Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
Biochem Biophys Res Commun ; 625: 122-127, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35961135

RESUMO

Human immunodeficiency virus-1 (HIV-1) infection leads to the development of acquired immunodeficiency syndrome (AIDS). To establish a productive infection, HIV-1 hijacks the cellular machinery and modulates various physiological processes to propagate itself. The pathways altered by HIV-1 include cell cycle, autophagy, apoptosis, cell stress pathways, immune response, antiviral response, etc. Zipper interacting protein kinase (ZIPK) is a member of the death-associated protein kinase (DAPK) family of proteins, known to be one of the key regulators of cell death and cell survival pathways. ZIPK is also involved in regulating many cellular processes that are altered during HIV-1 infection; thus, we have explored the functional role of ZIPK in HIV-1 infection. Our results show that ZIPK protein expression is downregulated during HIV-1 infection in Nef dependent manner. Overexpression of ZIPK leads to downregulation in LTR-driven gene expression and virus production, whereas ZIPK knockdown induces viral gene expression and replication. HIV-1 promoter activity is reportedly enhanced by Nef-mediated activation of some transcription factors like NFκB and STAT3. ZIPK is reported to inhibit the STAT3 activity by phosphorylating it at ser-727. Our results show that STAT3 (ser-727) phosphorylation is decreased upon overexpression of Nef with simultaneous downregulation of ZIPK expression. We finally show that HIV-1 Nef interacts with ZIPK and induces its proteasomal degradation. Overall, our data suggests that Nef is involved in downregulation of ZIPK thereby increasing the virus production through rescue of STAT3 activity.


Assuntos
Produtos do Gene nef , HIV-1 , Proteínas Quinases Associadas com Morte Celular , Produtos do Gene nef/fisiologia , HIV-1/genética , Humanos , Proteínas Quinases , Proteínas Virais , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
2.
PLoS Pathog ; 14(8): e1007269, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30125328

RESUMO

SERINC5 is a host restriction factor that impairs infectivity of HIV-1 and other primate lentiviruses and is counteracted by the viral accessory protein Nef. However, the importance of SERINC5 antagonism for viral replication and cytopathicity remained unclear. Here, we show that the Nef protein of the highly divergent SIVcol lineage infecting mantled guerezas (Colobus guereza) is a potent antagonist of SERINC5, although it lacks the CD4, CD3 and CD28 down-modulation activities exerted by other primate lentiviral Nefs. In addition, SIVcol Nefs decrease CXCR4 cell surface expression, suppress TCR-induced actin remodeling, and counteract Colobus but not human tetherin. Unlike HIV-1 Nef proteins, SIVcol Nef induces efficient proteasomal degradation of SERINC5 and counteracts orthologs from highly divergent vertebrate species, such as Xenopus frogs and zebrafish. A single Y86F mutation disrupts SERINC5 and tetherin antagonism but not CXCR4 down-modulation by SIVcol Nef, while mutation of a C-proximal di-leucine motif has the opposite effect. Unexpectedly, the Y86F change in SIVcol Nef had little if any effect on viral replication and CD4+ T cell depletion in preactivated human CD4+ T cells and in ex vivo infected lymphoid tissue. However, SIVcol Nef increased virion infectivity up to 10-fold and moderately increased viral replication in resting peripheral blood mononuclear cells (PBMCs) that were first infected with HIV-1 and activated three or six days later. In conclusion, SIVcol Nef lacks several activities that are conserved in other primate lentiviruses and utilizes a distinct proteasome-dependent mechanism to counteract SERINC5. Our finding that evolutionarily distinct SIVcol Nefs show potent anti-SERINC5 activity supports a relevant role of SERINC5 antagonism for viral fitness in vivo. Our results further suggest this Nef function is particularly important for virion infectivity under conditions of limited CD4+ T cell activation.


Assuntos
Linfócitos T CD4-Positivos/virologia , Produtos do Gene nef/fisiologia , HIV-1/fisiologia , Tecido Linfoide/virologia , Proteínas de Membrana/metabolismo , Replicação Viral/genética , Animais , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Colobus/virologia , Células HEK293 , Humanos , Células Jurkat , Proteínas de Membrana/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Vírus da Imunodeficiência Símia/genética
4.
Curr HIV Res ; 9(7): 474-89, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22103831

RESUMO

The Nef protein is an essential factor for lentiviral pathogenesis in humans and other simians. Despite a multitude of functions attributed to this protein, the exact role of Nef in disease progression remains unclear. One of its most intriguing functions is the ability of Nef to enhance the infectivity of viral particles. In this review we will discuss current insights in the mechanism of this well-known, yet poorly understood Nef effect. We will elaborate on effects of Nef, on both virion biogenesis and the early stage of the cellular infection, that might be involved in infectivity enhancement. In addition, we provide an overview of different HIV-1 Nef domains important for optimal infectivity and briefly discuss some possible sources of the frequent discrepancies in the field. Hereby we aim to contribute to a better understanding of this highly conserved and therapeutically attractive Nef function.


Assuntos
Produtos do Gene nef/fisiologia , HIV/patogenicidade , Vírion/patogenicidade , Replicação Viral/fisiologia , Animais , Produtos do Gene nef/genética , HIV/genética , HIV-1 , Humanos , Macaca mulatta , Mutação
5.
Curr HIV Res ; 9(7): 514-23, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22103835

RESUMO

The multifunctional Nef protein of primate lentiviruses is commonly considered an early viral factor that down-modulates various receptors from the cell surface and modulates several signaling pathways to facilitate viral immune evasion and to render the cell conducive for viral replication. However, Nef also acts during the late stages of infection, e.g. by increasing the infectivity of progeny virions. Just recently, it has become clear that many primate lentiviruses that have been detected in about 40 different monkey and ape species also use Nef to antagonize tetherin (BST2/CD317), a cellular factor that inhibits virus release by tethering nascent viral particles to the cell surface. Exceptions are some simian immunodeficiency viruses (SIVs) infecting Cercopithecus monkeys that employ their accessory Vpu protein to counteract the restriction by tetherin. Furthermore, pandemic HIV-1 group M strains switched from Nef to Vpu and HIV-2 group A isolates from Nef to Env after zoonotic transmission from chimpanzees and sooty mangabeys, respectively, to antagonize the tetherin restriction in humans. These evolutionary switches were most likely enforced by a deletion in the cytoplasmic domain of the human tetherin orthologue that confers resistance to Nef. Here, we summarize some of our current knowledge about Nef-mediated tetherin antagonism.


Assuntos
Produtos do Gene nef/fisiologia , HIV/fisiologia , Infecções por Lentivirus/imunologia , Lentivirus de Primatas/fisiologia , Primatas/virologia , Animais , Antígenos CD/fisiologia , Evolução Biológica , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/fisiologia , HIV-1/fisiologia , HIV-2/fisiologia , Humanos , Lentivirus de Primatas/patogenicidade , Especificidade da Espécie , Proteínas Virais Reguladoras e Acessórias/fisiologia , Liberação de Vírus/fisiologia
6.
Curr HIV Res ; 9(7): 531-42, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22103837

RESUMO

A bewildering number of host cell proteins associated with Nef can be found in the literature and in the public protein interaction databases. However, only in a few of these cases, including binding of Nef to certain Src homology-3 (SH3) domain proteins, is the interaction understood in any molecular detail or even known to be direct. Indeed, SH3 binding capacity by Nef is required for many of the other protein interactions to take place, suggesting that a large proportion of the latter is indirectly coupled to Nef via an SH3 protein. Accordingly, the proline-rich SH3 binding site, the "PxxP motif", is one of the key functional determinants of Nef. It is highly conserved among the lentiviral Nef proteins, and mutations disrupting it abrogate the majority of the known effects of Nef on host cell physiology. This review summarizes the current understanding as well as the outstanding gaps in our knowledge regarding the relevant SH3 protein partners and SH3-dependent cellular functions of Nef. The roles of these interactions in the pathogenesis of AIDS and their potential as targets for antiviral drug development are also discussed.


Assuntos
Produtos do Gene nef/fisiologia , HIV/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Vírus da Imunodeficiência Símia/fisiologia , Domínios de Homologia de src , Síndrome da Imunodeficiência Adquirida/imunologia , Animais , Produtos do Gene nef/química , HIV/patogenicidade , Humanos , Macrófagos/metabolismo , Vírus da Imunodeficiência Símia/patogenicidade , Linfócitos T/metabolismo
7.
Angiology ; 61(7): 669-78, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20566577

RESUMO

Human immunodeficiency virus (HIV)-infected patients have increased rates of atherosclerotic cardiovascular diseases because the highly active antiretroviral therapy (HAART) decreased the morbidity and mortality of the disease. Endothelial dysfunction is possibly the most plausible link between HIV infection and related expression of cell adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) on the endothelial cells. HIV-1 accessory protein negative regulate factor (Nef) has been shown to be very important for high virus replication and disease progression. Nef could upregulate the expression of ICAM-1 in the pathogenesis of HIV infection. Here, we provide evidence that the HIV-1 Nef can transcriptionally induce the expression of ICAM-1 in stable expressed Nef vascular endothelial cells. Nef-induced ICAM-1 upregulation requires the activation of the downstream kinase extracellular signal-regulated kinase (ERK). Flow cytometry (FCM) results showed that the percentage of ICAM-1 positive cells in Nef-expressed cells and control cells was (35.3% +/- 2.2%) and (12.5% +/- 0.8%), respectively (P < .01). Furthermore, inhibition of Nef activity by ERK mitogen-activated protein kinase (MAPK) inhibitor effectively blocked ICAM-1 upregulation, suggesting that ERK MAPK activation is an important initiating event in Nef-mediated ICAM-1 expression in Nef-expressed cells. These data demonstrate an important signaling event of Nef in HIV-1 pathogenesis.


Assuntos
Células Endoteliais/metabolismo , Infecções por HIV/genética , Molécula 1 de Adesão Intercelular/biossíntese , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Western Blotting , Células Cultivadas , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Regulação da Expressão Gênica/fisiologia , Produtos do Gene nef/fisiologia , Infecções por HIV/metabolismo , HIV-1 , Humanos , Molécula 1 de Adesão Intercelular/genética , Células Jurkat/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Transdução de Sinais , Transcrição Gênica , Transfecção , Regulação para Cima/fisiologia
8.
Trends Cell Biol ; 20(3): 160-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20071173

RESUMO

p21-activated protein kinases (Paks) are cytosolic serine/threonine protein kinases that act as effectors for small (p21) GTPases of the Cdc42 and Rac families. It has long been established that Paks play a major role in a host of vital cellular functions such as proliferation, survival and motility, and abnormal Pak function is associated with a number of human diseases. Here, we discuss emerging evidence that these enzymes also play a major role in the entry, replication and spread of many important pathogenic human viruses, including HIV. Careful assessment of the potential role of Paks in antiviral immunity will be pivotal to evaluate thoroughly the potential of agents that inhibit Pak as a new class of anti-viral therapeutics.


Assuntos
Viroses/fisiopatologia , Quinases Ativadas por p21/fisiologia , Adenoviridae/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Apoptose/fisiologia , Ativação Enzimática , Produtos do Gene nef/fisiologia , Hepadnaviridae/fisiologia , Herpesviridae/fisiologia , Humanos , Dados de Sequência Molecular , Poxviridae/fisiologia , Alinhamento de Sequência , Internalização do Vírus , Replicação Viral/fisiologia
9.
J Virol ; 83(22): 11673-81, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19726508

RESUMO

The Vpu accessory gene that originated in the primate lentiviral lineage leading to human immunodeficiency virus type 1 is an antagonist of human tetherin/BST-2 restriction. Most other primate lentivirus lineages, including the lineage represented by simian immunodeficiency virus SIVagm from African green monkeys (AGMs), do not encode Vpu. While some primate lineages encode gene products other than Vpu that overcome tetherin/BST-2, we find that SIVagm does not antagonize physiologically relevant levels of AGM tetherin/BST-2. AGM tetherin/BST-2 can be induced by low levels of type I interferon and can potently restrict two independent strains of SIVagm. Although SIVagm Nef had an effect at low levels of AGM tetherin/BST-2, simian immunodeficiency virus SIVmus Vpu, from a virus that infects the related monkey Cercopithecus cephus, is able to antagonize even at high levels of AGM tetherin/BST-2 restriction. We propose that since the replication of SIVagm does not induce interferon production in vivo, tetherin/BST-2 is not induced, and therefore, SIVagm does not need Vpu. This suggests that primate lentiviruses evolve tetherin antagonists such as Vpu or Nef only if they encounter tetherin during the typical course of natural infection.


Assuntos
Chlorocebus aethiops/virologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Western Blotting , Linhagem Celular , Produtos do Gene nef/fisiologia , Interferon alfa-2 , Interferon beta-1b , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Dados de Sequência Molecular , Proteínas Recombinantes , Proteínas dos Retroviridae/genética , Proteínas dos Retroviridae/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/fisiologia
10.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L729-37, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19648286

RESUMO

Golgi dysfunction has been previously investigated as a mechanism involved in monocrotaline-induced pulmonary hypertension (PAH). In the present study, we addressed whether Golgi dysfunction might occur in pulmonary vascular cells in idiopathic PAH (IPAH) and whether there might be a causal relationship between trafficking dysfunction and vasculopathies of PAH. Quantitative immunostaining for the Golgi tethers giantin and p115 on human lung tissue from patients with IPAH (n = 6) compared with controls demonstrated a marked cytoplasmic dispersal of giantin- and p115-bearing vesicular elements in vascular cells in the proliferative, obliterative, and plexiform lesions in IPAH and an increase in the amounts of these Golgi tethers/matrix proteins per cell. The causality question was approached by genetic means using human immunodeficiency virus (HIV)-Nef, a protein that disrupts endocytic and trans-Golgi trafficking. Macaques infected with a chimeric simian immunodeficiency virus (SIV) containing the HIV-nef gene (SHIV-nef), but not the nonchimeric SIV virus containing the endogenous SIV-nef gene, displayed pulmonary arterial vasculopathies similar to those in human IPAH. Giantin and p115 levels and their subcellular distribution in pulmonary vascular cells in lungs of SHIV-nef infected macaques (n = 4) were compared with SIV-infected (n = 3) and an uninfected macaque control. Only macaques infected with chimeric SHIV-nef showed pulmonary vascular lesions containing cells with dramatic cytoplasmic dispersal and an increase in giantin and p115. Specifically, the HIV-Nef-positive cells showed increased giantin, p115, and the activated transcription factor PY-STAT3. These data represent the first test of the Golgi dysfunction hypothesis in IPAH and place trafficking and Golgi disruption in the chain of causality of pulmonary vasculopathies in the macaque model.


Assuntos
Endotélio Vascular/patologia , Produtos do Gene nef/fisiologia , Complexo de Golgi/patologia , Hipertensão Pulmonar/patologia , Macaca/virologia , Artéria Pulmonar/patologia , Doenças Vasculares/patologia , Animais , Estudos de Casos e Controles , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Imunofluorescência , Complexo de Golgi/metabolismo , Proteínas da Matriz do Complexo de Golgi , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Humanos , Hipertensão Pulmonar/metabolismo , Proteínas de Membrana/metabolismo , Artéria Pulmonar/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/isolamento & purificação , Doenças Vasculares/metabolismo
11.
J Immunol ; 183(4): 2415-24, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19620308

RESUMO

HIV-1 Nef has been reported to disrupt MHC class II (MHCII)-mediated Ag presentation by a dual strategy that comprises a reduction in cell surface levels of peptide-loaded mature MHCII molecules and a up-regulation of immature MHCII molecules. We show that Nef achieves relocation of MHCII away from the cell surface in monocytic cells by both delaying its transport to the cell surface and by accelerating endocytic removal of cell surface MHCII to a lysosomal compartment. Nef-induced MHCII endocytosis is cholesterol-sensitive but clathrin- and dynamin-independent. Internalized MHCII molecules traverse the early endosomal system and colocalize with pinocytic cargo before reaching lysosomes. Nef-triggered MHCII endocytosis requires Rab5 activity and lyst function, whereas lysosomal trafficking of internalized MHCII molecules requires Rab7 activity. We further show that a similar pathway can remove peptide-MHCII complexes from the surface of monocytic cells not expressing Nef. Our data suggest that Nef uses mechanisms involved in normal MHCII recycling and turnover to mediate the delivery of cell surface MHCII to a lysosomal destination. Thus, Nef-mediated endocytosis of MHCII provides a novel perspective on the regulation of normal MHCII trafficking.


Assuntos
Membrana Celular/metabolismo , Endocitose/imunologia , Produtos do Gene nef/fisiologia , HIV-1/imunologia , Antígenos HLA-D/metabolismo , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/virologia , Células Cultivadas , Endocitose/genética , HIV-1/genética , Antígenos HLA-D/biossíntese , Antígenos HLA-D/fisiologia , Humanos , Lisossomos/imunologia , Lisossomos/metabolismo , Lisossomos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/genética , Células U937
12.
Nat Rev Microbiol ; 7(6): 467-76, 2009 06.
Artigo em Inglês | MEDLINE | ID: mdl-19305418

RESUMO

In the subset of primate lentiviruses that contain a vpu gene - HIV-1 and its simian precursors - the Nef protein has lost the ability to down-modulate CD3, block T cell activation and suppress programmed death. Vpu counteracts a host restriction factor induced by the inflammatory cytokine interferon-alpha. I propose that the acquisition of vpu may have allowed the viral lineage that gave rise to HIV-1 to evolve towards greater pathogenicity by removing the selective pressure for a protective Nef function that prevents damagingly high levels of immune activation.


Assuntos
Evolução Molecular , HIV-1/patogenicidade , Animais , Produtos do Gene nef/metabolismo , Produtos do Gene nef/fisiologia , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Humanos , Modelos Biológicos , Vírus da Imunodeficiência Símia/metabolismo , Vírus da Imunodeficiência Símia/patogenicidade , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Virais Reguladoras e Acessórias/fisiologia
13.
Traffic ; 9(11): 1925-35, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18764822

RESUMO

The Nef protein of HIV-1 removes the immune costimulatory proteins CD80 and CD86 from the cell surface by a unique clathrin- and dynamin-independent, actin-based endocytic pathway that deploys coupled activation of c-src and Rac. In this study, we show that, similar to major histocompatibility complex class I (MHCI), Nef subsequently reroutes CD80 and CD86 to the Golgi region. However, not only are CD80/CD86 internalized by a different mechanism from MHCI but also the vesicular pathway of Golgi delivery for CD80/CD86 is distinct from that employed for MHCI. While MHCI passes through early endosomal and sorting compartments marked by Rab5/early embryonic antigen 1 and ADP ribosylation factor 6, respectively, CD80 and CD86 enter endocytic vesicles that do not acquire conventional early endosomal markers but remain accessible to fluid probes. Rather than being delivered to preexisting Rab11-positive recycling compartments, these vesicles recruit Rab11 de novo. Rab11 activity is also necessary for the delivery of CD80/CD86 in these transitional vesicles to the Golgi region. These data reveal an unusual pathway of endocytic vesicular traffic to the Golgi and its recruitment in a viral immune evasion strategy.


Assuntos
Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Endocitose , Produtos do Gene nef/fisiologia , Complexo de Golgi/metabolismo , HIV-1/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Fator 6 de Ribosilação do ADP , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Células U937
14.
J Virol ; 82(16): 7758-67, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18524831

RESUMO

The human immunodeficiency virus type 1 (HIV-1) Nef protein upregulates the expression of the invariant chain (Ii)/major histocompatibility complex class II (MHC-II) complex at the cell surface. This complex appears to reach the antigen-loading endosomal compartment at least in part via an indirect pathway in which it is internalized from the cell surface via the adaptor protein 2 (AP-2) complex. Here we provide evidence for a competition model to explain how Nef upregulates the expression of Ii at the cell surface. In this model, Nef and Ii compete for binding to AP-2. In support of this model, Nef decreased the rate of internalization of Ii from the cell surface. The AP-binding dileucine motif in Nef, ENTSLL(165), was necessary and sufficient for the upregulation of Ii. In addition, two leucine-based AP-binding motifs in the Ii cytoplasmic tail, DDQRDLI(8) and EQLPML(17), were critical for the efficient upregulation of Ii by Nef. Experiments using Nef variants in which the native dileucine-based sorting motif was replaced with similar motifs from cellular transmembrane proteins allowed modulation of AP-binding specificity. Analysis of these variants suggested that the binding of Nef to AP-2 is sufficient to upregulate Ii at the plasma membrane. Finally, interference with the expression of AP-2 caused an upregulation of Ii at the plasma membrane, and this decreased the effect of Nef. These data indicate that Nef usurps AP-2 complexes to dysregulate Ii trafficking and potentially interfere with antigen presentation in the context of MHC-II.


Assuntos
Regulação Viral da Expressão Gênica , Produtos do Gene nef/biossíntese , Produtos do Gene nef/fisiologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Complexo 2 de Proteínas Adaptadoras/metabolismo , Motivos de Aminoácidos , Apresentação de Antígeno , Ligação Competitiva , Linfócitos T CD4-Positivos/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Antígenos de Histocompatibilidade Classe II/química , Humanos , Leucina/química , Leucócitos Mononucleares/metabolismo , Modelos Biológicos
16.
J Virol ; 82(6): 2918-29, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094167

RESUMO

Alterations of T-cell receptor signaling by human immunodeficiency virus type 1 (HIV-1) Nef involve its association with a highly active subpopulation of p21-activated kinase 2 (PAK2) within a dynamic signalosome assembled in detergent-insoluble membrane microdomains. Nef-PAK2 complexes contain the GTPases Rac and Cdc42 as well as a factor providing guanine nucleotide exchange factor (GEF) activity for Rac/Cdc42. However, the identity of this GEF has remained controversial. Previous studies suggested the association of Nef with at least three independent GEFs, Vav, DOCK2/ELMO1, and betaPix. Here we used a broad panel of approaches to address which of these GEFs is involved in the functional interaction of Nef with PAK2 activity. Biochemical fractionation and confocal microscopy revealed that Nef recruits Vav1, but not DOCK2/ELMO1 or betaPix, to membrane microdomains. Transient RNAi knockdown, analysis of cell lines defective for expression of Vav1 or DOCK2 as well as use of a betaPix binding-deficient PAK2 variant confirmed a role for Vav1 but not DOCK2 or betaPix in Nef's association with PAK2 activity. Nef-mediated microdomain recruitment of Vav1 occurred independently of the Src homology 3 domain binding PxxP motif, which is known to connect Nef to many cellular signaling processes. Instead, a recently described protein interaction surface surrounding Nef residue F195 was identified as critical for Nef-mediated raft recruitment of Vav1. These results identify Vav1 as a relevant component of the Nef-PAK2 signalosome and provide a molecular basis for the role of F195 in formation of a catalytically active Nef-PAK2 complex.


Assuntos
Produtos do Gene nef/fisiologia , HIV-1/fisiologia , Proteínas Proto-Oncogênicas c-vav/metabolismo , Quinases Ativadas por p21/metabolismo , Sequência de Bases , Membrana Celular/metabolismo , Ligação Proteica , Interferência de RNA
18.
J Virol ; 81(20): 11426-40, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17670831

RESUMO

Primary CD4(+) T lymphocytes, supporting in vitro human immunodeficiency virus type 1 (HIV-1) replication, are destined to die by apoptosis. We explored the initial molecular events that act upstream from mitochondrial dysfunction in CD4(+) T lymphocytes exposed to the HIV-1(LAI) strain. We tracked by immunofluorescence the cells expressing the p24 viral antigen and used Percoll density gradients to isolate a nonapoptotic CD4(+) T-cell subset with a high inner mitochondrial transmembrane potential (DeltaPsim) but no outer mitochondrial membrane (OMM) rupture. In most p24(+) (but not bystander p24(-)) cells of this subset, the lysosomes were undergoing limited membrane permeabilization, allowing the lysosomal efflux of cathepsins (Cat) to the cytosol. This was also induced by HIV-1 isolates from infected patients. Using pepstatin A to inhibit Cat-D enzymatic activity and Cat-D small interfering RNA to silence the Cat-D gene, we demonstrate that once released into the cytosol, Cat-D induces the conformational change of Bax and its insertion into the OMM. Inhibition of Cat-D activity/expression also conferred a transient survival advantage upon productively HIV-1-infected cells, indicating that Cat-D is an early death factor. The transfection of activated CD4(+) T lymphocytes with a Nef expression vector rapidly induced the permeabilization of lysosomes and the release of Cat-D, with these two events preceding OMM rupture. These results reveal a previously undocumented mechanism in which Nef acts as an internal cytopathic factor and strongly suggest that this viral protein may behave similarly in the context of productive HIV-1 infection in CD4(+) T lymphocytes.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/virologia , Produtos do Gene nef/fisiologia , HIV-1/patogenicidade , Lisossomos/virologia , Linfócitos T CD4-Positivos/patologia , Células Cultivadas , Infecções por HIV/patologia , Humanos , Membranas Intracelulares/virologia , Lisossomos/ultraestrutura , Permeabilidade
19.
Curr Drug Discov Technol ; 4(1): 12-23, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17630924

RESUMO

Human immunodeficiency virus (HIV)-1 Nef is a regulatory protein critically involved in AIDS pathogenesis. We previously demonstrated that extracellular Nef is efficiently internalized by human primary monocyte-derived macrophages (MDMs), thereby activating a number of transcription factors including STATs, MAPKs, IRF-3, and NF-kappaB. Such an activation state leads to the release of inflammatory factors whose paracrine effects deserve deep consideration. Here, we demonstrate that quiescent CD4 lymphocytes undergo cell activation when cultivated in supernatants from autologous MDMs treated with extracellular wt Nef but not with its counterpart mutated in the (72)PxxP(75) polyproline domain. Of a pathogenetic relevance, this effect coupled with the sensitization of quiescent CD4 lymphocytes to HIV-1 infection. By microarray assay, we found that the CCL24/eotaxin-2 gene was up-regulated in MDMs treated with wt Nef but not with the (72)AxxA(75) mutant. In addition, the higher transcription activity correlated with a significant increase of the CCL24/Eotaxin-2 release. Finally, we observed that anti-CCL24/eotaxin-2 antibodies efficiently neutralized the stimulatory effect on CD4 lymphocytes of supernatants from MDMs treated with extracellular Nef. Overall, these data support the idea that CCL24/eotaxin-2 is part of the mechanism of CD4 lymphocyte activation paracrinally induced by Nef.


Assuntos
Quimiocinas CC/fisiologia , Produtos do Gene nef/fisiologia , HIV-1/patogenicidade , Adulto , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Quimiocina CCL24 , Quimiocinas CC/biossíntese , Produtos do Gene nef/genética , Produtos do Gene nef/metabolismo , HIV-1/fisiologia , Humanos , Leucócitos Mononucleares , Ativação Linfocitária , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Comunicação Parácrina , Peptídeos/genética , Transcrição Gênica , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana
20.
J Immunol ; 178(9): 5762-8, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17442960

RESUMO

The HIV-Nef protein has been implicated in generating high viral loads and T cell activation. Transgenic (tg) mice with constitutive T cell-specific Nef expression show a dramatic reduction in T cell number and highly increased T cell turnover. Previous studies in Nef tg mice attributed this T cell activation to a direct effect of Nef at the cellular level. Given the strongly reduced peripheral T cell numbers, we examined whether this enhanced T cell division might instead be lymphopenia induced. Adoptively transferred naive wild-type T cells into lymphopenic Nef tg mice showed high T cell turnover and obtained the same effector/memory phenotype as the autologous Nef tg T cells, supporting the idea that the microenvironment determines the phenotype of the T cells present. Moreover, in bone marrow chimeras from mixtures of wild-type and Nef tg bone marrow, with a full T cell compartment containing a small proportion of Nef tg T cells, Nef tg T cells kept a naive phenotype. These results demonstrate that T cell activation in the Nef tg mice is lymphopenia induced rather than due to a direct T cell-activating effect of Nef.


Assuntos
Produtos do Gene nef/fisiologia , HIV/fisiologia , Ativação Linfocitária , Linfopenia/imunologia , Linfócitos T/imunologia , Animais , Células da Medula Óssea/imunologia , Proliferação de Células , Quimera/imunologia , Produtos do Gene nef/genética , HIV/genética , Contagem de Linfócitos , Camundongos , Camundongos Transgênicos , Produtos do Gene nef do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...