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










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 13(8)2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34452519

RESUMO

Strategies to combat COVID-19 require multiple ways to protect vulnerable people from infection. SARS-CoV-2 is an airborne pathogen and the nasal cavity is a primary target of infection. The K18-hACE2 mouse model was used to investigate the anti-SARS-CoV-2 efficacy of astodrimer sodium formulated in a mucoadhesive nasal spray. Animals received astodrimer sodium 1% nasal spray or PBS intranasally, or intranasally and intratracheally, for 7 days, and they were infected intranasally with SARS-CoV-2 after the first product administration on Day 0. Another group was infected intranasally with SARS-CoV-2 that had been pre-incubated with astodrimer sodium 1% nasal spray or PBS for 60 min before the neutralisation of test product activity. Astodrimer sodium 1% significantly reduced the viral genome copies (>99.9%) and the infectious virus (~95%) in the lung and trachea vs. PBS. The pre-incubation of SARS-CoV-2 with astodrimer sodium 1% resulted in a significant reduction in the viral genome copies (>99.9%) and the infectious virus (>99%) in the lung and trachea, and the infectious virus was not detected in the brain or liver. Astodrimer sodium 1% resulted in a significant reduction of viral genome copies in nasal secretions vs. PBS on Day 7 post-infection. A reduction in the viral shedding from the nasal cavity may result in lower virus transmission rates. Viraemia was low or undetectable in animals treated with astodrimer sodium 1% or infected with treated virus, correlating with the lack of detectable viral replication in the liver. Similarly, low virus replication in the nasal cavity after treatment with astodrimer sodium 1% potentially protected the brain from infection. Astodrimer sodium 1% significantly reduced the pro-inflammatory cytokines IL-6, IL-1α, IL-1ß, TNFα and TGFß and the chemokine MCP-1 in the serum, lung and trachea vs. PBS. Astodrimer sodium 1% nasal spray blocked or reduced SARS-CoV-2 replication and its sequelae in K18-hACE2 mice. These data indicate a potential role for the product in preventing SARS-CoV-2 infection or for reducing the severity of COVID-19.


Assuntos
Antivirais/administração & dosagem , Tratamento Farmacológico da COVID-19 , Dendrímeros/administração & dosagem , Cavidade Nasal/virologia , Sprays Nasais , Polilisina/administração & dosagem , SARS-CoV-2/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Antivirais/uso terapêutico , Encéfalo/virologia , COVID-19/prevenção & controle , COVID-19/virologia , Dendrímeros/uso terapêutico , Modelos Animais de Doenças , Feminino , Fígado/virologia , Masculino , Camundongos , Camundongos Transgênicos , Polilisina/uso terapêutico , Sistema Respiratório/virologia , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/fisiologia , Carga Viral/efeitos dos fármacos , Viremia , Replicação Viral/efeitos dos fármacos
2.
Antiviral Res ; 192: 105122, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34186107

RESUMO

There are, besides remdesivir, no approved antivirals for the treatment of SARS-CoV-2 infections. To aid in the search for antivirals against this virus, we explored the use of human tracheal airway epithelial cells (HtAEC) and human small airway epithelial cells (HsAEC) grown at the air-liquid interface (ALI). These cultures were infected at the apical side with one of two different SARS-CoV-2 isolates. Each virus was shown to replicate to high titers for extended periods of time (at least 8 days) and, in particular an isolate with the D614G in the spike (S) protein did so more efficiently at 35 °C than 37 °C. The effect of a selected panel of reference drugs that were added to the culture medium at the basolateral side of the system was explored. Remdesivir, GS-441524 (the parent nucleoside of remdesivir), EIDD-1931 (the parent nucleoside of molnupiravir) and IFN (ß1 and λ1) all resulted in dose-dependent inhibition of viral RNA and infectious virus titers collected at the apical side. However, AT-511 (the free base form of AT-527 currently in clinical testing) failed to inhibit viral replication in these in vitro primary cell models. Together, these results provide a reference for further studies aimed at selecting SARS-CoV-2 inhibitors for further preclinical and clinical development.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , SARS-CoV-2/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos/métodos , Células Epiteliais/virologia , Humanos , RNA Viral , SARS-CoV-2/isolamento & purificação , Células Vero
3.
Antiviral Res ; 191: 105089, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34010661

RESUMO

An effective response to the ongoing coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will involve a range of complementary preventive modalities. The current studies were conducted to evaluate the in vitro SARS-CoV-2 antiviral and virucidal (irreversible) activity of astodrimer sodium, a dendrimer with broad spectrum antimicrobial activity, including against enveloped viruses in in vitro and in vivo models, that is marketed for antiviral and antibacterial applications. We report that astodrimer sodium inhibits replication of SARS-CoV-2 in Vero E6 and Calu-3 cells, with 50% effective concentrations (EC50) for i) reducing virus-induced cytopathic effect of 0.002-0.012 mg/mL in Vero E6 cells, and ii) infectious virus release by plaque assay of 0.019-0.032 mg/mL in Vero E6 cells and 0.030-0.037 mg/mL in Calu-3 cells. The selectivity index (SI) in these assays was as high as 2197. Astodrimer sodium was also virucidal, irreversibly reducing SARS-CoV-2 infectivity by >99.9% (>3 log10) within 1 min of exposure, and up to >99.999% (>5 log10) shown at astodrimer sodium concentrations of 10-30 mg/mL in Vero E6 and Calu-3 cell lines. Astodrimer sodium also inhibited infection in a primary human airway epithelial cell line. The data were similar for all investigations and were consistent with the potent antiviral and virucidal activity of astodrimer sodium being due to irreversible inhibition of virus-host cell interactions, as previously demonstrated for other viruses. Further studies will confirm if astodrimer sodium binds to SARS-CoV-2 spike protein and physically blocks initial attachment of the virus to the host cell. Given the in vitro effectiveness and significantly high SI, astodrimer sodium warrants further investigation for potential as a topically administered agent for SARS-CoV-2 therapeutic applications.


Assuntos
Antivirais/farmacologia , Dendrímeros/farmacologia , Polilisina/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Efeito Citopatogênico Viral , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Células Vero
4.
PLoS One ; 11(4): e0152036, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27104614

RESUMO

Shortened current direct-acting antiviral (DAA) therapies while less expensive, have not provided satisfactory efficacy in naïve cirrhotics, treatment experienced non-cirrhotics or even genotype-3 (GT3)-infected patients. Since DAA regimens consist of the same classes of inhibitors-NS5A (NS5Ai) and NS5B (NS5Bi) +/- NS3 (NS3i) inhibitors-it is likely that their costs will be high and will provide similar degrees of protection. Integrating drugs with distinct mechanisms of action (MoA) into DAA regimens could provide the solution for shortening the period of treatment. One such class of agents is the cyclophilin inhibitors (CypI), which has shown efficacy in patients. Resistance-associated variants persist for years post-treatment in patients exposed to NS5Ai or NS5Bi who fail to achieve a sustained virologic response, impairing their chance for cure on retreatment with existing DAA combinations. Because of their high barrier to resistance, CypI may be particularly useful as a rescue therapy for patients who have relapsed with DAA resistance-associated variants. In this study, we analyzed the anti-HCV properties of the novel cyclosporine A (CsA) derivate-STG-175. The non-immunosuppressive STG-175 possesses a high (EC50 11.5-38.9 nM) multi-genotypic (GT1a to 4a) anti-HCV activity. STG-175 clears cells from HCV since no viral replication rebound was observed after cessation of drug treatment. It presents a higher barrier to resistance than other CypI or selected DAAs. HCV variants, which emerged under STG-175 pressure, are only ~2-fold resistant to the drug. No cross-resistance was observed with DAAs STG-175 was efficacious against DAA-resistant HCV variants. Drug combination studies revealed that STG-175 provides additive and synergistic effects against GT1a to 4a. STG-175 inhibits the infection of HCV, HIV-1 and HBV in mono-, dual- and triple-infection settings. Altogether these results suggest that the new CypI STG-175 represents an attractive drug partner for IFN-free DAA regimens for the treatment of HCV and co-infections.


Assuntos
Antivirais/farmacologia , Ciclofilinas/antagonistas & inibidores , Ciclosporinas/farmacologia , Hepacivirus/efeitos dos fármacos , Linhagem Celular , Humanos
5.
PLoS One ; 10(8): e0134707, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26263487

RESUMO

HCV-related liver disease is the main cause of morbidity and mortality of HCV/HIV-1 co-infected patients. Despite the recent advent of anti-HCV direct acting antivirals (DAAs), the treatment of HCV/HIV-1 co-infected patients remains a challenge, as these patients are refractory to most therapies and develop liver fibrosis, cirrhosis and liver cancer more often than HCV mono-infected patients. Until the present study, there was no suitable in vitro assay to test the inhibitory activity of drugs on HCV/HIV-1 co-infection. Here we developed a novel in vitro "co-infection" model where HCV and HIV-1 concurrently replicate in their respective main host target cells--human hepatocytes and CD4+ T-lymphocytes. Using this co-culture model, we demonstrate that cyclophilin inhibitors (CypI), including a novel cyclosporin A (CsA) analog, CPI-431-32, simultaneously inhibits replication of both HCV and HIV-1 when added pre- and post-infection. In contrast, the HIV-1 protease inhibitor nelfinavir or the HCV NS5A inhibitor daclatasvir only blocks the replication of a single virus in the "co-infection" system. CPI-431-32 efficiently inhibits HCV and HIV-1 variants, which are normally resistant to DAAs. CPI-431-32 is slightly, but consistently more efficacious than the most advanced clinically tested CypI--alisporivir (ALV)--at interrupting an established HCV/HIV-1 co-infection. The superior antiviral efficacy of CPI-431-32 over ALV correlates with its higher potency inhibition of cyclophilin A (CypA) isomerase activity and at preventing HCV NS5A-CypA and HIV-1 capsid-CypA interactions known to be vital for replication of the respective viruses. Moreover, we obtained evidence that CPI-431-32 prevents the cloaking of both the HIV-1 and HCV genomes from cellular sensors. Based on these results, CPI-431-32 has the potential, as a single agent or in combination with DAAs, to inhibit both HCV and HIV-1 infections.


Assuntos
Antivirais/farmacologia , Ciclofilinas/antagonistas & inibidores , Ciclosporinas/farmacologia , HIV-1/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Coinfecção , Ciclofilinas/metabolismo , Farmacorresistência Viral/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Humanos , Ligação Proteica/efeitos dos fármacos , Transcrição Reversa/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
6.
PLoS One ; 9(2): e88866, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24533158

RESUMO

Hepatitis C virus (HCV) infection is a major cause of liver disease. The molecular machinery of HCV assembly and particle release remains obscure. A better understanding of the assembly events might reveal new potential antiviral strategies. It was suggested that the nonstructural protein 5A (NS5A), an attractive recent drug target, participates in the production of infectious particles as a result of its interaction with the HCV core protein. However, prior to the present study, the NS5A-binding site in the viral core remained unknown. We found that the D1 domain of core contains the NS5A-binding site with the strongest interacting capacity in the basic P38-K74 cluster. We also demonstrated that the N-terminal basic residues of core at positions 50, 51, 59 and 62 were required for NS5A binding. Analysis of all substitution combinations of R50A, K51A, R59A, and R62A, in the context of the HCVcc system, showed that single, double, triple, and quadruple mutants were fully competent for viral RNA replication, but deficient in secretion of viral particles. Furthermore, we found that the extracellular and intracellular infectivity of all the mutants was abolished, suggesting a defect in the formation of infectious particles. Importantly, we showed that the interaction between the single and quadruple core mutants and NS5A was impaired in cells expressing full-length HCV genome. Interestingly, mutations of the four basic residues of core did not alter the association of core or NS5A with lipid droplets. This study showed for the first time that basic residues in the D1 domain of core that are critical for the formation of infectious extracellular and intracellular particles also play a role in core-NS5A interactions.


Assuntos
Hepacivirus/metabolismo , Proteínas do Core Viral/química , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Hepacivirus/patogenicidade , Humanos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas do Core Viral/genética
7.
Viruses ; 5(3): 981-97, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23524389

RESUMO

DEB025 (alisporivir) is a synthetic cyclosporine with inhibitory activity against human immunodeficiency virus type-1 (HIV-1) and hepatitis C virus (HCV). It binds to cyclophilin A (CypA) and blocks essential functions of CypA in the viral replication cycles of both viruses. DEB025 inhibits clinical HIV-1 isolates in vitro and decreases HIV-1 virus load in the majority of patients. HIV-1 isolates being naturally resistant to DEB025 have been detected in vitro and in nonresponder patients. By sequence analysis of their capsid protein (CA) region, two amino acid polymorphisms that correlated with DEB025 resistance were identified: H87Q and I91N, both located in the CypA-binding loop of the CA protein of HIV-1. The H87Q change was by far more abundant than I91N. Additional polymorphisms in the CypA-binding loop (positions 86, 91 and 96), as well as in the N-terminal loop of CA were detected in resistant isolates and are assumed to contribute to the degree of resistance. These amino acid changes may modulate the conformation of the CypA-binding loop of CA in such a way that binding and/or isomerase function of CypA are no longer necessary for virus replication. The resistant HIV-1 isolates thus are CypA-independent.


Assuntos
Fármacos Anti-HIV/farmacologia , Proteínas do Capsídeo/genética , Farmacorresistência Viral , Infecções por HIV/virologia , HIV-1/genética , Polimorfismo Genético , Sequência de Aminoácidos , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Ciclosporina , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , HIV-1/metabolismo , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto/efeitos dos fármacos
8.
Pharm Res ; 29(11): 3156-68, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22736232

RESUMO

PURPOSE: Use of coital-dependent products to prevent HIV-1 transmission has resulted in mixed success. We hypothesize that incorporation of antiviral drug candidates into a novel controlled delivery system will prolong their activity, making their use coital independent, thus increasing their chance of prophylactic success. METHODS: Tenofovir, emtricitabine, and C5A peptide HIV microbicides were mechanically incorporated into matrices comprising a series of subliming solids. Matrix sublimation rates and drug release rates were measured in three in vitro and one in vivo environments intended to model human vaginal interior. Antiviral activity studies evaluating matrix incorporated microbicides were performed using in vitro cell cultures and human ectocervical explants. RESULTS: Drug release rates were identical to matrix sublimation rates, and were zero order. Differences in matrix material sublimation enthalpies determined drug release and matrix erosion rates in a thermodynamically definable manner, in vitro and in vivo. Durations of release ranging from several days to several months were readily achieved. Prolonged duration of anti HIV-1 activity was shown for matrix incorporated microbicides, using ectocervical explant and cell culture models of HIV-1 infection. CONCLUSION: Subliming solid matrices show promise as a delivery system providing multi month intravaginal release of a wide range of HIV-1 microbicides.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/química , Sistemas de Liberação de Medicamentos/métodos , Infecções por HIV/prevenção & controle , Adenina/administração & dosagem , Adenina/análogos & derivados , Animais , Células Cultivadas , Dispositivos Anticoncepcionais Femininos , Preparações de Ação Retardada , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Emtricitabina , Feminino , Infecções por HIV/transmissão , HIV-1/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Organofosfonatos/administração & dosagem , Ovinos , Relação Estrutura-Atividade , Sublimação Química , Linfócitos T/efeitos dos fármacos , Tenofovir
9.
PLoS One ; 6(5): e18917, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21573158

RESUMO

Genital herpes is a major risk factor in acquiring human immunodeficiency virus type-1 (HIV-1) infection and is caused by both Herpes Simplex virus type 1 (HSV-1) and HSV-2. The amphipathic peptide C5A, derived from the non-structural hepatitis C virus (HCV) protein 5A, was shown to prevent HIV-1 infection but neither influenza nor vesicular stomatitis virus infections. Here we investigated the antiviral function of C5A on HSV infections. C5A efficiently inhibited both HSV-1 and HSV-2 infection in epithelial cells in vitro as well as in an ex vivo epidermal infection model. C5A destabilized the integrity of the viral HSV membrane. Furthermore, drug resistant HSV strains were inhibited by this peptide. Notably, C5A-mediated neutralization of HSV-1 prevented HIV-1 transmission. An in vitro HIV-1 transmigration assay was developed using primary genital epithelial cells and HSV infection increased HIV-1 transmigration. Treatment with C5A abolished HIV-1 transmigration by preventing HSV infection and by preserving the integrity of the genital epithelium that was severely compromised by HSV infection. In conclusion, this study demonstrates that C5A represents a multipurpose microbicide candidate, which neutralizes both HIV-1 and HSV, and which may interfere with HIV-1 transmission through the genital epithelium.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Células Cultivadas , Chlorocebus aethiops , HIV-1/efeitos dos fármacos , Humanos , Células Vero
10.
J Hepatol ; 53(1): 50-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20451281

RESUMO

BACKGROUND & AIMS: The cyclophilin (Cyp) inhibitors - cyclosporine A (CsA), NIM811, Debio 025, and SCY 635 - block HCV replication both in vitro and in vivo, and represent a novel class of potent anti-HCV agents. We and others showed that HCV relies on cyclophilin A (CypA) to replicate. We demonstrated that the hydrophobic pocket of CypA, where Cyp inhibitors bind, and which controls the isomerase activity of CypA, is critical for HCV replication. Recent studies showed that under Cyp inhibitor selection, mutations arose in the HCV nonstructural 5A (NS5A) protein. This led us to postulate that CypA assists HCV by acting on NS5A. METHODS: We tested this hypothesis by developing several interaction assays including GST pull-down assays, ELISA, and mammalian two-hybrid binding assays. RESULTS: We demonstrated that full-length NS5A and CypA form a stable complex. Remarkably, CsA prevents the CypA-NS5A interaction in a dose-dependent manner. Importantly, the CypA-NS5A interaction is conserved among genotypes and is interrupted by CsA. Surprisingly, the NS5A mutant protein, which arose in CsA-resistant HCV variants, behaves similarly to wild-type NS5A in terms of both CypA binding and CsA-mediated release from CypA. This latter finding suggests that HCV resistance to CsA does not correlate with a resistance of the CypA-NS5A interaction to Cyp inhibitors. Moreover, we found that CypA, devoid of its isomerase activity, fails to bind NS5A. CONCLUSIONS: Altogether these data suggest that CypA, via its isomerase pocket, binds directly to NS5A, and most importantly, that disrupting this interaction stops HCV replication.


Assuntos
Ciclofilina A/antagonistas & inibidores , Ciclosporina/farmacologia , Hepacivirus/efeitos dos fármacos , Proteínas não Estruturais Virais/efeitos dos fármacos , Substituição de Aminoácidos , Antivirais/farmacologia , Sítios de Ligação , Ciclofilina A/química , Ciclofilina A/genética , Farmacorresistência Viral/genética , Genes Virais , Hepacivirus/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Cinética , Mutagênese Sítio-Dirigida , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
11.
Antimicrob Agents Chemother ; 54(7): 2753-66, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20439611

RESUMO

In the absence of a vaccine, there is an urgent need for the development of safe and effective topical microbicides to prevent the sexual transmission of human immunodeficiency virus type 1 (HIV-1). In this study, we proposed to develop a novel class of microbicides using syndecan as the antiviral agent. Specifically, we generated a soluble syndecan-Fc hybrid molecule by fusing the ectodomain of syndecan-1 to the Fc domain of a human IgG. We then tested the syndecan-Fc hybrid molecule for various in vitro microbicidal anti-HIV-1 properties. Remarkably, the syndecan-Fc hybrid molecule possesses multiple attractive microbicidal properties: (i) it blocks HIV-1 infection of primary targets including T cells, macrophages, and dendritic cells (DC); (ii) it exhibits a broad range of antiviral activity against primary HIV-1 isolates, multidrug resistant HIV-1 isolates, HIV-2, and simian immunodeficiency virus (SIV); (iii) it prevents transmigration of HIV-1 through human primary genital epithelial cells; (iv) it prevents HIV-1 transfer from dendritic cells to CD4(+) T cells; (v) it is potent when added 2 h prior to addition of HIV-1 to target cells; (vi) it is potent at a low pH; (vii) it blocks HIV-1 infectivity when diluted in genital fluids; and (viii) it prevents herpes simplex virus infection. The heparan sulfate chains of the syndecan-Fc hybrid molecule are absolutely required for HIV-1 neutralization. Several lines of evidence suggest that the highly conserved Arg298 in the V3 region of gp120 serves as the locus for the syndecan-Fc hybrid molecule neutralization. In conclusion, this study suggests that the syndecan-Fc hybrid molecule represents the prototype of a new generation of microbicidal agents that may have promise for HIV-1 prevention.


Assuntos
Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Sindecana-1/metabolismo , Sindecana-1/farmacologia , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Células Dendríticas/virologia , Ensaio de Imunoadsorção Enzimática , Infecções por HIV/tratamento farmacológico , Infecções por HIV/prevenção & controle , Humanos , Concentração de Íons de Hidrogênio , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Macrófagos/virologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Sindecana-1/genética , Sindecana-1/uso terapêutico , Linfócitos T/virologia , Integração Viral/efeitos dos fármacos
12.
J Gen Virol ; 91(Pt 5): 1189-93, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20107018

RESUMO

The mechanisms by which cyclophilin A (CypA) governs hepatitis C virus (HCV) replication remain unknown. Since CypA binds two essential components of the HCV replication complex (RC)--the polymerase NS5B and the phosphoprotein NS5A--we asked in this study whether CypA regulates their RC association. We found that CypA, via its isomerase pocket, locates in a protease-resistant compartment similar to that where HCV replicates. CypA association with this compartment is not mediated by HCV. Moreover, CypA depletion of RC does not influence NS5A and NS5B RC association, arguing against a model where CypA governs HCV replication by recruiting NS5A or NS5B into RC.


Assuntos
Ciclofilina A/metabolismo , Hepacivirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Linhagem Celular , Hepatócitos/virologia , Humanos
13.
Proc Natl Acad Sci U S A ; 105(14): 5525-30, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18378908

RESUMO

In the absence of an effective vaccine, there is an urgent need for safe and effective antiviral agents to prevent transmission of HIV. Here, we report that an amphipathic alpha-helical peptide derived from the hepatitis C virus NS5A anchor domain (designated C5A in this article) that has been shown to be virocidal for the hepatitis C virus (HCV) also has potent antiviral activity against HIV. C5A exhibits a broad range of antiviral activity against HIV isolates, and it prevents infection of the three in vivo targets of HIV: CD4(+) T lymphocytes, macrophages, and dendritic cells by disrupting the integrity of the viral membrane and capsid core while preserving the integrity of host membranes. C5A can interrupt an ongoing T cell infection, and it can prevent transmigration of HIV through primary genital epithelial cells, infection of mucosal target cells and transfer from dendritic cells to T cells ex vivo, justifying future experiments to determine whether C5A can prevent HIV transmission in vivo.


Assuntos
Infecções por HIV/prevenção & controle , HIV/efeitos dos fármacos , Hepacivirus/química , Fragmentos de Peptídeos/farmacologia , Proteínas não Estruturais Virais/farmacologia , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Células Dendríticas/virologia , Humanos , Macrófagos/virologia , Fragmentos de Peptídeos/uso terapêutico , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/uso terapêutico
14.
Antimicrob Agents Chemother ; 52(4): 1302-17, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18212100

RESUMO

Debio-025 is a synthetic cyclosporine with no immunosuppressive capacity but a high inhibitory potency against cyclophilin A (CypA)-associated cis-trans prolyl isomerase (PPIase) activity. A lack of immunosuppressive effects compared to that of cyclosporine was demonstrated both in vitro and in vivo. For three cyclosporines, the inhibitory potential against PPIase activity was quantitatively correlated with that against human immunodeficiency virus type 1 (HIV-1) replication. Debio-025 selectively inhibited the replication of HIV-1 in a CD4+ cell line and in peripheral blood mononuclear cells: potent activity was demonstrated against clinical isolates of various HIV-1 subtypes, including isolates with multidrug resistance to reverse transcriptase and protease inhibitors. Simian immunodeficiency virus and HIV-2 strains were generally resistant to inhibition by Debio-025; however, some notable exceptions of sensitive HIV-2 clinical isolates were detected. In two-drug combination studies, additive inhibitory effects were found between Debio-025 and 19 clinically used drugs of different classes. Clinical HIV-1 isolates that are naturally resistant to Debio-025 and that do not depend on CypA for infection were identified. Comparison of the amino acid sequences of the CypA binding domain of the capsid (CA) protein from Debio-025-sensitive and -resistant HIV-1 isolates indicated that resistance was mostly associated with an H87Q/P exchange. Mechanistically, cyclosporines competitively inhibit the binding of CypA to the HIV-1 CA protein, which is an essential interaction required for early steps in HIV-1 replication. By real-time PCR we demonstrated that early reverse transcription is reduced in the presence of Debio-025 and that late reverse transcription is almost completely blocked. Thus, Debio-025 seems to interfere with the function of CypA during the progression/completion of HIV-1 reverse transcription.


Assuntos
Ciclofilinas/metabolismo , Ciclosporina/farmacologia , HIV-1/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Ciclosporina/síntese química , Ciclosporina/química , Ciclosporina/metabolismo , HIV-1/patogenicidade , HIV-1/fisiologia , Humanos , Células Jurkat , Leucócitos Mononucleares/virologia , Testes de Sensibilidade Microbiana/métodos
15.
J Virol ; 81(1): 395-405, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17050597

RESUMO

Although the transport of human immunodeficiency virus type 1 (HIV-1) through the epithelium is critical for HIV-1 colonization, the mechanisms controlling this process remain obscure. In the present study, we investigated the transcellular migration of HIV-1 as a cell-free virus through primary genital epithelial cells (PGECs). The absence of CD4 on PGECs implicates an unusual entry pathway for HIV-1. We found that syndecans are abundantly expressed on PGECs and promote the initial attachment and subsequent entry of HIV-1 through PGECs. Although CXCR4 and CCR5 do not contribute to HIV-1 attachment, they enhance viral entry and transcytosis through PGECs. Importantly, HIV-1 exploits both syndecans and chemokine receptors to ensure successful cell-free transport through the genital epithelium. HIV-1-syndecan interactions rely on specific residues in the V3 of gp120 and specific sulfations within syndecans. We found no obvious correlation between coreceptor usage and the capacity of the virus to transcytose. Since viruses isolated after sexual transmission are mainly R5 viruses, this suggests that the properties conferring virus replication after transmission are distinct from those conferring cell-free virus transcytosis through the genital epithelium. Although we found that cell-free HIV-1 crosses PGECs as infectious particles, the efficiency of transcytosis is extremely poor (less than 0.02% of the initial inoculum). This demonstrates that the genital epithelium serves as a major barrier against HIV-1. Although one cannot exclude the possibility that limited passage of cell-free HIV-1 transcytosis through an intact genital epithelium occurs in vivo, it is likely that the establishment of infection via cell-free HIV-1 transmigration is a rare event.


Assuntos
Células Epiteliais/virologia , HIV-1/fisiologia , Vagina/virologia , Transporte Biológico , Antígenos CD4/metabolismo , Antígenos CD4/fisiologia , Células Cultivadas , Feminino , Galactosilceramidas/fisiologia , Proteína gp120 do Envelope de HIV/metabolismo , Heparina/análogos & derivados , Heparina/fisiologia , Humanos , Lectinas Tipo C/fisiologia , Proteoglicanas/fisiologia , Receptores CCR5/metabolismo , Receptores CCR5/fisiologia , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiologia , Sindecanas/metabolismo , Vagina/citologia
16.
J Biol Chem ; 281(48): 37025-33, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17028189

RESUMO

The TRIM5alpha (tripartite motif 5alpha protein) has been linked to the cross-species restriction in human immunodeficiency virus type 1 (HIV-1) infection of non-human cells, but the mechanism by which this occurs remains to be fully elucidated. Here we demonstrate that the capsid (CA) protein of HIV-1 is more rapidly degraded in cells expressing monkey TRIM5alpha than in cells expressing human TRIM5alpha. Other proteins encoded by Gag and Pol are not subject to TRIM5alpha-mediated accelerated degradation. The accelerated CA degradation by TRIM5alpha apparently occurs via a nonproteosomal pathway. TRIM5alpha selectively accelerates degradation of the CA population, which reached the cytosol of restrictive cells, but not the CA population, which ended into the vesicular compartment. Given that cytosolic CA represents "productively" entered cores, whereas vesicular CA represents "nonproductively" entered cores, our findings suggest that TRIM5alpha interrupts the infectious pathway of HIV-1 by acting on the incoming cytosolic CA. The mode of viral entry does not influence the accelerated degradation of cytosolic CA by TRIM5alpha. Thus, this study reveals a correlation between TRIM5alpha-mediated HIV-1 restriction and a selective degradation of cytosolic CA normally associated with productive viral entry.


Assuntos
Capsídeo/fisiologia , Citosol/virologia , HIV-1/metabolismo , Proteínas/fisiologia , Animais , Fatores de Restrição Antivirais , Linfócitos T CD4-Positivos/virologia , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular , Citosol/metabolismo , Produtos do Gene gag/metabolismo , Produtos do Gene pol/metabolismo , Humanos , Macaca mulatta , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/metabolismo , Fatores de Tempo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Replicação Viral
17.
J Biol Chem ; 280(48): 40293-300, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16199531

RESUMO

In this study, we asked if a naturally occurring HIV-1 variant exists that circumvents CypA dependence in human cells. To address this issue, we sought viruses for CypA independence using Debio-025, a cyclosporine A (CsA) analog that disrupts CypA-capsid interaction. Surprisingly, viral variants from the Main group replicate even in the presence of the drug. Sequencing analyses revealed that these viruses encode capsid substitutions within the CypA-binding site (V86P/H87Q/I91V/M96I). When we introduced these substitutions into viruses that normally rely on CypA for replication, these mutants no longer depended on CypA, suggesting that naturally occurring capsid substitutions obviate the need for CypA. This is the first demonstration that isolates from the Main group naturally develop CypA-independent strategies to replicate in human cells. Surprisingly, we found that these capsid substitutions render HIV-1 capable of infecting Owl monkey (OMK) cells that highly restrict HIV-1. OMK cell resistance to HIV-1 is mediated via TRIM-Cyp, which arose from a retrotransposition of CypA into the TRIM5 alpha gene. Interestingly, saturation experiments suggest that the Pro86/Gln87/Val91/Ile96 capsid core is "invisible" to TRIM-Cyp. This study demonstrates that specific capsid substitutions can release HIV-1 from both CypA dependence in human cells and TRIM-Cyp restriction in monkey cells.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Ciclofilina A/química , HIV-1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Aotidae , Sítios de Ligação , Western Blotting , Linhagem Celular , Ciclofilinas/química , Humanos , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Polimorfismo Genético , Retroelementos , Fatores de Tempo
18.
J Biol Chem ; 280(47): 39493-504, 2005 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-16157597

RESUMO

HIV-1 has maximized its utilization of syndecans. It uses them as in cis receptors to infect macrophages and as in trans receptors to infect T-lymphocytes. In this study, we investigated at a molecular level the mechanisms that control HIV-1-syndecan interactions. We found that a single conserved arginine (Arg-298) in the V3 region of gp120 governs HIV-1 binding to syndecans. We found that an amine group on the side chain of this residue is necessary for syndecan utilization by HIV-1. Furthermore, we showed that HIV-1 binds syndecans via a 6-O sulfation, demonstrating that this binding is not the result of random interactions between basic residues and negative charges, but the result of specific contacts between gp120 and a well defined sulfation in syndecans. Surprisingly, we found that Arg-298, which mediates HIV-1 binding to syndecans, also mediates HIV-1 binding to CCR5. We postulated that HIV-1 recognizes similar motifs on syndecans and CCR5. Supporting this hypothesis, we obtained several lines of evidence that suggest that the 6-O sulfation recognized by HIV-1 on syndecans mimics the sulfated tyrosines recognized by HIV-1 in the N terminus of CCR5. Our finding that CCR5 and syndecans are exploited by HIV-1 via a single determinant echoes the mechanisms by which chemokines utilize these two disparate receptors and suggests that the gp120/chemokine mimicry may represent a common strategy in microbial pathogenesis.


Assuntos
Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanas/metabolismo , Receptores CCR5/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Arginina/química , Sítios de Ligação , Linhagem Celular , Sequência Conservada , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , HIV-1/patogenicidade , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Modelos Biológicos , Mimetismo Molecular , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteoglicanas/química , Proteoglicanas/genética , Receptores CCR5/química , Receptores CCR5/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sulfatos/química , Sindecanas
19.
Virology ; 325(2): 389-98, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15246277

RESUMO

Polyanions are potent HIV-1 entry inhibitors. Nevertheless, resistant viruses may emerge under polyanion inhibitory pressure. Specifically, a polyanion-resistant virus replicates in T cells even in the presence of high concentrations of polyanions. We found that although the polyanion-resistant virus grows in suspension CD4+ T cells efficiently, it infects nonlymphocytic adherent CD4+ cells poorly. Given that a main distinction between suspension and adherent cells is the absence or presence of cell-surface heparan sulfate proteoglycan (HSPG), we investigated if the failure of the polyanion-resistant virus to infect adherent CD4+ cells arises from its inability to bind HSPG. We found that the emergence of mutations in gp120 associated with polyanion resistance resulted in a decreased capacity of HIV-1 to bind HSPG. We also found that the polycation polybrene rescued the capacity of the polyanion-resistant virus to bind HSPG and to infect adherent CD4+ cells. The identification of this virus, unable to bind HSPG, provides a convenient probe to measure the impact of HIV-1-HSPG interactions in vivo. Altogether, these findings suggest that polyanion-resistance narrows the range of potential target cells for HIV-1 in the host. This reinforces the hypothesis that cell-free or cell-associated polyanions such as HSPG possess the capacity to modulate HIV-1 pathogenesis.


Assuntos
Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , Polímeros/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Farmacorresistência Viral/genética , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/genética , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/genética , HIV-1/patogenicidade , HIV-1/fisiologia , Proteoglicanas de Heparan Sulfato/metabolismo , Brometo de Hexadimetrina/farmacologia , Humanos , Técnicas In Vitro , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Polieletrólitos , Virulência/efeitos dos fármacos , Virulência/genética , Replicação Viral
20.
J Virol ; 78(12): 6567-84, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15163749

RESUMO

As a neurotropic virus, human immunodeficiency virus type 1 (HIV-1) invades the brain and causes severe neuronal, astrocyte, and myelin damage in AIDS patients. To gain access to the brain, HIV-1 must migrate through brain microvascular endothelial cells (BMECs), which compose the blood-brain barrier (BBB). Given that BMECs lack the entry receptor CD4, HIV-1 must use receptors distinct from CD4 to enter these cells. We previously reported that cell surface proteoglycans serve as major HIV-1 receptors on primary human endothelial cells. In this study, we examined whether proteoglycans also impact cell-free HIV-1 invasion of the brain. Using an artificial BBB transmigration assay, we found that both heparan and chondroitin sulfate proteoglycans (HSPGs and CSPGs, respectively) are abundantly expressed on primary BMECs and promote HIV-1 attachment and entry. In contrast, the classical entry receptors, CXCR4 and CCR5, only moderately enhanced these processes. HSPGs and CSPGs captured HIV-1 in a gp120-dependent manner. However, no correlation between coreceptor usage and transmigration was identified. Furthermore, brain-derived viruses did not transmigrate more efficiently than lymphoid-derived viruses, suggesting that the ability of HIV-1 to replicate in the brain does not correlate with its capacity to migrate through the BBB as cell-free virus. Given that HIV-1-proteoglycan interactions are based on electrostatic contacts between basic residues in gp120 and sulfate groups in proteoglycans, HIV-1 may exploit these interactions to rapidly enter and migrate through the BBB to invade the brain.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/virologia , Proteoglicanas de Sulfatos de Condroitina/metabolismo , HIV-1/patogenicidade , Heparina/análogos & derivados , Heparina/metabolismo , Proteoglicanas/metabolismo , Receptores de HIV/metabolismo , Barreira Hematoencefálica/virologia , Células Cultivadas , Células Endoteliais/virologia , Endotélio Vascular/citologia , Endotélio Vascular/virologia , HIV-1/fisiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...