Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Virol ; 94(8)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-31996435

RESUMO

Argentine hemorrhagic fever is a potentially lethal disease that is caused by Junin virus (JUNV). There are currently around 5 million individuals at risk of infection within regions of endemicity in Argentina. The live attenuated vaccine strain Candid #1 (Can) is approved for use in regions of endemicity and has substantially decreased the number of annual Argentine hemorrhagic fever (AHF) cases. The glycoprotein (GPC) gene is primarily responsible for attenuation of the Can strain, and we have shown that the absence of an N-linked glycosylation motif in the subunit G1 of the glycoprotein complex of Can, which is otherwise present in the wild-type pathogenic JUNV, causes GPC retention in the endoplasmic reticulum (ER). Here, we show that Can GPC aggregates in the ER of infected cells, forming incorrect cross-chain disulfide bonds, which results in impaired GPC processing into G1 and G2. The GPC fails to cleave into its G1 and G2 subunits and is targeted for degradation within lysosomes. Cells infected with the wild-type Romero (Rom) strain do not produce aggregates that are observed in Can infection, and the stress on the ER remains minimal. While the mutation of the N-linked glycosylation motif (T168A) is primarily responsible for the formation of aggregates, other mutations within G1 that occurred earlier in the passage history of the Can strain also contribute to aggregation of the GPC within the ER.IMPORTANCE The development of vaccines and therapeutics to combat viral hemorrhagic fevers remains a top priority within the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. The Can strain, derived from the pathogenic XJ strain of JUNV, has been demonstrated to be both safe and protective against AHF. While the vaccine strain is approved for use in regions of endemicity within Argentina, the mechanisms of Can attenuation have not been elucidated. A better understanding of the viral genetic determinants of attenuation will improve our understanding of the mechanisms contributing to disease pathogenesis and provide critical information for the rational design of live attenuated vaccine candidates for other viral hemorrhagic fevers.


Assuntos
Estresse do Retículo Endoplasmático/imunologia , Glicoproteínas/imunologia , Vírus Junin/imunologia , Lisossomos/metabolismo , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Animais , Autofagia , Encéfalo/metabolismo , Chlorocebus aethiops , Retículo Endoplasmático/imunologia , Glicoproteínas/genética , Glicosilação , Células HEK293 , Febre Hemorrágica Americana/virologia , Febres Hemorrágicas Virais/prevenção & controle , Humanos , Vírus Junin/genética , Camundongos , Mutação , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
2.
Nat Commun ; 9(1): 1884, 2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29760382

RESUMO

While five arenaviruses cause human hemorrhagic fevers in the Western Hemisphere, only Junin virus (JUNV) has a vaccine. The GP1 subunit of their envelope glycoprotein binds transferrin receptor 1 (TfR1) using a surface that substantially varies in sequence among the viruses. As such, receptor-mimicking antibodies described to date are type-specific and lack the usual breadth associated with this mode of neutralization. Here we isolate, from the blood of a recipient of the live attenuated JUNV vaccine, two antibodies that cross-neutralize Machupo virus with varying efficiency. Structures of GP1-Fab complexes explain the basis for efficient cross-neutralization, which involves avoiding receptor mimicry and targeting a conserved epitope within the receptor-binding site (RBS). The viral RBS, despite its extensive sequence diversity, is therefore a target for cross-reactive antibodies with activity against New World arenaviruses of public health concern.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Arenavirus do Novo Mundo/imunologia , Febre Hemorrágica Americana/prevenção & controle , Fragmentos Fab das Imunoglobulinas/química , Vírus Junin/imunologia , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Arenavirus do Novo Mundo/genética , Sítios de Ligação de Anticorpos , Reações Cruzadas , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Células HEK293 , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , Humanos , Soros Imunes/química , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Vírus Junin/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/imunologia , Receptores da Transferrina/química , Receptores da Transferrina/genética , Receptores da Transferrina/imunologia , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem
3.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070682

RESUMO

The Candid#1 strain of Junín virus was developed using a conventional attenuation strategy of serial passage in nonhost animals and cultured cells. The live-attenuated Candid#1 vaccine is used in Argentina to protect at-risk individuals against Argentine hemorrhagic fever, but it has not been licensed in the United States. Recent studies have revealed that Candid#1 attenuation is entirely dependent on a phenylalanine-to-isoleucine substitution at position 427 in the fusion subunit (GP2) of the viral envelope glycoprotein complex (GPC), thereby raising concerns regarding the potential for reversion to virulence. In this study, we report the identification and characterization of an intragenic epistatic interaction between the attenuating F427I mutation in GP2 and a lysine-to-serine mutation at position 33 in the stable signal peptide (SSP) subunit of GPC, and we demonstrate the utility of this interaction in creating an evolutionary barrier against reversion to the pathogenic genotype. In the presence of the wild-type F427 residue, the K33S mutation abrogates the ability of ectopically expressed GPC to mediate membrane fusion at endosomal pH. This defect is rescued by the attenuating F427I mutation. We show that the recombinant Candid#1 (rCan) virus bearing K33S GPC is viable and retains its attenuated genotype under cell culture conditions that readily select for reversion in the parental rCan virus. If back-mutation to F427 offers an accessible pathway to increase fitness in rCan, reversion in K33S-GPC rCan is likely to be lethal. The epistatic interaction between K33S and F427I thus may minimize the likelihood of reversion and enhance safety in a second-generation Candid#1 vaccine.IMPORTANCE The live-attenuated Candid#1 vaccine strain of Junín virus is used to protect against Argentine hemorrhagic fever. Recent findings that a single missense mutation in the viral envelope glycoprotein complex (GPC) is responsible for attenuation raise the prospect of facile reversion to pathogenicity. Here, we characterize a genetic interaction between GPC subunits that evolutionarily forces retention of the attenuating mutation. By incorporating this secondary mutation into Candid#1 GPC, we hope to minimize the likelihood of reversion and enhance safety in a second-generation Candid#1 vaccine. A similar approach may guide the design of live-attenuated vaccines against Lassa and other arenaviral hemorrhagic fevers.


Assuntos
Epistasia Genética , Glicoproteínas/genética , Vírus Junin/genética , Vírus Junin/imunologia , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Animais , Chlorocebus aethiops , Evolução Molecular , Genótipo , Febre Hemorrágica Americana/prevenção & controle , Humanos , Vírus Junin/metabolismo , Vírus Junin/patogenicidade , Fusão de Membrana , Mutação , Vacinas Atenuadas/genética , Células Vero , Virulência , Internalização do Vírus
4.
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
5.
J Gen Virol ; 97(4): 855-866, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26769540

RESUMO

The Arenaviridae is a diverse and growing family of viruses that already includes more than 25 distinct species. While some of these viruses have a significant impact on public health, others appear to be non-pathogenic. At present little is known about the host cell responses to infection with different arenaviruses, particularly those found in the New World; however, apoptosis is known to play an important role in controlling infection of many viruses. Here we show that infection with Tacaribe virus (TCRV), which is widely considered the prototype for non-pathogenic arenaviruses, leads to stronger induction of apoptosis than does infection with its human-pathogenic relative Junín virus. TCRV-induced apoptosis occurred in several cell types during late stages of infection and was shown to be caspase-dependent, involving the activation of caspases 3, 7, 8 and 9. Further, UV-inactivated TCRV did not induce apoptosis, indicating that the activation of this process is dependent on active viral replication/transcription. Interestingly, when apoptosis was inhibited, growth of TCRV was not enhanced, indicating that apoptosis does not have a direct negative effect on TCRV infection in vitro. Taken together, our data identify and characterize an important virus-host cell interaction of the prototypic, non-pathogenic arenavirus TCRV, which provides important insight into the growing field of arenavirus research aimed at better understanding the diversity in responses to different arenavirus infections and their functional consequences.


Assuntos
Arenavirus do Novo Mundo/genética , Caspases/genética , Interações Hospedeiro-Patógeno , Macrófagos/virologia , Replicação Viral/genética , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/genética , Apoptose/imunologia , Arenavirus do Novo Mundo/efeitos dos fármacos , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/efeitos da radiação , Camptotecina/farmacologia , Caspases/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação da Expressão Gênica , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Vírus Junin/genética , Vírus Junin/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/imunologia , Cultura Primária de Células , Transdução de Sinais , Tubulina (Proteína)/genética , Tubulina (Proteína)/imunologia , Raios Ultravioleta , Células Vero , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação
6.
Appl Microbiol Biotechnol ; 100(1): 125-33, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26386688

RESUMO

Two new vaccine candidates against dengue virus (DENV) infection were generated by fusing the coding sequences of the self-budding Z protein from Junin virus (Z-JUNV) to those of two cryptic peptides (Z/DENV-P1 and Z/DENV-P2) conserved on the envelope protein of all serotypes of DENV. The capacity of these chimeras to generate virus-like particles (VLPs) and to induce virus-neutralizing antibodies in mice was determined. First, recombinant proteins that displayed reactivity with a Z-JUNV-specific serum by immunofluorescence were detected in HEK-293 cells transfected with each of the two plasmids and VLP formation was also observed by transmission electron microscopy. Next, we determined the presence of antibodies against the envelope peptides of DENV in the sera of immunized C57BL/6 mice. Results showed that those animals that received Z/DENV-P2 DNA coding sequences followed by a boost with DENV-P2 synthetic peptides elicited significant specific antibody titers (≥6.400). Finally, DENV plaque-reduction neutralization tests (PRNT) were performed. Although no significant protective effect was observed when using sera of Z/DENV-P1-immunized animals, antibodies raised against vaccine candidate Z/DENV-P2 (diluted 1:320) were able to reduce in over 50 % the number of viral plaques generated by infectious DENV particles. This reduction was comparable to that of the 4G2 DENV-specific monoclonal cross-reactive (all serotypes) neutralizing antibody. We conclude that Z-JUNV-VLP is a valid carrier to induce antibody-mediated immune responses in mice and that Z/DENV-P2 is not only immunogenic but also protective in vitro against infection of cells with DENV, deserving further studies. On the other side, DENV's fusion peptide-derived chimera Z/DENV-P1 did not display similar protective properties.


Assuntos
Anticorpos Neutralizantes/sangue , Vacinas contra Dengue/imunologia , Vírus da Dengue/genética , Portadores de Fármacos , Vírus Junin/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Camundongos Endogâmicos C57BL , Testes de Neutralização , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral
7.
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
8.
J Virol ; 85(24): 13457-62, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21976641

RESUMO

Candid#1 (Cd1) is an attenuated vaccine strain of Junin virus, the causative agent of Argentine hemorrhagic fever. Although several substitutions are present in Cd1, their importance for attenuation has not been established. We functionally characterized the substitutions present in the Cd1 glycoprotein (GP) and identified F427I in the transmembrane domain of the GP2 subunit as reducing infectivity in a reconstituted viral system. We further showed that this phenotype derives from the destabilization of the GP metastable conformation. Lastly, we identified an increased dependence of Cd1 GP on human transferrin receptor type 1 (hTfR-1) for entry, which may affect the tropism of the attenuated strain in vivo.


Assuntos
Antígenos CD/metabolismo , Vírus Junin/patogenicidade , Glicoproteínas de Membrana/metabolismo , Receptores da Transferrina/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Fatores de Virulência/metabolismo , Internalização do Vírus , Substituição de Aminoácidos , Animais , Linhagem Celular , Humanos , Vírus Junin/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Conformação Proteica , Vacinas Atenuadas/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Fatores de Virulência/química , Fatores de Virulência/genética
9.
J Virol ; 85(1): 112-22, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980515

RESUMO

The Arenaviridae are a diverse and globally distributed collection of viruses that are maintained primarily by rodent reservoirs. Junin virus (JUNV) and Lassa virus (LASV) can both cause significant outbreaks of severe and often fatal human disease throughout their respective areas of endemicity. In an effort to improve upon the existing live attenuated JUNV Candid1 vaccine, we generated a genetically homogenous stock of this virus from cDNA copies of the virus S and L segments by using a reverse genetics system. Further, these cDNAs were used in combination with LASV cDNAs to successfully generate two recombinant Candid1 JUNV/LASV chimeric viruses (via envelope glycoprotein [GPC] exchange). It was found that while the GPC extravirion domains were readily exchangeable, homologous stable signal peptide (SSP) and G2 transmembrane and cytoplasmic tail domains were essential for correct GPC maturation and production of infectious chimeric viruses. The switching of the JUNV and LASV G1/G2 ectodomains within the Candid1 vaccine background did not alter the attenuated phenotype of the vaccine strain in a lethal mouse model. These recombinant chimeric viruses shed light on the fundamental requirements of arenavirus GPC maturation and may serve as a strategy for the development of bivalent JUNV and LASV vaccine candidates.


Assuntos
Glicoproteínas/genética , Vírus Junin/genética , Vírus Lassa/genética , Recombinação Genética , Proteínas do Envelope Viral/genética , Vacinas Virais , Animais , Infecções por Arenaviridae/mortalidade , Infecções por Arenaviridae/prevenção & controle , Infecções por Arenaviridae/virologia , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Vírus Junin/metabolismo , Vírus Junin/patogenicidade , Vírus Lassa/metabolismo , Vírus Lassa/patogenicidade , Camundongos , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
10.
J Biol Chem ; 286(8): 6192-200, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21159779

RESUMO

Arenaviruses are responsible for acute hemorrhagic fevers worldwide and are recognized to pose significant threats to public health and biodefense. Small molecule compounds have recently been discovered that inhibit arenavirus entry and protect against lethal infection in animal models. These chemically distinct inhibitors act on the tripartite envelope glycoprotein (GPC) through its unusual stable signal peptide subunit to stabilize the complex against pH-induced activation of membrane fusion in the endosome. Here, we report the production and characterization of the intact transmembrane GPC complex of Junín arenavirus and its interaction with these inhibitors. The solubilized GPC is antigenically indistinguishable from the native protein and forms a homogeneous trimer in solution. When reconstituted into a lipid bilayer, the purified complex interacts specifically with its cell-surface receptor transferrin receptor-1. We show that small molecule entry inhibitors specific to New World or Old World arenaviruses bind to the membrane-associated GPC complex in accordance with their respective species selectivities and with dissociation constants comparable with concentrations that inhibit GPC-mediated membrane fusion. Furthermore, competitive binding studies reveal that these chemically distinct inhibitors share a common binding pocket on GPC. In conjunction with previous genetic studies, these findings identify the pH-sensing interface of GPC as a highly vulnerable target for antiviral intervention. This work expands our mechanistic understanding of arenavirus entry and provides a foundation to guide the development of small molecule compounds for the treatment of arenavirus hemorrhagic fevers.


Assuntos
Antígenos CD/metabolismo , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Vírus Junin/metabolismo , Receptores da Transferrina/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/metabolismo , Animais , Antígenos CD/genética , Antivirais/química , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/metabolismo , Chlorocebus aethiops , Humanos , Vírus Junin/genética , Receptores da Transferrina/genética , Células Vero , Proteínas do Envelope Viral/genética , Internalização do Vírus/efeitos dos fármacos
11.
Rev. Soc. Bras. Med. Trop ; 43(3): 229-233, May-June 2010. ilus, mapas, tab
Artigo em Inglês | LILACS | ID: lil-548514

RESUMO

INTRODUCTION: Arenavirus hemorrhagic fever is a severe emerging disease. METHODS: Considering that the levels of antibodies against arenavirus in the Brazilian population are completely unknown, we have standardized an ELISA test for detecting IgG antibodies using a recombinant nucleoprotein from the Junin virus as the antigen. This protein was obtained by inserting the gene of the Junin virus nucleoprotein into the genome of Autographa californica nucleopolyhedrovirus, using the Bac-to-Bac baculovirus expression system. This recombinant baculovirus was used to infect S. frugiperda cells (SF9). RESULTS: The infection resulted in synthesis of high concentrations of recombinant protein. This protein was detected on 12.5 percent polyacrylamide gel and by means of Western blot. Using the standardized ELISA test, 343 samples from the population of Nova Xavantina were analyzed. We observed that 1.4 percent of the serum samples (five samples) presented antibody titers against arenavirus. CONCLUSIONS: These results show the population studied may present exposure to arenavirus infection.


INTRODUÇÃO: A febre hemorrágica por Arenavirus é uma severa doença emergente. MÉTODOS: Considerando que os níveis de anticorpos contra Arenavirus na população brasileira é totalmente desconhecido, nos padronizamos um teste de ELISA para detecção de anticorpos IgG usando uma nucleoproteína recombinante do vírus Junin como antígeno. Esta proteína foi obtida pela inserção do gene da nucleoproteína do vírus Junin no genoma do vírus Autographa californica nucleopolyhedrovirus, utilizando o sistema de expressão em Baculovírus, Bac-To-Bac. Este baculovirus recombinante foi utilizado para infecção de células de S. frugiperda (Sf9). RESULTADOS: A infecção resultou na produção de altas concentrações de proteína recombinante. Esta proteína foi detectada em gel de poliacrilamida 12,5 por cento, e em Western blot. Utilizando o teste de ELISA padronizado, foram analizadas 343 amostras provenientes da população de Nova Xavantina. Observamos que 1,4 por cento dos soros (5 amostras) apresentavam títulos de anticorpos contra arenavírus. CONCLUSÕES: Estes resultados sugerem que a população estudada pode estar sendo exposta a infecções por arenavírus.


Assuntos
Adulto , Feminino , Humanos , Masculino , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Infecções por Arenaviridae/diagnóstico , Arenavirus/imunologia , Ensaio de Imunoadsorção Enzimática/normas , Vírus Junin/imunologia , Arenavirus/genética , Brasil , Ensaio de Imunoadsorção Enzimática/métodos , Imunoglobulina G/sangue , Vírus Junin/genética , Nucleoproteínas/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
12.
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
13.
Virus Res ; 143(1): 106-13, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19463727

RESUMO

Our previous studies reported the inhibitory action against arenaviruses of antiretroviral zinc finger-reactive compounds provided by the National Cancer Institute (USA). These compounds were able to inactivate virions as well as to reduce virus yields from infected cells. Here, the inactivation of the arenavirus Junín (JUNV), agent of Argentine hemorrhagic fever, by the aromatic disulfide NSC20625 was analyzed. The treatment of purified JUNV with this compound eliminated infectivity apparently through irreversible modifications in the matrix Z protein detected by: (a) alterations in the electrophoretic migration profile of Z under non-reducing conditions; (b) an electrodense labeling in the internal layer beneath the envelope and around the matrix Z protein, in negatively stained preparations; (c) changes in the subcellular localization of Z in cells transfected with a recombinant fusion protein JUNVZ-eGFP. The infection of Vero cells with JUNV inactivated particles was blocked at the uncoating of viral nucleocapsid from endosomes, providing new evidence for a functional role of Z in this stage of arenavirus cycle. Furthermore, the inactivated JUNV particles retained the immunoreactivity of the surface glycoprotein GP1 suggesting that this disulfide may be useful in the pursuit of an inactivating agent to obtain a vaccine antigen or diagnostic tool.


Assuntos
Infecções por Arenaviridae/tratamento farmacológico , Compostos Azo/farmacologia , Dissulfetos/farmacologia , Guanidinas/farmacologia , Vírus Junin/efeitos dos fármacos , Vírion/efeitos dos fármacos , Dedos de Zinco , Animais , Fármacos Anti-HIV/farmacologia , Infecções por Arenaviridae/metabolismo , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Chlorocebus aethiops , Proteínas de Fluorescência Verde , Vírus Junin/genética , Microscopia Eletrônica de Transmissão , Proteínas do Nucleocapsídeo/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Células Vero , Vírion/ultraestrutura , Inativação de Vírus
14.
J Virol ; 83(9): 4121-6, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19224989

RESUMO

The mature arenavirus envelope glycoprotein GPC is a tripartite complex comprising a stable signal peptide (SSP) in addition to the receptor-binding (G1) and transmembrane fusion (G2) subunits. We have shown previously that SSP is a key element in GPC-mediated membrane fusion, and that GPC sensitivity to acidic pH is modulated in part through the lysine residue at position 33 in the ectodomain loop of SSP (J. York and J. H. Nunberg, J. Virol. 80:7775-7780, 2006). A glutamine substitution at this position stabilizes the native GPC complex and thereby prevents the induction of pH-dependent membrane fusion. In efforts to identify the intersubunit interactions of K33, we performed alanine-scanning mutagenesis at charged residues in the membrane-proximal ectodomain of G2 and determined the ability of these mutations to rescue the fusion deficiency in K33Q GPC. Four second-site mutations that specifically complement K33Q were identified (D400A, E410A, R414A, and K417A). Moreover, complementation was also observed at three hydrophobic positions in the membrane-spanning domain of G2 (F427, W428, and F438). Interestingly, all of the complementing mutations restored wild-type pH sensitivity to the K33Q mutant, while none themselves affected the pH of membrane fusion. Our studies demonstrate a specific interaction between SSP and G2 that is involved in priming the native GPC complex for pH-induced membrane fusion. Importantly, this pH-dependent interaction has been shown to be vulnerable to small-molecule compounds that stabilize the native complex and prevent the activation of membrane fusion. A detailed mechanistic understanding of the control of GPC-mediated membrane fusion will be important in guiding the development of effective therapeutics against arenaviral hemorrhagic fever.


Assuntos
Glicoproteínas/metabolismo , Vírus Junin/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/genética , Concentração de Íons de Hidrogênio , Vírus Junin/genética , Dados de Sequência Molecular , Mutação/genética , Peptídeos/genética , Peptídeos/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Alinhamento de Sequência , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
15.
J Virol ; 81(24): 13385-91, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17928348

RESUMO

The envelope glycoprotein of the Junín arenavirus (GP-C) mediates entry into target cells through a pH-dependent membrane fusion mechanism. Unlike other class I viral fusion proteins, the mature GP-C complex retains a cleaved, 58-amino-acid signal peptide (SSP) as an essential subunit, required both for trafficking of GP-C to the cell surface and for the activation of membrane fusion. SSP has been shown to associate noncovalently in GP-C via the cytoplasmic domain (CTD) of the transmembrane fusion subunit G2. In this report we investigate the molecular basis for this intersubunit interaction. We identify an invariant series of six cysteine and histidine residues in the CTD of G2 that is essential for incorporation of SSP in the GP-C complex. Moreover, we show that a CTD peptide fragment containing His-447, His-449, and Cys-455 specifically binds Zn(2+) at subnanomolar concentrations. Together, these results suggest a zinc finger-like domain structure in the CTD of G2. We propose that the remaining residues in the series (His-459, Cys-467, and Cys-469) form an intersubunit zinc-binding center that incorporates Cys-57 of SSP. This unusual motif may act to retain SSP in the GP-C complex and position the ectodomain loop of SSP for its role in modulating membrane fusion activity. The unique tripartite organization of GP-C could provide novel molecular targets for therapeutic intervention in arenaviral disease.


Assuntos
Glicoproteínas/metabolismo , Vírus Junin/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Zinco/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/genética , Concentração de Íons de Hidrogênio , Vírus Junin/química , Vírus Junin/genética , Fusão de Membrana , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Dedos de Zinco/genética
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.
J Virol ; 80(15): 7775-80, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16840359

RESUMO

The envelope glycoprotein of the arenaviruses (GP-C) is unusual in that the mature complex retains the cleaved, 58-amino-acid signal peptide. Association of this stable signal peptide (SSP) has been shown to be essential for intracellular trafficking and proteolytic maturation of the GP-C complex. We identify here a specific and previously unrecognized role of SSP in pH-dependent membrane fusion. Amino acid substitutions that alter the positive charge at lysine K33 in SSP affect the ability of GP-C to mediate cell-cell fusion and the threshold pH at which membrane fusion is triggered. Based on the presumed location of K33 at or near the luminal domain of SSP, we postulate that SSP interacts with the membrane-proximal or transmembrane regions of the G2 fusion protein. This unique organization of the GP-C complex may suggest novel strategies for intervention in arenavirus infection.


Assuntos
Glicoproteínas/metabolismo , Vírus Junin/metabolismo , Fusão de Membrana , Sinais Direcionadores de Proteínas/fisiologia , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Fusão Celular , Chlorocebus aethiops , Glicoproteínas/genética , Concentração de Íons de Hidrogênio , Vírus Junin/genética , Lisina/genética , Lisina/metabolismo , Dados de Sequência Molecular , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas do Envelope Viral/genética
18.
Virus Genes ; 32(1): 37-41, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16525733

RESUMO

Junin virus strain Candid #1 was developed as a live attenuated vaccine for Argentine haemorrhagic fever. In this paper, we report the nucleotide sequences of L RNA of Candid #1 and examine the relationship to its more virulent ancestors Junin virus XJ#44 and XJ 13 (prototype) and other closely and distantly related arenaviruses. Comparisons of the nucleotide and amino acid sequences of L and Z genes of Candid #1 and its progenitor strains revealed twelve point mutations in the L polypeptide that are unique to the vaccine strain. These changes could be provisionally associated with the attenuated phenotype. In contrast, Z ORF was completely conserved among all strains.


Assuntos
Vírus Junin/genética , Vírus Junin/imunologia , Animais , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/prevenção & controle , Genoma Viral , Cobaias , Humanos , Vírus Junin/patogenicidade , Camundongos , Dados de Sequência Molecular , Filogenia , Mutação Puntual , RNA Viral/genética , Especificidade da Espécie , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas Virais/genética , Vacinas Virais/genética , Vacinas Virais/imunologia
19.
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
20.
Arch Virol ; 147(8): 1543-57, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12181674

RESUMO

Two Vero cell lines persistently infected with XJCl3 and Cl67 strains of Junin virus and named V3 and V7, respectively, have been characterized with respect to the presence and expression of the nucleoprotein (N) and the glycoprotein precursor (GPC) viral genes. After the acute phase of infection, where a marked CPE and high titers of virus were obtained, JV persistently infected cells became morphologically undistinguishable from Vero cells and virus production dropped to undetectable levels. V3 and V7 were resistant to the superinfection with antigenically related viruses. This fact could not be attributed to the presence of defective interfering particles or non-infectious virus in the supernatant. Expression of N was consistently detected in both cultures and accumulation of two degradation products of N was evident during the late passages. Although no G1 (main surface glycoprotein) expression was observed, a marked fusogenic capacity was detected in both cultures indicating at least, the synthesis of a GPC derived fusogenic glycoprotein. Cell lysates from V3 and V7 subjected to RT-PCR, using specific primers for N gene, or to a nested RT-PCR using specific primers for GPC (G1 region) confirmed the presence of both viral genes. No viral DNA sequences could be detected in JV persistently infected cells.


Assuntos
Antígenos Virais/biossíntese , Vírus Junin/imunologia , Animais , Chlorocebus aethiops , Vírus Junin/genética , Proteínas do Nucleocapsídeo/genética , RNA Viral/análise , Células Vero , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA