Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS Pathog ; 7(9): e1002232, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21931550

RESUMO

The highly pathogenic Old World arenavirus Lassa virus (LASV) and the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) use α-dystroglycan as a cellular receptor and enter the host cell by an unusual endocytotic pathway independent of clathrin, caveolin, dynamin, and actin. Upon internalization, the viruses are delivered to acidified endosomes in a Rab5-independent manner bypassing classical routes of incoming vesicular trafficking. Here we sought to identify cellular factors involved in the unusual and largely unknown entry pathway of LASV and LCMV. Cell entry of LASV and LCMV required microtubular transport to late endosomes, consistent with the low fusion pH of the viral envelope glycoproteins. Productive infection with recombinant LCMV expressing LASV envelope glycoprotein (rLCMV-LASVGP) and LCMV depended on phosphatidyl inositol 3-kinase (PI3K) as well as lysobisphosphatidic acid (LBPA), an unusual phospholipid that is involved in the formation of intraluminal vesicles (ILV) of the multivesicular body (MVB) of the late endosome. We provide evidence for a role of the endosomal sorting complex required for transport (ESCRT) in LASV and LCMV cell entry, in particular the ESCRT components Hrs, Tsg101, Vps22, and Vps24, as well as the ESCRT-associated ATPase Vps4 involved in fission of ILV. Productive infection with rLCMV-LASVGP and LCMV also critically depended on the ESCRT-associated protein Alix, which is implicated in membrane dynamics of the MVB/late endosomes. Our study identifies crucial cellular factors implicated in Old World arenavirus cell entry and indicates that LASV and LCMV invade the host cell passing via the MVB/late endosome. Our data further suggest that the virus-receptor complexes undergo sorting into ILV of the MVB mediated by the ESCRT, possibly using a pathway that may be linked to the cellular trafficking and degradation of the cellular receptor.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Vírus Lassa/patogenicidade , Vírus da Coriomeningite Linfocítica/patogenicidade , Corpos Multivesiculares/virologia , Internalização do Vírus , ATPases Associadas a Diversas Atividades Celulares , Animais , Transporte Biológico , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Distroglicanas/metabolismo , Endocitose , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Endossomos/metabolismo , Células HEK293 , Humanos , Immunoblotting , Lisofosfolipídeos/metabolismo , Monoglicerídeos/metabolismo , Mutação , Fosfatos de Fosfatidilinositol/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptores Virais/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transferrina/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
2.
Cell Microbiol ; 14(7): 1122-34, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22405130

RESUMO

The arenavirus Lassa virus (LASV) causes a severe haemorrhagic fever with high mortality in man. The cellular receptor for LASV is dystroglycan (DG). DG is a ubiquitous receptor for extracellular matrix (ECM) proteins, which cooperates with ß1 integrins to control cell-matrix interactions. Here, we investigated whether LASV binding to DG triggers signal transduction, mimicking the natural ligands. Engagement of DG by LASV resulted in the recruitment of the adaptor protein Grb2 and the protein kinase MEK1 by the cytoplasmic domain of DG without activating the MEK/ERK pathway, indicating assembly of an inactive signalling complex. LASV binding to cells however affected the activation of the MEK/ERK pathway via α6ß1 integrins. The virus-induced perturbation of α6ß1 integrin signalling critically depended on high-affinity LASV binding to DG and DG's cytoplasmic domain, indicating that LASV-receptor binding perturbed signalling cross-talk between DG and ß1 integrins.


Assuntos
Distroglicanas/metabolismo , Matriz Extracelular/metabolismo , Vírus Lassa/patogenicidade , Receptores Virais/metabolismo , Transdução de Sinais , Ligação Viral , Linhagem Celular , Humanos , Integrina beta1/metabolismo , Vírus Lassa/fisiologia , Modelos Biológicos
3.
J Virol ; 84(1): 573-84, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846507

RESUMO

A crucial step in the arenavirus life cycle is the biosynthesis of the viral envelope glycoprotein (GP) responsible for virus attachment and entry. Processing of the GP precursor (GPC) by the cellular proprotein convertase site 1 protease (S1P), also known as subtilisin-kexin-isozyme 1 (SKI-1), is crucial for cell-to-cell propagation of infection and production of infectious virus. Here, we sought to evaluate arenavirus GPC processing by S1P as a target for antiviral therapy using a recently developed peptide-based S1P inhibitor, decanoyl (dec)-RRLL-chloromethylketone (CMK), and the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). To control for off-target effects of dec-RRLL-CMK, we employed arenavirus reverse genetics to introduce a furin recognition site into the GPC of LCMV. The rescued mutant virus grew to normal titers, and the processing of its GPC critically depended on cellular furin, but not S1P. Treatment with the S1P inhibitor dec-RRLL-CMK resulted in specific blocking of viral spread and virus production of LCMV. Combination of the protease inhibitor with ribavirin, currently used clinically for treatment of human arenavirus infections, resulted in additive drug effects. In cells deficient in S1P, the furin-dependent LCMV variant established persistent infection, whereas wild-type LCMV underwent extinction without the emergence of S1P-independent escape variants. Together, the potent antiviral activity of an inhibitor of S1P-dependent GPC cleavage, the additive antiviral effect with ribavirin, and the low probability of emergence of S1P-independent viral escape variants make S1P-mediated GPC processing by peptide-derived inhibitors a promising strategy for the development of novel antiarenaviral drugs.


Assuntos
Antivirais/farmacologia , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Pró-Proteína Convertases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Proteínas do Envelope Viral/metabolismo , Animais , Arenavirus/efeitos dos fármacos , Linhagem Celular , Sistemas de Liberação de Medicamentos/métodos , Sinergismo Farmacológico , Glicoproteínas/metabolismo , Humanos , Peptídeo Hidrolases/metabolismo , Peptídeos/farmacologia , Pró-Proteína Convertases/metabolismo , Inibidores de Proteases/uso terapêutico , Ribavirina/farmacologia , Serina Endopeptidases/metabolismo
4.
Mol Biol Cell ; 18(11): 4493-507, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17761532

RESUMO

alpha-Dystroglycan (alpha-DG) is an important cellular receptor for extracellular matrix (ECM) proteins as well as the Old World arenaviruses lymphocytic choriomeningitis virus (LCMV) and the human pathogenic Lassa fever virus (LFV). Specific O-glycosylation of alpha-DG is critical for its function as receptor for ECM proteins and arenaviruses. Here, we investigated the impact of arenavirus infection on alpha-DG expression. Infection with an immunosuppressive LCMV isolate caused a marked reduction in expression of functional alpha-DG without affecting biosynthesis of DG core protein or global cell surface glycoprotein expression. The effect was caused by the viral glycoprotein (GP), and it critically depended on alpha-DG binding affinity and GP maturation. An equivalent effect was observed with LFVGP. Viral GP was found to associate with a complex between DG and the glycosyltransferase LARGE in the Golgi. Overexpression of LARGE restored functional alpha-DG expression in infected cells. We provide evidence that virus-induced down-modulation of functional alpha-DG perturbs DG-mediated assembly of laminin at the cell surface, affecting normal cell-matrix interactions.


Assuntos
Arenavirus do Velho Mundo/fisiologia , Distroglicanas/metabolismo , Regulação da Expressão Gênica , Animais , Linhagem Celular , Chlorocebus aethiops , Glicosilação , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Humanos , Laminina/metabolismo , Camundongos , Ligação Proteica , Transcrição Gênica/genética , Proteínas Virais/metabolismo
5.
J Virol ; 82(3): 1505-17, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18045945

RESUMO

In contrast to most enveloped viruses that enter the host cell via clathrin-dependent endocytosis, the Old World arenavirus lymphocytic choriomeningitis virus (LCMV) enters cells via noncoated vesicles that deliver the virus to endosomes, where pH-dependent membrane fusion occurs. Here, we investigated the initial steps of LCMV infection. We found that the attachment of LCMV to its cellular receptor alpha-dystroglycan occurs rapidly and is not dependent on membrane cholesterol. However, subsequent virus internalization is sensitive to cholesterol depletion, indicating the involvement of a cholesterol-dependent pathway. We provide evidence that LCMV entry involves an endocytotic pathway that is independent of clathrin and caveolin and that does not require the GTPase dynamin. In addition, neither the structural integrity nor the dynamics of the actin cytoskeleton are required for infection. These findings indicate that the prototypic Old World arenavirus LCMV uses a mechanism of entry that is different from clathrin-mediated endocytosis, which is used by the New World arenavirus Junin virus, and pathways used by other enveloped viruses.


Assuntos
Endocitose/fisiologia , Vírus da Coriomeningite Linfocítica/fisiologia , Internalização do Vírus , Actinas/metabolismo , Animais , Caveolinas/metabolismo , Linhagem Celular , Membrana Celular/química , Chlorocebus aethiops , Colesterol/fisiologia , Clatrina/metabolismo , Cricetinae , Dinaminas/metabolismo , Distroglicanas/metabolismo , Humanos , Mesocricetus , Ligação Viral
6.
J Virol ; 82(12): 6045-51, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400865

RESUMO

The cellular proprotein convertase site 1 protease (S1P) has been implicated in the proteolytic processing of the glycoproteins (GPs) of Old World arenaviruses. Here we report that S1P is also involved in the processing of the GPs of the genetically more-distant South American hemorrhagic fever viruses Guanarito, Machupo, and Junin. Efficient cleavage of Guanarito virus GP, whose protease recognition sites deviate from the reported S1P consensus sequence, indicates a broader specificity of S1P than anticipated. Lack of GP processing of Junin virus dramatically reduced production of infectious virus and prevented cell-to-cell propagation. Infection of S1P-deficient cells resulted in viral persistence over several weeks without the emergence of escape variants able to use other cellular proteases for GP processing.


Assuntos
Arenavirus do Novo Mundo/metabolismo , Glicoproteínas/metabolismo , Pró-Proteína Convertases/metabolismo , Processamento de Proteína Pós-Traducional , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteínas de Fluorescência Verde/metabolismo , Humanos , Rim/citologia , Luciferases/análise , Luciferases/metabolismo , Dados de Sequência Molecular , Mutação , Pró-Proteína Convertases/química , Pró-Proteína Convertases/genética , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/genética , Transfecção , Células Vero
7.
J Virol ; 82(15): 7677-87, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18508885

RESUMO

The Old World arenavirus Lassa virus (LASV) is the causative agent of severe viral hemorrhagic fever (VHF) in humans and is the most prevalent human pathogen among arenaviruses. The present study investigated the largely unknown mechanisms of cell entry of LASV, a process know to be mediated solely by the virus envelope glycoprotein (GP). To circumvent biosafety restrictions associated with the use of live LASV, we used reverse genetics to generate a recombinant variant of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) expressing the LASV GP (rLCMV-LASVGP). The rescued rLCMV-LASVGP grew to titers comparable to that of LCMV and showed the receptor binding characteristics of LASV. We used rLCMV-LASVGP to characterize the cellular mechanisms of LASV entry in the context of a productive arenavirus infection. The kinetics of pH-dependent membrane fusion of rLCMV-LASVGP resembled those of the human-pathogenic New World arenavirus Junin virus (JUNV) and other enveloped viruses that use clathrin-mediated endocytosis for entry. However, rLCMV-LASVGP entered cells predominantly via a clathrin-, caveolin-, and dynamin-independent endocytotic pathway similar to the one recently described for LCMV. Productive infection of rLCMV-LASVGP was only mildly affected by a dominant negative mutant of Rab5 and was independent of Rab7, suggesting an unusual mechanism of delivery to endosomes. In addition, rLCMV-LASVGP infection was independent of actin but required intact microtubules. Our data indicate that LASV enters cells via a pathway distinct from the one used by human-pathogenic New World arenaviruses.


Assuntos
Arenavirus do Velho Mundo/fisiologia , Vírus Lassa/genética , Vírus da Coriomeningite Linfocítica/genética , Internalização do Vírus , Actinas/genética , Animais , Arenavirus do Novo Mundo/fisiologia , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Endocitose , Endossomos/virologia , Humanos , Concentração de Íons de Hidrogênio , Microtúbulos/virologia , Fatores de Tempo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/genética , proteínas de unión al GTP Rab7
8.
Cell Microbiol ; 10(4): 828-35, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18182084

RESUMO

The arenaviruses Lassa virus (LASV) in Africa and Machupo (MACV), Guanarito (GTOV) and Junin viruses (JUNV) in South America cause severe haemorrhagic fevers in humans with fatality rates of 15-35%. The present review focuses on the first steps of infection with human pathogenic arenaviruses, the interaction with their cellular receptor molecules and subsequent entry into the host cell. While similarities exist in genomic organization, structure and clinical disease caused by pathogenic Old World and New World arenaviruses these pathogens use different primary receptors. The Old World arenaviruses employ alpha-dystroglycan, a cellular receptor for proteins of the extracellular matrix, and the human pathogenic New World arenaviruses use the cellular cargo receptor transferrin receptor 1. While the New World arenavirus JUNV enters cells via clathrin-dependent endocytosis, evidence occurred for clathrin-independent entry of the prototypic Old World arenavirus lymphocytic choriomeningitis virus. Upon internalization, arenaviruses are delivered to the endosome, where pH-dependent membrane fusion is mediated by the envelope glycoprotein (GP). While arenavirus GPs share characteristics with class I fusion GPs of other enveloped viruses, unusual mechanistic features of GP-mediated membrane fusion have recently been discovered for arenaviruses with important implications for viral entry.


Assuntos
Arenavirus/crescimento & desenvolvimento , Receptores de Superfície Celular/fisiologia , Animais , Arenavirus/metabolismo , Arenavirus do Novo Mundo/crescimento & desenvolvimento , Arenavirus do Novo Mundo/metabolismo , Arenavirus do Velho Mundo/crescimento & desenvolvimento , Arenavirus do Velho Mundo/metabolismo , Endocitose/fisiologia , Humanos , Modelos Biológicos , Ligação Viral
9.
Virology ; 372(1): 107-17, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18022208

RESUMO

The prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) represents a powerful experimental model for the study of the basic virology and pathogenesis of arenaviruses. In the present study, we used the LCMV model to evaluate the anti-viral potential of phosphorothioate oligonucleotides against arenaviruses. Our findings indicate that amphipathic DNA polymers (APs) are potent inhibitors of infection with a series of LCMV isolates with IC(50) in the low nanomolar range. APs target the surface glycoprotein (GP) of LCMV and block viral entry and cell-cell propagation of the virus, without affecting later steps in replication or release of progeny virus from infected cells. The anti-viral action of APs is sequence-independent but is critically dependent on their size and hydrophobicity. Mechanistically, we provide evidence that APs disrupt the interaction between LCMVGP and its cellular receptor, alpha-dystroglycan. Exposure of LCMV to APs does not affect the stability of the GP virion spike and has no effect on the conformation of a neutralizing antibody epitope, suggesting rather subtle changes in the conformation and/or conformational dynamics of the viral GP.


Assuntos
Antivirais/farmacologia , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Oligonucleotídeos Fosforotioatos/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Distroglicanas/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Vírus da Coriomeningite Linfocítica/patogenicidade , Glicoproteínas de Membrana/metabolismo , Células Vero
10.
J Biol Chem ; 283(27): 18734-42, 2008 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-18474596

RESUMO

Viral hemorrhagic fevers caused by the arenaviruses Lassa virus in Africa and Machupo, Guanarito, Junin, and Sabia virus in South America are among the most devastating emerging human diseases with fatality rates of 15-35% and a limited antiviral therapeutic repertoire available. Here we used high throughput screening of synthetic combinatorial small molecule libraries to identify inhibitors of arenavirus infection using pseudotyped virion particles bearing the glycoproteins (GPs) of highly pathogenic arenaviruses. Our screening efforts resulted in the discovery of a series of novel small molecule inhibitors of viral entry that are highly active against both Old World and New World hemorrhagic arenaviruses. We observed potent inhibition of infection of human and primate cells with live hemorrhagic arenaviruses (IC(50)=500-800 nm). Investigations of the mechanism of action revealed that the candidate compounds efficiently block pH-dependent fusion by the arenavirus GPs (IC(50) of 200-350 nm). Although our lead compounds were potent against phylogenetically distant arenaviruses, they did not show activity against other enveloped viruses with class I viral fusion proteins, indicating specificity for arenavirus GP-mediated membrane fusion.


Assuntos
Antivirais/química , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Arenavirus , Doenças Transmissíveis Emergentes/tratamento farmacológico , Febres Hemorrágicas Virais/tratamento farmacológico , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Antivirais/uso terapêutico , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Células HeLa , Humanos , Células Vero , Vírion/metabolismo
11.
J Virol ; 81(11): 5685-95, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17360738

RESUMO

alpha-Dystroglycan (DG) is an important cellular receptor for extracellular matrix (ECM) proteins and also serves as the receptor for Old World arenaviruses Lassa fever virus (LFV) and lymphocytic choriomeningitis virus (LCMV) and clade C New World arenaviruses. In the host cell, alpha-DG is subject to a remarkably complex pattern of O glycosylation that is crucial for its interactions with ECM proteins. Two of these unusual sugar modifications, protein O mannosylation and glycan modifications involving the putative glycosyltransferase LARGE, have recently been implicated in arenavirus binding. Considering the complexity of alpha-DG O glycosylation, our present study was aimed at the identification of the specific O-linked glycans on alpha-DG that are recognized by arenaviruses. As previously shown for LCMV, we found that protein O mannosylation of alpha-DG is crucial for the binding of arenaviruses of distinct phylogenetic origins, including LFV, Mobala virus, and clade C New World arenaviruses. In contrast to the highly conserved requirement for O mannosylation, more generic O glycans present on alpha-DG are dispensable for arenavirus binding. Despite the critical role of O-mannosyl glycans for arenavirus binding under normal conditions, the overexpression of LARGE in cells deficient in O mannosylation resulted in highly glycosylated alpha-DG that was functional as a receptor for arenaviruses. Thus, modifications by LARGE but not O-mannosyl glycans themselves are most likely the crucial structures recognized by arenaviruses. Together, the data demonstrate that arenaviruses recognize the same highly conserved O-glycan structures on alpha-DG involved in ECM protein binding, indicating a strikingly similar mechanism of receptor recognition by pathogen- and host-derived ligands.


Assuntos
Arenavirus do Novo Mundo/metabolismo , Arenavirus do Velho Mundo/metabolismo , Distroglicanas/metabolismo , Mimetismo Molecular , Receptores Virais/metabolismo , Animais , Arenavirus do Novo Mundo/química , Arenavirus do Velho Mundo/química , Linhagem Celular Tumoral , Chlorocebus aethiops , Distroglicanas/química , Glicosilação , Humanos , Células Jurkat , Vírus Lassa/metabolismo , Ligantes , Vírus da Coriomeningite Linfocítica/química , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Coelhos , Receptores Virais/química , Células Vero
12.
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
13.
J Virol ; 80(18): 9082-92, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16940520

RESUMO

Neonatal infection of most mouse strains with lymphocytic choriomeningitis virus (LCMV) leads to a life-long persistent infection characterized by high virus loads in the central nervous system (CNS) in the absence of inflammation and tissue destruction. These mice, however, exhibit impaired learning and memory. The occurrence of cognitive defects in the absence of overt CNS pathology led us to the hypothesis that chronic virus infection may contribute to neuronal dysfunction by altering the host's gene expression profile. To test this hypothesis, we examined the impact of LCMV persistence on host gene expression in the CNS. To model the natural route of human congenital CNS infection observed with a variety of viruses, we established a persistently infected mouse colony where the virus was maintained via vertical transmission from infected mothers to offspring (LCMV-cgPi). LCMV-cgPi mice exhibited a lifelong persistent infection involving the CNS; the infection was associated with impaired spatial-temporal learning. Despite high viral loads in neurons of the brains of adult LCMV-cgPi mice, we detected changes in the host's CNS gene expression for only 75 genes, 56 and 19 being significantly induced and reduced, respectively. The majority of the genes induced in the brain of LCMV-cgPi mice were interferon (IFN)-stimulated genes (ISGs) and included the transcription factors STAT1 and IRF9, the ISG15 protease UBP43, and the glucocorticoid attenuated-response genes GARG16 and GARG49. Based on their crucial role in antiviral defense, these ISGs may play an important role in limiting viral spread and replication. However, since IFNs have also been implicated in adverse effects on neuronal function, the chronic induction of some ISGs may also contribute to the observed cognitive impairment.


Assuntos
Sistema Nervoso Central/metabolismo , Regulação da Expressão Gênica , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Animais , Feminino , Coriomeningite Linfocítica/congênito , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , RNA/metabolismo , Distribuição Tecidual , Transcrição Gênica
14.
J Virol ; 79(5): 2880-90, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15709007

RESUMO

Developmentally aged chicken embryo cells which hyperproduce interferon (IFN) when induced were used to quantify IFN production and its suppression by eight strains of type A influenza viruses (AIV). Over 90% of the IFN-inducing or IFN induction-suppressing activity of AIV populations resided in noninfectious particles. The IFN-inducer moiety of AIV appears to preexist in, or be generated by, virions termed IFN-inducing particles (IFP) and was detectable under conditions in which a single molecule of double-stranded RNA introduced into a cell via endocytosis induced IFN, whereas single-stranded RNA did not. Some AIV strains suppressed IFN production, an activity that resided in a noninfectious virion termed an IFN induction-suppressing particle (ISP). The ISP phenotype was dominant over the IFP phenotype. Strains of AIV varied 100-fold in their capacity to induce IFN. AIV genetically compromised in NS1 expression induced about 20 times more IFN than NS1-competent parental strains. UV irradiation further enhanced the IFN-inducing capacity of AIV up to 100-fold, converting ISP into IFP and IFP into more efficient IFP. AIV is known to prevent IFN induction and/or production by expressing NS1 from a small UV target (gene NS). Evidence is presented for an additional downregulator of IFN production, identified as a large UV target postulated to consist of AIV polymerase genes PB1 + PB2 + PA, through the ensuing action of their cap-snatching endonuclease on pre-IFN-mRNA. The products of both the small and large UV targets act in concert to regulate IFN induction and/or production. Knowledge of the IFP/ISP phenotype may be useful in the development of attenuated AIV strains that maximally induce cytokines favorable to the immune response.


Assuntos
Vírus da Influenza A/imunologia , Interferons/biossíntese , Animais , Células Cultivadas , Embrião de Galinha , Relação Dose-Resposta à Radiação , Genoma Viral , Temperatura Alta , Tolerância Imunológica , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Vírus da Influenza A/efeitos da radiação , Cinética , Proteínas Repressoras/metabolismo , Especificidade da Espécie , Raios Ultravioleta
15.
J Virol ; 79(10): 5979-87, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15857984

RESUMO

The cellular receptor for the Old World arenaviruses Lassa fever virus (LFV) and lymphocytic choriomeningitis virus (LCMV) has recently been identified as alpha-dystroglycan (alpha-DG), a cell surface receptor that provides a molecular link between the extracellular matrix and the actin-based cytoskeleton. In the present study, we show that LFV binds to alpha-DG with high affinity in the low-nanomolar range. Recombinant vesicular stomatitis virus pseudotyped with LFV glycoprotein (GP) adopted the receptor binding characteristics of LFV and depended on alpha-DG for infection of cells. Mapping of the binding site of LFV on alpha-DG revealed that LFV binding required the same domains of alpha-DG that are involved in the binding of LCMV. Further, LFV was found to efficiently compete with laminin alpha1 and alpha2 chains for alpha-DG binding. Together with our previous studies on receptor binding of the prototypic immunosuppressive LCMV isolate LCMV clone 13, these findings indicate a high degree of conservation in the receptor binding characteristics between the highly human-pathogenic LFV and murine-immunosuppressive LCMV isolates.


Assuntos
Distroglicanas/metabolismo , Febre Lassa/virologia , Vírus Lassa/metabolismo , Receptores Virais/metabolismo , Sítios de Ligação , Células HeLa , Humanos , Vírus Lassa/fisiologia , Ligação Proteica , Estrutura Terciária de Proteína , Replicação Viral
16.
J Virol ; 79(22): 14282-96, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16254363

RESUMO

The receptor for lymphocytic choriomeningitis virus (LCMV), the human pathogenic Lassa fever virus (LFV), and clade C New World arenaviruses is alpha-dystroglycan (alpha-DG), a cell surface receptor for proteins of the extracellular matrix (ECM). Specific posttranslational modification of alpha-DG by the glycosyltransferase LARGE is critical for its function as an ECM receptor. In the present study, we show that LARGE-dependent modification is also crucial for alpha-DG's function as a cellular receptor for arenaviruses. Virus binding involves the mucin-type domain of alpha-DG and depends on modification by LARGE. A crucial role of the LARGE-dependent glycosylation of alpha-DG for virus binding is found for several isolates of LCMV, LFV, and the arenaviruses Mobala and Oliveros. Since the posttranslational modification by LARGE is crucial for alpha-DG recognition by both arenaviruses and the host-derived ligand laminin, it also influences competition between virus and laminin for alpha-DG. Hence, LARGE-dependent glycosylation of alpha-DG has important implications for the virus-host cell interaction and the pathogenesis of LFV in humans.


Assuntos
Arenaviridae/fisiologia , RNA Polimerases Dirigidas por DNA/metabolismo , Distroglicanas/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Arenaviridae/metabolismo , Sequência de Bases , Cruzamentos Genéticos , Primers do DNA , Feminino , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Virais/metabolismo
17.
Virology ; 325(2): 432-45, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15246281

RESUMO

Long-term infections with viruses permit the generation of variants that evolve specific growth advantages in certain tissues and may show altered disease potentials. The selection of such variants is influenced by the host tissue and often involves virus-receptor interactions. Here we report studies of receptor usage by several lymphocytic choriomeningitis virus (LCMV) isolates that expressed different disease patterns. Consistent with our previous studies, we found that, with one exception, multiple LCMV variants that cause suppression of immune responses bound with high affinity to their cellular receptor alpha-dystroglycan (alpha-DG) and were dependent on alpha-DG for entry and infection. The exception also bound strongly to alpha-DG but was not dependent on alpha-DG for entry and infection. In contrast, those variants of LCMV that do not suppress the immune response either displayed low or no binding affinity for alpha-DG and used alternative receptors in addition to or instead of alpha-DG for entry and infection. For all alpha-DG binding variants, alpha-DG represents the preferred receptor in DG-expressing cells, as soluble alpha-DG blocked their infection of DG-deficient cells, indicating that binding of alpha-DG to the viral glycoprotein (GP) at the virion surface interferes with the GP's interaction with the alternative receptor. Biochemical characterization of the alternative receptor(s) for LCMV indicated that they are either protein(s) or protein-bound entities.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Glicoproteínas de Membrana/fisiologia , Receptores Virais/fisiologia , Animais , Células Cultivadas , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Distroglicanas , Glicoproteínas/metabolismo , Técnicas In Vitro , Vírus da Coriomeningite Linfocítica/isolamento & purificação , Vírus da Coriomeningite Linfocítica/fisiologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA