Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Mol Biol ; 434(6): 167421, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-34954236

RESUMO

Human SERINC5 (SER5) protein is a recently described restriction factor against human immunodeficiency virus-1 (HIV-1), which is antagonized by HIV-1 Nef protein. Other retroviral accessory proteins such as the glycosylated Gag (glycoGag) from the murine leukemia virus (MLV) can also antagonize SER5. In addition, some viruses escape SER5 restriction by expressing a SER5-insensitive envelope (Env) glycoprotein. Here, we studied the activity of human and feline SER5 on HIV-1 and on the two pathogenic retroviruses in cats, feline immunodeficiency virus (FIV) and feline leukemia virus (FeLV). HIV-1 in absence of Nef is restricted by SER5 from domestic cats and protected by its Nef protein. The sensitivity of feline retroviruses FIV and FeLV to human and feline SER5 is considerably different: FIV is sensitive to feline and human SER5 and lacks an obvious mechanism to counteract SER5 activity, while FeLV is relatively resistant to SER5 inhibition. We speculated that similar to MLV, FeLV-A or FeLV-B express glycoGag proteins and investigated their function against human and feline SER5 in wild type and envelope deficient virus variants. We found that the endogenous FeLV recombinant virus, FeLV-B but not wild type exogenous FeLV-A envelope mediates a strong resistance against human and feline SER5. GlycoGag has an additional but moderate role to enhance viral infectivity in the presence of SER5 that seems to be dependent on the FeLV envelope. These findings may explain, why in vivo FeLV-B has a selective advantage and causes higher FeLV levels in infected cats compared to infections of FeLV-A only.


Assuntos
HIV-1 , Vírus da Imunodeficiência Felina , Vírus da Leucemia Felina , Proteínas de Membrana , Proteínas do Envelope Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana , Animais , Gatos , Glicosilação , HIV-1/fisiologia , Humanos , Vírus da Imunodeficiência Felina/fisiologia , Vírus da Leucemia Felina/fisiologia , Proteínas de Membrana/fisiologia , Proteínas do Envelope Viral/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia
2.
J Virol ; 95(16): e0058821, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34037423

RESUMO

Serine incorporator 5 (SERINC5) reduces the infectivity of progeny HIV-1 virions by incorporating into the outer host-derived viral membrane during egress. To counter SERINC5, the HIV-1 accessory protein Nef triggers SERINC5 internalization by engaging the adaptor protein 2 (AP-2) complex using the [D/E]xxxL[L/I]167 Nef dileucine motif. Nef also engages AP-2 via its dileucine motif to downregulate the CD4 receptor. Although these two Nef functions are related, the mechanisms governing SERINC5 downregulation are incompletely understood. Here, we demonstrate that two primary Nef isolates, referred to as 2410 and 2391 Nef, acquired from acutely HIV-1 infected women from Zimbabwe, both downregulate CD4 from the cell surface. However, only 2410 Nef retains the ability to downregulate cell surface SERINC5. Using a series of Nef chimeras, we mapped the region of 2391 Nef responsible for the functional uncoupling of these two antagonistic pathways to the dileucine motif. Modifications of the first and second x positions of the 2410 Nef dileucine motif to asparagine and aspartic acid residues, respectively (ND164), impaired cell surface SERINC5 downregulation, which resulted in reduced infectious virus yield in the presence of SERINC5. The ND164 mutation additionally partially impaired, but did not completely abrogate, Nef-mediated cell surface CD4 downregulation. Furthermore, the patient infected with HIV-1 encoding 2391 Nef had stable CD4+ T cell counts, whereas infection with HIV-1 encoding 2410 Nef resulted in CD4+ T cell decline and disease progression. IMPORTANCE A contributing factor to HIV-1 persistence is evasion of the host immune response. HIV-1 uses the Nef accessory protein to evade the antiviral roles of the adaptive and intrinsic innate immune responses. Nef targets SERINC5, a restriction factor which potently impairs HIV-1 infection by triggering SERINC5 removal from the cell surface. The molecular determinants underlying this Nef function remain incompletely understood. Recent studies have found a correlation between the extent of Nef-mediated SERINC5 downregulation and the rate of disease progression. Furthermore, single-residue polymorphisms outside the known Nef functional motifs can modulate SERINC5 downregulation. The identification of a naturally occurring Nef polymorphism impairing SERINC5 downregulation in this study supports a link between Nef downregulation of SERINC5 and the rate of plasma CD4+ T cell decline. Moreover, the observed functional impairments of this polymorphism could provide clues to further elucidate unknown aspects of the SERINC5 antagonistic pathway via Nef.


Assuntos
Antígenos CD4/metabolismo , Infecções por HIV/virologia , HIV-1/patogenicidade , Proteínas de Membrana/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Motivos de Aminoácidos , Linfócitos T CD4-Positivos/patologia , Progressão da Doença , Regulação para Baixo , Feminino , Infecções por HIV/metabolismo , HIV-1/genética , Humanos , Mutação , Polimorfismo Genético , Vírion , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo
3.
Virus Genes ; 55(4): 541-544, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31093843

RESUMO

Almost 80% of viral transcripts during early HIV-1 infection encode the Nef protein, which has been implicated in altering expression of a number of genes. In this study, we infected primary human CD4+ T cells with pseudotyped Nef-containing or Nef-deleted (Δ-nef) NL4-3 virus and used RNA-Sequencing (RNA-Seq) for transcriptomic analysis. Our results showed that the interferon response, IL-15 and JAK/STAT signaling, as well as genes involved in metabolism, apoptosis, cell cycle regulation, and ribosome biogenesis were all altered in the presence of Nef. These early Nef-mediated transcriptional alterations may play a role in priming the host cell for cellular activation and viral replication.


Assuntos
Linfócitos T CD4-Positivos/virologia , Regulação Viral da Expressão Gênica , HIV-1/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Humanos , Replicação Viral
4.
Proc Natl Acad Sci U S A ; 116(12): 5705-5714, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30842281

RESUMO

The T cell Ig and mucin domain (TIM) proteins inhibit release of HIV-1 and other enveloped viruses by interacting with cell- and virion-associated phosphatidylserine (PS). Here, we show that the Nef proteins of HIV-1 and other lentiviruses antagonize TIM-mediated restriction. TIM-1 more potently inhibits the release of Nef-deficient relative to Nef-expressing HIV-1, and ectopic expression of Nef relieves restriction. HIV-1 Nef does not down-regulate the overall level of TIM-1 expression, but promotes its internalization from the plasma membrane and sequesters its expression in intracellular compartments. Notably, Nef mutants defective in modulating membrane protein endocytic trafficking are incapable of antagonizing TIM-mediated inhibition of HIV-1 release. Intriguingly, depletion of SERINC3 or SERINC5 proteins in human peripheral blood mononuclear cells (PBMCs) attenuates TIM-1 restriction of HIV-1 release, in particular that of Nef-deficient viruses. In contrast, coexpression of SERINC3 or SERINC5 increases the expression of TIM-1 on the plasma membrane and potentiates TIM-mediated inhibition of HIV-1 production. Pulse-chase metabolic labeling reveals that the half-life of TIM-1 is extended by SERINC5 from <2 to ∼6 hours, suggesting that SERINC5 stabilizes the expression of TIM-1. Consistent with a role for SERINC protein in potentiating TIM-1 restriction, we find that MLV glycoGag and EIAV S2 proteins, which, like Nef, antagonize SERINC-mediated diminishment of HIV-1 infectivity, also effectively counteract TIM-mediated inhibition of HIV-1 release. Collectively, our work reveals a role of Nef in antagonizing TIM-1 and highlights the complex interplay between Nef and HIV-1 restriction by TIMs and SERINCs.


Assuntos
Infecções por HIV/metabolismo , Receptor Celular 1 do Vírus da Hepatite A/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Membrana Celular/metabolismo , Regulação para Baixo , Células HEK293 , Soropositividade para HIV , HIV-1/metabolismo , HIV-1/patogenicidade , Receptor Celular 1 do Vírus da Hepatite A/antagonistas & inibidores , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Leucócitos Mononucleares/metabolismo , Glicoproteínas de Membrana , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Transporte Proteico , Receptores de Superfície Celular/metabolismo , Vírion/metabolismo , Replicação Viral/efeitos dos fármacos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo
5.
PLoS Comput Biol ; 14(9): e1006409, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30192744

RESUMO

Mutational correlation patterns found in population-level sequence data for the Human Immunodeficiency Virus (HIV) and the Hepatitis C Virus (HCV) have been demonstrated to be informative of viral fitness. Such patterns can be seen as footprints of the intrinsic functional constraints placed on viral evolution under diverse selective pressures. Here, considering multiple HIV and HCV proteins, we demonstrate that these mutational correlations encode a modular co-evolutionary structure that is tightly linked to the structural and functional properties of the respective proteins. Specifically, by introducing a robust statistical method based on sparse principal component analysis, we identify near-disjoint sets of collectively-correlated residues (sectors) having mostly a one-to-one association to largely distinct structural or functional domains. This suggests that the distinct phenotypic properties of HIV/HCV proteins often give rise to quasi-independent modes of evolution, with each mode involving a sparse and localized network of mutational interactions. Moreover, individual inferred sectors of HIV are shown to carry immunological significance, providing insight for guiding targeted vaccine strategies.


Assuntos
Infecções por HIV/virologia , HIV-1 , Hepacivirus , Hepatite C/virologia , Algoritmos , Alelos , Biologia Computacional , Simulação por Computador , Análise Mutacional de DNA , DNA Viral , Progressão da Doença , Evolução Molecular , Proteína do Núcleo p24 do HIV/fisiologia , Antígenos HLA/química , Humanos , Sistema Imunitário , Distribuição Normal , Fenótipo , Análise de Componente Principal , Domínios Proteicos , Relação Estrutura-Atividade , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia
6.
Cell ; 174(3): 659-671.e14, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30053425

RESUMO

The HIV accessory protein Nef counteracts immune defenses by subverting coated vesicle pathways. The 3.7 Å cryo-EM structure of a closed trimer of the clathrin adaptor AP-1, the small GTPase Arf1, HIV-1 Nef, and the cytosolic tail of the restriction factor tetherin suggested a mechanism for inactivating tetherin by Golgi retention. The 4.3 Å structure of a mutant Nef-induced dimer of AP-1 showed how the closed trimer is regulated by the dileucine loop of Nef. HDX-MS and mutational analysis were used to show how cargo dynamics leads to alternative Arf1 trimerization, directing Nef targets to be either retained at the trans-Golgi or sorted to lysosomes. Phosphorylation of the NL4-3 M-Nef was shown to regulate AP-1 trimerization, explaining how O-Nefs lacking this phosphosite counteract tetherin but most M-Nefs do not. These observations show how the higher-order organization of a vesicular coat can be allosterically modulated to direct cargoes to distinct fates.


Assuntos
Fator de Transcrição AP-1/ultraestrutura , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/ultraestrutura , Fator 1 de Ribosilação do ADP/metabolismo , Fator 1 de Ribosilação do ADP/ultraestrutura , Proteínas Adaptadoras de Transporte Vesicular , Antígeno 2 do Estroma da Médula Óssea/metabolismo , Antígeno 2 do Estroma da Médula Óssea/ultraestrutura , Clatrina , Complexo de Golgi , Células HEK293 , HIV-1 , Humanos , Transporte Proteico/fisiologia , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-1/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia
7.
Retrovirology ; 15(1): 6, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29329537

RESUMO

BACKGROUND: The HIV-1 accessory proteins Nef and Vpu alter cell surface levels of multiple host proteins to modify the immune response and increase viral persistence. Nef and Vpu can downregulate cell surface levels of the co-stimulatory molecule CD28, however the mechanism of this function has not been completely elucidated. RESULTS: Here, we provide evidence that Nef and Vpu decrease cell surface and total cellular levels of CD28. Moreover, using inhibitors we implicate the cellular degradation machinery in the downregulation of CD28. We shed light on the mechanisms of CD28 downregulation by implicating the Nef LL165 and DD175 motifs in decreasing cell surface CD28 and Nef DD175 in decreasing total cellular CD28. Moreover, the Vpu LV64 and S52/56 motifs were required for cell surface CD28 downregulation, while, unlike for CD4 downregulation, Vpu W22 was dispensable. The Vpu S52/56 motif was also critical for Vpu-mediated decreases in total CD28 protein level. Finally, the ability of Vpu to downregulate CD28 is conserved between multiple group M Vpu proteins and infection with viruses encoding or lacking Nef and Vpu have differential effects on activation upon stimulation. CONCLUSIONS: We report that Nef and Vpu downregulate cell surface and total cellular CD28 levels. We identified inhibitors and mutations within Nef and Vpu that disrupt downregulation, shedding light on the mechanisms utilized to downregulate CD28. The conservation and redundancy between the abilities of two HIV-1 proteins to downregulate CD28 highlight the importance of this function, which may contribute to the development of latently infected cells.


Assuntos
Antígenos CD28/genética , Linfócitos T CD4-Positivos/imunologia , Regulação para Baixo , Infecções por HIV/imunologia , HIV-1/fisiologia , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Motivos de Aminoácidos/genética , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Membrana Celular/metabolismo , Células Cultivadas , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Interações Hospedeiro-Patógeno , Proteínas do Vírus da Imunodeficiência Humana/química , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Ativação Linfocitária , Lisossomos/metabolismo , Mutação , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
8.
Immunol Lett ; 187: 19-26, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28479111

RESUMO

Vaccines currently available for AIDS show poor efficiency, demonstrating the need for new strategies to increase their immunogenicity. In this study, the HIV-1P24-Nef peptide was used as a model vaccine, followed by utilization of a novel strategy to increase its immunogenicity. There is a growing interest in using TLR agonists for vaccine formulations. Such molecules bind to their receptors on immune cells, especially the cell surface of antigen presenting cells, thereby activating these cells and inflammatory responses. In the present study, FLiC (flagellin molecule sequence from Pseudomonas aeruginosa) was used as a TLR5 agonist. In addition, PLGA nanoparticles were used as a transmitter system to enhance vaccine efficiency and its effective transfer to immune systems. In light of this, the P24-Nef peptide was conjugated to FLiC through chemical reactions. The HIV-1P24-Nef/FLiC conjugate was constructed as a nano-vaccine using PLGA particles. Subsequently, mice were immunized intradermally three times with three-week intervals with HIV-p24-Nef/FLiC/PLGA, HIV-p24-Nef/PLGA, FLiC/PLGA, PLGA, and PBS in two doses (20 and 5µg). Three weeks after the last booster injection, cell proliferation was assessed using the Brdu/ELISA assay, and cytotoxicity was evaluated by CFSE and splenocyte cytokine secretion (IL-4 and IFN-γ); in addition, IgG1 and IgG2a antibody isotype titers were determined using a commercial ELISA kit. Our results showed that Co-utilization of TLR5 and nano-particles not only improves vaccine immunogenicity but also decreases the immunogenic dose of vaccine candidate required. We showed that the immune system was effectively stimulated via the nano-vaccination strategy using the TLR5 agonists. The effect of this strategy showed variations in different parameters of the immune system; in this regard, cellular immune responses had a higher stimulation level, compared with humoral immune responses.


Assuntos
Vacinas contra a AIDS/farmacologia , HIV-1/imunologia , Imunogenicidade da Vacina , Peptídeos/farmacologia , Receptor 5 Toll-Like/agonistas , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Vacinas contra a AIDS/imunologia , Animais , Relação Dose-Resposta a Droga , Relação Dose-Resposta Imunológica , Feminino , Anticorpos Anti-HIV/imunologia , Humanos , Imunoglobulina G/imunologia , Interferon gama/imunologia , Interleucina-4/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia
9.
J Virol ; 90(23): 10915-10927, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27681140

RESUMO

SERINC3 (serine incorporator 3) and SERINC5 are recently identified host cell inhibitors of HIV-1 particle infectivity that are counteracted by the viral pathogenesis factor Nef. Here we confirm that HIV-1 Nef, but not HIV-1 Vpu, antagonizes the particle infectivity restriction of SERINC5. SERINC5 antagonism occurred in parallel with other Nef activities, including cell surface receptor downregulation, trans-Golgi network targeting of Lck, and inhibition of host cell actin dynamics. Interaction motifs with host cell endocytic machinery and the Nef-associated kinase complex, as well as CD4 cytoplasmic tail/HIV-1 protease, were identified as essential Nef determinants for SERINC5 antagonism. Characterization of antagonism-deficient Nef mutants revealed that counteraction of SERINC5 occurs in the absence of retargeting of the restriction factor to intracellular compartments and reduction of SERINC5 cell surface density is insufficient for antagonism. Consistent with virion incorporation of SERINC5 being a prerequisite for its antiviral activity, the infectivity of HIV-1 particles produced in the absence of a SERINC5 antagonist decreased with increasing amounts of virion SERINC5. At low levels of SERINC5 expression, enhancement of virion infectivity by Nef was associated with reduced virion incorporation of SERINC5 and antagonism-defective Nef mutants failed to exclude SERINC5 from virions. However, at elevated levels of SERINC5 expression, Nef maintained infectious HIV particles, despite significant virion incorporation of the restriction factor. These results suggest that in addition to virion exclusion, Nef employs a cryptic mechanism to antagonize virion-associated SERINC5. The involvement of common determinants suggests that the antagonism of Nef to SERINC5 and the downregulation of cell surface CD4 by Nef involve related molecular mechanisms. IMPORTANCE: HIV-1 Nef critically determines virus spread and disease progression in infected individuals by incompletely defined mechanisms. SERINC3 and SERINC5 were recently identified as potent inhibitors of HIV particle infectivity whose antiviral activity is antagonized by HIV-1 Nef. To address the mechanism of SERINC5 antagonism, we identified four molecular determinants of Nef antagonism that are all linked to the mechanism by which Nef downregulates cell surface CD4. Functional characterization of these mutants revealed that endosomal targeting and cell surface downregulation of SERINC5 are dispensable and insufficient for antagonism, respectively. In contrast, virion exclusion and antagonism of SERINC5 were correlated; however, Nef was also able to enhance the infectivity of virions that incorporated robust levels of SERINC5. These results suggest that the antagonism of HIV-1 Nef to SERINC5 restriction of virion infectivity is mediated by a dual mechanism that is related to CD4 downregulation.


Assuntos
HIV-1/fisiologia , HIV-1/patogenicidade , Proteínas de Membrana/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Antígenos CD4/metabolismo , Regulação para Baixo , Genes Virais , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Proteínas de Membrana/genética , Mutação , Vírion/genética , Vírion/fisiologia , Virulência/genética , Virulência/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
10.
Invest. clín ; 57(3): 304-317, Sept. 2016. ilus, graf
Artigo em Inglês | LILACS | ID: biblio-841120

RESUMO

Nef -HIV-1 has been shown to be involved in NADPH complex interaction and superoxide production. The aim of this work was to study the domains involved in the interaction between Nef and p22-phox. Two approaches were used: 1) in silico modelling, to determine the potential binding motifs and design Nef truncated forms and 2) functional assays. The results showed that GFPVT 68-72, FPDW 121-124 and REVLE 179-183 on Nef are critical for p22-phox (RPQIG 142-146 and PGGP 181-184) docking. However, only the region containing FPDW 121-124 on Nef is able to induce superoxide production. Understanding the molecular mechanisms involved in generating oxidative stress during HIV infection, is critical for therapeutic intervention, in order to minimize viral replication and dissemination.


Se ha evidenciado que Nef-VIH-1 está involucrado en la interacción con el complejo NADPH y la producción de superóxido. El objetivo de este trabajo fue identificar los dominios implicados en la interacción entre Nef y p22-phox. Se utilizaron dos estrategias: 1) análisis in silico para determinar los posibles motivos de unión y el diseño Nef formas truncadas y 2) ensayos funcionales. Los resultados mostraron que GFPVT 68-72, FPDW 121 a 124 y 179 a 183 REVLE de Nef son críticos para su unión con p22-phox (RPQIG 142-146 y 181-184 PGGP). Sin embargo, sólo la región que contiene FPDW 121-124 en Nef, es capaz de inducir la producción de superóxido. La comprensión de los mecanismos moleculares implicados en la generación de estrés oxidativo durante la infección por VIH, es crítico para la intervención terapéutica, con el fin de minimizar la replicación y la propagación viral.


Assuntos
Humanos , Espécies Reativas de Oxigênio , NADPH Oxidases/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia
11.
AIDS Rev ; 18(3): 158-165, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27438576

RESUMO

In vitro studies have demonstrated that HIV-1 Nef has several important activities, promoting viral replication and pathogenesis. These activities include downregulation of cell surface molecules CD4 and major histocompatibility complex class I, enhancement of viral infectivity, activation of p21-activated kinase 2, and inhibition of immunoglobulin class switching. But how important each in vitro activity is to in vivo Nef function remains elusive. To address this question, several small animal models have been developed in the past two decades, such as Nef transgenic mice, SCID-hu mice, and humanized mice. Each of those models has its own pros and cons. Easy access and relative inexpensiveness have made small animal models the favorite models for HIV research. This review will be focused on the recent progress in the understanding of the in vivo functions of HIV-1 Nef obtained from studies using these small animal models.


Assuntos
Modelos Animais de Doenças , HIV-1/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Animais , Camundongos , Replicação Viral/fisiologia
12.
Invest Clin ; 57(3): 304-17, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29938962

RESUMO

Nef -HIV-1 has been shown to be involved in NADPH complex interaction and superoxide production. The aim of this work was to study the domains involved in the interaction between Nef and p22-phox. Two approaches were used: 1) in silico modelling, to determine the potential binding motifs and design Nef truncated forms and 2) functional assays. The results showed that GFPVT 68-72, FPDW 121-124 and REVLE 179-183 on Nef are critical for p22-phox (RPQIG 142-146 and PGGP 181-184) docking. However, only the region containing FPDW 121-124 on Nef is able to induce superoxide production. Understanding the molecular mechanisms involved in generating oxidative stress during HIV infection, is critical for therapeutic intervention, in order to minimize viral replication and dissemination.


Assuntos
NADPH Oxidases/fisiologia , Espécies Reativas de Oxigênio , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Humanos
13.
PLoS One ; 10(12): e0145239, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26700863

RESUMO

To find out new determinants required for Nef activity we performed a functional alanine scanning analysis along a discrete but highly conserved region at the core of HIV-1 Nef. We identified the GPG-motif, located at the 121-137 region of HIV-1 NL4.3 Nef, as a novel protein signature strictly required for the p56Lck dependent Nef-induced CD4-downregulation in T-cells. Since the Nef-GPG motif was dispensable for CD4-downregulation in HeLa-CD4 cells, Nef/AP-1 interaction and Nef-dependent effects on Tf-R trafficking, the observed effects on CD4 downregulation cannot be attributed to structure constraints or to alterations on general protein trafficking. Besides, we found that the GPG-motif was also required for Nef-dependent inhibition of ring actin re-organization upon TCR triggering and MHCI downregulation, suggesting that the GPG-motif could actively cooperate with the Nef PxxP motif for these HIV-1 Nef-related effects. Finally, we observed that the Nef-GPG motif was required for optimal infectivity of those viruses produced in T-cells. According to these findings, we propose the conserved GPG-motif in HIV-1 Nef as functional region required for HIV-1 infectivity and therefore with a potential interest for the interference of Nef activity during HIV-1 infection.


Assuntos
Motivos de Aminoácidos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , HIV-1/genética , HIV-1/patogenicidade , Células HeLa , Humanos , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Linfócitos T/virologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
15.
Cell Cycle ; 14(18): 2899-904, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26176554

RESUMO

Viruses often exploit autophagy, a common cellular process of degradation of damaged proteins, organelles, and pathogens, to avoid destruction. HIV-1 dysregulates this process in several cell types by means of Nef protein. Nef is a small HIV-1 protein which is expressed abundantly in astrocytes of HIV-1-infected brains and has been suggested to have a role in the pathogenesis of HIV-Associated Neurocognitive Disorders (HAND). In order to explore its effect in the CNS with respect to autophagy, HIV-1 Nef was expressed in primary human fetal astrocytes (PHFA) using an adenovirus vector (Ad-Nef). We observed that Nef expression triggered the accumulation of autophagy markers, ATG8/LC3 and p62 (SQSMT1). Similar results were obtained with Bafilomycin A1, an autophagy inhibitor which blocks the fusion of autophagosome to lysosome. Furthermore co-expression of tandem LC3 vector (mRFP-EGFP-LC3) and Ad-Nef in these cells produced mainly yellow puncta (mRFP+, EGFP+) strongly suggesting that autophagosome fusion to lysosome is blocked in PHFA cells in the presence of Nef. Together these data indicate that HIV-1 Nef mimics Bafilomycin A1 and blocks the last step of autophagy thereby helping HIV-1 virus to avoid autophagic degradation in human astrocytes.


Assuntos
Astrócitos/patologia , Autofagia , HIV-1/patogenicidade , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Astrócitos/virologia , Família da Proteína 8 Relacionada à Autofagia , Biomarcadores/metabolismo , Humanos , Macrolídeos/farmacologia , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Ligação a RNA/metabolismo
16.
PLoS One ; 10(4): e0125619, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25915798

RESUMO

The Human Immunodeficiency Virus type 1 (HIV-1) accessory protein Nef interacts with a multitude of cellular proteins, manipulating the host membrane trafficking machinery to evade immune surveillance. Nef interactions have been analyzed using various in vitro assays, co-immunoprecipitation studies, and more recently mass spectrometry. However, these methods do not evaluate Nef interactions in the context of viral infection nor do they define the sub-cellular location of these interactions. In this report, we describe a novel bimolecular fluorescence complementation (BiFC) lentiviral expression tool, termed viral BiFC, to study Nef interactions with host cellular proteins in the context of viral infection. Using the F2A cleavage site from the foot and mouth disease virus we generated a viral BiFC expression vector capable of concurrent expression of Nef and host cellular proteins; PACS-1, MHC-I and SNX18. Our studies confirmed the interaction between Nef and PACS-1, a host membrane trafficking protein involved in Nef-mediated immune evasion, and demonstrated co-localization of this complex with LAMP-1 positive endolysosomal vesicles. Furthermore, we utilized viral BiFC to localize the Nef/MHC-I interaction to an AP-1 positive endosomal compartment. Finally, viral BiFC was observed between Nef and the membrane trafficking regulator SNX18. This novel demonstration of an association between Nef and SNX18 was localized to AP-1 positive vesicles. In summary, viral BiFC is a unique tool designed to analyze the interaction between Nef and host cellular proteins by mapping the sub-cellular locations of their interactions during viral infection.


Assuntos
Fluorescência , Vesículas Transportadoras/fisiologia , Integração Viral/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Western Blotting , Citometria de Fluxo , Genes MHC Classe I/fisiologia , Células HEK293 , HIV-1/fisiologia , Células HeLa , Humanos , Células Jurkat , Lentivirus , Transporte Proteico/fisiologia , Nexinas de Classificação/fisiologia , Fator de Transcrição AP-1/fisiologia , Vesículas Transportadoras/virologia , Proteínas de Transporte Vesicular/fisiologia , Replicação Viral/fisiologia
17.
Blood ; 125(10): 1512-3, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25745180

RESUMO

In this issue of Blood, Vérollet et al show that expression of the HIV-1­derived protein Nef alters the migratory mode adopted by macrophages, enhancing macrophage tissue infiltration and explaining the observed accumulation of tissue-resident macrophages in some HIV-infected patients.


Assuntos
HIV-1/patogenicidade , Macrófagos/fisiologia , Macrófagos/virologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Animais , Humanos
18.
Biochim Biophys Acta ; 1850(4): 733-41, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25585010

RESUMO

BACKGROUND: Many viral genomes encode a limited number of proteins, illustrating their innate efficiency in bypassing host immune surveillance. This concept of genomic efficiency is exemplified by the 9 kb RNA genome of human immunodeficiency virus 1 (HIV-1), encoding 15 proteins sub-divided according to function. The enzymatic group includes proteins such as the drug targets reverse transcriptase and protease. In contrast, the accessory proteins lack any known enzymatic or structural function, yet are essential for viral fitness and HIV-1 pathogenesis. Of these, the HIV-1 accessory protein Nef is a master manipulator of host cellular processes, ensuring efficient counterattack against the host immune response, as well as long-term evasion of immune surveillance. In particular, the ability of Nef to downmodulate major histocompatibility complex class I (MHC-I) is a key cellular event that enables HIV-1 to bypass the host's defenses by evading the adaptive immune response. SCOPE OF REVIEW: In this article, we briefly review how various pathogenic viruses control cell-surface MHC-I, and then focus on the mechanisms and implications of HIV-1 Nef-mediated MHC-I downregulation via modulation of the host membrane trafficking machinery. CONCLUSION: The extensive interaction network formed between Nef and numerous membrane trafficking regulators suggests that Nef's role in evading the immune surveillance system intersects multiple host membrane trafficking pathways. SIGNIFICANCE: Nef's ability to evade the immune surveillance system is linked to AIDS pathogenesis. Thus, a complete understanding of the molecular pathways that are subverted by Nef in order to downregulate MHC-I will enhance our understanding of HIV-1's progression to AIDS.


Assuntos
Evasão da Resposta Imune , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Síndrome da Imunodeficiência Adquirida/etiologia , Transporte Biológico , Membrana Celular/metabolismo , Regulação para Baixo , Endocitose , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Transporte Proteico
19.
Blood ; 125(10): 1611-22, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25527710

RESUMO

Macrophages are motile leukocytes, targeted by HIV-1, thought to play a critical role in host dissemination of the virus. However, whether infection impacts their migration capacity remains unknown. We show that 2-dimensional migration and the 3-dimensional (3D) amoeboid migration mode of HIV-1-infected human monocyte-derived macrophages were inhibited, whereas the 3D mesenchymal migration was enhanced. The viral protein Nef was necessary and sufficient for all HIV-1-mediated effects on migration. In Nef transgenic mice, tissue infiltration of macrophages was increased in a tumor model and in several tissues at steady state, suggesting a dominant role for mesenchymal migration in vivo. The mesenchymal motility involves matrix proteolysis and podosomes, cell structures constitutive of monocyte-derived cells. Focusing on the mechanisms used by HIV-1 Nef to control the mesenchymal migration, we show that the stability, size, and proteolytic function of podosomes are increased via the phagocyte-specific kinase Hck and Wiskott-Aldrich syndrome protein (WASP), 2 major regulators of podosomes. In conclusion, HIV-1 reprograms macrophage migration, which likely explains macrophage accumulation in several patient tissues, which is a key step for virus spreading and pathogenesis. Moreover, Nef points out podosomes and the Hck/WASP signaling pathway as good candidates to control tissue infiltration of macrophages, a detrimental phenomenon in several diseases.


Assuntos
HIV-1/patogenicidade , Macrófagos/fisiologia , Macrófagos/virologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Animais , Linhagem Celular Tumoral , Estruturas da Membrana Celular/patologia , Estruturas da Membrana Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Reprogramação Celular/fisiologia , Infecções por HIV/patologia , Infecções por HIV/fisiopatologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-hck/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
20.
Cytokine Growth Factor Rev ; 26(2): 159-73, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25529283

RESUMO

Several viruses manipulate host innate immune responses to avoid immune recognition and improve viral replication and spreading. The viral protein Nef of Human Immunodeficiency Virus is mainly involved in this "hijacking" activity and is a well established virulence factor. In the last few years there have been remarkable advances in outlining a defined framework of its functions. In particular Nef appears to be a shuttling molecular adaptor able to exert its effects both on infected and non infected bystander cell. In addition it is emerging fact that it has an important impact on the chemo-cytokine network. Nef protein represents an interesting new target to develop therapeutic drugs for treatment of seropositive patients. In this review we have tried to provide a unifying view of the multiple functions of this viral protein on the basis of recently available experimental data.


Assuntos
Citocinas/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/fisiologia , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Antígenos CD4/imunologia , Citocinas/metabolismo , HIV-1/imunologia , Humanos , Macrófagos/imunologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/sangue , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...