Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 558, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35091550

RESUMO

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Assuntos
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Febre Hemorrágica Americana/metabolismo , Receptores da Transferrina/metabolismo , Proteínas do Envelope Viral/metabolismo , Células A549 , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Antígenos CD/imunologia , Arenaviridae/efeitos dos fármacos , Arenaviridae/fisiologia , Chlorocebus aethiops , Febre Hemorrágica Americana/prevenção & controle , Febre Hemorrágica Americana/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus Junin/efeitos dos fármacos , Vírus Junin/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Simulação de Acoplamento Molecular , Ligação Proteica/efeitos dos fármacos , Receptores da Transferrina/antagonistas & inibidores , Receptores da Transferrina/imunologia , Células Vero
2.
J Gen Virol ; 101(6): 573-586, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32375950

RESUMO

Bone marrow stromal cell antigen-2 (BST-2), also known as tetherin, is an interferon-inducible membrane-associated protein. It effectively targets enveloped viruses at the release step of progeny viruses from host cells, thereby restricting the further spread of viral infection. Junin virus (JUNV) is a member of Arenaviridae, which causes Argentine haemorrhagic fever that is associated with a high rate of mortality. In this study, we examined the effect of human BST-2 on the replication and propagation of JUNV. The production of JUNV Z-mediated virus-like particles (VLPs) was significantly inhibited by over-expression of BST-2. Electron microscopy analysis revealed that BST-2 functions by forming a physical link that directly retains VLPs on the cell surface. Infection using JUNV showed that infectious JUNV production was moderately inhibited by endogenous or exogenous BST-2. We also observed that JUNV infection triggers an intense interferon response, causing an upregulation of BST-2, in infected cells. However, the expression of cell surface BST-2 was reduced upon infection. Furthermore, the expression of JUNV nucleoprotein (NP) partially recovered VLP production from BST-2 restriction, suggesting that the NP functions as an antagonist against antiviral effect of BST-2. We further showed that JUNV NP also rescued the production of Ebola virus VP40-mediated VLP from BST-2 restriction as a broad spectrum BST-2 antagonist. To our knowledge, this is the first report showing that an arenavirus protein counteracts the antiviral function of BST-2.


Assuntos
Antígenos CD/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Vírus Junin/fisiologia , Nucleoproteínas/metabolismo , Proteínas do Core Viral/metabolismo , Liberação de Vírus/fisiologia , Células A549 , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Interferons/farmacologia , Vírus Junin/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
3.
J Virol ; 90(18): 8341-50, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27412594

RESUMO

UNLABELLED: Arenaviruses are responsible for severe and often fatal hemorrhagic disease. In the absence of effective antiviral therapies and vaccines, these viruses pose serious threats to public health and biodefense. Arenaviruses enter the host cell by fusion of the viral and endosomal membranes, a process mediated by the virus envelope glycoprotein GPC. Unlike other class I viral fusion proteins, GPC retains its stable signal peptide (SSP) as an essential third subunit in the mature complex. SSP spans the membrane twice and is myristoylated at its cytoplasmic N terminus. Mutations that abolish SSP myristoylation have been shown to reduce pH-induced cell-cell fusion activity of ectopically expressed GPC to ∼20% of wild-type levels. In order to examine the role of SSP myristoylation in the context of the intact virus, we used reverse genetics to generate Junín viruses (Candid #1 isolate) in which the critical glycine-2 residue in SSP was either replaced by alanine (G2A) or deleted (ΔG2). These mutant viruses produced smaller foci of infection in Vero cells and showed an ∼5-fold reduction in specific infectivity, commensurate with the defect in cell-cell fusion. However, virus assembly and GPC incorporation into budded virions were unaffected. Our findings suggest that the myristate moiety is cryptically disposed in the prefusion GPC complex and may function late in the fusion process to promote merging of the viral and cellular membranes. IMPORTANCE: Hemorrhagic fever arenaviruses pose significant threats to public health and biodefense. Arenavirus entry into the host cell is promoted by the virus envelope glycoprotein GPC. Unlike other viral envelope glycoproteins, GPC contains a myristoylated stable signal peptide (SSP) as an essential third subunit. Myristoylation has been shown to be important for the membrane fusion activity of recombinantly expressed GPC. Here, we use reverse genetics to study the role of SSP myristoylation in the context of the intact virion. We find that nonmyristoylated GPC mutants of the Candid #1 strain of Junín virus display a commensurate deficiency in their infectivity, albeit without additional defects in virion assembly and budding. These results suggest that SSP myristoylation may function late in the fusion process to facilitate merging of the viral and cellular membranes. Antiviral agents that target this novel aspect of GPC membrane fusion may be useful in the treatment of arenavirus hemorrhagic fevers.


Assuntos
Vírus Junin/fisiologia , Glicoproteínas de Membrana/metabolismo , Ácido Mirístico/metabolismo , Processamento de Proteína Pós-Traducional , Sinais Direcionadores de Proteínas , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus , Internalização do Vírus , Substituição de Aminoácidos , Animais , Chlorocebus aethiops , Glicoproteínas/genética , Glicoproteínas/metabolismo , Vírus Junin/genética , Glicoproteínas de Membrana/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Genética Reversa , Deleção de Sequência , Células Vero , Proteínas do Envelope Viral/genética , Vírion/metabolismo
4.
J Virol ; 89(14): 7079-88, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25926656

RESUMO

UNLABELLED: The arenavirus family includes several important pathogens that cause severe and sometimes fatal diseases in humans. The highly pathogenic Old World (OW) arenavirus Lassa fever virus (LASV) is the causative agent of Lassa fever (LF) disease in humans. LASV infections in severe cases are generally immunosuppressive without stimulating interferon (IFN) induction, a proinflammatory response, or T cell activation. However, the host innate immune responses to highly pathogenic New World (NW) arenaviruses are not well understood. We have previously shown that the highly pathogenic NW arenavirus, Junin virus (JUNV), induced an IFN response in human A549 cells. Here, we report that Machupo virus (MACV), another highly pathogenic NW arenavirus, also induces an IFN response. Importantly, both pathogenic NW arenaviruses, in contrast to the OW highly pathogenic arenavirus LASV, readily elicited an IFN response in human primary dendritic cells and A549 cells. Coinfection experiments revealed that LASV could potently inhibit MACV-activated IFN responses even at 6 h after MACV infection, while the replication levels of MACV and LASV were not affected by virus coinfection. Our results clearly demonstrated that although all viruses studied herein are highly pathogenic to humans, the host IFN responses toward infections with the NW arenaviruses JUNV and MACV are quite different from responses to infections with the OW arenavirus LASV, a discovery that needs to be further investigated in relevant animal models. This finding might help us better understand various interplays between the host immune system and highly pathogenic arenaviruses as well as distinct mechanisms underlying viral pathogenesis. IMPORTANCE: Infections of humans with the highly pathogenic OW LASV are accompanied by potent suppression of interferon or proinflammatory cytokine production. In contrast, infections with the highly pathogenic NW arenavirus JUNV are associated with high levels of IFNs and cytokines in severe and fatal cases. Arenaviruses initially target macrophages and dendritic cells, which are potent IFN/cytokine-producers. In human macrophages, JUNV reportedly does not trigger IFN responses. We here demonstrated that JUNV activated IFN responses in human dendritic cells. MACV, another highly pathogenic NW arenavirus, also activated IFN responses. LASV did not induce detectable IFN responses, in spite of higher replication levels, and blocked the MACV-triggered IFN response in a coinfection assay. Although these viruses are highly pathogenic to humans, our study highlights distinct innate immune responses to infections with the NW arenaviruses JUNV and MACV and to infection with the OW arenavirus LASV and provides important insights into the virus-host interaction and pathogenesis.


Assuntos
Arenavirus do Novo Mundo/imunologia , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Interferons/biossíntese , Vírus Junin/imunologia , Arenavirus do Novo Mundo/fisiologia , Células Cultivadas , Células Dendríticas/virologia , Células Epiteliais/virologia , Humanos , Vírus Junin/fisiologia , Replicação Viral
5.
J Virol ; 89(11): 5949-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25810546

RESUMO

UNLABELLED: The New World arenavirus Junin virus (JUNV) is the causative agent of Argentine hemorrhagic fever (AHF), a potentially deadly disease endemic to central regions of Argentina. The live-attenuated Candid #1 (Can) strain of JUNV is currently used to vaccinate the human population at risk. However, the mechanism of attenuation of this strain is still largely unknown. Therefore, the identification and functional characterization of viral genetic determinants dictating JUNV virulence or attenuation would significantly improve the understanding of the mechanisms underlying AHF and facilitate the development of novel, more effective, and safer vaccines. Here, we utilized a reverse genetics approach to generate recombinant JUNV (rJUNV) strains encoding different gene combinations of the pathogenic Romero (Rom) and attenuated Can strains of JUNV. All strains of rJUNV exhibited in vitro growth kinetics similar to those of their parental counterparts. Analysis of virulence of the rJUNV in a guinea pig model of lethal infection that closely reproduces the features of AHF identified the envelope glycoproteins (GPs) as the major determinants of pathogenesis and attenuation of JUNV. Accordingly, rJUNV strains expressing the full-length GPs of Rom and Can exhibited virulent and attenuated phenotypes, respectively, in guinea pigs. Mutation F427I in the transmembrane region of JUNV envelope glycoprotein GP2 has been shown to attenuate the neurovirulence of JUNV in suckling mice. We document that in the guinea pig model of AHF, mutation F427I in GP2 is also highly attenuating but insufficient to prevent virus dissemination and development of mild clinical and pathological symptoms, indicating that complete attenuation of JUNV requires additional mutations present in Can glycoprotein precursor (GPC). IMPORTANCE: Development of antiviral strategies against viral hemorrhagic fevers, including AHF, is one of the top priorities within the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Live-attenuated Candid #1 strain, derived from the 44th mouse brain passage of the prototype XJ strain of JUNV, has been demonstrated to be safe, immunogenic, and highly protective and is currently licensed for human use in Argentina. However, the bases for the attenuated phenotype of Candid #1 have not been established. Therefore, the identification and functional characterization of viral genetic factors implicated in JUNV pathogenesis and attenuation would significantly improve the understanding of the molecular mechanisms underlying AHF and facilitate the development of novel antiviral strategies.


Assuntos
Glicoproteínas/metabolismo , Febre Hemorrágica Americana/virologia , Vírus Junin/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Modelos Animais de Doenças , Glicoproteínas/genética , Cobaias , Febre Hemorrágica Americana/patologia , Vírus Junin/genética , Genética Reversa , Proteínas do Envelope Viral/genética , Virulência , Fatores de Virulência
6.
Am J Trop Med Hyg ; 90(6): 993-1002, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24710609

RESUMO

Junín virus (JUNV) is endemic to the fertile Pampas of Argentina, maintained in nature by the rodent host Calomys musculinus, and the causative agent of Argentine hemorrhagic fever (AHF), which is characterized by vascular dysfunction and fluid distribution abnormalities. Clinical as well as experimental studies implicate involvement of the endothelium in the pathogenesis of AHF, although little is known of its role. JUNV has been shown to result in productive infection of endothelial cells (ECs) in vitro with no visible cytopathic effects. In this study, we show that direct JUNV infection of primary human ECs results in increased vascular permeability as measured by electric cell substrate impedance sensing and transwell permeability assays. We also show that EC adherens junctions are disrupted during virus infection, which may provide insight into the role of the endothelium in the pathogenesis of AHF and possibly, other viral hemorrhagic fevers.


Assuntos
Junções Aderentes/virologia , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Febre Hemorrágica Americana/virologia , Vírus Junin/fisiologia , Sigmodontinae/virologia , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Cateninas/metabolismo , Permeabilidade da Membrana Celular , Quimiocina CCL2/metabolismo , Reservatórios de Doenças , Fluoresceína-5-Isotiocianato/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-6/metabolismo , Permeabilidade , delta Catenina
7.
J Virol ; 88(14): 7703-14, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24760892

RESUMO

Successful adaptive immunity to virus infection often depends on the initial innate response. Previously, we demonstrated that Junín virus, the etiological agent responsible for Argentine hemorrhagic fever (AHF), activates an early innate immune response via an interaction between the viral glycoprotein and Toll-like receptor 2 (TLR2). Here we show that TLR2/6 but not TLR1/2 heterodimers sense Junín virus glycoprotein and induce a cytokine response, which in turn upregulates the expression of the RNA helicases RIG-I and MDA5. NF-κB and Erk1/2 were important in the cytokine response, since both proteins were phosphorylated as a result of the interaction of virus with TLR2, and treatment with an Erk1/2-specific inhibitor blocked cytokine production. We show that the Junín virus glycoprotein activates cytokine production in a human macrophage cell line as well. Moreover, we show that TLR2-mediated immune response plays a role in viral clearance because wild-type mice cleared Candid 1 (JUNV C1), the vaccine strain of Junín virus, more rapidly than did TLR2 knockout mice. This clearance correlated with the generation of Junín virus-specific CD8(+) T cells. However, infected wild-type and TLR2 knockout mice developed TLR2-independent blocking antibody responses with similar kinetics. We also show that microglia and astrocytes but not neurons are susceptible to infection with JUNV C1. Although JUNV C1 infection of the brain also triggered a TLR2-dependent cytokine response, virus levels were equivalent in wild-type and TLR2 knockout mice. Importance: Junín virus is transmitted by rodents native to Argentina and is associated with both systemic disease and, in some patients, neurological symptoms. Humans become infected when they inhale aerosolized Junín virus. AHF has a 15 to 30% mortality rate, and patients who clear the infection develop a strong antibody response to Junín virus. Here we investigated what factors determine the immune response to Junín virus. We show that a strong initial innate immune response to JUNV C1 determines how quickly mice can clear systemic infection and that this depended on the cellular immune response. In contrast, induction of an innate immune response in the brain had no effect on virus infection levels. These findings may explain how the initial immune response to Junín virus infection could determine different outcomes in humans.


Assuntos
Imunidade Adaptativa , Sangue/imunologia , Encéfalo/imunologia , Imunidade Inata , Vírus Junin/imunologia , Receptor 2 Toll-Like/imunologia , Replicação Viral , Animais , Sangue/virologia , Encéfalo/virologia , Linhagem Celular , Citocinas/metabolismo , Humanos , Vírus Junin/fisiologia , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Biochem Biophys Res Commun ; 441(3): 612-617, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24183720

RESUMO

The target cell tropism of enveloped viruses is regulated by interactions between viral proteins and cellular receptors determining susceptibility at a host cell, tissue or species level. However, a number of additional cell-surface moieties can also bind viral envelope glycoproteins and could act as capture receptors, serving as attachment factors to concentrate virus particles on the cell surface, or to disseminate the virus infection to target organs or susceptible cells within the host. Here, we used Junín virus (JUNV) or JUNV glycoprotein complex (GPC)-pseudotyped particles to study their ability to be internalized by the human C-type lectins hDC- or hL-SIGN. Our results provide evidence that hDC- and hL-SIGN can mediate the entry of Junín virus into cells, and may play an important role in virus infection and dissemination in the host.


Assuntos
Moléculas de Adesão Celular/metabolismo , Febre Hemorrágica Americana/metabolismo , Febre Hemorrágica Americana/virologia , Interações Hospedeiro-Patógeno , Vírus Junin/fisiologia , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Internalização do Vírus , Células 3T3 , Animais , Antígenos CD/metabolismo , Chlorocebus aethiops , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Receptores da Transferrina/metabolismo , Células Vero , Proteínas do Envelope Viral/metabolismo
9.
Sci Transl Med ; 5(204): 204ra131, 2013 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-24068738

RESUMO

New World hemorrhagic fever arenavirus infection results in 15 to 30% mortality in humans. We performed a high-throughput small interfering RNA screen with Junín virus glycoprotein-pseudotyped viruses to find potential host therapeutic targets. Voltage-gated calcium channel (VGCC) subunits, for which there are Food and Drug Administration (FDA)-approved drugs, were identified in the screen. Knockdown of VGCC subunits or treatment with channel blockers diminished Junín virus-cell fusion and entry into cells and thereby decreased infection. Gabapentin, an FDA-approved drug used to treat neuropathic pain that targets the α2δ2 subunit, inhibited infection of mice by the Candid 1 vaccine strain of the virus. These findings demonstrate that VGCCs play a role in virus infection and have the potential to lead to therapeutic intervention of New World arenavirus infection.


Assuntos
Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Ensaios de Triagem em Larga Escala , Vírus Junin/fisiologia , Terapia de Alvo Molecular , RNA Interferente Pequeno/genética , Internalização do Vírus , Aminas/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Fusão Celular , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Chlorocebus aethiops , Ácidos Cicloexanocarboxílicos/farmacologia , Gabapentina , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno/genética , Humanos , Vírus Junin/efeitos dos fármacos , Camundongos , Ligação Proteica/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Interferência de RNA , Receptores da Transferrina/metabolismo , Células Vero , Internalização do Vírus/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia
10.
Biochem Biophys Res Commun ; 430(3): 912-7, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23261443

RESUMO

Arenavirus morphogenesis and budding occurs at cellular plasma membrane; however, the nature of membrane assembly sites remains poorly understood. In this study we examined the effect of different cholesterol-lowering agents on Junín virus (JUNV) multiplication. We found that cholesterol cell depletion reduced JUNV glycoproteins (GPs) membrane expression and virus budding. Analysis of membrane protein insolubility in Triton X-100 suggested that JUNV GPs associate with cholesterol enriched membranes. Rafts dissociation conditions as warm detergent extraction and cholesterol removal by methyl-ß-cyclodextrin compound showed to impair GPs cholesterol enriched membrane association. Analysis of GPs transfected cells showed similar results suggesting that membrane raft association is independent of other viral proteins.


Assuntos
Membrana Celular/química , Membrana Celular/virologia , Colesterol/química , Vírus Junin/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Replicação Viral/fisiologia , Animais , Anticolesterolemiantes/farmacologia , Chlorocebus aethiops , Cães , Células Madin Darby de Rim Canino , Células Vero , Replicação Viral/efeitos dos fármacos
11.
Viruses ; 4(10): 2317-39, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-23202466

RESUMO

Junín virus, the etiological agent of Argentine hemorrhagic fever, causes significant morbidity and mortality. The virus is spread through the aerosolization of host rodent excreta and endemic to the humid pampas of Argentina. Recently, significant progress has been achieved with the development of new technologies (e.g. reverse genetics) that have expanded knowledge about the pathogenesis and viral replication of Junín virus. We will review the pathogenesis of Junín virus in various animal models and the role of innate and adaptive immunity during infection. We will highlight current research regarding the role of molecular biology of Junín virus in elucidating virus attenuation. We will also summarize current knowledge on Junín virus pathogenesis focusing on the recent development of vaccines and potential therapeutics.


Assuntos
Infecções por Arenaviridae/imunologia , Vírus Junin/patogenicidade , Replicação Viral , Imunidade Adaptativa , Animais , Anticorpos Antivirais , Infecções por Arenaviridae/patologia , Infecções por Arenaviridae/prevenção & controle , Infecções por Arenaviridae/virologia , Modelos Animais de Doenças , Transmissão de Doença Infecciosa/prevenção & controle , Humanos , Imunidade Inata , Vírus Junin/imunologia , Vírus Junin/fisiologia , Glicoproteínas de Membrana/imunologia , Roedores/virologia , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Internalização do Vírus
12.
J Virol ; 86(20): 11301-10, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22875974

RESUMO

Arenaviruses are responsible for acute hemorrhagic fevers with high mortality and pose significant threats to public health and biodefense. These enveloped negative-sense RNA viruses replicate in the cell cytoplasm and express four proteins. To better understand how these proteins insinuate themselves into cellular processes to orchestrate productive viral replication, we have identified and characterized novel cytosolic structures involved in arenavirus replication and transcription. In cells infected with the nonpathogenic Tacaribe virus or the attenuated Candid#1 strain of Junín virus, we find that newly synthesized viral RNAs localize to cytosolic puncta containing the nucleoprotein (N) of the virus. Density gradient centrifugation studies reveal that these replication-transcription complexes (RTCs) are associated with cellular membranes and contain full-length genomic- and antigenomic-sense RNAs. Viral mRNAs segregate at a higher buoyant density and are likewise scant in immunopurified RTCs, consistent with their translation on bulk cellular ribosomes. In addition, confocal microscopy analysis reveals that RTCs contain the lipid phosphatidylinositol-4-phosphate and proteins involved in cellular mRNA metabolism, including the large and small ribosomal subunit proteins L10a and S6, the stress granule protein G3BP1, and a subset of translation initiation factors. Elucidating the structure and function of RTCs will enhance our understanding of virus-cell interactions that promote arenavirus replication and mitigate against host cell immunity. This knowledge may lead to novel intervention strategies to limit viral virulence and pathogenesis.


Assuntos
Infecções por Arenaviridae/virologia , Arenavirus do Novo Mundo/fisiologia , Citosol/virologia , Vírus Junin/fisiologia , RNA Viral/biossíntese , Replicação Viral , Animais , Arenavirus do Novo Mundo/patogenicidade , Membrana Celular/virologia , Centrifugação com Gradiente de Concentração , Chlorocebus aethiops , Citosol/ultraestrutura , Vírus Junin/patogenicidade , Nucleoproteínas , Fosfatos de Fosfatidilinositol/análise , RNA Viral/genética , Transcrição Gênica , Células Vero
13.
J Virol ; 84(12): 6119-29, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20392854

RESUMO

The arenavirus envelope glycoprotein (GPC) initiates infection in the host cell through pH-induced fusion of the viral and endosomal membranes. As in other class I viral fusion proteins, this process proceeds through a structural reorganization in GPC in which the ectodomain of the transmembrane fusion subunit (G2) engages the host cell membrane and subsequently refolds to form a highly stable six-helix bundle structure that brings the two membranes into apposition for fusion. Here, we describe a G2-directed monoclonal antibody, F100G5, that prevents membrane fusion by binding to an intermediate form of the protein on the fusion pathway. Inhibition of syncytium formation requires that F100G5 be present concomitant with exposure of GPC to acidic pH. We show that F100G5 recognizes neither the six-helix bundle nor the larger trimer-of-hairpins structure in the postfusion form of G2. Rather, Western blot analysis using recombinant proteins and a panel of alanine-scanning GPC mutants revealed that F100G5 binding is dependent on an invariant lysine residue (K283) near the N terminus of G2, in the so-called fusion peptide that inserts into the host cell membrane during the fusion process. The F100G5 epitope is located in the internal segment of the bipartite GPC fusion peptide, which also contains four conserved cysteine residues, raising the possibility that this fusion peptide may be highly structured. Collectively, our studies indicate that F100G5 identifies an on-path intermediate form of GPC. Binding to the transiently exposed fusion peptide may interfere with G2 insertion into the host cell membrane. Strategies to effectively target fusion peptide function in the endosome may lead to novel classes of antiviral agents.


Assuntos
Anticorpos/farmacologia , Glicoproteínas/imunologia , Vírus Junin/fisiologia , Fusão de Membrana/efeitos dos fármacos , Proteínas Virais de Fusão/imunologia , Sequência de Aminoácidos , Animais , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/genética , Concentração de Íons de Hidrogênio , Vírus Junin/química , Vírus Junin/efeitos dos fármacos , Vírus Junin/genética , Dados de Sequência Molecular , Alinhamento de Sequência , Células Vero , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética
14.
Virus Res ; 145(1): 166-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19595723

RESUMO

In this paper we demonstrate that infection of cell cultures with the arenavirus Junín (JUNV), agent of the argentine haemorrhagic fever, leads to the activation of PI3K/Akt signalling pathway. Phosphorylation of Akt occurs early during JUNV infection of Vero cells and is blocked by the PI3K inhibitor, Ly294002. Infection of cells with UV-irradiated JUNV redeemed the pattern of stimulation observed for infectious virus indicating that an early stage of multiplication cycle would be enough to trigger activation. Treatment of cells with chlorpromazine abrogated phosphorylation of Akt upon JUNV infection suggesting virus internalization as responsible for activation. Inhibition of Akt phosphorylation by Ly294002 impaired viral protein synthesis and expression leading to a reduced infectious virus yield without blocking the onset of persistent stage of infection. This impairment is linked to a reduced amount of virus bound to cells probably due to a blockage on the recycling of transferrin cell-receptor, employed by the virus to adsorb to the cell surface. Early Akt activation was also observed in BHK-21 and A549 JUNV infected cells suggesting an important role of PI3K/Akt signalling in JUNV multiplication in vitro.


Assuntos
Febre Hemorrágica Americana/metabolismo , Vírus Junin/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Cromonas/farmacologia , Cricetinae , Inibidores Enzimáticos/farmacologia , Febre Hemorrágica Americana/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus Junin/efeitos dos fármacos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Células Vero , Internalização do Vírus
15.
J Virol ; 81(8): 4331-7, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17267481

RESUMO

The stable signal peptide (SSP) of the GP-C envelope glycoprotein of the Junín arenavirus plays a critical role in trafficking of the GP-C complex to the cell surface and in its membrane fusion activity. SSP therefore may function on both sides of the lipid membrane. In this study, we have investigated the membrane topology of SSP by confocal microscopy of cells treated with the detergent digitonin to selectively permeabilize the plasma membrane. By using an affinity tag to mark the termini of SSP in the properly assembled GP-C complex, we find that both the N and C termini reside in the cytosol. Thus, SSP adopts a bitopic topology in which the C terminus is translocated from the lumen of the endoplasmic reticulum to the cytoplasm. This model is supported by (i) the presence of two conserved hydrophobic regions in SSP (hphi1 and hphi2) and (ii) our previous demonstration that lysine-33 in the ectodomain loop is essential for pH-dependent membrane fusion. Moreover, we demonstrate that the introduction of a charged side chain or single amino acid deletion in the membrane-spanning hphi2 region significantly diminishes SSP association in the GP-C complex and abolishes membrane fusion activity. Taken together, our results suggest that bitopic membrane insertion of SSP is centrally important in the assembly and function of the tripartite GP-C complex.


Assuntos
Membrana Celular/química , Membrana Celular/virologia , Citoplasma/química , Vírus Junin/química , Sinais Direcionadores de Proteínas , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Fusão Celular , Detergentes/farmacologia , Digitonina/farmacologia , Interações Hidrofóbicas e Hidrofílicas , Vírus Junin/fisiologia , Microscopia Confocal , Modelos Moleculares , Dados de Sequência Molecular , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus/fisiologia
16.
Virology ; 359(1): 72-81, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17045626

RESUMO

The arenavirus envelope glycoprotein (GP-C) retains a cleaved and stable signal peptide (SSP) as an essential subunit of the mature complex. This 58-amino-acid residue peptide serves as a signal sequence and is additionally required to enable transit of the assembled GP-C complex to the Golgi, and for pH-dependent membrane fusion activity. We have investigated the C-terminal region of the Junín virus SSP to study the role of the cellular signal peptidase (SPase) in generating SSP. Site-directed mutagenesis at the cleavage site (positions -1 and -3) reveals a pattern of side-chain preferences consistent with those of SPase. Although position -2 is degenerate for SPase cleavage, this residue in the arenavirus SSP is invariably a cysteine. In the Junín virus, this cysteine is not involved in disulfide bonding. We show that replacement with alanine or serine is tolerated for SPase cleavage but prevents the mutant SSP from associating with GP-C and enabling transport to the cell surface. Conversely, an arginine mutation at position -1 that prevents SPase cleavage is fully compatible with GP-C-mediated membrane fusion activity when the mutant SSP is provided in trans. These results point to distinct roles of SSP sequences in SPase cleavage and GP-C biogenesis. Further studies of the unique structural organization of the GP-C complex will be important in identifying novel opportunities for antiviral intervention against arenaviral hemorrhagic disease.


Assuntos
Vírus Junin/fisiologia , Proteínas de Membrana/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Chlorocebus aethiops , Vírus Junin/genética , Fusão de Membrana , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Precursores de Proteínas/química , Precursores de Proteínas/imunologia , Precursores de Proteínas/metabolismo , Sinais Direcionadores de Proteínas/genética , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
17.
Virology ; 343(2): 267-74, 2005 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-16169032

RESUMO

The G2 fusion subunit of the Junín virus envelope glycoprotein GP-C contains two hydrophobic heptad-repeat regions that are postulated to form a six-helix bundle structure required for the membrane fusion activity of Class I viral fusion proteins. We have investigated the role of these heptad-repeat regions and, specifically, the importance of the putative interhelical a and d position sidechains by using alanine-scanning mutagenesis. All the mutant glycoproteins were expressed and transported to the cell surface. Proteolytic maturation at the subtilisin kexin isozyme-1/site-1-protease (SKI-1/S1P) cleavage site was observed in all but two of the mutants. Among the adequately cleaved mutant glycoproteins, four positions in the N-terminal region (I333, L336, L347 and L350) and two positions in the C-terminal region (R392 and W395) were shown to be important determinants of cell-cell fusion. Taken together, our results indicate that alpha-helical coiled-coil structures are likely critical in promoting arenavirus membrane fusion. These findings support the inclusion of the arenavirus GP-C among the Class I viral fusion proteins and suggest pharmacologic and immunologic strategies for targeting arenavirus infection and hemorrhagic fever.


Assuntos
Vírus Junin/genética , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Humanos , Concentração de Íons de Hidrogênio , Vírus Junin/patogenicidade , Vírus Junin/fisiologia , Fusão de Membrana , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Insercional , Estrutura Terciária de Proteína , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas do Envelope Viral/química , Proteínas Virais de Fusão/química
18.
J Gen Virol ; 86(Pt 5): 1475-1479, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831960

RESUMO

Junin virus (JUNV), the causative agent of Argentine haemorrhagic fever, is a human pathogen that naturally enters the body through the epithelial cells of the respiratory and digestive tracts. The interaction of JUNV with two types of polarized epithelial cultures, Vero C1008 and A549, was investigated. Radioactive virus-binding assays showed that JUNV infects polarized lines preferentially through the apical surface. High-level expression of viral nucleoprotein was detected in polarized cell lines infected through the apical domain. Virus production from apical media was about 100-fold higher than that found into the basolateral medium. Confocal-immunofluorescence analysis revealed high-level expression of glycoprotein at the apical-membrane surface. Disruption of the microtubule network by colchicine impaired JUNV vectorial release. This is the first study to analyse the interaction between a member of the virus family Arenaviridae and polarized epithelial cells, showing preferential entry and release from the apical plasma membrane.


Assuntos
Células Epiteliais/virologia , Vírus Junin/fisiologia , Animais , Membrana Celular/química , Polaridade Celular , Células Cultivadas , Chlorocebus aethiops , Expressão Gênica , Humanos , Microscopia Confocal , Proteínas do Nucleocapsídeo/análise , Proteínas do Nucleocapsídeo/biossíntese , Células Vero
19.
J Clin Virol ; 32(4): 286-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15780806

RESUMO

BACKGROUND: Junin virus (JV), a member of the Arenaviridae family, is the etiological agent of Argentine hemorrhagic fever (AHF). A low pH-pulse, induces fusion of Vero cells infected with JV to form syncytia, whose production can be inhibited by neutralizing antibodies against the JV major glycoprotein. OBJECTIVES: To characterize the existence of an antifusogenic activity present in sera obtained from natural infections of AHF over a 20-year period and to study both the fusogenic activity of one pathogenic and two attenuated strains of JV in Vero cells, at different pH. The study sample consisted of sera obtained from two provinces in the Argentine Republic. Vero cells grown in monolayers, were infected with different strains of JV and a 2 h pulse, at different pH, was performed. Syncytium production was evaluated 12 h later, after staining with Giemsa. Neutralization tests against the attenuated strain XJCl3 were carried out and the antifusogenic activity of immunosera was studied by incubating serum with JV-infected Vero cells. Also the fusion activity in Vero cells infected with three JV strains was assayed. RESULTS AND CONCLUSIONS: A pathogenic strain XJ exhibited the highest fusogenic activity at pH 5. Syncytium formation was prevented by patients' sera obtained from different geographical locations, independently of time of infection. However, when Vero cells were infected with XJ, a significant reduction of syncytium production was observed, though the level of inhibition was lower than that detected in other JV strains-infected cells. These results could be explained by the existence of a conserved domain on JV proteins and also antigenic heterogeneity among strains.


Assuntos
Fusão Celular , Febre Hemorrágica Americana/imunologia , Soros Imunes/farmacologia , Vírus Junin/fisiologia , Animais , Especificidade de Anticorpos , Linhagem Celular Tumoral , Chlorocebus aethiops , Células Gigantes/efeitos dos fármacos , Células Gigantes/virologia , Febre Hemorrágica Americana/sangue , Humanos , Concentração de Íons de Hidrogênio , Vírus Junin/imunologia , Vírus Junin/patogenicidade , Células Vero
20.
Arch Virol ; 146(2): 251-63, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11315636

RESUMO

The role of vacuolar-proton ATPase (V-H+ ATPAse) on Junin virus (JV) replication was evaluated by analyzing the effect of specific inhibitors of the enzyme activity on different steps of virus multiplication cycle. The presence of the macrolide antibiotics bafilomycin A1 and concanamycin A during the first two hours of infection caused a significant reduction of extracellular infectious virus production and viral protein expression in Vero and BHK-21 cells. The inhibitory action of the compounds was mainly exerted at an early stage of the JV multiplication cycle, without affecting virus attachment to the cell but preventing virus penetration. A correlation between the inhibitory action of the compounds on intracellular compartments acidification and the reduction of JV yield was observed. The addition of concanamycin A at different times after infection indicated that the compound also interferes with the release of infectious particles to the extracellular medium. Although, intracellular transport of JV glycoproteins to the cell membrane, seems not to be affected as revealed by immunofluorescence staining. The results confirm that JV enters into the cell through the endocytic pathway as previously suggested by using lysosomotropic compounds.


Assuntos
Antibacterianos/farmacologia , Inibidores Enzimáticos/farmacologia , Vírus Junin/fisiologia , Macrolídeos , ATPases Translocadoras de Prótons/metabolismo , ATPases Vacuolares Próton-Translocadoras , Replicação Viral , Adsorção , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Cricetinae , Técnica Indireta de Fluorescência para Anticorpo , Transporte Proteico/efeitos dos fármacos , ATPases Translocadoras de Prótons/antagonistas & inibidores , Células Vero , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA