Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Virol ; 89(9): 4827-36, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25673710

RESUMO

UNLABELLED: Previously, we reported that a mutant of Tat referred to as Nullbasic inhibits HIV-1 reverse transcription although the mechanism of action is unknown. Here we show that Nullbasic is a reverse transcriptase (RT) binding protein that targets the reverse transcription complex rather than directly inhibiting RT activity. An interaction between Nullbasic and RT was observed by using coimmunoprecipitation and pulldown assays, and a direct interaction was measured by using a biolayer interferometry assay. Mixtures of recombinant 6×His-RT and Nullbasic-FLAG-V5-6×His at molar ratios of up to 1:20,000 did not inhibit RT activity in standard homopolymer primer template assays. An analysis of virus made by cells that coexpressed Nullbasic showed that Nullbasic copurified with virus particles, indicating that it was a virion protein. In addition, analysis of reverse transcription complexes (RTCs) isolated from cells infected with wild type or Nullbasic-treated HIV-1 showed that Nullbasic reduced the levels of viral DNA in RTC fractions. In addition, a shift in the distribution of viral DNA and CAp24 to less-dense non-RTC fractions was observed, indicating that RTC activity from Nullbasic-treated virus was impaired. Further analysis showed that viral cores isolated from Nullbasic-treated HIV undergo increased disassembly in vitro compared to untreated HIV-1. To our knowledge, this is the first description of an antiviral protein that inhibits reverse transcription by targeting the RTC and affecting core stability. IMPORTANCE: HIV-1 infection is treated by using combinations of antiretroviral drugs that target independent steps of virus replication. A newly described antiviral protein called Nullbasic can also inhibit a combination of different steps in virus replication (transcription, reverse transcription, and Rev-mediated viral mRNA transport), although the precise mechanism of action is unknown. This study shows that Nullbasic can inhibit reverse transcription by binding to the viral enzyme called reverse transcriptase, which results in accelerated uncoating of the viral core and instability of the viral apparatus called the reverse transcription complex (RTC). This unique antiviral activity may inform development of other RTC inhibitors, as well as providing a unique investigative tool for dissecting the RTC cellular composition.


Assuntos
HIV-1/fisiologia , Transcrição Reversa , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Centrifugação , Transcriptase Reversa do HIV/antagonistas & inibidores , Transcriptase Reversa do HIV/metabolismo , HIV-1/genética , Imunoprecipitação , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
2.
Proc Natl Acad Sci U S A ; 109(24): 9587-92, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22628567

RESUMO

Cellular proteins have been implicated as important for HIV-1 reverse transcription, but whether any are reverse transcription complex (RTC) cofactors or affect reverse transcription indirectly is unclear. Here we used protein fractionation combined with an endogenous reverse transcription assay to identify cellular proteins that stimulated late steps of reverse transcription in vitro. We identified 25 cellular proteins in an active protein fraction, and here we show that the eEF1A and eEF1G subunits of eukaryotic elongation factor 1 (eEF1) are important components of the HIV-1 RTC. eEF1A and eEF1G were identified in fractionated human T-cell lysates as reverse transcription cofactors, as their removal ablated the ability of active protein fractions to stimulate late reverse transcription in vitro. We observed that the p51 subunit of reverse transcriptase and integrase, two subunits of the RTC, coimmunoprecipitated with eEF1A and eEF1G. Moreover eEF1A and eEF1G associated with purified RTCs and colocalized with reverse transcriptase following infection of cells. Reverse transcription in cells was sharply down-regulated when eEF1A or eEF1G levels were reduced by siRNA treatment as a result of reduced levels of RTCs in treated cells. The combined evidence indicates that these eEF1 subunits are critical RTC stability cofactors required for efficient completion of reverse transcription. The identification of eEF1 subunits as unique RTC components provides a basis for further investigations of reverse transcription and trafficking of the RTC to the nucleus.


Assuntos
Transcriptase Reversa do HIV/metabolismo , HIV-1/enzimologia , Fator 1 de Elongação de Peptídeos/metabolismo , Linhagem Celular , Cromatografia Líquida , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Humanos , Imunoprecipitação , Fator 1 de Elongação de Peptídeos/genética , RNA Interferente Pequeno , Espectrometria de Massas em Tandem , Transcrição Gênica
3.
Virol J ; 10: 207, 2013 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-23800116

RESUMO

BACKGROUND: Protein arginine methyltransferase 6 (PRMT6) can methylate the HIV-1 Tat, Rev and nucleocapsid proteins in a manner that diminishes each of their functions in in vitro assays, and increases the stability of Tat in human cells. In this study, we explored the relationship between PRMT6 and HIV-1 Tat by determining the domains in each protein required for interaction. METHODS: Through domain mapping and immunoprecipitation experiments, we determined that both the amino and carboxyl termini of PRMT6, and the activation domain within Tat are essential for interaction. Mutation of the basic domain of Tat did not affect the ability of PRMT6 to interact with Tat. RESULTS: We next used the A549 human alveolar adenocarcinoma cell line, which naturally expresses undetectable levels of PRMT6, as a model for testing the effects of PRMT6 on Tat stability, transactivation, and HIV-1 replication. As previously observed, steady state levels and the protein half-life of Tat were increased by the ectopic expression of PRMT6. However, no down regulation of Tat transactivation function was observed, even with over 300-fold molar excess of PRMT6 plasmid. We also observed no negative effect on HIV-1 infectivity when A549 producer cells overexpressed PRMT6. CONCLUSIONS: We show that PRMT6 requires the activation domain, but surprisingly not the basic domain, of Tat for protein interaction. This interaction between Tat and PRMT6 may impact upon pathogenic effects attributed to Tat during HIV-1 infection other than its function during transactivation.


Assuntos
HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Ativação Transcricional , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Células Epiteliais/virologia , Humanos , Imunoprecipitação , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína
4.
J Virol ; 83(22): 11694-703, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19726520

RESUMO

Arginine methylation of human immunodeficiency virus type 1 (HIV-1) Tat protein downregulates its key function in viral-gene transactivation. The fate of methylated Tat is unknown, so it is unclear whether methylated Tat is degraded or persists in the cell for additional functions. Here we show that the arginine methyltransferase PRMT6 increases Tat protein half-life by 4.7-fold. Tat stabilization depends on the catalytic activity of PRMT6 and requires arginine methylation within the Tat basic domain. In contrast, HIV-1 Rev, which is also methylated by PRMT6, is completely refractory to the stabilizing effect. Proteasome inhibition and silencing experiments demonstrated that Tat can be degraded by a REGgamma-independent proteasome, against which PRMT6 appears to act to increase Tat half-life. Our data reveal a proteasome-dependent Tat degradation pathway that is inhibited by arginine methylation. The stabilizing action of PRMT6 could allow Tat to persist within the cell and the extracellular environment and thereby enable functions implicated in AIDS-related cancer, neurodegeneration, and T-cell death.


Assuntos
HIV-1/fisiologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/fisiologia , Arginina/metabolismo , Western Blotting , Linhagem Celular , Células HeLa , Humanos , Metilação , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/fisiologia
5.
Retrovirology ; 3: 92, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17169163

RESUMO

BACKGROUND: Protein methylation is recognized as a major protein modification pathway regulating diverse cellular events such as protein trafficking, transcription, and signal transduction. More recently, protein arginine methyltransferase activity has been shown to regulate HIV-1 transcription via Tat. In this study, adenosine periodate (AdOx) was used to globally inhibit protein methyltransferase activity so that the effect of protein methylation on HIV-1 infectivity could be assessed. RESULTS: Two cell culture models were used: HIV-1-infected CEM T-cells and HEK293T cells transfected with a proviral DNA plasmid. In both models, AdOx treatment of cells increased the levels of virion in culture supernatant. However, these viruses had increased levels of unprocessed or partially processed Gag-Pol, significantly increased diameter, and displayed reduced infectivity in a MAGI X4 assay. AdOx reduced infectivity equally in both dividing and non-dividing cells. However, infectivity was further reduced if Vpr was deleted suggesting virion proteins, other than Vpr, were affected by protein methylation. Endogenous reverse transcription was not inhibited in AdOx-treated HIV-1, and infectivity could be restored by pseudotyping HIV with VSV-G envelope protein. These experiments suggest that AdOx affects an early event between receptor binding and uncoating, but not reverse transcription. CONCLUSION: Overall, we have shown for the first time that protein methylation contributes towards maximal virus infectivity. Furthermore, our results also indicate that protein methylation regulates HIV-1 infectivity in a complex manner most likely involving the methylation of multiple viral or cellular proteins and/or multiple steps of replication.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacologia , HIV-1/patogenicidade , Metiltransferases/antagonistas & inibidores , Ácido Periódico/farmacologia , Proteínas/metabolismo , Adenosina/química , Linhagem Celular , Proteínas de Fusão gag-pol/metabolismo , Produtos do Gene vpr/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/ultraestrutura , Células HeLa , Humanos , Metilação , Linfócitos T/virologia , Vírion/efeitos dos fármacos , Vírion/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
6.
Hum Gene Ther ; 24(3): 270-82, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23298160

RESUMO

Here we show potent inhibition of HIV-1 replication in a human T cell line and primary human CD4(+) cells by expressing a single antiviral protein. Nullbasic is a mutant form of the HIV-1 Tat protein that was previously shown to strongly inhibit HIV-1 replication in nonhematopoietic cell lines by targeting three steps of HIV-1 replication: reverse transcription, transport of viral mRNA, and trans-activation of HIV-1 gene expression. Here we investigated gene delivery of Nullbasic, using lentiviral and retroviral vectors. Although Nullbasic could be delivered by lentiviral vectors to target cells, transduction efficiencies were sharply reduced primarily because of negative effects on reverse transcription mediated by Nullbasic. However, Nullbasic did not inhibit transduction of HEK293T cells by a murine leukemia virus (MLV)-based retroviral vector. Therefore, MLV-based virus-like particles were used to transduce and express Nullbasic-EGFP or EGFP in Jurkat cells, a human leukemia T cell line, and Nullbasic-ZsGreen1 or ZsGreen1 in primary human CD4(+) cells. HIV-1 replication kinetics were similar in parental Jurkat and Jurkat-EGFP cells, but were strongly attenuated in Jurkat-Nullbasic-EGFP cells. Similarly, virus replication in primary CD4(+) cells expressing a Nullbasic-ZsGreen1 fusion protein was inhibited by approximately 8- to 10-fold. These experiments demonstrate the potential of Nullbasic, which has unique inhibitory activity, as an antiviral agent against HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , HIV-1/genética , HIV-1/imunologia , Mutação , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Linhagem Celular , Ordem dos Genes , Vetores Genéticos/genética , Infecções por HIV/imunologia , Humanos , Células Jurkat , Lentivirus/genética , Vírus da Leucemia Murina/genética , Transcrição Reversa , Transdução Genética , Vírion , Replicação Viral
7.
PLoS One ; 7(12): e51466, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251541

RESUMO

Nullbasic, a mutant of the HIV-1 Tat protein, has anti-HIV-1 activity through mechanisms that include inhibition of Rev function and redistribution of the HIV-1 Rev protein from the nucleolus to the nucleoplasm and cytoplasm. Here we investigate the mechanism of this effect for the first time, establishing that redistribution of Rev by Nullbasic is not due to direct interaction between the two proteins. Rather, Nullbasic affects subcellular localization of cellular proteins that regulate Rev trafficking. In particular, Nullbasic induced redistribution of exportin 1 (CRM1), nucleophosmin (B23) and nucleolin (C23) from the nucleolus to the nucleus when Rev was coexpressed, but never in its absence. Inhibition of the Rev:CRM1 interaction by leptomycin B or a non-interacting RevM10 mutant completely blocked redistribution of Rev by Nullbasic. Finally, Nullbasic did not inhibit importin ß- or transportin 1-mediated nuclear import, suggesting that cytoplasmic accumulation of Rev was due to increased export by CRM1. Overall, our data support the conclusion that CRM1-dependent subcellular redistribution of Rev from the nucleolus by Nullbasic is not through general perturbation of either nuclear import or export. Rather, Nullbasic appears to interact with and disrupt specific components of a Rev trafficking complex required for its nucleocytoplasmic shuttling and, in particular, its nucleolar accumulation.


Assuntos
Fármacos Anti-HIV/metabolismo , HIV-1/metabolismo , Carioferinas/metabolismo , Proteínas Mutantes/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , beta Carioferinas/metabolismo , Produtos do Gene rev do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Nucléolo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosfoproteínas/metabolismo , Ligação Proteica , Transporte Proteico , Proteínas de Ligação a RNA/metabolismo , Frações Subcelulares/metabolismo , Proteína Exportina 1 , Nucleolina
8.
Curr HIV Res ; 5(5): 473-83, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17896967

RESUMO

The role of Tat in HIV-1 reverse transcription has been controversial largely because different studies have observed disparate effects of the Tat protein on reverse transcription. Studies of HIV-1 lacking a functional tat gene demonstrated a decrease in reverse transcription efficiency following infection of T-cells however, in vitro recombinant Tat(1-86) has been shown to inhibit RT activity. Here we show that 20-200 nM of both N-terminally histidine-tagged recombinant Tat(1-72) and Tat(1-86) stimulated reverse transcription by HIV-1 reverse transcriptase (RT) in vitro by 2-3 fold. However, both Tat species were efficient inhibitors of RT activity at 400 nM. The lower concentrations of Tat increased reverse transcription efficiency by facilitating multiple rounds of DNA synthesis, and this increase was either not seen or reduced when Tat proteins with multiply-mutated cysteine or basic domains were used. Tat-enhanced reverse transcription occurred in a RNA-independent manner, and required formation of a Tat-RT complex. Pull-down and immunoprecipitation experiments confirmed that Tat could interact with the RT p51 subunit, and mammalian two-hybrid experiments showed interaction between Tat and both the p51 and p66 subunits. Together these results provide evidence that Tat can stimulate reverse transcription through an interaction with RT.


Assuntos
Ativação Enzimática , Produtos do Gene tat/metabolismo , Transcriptase Reversa do HIV/metabolismo , HIV-1/enzimologia , Substituição de Aminoácidos/genética , DNA Viral/biossíntese , Produtos do Gene tat/genética , Transcriptase Reversa do HIV/antagonistas & inibidores , HIV-1/metabolismo , Humanos , Imunoprecipitação , Mutação de Sentido Incorreto , Ligação Proteica , Mapeamento de Interação de Proteínas , Técnicas do Sistema de Duplo-Híbrido , Produtos do Gene tat do Vírus da Imunodeficiência Humana
9.
Curr Drug Targets ; 7(12): 1595-606, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17168834

RESUMO

More than fifteen years following the description of Tat as a critical HIV gene expression regulatory protein, additional roles for Tat in HIV replication have been described, including reverse transcription. Tat achieves function through direct interaction with viral proteins, including reverse transcriptase, and numerous cellular proteins including cyclin T1, RNA polymerase II, protein kinase R (PKR), p300/CBP, and P/CAF. Despite our advanced knowledge of how Tat operates, this has not yet resulted in the discovery of effective agents capable of targeting various Tat functions. Nevertheless, Tat remains an attractive, virus-specific molecule and detailed understanding of specific protein interaction holds promise for future drug discovery.


Assuntos
Fármacos Anti-HIV/farmacologia , Produtos do Gene tat/antagonistas & inibidores , Produtos do Gene tat/fisiologia , Éxons , Regulação Viral da Expressão Gênica , Produtos do Gene tat/química , Produtos do Gene tat/genética , Repetição Terminal Longa de HIV , HIV-1/genética , Humanos , Fosforilação , Fator B de Elongação Transcricional Positiva/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Ativação Transcricional , eIF-2 Quinase/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana
10.
J Virol ; 77(18): 9912-21, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941901

RESUMO

The human immunodeficiency virus type 1 (HIV-1) Tat protein enhances reverse transcription, but it is not known whether Tat acts directly on the reverse transcription complex or through indirect mechanisms. Since processing of Tat by HIV protease (PR) might mask its presence and, at least in part, explain this lack of data, we asked whether Tat can be cleaved by PR. We used a rabbit reticulocyte lysate (RRL) system to make Tat and PR. HIV-1 PR is expressed as a Gag-Pol fusion protein, and a PR-inactivated Gag-Pol is also expressed as a control. We showed that Tat is specifically cleaved in the presence of PR, producing a protein of approximately 5 kDa. This result suggested that the cleavage site was located in or near the Tat basic domain (amino acids 49 to 57), which we have previously shown to be important in reverse transcription. We created a panel of alanine-scanning mutations from amino acids 45 to 54 in Tat and evaluated functional parameters, including transactivation, reverse transcription, and cleavage by HIV-1 PR. We showed that amino acids 49 to 52 (RKKR) are absolutely required for Tat function in reverse transcription, that mutation of this domain blocks cleavage by HIV-1 PR, and that other pairwise mutations in this region modulate reverse transcription and proteolysis in strikingly similar degrees. Mutation of Tat Y47G48 to AA also down-regulated Tat-stimulated reverse transcription but had little effect on transactivation or proteolysis by HIV PR, suggesting that Y47 is critical for reverse transcription. We altered the tat gene of the laboratory strain NL4-3 to Y47D and Y47N so that overlapping reading frames were not affected and showed that Y47D greatly diminished virus replication and conveyed a reverse transcription defect. We hypothesize that a novel, cleaved form of Tat is present in the virion and that it requires Y47 for its role in support of efficient reverse transcription.


Assuntos
Produtos do Gene tat/fisiologia , Protease de HIV/fisiologia , HIV-1/fisiologia , Transcrição Gênica , Replicação Viral , Animais , Linhagem Celular , Produtos do Gene tat/química , HIV-1/genética , Humanos , Coelhos , Reticulócitos/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana
11.
Virology ; 300(2): 226-35, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12350353

RESUMO

The tat gene is required by HIV-1 for efficient reverse transcription and this function of Tat can be distinguished from its role in transcription by RNA polymerase II using tat point mutations that abrogate each function independently. The mechanism of Tat's role in reverse transcription, however, is not known, nor is it known whether this role is conserved among trans-activating factors in other retroviruses. Here we examine the abilities of heterologous viral trans-activating proteins from jembrana disease virus (jTat), HIV-2 (Tat2), and equine infectious anemia virus (eTat) to substitute for HIV-1 Tat (Tat1) and restore reverse transcription in HIV-1 carrying an inactivated tat gene. Natural endogenous reverse transcription assays showed that trans-activators from some retroviruses (Tat2 and jTat, but not eTat) could substitute for Tat1 in complementation of HIV-1 reverse transcription. Finally, we show that Y47 is critical for Tat1 to function in reverse transcription, but not HIV-1 gene expression. We mutated the homologous position in jTat to H62Y and found it did not improve its ability to stimulate reverse transcription, but an H62A mutation did inhibit jTat complementation. These data highlight the finding that the role of Tat in reverse transcription is not related to trans-activation and demonstrate that other tat genes conserve this function.


Assuntos
Genes tat/fisiologia , HIV-1/genética , Lentivirus/genética , Transcrição Gênica , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Repetição Terminal Longa de HIV , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Viral/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA