Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31776272

RESUMO

The human immunodeficiency virus type 1 (HIV-1) accessory protein Vpr enhances viral replication in both macrophages and, to a lesser extent, cycling T cells. Virion-packaged Vpr is released in target cells shortly after entry, suggesting it is required in the early phase of infection. Previously, we described REAF (RNA-associated early-stage antiviral factor; RPRD2), a constitutively expressed protein that potently restricts HIV replication at or during reverse transcription. Here, we show that a virus without an intact vpr gene is more highly restricted by REAF and, using delivery by virus-like particles (VLPs), that Vpr alone is sufficient for REAF degradation in primary macrophages. REAF is more highly expressed in macrophages than in cycling T cells, and we detected, by coimmunoprecipitation assay, an interaction between Vpr protein and endogenous REAF. Vpr acts quickly during the early phase of replication and induces the degradation of REAF within 30 min of viral entry. Using Vpr F34I and Q65R viral mutants, we show that nuclear localization and interaction with cullin 4A-DBB1 (DCAF1) E3 ubiquitin ligase are required for REAF degradation by Vpr. In response to infection, cells upregulate REAF levels. This response is curtailed in the presence of Vpr. These findings support the hypothesis that Vpr induces the degradation of a factor, REAF, that impedes HIV infection in macrophages.IMPORTANCE For at least 30 years, it has been known that HIV-1 Vpr, a protein carried in the virion, is important for efficient infection of primary macrophages. Vpr is also a determinant of the pathogenic effects of HIV-1 in vivo A number of cellular proteins that interact with Vpr have been identified. So far, it has not been possible to associate these proteins with altered viral replication in macrophages or to explain why Vpr is carried in the virus particle. Here, we show that Vpr mitigates the antiviral effects of REAF, a protein highly expressed in primary macrophages and one that inhibits virus replication during reverse transcription. REAF is degraded by Vpr within 30 min of virus entry in a manner dependent on the nuclear localization of Vpr and its interaction with the cell's protein degradation machinery.


Assuntos
Antivirais/metabolismo , HIV-1/metabolismo , Replicação Viral/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Produtos do Gene vpr/metabolismo , Produtos do Gene vpr/fisiologia , Células HEK293 , Infecções por HIV/virologia , HIV-1/fisiologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/metabolismo , Cultura Primária de Células , Ubiquitina-Proteína Ligases/metabolismo , Vírion/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
2.
Nat Immunol ; 12(10): 975-83, 2011 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-21874023

RESUMO

APOBEC3G (A3G) is an intrinsic antiviral factor that inhibits the replication of human immunodeficiency virus (HIV) by deaminating cytidine residues to uridine. This causes guanosine-to-adenosine hypermutation in the opposite strand and results in inactivation of the virus. HIV counteracts A3G through the activity of viral infectivity factor (Vif), which promotes degradation of A3G. We report that viral protein R (Vpr), which interacts with a uracil glycosylase, also counteracted A3G by diminishing the incorporation of uridine. However, this process resulted in activation of the DNA-damage-response pathway and the expression of natural killer (NK) cell-activating ligands. Our results show that pathogen-induced deamination of cytidine and the DNA-damage response to virus-mediated repair of the incorporation of uridine enhance the recognition of HIV-infected cells by NK cells.


Assuntos
Citidina Desaminase/fisiologia , HIV/imunologia , Células Matadoras Naturais/imunologia , Linfócitos T/virologia , Desaminase APOBEC-3G , Células Cultivadas , Citotoxicidade Imunológica , Dano ao DNA , Produtos do Gene vpr/fisiologia , Humanos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Uridina/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-21710851

RESUMO

The presence of siRNA against adapter-related protein complex 2 alpha 1 subunit (AP2alpha) enhances human immunodeficiency virus type 1 (HIV-1) replication by up-regulating nuclear transport of viral genome. In this report, we examined possible viral factors involved in AP2alpha-mediated regulation of HIV-1 replication, namely, Gag matrix protein (MA), integrase (IN) and Vpr. Replication of mutant viruses lacking the nucleophilic property of one of these viral proteins was significantly enhanced by treating cells with AP2alpha siRNA, indicating that Gag MA, IN or Vpr is not specifically involved in AP2alpha-mediated enhancement of viral replication. In contrast, AP2alpha siRNA showed no effect on the level of gene transduction mediated by HIV-1-derived lentiviral vector (LV). Although virus-like LV particle and parental HIV-1 particle are composed of almost equivalent viral structural proteins, LV particles lack three accessory proteins, Vif, Vpr and Vpu, and a large portion of the HIV-1 genome. Vif, Vpr and Vpu were dispensable for AP2alpha siRNA-mediated enhancement of HIV-1 replication, indicating that a particular part of the HIV-1 genomic fragment deleted in the LV genome might be required for the enhancing effect of AP2alpha siRNA on viral replication. Taken together, these results suggest that an as yet undetermined gene fragment of the HIV-1 genome is involved in AP2alpha-mediated regulation of HIV-1 replication.


Assuntos
Complexo 2 de Proteínas Adaptadoras/fisiologia , Subunidades alfa do Complexo de Proteínas Adaptadoras/fisiologia , Produtos do Gene gag/fisiologia , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Integrases/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia , Complexo 2 de Proteínas Adaptadoras/genética , Subunidades alfa do Complexo de Proteínas Adaptadoras/genética , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Produtos do Gene gag/genética , Produtos do Gene vpr/genética , HIV-1/genética , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Humanos , Integrases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia
4.
Biol Direct ; 3: 17, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18445273

RESUMO

BACKGROUND: Despite continuing advances in our understanding of AIDS pathogenesis, the mechanism of CD4+ T cell depletion in HIV-1-infected individuals remains unclear. The HIV-1 Vpr accessory protein causes cell death, likely through a mechanism related to its ability to arrest cells in the G2,M phase. Recent evidence implicated the scaffold protein, 14-3-3, in Vpr cell cycle blockade. RESULTS: We found that in human T cells, 14-3-3 plays an active role in mediating Vpr-induced cell cycle arrest and reveal a dramatic increase in the amount of Cdk1, Cdc25C, and CyclinB1 bound to 14-3-3 theta during Vprv-induced G2,M arrest. By contrast, a cell-cycle-arrest-dead Vpr mutant failed to augment 14-3-3 theta association with Cdk1 and CyclinB1. Moreover, G2,M arrest caused by HIV-1 infection strongly correlated with a disruption in 14-3-3 theta binding to centrosomal proteins, Plk1 and centrin. Finally, Vpr caused elevated levels of CyclinB1, Plk1, and Cdk1 in a complex with the nuclear transport and spindle assembly protein, importin beta. CONCLUSION: Thus, our data reveal a new facet of Vpr-induced cell cycle arrest involving previously unrecognized abnormal rearrangements of multiprotein assemblies containing key cell cycle regulatory proteins.


Assuntos
Proteínas 14-3-3/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Proteínas 14-3-3/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/virologia , Proteínas de Ciclo Celular/fisiologia , Morte Celular/fisiologia , Células Clonais , Técnicas de Silenciamento de Genes , Produtos do Gene vpr/metabolismo , Humanos , Células Jurkat , Ligação Proteica/fisiologia , Replicação Viral/fisiologia
5.
J Virol ; 82(5): 2528-42, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094160

RESUMO

Human immunodeficiency virus type 1 (HIV-1)-infected macrophages damage mature neurons in the brain, although their effect on neuronal development has not been clarified. In this study, we show that HIV-1-infected macrophages produce factors that impair the development of neuronal precursor cells and that soluble viral protein R (Vpr) is one of the factors that has the ability to suppress axonal growth. Cell biological analysis revealed that extracellularly administered recombinant Vpr (rVpr) clearly accumulated in mitochondria where a Vpr-binding protein adenine nucleotide translocator localizes and also decreased the mitochondrial membrane potential, which led to ATP synthesis. The depletion of ATP synthesis reduced the transportation of mitochondria within neurites. This mitochondrial dysfunction inhibited axonal growth even when the frequency of apoptosis was not significant. We also found that point mutations of arginine (R) residues to alanine (A) residues at positions 73, 77, and 80 rendered rVpr incapable of causing mitochondrial membrane depolarization and axonal growth inhibition. Moreover, the Vpr-induced inhibition was suppressed after treatment with a ubiquinone analogue (ubiquinone-10). Our results suggest that soluble Vpr is a major viral factor that causes a disturbance in neuronal development through the induction of mitochondrial dysfunction. Since ubiquinone-10 protects the neuronal plasticity in vitro, it may be a therapeutic agent that can offer defense against HIV-1-associated neurological disease.


Assuntos
Axônios , Produtos do Gene vpr/fisiologia , HIV-1/metabolismo , Mitocôndrias/fisiologia , Animais , Meios de Cultivo Condicionados , Imuno-Histoquímica , Potenciais da Membrana , Camundongos
6.
J Virol ; 82(6): 2904-17, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18160429

RESUMO

Human immunodeficiency virus type 1 (HIV-1) Vpr induces cell cycle G(2) arrest in fission yeast (Schizosaccharomyces pombe) and mammalian cells, suggesting the cellular pathway(s) targeted by Vpr is conserved among eukaryotes. Our previous studies in fission yeast demonstrated that Vpr induces G(2) arrest in part through inhibition of Cdc25, a Cdc2-specific phosphatase that promotes G(2)/M transition. The goal of this study was to further elucidate molecular mechanism underlying the inhibitory effect of Vpr on Cdc25. We show here that, similar to the DNA checkpoint controls, expression of vpr promotes subcellular relocalization of Cdc25 from nuclear to cytoplasm and thereby prevents activation of Cdc2 by Cdc25. Vpr-induced nuclear exclusion of Cdc25 appears to depend on the serine/threonine phosphorylation of Cdc25 and the presence of Rad24/14-3-3 protein, since amino acid substitutions of the nine possible phosphorylation sites of Cdc25 with Ala (9A) or deletion of the rad24 gene abolished nuclear exclusion induced by Vpr. Interestingly, Vpr is still able to promote Cdc25 nuclear export in mutants defective in the checkpoints (rad3 and chk1/cds1), the kinases that are normally required for Cdc25 phosphorylation and nuclear exclusion of Cdc25, suggesting that others kinase(s) might modulate phosphorylation of Cdc25 for the Vpr-induced G(2) arrest. We report here that this kinase is Srk1. Deletion of the srk1 gene blocks the nuclear exclusion of Cdc25 caused by Vpr. Overexpression of srk1 induces cell elongation, an indication of cell cycle G(2) delay, in a similar fashion to Vpr; however, no additive effect of cell elongation was observed when srk1 and vpr were coexpressed, indicating Srk1 and Vpr are likely affecting the cell cycle G(2)/M transition through the same cellular pathway. Immunoprecipitation further shows that Vpr and Srk1 are part of the same protein complex. Consistent with our findings in fission yeast, depletion of the MK2 gene, a human homologue of Srk1, either by small interfering RNA or an MK2 inhibitor suppresses Vpr-induced cell cycle G(2) arrest in mammalian cells. Collectively, our data suggest that Vpr induces cell cycle G(2) arrest at least in part through a Srk1/MK2-mediated mechanism.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Fúngicas/metabolismo , Fase G2 , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas de Schizosaccharomyces pombe/fisiologia , ras-GRF1/metabolismo , Compartimento Celular , Linhagem Celular , Citoplasma/enzimologia , Humanos , Fosforilação , Schizosaccharomyces/enzimologia , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo
7.
Mol Endocrinol ; 22(2): 234-47, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17932108

RESUMO

HIV-1-infected patients may develop lipodystrophy and insulin resistance. We investigated the effect of the HIV-1 accessory protein viral protein R (Vpr) on the activity of the peroxisome proliferator-activating receptor-gamma (PPARgamma), a key regulator of adipocyte differentiation and tissue insulin sensitivity. We studied expression of PPARgamma-responsive reporter genes in 3T3-L1 mouse adipocytes. We investigated Vpr interaction with the PPAR/retinoid X receptor (RXR)-binding site of the c-Cbl-associating protein (CAP) gene using the chromatin immunoprecipitation assay as well as the interaction of Vpr and PPARgamma using coimmunoprecipitation. Finally, we studied the ability of exogenous Vpr protein to enter cultured adipocytes and retard differentiation. We found that Vpr suppressed PPARgamma-induced transactivation in both undifferentiated and differentiated 3T3-L1 cells. Transcriptional suppression by Vpr required an intact LXXLL coactivator motif. Vpr suppressed mRNA expression of PPARgamma-responsive genes in undifferentiated 3T3-L1 cells and associated with the PPAR/RXR-binding site located in the promoter region of the CAP gene. Vpr interacted with the ligand-binding domain of PPARgamma in an agonist-dependent fashion in vitro. Vpr delivered either by an expression plasmid or as protein added to media suppressed PPARgamma agonist-induced adipocyte differentiation, assessed as lipid accumulation and mRNA expression of the adipocyte differentiation marker adipocyte P2 in 3T3-L1 cells. In conclusion, circulating Vpr or, alternatively, Vpr produced as a consequence of direct infection of adipocytes could suppress in vivo differentiation of preadipocytes by acting as a corepressor of PPARgamma-mediated gene transcription. Vpr may alter sensitivity to insulin and thereby contribute to the development of lipodystrophy and insulin resistance observed in HIV-1-infected patients.


Assuntos
Adipócitos/citologia , Diferenciação Celular/genética , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , PPAR gama/genética , Células 3T3-L1 , Animais , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Produtos do Gene vpr/genética , HIV-1/genética , Células HeLa , Humanos , Imunoprecipitação , Metabolismo dos Lipídeos/genética , Lipodistrofia/metabolismo , Lipodistrofia/virologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR delta/genética , PPAR delta/metabolismo , PPAR gama/metabolismo , Reação em Cadeia da Polimerase , Ligação Proteica , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Transcrição Gênica
10.
Proc Natl Acad Sci U S A ; 104(28): 11778-83, 2007 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-17609381

RESUMO

The replication of viruses depends on the cell cycle status of the infected cells. Viruses have evolved functions that alleviate restrictions imposed on their replication by the host. Vpr, an accessory factor of primate lentiviruses, arrests cells at the DNA damage checkpoint in G2 phase of the cell cycle, but the mechanism underlying this effect has remained elusive. Here we report that Vpr proteins of both the human (HIV-1) and the distantly related simian (SIVmac) immunodeficiency viruses specifically associate with a protein complex comprising subunits of E3 ubiquitin ligase assembled on Cullin-4 scaffold (Cul4-DDB1[VprBP]). We show that Vpr binding to Cul4-DDB1[VprBP] leads to increased neddylation and elevated intrinsic ubiquitin ligase activity of this E3. This effect is mediated through the VprBP subunit of the complex, which recently has been suggested to function as a substrate receptor for Cul4. We also demonstrate that VprBP regulates G1 phase and is essential for the completion of DNA replication in S phase. Furthermore, the ability of Vpr to arrest cells in G2 phase correlates with its ability to interact with Cul4-DDB1[VprBP] E3 complex. Our studies identify the Cul4-DDB1[VprBP] E3 ubiquitin ligase complex as the downstream effector of lentiviral Vpr for the induction of cell cycle arrest in G2 phase and suggest that Vpr may use this complex to perturb other aspects of the cell cycle and DNA metabolism in infected cells.


Assuntos
Ciclo Celular/fisiologia , Proteínas Culina/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Vírus da Imunodeficiência Símia/fisiologia , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal , Animais , Complexo do Signalossomo COP9 , Proteínas de Transporte/metabolismo , Linhagem Celular , Fatores de Iniciação em Eucariotos/metabolismo , Fase G2/fisiologia , Produtos do Gene vpr/metabolismo , Humanos , Ligação Proteica , Subunidades Proteicas/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
11.
J Biol Chem ; 282(37): 27046-27057, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17620334

RESUMO

The roles of the HIV1 protein Vpr in virus replication and pathogenesis remain unclear. Expression of Vpr in dividing cells causes cell cycle arrest in G(2). Vpr also facilitates low titer infection of terminally differentiated macrophages, enhances transcription, promotes apoptosis, and targets cellular uracil N-glycosylase for degradation. Using co-immunoprecipitation and tandem mass spectroscopy, we found that HIV1 Vpr engages a DDB1- and cullin4A-containing ubiquitin-ligase complex through VprBP/DCAF1. HIV2 Vpr has two Vpr-like proteins, Vpr and Vpx, which cause G(2) arrest and facilitate macrophage infection, respectively. HIV2 Vpr, but not Vpx, engages the same set of proteins. We further demonstrate that the interaction between Vpr and the ubiquitin-ligase components as well as further assembly of the ubiquitin-ligase are necessary for Vpr-mediated G(2) arrest. Our data support a model in which Vpr engages the ubiquitin ligase to deplete a cellular factor that is required for cell cycle progression into mitosis. Vpr, thus, functions like the HIV1 proteins Vif and Vpu to usurp cellular ubiquitin ligases for viral functions.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas Culina/fisiologia , Proteínas de Ligação a DNA/fisiologia , Fase G2 , Produtos do Gene vpr/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Células Cultivadas , Humanos , Imunoprecipitação , Uracila-DNA Glicosidase/metabolismo
12.
Virol J ; 4: 57, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17559673

RESUMO

HIV-1 Vpr is a viral accessory protein that activates ATR through the induction of DNA replication stress. ATR activation results in cell cycle arrest in G2 and induction of apoptosis. In the present study, we investigate the role of the ubiquitin/proteasome system (UPS) in the above activity of Vpr. We report that the general function of the UPS is required for Vpr to induce G2 checkpoint activation, as incubation of Vpr-expressing cells with proteasome inhibitors abolishes this effect. We further investigated in detail the specific E3 ubiquitin ligase subunits that Vpr manipulates. We found that Vpr binds to the DCAF1 subunit of a cullin 4a/DDB1 E3 ubiquitin ligase. The carboxy-terminal domain Vpr(R80A) mutant, which is able to bind DCAF1, is inactive in checkpoint activation and has dominant-negative character. In contrast, the mutation Q65R, in the leucine-rich domain of Vpr that mediates DCAF1 binding, results in an inactive Vpr devoid of dominant negative behavior. Thus, the interaction of Vpr with DCAF1 is required, but not sufficient, for Vpr to cause G2 arrest. We propose that Vpr recruits, through its carboxy terminal domain, an unknown cellular factor that is required for G2-to-M transition. Recruitment of this factor leads to its ubiquitination and degradation, resulting in failure to enter mitosis.


Assuntos
Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Linhagem Celular , Fase G2 , Produtos do Gene vpr/isolamento & purificação , HIV-1/efeitos dos fármacos , Células HeLa/citologia , Células HeLa/fisiologia , Células HeLa/virologia , Humanos , Rim , Oligopeptídeos/farmacologia , RNA Interferente Pequeno/genética , RNA Viral/genética , Transfecção , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
13.
J Virol ; 81(17): 9572-6, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17553868

RESUMO

Mutational analysis of the four conserved proline residues in human immunodeficiency virus type 1 (HIV-1) Vpr reveals that only Pro-35 is required for efficient replication of R5-tropic, but not of X4-tropic, viruses in human lymphoid tissue (HLT) cultivated ex vivo. While Vpr-mediated apoptosis and G(2) cell cycle arrest, as well as the expression and subcellular localization of Vpr, were independent, the capacity for encapsidation of Vpr into budding virions was dependent on Pro-35. (1)H nuclear magnetic resonance data suggest that mutation of Pro-35 causes a conformational change in the hydrophobic core of the molecule, whose integrity is required for the encapsidation of Vpr, and thus, Pro-35 supports the replication of R5-tropic HIV-1 in HLT.


Assuntos
Produtos do Gene vpr/fisiologia , HIV-1/crescimento & desenvolvimento , Tecido Linfoide/virologia , Estrutura Secundária de Proteína/genética , Replicação Viral/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Produtos do Gene vpr/química , Produtos do Gene vpr/genética , Células HeLa , Humanos , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Prolina , Montagem de Vírus/genética , Montagem de Vírus/fisiologia , Replicação Viral/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
14.
J Virol ; 81(17): 8878-90, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17553871

RESUMO

The mechanism of CD4(+) T-cell depletion in human immunodeficiency virus type 1 (HIV-1)-infected individuals remains unknown, although mounting evidence suggests that direct viral cytopathicity contributes to this loss. The HIV-1 Vpr accessory protein causes cell death and arrests cells in the G(2)/M phase; however, the molecular mechanism underlying these properties is not clear. Mutation of hydrophobic residues on the surface of its third alpha-helix disrupted Vpr toxicity, G(2)/M arrest induction, nuclear localization, and self-association, implicating this region in multiple Vpr functions. Cytopathicity by virion-delivered mutant Vpr protein correlated with G(2)/M arrest induction but not nuclear localization or self-association. However, infection with whole virus encoding these Vpr mutants did not abrogate HIV-1-induced cell killing. Rather, mutant Vpr proteins that are impaired for G(2)/M block still prevented infected cell proliferation, and this property correlated with the death of infected cells. Chemical agents that inhibit infected cells from entering G(2)/M also did not reduce HIV-1 cytopathicity. Combined, these data implicate Vpr in HIV-1 killing through a mechanism involving inhibiting cell division but not necessarily in G(2)/M. Thus, the hydrophobic region of the third alpha-helix of Vpr is crucial for mediating G(2)/M arrest, nuclear localization, and self-association but dispensable for HIV-1 cytopathicity due to residual cell proliferation blockade mediated by a separate region of the protein.


Assuntos
Linfócitos T CD4-Positivos/virologia , Ciclo Celular , Efeito Citopatogênico Viral , Produtos do Gene vpr/fisiologia , HIV-1/patogenicidade , Substituição de Aminoácidos/genética , Linfócitos T CD4-Positivos/citologia , Morte Celular , Linhagem Celular Tumoral , Produtos do Gene vpr/química , Produtos do Gene vpr/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células Jurkat , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Ligação Proteica/genética , Estrutura Secundária de Proteína , Transporte Proteico/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
15.
J Neurosci ; 27(14): 3703-11, 2007 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-17409234

RESUMO

Despite the introduction of highly active antiretroviral therapy, dementia caused by human immunodeficiency virus-1 (HIV-1) infection remains a devastating and common neurological disorder. Although the mechanisms governing neurodegeneration during HIV-1 infection remain uncertain, the HIV-1 accessory protein, viral protein R (Vpr), has been proposed as a neurotoxic protein. Herein, we report that Vpr protein and transcript were present in the brains of HIV-infected persons. Moreover, soluble Vpr caused neuronal apoptosis, involving cytochrome c extravasation, p53 induction, and activation of caspase-9 while exerting a depressive effect on whole-cell currents in neurons (p < 0.05), which was inhibited by iberiotoxin. Vpr-activated glial cells secreted neurotoxins in a concentration-dependent manner (p < 0.001). Transgenic (Tg) mice expressing Vpr in brain monocytoid cells displayed the transgene principally in the basal ganglia (p < 0.05) and cerebral cortex (p < 0.01) compared with hindbrain expression. Vpr was released from cultured transgenic macrophages, which was cytotoxic to neurons and was blocked by anti-Vpr antibody (p < 0.05). Neuronal injury was observed in Tg animals compared with wild-type littermates, chiefly affecting GAD65 (p < 0.01) and vesicular acetylcholine transferase (p < 0.001) immunopositive neuronal populations in the basal ganglia. There was also a loss of subcortical synaptophysin (p < 0.001) immunoreactivity as well as an increase in activated caspase-3, which was accompanied by a hyperexcitable neurobehavioral phenotype (p < 0.05). Thus, HIV-1 Vpr caused neuronal death through convergent pathogenic mechanisms with ensuing in vivo neurodegeneration, yielding new insights into the mechanisms by which HIV-1 injures the nervous system.


Assuntos
Apoptose/fisiologia , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Degeneração Neural/metabolismo , Neurônios/metabolismo , Animais , Linhagem Celular Tumoral , Produtos do Gene vpr/biossíntese , HIV-1/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Degeneração Neural/patologia , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
16.
DNA Cell Biol ; 26(2): 116-31, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17328670

RESUMO

The human immunodeficiency virus type I (HIV-1) accessory protein Vpr has been associated with the induction of programmed cell death (apoptosis) and cell-cycle arrest. Studies have shown the apoptotic effect of Vpr on primary and established cell lines and on diverse tissues including the central nervous system (CNS) in vitro. However, the relevance of the effect of Vpr observed in vitro to HIV-1 neuropathogenesis in vivo, remains unknown. Due to the narrow host range of HIV-1 infection, no animal model is currently available. This has prompted us to consider a small animal model to evaluate the effects of Vpr on CNS in vivo through surrogate viruses expressing HIV-1Vpr. A single round of replication competent viral vectors, expressing Vpr, were used to investigate the apoptosis-inducing capabilities of HIV-1Vpr in vivo. Viral particles pseudotyped with VSV-G or N2c envelopes were generated from spleen necrosis virus (SNV) and HIV-1-based vectors to transduce CNS cells. The in vitro studies have demonstrated that Vpr generated by SNV vectors had less apoptotic effects on CNS cells compared with Vpr expressed by HIV-1 vectors. The in vivo study has suggested that viral particles, expressing Vpr generated by HIV-1-based vectors, when delivered through the ventricle, caused loss of neurons and dendritic processes in the cortical region. The apoptotic effect was extended beyond the cortical region and affected the hippocampus neurons, the lining of the choroids plexus, and the cerebellum. However, the effect of Vpr, when delivered through the cortex, showed neuronal damage only around the site of injection. Interestingly, the number of apoptotic neurons were significantly higher with HIV-1 vectors expressing Vpr than by the SNV vectors. This may be due to the differences in the proteins expressed by these viral vectors. These results suggest that Vpr induces apoptosis in CNS cells in vitro and in vivo. To our knowledge, this is the first study to investigate the apoptosis-inducing capabilities of HIV-1Vpr in vivo in neonatal mice. We propose that this, in expensive animal model, may be of value to design-targeted neuroprotective therapeutics.


Assuntos
Apoptose , Encéfalo/patologia , Produtos do Gene vpr/fisiologia , HIV-1/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Células Cultivadas , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oligodendroglia/patologia , Vírus do Infarto Esplênico do Pato de Trager/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
17.
Proc Natl Acad Sci U S A ; 104(10): 4130-5, 2007 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-17360488

RESUMO

The Vpr accessory protein of HIV-1 induces a response similar to that of DNA damage. In cells expressing Vpr, the DNA damage sensing kinase, ATR, is activated, resulting in G(2) arrest and apoptosis. In addition, Vpr causes rapid degradation of the uracil-DNA glycosylases UNG2 and SMUG1. Although several cellular proteins have been reported to bind to Vpr, the mechanism by which Vpr mediates its biological effects is unknown. Using tandem affinity purification and mass spectrometry, we identified a predominant cellular protein that binds to Vpr as the damage-specific DNA-binding protein 1 (DDB1). In addition to its role in the repair of damaged DNA, DDB1 is a component of an E3 ubiquitin ligase that degrades numerous cellular substrates. Interestingly, DDB1 is targeted by specific regulatory proteins of other viruses, including simian virus 5 and hepatitis B. We show that the interaction with DDB1 mediates Vpr-induced apoptosis and UNG2/SMUG1 degradation and impairs the repair of UV-damaged DNA, which could account for G(2) arrest and apoptosis. The interaction with DDB1 may explain several of the diverse biological functions of Vpr and suggests potential roles for Vpr in HIV-1 replication.


Assuntos
DNA Glicosilases/metabolismo , Proteínas de Ligação a DNA/química , Produtos do Gene vpr/fisiologia , Apoptose , Linhagem Celular , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Genes vpr/genética , HIV-1/metabolismo , Humanos , Ligação Proteica , Fatores de Tempo , Ubiquitina-Proteína Ligases/metabolismo , Raios Ultravioleta , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
18.
Virus Res ; 126(1-2): 76-85, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17349711

RESUMO

Human immunodeficiency virus type 1 (HIV-1) Vpr is known to dysregulate host cellular functions through its interaction with cellular proteins. Using a protein array we assessed Vpr-mediated differential regulation of host cellular proteins expression. Results demonstrated that Vpr differentially regulated host factors that are involved in functions, such as cell proliferation, differentiation and apoptosis. One of the most highly downregulated proteins attained was the sodium hydrogen exchanger, isoform 1 (NHE1), which showed a significant (60%) decrease in HIV-1 Vpr(+) virus infected cells as compared to HIV-1 Vpr(-) virus infected control. NHE1 downregulation further led to acidification of cells and was directly correlated with loss of ezrin, radixin and moesin (ERM) protein complex and decreased AKT phosphorylation. Vpr-mediated NHE1 dyregulation is in part through GR pathway as GR antagonist, mifepristone reversed Vpr-induced NHE1 downregulation.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , HIV-1/patogenicidade , Trocadores de Sódio-Hidrogênio/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas do Citoesqueleto/metabolismo , Regulação para Baixo/efeitos dos fármacos , Produtos do Gene vpr/genética , Genes vpr , HIV-1/genética , Células HeLa , Antagonistas de Hormônios/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Líquido Intracelular/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Mifepristona/farmacologia , Modelos Biológicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
19.
Cell Cycle ; 6(2): 182-8, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17314515

RESUMO

How the HIV1 Vpr protein initiates the host cell response leading to cell cycle arrest in G(2) has remained unknown. Here, we show that recruitment of DCAF1/VprBP by Vpr is essential for its cytostatic activity, which can be abolished either by single mutations of Vpr that impair DCAF1 binding, or by siRNA-mediated silencing of DCAF1. Furthermore, DCAF1 bridges Vpr to DDB1, a core subunit of Cul4 ubiquitin ligases. Altogether these results point to a mechanism where Vpr triggers G(2) arrest by hijacking the Cul4/DDB1(DCAF1) ubiquitin ligase. We further show that, Vpx, a non-cytostatic Vpr-related protein acquired by HIV2 and SIV, also binds DCAF1 through a conserved motif. Thus, Vpr from HIV1 and Vpx from SIV recruit DCAF1 with different physiological outcomes for the host cell. This in turn suggests that both proteins have evolved to preserve interaction with the same Cul4 ubiquitin ligase while diverging in the recognition of host substrates targeted for proteasomal degradation.


Assuntos
Ciclo Celular/fisiologia , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Proteínas Culina/fisiologia , Citotoxinas/fisiologia , Citotoxinas/toxicidade , Proteínas de Ligação a DNA/fisiologia , Produtos do Gene vpr/toxicidade , Células HeLa , Humanos , Dados de Sequência Molecular , Transporte Proteico/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
20.
FEBS Lett ; 581(3): 535-40, 2007 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-17254575

RESUMO

Viral protein R (Vpr) from the human immunodeficiency virus induces cell cycle arrest in proliferating cells, stimulates virus transcription, and regulates activation and apoptosis of infected T-lymphocytes. We report that Jurkat cells overexpressing full-length gelsolin show resistance to Vpr-induced T-cell apoptosis with abrogation of mitochondrial membrane potential loss and the release of cytochrome c. Co-immunoprecipitation assays in HEK293T cells demonstrated that overexpression of full-length or segment 5 (G5) but not G5-deleted gelsolin (DeltaG5) bound to the voltage-dependent anion channel (VDAC), and that the G5 subunit can inhibit HIV-1-Vpr-binding to VDAC. We also confirmed that full-length gelsolin has the same effect in Jurkat cells. Clonogenic analysis showed that transfection of G5 but not DeltaG5 cDNA protects Jurkat T cells from HIV-Vpr-Tet induced T-cell apoptosis and promoted cell survival, as did full-length gelsolin. These results suggest that the gelsolin G5 domain inhibits HIV-Vpr-induced T-cell apoptosis by blocking the interaction between Vpr and VDAC, and might be used as a protective treatment against HIV-Vpr-induced T-cell apoptosis.


Assuntos
Gelsolina/fisiologia , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , HIV-1/patogenicidade , Linfócitos T/patologia , Linfócitos T/virologia , Canal de Ânion 1 Dependente de Voltagem/fisiologia , Apoptose , Sequência de Bases , Linhagem Celular , DNA Complementar/genética , Gelsolina/química , Gelsolina/genética , Humanos , Técnicas In Vitro , Células Jurkat , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Linfócitos T/fisiologia , Transfecção , Canal de Ânion 1 Dependente de Voltagem/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...