Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(12): e0261971, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34965271

RESUMO

MicroRNAs (miRNAs) are important molecules that mediate virus-host interactions, mainly by regulating gene expression via gene silencing. Here, we demonstrated that HIV-1 infection upregulated miR-210-5p in HIV-1-inoculated cell lines and in the serum of HIV-1-infected individuals. Luciferase reporter assays and western blotting confirmed that a target protein of miR-210-5p, TGIF2, is regulated by HIV-1 infection. Furthermore, HIV-1 Vpr protein induced miR-210-5p expression. The use of a miR-210-5p inhibitor and TGIF2 overexpression showed that Vpr upregulated miR-210-5p and thereby downregulated TGIF2, which might be one of the mechanisms used by Vpr to induce G2 arrest. Moreover, we identified a transcription factor, NF-κB p50, which upregulated miR-210-5p in response to Vpr protein. In conclusion, we identified a mechanism whereby miR-210-5p, which is induced upon HIV-1 infection, targets TGIF2. This pathway was initiated by Vpr protein activating NF-κB p50, which promoted G2 arrest. These alterations orchestrated by miRNA provide new evidence on how HIV-1 interacts with its host during infection and increase our understanding of the mechanism by which Vpr regulates the cell cycle.


Assuntos
Infecções por HIV , HIV-1/imunologia , Proteínas de Homeodomínio/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , MicroRNAs/imunologia , Proteínas Repressoras/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Linhagem Celular , Pontos de Checagem da Fase G2 do Ciclo Celular , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos
2.
Viruses ; 13(11)2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34835114

RESUMO

Human immunodeficiency virus type 1 (HIV-1) modulates the host cell cycle. The HIV-1 accessory protein Vpr arrests the cell cycle at the G2 phase in dividing cells, and the ability of Vpr to induce G2 arrest is well conserved among primate lentiviruses. Additionally, Vpr-mediated G2 arrest likely correlates with enhanced HIV-1 infection in monocyte-derived macrophages. Here, we screened small-interfering RNA to reveal candidates that suppress Vpr-induced G2 arrest and identified Huntingtin-interacting protein 1 (HIP1) required for efficient G2 arrest. Interestingly, HIP1 was not essential for Vpr-induced DNA double-strand breaks, which are required for activation of the DNA-damage checkpoint and G2 arrest. Furthermore, HIP1 knockdown suppressed HIV-1 infection in monocyte-derived macrophages. This study identifies HIP1 as a factor promoting Vpr-induced G2 arrest and HIV-1 infection in macrophages.


Assuntos
Proteínas de Ligação a DNA/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , RNA Interferente Pequeno , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Pontos de Checagem do Ciclo Celular , Células HEK293 , Células HeLa , Humanos , Macrófagos , Monócitos , Replicação Viral
3.
Elife ; 92020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33300875

RESUMO

HIV-1 must replicate in cells that are equipped to defend themselves from infection through intracellular innate immune systems. HIV-1 evades innate immune sensing through encapsidated DNA synthesis and encodes accessory genes that antagonize specific antiviral effectors. Here, we show that both particle associated, and expressed HIV-1 Vpr, antagonize the stimulatory effect of a variety of pathogen associated molecular patterns by inhibiting IRF3 and NF-κB nuclear transport. Phosphorylation of IRF3 at S396, but not S386, was also inhibited. We propose that, rather than promoting HIV-1 nuclear import, Vpr interacts with karyopherins to disturb their import of IRF3 and NF-κB to promote replication in macrophages. Concordantly, we demonstrate Vpr-dependent rescue of HIV-1 replication in human macrophages from inhibition by cGAMP, the product of activated cGAS. We propose a model that unifies Vpr manipulation of nuclear import and inhibition of innate immune activation to promote HIV-1 replication and transmission.


Assuntos
Infecções por HIV/imunologia , Evasão da Resposta Imune/fisiologia , Imunidade Inata/imunologia , Replicação Viral/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Transporte Ativo do Núcleo Celular/fisiologia , Infecções por HIV/transmissão , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Carioferinas/imunologia , Carioferinas/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , NF-kappa B/imunologia , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
4.
Viruses ; 12(8)2020 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-32726944

RESUMO

HIV infects several cell types in the body, including CD4+ T cells and macrophages. Here we review the role of macrophages in HIV infection and describe complex interactions between viral proteins and host defenses in these cells. Macrophages exist in many forms throughout the body, where they play numerous roles in healthy and diseased states. They express pattern-recognition receptors (PRRs) that bind viral, bacterial, fungal, and parasitic pathogens, making them both a key player in innate immunity and a potential target of infection by pathogens, including HIV. Among these PRRs is mannose receptor, a macrophage-specific protein that binds oligosaccharides, restricts HIV replication, and is downregulated by the HIV accessory protein Vpr. Vpr significantly enhances infection in vivo, but the mechanism by which this occurs is controversial. It is well established that Vpr alters the expression of numerous host proteins by using its co-factor DCAF1, a component of the DCAF1-DDB1-CUL4 ubiquitin ligase complex. The host proteins targeted by Vpr and their role in viral replication are described in detail. We also discuss the structure and function of the viral protein Env, which is stabilized by Vpr in macrophages. Overall, this literature review provides an updated understanding of the contributions of macrophages and Vpr to HIV pathogenesis.


Assuntos
Infecções por HIV/imunologia , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/virologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Animais , Infecções por HIV/patologia , HIV-1/imunologia , Humanos , Macrófagos/imunologia , Camundongos , Replicação Viral
5.
Biol Cell ; 111(12): 308-318, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31628772

RESUMO

BACKGROUND INFORMATION: Autophagy is induced during HIV-1 entry into CD4 T cells by the fusion of the membranes triggered by the gp41 envelope glycoprotein. This anti-HIV-1 mechanism is inhibited by the viral infectivity factor (Vif) neosynthesized after HIV-1 integration to allow viral replication. However, autophagy is very rapidly controlled after HIV-1 entry by a still unknown mechanism. As HIV-1 viral protein R (Vpr) is the only auxiliary protein found within the virion in substantial amount, we studied its capability to control the early steps of HIV-1 envelope-mediated autophagy. RESULTS: We demonstrated that ectopic Vpr inhibits autophagy in both the Jurkat CD4 T cell line and HEK.293T cells. Interestingly, Vpr coming from the virus also blocks autophagy in CD4 T cells, the main cell target of HIV-1. Furthermore, Vpr decreases the expression level of two essential autophagy proteins (ATG), LC3B and Beclin-1, and an important autophagy-related protein, BNIP3 as well as the level of their mRNA. We also demonstrated in HEK.293T cells that Vpr degrades the FOXO3a transcription factor through the ubiquitin proteasome system. CONCLUSION: Vpr, the only well-expressed HIV-1 auxiliary protein incorporated into viruses, is able to negatively control autophagy induced during HIV-1 entry into CD4 T cells. SIGNIFICANCE: We provide insights of how HIV-1 controls autophagy very early after its entry into CD4 T cells and discovered a new function of Vpr. These results open the route to a better understanding of the roles of Vpr during HIV-1 infection through FOXO3a degradation and could be important to consider additional therapies that counteract the role of Vpr on autophagy.


Assuntos
Autofagia/imunologia , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , HIV-1 , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Proteína Beclina-1/imunologia , Linfócitos T CD4-Positivos/citologia , Células HEK293 , HIV-1/imunologia , HIV-1/fisiologia , Humanos , Células Jurkat , Proteínas de Membrana/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proteínas Supressoras de Tumor/imunologia , Replicação Viral
6.
Nat Microbiol ; 4(12): 2552-2564, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31659299

RESUMO

Innate immunity is the first line of host defence against pathogens. Suppression of innate immune responses is essential for the survival of all viruses. However, the interplay between innate immunity and HIV/SIV is only poorly characterized. We have discovered Vpx as a novel inhibitor of innate immune activation that associates with STING signalosomes and interferes with the nuclear translocation of NF-κB and the induction of innate immune genes. This new function of Vpx could be separated from its role in mediating degradation of the antiviral factor SAMHD1, and is conserved among diverse HIV-2/SIV Vpx. Vpx selectively suppressed cGAS-STING-mediated nuclear factor-κB signalling. Furthermore, Vpx and Vpr had complementary activities against cGAS-STING activity. Since SIVMAC lacking both Vpx and Vpr was less pathogenic than SIV deficient for Vpr or Vpx alone, suppression of innate immunity by HIV/SIV is probably a key pathogenic determinant, making it a promising target for intervention.


Assuntos
HIV-2/metabolismo , Proteínas de Membrana/metabolismo , NF-kappa B/metabolismo , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Vírus da Imunodeficiência Símia/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Feminino , Células HEK293 , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-2/genética , Células HeLa , Humanos , Imunidade Inata/genética , Masculino , Camundongos Endogâmicos BALB C , Proteína 1 com Domínio SAM e Domínio HD/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/imunologia , Adulto Jovem , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
7.
J Neuroimmune Pharmacol ; 12(2): 233-248, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27726055

RESUMO

Human Immunodeficiency virus (HIV) enters the brain soon after seroconversion and induces chronic neuroinflammation by infecting and activating brain macrophages. Inflammasomes are cytosolic protein complexes that mediate caspase-1 activation and ensuing cleavage and release of IL-1ß and -18 by macrophages. Our group recently showed that HIV-1 infection of human microglia induced inflammasome activation in NLRP3-dependent manner. The HIV-1 viral protein R (Vpr) is an accessory protein that is released from HIV-infected cells, although its effects on neuroinflammation are undefined. Infection of human microglia with Vpr-deficient HIV-1 resulted in reduced caspase-1 activation and IL-1ß production, compared to cells infected with a Vpr-encoding HIV-1 virus. Vpr was detected at low nanomolar concentrations in cerebrospinal fluid from HIV-infected patients and in supernatants from HIV-infected primary human microglia. Exposure of human macrophages to Vpr caused caspase-1 cleavage and IL-1ß release with reduced cell viability, which was dependent on NLRP3 expression. Increased NLRP3, caspase-1, and IL-1ß expression was evident in HIV-1 Vpr transgenic mice compared to wild-type littermates, following systemic immune stimulation. Treatment with the caspase-1 inhibitor, VX-765, suppressed NLRP3 expression with reduced IL-1ß expression and associated neuroinflammation. Neurobehavioral deficits showed improvement in Vpr transgenic animals treated with VX-765. Thus, Vpr-induced NLRP3 inflammasome activation, which contributed to neuroinflammation and was abrogated by caspase-1 inhibition. This study provides a new therapeutic perspective for HIV-associated neuropsychiatric disease.


Assuntos
HIV-1/metabolismo , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Adulto , Idoso de 80 Anos ou mais , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Feto , HIV-1/imunologia , Humanos , Inflamassomos/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Pessoa de Meia-Idade , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
8.
Cell Rep ; 17(2): 413-424, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27705790

RESUMO

Several pattern-recognition receptors sense HIV-1 replication products and induce type I interferon (IFN-I) production under specific experimental conditions. However, it is thought that viral sensing and IFN induction are virtually absent in the main target cells of HIV-1 in vivo. Here, we show that activated CD4+ T cells sense HIV-1 infection through the cytosolic DNA sensor cGAS and mount a bioactive IFN-I response. Efficient induction of IFN-I by HIV-1 infection requires proviral integration and is regulated by newly expressed viral accessory proteins: Vpr potentiates, while Vpu suppresses cGAS-dependent IFN-I induction. Furthermore, Vpr also amplifies innate sensing of HIV-1 infection in Vpx-treated dendritic cells. Our results identify cGAS as mediator of an IFN-I response to HIV-1 infection in CD4+ T cells and demonstrate that this response is modulated by the viral accessory proteins Vpr and Vpu. Thus, viral innate immune evasion is incomplete in the main target cells of HIV-1.


Assuntos
Infecções por HIV/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Interferon Tipo I/imunologia , Nucleotidiltransferases/genética , Proteínas Virais Reguladoras e Acessórias/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Células HEK293 , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/patogenicidade , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Imunidade Inata/genética , Interferon Tipo I/genética , Lentivirus/genética , Nucleotidiltransferases/imunologia , Proteínas Virais Reguladoras e Acessórias/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
9.
J Neurovirol ; 22(4): 403-15, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27056720

RESUMO

It is increasingly evident that the human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) has a unique role in neuropathogenesis. Its ability to induce G2/M arrest coupled with its capacity to increase viral gene transcription gives it a unique role in sustaining viral replication and aiding in the establishment and maintenance of a systemic infection. The requirement of Vpr for HIV-1 infection and replication in cells of monocytic origin (a key lineage of cells involved in HIV-1 neuroinvasion) suggests an important role in establishing and sustaining infection in the central nervous system (CNS). Contributions of Vpr to neuropathogenesis can be expanded further through (i) naturally occurring HIV-1 sequence variation that results in functionally divergent Vpr variants; (ii) the dual activities of Vpr as a intracellular protein delivered and expressed during HIV-1 infection and as an extracellular protein that can act on neighboring, uninfected cells; (iii) cell type-dependent consequences of Vpr expression and exposure, including cell cycle arrest, metabolic dysregulation, and cytotoxicity; and (iv) the effects of Vpr on exosome-based intercellular communication in the CNS. Revealing that the effects of this pleiotropic viral protein is an essential part of a greater understanding of HIV-1-associated pathogenesis and potential approaches to treating and preventing disease caused by HIV-1 infection.


Assuntos
Sistema Nervoso Central/virologia , Regulação Viral da Expressão Gênica , Infecções por HIV/virologia , HIV-1/genética , Interações Hospedeiro-Patógeno/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Efeito Espectador/genética , Efeito Espectador/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/imunologia , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/patologia , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Humanos , Monócitos/imunologia , Monócitos/patologia , Monócitos/virologia , Neurônios/imunologia , Neurônios/patologia , Neurônios/virologia , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/virologia , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
10.
J Gen Virol ; 97(4): 813-824, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26764023

RESUMO

Human immunodeficiency virus (HIV) is the causative agent of the deadly disease AIDS, which is characterized by the progressive decline of CD4(+)T-cells. HIV-1-encoded proteins such as envelope gp120 (glycoprotein gp120), Tat (trans-activator of transcription), Nef (negative regulatory factor), Vpr (viral protein R), Vpu (viral protein unique) and protease are known to be effective in modulating host cell signalling pathways that lead to an alteration in apoptosis of both HIV-infected and uninfected bystander cells. Depending on the stage of the virus life cycle and host cell type, these viral proteins act as mediators of pro- or anti-apoptotic signals. HIV latency in viral reservoirs is a persistent phenomenon that has remained beyond the control of the human immune system. To cure HIV infections completely, it is crucial to reactivate latent HIV from cellular pools and to drive these apoptosis-resistant cells towards death. Several previous studies have reported the role of HIV-encoded proteins in apoptosis modulation, but the molecular basis for apoptosis evasion of some chronically HIV-infected cells and reactivated latently HIV-infected cells still needs to be elucidated. The current review summarizes our present understanding of apoptosis modulation in HIV-infected cells, uninfected bystander cells and latently infected cells, with a focus on highlighting strategies to activate the apoptotic pathway to kill latently infected cells.


Assuntos
Regulação da Expressão Gênica/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Interações Hospedeiro-Patógeno , Latência Viral/imunologia , Apoptose/genética , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/patologia , Infecções por HIV/virologia , Protease de HIV/genética , Protease de HIV/imunologia , HIV-1/genética , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Transdução de Sinais , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
11.
J Virol ; 89(23): 12118-30, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26401039

RESUMO

UNLABELLED: The HIV-1 accessory protein Vpr displays different activities potentially impacting viral replication, including the arrest of the cell cycle in the G2 phase and the stimulation of apoptosis and DNA damage response pathways. Vpr also modulates cytokine production by infected cells, but this property remains partly characterized. Here, we investigated the effect of Vpr on the production of the proinflammatory cytokine tumor necrosis factor (TNF). We report that Vpr significantly increases TNF secretion by infected lymphocytes. De novo production of Vpr is required for this effect. Vpr mutants known to be defective for G2 cell cycle arrest induce lower levels of TNF secretion, suggesting a link between these two functions. Silencing experiments and the use of chemical inhibitors further implicated the cellular proteins DDB1 and TAK1 in this activity of Vpr. TNF secreted by HIV-1-infected cells triggers NF-κB activity in bystander cells and allows viral reactivation in a model of latently infected cells. Thus, the stimulation of the proinflammatory pathway by Vpr may impact HIV-1 replication in vivo. IMPORTANCE: The role of the HIV-1 accessory protein Vpr remains only partially characterized. This protein is important for viral pathogenesis in infected individuals but is dispensable for viral replication in most cell culture systems. Some of the functions described for Vpr remain controversial. In particular, it remains unclear whether Vpr promotes or instead prevents proinflammatory and antiviral immune responses. In this report, we show that Vpr promotes the release of TNF, a proinflammatory cytokine associated with rapid disease progression. Using Vpr mutants or inhibiting selected cellular genes, we show that the cellular proteins DDB1 and TAK1 are involved in the release of TNF by HIV-infected cells. This report provides novel insights into how Vpr manipulates TNF production and helps clarify the role of Vpr in innate immune responses and inflammation.


Assuntos
Regulação da Expressão Gênica/imunologia , HIV-1/imunologia , Linfócitos T/virologia , Fator de Necrose Tumoral alfa/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Humanos , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
J Virol ; 89(20): 10656-67, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26269184

RESUMO

UNLABELLED: Expression of the cytoprotective enzyme heme oxygenase-1 (HO-1) is significantly reduced in the brain prefrontal cortex of HIV-positive individuals with HIV-associated neurocognitive disorders (HAND). Furthermore, this HO-1 deficiency correlates with brain viral load, markers of macrophage activation, and type I interferon responses. In vitro, HIV replication in monocyte-derived macrophages (MDM) selectively reduces HO-1 protein and RNA expression and induces production of neurotoxic levels of glutamate; correction of this HO-1 deficiency reduces neurotoxic glutamate production without an effect on HIV replication. We now demonstrate that macrophage HO-1 deficiency, and the associated neurotoxin production, is a conserved feature of infection with macrophage-tropic HIV-1 strains that correlates closely with the extent of replication, and this feature extends to HIV-2 infection. We further demonstrate that this HO-1 deficiency does not depend specifically upon the HIV-1 accessory genes nef, vpr, or vpu but rather on HIV replication, even when markedly limited. Finally, antiretroviral therapy (ART) applied to MDM after HIV infection is established does not prevent HO-1 loss or the associated neurotoxin production. This work defines a predictable relationship between HIV replication, HO-1 loss, and neurotoxin production in MDM that likely reflects processes in place in the HIV-infected brains of individuals receiving ART. It further suggests that correcting this HO-1 deficiency in HIV-infected MDM could provide neuroprotection above that provided by current ART or proposed antiviral therapies directed at limiting Nef, Vpr, or Vpu functions. The ability of HIV-2 to reduce HO-1 expression suggests that this is a conserved phenotype among macrophage-tropic human immunodeficiency viruses that could contribute to neuropathogenesis. IMPORTANCE: The continued prevalence of HIV-associated neurocognitive disorders (HAND) underscores the need for adjunctive therapy that targets the neuropathological processes that persist in antiretroviral therapy (ART)-treated HIV-infected individuals. To this end, we previously identified one such possible process, a deficiency of the antioxidative and anti-inflammatory enzyme heme oxygenase-1 (HO-1) in the brains of individuals with HAND. In the present study, our findings suggest that the HO-1 deficiency associated with excess glutamate production and neurotoxicity in HIV-infected macrophages is a highly conserved phenotype of macrophage-tropic HIV strains and that this phenotype can persist in the macrophage compartment in the presence of ART. This suggests a plausible mechanism by which HIV infection of brain macrophages in ART-treated individuals could exacerbate oxidative stress and glutamate-induced neuronal injury, each of which is associated with neurocognitive dysfunction in infected individuals. Thus, therapies that rescue the HO-1 deficiency in HIV-infected individuals could provide additional neuroprotection to ART.


Assuntos
Anemia Hemolítica/virologia , Ácido Glutâmico/toxicidade , Transtornos do Crescimento/virologia , HIV-1/patogenicidade , HIV-2/patogenicidade , Heme Oxigenase-1/deficiência , Distúrbios do Metabolismo do Ferro/virologia , Macrófagos/virologia , Anemia Hemolítica/genética , Anemia Hemolítica/imunologia , Animais , Fármacos Anti-HIV/farmacologia , Farmacorresistência Viral/genética , Farmacorresistência Viral/imunologia , Expressão Gênica , Ácido Glutâmico/biossíntese , Transtornos do Crescimento/genética , Transtornos do Crescimento/imunologia , HIV-1/imunologia , HIV-2/imunologia , Heme Oxigenase-1/genética , Heme Oxigenase-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Neuroglia/efeitos dos fármacos , Neuroglia/imunologia , Neuroglia/virologia , Fenótipo , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/imunologia , Replicação Viral/efeitos dos fármacos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
13.
PLoS Pathog ; 11(7): e1005054, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26186441

RESUMO

Vpr is a conserved primate lentiviral protein that promotes infection of T lymphocytes in vivo by an unknown mechanism. Here we demonstrate that Vpr and its cellular co-factor, DCAF1, are necessary for efficient cell-to-cell spread of HIV-1 from macrophages to CD4+ T lymphocytes when there is inadequate cell-free virus to support direct T lymphocyte infection. Remarkably, Vpr functioned to counteract a macrophage-specific intrinsic antiviral pathway that targeted Env-containing virions to LAMP1+ lysosomal compartments. This restriction of Env also impaired virological synapses formed through interactions between HIV-1 Env on infected macrophages and CD4 on T lymphocytes. Treatment of infected macrophages with exogenous interferon-alpha induced virion degradation and blocked synapse formation, overcoming the effects of Vpr. These results provide a mechanism that helps explain the in vivo requirement for Vpr and suggests that a macrophage-dependent stage of HIV-1 infection drives the evolutionary conservation of Vpr.


Assuntos
Linfócitos T CD4-Positivos/virologia , HIV-1/imunologia , Macrófagos/virologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Linfócitos T CD4-Positivos/imunologia , Humanos , Interferon-alfa/metabolismo , Macrófagos/metabolismo , Vírion/metabolismo
14.
Cell Host Microbe ; 16(6): 722-35, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25464830

RESUMO

The HIV-1 accessory protein Vpr enhances infection of primary macrophages through unknown mechanisms. Recent studies demonstrated that Vpr interactions with the cellular DCAF1-DDB1-CUL4 E3 ubiquitin ligase complex limit activation of innate immunity and interferon (IFN) induction. We describe a restriction mechanism that targets the HIV-1 envelope protein Env, but is overcome by Vpr and its interaction with DCAF1. This restriction is active in the absence of Vpr in HIV-1-infected primary macrophages and macrophage-epithelial cell heterokaryons, but not epithelial cell lines. HIV-1-infected macrophages lacking Vpr express more IFN following infection, target Env for lysosomal degradation, and produce fewer Env-containing virions. Conversely, Vpr expression reduces IFN induction, rescues Env expression, and enhances virion release. Addition of IFN or silencing DCAF1 reduces the amount of cell-associated Env and virion production in wild-type HIV-1-infected primary macrophages. These findings provide insight into an IFN-stimulated macrophage-specific restriction pathway targeting HIV-1 Env that is counteracted by Vpr.


Assuntos
Infecções por HIV/virologia , HIV-1/imunologia , Macrófagos/virologia , Vírion/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/genética , HIV-1/fisiologia , Células HeLa , Humanos , Interferons/imunologia , Macrófagos/imunologia , Proteínas Serina-Treonina Quinases , Ubiquitina-Proteína Ligases , Vírion/genética , Vírion/fisiologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
15.
PLoS One ; 9(12): e113729, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25436999

RESUMO

The targeting of HIV-1 using antibodies is of high interest as molecular tools to better understand the biology of the virus or as a first step toward the design of new inhibitors targeting critical viral intracellular proteins. Small and highly stable llama-derived single-domain antibodies can often be functionally expressed as intracellular antibodies in the cytoplasm of eukaryotic cells. Using a selection method based on the Sos Recruitment System, a cytoplasmic yeast two-hybrid approach, we have isolated single-domain antibodies able to bind HIV-1 Vpr and Capside proteins in the yeast cytoplasm. One anti-Vpr single domain antibody was able to bind the HIV-1 regulatory Vpr protein in the cytoplasm of eukaryotic cells, leading to its delocalization from the nucleus to the cytoplasm. To our knowledge, this is the first description of a functional single-domain intrabody targeting HIV-1 Vpr, isolated using an in vivo cytoplasmic selection method that alleviates some limitations of the conventional yeast two-hybrid system.


Assuntos
Citoplasma/metabolismo , Anticorpos de Domínio Único/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Núcleo Celular/metabolismo , HIV-1/metabolismo , Células HeLa , Humanos , Ligação Proteica , Saccharomyces cerevisiae/metabolismo , Transfecção
16.
Virology ; 439(1): 47-56, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23453579

RESUMO

The human immunodeficiency virus type I (HIV-1) Vpr plays an essential role in viral replication. A number of studies have reported that Vpr modulates the nuclear factor-κB (NF-κB) pathway. Yet, the reported effects of Vpr on NF-κB signaling are controversial. In this study, we investigate the interplay between Vpr and NF-κB pathway. We discover that HIV-1 infection elevates the phosphorylation of IκBα and p100, and that this increase is greatly reduced when a Vpr-negative HIV-1 is used for infection. Our data further show that Vpr regulates the activity of IKKα/ß through interactions. In addition, Vpr modulates the phosphorylation of p65 and p100, suggesting that Vpr activates both canonical and noncanonical NF-κB pathway. Knock down of endogenous IKKα/ß result in a decrease in Vpr-mediated NF-κB and HIV-1 LTR activation. Given that Vpr is present in HIV-1 particles, our data suggest that Vpr activates the NF-κB pathway immediately after HIV-1 entry.


Assuntos
HIV-1/fisiologia , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Processamento de Proteína Pós-Traducional , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Endonucleases , HIV-1/imunologia , Humanos , Quinase I-kappa B/imunologia , NF-kappa B/imunologia , Proteínas Nucleares/metabolismo , Fosforilação , Mapeamento de Interação de Proteínas , Transdução de Sinais , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
17.
Bing Du Xue Bao ; 28(2): 151-7, 2012 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-22519177

RESUMO

To develop polyclonal antibodies against predicted B cell epitopes in HIV-1 accessory protein Vpr, the prepared consensus Vpr amino acid sequence was used to predict potential B cell epitopes by online softwares (ABCpred and Bcepred), the synthesized polypeptides of B-cell epitopes were subsequently conjugated with keyhole limpet hemocyanin (KLH) and then used to immunize rabbits. The antibody titers were determined by ELISA, and antibody specifity was analyzed by Western-Blotting and immunoprecipitation, respectively. Amino acid residues 3-19 (N) and 82-95 (C) of Vpr were predicted as the potential B cell epitopes. After inoculation of the conjugation of synthesized peptide to KLH, the antibody titers in rabbit sera against N and C peptides reached more than 1:100000 by ELISA. Western-Blotting analysis showed that the polyclonal antibodies reacted with both wild Vpr and fusion protein of GFP with Vpr, no matter Vpr was derived from HIV-1 B subtype or CRF07_BC recombinant form; Immunoprecipitation analysis showed similar reactions to Western-Blotting results. Two B cell epitopes of Vpr were successfully predicted by Bio-informatics methods and polyclonal antibodies against those peptides could be successfully prepared.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Peptídeos/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Infecções por HIV/sangue , Infecções por HIV/virologia , HIV-1/genética , Humanos , Peptídeos/genética , Coelhos , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
18.
J Immunol ; 187(11): 5865-78, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22068233

RESUMO

Monocytic cells survive HIV replication and consequent cytopathic effects because of their decreased sensitivity to HIV-induced apoptosis. However, the mechanism underlying this resistance to apoptosis remains poorly understood. Lymphocytic cells are exposed to microbial products because of their translocation from the gut in persons with chronic HIV infections or following coinfections. We hypothesized that activation of monocytic cells by such microbial products through interaction with corresponding TLRs may confer antiapoptotic signals. Using HIV-viral protein R (Vpr)(52-96) peptide as a model apoptosis-inducing agent, we demonstrated that unlike monocyte-derived macrophages, undifferentiated primary human monocytes and promonocytic THP-1 cells are highly susceptible to Vpr(52-96)-induced apoptosis. Interestingly, monocytes and THP-1 cells stimulated with TLR9 agonist CpG induced almost complete resistance to Vpr(52-96)-induced apoptosis, albeit through a TLR9-independent signaling pathway. Moreover, CpG selectively induced the antiapoptotic cellular inhibitor of apoptosis (c-IAP)-2 protein and inhibition of the c-IAP-2 gene by either specific small interfering RNA or synthetic second mitochondrial activator of caspases mimetic reversed CpG-induced resistance against Vpr(52-96)-mediated apoptosis. We demonstrated that c-IAP-2 is regulated by the JNK and calcium signaling pathway, in particular calmodulin-dependent protein kinase-II. Furthermore, inhibition of JNK and the calcium signaling including the calmodulin-dependent protein kinase-II by either pharmacological inhibitors or their specific small interfering RNAs reversed CpG-induced protection against Vpr(52-96)-mediated apoptosis. We also show that CpG induced JNK phosphorylation through activation of the calcium signaling pathway. Taken together, our results suggest that CpG-induced protection may be mediated by c-IAP-2 through the calcium-activated JNK pathway via what appeared to be TLR9-independent signaling pathways.


Assuntos
Proteínas Inibidoras de Apoptose/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , Monócitos/virologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Apoptose/imunologia , Western Blotting , Linhagem Celular , Separação Celular , Citometria de Fluxo , Humanos , Microscopia Confocal , Monócitos/metabolismo , Oligonucleotídeos , Transdução de Sinais , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/metabolismo , Transfecção , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
19.
Retrovirology ; 8: 44, 2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21645334

RESUMO

BACKGROUND: Viral diversity and abundance are defining properties of human immunodeficiency virus (HIV)-1's biology and pathogenicity. Despite the increasing availability of antiretroviral therapy, HIV-associated dementia (HAD) continues to be a devastating consequence of HIV-1 infection of the brain although the underlying disease mechanisms remain uncertain. Herein, molecular diversity within the HIV-1 non-structural gene, Vpr, was examined in RNA sequences derived from brain and blood of HIV/AIDS patients with or without HIV-associated dementia (HAD) together with the ensuing pathobiological effects. RESULTS: Cloned brain- and blood-derived full length vpr alleles revealed that amino acid residue 77 within the brain-derived alleles distinguished HAD (77Q) from non-demented (ND) HIV/AIDS patients (77R) (p < 0.05) although vpr transcripts were more frequently detected in HAD brains (p < 0.05). Full length HIV-1 clones encoding the 77R-ND residue induced higher IFN-α, MX1 and BST-2 transcript levels in human glia relative to the 77Q-HAD encoding virus (p < 0.05) but both viruses exhibited similar levels of gene expression and replication. Myeloid cells transfected with 77Q-(pVpr77Q-HAD), 77R (pVpr77R-ND) or Vpr null (pVpr(-))-containing vectors showed that the pVpr77R-ND vector induced higher levels of immune gene expression (p < 0.05) and increased neurotoxicity (p < 0.05). Vpr peptides (amino acids 70-96) containing the 77Q-HAD or 77R-ND motifs induced similar levels of cytosolic calcium activation when exposed to human neurons. Human glia exposed to the 77R-ND peptide activated higher transcript levels of IFN-α, MX1, PRKRA and BST-2 relative to 77Q-HAD peptide (p < 0.05). The Vpr 77R-ND peptide was also more neurotoxic in a concentration-dependent manner when exposed to human neurons (p < 0.05). Stereotaxic implantation of full length Vpr, 77Q-HAD or 77R-ND peptides into the basal ganglia of mice revealed that full length Vpr and the 77R-ND peptide caused greater neurobehavioral deficits and neuronal injury compared with 77Q-HAD peptide-implanted animals (p < 0.05). CONCLUSIONS: These observations underscored the potent neuropathogenic properties of Vpr but also indicated viral diversity modulates innate neuroimmunity and neurodegeneration.


Assuntos
Encéfalo/virologia , HIV-1/imunologia , HIV-1/patogenicidade , Polimorfismo Genético , Tropismo Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/biossíntese , Sangue/virologia , Proteínas Ligadas por GPI/biossíntese , Proteínas de Ligação ao GTP/biossíntese , Perfilação da Expressão Gênica , HIV-1/genética , Humanos , Imunidade Inata , Interferon-alfa/biossíntese , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas de Resistência a Myxovirus , Neuroglia/imunologia , Neuroglia/virologia , RNA Viral/genética , Análise de Sequência de DNA , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
20.
FASEB J ; 25(7): 2211-20, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21427211

RESUMO

Immunosuppressive lentivirus infections, including human, simian, and feline immunodeficiency viruses (HIV, SIV, and FIV, respectively), cause the acquired immunodeficiency syndrome (AIDS), frequently associated with AIDS enteropathy. Herein, we investigated the extent to which lentivirus infections affected mucosal integrity and intestinal permeability in conjunction with immune responses and activation of endoplasmic reticulum (ER) stress pathways. Duodenal biopsies from individuals with HIV/AIDS exhibited induction of IL-1ß, CD3ε, HLA-DRA, spliced XBP-1(Xbp-1s), and CHOP expression compared to uninfected persons (P<0.05). Gut epithelial cells exposed to HIV-1 Vpr demonstrated elevated TNF-α, IL-1ß, spliced Xbp-1s, and CHOP expression (P<0.05) together with calcium activation and disruption of epithelial cell monolayer permeability. In addition to reduced blood CD4(+) T lymphocyte levels, viral loads in the gut and plasma were high in FIV-infected animals (P<0.05). FIV-infected animals also exhibited a failure to gain weight and increased lactulose/mannitol ratios compared with uninfected animals (P<0.05). Proinflammatory and ER stress gene expression were activated in the ileum of FIV-infected animals (P<0.05), accompanied by intestinal epithelial damage with loss of epithelial cells and leukocyte infiltration of the lamina propria. Lentivirus infections cause gut inflammation and ensuing damage to intestinal epithelial cells, likely through induction of ER stress pathways, resulting in disruption of gut functional integrity.


Assuntos
Retículo Endoplasmático/metabolismo , Enterite/genética , Células Epiteliais/metabolismo , Enteropatia por HIV/genética , Animais , Complexo CD3/genética , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Gatos , Linhagem Celular Tumoral , Duodeno/imunologia , Duodeno/metabolismo , Duodeno/virologia , Retículo Endoplasmático/imunologia , Enterite/imunologia , Enterite/virologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Expressão Gênica , Enteropatia por HIV/imunologia , Enteropatia por HIV/virologia , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/fisiologia , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Cadeias alfa de HLA-DR , Interações Hospedeiro-Patógeno , Humanos , Vírus da Imunodeficiência Felina/imunologia , Vírus da Imunodeficiência Felina/fisiologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Infecções por Lentivirus/genética , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/virologia , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Viral/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...