry-related lymphotropism determinants. The identification of genetic determinants within E1E2 represents a first step for a better understanding of the complex relationship between HCV infection, viral persistence, and extrahepatic disorders. IMPORTANCE: Hepatitis C virus (HCV) mainly replicates within the liver. However, it has been shown that patient-derived HCV particles can slightly infect lymphocytes in vitro and in vivo, highlighting the existence of lymphotropism determinants within HCV viral proteins. We isolated HCV envelope glycoproteins from patient B lymphocytes that conferred to a nonlymphotropic HCV the ability to enter B cells, thus providing a platform for characterization of HCV entry into lymphocytes. This unusual tropism was accompanied by a loss of entry function into hepatocytes, suggesting that HCV lymphotropic variants likely constitute a distinct but parallel source for viral persistence and immune escape within chronically infected patients. Moreover, the level of genetic divergence of B-cell-derived envelopes correlated with their degree of lymphotropism, underlining a long-term specialization of some viral populations for B-lymphocytes. Consequently, the clearance of both hepatotropic and nonhepatotropic HCV populations may be important for effective treatment of chronically infected patients.


Assuntos
Linfócitos B/virologia , Hepacivirus/fisiologia , Hepatite C Crônica/virologia , Proteínas do Envelope Viral/metabolismo , Tropismo Viral , Internalização do Vírus , Linhagem Celular , Hepacivirus/isolamento & purificação , Hepatócitos/virologia , Humanos , Transdução Genética
5.
J Virol ; 89(23): 12131-44, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26401036

RESUMO

UNLABELLED: Hepatitis C virus (HCV) only infects humans and chimpanzees, while GB virus B (GBV-B), another hepatotropic hepacivirus, infects small New World primates (tamarins and marmosets). In an effort to develop an immunocompetent small primate model for HCV infection to study HCV pathogenesis and vaccine approaches, we investigated the HCV life cycle step(s) that may be restricted in small primate hepatocytes. First, we found that replication-competent, genome-length chimeric HCV RNAs encoding GBV-B structural proteins in place of equivalent HCV sequences designed to allow entry into simian hepatocytes failed to induce viremia in tamarins following intrahepatic inoculation, nor did they lead to progeny virus in permissive, transfected human Huh7.5 hepatoma cells upon serial passage. This likely reflected the disruption of interactions between distantly related structural and nonstructural proteins that are essential for virion production, whereas such cross talk could be restored in similarly designed HCV intergenotypic recombinants via adaptive mutations in NS3 protease or helicase domains. Next, HCV entry into small primate hepatocytes was examined directly using HCV-pseudotyped retroviral particles (HCV-pp). HCV-pp efficiently infected tamarin hepatic cell lines and primary marmoset hepatocyte cultures through the use of the simian CD81 ortholog as a coreceptor, indicating that HCV entry is not restricted in small New World primate hepatocytes. Furthermore, we observed genomic replication and modest virus secretion following infection of primary marmoset hepatocyte cultures with a highly cell culture-adapted HCV strain. Thus, HCV can successfully complete its life cycle in primary simian hepatocytes, suggesting the possibility of adapting some HCV strains to small primate hosts. IMPORTANCE: Hepatitis C virus (HCV) is an important human pathogen that infects over 150 million individuals worldwide and leads to chronic liver disease. The lack of a small animal model for this infection impedes the development of a preventive vaccine and pathogenesis studies. In seeking to establish a small primate model for HCV, we first attempted to generate recombinants between HCV and GB virus B (GBV-B), a hepacivirus that infects small New World primates (tamarins and marmosets). This approach revealed that the genetic distance between these hepaciviruses likely prevented virus morphogenesis. We next showed that HCV pseudoparticles were able to infect tamarin or marmoset hepatocytes efficiently, demonstrating that there was no restriction in HCV entry into these simian cells. Furthermore, we found that a highly cell culture-adapted HCV strain was able to achieve a complete viral cycle in primary marmoset hepatocyte cultures, providing a promising basis for further HCV adaptation to small primate hosts.


Assuntos
Vírus GB B/fisiologia , Hepacivirus/fisiologia , Estágios do Ciclo de Vida/fisiologia , Modelos Animais , Primatas/virologia , Internalização do Vírus , Animais , Sequência de Bases , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Células HEK293 , Hepacivirus/genética , Hepatócitos/virologia , Especificidade de Hospedeiro , Humanos , Immunoblotting , Dados de Sequência Molecular , Plasmídeos/genética , Análise de Sequência de DNA , Viremia
6.
Mol Ther ; 23(11): 1734-1747, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26281898

RESUMO

The development of lentiviral vectors (LVs) for expression of a specific antibody can be achieved through the transduction of mature B-cells. This approach would provide a versatile tool for active immunotherapy strategies for infectious diseases or cancer, as well as for protein engineering. Here, we created a lentiviral expression system mimicking the natural production of these two distinct immunoglobulin isoforms. We designed a LV (FAM2-LV) expressing an anti-HCV-E2 surface glycoprotein antibody (AR3A) as a membrane-anchored Ig form or a soluble Ig form, depending on the B-cell maturation status. FAM2-LV induced high-level and functional membrane expression of the transgenic antibody in a nonsecretory B-cell line. In contrast, a plasma cell (PC) line transduced with FAM2-LV preferentially produced the secreted transgenic antibody. Similar results were obtained with primary B-cells transduced ex vivo. Most importantly, FAM2-LV transduced primary B-cells efficiently differentiated into PCs, which secreted the neutralizing anti-HCV E2 antibody upon adoptive transfer into immunodeficient NSG (NOD/SCIDγc(-/-)) recipient mice. Altogether, these results demonstrate that the conditional FAM2-LV allows preferential expression of the membrane-anchored form of an antiviral neutralizing antibody in B-cells and permits secretion of a soluble antibody following B-cell maturation into PCs in vivo.


Assuntos
Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Vetores Genéticos , Imunoglobulina G/imunologia , Ativação Linfocitária , Animais , Citotoxicidade Celular Dependente de Anticorpos , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Hepacivirus/imunologia , Humanos , Lentivirus , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Transporte Proteico , Receptores de IgG/metabolismo , Transdução Genética , Proteínas do Envelope Viral/imunologia
7.
J Biol Chem ; 290(38): 23173-87, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26224633

RESUMO

Lipoprotein components are crucial factors for hepatitis C virus (HCV) assembly and entry. As hepatoma cells producing cell culture-derived HCV (HCVcc) particles are impaired in some aspects of lipoprotein metabolism, it is of upmost interest to biochemically and functionally characterize the in vivo produced viral particles, particularly regarding how lipoprotein components modulate HCV entry by lipid transfer receptors such as scavenger receptor BI (SR-BI). Sera from HCVcc-infected liver humanized FRG mice were separated by density gradients. Viral subpopulations, termed HCVfrg particles, were characterized for their physical properties, apolipoprotein association, and infectivity. We demonstrate that, in contrast to the widely spread distribution of apolipoproteins across the different HCVcc subpopulations, the most infectious HCVfrg particles are highly enriched in apoE, suggesting that such apolipoprotein enrichment plays a role for entry of in vivo derived infectious particles likely via usage of apolipoprotein receptors. Consistent with this salient feature, we further reveal previously undefined functionalities of SR-BI in promoting entry of in vivo produced HCV. First, unlike HCVcc, SR-BI is a particularly limiting factor for entry of HCVfrg subpopulations of very low density. Second, HCVfrg entry involves SR-BI lipid transfer activity but not its capacity to bind to the viral glycoprotein E2. In conclusion, we demonstrate that composition and biophysical properties of the different subpopulations of in vivo produced HCVfrg particles modulate their levels of infectivity and receptor usage, hereby featuring divergences with in vitro produced HCVcc particles and highlighting the powerfulness of this in vivo model for the functional study of the interplay between HCV and liver components.


Assuntos
Hepacivirus/metabolismo , Hepatite C/metabolismo , Fígado/virologia , Internalização do Vírus , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Hepacivirus/genética , Hepatite C/genética , Hepatite C/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
8.
J Virol ; 89(16): 8346-64, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26041282

RESUMO

UNLABELLED: In our study, we characterized the effect of monensin, an ionophore that is known to raise the intracellular pH, on the hepatitis C virus (HCV) life cycle. We showed that monensin inhibits HCV entry in a pangenotypic and dose-dependent manner. Monensin induces an alkalization of intracellular organelles, leading to an inhibition of the fusion step between viral and cellular membranes. Interestingly, we demonstrated that HCV cell-to-cell transmission is dependent on the vesicular pH. Using the selective pressure of monensin, we selected a monensin-resistant virus which has evolved to use a new entry route that is partially pH and clathrin independent. Characterization of this mutant led to the identification of two mutations in envelope proteins, the Y297H mutation in E1 and the I399T mutation in hypervariable region 1 (HVR1) of E2, which confer resistance to monensin and thus allow HCV to use a pH-independent entry route. Interestingly, the I399T mutation introduces an N-glycosylation site within HVR1 and increases the density of virions and their sensitivity to neutralization with anti-apolipoprotein E (anti-ApoE) antibodies, suggesting that this mutation likely induces conformational changes in HVR1 that in turn modulate the association with ApoE. Strikingly, the I399T mutation dramatically reduces HCV cell-to-cell spread. In summary, we identified a mutation in HVR1 that overcomes the vesicular pH dependence, modifies the biophysical properties of particles, and drastically reduces cell-to-cell transmission, indicating that the regulation by HVR1 of particle association with ApoE might control the pH dependence of cell-free and cell-to-cell transmission. Thus, HVR1 and ApoE are critical regulators of HCV propagation. IMPORTANCE: Although several cell surface proteins have been identified as entry factors for hepatitis C virus (HCV), the precise mechanisms regulating its transmission to hepatic cells are still unclear. In our study, we used monensin A, an ionophore that is known to raise the intracellular pH, and demonstrated that cell-free and cell-to-cell transmission pathways are both pH-dependent processes. We generated monensin-resistant viruses that displayed different entry routes and biophysical properties. Thanks to these mutants, we highlighted the importance of hypervariable region 1 (HVR1) of the E2 envelope protein for the association of particles with apolipoprotein E, which in turn might control the pH dependency of cell-free and cell-to-cell transmission.


Assuntos
Hepacivirus/fisiologia , Ionóforos/farmacologia , Monensin/farmacologia , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Internalização do Vírus/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Farmacorresistência Viral/genética , Técnica Indireta de Fluorescência para Anticorpo , Hepacivirus/genética , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Mutação de Sentido Incorreto/genética , Testes de Neutralização , Proteínas Virais/metabolismo
9.
J Virol ; 89(1): 535-44, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25339764

RESUMO

UNLABELLED: Bone marrow stromal cell antigen 2 (BST2) is a cellular restriction factor with a broad antiviral activity. In sheep, the BST2 gene is duplicated into two paralogs termed oBST2A and oBST2B. oBST2A impedes viral exit of the Jaagsiekte sheep retroviruses (JSRV), most probably by retaining virions at the cell membrane, similar to the "tethering" mechanism exerted by human BST2. In this study, we provide evidence that unlike oBST2A, oBST2B is limited to the Golgi apparatus and disrupts JSRV envelope (Env) trafficking by sequestering it. In turn, oBST2B leads to a reduction in Env incorporation into viral particles, which ultimately results in the release of virions that are less infectious. Furthermore, the activity of oBST2B does not seem to be restricted to retroviruses, as it also acts on vesicular stomatitis virus glycoproteins. Therefore, we suggest that oBST2B exerts antiviral activity using a mechanism distinct from the classical tethering restriction observed for oBST2A. IMPORTANCE: BST2 is a powerful cellular restriction factor against a wide range of enveloped viruses. Sheep possess two paralogs of the BST2 gene called oBST2A and oBST2B. JSRV, the causative agent of a transmissible lung cancer of sheep, is known to be restricted by oBST2A. In this study, we show that unlike oBST2A, oBST2B impairs the normal cellular trafficking of JSRV envelope glycoproteins by sequestering them within the Golgi apparatus. We also show that oBST2B decreases the incorporation of envelope glycoprotein into JSRV viral particles, which in turn reduces virion infectivity. In conclusion, oBST2B exerts a novel antiviral activity that is distinct from those of BST2 proteins of other species.


Assuntos
Retrovirus Jaagsiekte de Ovinos/imunologia , Retrovirus Jaagsiekte de Ovinos/fisiologia , Glicoproteínas de Membrana/imunologia , Proteínas do Envelope Viral/antagonistas & inibidores , Vírion/metabolismo , Montagem de Vírus , Animais , Complexo de Golgi/metabolismo , Transporte Proteico , Ovinos
10.
Blood ; 124(8): 1221-31, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-24951430

RESUMO

Hematopoietic stem cell (HSC)-based gene therapy holds promise for the cure of many diseases. The field is now moving toward the use of lentiviral vectors (LVs) as evidenced by 4 successful clinical trials. These trials used vesicular-stomatitis-virus-G protein (VSV-G)-LVs at high doses combined with strong cytokine-cocktail stimulation to obtain therapeutically relevant transduction levels; however, they might compromise the HSC character. Summarizing all these disadvantages, alternatives to VSV-G-LVs are urgently needed. We generated here high-titer LVs pseudotyped with a baboon retroviral envelope glycoprotein (BaEV-LVs), resistant to human complement. Under mild cytokine prestimulation to preserve the HSC characteristics, a single BaEV-LV application at a low dose, resulted in up to 90% of hCD34(+) cell transduction. Even more striking was that these new BaEV-LVs allowed, at low doses, efficient transduction of up to 30% of quiescent hCD34(+) cells, whereas high-dose VSV-G-LVs were insufficient. Importantly, reconstitution of NOD/Lt-SCID/γc(-/-) (NSG) mice with BaEV-LV-transduced hCD34(+) cells maintained these high transduction levels in all myeloid and lymphoid lineages, including early progenitors. This transduction pattern was confirmed or even increased in secondary NSG recipient mice. This suggests that BaEV-LVs efficiently transduce true HSCs and could improve HSC-based gene therapy, for which high-level HSC correction is needed for life-long cure.


Assuntos
Betaretrovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Células-Tronco Hematopoéticas , Lentivirus/genética , Transdução Genética , Proteínas do Envelope Viral/genética , Animais , Antígenos CD34 , Linhagem Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Macaca , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID
11.
PLoS One ; 9(6): e99601, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24927415

RESUMO

Hepatitis C virus (HCV) is a leading cause of cirrhosis and liver cancer worldwide. We recently characterized for the first time the expression of Signaling Lymphocyte Activating Molecule 3 (SLAMF3) in human hepatocytes and here, we report that SLAMF3 interacts with the HCV viral protein E2 and is implicated in HCV entry process. We found a strong correlation between SLAMF3 expression level and hepatocyte susceptibility to HCV infection. The use of specific siRNAs to down-modulate SLAMF3 expression and SLAMF3-blocking antibodies both decreased the hepatocytes susceptibility to HCV infection. Moreover, SLAMF3 over-expression significantly increased susceptibility to HCV infection. Interestingly, experiments with peptides derived from each SLAMF3 domain showed that the first N-terminal extracellular domain is essential for interaction with HCV particles. Finally, we showed that recombinant HCV envelop protein E2 can bind SLAMF3 and that anti-SLAMF3 antibodies inhibited specifically this interaction. Overall, our results revealed that SLAMF3 plays a role during HCV entry, likely by enhancing entry of viral particle within hepatocytes.


Assuntos
Antígenos CD/metabolismo , Hepacivirus/fisiologia , Hepatite C/virologia , Proteínas do Envelope Viral/metabolismo , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/imunologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Hepatite C/genética , Hepatite C/metabolismo , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária , Tetraspanina 28/farmacologia , Proteínas do Envelope Viral/imunologia
12.
PLoS One ; 9(3): e92140, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24646941

RESUMO

Hepatitis C virus (HCV) establishes infection using host lipid metabolism pathways that are thus considered potential targets for indirect anti-HCV strategies. HCV enters the cell via clathrin-dependent endocytosis, interacting with several receptors, and virus-cell fusion, which depends on acidic pH and the integrity of cholesterol-rich domains of the hepatocyte membrane. The ATP-binding Cassette Transporter A1 (ABCA1) mediates cholesterol efflux from hepatocytes to extracellular Apolipoprotein A1 and moves cholesterol within cell membranes. Furthermore, it generates high-density lipoprotein (HDL) particles. HDL protects against arteriosclerosis and cardiovascular disease. We show that the up-regulation of ABCA1 gene expression and its cholesterol efflux function in Huh7.5 hepatoma cells, using the liver X receptor (LXR) agonist GW3965, impairs HCV infection and decreases levels of virus produced. ABCA1-stimulation inhibited HCV cell entry, acting on virus-host cell fusion, but had no impact on virus attachment, replication, or assembly/secretion. It did not affect infectivity or properties of virus particles produced. Silencing of the ABCA1 gene and reduction of the specific cholesterol efflux function counteracted the inhibitory effect of the GW3965 on HCV infection, providing evidence for a key role of ABCA1 in this process. Impaired virus-cell entry correlated with the reorganisation of cholesterol-rich membrane microdomains (lipid rafts). The inhibitory effect could be reversed by an exogenous cholesterol supply, indicating that restriction of HCV infection was induced by changes of cholesterol content/distribution in membrane regions essential for virus-cell fusion. Stimulation of ABCA1 expression by GW3965 inhibited HCV infection of both human primary hepatocytes and isolated human liver slices. This study reveals that pharmacological stimulation of the ABCA1-dependent cholesterol efflux pathway disrupts membrane cholesterol homeostasis, leading to the inhibition of virus-cell fusion and thus HCV cell entry. Therefore besides other beneficial roles, ABCA1 might represent a potential target for HCV therapy.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/genética , Hepacivirus/fisiologia , Hepatite C/genética , Hepatite C/virologia , Regulação para Cima/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Benzoatos/farmacologia , Benzilaminas/farmacologia , Ciclo Celular/efeitos dos fármacos , Fusão Celular , Linhagem Celular Tumoral , Colesterol/metabolismo , Células HEK293 , Hepacivirus/efeitos dos fármacos , Hepacivirus/patogenicidade , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Microdomínios da Membrana/metabolismo , RNA Viral/metabolismo , Receptores Virais/metabolismo , Regulação para Cima/efeitos dos fármacos , Vírion/efeitos dos fármacos , Vírion/metabolismo , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
13.
Hepatology ; 59(3): 776-88, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24038151

RESUMO

UNLABELLED: Hepatitis C virus (HCV) envelope glycoproteins E1 and E2 are important mediators for productive cell entry. However, knowledge about their structure, intra- or intermolecular dialogs, and conformational changes is scarce, limiting the design of therapeutic strategies targeting E1E2. Here we sought to investigate how certain domains of E1 and E2 have coevolved to optimize their interactions to promote efficient HCV entry. For this purpose we generated chimeric E1E2 heterodimers derived from two HCV 1a strains to identify and characterize crosstalk between their domains. We found an E1E2 combination that drastically impaired the infectivity of cell culture-derived HCV particles, whereas the reciprocal E1E2 combination led to increased infectivity. Using HCV pseudoparticle assays, we confirmed the opposing entry phenotypes of these heterodimers. By mutagenesis analysis, we identified a particular crosstalk between three amino acids of E1 and the domain III of E2. Its modulation leads to either a full restoration of the functionality of the suboptimal heterodimer or a destabilization of the functional heterodimer. Interestingly, we found that this crosstalk modulates E1E2 binding to HCV entry receptors SR-BI and CD81. In addition, we found for the first time that E1E2 complexes can interact with the first extracellular loop of Claudin-1, whereas soluble E2 did not. These results highlight the critical role of E1 in the modulation of HCV binding to receptors. Finally, we demonstrated that this crosstalk is involved in membrane fusion. CONCLUSIONS: These results reveal a multifunctional and crucial interaction between E1 and E2 for HCV entry into cells. Our study highlights the role of E1 as a modulator of HCV binding to receptors and membrane fusion, underlining its potential as an antiviral target.


Assuntos
Hepacivirus/metabolismo , Hepatite C/virologia , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Carcinoma Hepatocelular , Claudina-1/metabolismo , Dimerização , Células HEK293 , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Humanos , Neoplasias Hepáticas , Fusão de Membrana/fisiologia , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Receptores Depuradores Classe B/metabolismo , Tetraspanina 28/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
14.
Hepatology ; 58(1): 86-97, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23348596

RESUMO

UNLABELLED: Hepatitis C virus (HCV) is a major cause of chronic liver disease. Despite recent success in improving anti-HCV therapy, additional progress is still needed to develop cheaper and interferon (IFN)-free treatments. Here, we report that ferroquine (FQ), an antimalarial ferrocenic analog of chloroquine, is a novel inhibitor of HCV. FQ potently inhibited HCV infection of hepatoma cell lines by affecting an early step of the viral life cycle. The antiviral activity of FQ on HCV entry was confirmed with pseudoparticles expressing HCV envelope glycoproteins E1 and E2 from six different genotypes. In addition to its effect on HCV entry, FQ also inhibited HCV RNA replication, albeit at a higher concentration. We also showed that FQ has no effect on viral assembly and virion secretion. Using a binding assay at 4°C, we showed that FQ does not prevent attachment of the virus to the cell surface. Furthermore, virus internalization was not affected by FQ, whereas the fusion process was impaired in the presence of FQ as shown in a cell-cell fusion assay. Finally, virus with resistance to FQ was selected by sequential passage in the presence of the drug, and resistance was shown to be conferred by a single mutation in E1 glycoprotein (S327A). By inhibiting cell-free virus transmission using a neutralizing antibody, we also showed that FQ inhibits HCV cell-to-cell spread between neighboring cells. Combinations of FQ with IFN, or an inhibitor of HCV NS3/4A protease, also resulted in additive to synergistic activity. CONCLUSION: FQ is a novel, interesting anti-HCV molecule that could be used in combination with other direct-acting antivirals.


Assuntos
Aminoquinolinas/farmacologia , Antivirais/uso terapêutico , Compostos Ferrosos/farmacologia , Hepacivirus/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Hepacivirus/genética , Hepatite C/prevenção & controle , Humanos , Interferon-alfa/administração & dosagem , Metalocenos , Prolina/administração & dosagem , Prolina/análogos & derivados , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/antagonistas & inibidores
15.
J Biol Chem ; 287(37): 31242-57, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22767607

RESUMO

Hepatitis C virus (HCV) particles assemble along the very low density lipoprotein pathway and are released from hepatocytes as entities varying in their degree of lipid and apolipoprotein (apo) association as well as buoyant densities. Little is known about the cell entry pathway of these different HCV particle subpopulations, which likely occurs by regulated spatiotemporal processes involving several cell surface molecules. One of these molecules is the scavenger receptor BI (SR-BI), a receptor for high density lipoprotein that can bind to the HCV glycoprotein E2. By studying the entry properties of infectious virus subpopulations differing in their buoyant densities, we show that these HCV particles utilize SR-BI in a manifold manner. First, SR-BI mediates primary attachment of HCV particles of intermediate density to cells. These initial interactions involve apolipoproteins, such as apolipoprotein E, present on the surface of HCV particles, but not the E2 glycoprotein, suggesting that lipoprotein components in the virion act as host-derived ligands for important entry factors such as SR-BI. Second, we found that in contrast to this initial attachment, SR-BI mediates entry of HCV particles independent of their buoyant density. This function of SR-BI does not depend on E2/SR-BI interaction but relies on the lipid transfer activity of SR-BI, probably by facilitating entry steps along with other HCV entry co-factors. Finally, our results underscore a third function of SR-BI governed by specific residues in hypervariable region 1 of E2 leading to enhanced cell entry and depending on SR-BI ability to bind to E2.


Assuntos
Apolipoproteínas E/metabolismo , Hepacivirus/fisiologia , Receptores Depuradores Classe B/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Apolipoproteínas E/genética , Linhagem Celular Tumoral , Humanos , Camundongos , Estrutura Terciária de Proteína , Ratos , Receptores Depuradores Classe B/genética , Proteínas do Envelope Viral/genética
16.
Biochem J ; 443(1): 85-94, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22240035

RESUMO

The protonation of histidine in acidic environments underpins its role in regulating the function of pH-sensitive proteins. For pH-sensitive viral fusion proteins, histidine protonation in the endosome leads to the activation of their membrane fusion function. The HCV (hepatitis C virus) glycoprotein E1-E2 heterodimer mediates membrane fusion within the endosome, but the roles of conserved histidine residues in the formation of a functional heterodimer and in sensing pH changes is unknown. We examined the functional roles of conserved histidine residues located within E1 and E2. The E1 mutations, H222A/R, H298R and H352A, disrupted E1-E2 heterodimerization and reduced virus entry. A total of five out of six histidine residues located within the E2 RBD (receptor-binding domain) were important for the E2 fold, and their substitution with arginine or alanine caused aberrant heterodimerization and/or CD81 binding. Distinct roles in E1-E2 heterodimerization and in virus entry were identified for His691 and His693 respectively within the membrane-proximal stem region. Viral entry and cell-cell fusion at neutral and low pH values were enhanced with H445R, indicating that the protonation state of His445 is a key regulator of HCV fusion. However, H445R did not overcome the block to virus entry induced by bafilomycin A1, indicating a requirement for an endosomal activation trigger in addition to acidic pH.


Assuntos
Hepacivirus/fisiologia , Tetraspanina 28/química , Proteínas do Envelope Viral/biossíntese , Internalização do Vírus , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência Conservada , Células HEK293 , Hepacivirus/patogenicidade , Histidina/genética , Humanos , Concentração de Íons de Hidrogênio , Mutagênese Sítio-Dirigida , Ligação Proteica , Dobramento de Proteína , Multimerização Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Vírion
17.
Virology ; 425(1): 31-9, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22280897

RESUMO

Hepatocytes are highly polarized cells where intercellular junctions, including tight junctions (TJs), determine the polarity. Recently, the TJ-associated proteins claudin-1 and occludin have been implicated in hepatitis C virus (HCV) entry and spread. Nevertheless, cell line-based experimental systems that exhibit hepatocyte-like polarity and permit robust infection and virion production are not currently available. Thus, we sought to determine whether cell line-based, Matrigel-embedded cultures could be used to study hepatitis C virus (HCV) infection and virion production in a context of hepatocyte-like polarized cells. In contrast to standard bidimensional cultures, Matrigel-cultured Huh-7 cells adopted hepatocyte polarization features forming a continuous network of functional proto-bile canaliculi structures. These 3D cultures supported HCV infection by JFH-1 virus and produced infective viral particles which shifted towards lower densities with higher associated specific infectivity. In conclusion, our findings describe a novel use of Matrigel to study the entire HCV cycle in a more relevant context.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura de Células/métodos , Colágeno/química , Hepacivirus/patogenicidade , Laminina/química , Proteoglicanas/química , Inclusão do Tecido/métodos , Vírion/metabolismo , Canalículos Biliares/ultraestrutura , Linhagem Celular Tumoral , Polaridade Celular , Combinação de Medicamentos , Hepacivirus/genética , Hepacivirus/metabolismo , Hepacivirus/ultraestrutura , Hepatite C/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Microscopia Confocal , Junções Íntimas/ultraestrutura , Vírion/ultraestrutura
18.
J Biol Chem ; 286(27): 23865-76, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21555519

RESUMO

Several conserved domains critical for E1E2 assembly and hepatitis C virus entry have been identified in E1 and E2 envelope glycoproteins. However, the role of less conserved domains involved in cross-talk between either glycoprotein must be defined to fully understand how E1E2 undergoes conformational changes during cell entry. To characterize such domains and to identify their functional partners, we analyzed a set of intergenotypic E1E2 heterodimers derived from E1 and E2 of different genotypes. The infectivity of virions indicated that Con1 E1 did not form functional heterodimers when associated with E2 from H77. Biochemical analyses demonstrated that the reduced infectivity was not related to alteration of conformation and incorporation of Con1 E1/H77 E2 heterodimers but rather to cell entry defects. Thus, we generated chimeric E1E2 glycoproteins by exchanging different domains of each protein in order to restore functional heterodimers. We found that both the ectodomain and transmembrane domain of E1 influenced infectivity. Site-directed mutagenesis highlighted the role of amino acids 359, 373, and 375 in transmembrane domain in entry. In addition, we identified one domain involved in entry within the N-terminal part of E1, and we isolated a motif at position 219 that is critical for H77 function. Interestingly, using additional chimeric E1E2 complexes harboring substitutions in this motif, we found that the transmembrane domain of E1 acts as a partner of this motif. Therefore, we characterized domains of E1 and E2 that have co-evolved inside a given genotype to optimize their interactions and allow efficient entry.


Assuntos
Hepacivirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Motivos de Aminoácidos , Linhagem Celular , Hepacivirus/genética , Humanos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/genética
19.
Nat Med ; 17(5): 589-95, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21516087

RESUMO

Hepatitis C virus (HCV) is a major cause of liver disease, but therapeutic options are limited and there are no prevention strategies. Viral entry is the first step of infection and requires the cooperative interaction of several host cell factors. Using a functional RNAi kinase screen, we identified epidermal growth factor receptor and ephrin receptor A2 as host cofactors for HCV entry. Blocking receptor kinase activity by approved inhibitors broadly impaired infection by all major HCV genotypes and viral escape variants in cell culture and in a human liver chimeric mouse model in vivo. The identified receptor tyrosine kinases (RTKs) mediate HCV entry by regulating CD81-claudin-1 co-receptor associations and viral glycoprotein-dependent membrane fusion. These results identify RTKs as previously unknown HCV entry cofactors and show that tyrosine kinase inhibitors have substantial antiviral activity. Inhibition of RTK function may constitute a new approach for prevention and treatment of HCV infection.


Assuntos
Receptores ErbB/fisiologia , Hepacivirus/fisiologia , Hepatite C/fisiopatologia , Hepatite C/virologia , Receptor EphA2/fisiologia , Internalização do Vírus , Animais , Antígenos CD/fisiologia , Antivirais/farmacologia , Sequência de Bases , Linhagem Celular , Claudina-1 , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Cloridrato de Erlotinib , Hepacivirus/efeitos dos fármacos , Hepatite C/prevenção & controle , Hepatite C/terapia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Ligantes , Proteínas de Membrana/fisiologia , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor EphA2/antagonistas & inibidores , Receptor EphA2/genética , Tetraspanina 28 , Internalização do Vírus/efeitos dos fármacos
20.
Adv Genet ; 73: 121-83, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21310296

RESUMO

Enveloped viruses penetrate their cell targets following the merging of their membrane with that of the cell. This fusion process is catalyzed by one or several viral glycoproteins incorporated on the membrane of the virus. These envelope glycoproteins (EnvGP) evolved in order to combine two features. First, they acquired a domain to bind to a specific cellular protein, named "receptor." Second, they developed, with the help of cellular proteins, a function of finely controlled fusion to optimize the replication and preserve the integrity of the cell, specific to the genus of the virus. Following the activation of the EnvGP either by binding to their receptors and/or sometimes the acid pH of the endosomes, many changes of conformation permit ultimately the action of a specific hydrophobic domain, the fusion peptide, which destabilizes the cell membrane and leads to the opening of the lipidic membrane. The comprehension of these mechanisms is essential to develop medicines of the therapeutic class of entry inhibitor like enfuvirtide (Fuzeon) against human immunodeficiency virus (HIV). In this chapter, we will summarize the different envelope glycoprotein structures that viruses develop to achieve membrane fusion and the entry of the virus. We will describe the different entry pathways and cellular proteins that viruses have subverted to allow infection of the cell and the receptors that are used. Finally, we will illustrate more precisely the recent discoveries that have been made within the field of the entry process, with a focus on the use of pseudoparticles. These pseudoparticles are suitable for high-throughput screenings that help in the development of natural or artificial inhibitors as new therapeutics of the class of entry inhibitors.


Assuntos
Fusão de Membrana , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Viroses/virologia , Vírus/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/virologia , Endocitose , Endossomos/metabolismo , Enfuvirtida , Proteína gp41 do Envelope de HIV/administração & dosagem , Proteína gp41 do Envelope de HIV/uso terapêutico , Inibidores da Fusão de HIV/administração & dosagem , Inibidores da Fusão de HIV/uso terapêutico , Ensaios de Triagem em Larga Escala , Humanos , Concentração de Íons de Hidrogênio , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/uso terapêutico , Ligação Proteica , Redobramento de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/genética , Vírion/química , Vírion/metabolismo , Vírion/fisiologia , Viroses/tratamento farmacológico , Viroses/metabolismo , Vírus/genética , Vírus/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA