Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(2): 588-93, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22203987

RESUMO

The ability of human CMV (HCMV) to enter and establish a latent infection in myeloid cells is crucial for survival and transmission in the human population. Initial pathogen binding and entry triggers a number of antiviral responses, including the activation of proapoptotic cell death pathways, which must be countered during latency establishment. However, mechanisms responsible for a prosurvival state in myeloid cells upon latent HCMV infection remain completely undefined. We hypothesized that the cellular antiapoptotic machinery must be initially activated by HCMV to promote early survival events upon entry. Here we show that HCMV transiently protects nonpermissive myeloid cells from chemical and virus entry induced cell death by up-regulating a key myeloid cell survival gene, myeloid cell leukemia (MCL)-1 protein. The induction of MCL-1 expression was independent of viral gene expression but dependent on activation of the ERK-MAPK pathway by viral glycoprotein B. Inhibition of ERK-MAPK signaling, inhibition of HCMV fusion, antibody-mediated neutralization of glycoprotein B signaling or expression of a shRNA against MCL-1 all correlated with increased cell death in response to virus infection or chemical stimulation. Finally we show that activation of ERK-MAPK signaling impacts on long-term latency and reactivation in hematopoietic cells. Thus, HCMV primes myeloid cells for from the initial virus-cell encounter. Given the importance of ERK and MCL-1 for myeloid cell survival, the successful establishment of HCMV latency in myeloid progenitors begins at the point of virus entry.


Assuntos
Infecções por Citomegalovirus/transmissão , Citomegalovirus/metabolismo , Regulação da Expressão Gênica/fisiologia , Células Mieloides/virologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/fisiologia , Internalização do Vírus , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Butadienos/farmacologia , Primers do DNA/genética , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Vetores Genéticos , Humanos , Imidazóis/farmacologia , Lentivirus , Proteína de Sequência 1 de Leucemia de Células Mieloides , Células Mieloides/metabolismo , Nitrilas/farmacologia , Piridinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas do Envelope Viral/metabolismo
2.
Proc Natl Acad Sci U S A ; 108(17): 6739-44, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21502533

RESUMO

The search for novel therapeutic interventions for viral disease is a challenging pursuit, hallmarked by the paucity of antiviral agents currently prescribed. Targeting of viral proteins has the inextricable challenge of rise of resistance. Safe and effective vaccines are not possible for many viral pathogens. New approaches are required to address the unmet medical need in this area. We undertook a cell-based high-throughput screen to identify leads for development of drugs to treat respiratory syncytial virus (RSV), a serious pediatric pathogen. We identified compounds that are potent (nanomolar) inhibitors of RSV in vitro in HEp-2 cells and in primary human bronchial epithelial cells and were shown to act postentry. Interestingly, two scaffolds exhibited broad-spectrum activity among multiple RNA viruses. Using the chemical matter as a probe, we identified the targets and identified a common cellular pathway: the de novo pyrimidine biosynthesis pathway. Both targets were validated in vitro and showed no significant cell cytotoxicity except for activity against proliferative B- and T-type lymphoid cells. Corollary to this finding was to understand the consequences of inhibition of the target to the host. An in vivo assessment for antiviral efficacy failed to demonstrate reduced viral load, but revealed microscopic changes and a trend toward reduced pyrimidine pools and findings in histopathology. We present here a discovery program that includes screen, target identification, validation, and druggability that can be broadly applied to identify and interrogate other host factors for antiviral effect starting from chemical matter of unknown target/mechanism of action.


Assuntos
Antivirais , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sinciciais Respiratórios/metabolismo , Animais , Antivirais/síntese química , Antivirais/química , Antivirais/farmacologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos B/virologia , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Cães , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Células Jurkat , Infecções por Vírus Respiratório Sincicial/patologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Linfócitos T/virologia , Células Vero
3.
J Virol ; 85(23): 12750-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21937636

RESUMO

Human cytomegalovirus (HCMV) remains a major cause of viral disease in immunosuppressed transplant patients. The ability of HCMV to establish lifelong infection in humans and reactivate with devastating clinical consequences underscores the importance of understanding the triggers of HCMV reactivation in mature myeloid cells. Dendritic cell (DC) differentiation is concomitant with the activation of cellular signaling pathways and inflammatory gene expression and also HCMV reactivation. Here, we show a major role for interleukin-6 (IL-6) through extracellular signal-regulated kinase-mitogen-activated protein kinase (ERK-MAPK) signaling upon DC differentiation to promote HCMV reactivation. IL-6 drives reactivation by transcriptional upregulation of the major immediate-early (IE) genes, resulting in efficient progression of the virus life cycle and, ultimately, higher titers of infectious virus. Furthermore, the interception of IL-6 signaling with biological inhibitors significantly abrogated HCMV reactivation from experimental latency. Crucially, using cells derived from healthy seropositive donors, we observed a key role for IL-6 during reactivation from natural latency ex vivo in interstitial DCs. Clinically, HCMV reactivation occurs in highly inflammatory environments (i.e., transplantation); thus, the implications of this study could potentially provide novel approaches for therapeutic intervention.


Assuntos
Infecções por Citomegalovirus/virologia , Células Dendríticas/virologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Interleucina-6/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ativação Viral/fisiologia , Western Blotting , Diferenciação Celular , Células Cultivadas , Citomegalovirus , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/metabolismo , Células Dendríticas/metabolismo , Regulação Viral da Expressão Gênica , Genes Precoces , Humanos , Interleucina-6/antagonistas & inibidores , Monócitos/citologia , Monócitos/metabolismo , Monócitos/virologia , Células Mieloides/citologia , Células Mieloides/metabolismo , Células Mieloides/virologia , RNA Mensageiro/genética , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Ativação Transcricional , Latência Viral
4.
J Virol ; 85(13): 6548-56, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21507975

RESUMO

Viral replication relies on the host to supply nucleosides. Host enzymes involved in nucleoside biosynthesis are potential targets for antiviral development. Ribavirin (a known antiviral drug) is such an inhibitor that suppresses guanine biosynthesis; depletion of the intracellular GTP pool was shown to be the major mechanism to inhibit flavivirus. Along similar lines, inhibitors of the pyrimidine biosynthesis pathway could be targeted for potential antiviral development. Here we report on a novel antiviral compound (NITD-982) that inhibits host dihydroorotate dehydrogenase (DHODH), an enzyme required for pyrimidine biosynthesis. The inhibitor was identified through screening 1.8 million compounds using a dengue virus (DENV) infection assay. The compound contains an isoxazole-pyrazole core structure, and it inhibited DENV with a 50% effective concentration (EC(50)) of 2.4 nM and a 50% cytotoxic concentration (CC(50)) of >5 µM. NITD-982 has a broad antiviral spectrum, inhibiting both flaviviruses and nonflaviviruses with nanomolar EC(90)s. We also show that (i) the compound inhibited the enzymatic activity of recombinant DHODH, (ii) an NITD-982 analogue directly bound to the DHODH protein, (iii) supplementing the culture medium with uridine reversed the compound-mediated antiviral activity, and (iv) DENV type 2 (DENV-2) variants resistant to brequinar (a known DHODH inhibitor) were cross resistant to NITD-982. Collectively, the results demonstrate that the compound inhibits DENV through depleting the intracellular pyrimidine pool. In contrast to the in vitro potency, the compound did not show any efficacy in the DENV-AG129 mouse model. The lack of in vivo efficacy is likely due to the exogenous uptake of pyrimidine from the diet or to a high plasma protein-binding activity of the current compound.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Pirimidinas/antagonistas & inibidores , Animais , Antivirais/química , Antivirais/farmacocinética , Chlorocebus aethiops , Efeito Citopatogênico Viral/efeitos dos fármacos , Dengue/virologia , Vírus da Dengue/enzimologia , Vírus da Dengue/patogenicidade , Vírus da Dengue/fisiologia , Di-Hidro-Orotato Desidrogenase , Modelos Animais de Doenças , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Pirimidinas/biossíntese , Sigmodontinae , Resultado do Tratamento , Células Vero , Replicação Viral/efeitos dos fármacos
6.
J Virol ; 84(14): 7185-94, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20444888

RESUMO

A putative latency-associated transcript (LUNA) complementary to the human cytomegalovirus (HCMV) UL81-82 region previously identified in seropositive donors' monocytes is also expressed during lytic infection. Thus, the LUNA promoter is active during both lytic and latent infection. Consequently, the mechanisms regulating this promoter may provide further insight into factors that determine whether the outcome of HCMV infection is latent or lytic. By transfection, the LUNA promoter exhibited low but reproducible activity. Substantial activation by virus infection suggested that a viral factor was important for LUNA expression during lytic infection. IE72, a known transactivator of viral promoters, activated the LUNA promoter in cotransfection assays. Furthermore, coinfection with wild-type HCMV but not an IE72 deletion virus (CR208) also activated the LUNA promoter. Finally, diminished LUNA gene expression in CR208 virus-infected cells supported a role for IE72 in LUNA gene expression. The initial regulation of herpesvirus immediate-early gene expression is associated with proteins found at cellular nuclear domain 10 (ND10) bodies, such as PML, hDaxx, and ATRX. hDaxx transfection repressed LUNA promoter activity. Furthermore, we observed binding of hDaxx to the LUNA promoter, which was abrogated by IE72 gene expression via direct interaction. Finally, we show that small interfering RNA (siRNA) knockdown of the hDaxx interaction partner ATRX rescued LUNA gene expression in CR208-infected cells. Overall, these data show that hDaxx/ATRX-mediated repression of LUNA during lytic infection absolutely requires IE72 gene expression. It also suggests that the targeting of cellular factors by IE72 is important throughout the different phases of HCMV gene expression during productive infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citomegalovirus/fisiologia , Regulação Viral da Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Virais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Proteínas Correpressoras , Citomegalovirus/genética , Humanos , Proteínas Imediatamente Precoces/genética , Recém-Nascido , Chaperonas Moleculares , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Transfecção , Proteínas Virais/genética , Latência Viral/genética
7.
J Virol ; 84(19): 10026-37, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660204

RESUMO

Cellular integrins were identified as human cytomegalovirus (HCMV) entry receptors and signaling mediators in both fibroblasts and endothelial cells. The goal of these studies was to determine the mechanism by which HCMV binds to cellular integrins to mediate virus entry. HCMV envelope glycoprotein B (gB) has sequence similarity to the integrin-binding disintegrin-like domain found in the ADAM (a disintegrin and metalloprotease) family of proteins. To test the ability of this region to bind to cellular integrins, we generated a recombinant soluble version of the gB disintegrin-like domain (gB-DLD). The gB-DLD protein bound to human fibroblasts in a specific, dose-dependent and saturable manner that required the expression of an intact beta1 integrin ectodomain. Furthermore, a physical association between gB-DLD and beta1 integrin was demonstrated through in vitro pull-down assays. The function of this interaction was shown by the ability of cell-bound gB-DLD to efficiently block HCMV entry and the infectivity of multiple in vivo target cells. Additionally, rabbit polyclonal antibodies raised against gB-DLD neutralized HCMV infection. Mimicry of the ADAM family disintegrin-like domain by HCMV gB represents a novel mechanism for integrin engagement by a virus and reveals a unique therapeutic target for HCMV neutralization. The strong conservation of the DLD across beta- and gammaherpesviruses suggests that integrin recognition and utilization may be a more broadly conserved feature throughout the Herpesviridae.


Assuntos
Citomegalovirus/fisiologia , Integrina beta1/fisiologia , Proteínas do Envelope Viral/fisiologia , Internalização do Vírus , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Sequência de Bases , Membrana Celular/fisiologia , Membrana Celular/virologia , Células Cultivadas , Citomegalovirus/genética , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Primers do DNA/genética , DNA Viral/genética , Desintegrinas/química , Desintegrinas/imunologia , Desintegrinas/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Domínios e Motivos de Interação entre Proteínas , Coelhos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
8.
Virol J ; 8: 329, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21711559

RESUMO

Host cyclophilin (cyp) inhibitors, such as NIM811, efficiently inhibit replication of hepatitis C virus (HCV) and have shown significant promise in recent clinical trials for the treatment of chronic HCV. It is therefore important to fully understand the mechanism of action of these therapeutic agents. Data obtained from comprehensive systems biology approaches have led to the hypothesis that the antiviral activity of cyclophilin inhibitors is mediated through impairing the cellular machinery on which HCV relies to traffic cofactors necessary for formation of the replication complex. Indeed, our results demonstrate when cyclophilins are inhibited by NIM811, lipid and protein trafficking within the VLDL pathway is impaired. Following treatment of replicon or HCV infected cells with NIM811, intracellular lipid droplets (LD) more than double in size and decrease in number. Changes in the LDs in response to cyclophilin inhibition are dependent upon expression of viral proteins. Additionally, in cells treated with NIM811, apoB accumulates in a crescent or ring shaped structure surrounding the enlarged LDs and is no longer secreted. Silencing of cypA or cyp40 using siRNA had a similar effect on LD size and apoB localization as compound treatment, suggesting these cyclophilins may play an important role in lipid and apoB trafficking. Interestingly, the decrease in apoB secretion correlates with a decrease in release of viral particles in HCV infected cells. Altogether, these results add a new level of complexity to the mechanism of action of cyclophilin inhibition, and suggest the role for cyclophilins in the virus life cycle extends beyond replication to virus release.


Assuntos
Antivirais/metabolismo , Apolipoproteínas B/metabolismo , Ciclofilinas/metabolismo , Ciclosporina/metabolismo , Hepacivirus/fisiologia , Liberação de Vírus , Ciclofilinas/antagonistas & inibidores , Humanos
9.
Antimicrob Agents Chemother ; 54(5): 1981-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20176894

RESUMO

The current standard of care for hepatitis C virus (HCV) infection, pegylated alpha interferon in combination with ribavirin, has a limited response rate and adverse side effects. Drugs targeting viral proteins are in clinical development, but they suffer from the development of high viral resistance. The inhibition of cellular proteins that are essential for viral amplification is thought to have a higher barrier to the emergence of resistance. Three cyclophilin inhibitors, the cyclosporine analogs DEBIO-025, SCY635, and NIM811, have shown promising results for the treatment of HCV infection in early clinical trials. In this study, we investigated the frequency and mechanism of resistance to cyclosporine (CsA), NIM811, and a structurally unrelated cyclophilin inhibitor, SFA-1, in replicon-containing Huh7 cells. Cross-resistance between all clones was observed. NIM811-resistant clones were selected only after obtaining initial resistance to either CsA or SFA-1. The time required to select resistance against cyclophilin inhibitors was significantly longer than that required for resistance selection against viral protein inhibitors, and the achievable resistance level was substantially lower. Resistance to cyclophilin inhibitors was mediated by amino acid substitutions in NS3, NS5A, and NS5B, with NS5A mutations conferring the majority of resistance. Mutation D320E in NS5A mediated most of the resistance conferred by NS5A. Taken together, the results indicate that there is a very low frequency and level of resistance to cyclophilin-binding drugs mediated by amino acid substitutions in three viral proteins. The interaction of cyclophilin with NS5A seems to be the most critical, since the NS5A mutations have the largest impact on resistance.


Assuntos
Ciclofilinas/antagonistas & inibidores , Ciclosporina/farmacologia , Farmacorresistência Viral/fisiologia , Hepacivirus/genética , Replicon/genética , Antivirais/farmacologia , Linhagem Celular , Ciclosporinas/farmacologia , Inibidores Enzimáticos/farmacologia , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/metabolismo , Humanos , Lactonas/farmacologia , Mutagênese Sítio-Dirigida , RNA Viral/metabolismo , Compostos de Espiro/farmacologia , Transfecção , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
10.
J Virol ; 83(8): 3891-903, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19193805

RESUMO

Glycoprotein B (gB) homologs are conserved throughout the family Herpesviridae and appear to serve essential, universal functions, as well as specific functions unique to a particular herpesvirus. Genetic analysis is a powerful tool to analyze protein function, and while it has been possible to generate virus mutants, complementation of essential virus knockouts has been problematic. Human cytomegalovirus (HCMV) gB (UL55) plays an essential role in the replication cycle of the virus. To define the function(s) of gB in HCMV infection, the BAC system was used to generate a recombinant virus in which the UL55 gene was replaced with galK (pAD/CreDeltaUL55). UL55 deletions in the viral genome have been made before, demonstrating that UL55 is an essential gene. However, without being able to successfully complement the genetic defect, a phenotypic analysis of the mutant virus was impossible. We generated fibroblasts expressing HCMV gB that complement pAD/CreDeltaUL55 and produce infectious virions lacking the UL55 gene but containing wild-type gB on the virion surface (DeltaUL55-gB HCMV). This is the first successful complementation of an HCMV mutant with a glycoprotein deleted. To characterize DeltaUL55 infection in the absence of gB, noncomplementing cells were infected with DeltaUL55-gB virus. All stages of gene expression were detected, and significant amounts of DNase-resistant viral DNA genomes, representing whole intact virions, were released into the infected cell supernatant. Gradient purification of these virions revealed they lacked gB but contained other viral structural proteins. The gB-null virions were able to attach to the cell surface similarly to wild-type gB-containing virions but were defective in virus entry and cell-to-cell spread. Glycoprotein B-null virions do, however, contain infectious DNA, as IE gene expression can be detected in fibroblasts following treatment of attached gB-null virions with a membrane fusion agent, polyethylene glycol. Taken together, our results indicate that gB is required for virus entry and cell-to-cell spread of the virus. However, HCMV gB is not absolutely required for virus attachment or assembly and egress from infected cells.


Assuntos
Citomegalovirus/fisiologia , Proteínas do Envelope Viral/fisiologia , Internalização do Vírus , Baculoviridae/genética , Células Cultivadas , Citomegalovirus/genética , Fibroblastos/virologia , Deleção de Genes , Genes Essenciais , Genes Virais , Teste de Complementação Genética , Humanos , Proteínas do Envelope Viral/genética , Montagem de Vírus , Ligação Viral
11.
J Virol ; 83(19): 10058-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19605471

RESUMO

Host factor pathways are known to be essential for hepatitis C virus (HCV) infection and replication in human liver cells. To search for novel host factor proteins required for HCV replication, we screened a subgenomic genotype 1b replicon cell line (Luc-1b) with a kinome and druggable collection of 20,779 siRNAs. We identified and validated several enzymes required for HCV replication, including class III phosphatidylinositol 4-kinases (PI4KA and PI4KB), carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD), and mevalonate (diphospho) decarboxylase. Knockdown of PI4KA could inhibit the replication and/or HCV RNA levels of the two subgenomic genotype 1b clones (SG-1b and Luc-1b), two subgenomic genotype 1a clones (SG-1a and Luc-1a), JFH-1 genotype 2a infectious virus (JFH1-2a), and the genomic genotype 1a (FL-1a) replicon. In contrast, PI4KB knockdown inhibited replication and/or HCV RNA levels of Luc-1b, SG-1b, and Luc-1a replicons. The small molecule inhibitor, PIK93, was found to block subgenomic genotype 1b (Luc-1b), subgenomic genotype 1a (Luc-1a), and genomic genotype 2a (JFH1-2a) infectious virus replication in the nanomolar range. PIK93 was characterized by using quantitative chemical proteomics and in vitro biochemical assays to demonstrate PIK93 is a bone fide PI4KA and PI4KB inhibitor. Our data demonstrate that genetic or pharmacological modulation of PI4KA and PI4KB inhibits multiple genotypes of HCV and represents a novel druggable class of therapeutic targets for HCV infection.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Fígado/virologia , Replicação Viral , 1-Fosfatidilinositol 4-Quinase/química , Antivirais/farmacologia , Ligação Competitiva , Linhagem Celular , Inativação Gênica , Genótipo , Humanos , Concentração Inibidora 50 , Espectrometria de Massas/métodos , Proteômica/métodos , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiazóis/farmacologia
12.
Trends Cell Biol ; 14(1): 5-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14729174

RESUMO

The ability of human cytomegalovirus (HCMV) to infect an extensive range of cell types has complicated efforts to identify cellular receptors for this significant pathogen. Recent findings demonstrate that epidermal growth factor receptor (EGFR) serves also as a receptor for HCMV. Additional evidence has shown that HCMV entry occurs in concert with immune detection through toll-like receptors. Here, the implications of EGFR activation, the existence of other receptors and the coordination of entry with the innate sensing are discussed.


Assuntos
Citomegalovirus/patogenicidade , Receptores ErbB/fisiologia , Glicoproteínas de Membrana/fisiologia , Receptores de Superfície Celular/fisiologia , Receptores Virais/metabolismo , Transporte Biológico , Citomegalovirus/metabolismo , Humanos , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Receptores Toll-Like , Proteínas do Envelope Viral/metabolismo
13.
Antimicrob Agents Chemother ; 52(9): 3267-75, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18591281

RESUMO

Chronic hepatitis C virus (HCV) infection remains a major global health burden while current interferon-based therapy is suboptimal. Efforts to develop more effective antiviral agents mainly focus on two viral targets: NS3-4A protease and NS5B polymerase. However, resistant mutants against these viral specific inhibitors emerge quickly both in vitro and in patients, particularly in the case of monotherapy. An alternative and complementary strategy is to target host factors such as cyclophilins that are also essential for viral replication. Future HCV therapies will most likely be combinations of multiple drugs of different mechanisms to maximize antiviral activity and to suppress the emergence of resistance. Here, the effects of combining a host cyclophilin inhibitor NIM811 with other viral specific inhibitors were investigated in vitro using HCV replicon. All of the combinations led to more pronounced antiviral effects than any single agent, with no significant increase of cytotoxicity. Moreover, the combination of NIM811 with a nucleoside (NM107) or a non-nucleoside (thiophene-2-carboxylic acid) polymerase inhibitor was synergistic, while the combination with a protease inhibitor (BILN2061) was additive. Resistant clones were selected in vitro with these inhibitors. Interestingly, it was much more difficult to develop resistance against NIM811 than viral specific inhibitors. No cross-resistance was observed among these inhibitors. Most notably, NIM811 was highly effective in blocking the emergence of resistance when used in combination with viral protease or polymerase inhibitors. Taken together, these results illustrate the significant advantages of combining inhibitors targeting both viral and host factors as key components of future HCV therapies.


Assuntos
Antivirais/farmacologia , Ciclosporina/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Inibidores de Proteases/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Antivirais/química , Linhagem Celular , Ciclofilinas/antagonistas & inibidores , Ciclosporina/química , Sinergismo Farmacológico , Quimioterapia Combinada , Hepacivirus/enzimologia , Hepacivirus/fisiologia , Humanos , Inibidores de Proteases/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos
14.
J Clin Virol ; 25 Suppl 2: S87-95, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12361760

RESUMO

BACKGROUND: All herpesviruses examined to date encode a heterodimeric envelope complex consisting of glycoprotein H (gH) and glycoprotein L (gL); however, co-expression of human cytomegalovirus (HCMV) gH and gL is not sufficient to reconstitute the high molecular weight complex seen in infected cells. Previously, we showed that HCMV encodes a third glycoprotein, gO, which associates with gH and gL to form an unusual tripartite complex. OBJECTIVES: The objective of this study was to reconstitute the HCMV gH-containing complex by co-expression of the gH (UL75), gL (UL115), and gO (UL74) genes. We co-expressed gH, gL, and gO in insect cells using a recombinant baculovirus, and in a mammalian system using triple plasmid transfection. Recombinant complexes from both systems were compared with those expressed in HCMV infected cells by SDS-PAGE and immunoblot or immunoprecipitation with antibodies to gH, gL, or gO. RESULTS: Insect cells infected with the triple gene baculovirus produced gH/gL heterodimers, gH/gL heteromultimers, and gO homomultimers, however, they did not produce detectable tripartite complex. In contrast, co-expression of gH, gL, and gO in mammalian cells produced high molecular weight complexes that closely resemble gH/gL/gO complexes formed in HCMV infected cells. Reduction of disulfide bonds resolved high molecular weight complexes into the three individual glycoproteins. Additionally, cell surface immunofluorescence proved that the complexes are expressed and displayed on the surface of transfected cells. CONCLUSIONS: Triple plasmid transfected cells produced high molecular weight complexes that co-migrated with endogenous HCMV gH/gL/gO complexes as analyzed by SDS-PAGE. In addition, several distinct, novel forms of the three glycoproteins were detected.


Assuntos
Citomegalovirus/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/patogenicidade , Imunofluorescência , Humanos , Immunoblotting , Insetos , Glicoproteínas de Membrana/genética , Transfecção , Proteínas do Envelope Viral/genética
15.
Antiviral Res ; 89(3): 246-56, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21277329

RESUMO

Human cytomegalovirus (HCMV) enters cells through a complex pathway involving the interaction of multiple viral glycoproteins and cellular receptors. While HCMV clinical isolates enter a wide range of cell types, entry has historically been studied using a laboratory strain of virus that can only infect fibroblasts. Herein, we have constructed a HCMV reporter strain that contains GFP fused to the abundant tegument protein pp65 to allow for the direct visualization of virus attachment and entry. Furthermore, the UL131 gene of this strain was restored to clinical isolate sequence to expand our studies of entry into physiologically relevant epithelial cell types. Using the HCMV-GFP reporter virus, we developed an image-based assay and screened a library containing 65,000 compounds for the inhibition of virus entry into fibroblasts. In addition to assessing the effect on virus entry, automated image analysis provided information on compound toxicity and whether the compounds acted as attachment or post-attachment inhibitors. To identify therapeutically viable inhibitors capable of blocking entry in multiple cell types, the inhibitors were screened further for their ability to inhibit virus entry into epithelial cells. Compounds were identified that were able to inhibit virus entry into both cell types at either attachment or post-attachment steps.


Assuntos
Antivirais/isolamento & purificação , Citomegalovirus/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Células Epiteliais/virologia , Fibroblastos/virologia , Virologia/métodos , Internalização do Vírus , Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Coloração e Rotulagem/métodos
16.
Cell Host Microbe ; 10(3): 197-209, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21925108

RESUMO

Human cytomegalovirus (CMV) is a common but difficult to treat infection of immunocompromised patients. MSL-109 is a human monoclonal IgG isolated from a CMV seropositive individual that recognizes the viral glycoprotein H (gH) surface antigen complexes that mediate entry. Although MSL-109 blocks CMV infection in vitro, it lacked sufficient efficacy in human trials, and CMV isolated from treated patients suggested the evolution of MSL-109 resistance. To understand how CMV escapes MSL-109, we characterized a MSL-109-resistant CMV strain. Our results elucidate a nongenetic escape mechanism in which the antibody is selectively taken up by infected cells and incorporated into assembling virions in a dose-dependent manner. The resistant virus then utilizes the Fc domain of the incorporated antibody to infect naive nonimmune cells. This resistance mechanism may explain the clinical failure of MSL-109, illustrate a general mechanism of viral antibody escape, and inform antiviral vaccine and therapeutic development.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Interações Hospedeiro-Patógeno , Vírion/fisiologia , Montagem de Vírus , Anticorpos Antivirais/metabolismo , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Humanos , Vírion/genética , Vírion/imunologia
17.
Virology ; 397(1): 43-55, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19932913

RESUMO

Three cyclophilin inhibitors (DEBIO-025, SCY635, and NIM811) are currently in clinical trials for hepatitis C therapy. The mechanism of action of these, however, is not completely understood. There are at least 16 cyclophilins expressed in human cells which are involved in a diverse set of cellular processes. Large-scale siRNA experiments, chemoproteomic assays with cyclophilin binding compounds, and mRNA profiling of HCV replicon containing cells were used to identify the cyclophilins that are instrumental to HCV replication. The previously reported cyclophilin A was confirmed and additional cyclophilin containing pathways were identified. Together, the experiments provide strong evidence that NIM811 reduces viral replication by inhibition of multiple cyclophilins and pathways with protein trafficking as the most strongly and persistently affected pathway.


Assuntos
Ciclofilinas/metabolismo , Hepacivirus/fisiologia , Interações Hospedeiro-Patógeno , Replicação Viral , Antivirais/química , Antivirais/farmacologia , Linhagem Celular , Ciclosporina/química , Ciclosporina/farmacologia , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Modelos Biológicos , Estrutura Molecular , Proteoma/análise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
18.
J Immunol ; 180(7): 4965-77, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18354222

RESUMO

Infection of permissive fibroblasts with human CMV (HCMV, AD169) is accompanied by a robust activation of innate immune defense. In this study, we show that inflammatory cytokine (IC) secretion and activation of the type I IFN pathway (alphabeta IFN) are initiated through distinct mechanisms. HCMV is recognized by TLR2 leading to the NF-kappaB activation and IC secretion. However, the IFN response to HCMV is not a TLR2-dependent process, as a dominant negative TLR2 does not affect the antiviral response to infection. Additionally, bafilomycin, an endosomal acidification inhibitor, has no effect on HCMV-induced IFN responses suggesting that IFN signaling is independent of endosomal resident TLRs. By contrast, disruption of lipid rafts by depletion of cellular cholesterol inhibits both HCMV entry as well as IFN responses. Cholesterol depletion had no effect on the induction of ICs by HCMV, illustrating a biological distinction at the cellular level with the initiation of innate immune pathways. Furthermore, HCMV entry inhibitors block IFN responses but not IC signaling. In particular, blocking the interaction of HCMV with beta(1) integrin diminished IFN signaling, suggesting that this virus-cell interaction or subsequent downstream steps in the entry pathway are critical for downstream signal transduction events. These data show that HCMV entry and IFN signaling are coordinated processes that require cholesterol-rich microdomains, whereas IC signaling is activated through outright sensing via TLR2. These findings further highlight the complexity and sophistication of innate immune responses at the earliest points in HCMV infection.


Assuntos
Citomegalovirus/imunologia , Imunidade Inata/imunologia , Ácidos/metabolismo , Linhagem Celular , Colesterol/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Fibroblastos , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon beta/farmacologia , Microssomos/metabolismo , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Internalização do Vírus
19.
J Virol ; 81(12): 6241-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17428848

RESUMO

Human cytomegalovirus (HCMV) can bind, fuse, and initiate gene expression in a diverse range of vertebrate cell types. This broad cellular tropism suggests that multiple receptors and/or universally distributed receptors mediate HCMV entry. Our laboratory has recently discovered that certain beta1 and beta3 integrin heterodimers are critical mediators of HCMV entry into permissive fibroblasts (A. L. Feire, H. Koss, and T. Compton, Proc. Natl. Acad. Sci. USA 101:15470-15475, 2004). It has also been reported that epidermal growth factor receptor (EGFR) is necessary for HCMV-mediated signaling and entry (X. Wang, S. M. Huong, M. L. Chiu, N. Raab-Traub, and E. E. Huang, Nature 424:456-461, 2003). Integrins are known to signal synergistically with growth factor receptors, and this coordination was recently reported for EGFR and beta3 integrins in the context of HCMV entry (X. Wang, D. Y. Huang, S. M. Huong, and E. S. Huang, Nat. Med. 11:515-521, 2005). However, EGFR-negative cell lines, such as hematopoietic cells, are known to be infected by HCMV. Therefore, we wished to confirm a role for EGFR in HCMV entry and then examine any interaction between beta1 integrins and EGFR during the entry process. Surprisingly, we were unable to detect any role for EGFR in the process of HCMV entry into fibroblast, epithelial, or endothelial cell lines. Additionally, HCMV did not activate the EGFR kinase in fibroblast cell lines. We first examined HCMV entry into two EGFR-positive or -negative cell lines but observed no increase in entry when EGFR was expressed to high levels. Physically blocking EGFR with a neutralizing antibody in fibroblast, epithelial, or endothelial cell lines or blocking EGFR kinase signaling with a chemical inhibitor in fibroblast cells did not inhibit virus entry. Lastly, we were unable to detect phosphorylation of EGFR in fibroblasts cells in response to HCMV stimulation. Our findings demonstrate that EGFR does not play a significant role in HCMV entry or signaling. These results suggest that specific integrin heterodimers either act alone as the primary entry receptors or interact in conjunction with an additional receptor(s), other than EGFR, to facilitate virus entry.


Assuntos
Citomegalovirus/metabolismo , Receptores ErbB/fisiologia , Internalização do Vírus , Fusão Celular , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Fibroblastos/virologia , Humanos , Testes de Neutralização , Quinazolinas , Transdução de Sinais , Fatores de Tempo , Tirfostinas/farmacologia , Proteínas Virais/metabolismo
20.
J Virol ; 80(22): 11105-14, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16956942

RESUMO

The Kaposi's sarcoma-associated herpesvirus (KSHV) envelope glycoprotein gpK8.1 contributes to cellular attachment through binding cell surface heparan sulfate proteoglycans. By using a soluble recombinant form of gpK8.1, we discovered that a consequence of gpK8.1 interaction with human fibroblasts is the induction of an antiviral response, as characterized by the activation of interferon regulatory factor 3 (IRF-3), production of interferon beta (IFN-beta), and expression of interferon-stimulated antiviral genes. In contrast, neither IFN-beta expression nor a functional antiviral response is observed in cells treated with KSHV virions. The interferon response induced by soluble gpK8.1 can be inhibited by simultaneous treatment with UV-inactivated virions, while the induction of an indicator inflammatory cytokine, interleukin-6, was readily evident in the response to both gpK8.1 and KSHV. In addition, KSHV virions abrogate gpK8.1-mediated activation of IRF-3, an early transcriptional regulator for cellular antiviral responses. Although innate immune responses are initiated during contact between gpK8.1 and cellular receptor(s), these results suggest that the virion contains one or more structural elements that selectively repress an effective antiviral response while allowing cellular responses favorable to the KSHV life cycle.


Assuntos
Glicoproteínas/imunologia , Herpesvirus Humano 8/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Interferons/biossíntese , Proteínas Virais/imunologia , Vírion/imunologia , Linhagem Celular , Fibroblastos/virologia , Regulação da Expressão Gênica , Herpesvirus Humano 8/crescimento & desenvolvimento , Humanos , Fator Regulador 3 de Interferon/biossíntese , Interleucina-6/biossíntese , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA