Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 98(2): e0197523, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38294249

RESUMO

The highly pathogenic arenavirus, Junín virus (JUNV), expresses three truncated alternative isoforms of its nucleoprotein (NP), i.e., NP53kD, NP47kD, and NP40kD. While both NP47kD and NP40kD have been previously shown to be products of caspase cleavage, here, we show that expression of the third isoform NP53kD is due to alternative in-frame translation from M80. Based on this information, we were able to generate recombinant JUNVs lacking each of these isoforms. Infection with these mutants revealed that, while all three isoforms contribute to the efficient control of caspase activation, NP40kD plays the predominant role. In contrast to full-length NP (i.e., NP65kD), which is localized to inclusion bodies, where viral RNA synthesis takes place, the loss of portions of the N-terminal coiled-coil region in these isoforms leads to a diffuse cytoplasmic distribution and a loss of function in viral RNA synthesis. Nonetheless, NP53kD, NP47kD, and NP40kD all retain robust interferon antagonistic and 3'-5' exonuclease activities. We suggest that the altered localization of these NP isoforms allows them to be more efficiently targeted by activated caspases for cleavage as decoy substrates, and to be better positioned to degrade viral double-stranded (ds)RNA species that accumulate in the cytoplasm during virus infection and/or interact with cytosolic RNA sensors, thereby limiting dsRNA-mediated innate immune responses. Taken together, this work provides insight into the mechanism by which JUNV leverages apoptosis during infection to generate biologically distinct pools of NP and contributes to our understanding of the expression and biological relevance of alternative protein isoforms during virus infection.IMPORTANCEA limited coding capacity means that RNA viruses need strategies to diversify their proteome. The nucleoprotein (NP) of the highly pathogenic arenavirus Junín virus (JUNV) produces three N-terminally truncated isoforms: two (NP47kD and NP40kD) are known to be produced by caspase cleavage, while, here, we show that NP53kD is produced by alternative translation initiation. Recombinant JUNVs lacking individual NP isoforms revealed that all three isoforms contribute to inhibiting caspase activation during infection, but cleavage to generate NP40kD makes the biggest contribution. Importantly, all three isoforms retain their ability to digest double-stranded (ds)RNA and inhibit interferon promoter activation but have a diffuse cytoplasmic distribution. Given the cytoplasmic localization of both aberrant viral dsRNAs, as well as dsRNA sensors and many other cellular components of innate immune activation pathways, we suggest that the generation of NP isoforms not only contributes to evasion of apoptosis but also robust control of the antiviral response.


Assuntos
Caspases , Citoplasma , Febre Hemorrágica Americana , Interações Hospedeiro-Patógeno , Imunidade Inata , Vírus Junin , Nucleoproteínas , Biossíntese de Proteínas , Humanos , Apoptose , Inibidores de Caspase/metabolismo , Caspases/metabolismo , Citoplasma/metabolismo , Citoplasma/virologia , Ativação Enzimática , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , Interferons/genética , Interferons/imunologia , Vírus Junin/genética , Vírus Junin/metabolismo , Vírus Junin/patogenicidade , Nucleoproteínas/biossíntese , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Viral/biossíntese , RNA Viral/genética , Replicação Viral
2.
mBio ; 13(2): e0265021, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35315691

RESUMO

Transmission of the New World hemorrhagic fever arenaviruses Junín virus (JUNV) and Machupo virus (MACV) to humans is facilitated, in part, by the interaction between the arenavirus GP1 glycoprotein and the human transferrin receptor 1 (hTfR1). We utilize a mouse model of live-attenuated immunization with envelope exchange viruses to isolate neutralizing monoclonal antibodies (NAbs) specific to JUNV GP1 and MACV GP1. Structures of two NAbs, termed JUN1 and MAC1, demonstrate that they neutralize through disruption of hTfR1 recognition. JUN1 utilizes a binding mode common to all characterized infection- and vaccine-elicited JUNV-specific NAbs, which involves mimicking hTfR1 binding through the insertion of a tyrosine into the receptor-binding site. In contrast, MAC1 undergoes a tyrosine-mediated mode of antigen recognition distinct from that used by the reported anti-JUNV NAbs and the only other characterized anti-MACV NAb. These data reveal the varied modes of GP1-specific recognition among New World arenaviruses by the antibody-mediated immune response. IMPORTANCE The GP1 subcomponent of the New World arenavirus GP is a primary target of the neutralizing antibody response, which has been shown to be effective in the prevention and treatment of infection. Here, we characterize the structural basis of the antibody-mediated immune response that arises from immunization of mice against Junín virus and Machupo virus, two rodent-borne zoonotic New World arenaviruses. We isolate a panel of GP1-specific monoclonal antibodies that recognize overlapping epitopes and exhibit neutralizing behavior, in vitro. Structural characterization of two of these antibodies indicates that antibody recognition likely interferes with GP1-mediated recognition of the transferrin receptor 1. These data provide molecular-level detail for a key region of vulnerability on the New World arenavirus surface and a blueprint for therapeutic antibody development.


Assuntos
Arenavirus do Novo Mundo , Vírus Junin , Animais , Anticorpos Monoclonais , Anticorpos Neutralizantes , Arenavirus do Novo Mundo/metabolismo , Imunização , Vírus Junin/metabolismo , Camundongos , Receptores da Transferrina , Tirosina
3.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33836604

RESUMO

The COVID-19 pandemic has reemphasized the need to identify safe and scalable therapeutics to slow or reverse symptoms of disease caused by newly emerging and reemerging viral pathogens. Recent clinical successes of monoclonal antibodies (mAbs) in therapy for viral infections demonstrate that mAbs offer a solution for these emerging biothreats. We have explored this with respect to Junin virus (JUNV), an arenavirus classified as a category A high-priority agent and the causative agent of Argentine hemorrhagic fever (AHF). There are currently no Food and Drug Administration-approved drugs available for preventing or treating AHF, although immune plasma from convalescent patients is used routinely to treat active infections. However, immune plasma is severely limited in quantity, highly variable in quality, and poses significant safety risks including the transmission of transfusion-borne diseases. mAbs offer a highly specific and consistently potent alternative to immune plasma that can be manufactured at large scale. We previously described a chimeric mAb, cJ199, that provided protection in a guinea pig model of AHF. To adapt this mAb to a format more suitable for clinical use, we humanized the mAb (hu199) and evaluated it in a cynomolgus monkey model of AHF with two JUNV isolates, Romero and Espindola. While untreated control animals experienced 100% lethality, all animals treated with hu199 at 6 d postinoculation (dpi) survived, and 50% of animals treated at 8 dpi survived. mAbs like hu199 may offer a safer, scalable, and more reproducible alternative to immune plasma for rare viral diseases that have epidemic potential.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Antivirais/farmacologia , Febre Hemorrágica Americana/prevenção & controle , Vírus Junin/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Cobaias , Febre Hemorrágica Americana/sangue , Humanos , Macaca fascicularis
4.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748396

RESUMO

Several clade B New World arenaviruses (NWAs) can cause severe and often fatal hemorrhagic fever, for which preventive and therapeutic measures are severely limited. These NWAs use human transferrin receptor 1 (hTfR1) as a host cell receptor for virus entry. The most prevalent of the pathogenic NWAs is Junín virus (JUNV), the etiological agent of Argentine hemorrhagic fever. Small animal models of JUNV infection are limited because most laboratory rodent species are refractory to disease. Only guinea pigs are known to develop disease following JUNV infection, but the underlying mechanisms are not well characterized. In the present study, we demonstrate marked susceptibility of Hartley guinea pigs to uniformly lethal disease when challenged with as few as 4 PFU of the Romero strain of JUNV. In vitro, we show that infection of primary guinea pig macrophages results in greater JUNV replication compared to infection of hamster or mouse macrophages. We provide evidence that the guinea pig TfR1 (gpTfR1) is the principal receptor for JUNV, while hamster and mouse orthologs fail to support viral entry/infection of pseudotyped murine leukemia viruses expressing pathogenic NWA glycoproteins or JUNV. Together, our results indicate that gpTfR1 serves as the primary receptor for pathogenic NWAs, enhancing viral infection in guinea pigs.IMPORTANCE JUNV is one of five known NWAs that cause viral hemorrhagic fever in humans. Countermeasures against JUNV infection are limited to immunization with the Candid#1 vaccine and immune plasma, which are available only in Argentina. The gold standard small animal model for JUNV infection is the guinea pig. Here, we demonstrate high sensitivity of this species to severe JUNV infection and identify gpTfR1 as the primary receptor. Use of hTfR1 for host cell entry is a feature shared by pathogenic NWAs. Our results show that expression of gpTfR1 or hTfR1 comparably enhances JUNV virus entry/infectivity. Our findings shed light on JUNV infection in guinea pigs as a model for human disease and suggest that similar pathophysiological mechanisms related to iron sequestration during infection and regulation of TfR1 expression may be shared between humans and guinea pigs. A better understanding of the underlying disease process will guide development of new therapeutic interventions.


Assuntos
Vírus Junin/imunologia , Vírus Junin/patogenicidade , Receptores da Transferrina/metabolismo , Animais , Arenavirus/imunologia , Arenavirus/patogenicidade , Células CHO , Chlorocebus aethiops , Cricetulus , Modelos Animais de Doenças , Feminino , Glicoproteínas/metabolismo , Cobaias/imunologia , Cobaias/metabolismo , Células HEK293 , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , Febres Hemorrágicas Virais/imunologia , Febres Hemorrágicas Virais/virologia , Humanos , Vírus Junin/metabolismo , Macrófagos/virologia , Masculino , Receptores da Transferrina/imunologia , Células Vero , Internalização do Vírus , Replicação Viral
5.
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
6.
Methods Mol Biol ; 1604: 209-215, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28986836

RESUMO

Independent expression of the VP40 or Z matrix proteins of filoviruses (marburgviruses and ebolaviruses) and arenaviruses (Lassa fever and Junín), respectively, gives rise to the production and release of virus-like particles (VLPs) that are morphologically identical to infectious virions. We can detect and quantify VLP production and egress in mammalian cells by transient transfection, SDS-PAGE, Western blotting, and live cell imaging techniques such as total internal reflection fluorescence (TIRF) microscopy. Since the VLP budding assay accurately mimics budding of infectious virus, this BSL-2 assay is safe and useful for the interrogation of both viral and host determinants required for budding and can be used as an initial screen to identify and validate small molecule inhibitors of virus release and spread.


Assuntos
Doença pelo Vírus Ebola/metabolismo , Liberação de Vírus/fisiologia , Animais , Arenavirus/genética , Arenavirus/metabolismo , Western Blotting , Ebolavirus/genética , Ebolavirus/metabolismo , Eletroforese em Gel de Poliacrilamida , Filoviridae/genética , Filoviridae/metabolismo , Doença pelo Vírus Ebola/genética , Humanos , Vírus Junin/genética , Vírus Junin/metabolismo , Febre Lassa/virologia , Marburgvirus/genética , Marburgvirus/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Liberação de Vírus/genética
7.
Methods Mol Biol ; 1604: 351-370, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28986848

RESUMO

Host restriction factors are cellular components that interfere with viral multiplication. They are up-regulated and expressed upon viral infection and in consequence their activity is specific. So far several important restriction factors have been described against diverse viruses. The cellular antiviral mechanisms defined include proteins with the ability to interfere with early steps of viral replication and others that have been shown to block viral morphogenesis. However, other strategies by which the antiviral action is exerted still remain elusive. An additional interesting matter is how viruses also developed ways to by-pass these host-specific obstacles. Thus, unusual cell localization or re-localization represents a frequent virus choice to evade the cellular surveillance. In the present chapter, we summarize methods to identify cell restriction factors, their antiviral activity, and possible subcellular locations where their activity can take place.


Assuntos
Febre Hemorrágica com Síndrome Renal/metabolismo , Animais , Dengue/virologia , Vírus da Dengue/metabolismo , Humanos , Vírus Junin/metabolismo , RNA Interferente Pequeno
8.
J Gen Virol ; 94(Pt 10): 2175-2183, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23884367

RESUMO

Junin virus (JUNV) has been identified as the aetiological agent of Argentine haemorrhagic fever (AHF), which is a serious public health problem with approximately 5 million people at risk. It is treated as a potential bioterrorism agent because of its rapid transmission by aerosols. JUNV is a negative-sense ssRNA virus that belongs to the genus Arenavirus within the family Arenaviridae, and its genomic RNA contains two segments encoding four proteins. Among these, the nucleoprotein (NP) has essential roles in viral RNA synthesis and immune suppression, but the molecular mechanisms of its actions are only partially understood. Here, we determined a 2.2 Å crystal structure of the C-terminal domain of JUNV NP. This structure showed high similarity to the Lassa fever virus (LASV) NP C-terminal domain. However, both the structure and function of JUNV NP showed differences compared with LASV NP. This study extends our structural insight into the negative-sense ssRNA virus NPs.


Assuntos
Vírus Junin/química , Vírus Junin/metabolismo , Nucleoproteínas/química , Nucleoproteínas/metabolismo , Sequência de Aminoácidos , Regulação Viral da Expressão Gênica/fisiologia , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Alinhamento de Sequência
9.
PLoS One ; 7(11): e51114, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226473

RESUMO

The membrane-anchored proteins of enveloped viruses form labile spikes on the virion surface, primed to undergo large-scale conformational changes culminating in virus-cell membrane fusion and viral entry. The prefusion form of these envelope glycoproteins thus represents an important molecular target for antiviral intervention. A critical roadblock to this endeavor has been our inability to produce the prefusion envelope glycoprotein trimer for biochemical and structural analysis. Through our studies of the GPC envelope glycoprotein of the hemorrhagic fever arenaviruses, we have shown that GPC is unique among class I viral fusion proteins in that the mature complex retains a stable signal peptide (SSP) in addition to the conventional receptor-binding and transmembrane fusion subunits. In this report we show that the recombinant GPC precursor can be produced as a discrete native-like trimer and that its proteolytic cleavage generates the mature glycoprotein. Proteoliposomes containing the cleaved GPC mediate pH-dependent membrane fusion, a characteristic feature of arenavirus entry. This reaction is inhibited by arenavirus-specific monoclonal antibodies and small-molecule fusion inhibitors. The in vitro reconstitution of GPC-mediated membrane-fusion activity offers unprecedented opportunities for biochemical and structural studies of arenavirus entry and its inhibition. To our knowledge, this report is the first to demonstrate functional reconstitution of membrane fusion by a viral envelope glycoprotein.


Assuntos
Glicoproteínas/metabolismo , Vírus Junin/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Chlorocebus aethiops , Furina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lipídeos/química , Proteínas Mutantes/metabolismo , Testes de Neutralização , Conformação Proteica , Proteolipídeos/metabolismo , Proteólise , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície , Células Vero , Eliminação de Partículas Virais
10.
BMC Biotechnol ; 12: 80, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23121996

RESUMO

BACKGROUND: Arenavirus matrix protein Z plays an important role in virus budding and is able to generate enveloped virus-like-particles (VLPs) in absence of any other viral proteins. In these VLPs, Z protein is associated to the plasma membrane inner surface by its myristoyl residue. Budding induction and vesicle formation properties can be exploited to generate enveloped VLPs platform. These structures can be designed to carry specific antigen in the inner side or on the surface of VLPs.Vaccines based on VLPs are a highly effective type of subunit vaccines that mimic the overall structure of virus particles in absence of viral nucleic acid, being noninfectious.In this work we assayed the capacity of Junin Z protein to produce VLPs carrying the green fluorescent protein (eGFP), as a model antigen. RESULTS: In this report the Junin Z protein ability to produce VLPs from 293T cells and its capacity to deliver a specific antigen (eGFP) fused to Z was evaluated. Confocal microscopy showed a particular membrane bending in cells expressing Z and a spot welded distribution in the cytoplasm. VLPs were detected by TEM (transmission electron microscopy) and were purified from cell supernatant. The proteinase protection assay demonstrated the VLPs integrity and the absence of degradation of the fused antigen, thus indicating its internal localization. Finally, immunization of mice with purified VLPs produced high titres of anti-eGFP antibodies compared to the controls. CONCLUSIONS: It was proved that VLPs can be generated from cells transfected with a fusion Junin virus Z-eGFP protein in absence of any other viral protein, and the capacity of Z protein to support fusions at the C-terminal, without impairing its budding activity, allowing vehiculization of specific antigens into VLPs.


Assuntos
Antígenos/metabolismo , Vírus Junin/metabolismo , Proteínas Virais/metabolismo , Vírion/metabolismo , Animais , Antígenos/genética , Antígenos/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transfecção , Proteínas Virais/genética , Proteínas Virais/imunologia , Vírion/imunologia , Vírion/ultraestrutura
11.
J Gen Virol ; 92(Pt 12): 2889-2899, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21813702

RESUMO

Stress granules (SGs) are ephemeral cytoplasmic aggregates containing stalled translation preinitiation complexes involved in mRNA storage and triage during the cellular stress response. SG formation is triggered by the phosphorylation of the alpha subunit of eIF2 (eIF2α), which provokes a dramatic blockage of protein translation. Our results demonstrate that acute infection of Vero cells with the arenavirus Junín (JUNV), aetiological agent of Argentine haemorrhagic fever, does not induce the formation of SGs. Moreover, JUNV negatively modulates SG formation in infected cells stressed with arsenite, and this inhibition correlates with low levels of eIF2α phosphorylation. Transient expression of JUNV nucleoprotein (N) or the glycoprotein precursor (GPC), but not of the matrix protein (Z), inhibits SG formation in a similar manner, comparable to infectious virus. Expression of N and GPC also impaired eIF2α phosphorylation triggered by arsenite. A moderate inhibition of SG formation was also observed when DTT and thapsigargin were employed as stress inducers. In contrast, no inhibition was observed when infected cells were treated with hippuristanol, a translational inhibitor and inducer of SGs that bypasses the requirement for eIF2α phosphorylation. Finally, we analysed SG formation in persistently JUNV-infected cells, where N and GPC are virtually absent and truncated N products are expressed abundantly. We found that persistently infected cells show a quite normal response to arsenite, with SG formation comparable to that of uninfected cells. This suggests that the presence of GPC and/or N is crucial to control the stress response upon JUNV infection of Vero cells.


Assuntos
Arsenitos/farmacologia , Fator de Iniciação 2 em Eucariotos/genética , Vírus Junin/genética , Vírus Junin/patogenicidade , Animais , Chlorocebus aethiops , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , Fator de Iniciação 2 em Eucariotos/antagonistas & inibidores , Fator de Iniciação 2 em Eucariotos/metabolismo , Febre Hemorrágica Americana/genética , Febre Hemorrágica Americana/metabolismo , Vírus Junin/metabolismo , Fosforilação , Plasmídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção/métodos , Células Vero
12.
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
13.
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
14.
J Biol Chem ; 286(2): 1528-36, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21068387

RESUMO

Arenaviruses cause acute hemorrhagic fevers with high mortality. Entry of the virus into the host cell is mediated by the viral envelope glycoprotein, GPC. In contrast to other class I viral envelope glycoproteins, the mature GPC complex contains a cleaved stable signal peptide (SSP) in addition to the canonical receptor-binding (G1) and transmembrane fusion (G2) subunits. SSP is critical for intracellular transport of the GPC complex to the cell surface and for its membrane-fusion activity. Previous studies have suggested that SSP is retained in GPC through interaction with a zinc-binding domain (ZBD) in the cytoplasmic tail of G2. Here we used NMR spectroscopy to determine the structure of Junín virus (JUNV) ZBD (G2 residues 445-485) and investigate its interaction with a conserved Cys residue (Cys-57) in SSP. We show that JUNV ZBD displays a novel fold containing two zinc ions. One zinc ion is coordinated by His-447, His-449, Cys-455, and His-485. The second zinc ion is coordinated by His-459, Cys-467, and Cys-469 and readily accepts Cys-57 from SSP as the fourth ligand. Our studies describe the structural basis for retention of the unique SSP subunit and suggest a mechanism whereby SSP is positioned in the GPC complex to modulate pH-dependent membrane fusion.


Assuntos
Vírus Junin/química , Glicoproteínas de Membrana/química , Proteínas do Envelope Viral/química , Internalização do Vírus , Zinco/química , Sequência de Aminoácidos , Sequência Conservada , Concentração de Íons de Hidrogênio , Vírus Junin/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/fisiologia , Sinais Direcionadores de Proteínas/fisiologia , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Zinco/metabolismo , Dedos de Zinco/fisiologia
15.
Biotechnol J ; 4(6): 866-70, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19548229

RESUMO

Junin arenavirus (JUNV) entry is dependent on clathrin-mediated pathways and it relies on the integrity of the actin cytoskeleton as well as the dynamics of microtubules. To determine the method of entry used by this human pathogen, we have demonstrated that in Vero cells JUNV is trafficked via the cellular dynamin 2 (dyn2) endocytic pathway and it is dependent on the Eps15 GTPase. In addition, we have shown that the virus travels through Rab5-mediated early and Rab7-mediated late endosomes in its pH-dependent entry. Altogether, this study gives further inside into the endocytic pathway utilized by the arenavirus JUNV.


Assuntos
Vírus Junin/fisiologia , Proteínas Virais/fisiologia , Internalização do Vírus , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Chlorocebus aethiops , Dinamina II/metabolismo , Endocitose/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Vírus Junin/metabolismo , Camundongos , Fosfoproteínas/metabolismo , Células Vero , Carga Viral , Proteínas Virais/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
16.
J Virol ; 83(11): 5606-14, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19321606

RESUMO

The New World arenaviruses, Junin, Machupo, Guanarito, Sabia, and Chapare, are associated with rapidly progressing severe hemorrhagic fever with a high rate of case fatality in various regions of South America. The threat of natural or deliberate outbreaks associated with these viruses makes the development of preventive or therapeutic measures important. Here we describe a Junin virus functional minigenome system and a reverse genetics system for production of infectious Junin virus. This robust, highly efficient system involves transfection of cells with only two plasmids which transcribe the virus S and L antigenomic RNAs. The utility of the system is demonstrated by generating Junin viruses which encode a glycoprotein precursor (GPC) containing the following: (i) the wild-type (SKI-1/S1P peptidase) cleavage site, (ii) no cleavage site, or (iii) a cleavage site where the SKI-1/S1P motif (RSLK) is replaced by a furin cleavage site (RRKR). In contrast to the wild-type virus, Junin virus lacking a GPC cleavage site replicated within successfully transfected cells but failed to yield infectious virus particles. This confirms observations with other arenaviruses suggesting that GPC cleavage is essential for arenavirus infectivity. In contrast, infectious Junin virus which encoded GPC cleaved by furin-like proteases was easily generated. The two-plasmid, high efficiency aspects of this Junin virus reverse genetics system show great promise for addressing important questions regarding arenavirus hemorrhagic fever disease and for development of precisely attenuated live arenavirus vaccines.


Assuntos
Glicoproteínas/metabolismo , Vírus Junin/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Genoma Viral/genética , Glicoproteínas/genética , Vírus Junin/genética , Mutação/genética , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Engenharia de Proteínas , RNA Viral/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Replicação Viral
17.
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
18.
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
19.
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
20.
Virology ; 349(2): 476-91, 2006 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-16574183

RESUMO

The New World arenaviruses Junin, Machupo, and Guanarito are the causative agents of hemorrhagic fevers (HF) with high mortality in humans. The cellular receptor for Old World arenaviruses and one subgroup of the New World arenaviruses (Clade C) have been identified as alpha-dystroglycan (alpha-DG). In contrast, the receptor(s) of the South American HF viruses, which belong to the Clade B New World arenaviruses, are currently unknown. To begin to characterize the cellular receptors used by these pathogens, we generated recombinant retroviral pseudotypes with the glycoproteins of Guanarito, Junin, and Machupo. Infection with the South American HF viruses is independent of alpha-DG and functional receptors for Guanarito, Junin, and Machupo were found on most human cell types and cells derived from non-human primate and rodents. Guanarito, Junin, and Machupo share a common receptor, which is distinct from the receptor(s) used by the closely related non-pathogenic Clade B virus Amapari, and the genetically more distant Clade A and C New World arenaviruses. We show that the cellular receptor(s) for the South American HF viruses are proteins or protein-linked entities and that infection is not dependent on protein-linked N-glycans, O-glycans, or glycosaminoglycans.


Assuntos
Arenavirus do Novo Mundo/metabolismo , Vírus Junin/metabolismo , Proteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Humanos , Camundongos , Polissacarídeos/fisiologia , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...