Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 1435: 29-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27188548

RESUMO

WNV infectious clones are valuable tools for elucidating WNV biology. Nevertheless, relatively few infectious WNV clones have been generated because their construction is hampered by the instability of flaviviral genomes. More recently, advances in cloning techniques as well as the development of several two-plasmid WNV infectious clone systems have facilitated the generation of WNV infectious clones. Here we described a protocol for recovering WNV from a two-plasmid system. In this approach, large quantities of these constructs are digested with restriction enzymes to produce complementary restriction sites at the 3' end of the upstream fragment and the 5' end of the downstream fragment. These fragments are then annealed to produce linear template for in vitro transcription to synthesize infectious RNA. The resulting RNA is transfected into cells and after several days WNV is recovered in the culture supernatant. This method can be used to generate virus from infectious clones encoding high- and low-pathogenicity strains of WNV, as well as chimeric virues.


Assuntos
Transfecção/métodos , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Vírus do Nilo Ocidental/genética , Animais , Chlorocebus aethiops , Células Clonais/virologia , Eletroporação , RNA Viral , Transcrição Gênica , Células Vero , Replicação Viral , Vírus do Nilo Ocidental/patogenicidade
2.
Arch Virol ; 159(11): 3113-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25023336

RESUMO

The viral determinants governing the varied neuropathogenicity of different West Nile virus (WNV) strains are poorly understood. Here, we generated an infectious clone (WNV-MAD(IC)) of the non-pathogenic strain WNV-MAD78 and compared its replication to that of parental WNV-MAD78 and a WNV-MAD78 infectious clone (WNV-MAD(TX-UTRs)) containing the 5' and 3' untranslated regions (UTRs) of the pathogenic strain WNV-TX. All three viruses replicated at similar rates and caused similar lethality in mice. Thus, the infectious clone is indistinguishable from parental virus in replication and neurovirulence, and the UTRs alone do not account for the increased virulence of WNV-TX compared to WNV-MAD78.


Assuntos
Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/fisiologia , Animais , Chlorocebus aethiops , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Regiões não Traduzidas , Células Vero , Virulência , Replicação Viral , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Vírus do Nilo Ocidental/patogenicidade
3.
J Gen Virol ; 95(Pt 9): 1991-2003, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24920724

RESUMO

The molecular basis for the increased resistance of astrocytes to a non-neuropathogenic strain of West Nile virus (WNV), WNV-MAD78, compared with the neuropathogenic strain WNV-NY remains unclear. Here, we demonstrated that the reduced susceptibility of astrocytes to WNV-MAD78 is due to a combination of both cellular activities as well as viral determinants. Analyses of the viral particle indicated that astrocyte-derived WNV-MAD78 particles were less infectious than those of WNV-NY. Additionally, inhibition of cellular furin-like proteases increased WNV-MAD78 infectious particle production in astrocytes, suggesting that high levels of furin-like protease activity within these cells acted in a cell- and strain-specific manner to inhibit WNV-MAD78 replication. Moreover, analysis of recombinant viruses indicated that the structural proteins of WNV-MAD78 were responsible for decreased particle infectivity and the corresponding reduction in infectious particle production compared with WNV-NY. Thus, the composition of the WNV virion was also a major determinant for viral fitness within astrocytes and may contribute to WNV propagation within the central nervous system. Whether the WNV-MAD78 structural genes reduce virus replication and particle infectivity through the same mechanism as the cellular furin-like protease activity or whether these two determinants function through distinct pathways remains to be determined.


Assuntos
Astrócitos/enzimologia , Astrócitos/virologia , Proteínas Estruturais Virais/imunologia , Replicação Viral/genética , Vírus do Nilo Ocidental/patogenicidade , Animais , Astrócitos/imunologia , Linhagem Celular , Sistema Nervoso Central/virologia , Chlorocebus aethiops , Furina/antagonistas & inibidores , Furina/metabolismo , Glicosilação , Humanos , Células Vero , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia
4.
Viruses ; 6(4): 1637-53, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24721788

RESUMO

West Nile virus (WNV) is a positive-sense RNA arbovirus responsible for recent outbreaks of severe neurological disease within the US and Europe. Large-scale analyses of antiviral compounds that inhibit virus replication have been limited due to the lack of an adequate WN reporter virus. Previous attempts to insert a reporter into the 3' untranslated region of WNV generated unstable viruses, suggesting that this region does not accommodate additional nucleotides. Here, we engineered two WNV infectious clones containing insertions at the Capsid (C)/Capsid Anchor (CA) junction of the viral polyprotein. Recombinant viruses containing a TAT(1-67) or Gaussia Luciferase (GLuc) gene at this location were successfully recovered. However, rapid loss of most, if not all, of the reporter sequence occurred for both viruses, indicating that the reporter viruses were not stable. While the GLuc viruses predominantly reverted back to wild-type WNV length, the TAT viruses retained up to 75 additional nucleotides of the reporter sequence. These additional nucleotides were stable over at least five passages and did not significantly alter WNV fitness. Thus, the C/CA junction of WNV can tolerate additional nucleotides, though insertions are subject to certain constraints.


Assuntos
Proteínas do Capsídeo/genética , Genes Reporter , Mutagênese Insercional , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/fisiologia , Instabilidade Genômica , Luciferases/genética , Genética Reversa , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
5.
Viruses ; 6(3): 1424-41, 2014 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-24662674

RESUMO

Although dermal fibroblasts are one of the first cell types exposed to West Nile virus (WNV) during a blood meal by an infected mosquito, little is known about WNV replication within this cell type. Here, we demonstrate that neuroinvasive, WNV-New York (WNV-NY), and nonneuroinvasive, WNV-Australia (WNV-AUS60) strains are able to infect and replicate in primary human dermal fibroblasts (HDFs). However, WNV-AUS60 replication and spread within HDFs was reduced compared to that of WNV-NY due to an interferon (IFN)-independent reduction in viral infectivity early in infection. Additionally, replication of both strains was constrained late in infection by an IFN-ß-dependent reduction in particle infectivity. Overall, our data indicates that human dermal fibroblasts are capable of supporting WNV replication; however, the low infectivity of particles produced from HDFs late in infection suggests that this cell type likely plays a limited role as a viral reservoir in vivo.


Assuntos
Fibroblastos/imunologia , Fibroblastos/virologia , Interferons/imunologia , Replicação Viral , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia , Células Cultivadas , Humanos , Tropismo Viral
6.
J Gen Virol ; 95(Pt 4): 862-867, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24413421

RESUMO

The neuroinflammatory response to West Nile virus (WNV) infection can be either protective or pathological depending on the context. Although several studies have examined chemokine profiles within brains of WNV-infected mice, little is known about how various cell types within the central nervous system (CNS) contribute to chemokine expression. Here, we assessed chemokine expression in brain microvascular endothelial cells and astrocytes, which comprise the major components of the blood-brain barrier (BBB), in response to a non-pathogenic (WNV-MAD78) and a highly pathogenic (WNV-NY) strain of WNV. Higher levels of the chemokine CCL5 were detected in WNV-MAD78-infected brain endothelial monolayers compared with WNV-NY-infected cells. However, the opposite profile was observed in WNV-infected astrocytes, indicating that pathogenic and non-pathogenic strains of WNV provoke different CCL5 profiles at the BBB. Thus, cells comprising the BBB may contribute to a dynamic pro-inflammatory response within the CNS that evolves as WNV infection progresses.


Assuntos
Astrócitos/virologia , Encéfalo/virologia , Quimiocina CCL5/biossíntese , Células Endoteliais/virologia , Interações Hospedeiro-Patógeno , Vírus do Nilo Ocidental/imunologia , Animais , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Camundongos
7.
J Neurovirol ; 20(2): 113-21, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23843081

RESUMO

The recent introduction of highly pathogenic strains of West Nile virus (WNV) into naïve populations in Europe, Israel, and the USA has resulted in a marked increase in both the number of reported cases and the severity of disease compared to previous outbreaks. The impact of the increased virulence of recently emerged strains of WNV is exacerbated by the fact that antiviral therapies and vaccines are not currently available for use in humans. A greater understanding of the viral and host factors involved in WNV-mediated neuropathology is necessary to facilitate the development of novel therapeutic approaches. This review summarizes the current state of knowledge of the role of the cell-intrinsic innate immune responses as well as the cell-mediated innate and adaptive immune responses in promoting the detection and clearance of WNV from the CNS.


Assuntos
Sistema Nervoso Central/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Imunidade Adaptativa , Animais , Sistema Nervoso Central/virologia , Quimiocinas/imunologia , Europa (Continente)/epidemiologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Israel/epidemiologia , Camundongos , Receptores de Quimiocinas/imunologia , Estados Unidos/epidemiologia , Virulência , Febre do Nilo Ocidental/epidemiologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia
8.
J Virol ; 87(5): 2814-22, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269784

RESUMO

The severity of West Nile virus (WNV) infection in immunocompetent animals is highly strain dependent, ranging from avirulent to highly neuropathogenic. Here, we investigate the nature of this strain-specific restriction by analyzing the replication of avirulent (WNV-MAD78) and highly virulent (WNV-NY) strains in neurons, astrocytes, and microvascular endothelial cells, which comprise the neurovascular unit within the central nervous system (CNS). We demonstrate that WNV-MAD78 replicated in and traversed brain microvascular endothelial cells as efficiently as WNV-NY. Likewise, similar levels of replication were detected in neurons. Thus, WNV-MAD78's nonneuropathogenic phenotype is not due to an intrinsic inability to replicate in key target cells within the CNS. In contrast, replication of WNV-MAD78 was delayed and reduced compared to that of WNV-NY in astrocytes. The reduced susceptibility of astrocytes to WNV-MAD78 was due to a delay in viral genome replication and an interferon-independent reduction in cell-to-cell spread. Together, our data suggest that astrocytes regulate WNV spread within the CNS and therefore are an attractive target for ameliorating WNV-induced neuropathology.


Assuntos
Astrócitos/virologia , Células Endoteliais/virologia , Replicação Viral , Vírus do Nilo Ocidental/fisiologia , Animais , Linhagem Celular , Sistema Nervoso Central/virologia , Chlorocebus aethiops , Humanos , Microvasos/citologia , Microvasos/virologia , Neurônios/virologia , Células Vero , Febre do Nilo Ocidental/transmissão , Febre do Nilo Ocidental/virologia
9.
PLoS One ; 7(9): e45800, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029249

RESUMO

Autophagy is a homeostatic process responsible for recycling cytosolic proteins and organelles. Moreover, this pathway contributes to the cell's intrinsic innate defenses. While many viruses have evolved mechanisms to antagonize the antiviral effects of the autophagy pathway, others subvert autophagy to facilitate replication. Here, we have investigated the role of autophagy in West Nile virus (WNV) replication. Experiments in cell lines derived from a variety of sources, including the kidney, liver, skin, and brain, indicated that WNV replication does not upregulate the autophagy pathway. Furthermore, WNV infection did not inhibit rapamycin-induced autophagy, suggesting that WNV does not disrupt the authophagy signaling cascade. Perturbation of the autophagy pathway by depletion of the major autophagy factors Atg5 or Atg7 had no effect on WNV infectious particle production, indicating that WNV does not require a functional autophagy pathway for replication. Taken together, the results of our study provide evidence that WNV, unlike several other viruses of the family Flaviviridae, does not significantly interact with the conventional autophagy pathway in mammalian cells.


Assuntos
Autofagia , Replicação Viral , Vírus do Nilo Ocidental/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Astrócitos/virologia , Proteína 5 Relacionada à Autofagia , Chlorocebus aethiops , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Fibroblastos/virologia , Técnicas de Silenciamento de Genes , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Cultura Primária de Células , Interferência de RNA , Proteína Sequestossoma-1 , Sindbis virus/fisiologia , Células Vero
10.
Virology ; 432(1): 232-8, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-22776165

RESUMO

The ability of viruses to control and/or evade the host antiviral response is critical to the establishment of a productive infection. One of the strategies utilized by West Nile virus (WNV) to circumvent the host response is to evade detection by the pathogen recognition receptor RIG-I early in infection. To begin elucidating the mechanisms by which WNV eludes detection, we undertook a systematic analysis of the WNV genome and antigenome to identify RIG-I-specific pathogen associated molecular patterns (PAMPs). Multiple segments of the WNV genome and anitigenome induced a RIG-I-specific antiviral response. However, incorporation of the stimulatory regions into larger RNAs substantially reduced their capacity to activate RIG-I. These results suggested that WNV evades the host response by sequestering RIG-I-specific PAMPs within the complete genome and antigenome at early times post-infection. Furthermore, activation of the RIG-I pathway may require the liberation of PAMPs by the cell's normal RNA processing pathways.


Assuntos
RNA Helicases DEAD-box/metabolismo , Genoma Viral , Receptores Imunológicos/metabolismo , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia , Linhagem Celular , Proteína DEAD-box 58 , Humanos , Evasão da Resposta Imune , Vírus do Nilo Ocidental/patogenicidade
11.
Viruses ; 2(2): 676-691, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21994652

RESUMO

The flavivirus genus includes viruses with a remarkable ability to produce disease on a large scale. The expansion and increased endemicity of dengue and West Nile viruses in the Americas exemplifies their medical and epidemiological importance. The rapid detection of viral infection and induction of the innate antiviral response are crucial to determining the outcome of infection. The intracellular pathogen receptors RIG-I and MDA5 play a central role in detecting flavivirus infections and initiating a robust antiviral response. Yet, these viruses are still capable of producing acute illness in humans. It is now clear that flaviviruses utilize a variety of mechanisms to modulate the interferon response. The non-structural proteins of the various flaviviruses reduce expression of interferon dependent genes by blocking phosphorylation, enhancing degradation or down-regulating expression of major components of the JAK/STAT pathway. Recent studies indicate that interferon modulation is an important factor in the development of severe flaviviral illness. This suggests that an increased understanding of viral-host interactions will facilitate the development of novel therapeutics to treat these viral infections and improved biological models to study flavivirus pathogenesis.

12.
J Virol ; 82(2): 609-16, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17977974

RESUMO

RIG-I and MDA5, two related pathogen recognition receptors (PRRs), are known to be required for sensing various RNA viruses. Here we investigated the roles that RIG-I and MDA5 play in eliciting the antiviral response to West Nile virus (WNV). Functional genomics analysis of WNV-infected fibroblasts from wild-type mice and RIG-I null mice revealed that the normal antiviral response to this virus occurs in two distinct waves. The initial response to WNV resulted in the expression of interferon (IFN) regulatory factor 3 target genes and IFN-stimulated genes, including several subtypes of alpha IFN. Subsequently, a second phase of IFN-dependent antiviral gene expression occurred very late in infection. In cells lacking RIG-I, both the initial and the secondary responses to WNV were delayed, indicating that RIG-I plays a critical role in initiating innate immunity against WNV. However, another PRR(s) was able to trigger a response to WNV in the absence of RIG-I. Disruption of both MDA5 and RIG-I pathways abrogated activation of the antiviral response to WNV, suggesting that MDA5 is involved in the host's defense against WNV infection. In addition, ablation of the function of IPS-1, an essential RIG-I and MDA5 adaptor molecule, completely disabled the innate antiviral response to WNV. Our data indicate that RIG-I and MDA5 are responsible for triggering downstream gene expression in response to WNV infection by signaling through IPS-1. We propose a model in which RIG-I and MDA5 operate cooperatively to establish an antiviral state and mediate an IFN amplification loop that supports immune effector gene expression during WNV infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , RNA Helicases DEAD-box/imunologia , Imunidade Inata , Transdução de Sinais , Vírus do Nilo Ocidental/imunologia , Animais , Células Cultivadas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/deficiência , Fibroblastos/virologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Helicase IFIH1 Induzida por Interferon , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Febre do Nilo Ocidental/imunologia
13.
J Virol ; 80(19): 9424-34, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973548

RESUMO

The emergence of West Nile virus (WNV) in the Western Hemisphere is marked by the spread of pathogenic lineage I strains, which differ from typically avirulent lineage II strains. To begin to understand the virus-host interactions that may influence the phenotypic properties of divergent lineage I and II viruses, we compared the genetic, pathogenic, and alpha/beta interferon (IFN-alpha/beta)-regulatory properties of a lineage II isolate from Madagascar (MAD78) with those of a new lineage I isolate from Texas (TX02). Full genome sequence analysis revealed that MAD78 clustered, albeit distantly, with other lineage II strains, while TX02 clustered with emergent North American isolates, more specifically with other Texas strains. Compared to TX02, MAD78 replicated at low levels in cultured human cells, was highly sensitive to the antiviral actions of IFN in vitro, and demonstrated a completely avirulent phenotype in wild-type mice. In contrast to TX02 and other pathogenic forms of WNV, MAD78 was defective in its ability to disrupt IFN-induced JAK-STAT signaling, including the activation of Tyk2 and downstream phosphorylation and nuclear translocation of STAT1 and STAT2. However, replication of MAD78 was rescued in cells with a nonfunctional IFN-alpha/beta receptor (IFNAR). Consistent with this finding, the virulence of MAD78 was unmasked upon infection of mice lacking IFNAR. Thus, control of the innate host response and IFN actions is a key feature of WNV pathogenesis and replication fitness.


Assuntos
Interferon-alfa/imunologia , Interferon beta/imunologia , Replicação Viral , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Linhagem Celular , Humanos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Camundongos , Dados de Sequência Molecular , Fenótipo , Filogenia , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/isolamento & purificação
14.
J Virol ; 80(6): 2913-23, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501100

RESUMO

The ability of viruses to control and/or evade the host antiviral response is critical to the establishment of a productive infection. We have previously shown that West Nile virus NY (WNV-NY) delays activation of interferon regulatory factor 3 (IRF-3), a transcription factor critical to the initiation of the antiviral response. Here we demonstrate that the delayed activation of IRF-3 is essential for WNV-NY to achieve maximum virus production. Furthermore, WNV-NY utilizes a unique mechanism to control activation of IRF-3. In contrast to many other viruses that impose a nonspecific block to the IRF-3 pathway, WNV-NY eludes detection by the host cell at early times postinfection. To better understand this process, we assessed the role of the pathogen recognition receptor (PRR) retinoic acid-inducible gene I (RIG-I) in sensing WNV-NY infection. RIG-I null mouse embryo fibroblasts (MEFs) retained the ability to respond to WNV-NY infection; however, the onset of the host response was delayed compared to wild-type (WT) MEFs. This suggests that RIG-I is involved in initially sensing WNV-NY infection, while other PRRs sustain and/or amplify the host response later in infection. The delayed initiation of the host response correlated with an increase in WNV-NY replication in RIG-I null MEFs compared to WT MEFs. Our data suggest that activation of the host response by RIG-I early in infection is important for controlling replication of WNV-NY. Furthermore, pathogenic strains of WNV may have evolved to circumvent stimulation of the host response until after replication is well under way.


Assuntos
Fator Regulador 3 de Interferon/antagonistas & inibidores , Transativadores/metabolismo , Ativação Transcricional , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Linhagem Celular , Humanos , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Ensaio de Placa Viral , Replicação Viral , Vírus do Nilo Ocidental/fisiologia
15.
J Virol ; 78(14): 7737-47, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220448

RESUMO

Recent outbreaks of West Nile Virus (WNV) have been associated with an increase in morbidity and mortality in humans, birds, and many other species. We have initiated studies to define the molecular mechanisms by which a recent pathogenic isolate of WNV evades the host cell innate antiviral response. Biochemical and microarray analyses demonstrated that WNV induced the expression of beta interferon (IFN-beta) and several IFN-stimulated genes late in infection of cultured human cells. The late expression of these antiviral genes was due to the delayed activation of the transcription factor IFN regulatory factor 3 (IRF-3). Despite this host response, WNV was still able to replicate efficiently. The effect of the IRF-3 pathway on WNV replication was assessed by examining virus replication and spread in cultures of wild-type or IRF-3-null mouse embryo fibroblasts. The absence of IRF-3 was marked by a significant increase in plaque size and a sustained production of infectious particles. Although the activation of the IRF-3 pathway was not sufficient to block virus replication, our results suggest that IRF-3 target genes function to constrain WNV infection and limit cell-to-cell virus spread.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Imunidade Inata , Fatores de Transcrição/metabolismo , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Fator Regulador 3 de Interferon , Interferon beta/metabolismo , Interferon beta/farmacologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Proteínas/genética , Proteínas/metabolismo , Fatores de Transcrição/genética , Células Vero , Ensaio de Placa Viral , Replicação Viral , Vírus do Nilo Ocidental/fisiologia
16.
J Virol ; 76(22): 11440-6, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388705

RESUMO

Productive entry of human immunodeficiency virus type 1 (HIV-1) into a host cell is believed to proceed via fusion of the viral envelope with the host cell's plasma membrane. Interestingly, the majority of HIV-1 particles that bind to the cell surface are taken up by the host cell via endocytosis; however, this mode of internalization generally does not result in infection. Presumably, virus particles remain trapped in the endocytic pathway and are eventually degraded. Here, we demonstrate that treatment of cells with various pharmacological agents known to elevate the pH of endosomes and lysosomes allows HIV-1 to efficiently enter and infect the host cell. Pretreatment of cells with bafilomycin A1 results in up to a 50-fold increase in the infectivity of HIV-1(SF2). Similarly, pretreatment of target cells with amantadine, concanamycin A, concanamycin B, chloroquine, and ammonium chloride resulted in increases in HIV-1 infectivity ranging between 2- and 15-fold. Analysis of receptor and coreceptor expression, HIV-long terminal repeat (LTR) transactivation, and transduction with amphotropic-pseudotyped murine leukemia virus (MLV)-based vectors suggests that the increase in infectivity is not artifactual. The increased infectivity under these conditions appears to be due to the ability of HIV-1 and MLV particles to enter via the endocytic pathway when spared from degradation in the late endosomes and lysosomes. These results could have significant implications for the administration of current and future lysosmotropic agents to patients with HIV disease.


Assuntos
Endocitose/efeitos dos fármacos , Endossomos/metabolismo , HIV-1/patogenicidade , Lisossomos/metabolismo , Macrolídeos , Amantadina/farmacologia , Animais , Antibacterianos/farmacologia , Linhagem Celular , Cloroquina/farmacologia , Endossomos/virologia , Repetição Terminal Longa de HIV , HIV-1/efeitos dos fármacos , HIV-1/genética , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Vírus da Leucemia Murina/genética , Lisossomos/virologia , Camundongos , Transdução Genética
17.
Microbes Infect ; 4(2): 189-99, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11880052

RESUMO

Primate lentiviruses encode a small protein designated Nef that has been shown to be a major determinant of virus pathogenicity. Nef regulates multiple host factors in order to optimize the cellular environment for virus replication. The mechanisms by which this small protein modulates distinct host cell properties provide intriguing insight into the intricate interaction between virus and host.


Assuntos
Produtos do Gene nef/fisiologia , HIV/patogenicidade , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Apoptose , Antígenos CD4/metabolismo , Regulação para Baixo , HIV/genética , HIV/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Ativação Linfocitária , Primatas/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...