Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Kidney Int ; 103(3): 529-543, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36565808

RESUMEN

Chronic kidney disease (CKD) is a common cause of morbidity in human immunodeficiency virus (HIV)-positive individuals. HIV infection leads to a wide spectrum of kidney cell damage, including tubular epithelial cell (TEC) injury. Among the HIV-1 proteins, the pathologic effects of viral protein R (Vpr) are well established and include DNA damage response, cell cycle arrest, and cell death. Several in vitro studies have unraveled the molecular pathways driving the cytopathic effects of Vpr in tubular epithelial cells. However, the in vivo effects of Vpr on tubular injury and CKD pathogenesis have not been thoroughly investigated. Here, we use a novel inducible tubular epithelial cell-specific Vpr transgenic mouse model to show that Vpr expression leads to progressive tubulointerstitial damage, interstitial inflammation and fibrosis, and tubular cyst development. Importantly, Vpr-expressing tubular epithelial cells displayed significant hypertrophy, aberrant cell division, and atrophy; all reminiscent of tubular injuries observed in human HIV-associated nephropathy (HIVAN). Single-cell RNA sequencing analysis revealed the Vpr-mediated transcriptomic responses in specific tubular subsets and highlighted the potential multifaceted role of p53 in the regulation of cell metabolism, proliferation, and death pathways in Vpr-expressing tubular epithelial cells. Thus, our study demonstrates that HIV Vpr expression in tubular cells is sufficient to induce HIVAN-like tubulointerstitial damage and fibrosis, independent of glomerulosclerosis and proteinuria. Additionally, as this new mouse model develops progressive CKD with diffuse fibrosis and kidney failure, it can serve as a useful tool to examine the mechanisms of kidney disease progression and fibrosis in vivo.


Asunto(s)
Nefropatía Asociada a SIDA , Productos del Gen vpr , Infecciones por VIH , VIH-1 , Insuficiencia Renal Crónica , Animales , Humanos , Ratones , Nefropatía Asociada a SIDA/genética , Modelos Animales de Enfermedad , Productos del Gen vpr/genética , Productos del Gen vpr/metabolismo , Productos del Gen vpr/farmacología , Infecciones por VIH/complicaciones , VIH-1/genética , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana , Ratones Transgénicos , Insuficiencia Renal Crónica/complicaciones
2.
PLoS Pathog ; 17(8): e1009775, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34339457

RESUMEN

Viruses have evolved means to manipulate the host's ubiquitin-proteasome system, in order to down-regulate antiviral host factors. The Vpx/Vpr family of lentiviral accessory proteins usurp the substrate receptor DCAF1 of host Cullin4-RING ligases (CRL4), a family of modular ubiquitin ligases involved in DNA replication, DNA repair and cell cycle regulation. CRL4DCAF1 specificity modulation by Vpx and Vpr from certain simian immunodeficiency viruses (SIV) leads to recruitment, poly-ubiquitylation and subsequent proteasomal degradation of the host restriction factor SAMHD1, resulting in enhanced virus replication in differentiated cells. To unravel the mechanism of SIV Vpr-induced SAMHD1 ubiquitylation, we conducted integrative biochemical and structural analyses of the Vpr protein from SIVs infecting Cercopithecus cephus (SIVmus). X-ray crystallography reveals commonalities between SIVmus Vpr and other members of the Vpx/Vpr family with regard to DCAF1 interaction, while cryo-electron microscopy and cross-linking mass spectrometry highlight a divergent molecular mechanism of SAMHD1 recruitment. In addition, these studies demonstrate how SIVmus Vpr exploits the dynamic architecture of the multi-subunit CRL4DCAF1 assembly to optimise SAMHD1 ubiquitylation. Together, the present work provides detailed molecular insight into variability and species-specificity of the evolutionary arms race between host SAMHD1 restriction and lentiviral counteraction through Vpx/Vpr proteins.


Asunto(s)
Proteínas Cullin/química , Productos del Gen vpr/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Proteína 1 que Contiene Dominios SAM y HD/química , Ubiquitinación , Replicación Viral , Secuencia de Aminoácidos , Animales , Microscopía por Crioelectrón , Proteínas Cullin/metabolismo , Productos del Gen vpr/genética , Proteína NEDD8/química , Proteína NEDD8/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
3.
Viruses ; 12(1)2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31952107

RESUMEN

Viral protein R (Vpr) is an accessory protein found in various primate lentiviruses, including human immunodeficiency viruses type 1 and 2 (HIV-1 and HIV-2) as well as simian immunodeficiency viruses (SIVs). Vpr modulates many processes during viral lifecycle via interaction with several of cellular targets. Previous studies showed that HIV-1 Vpr strengthened degradation of Mini-chromosome Maintenance Protein10 (MCM10) by manipulating DCAF1-Cul4-E3 ligase in proteasome-dependent pathway. However, whether Vpr from other primate lentiviruses are also associated with MCM10 degradation and the ensuing impact remain unknown. Based on phylogenetic analyses, a panel of primate lentiviruses Vpr/x covering main virus lineages was prepared. Distinct MCM10 degradation profiles were mapped and HIV-1, SIVmus and SIVrcm Vprs induced MCM10 degradation in proteasome-dependent pathway. Colocalization and interaction between MCM10 with these Vprs were also observed. Moreover, MCM10 2-7 interaction region was identified as a determinant region susceptible to degradation. However, MCM10 degradation did not alleviate DNA damage response induced by these Vpr proteins. MCM10 degradation by HIV-1 Vpr proteins was correlated with G2/M arrest, while induction of apoptosis and oligomerization formation of Vpr failed to alter MCM10 proteolysis. The current study demonstrated a distinct interplay pattern between primate lentiviruses Vpr proteins and MCM10.


Asunto(s)
Productos del Gen vpr/metabolismo , Lentivirus de los Primates/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Puntos de Control del Ciclo Celular , Daño del ADN , Productos del Gen vpr/genética , Células HEK293 , VIH-1/genética , VIH-1/fisiología , Células HeLa , Humanos , Lentivirus de los Primates/química , Proteínas de Mantenimiento de Minicromosoma/genética , Filogenia , Proteolisis , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/fisiología
4.
Sci Rep ; 9(1): 3937, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850685

RESUMEN

The discovery of tumor-associated antigens recognized by T lymphocytes opens the possibility of vaccinating cancer patients with defined antigens. However, one of the major limitation of peptide-based vaccines is the low immunogenicity of antigenic peptides. Interestingly, if these epitopes are directly delivered into the cytoplasm of antigen presenting cells, they can be efficiently presented via the direct MHC class I presentation pathway. To improve antigen entry, one promising approach is the use of cell penetrating peptides (CPPs). However, most studies use a covalent binding of the CPP with the antigen. In the present study, we focused on the C-terminal domain of Vpr which was previously demonstrated to efficiently deliver plasmid DNA into cells. We provide evidence that the peptides Vpr55-91 and Vpr55-82 possess the capacity of delivering proteins and epitopes into cell lines as well as into human primary dendritic cells, without the necessicity for a chemical linkage. Moreover, immunization of HLA-A2 transgenic mice with Vpr55-91 as the sole adjuvant is able to induce antigen-specific cytotoxic T lymphocytes against multiple tumor epitopes.


Asunto(s)
Péptidos de Penetración Celular/inmunología , Productos del Gen vpr/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Células CHO , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular , Péptidos de Penetración Celular/genética , Cricetulus , Sistemas de Liberación de Medicamentos , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Productos del Gen vpr/genética , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Células Hep G2 , Humanos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Transporte de Proteínas , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología
5.
Nat Microbiol ; 3(12): 1354-1361, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30297740

RESUMEN

Host factors that silence provirus transcription in CD4+ memory T cells help HIV-1 escape eradication by the host immune system and by antiviral drugs1. These same factors, however, must be overcome for HIV-1 to propagate. Here we show that Vpx and Vpr encoded by diverse primate immunodeficiency viruses activate provirus transcription. Vpx and Vpr are adaptor proteins for the DCAF1-CUL4A/B E3 ubiquitin ligase that degrade SAMHD1 and increase reverse transcription2-4. Nonetheless, Vpx and Vpr have effects on reporter gene expression that are not explained by SAMHD1 degradation5-8. A screen for factors that mimic these effects identified the human silencing hub (HUSH) complex, FAM208A (TASOR/RAP140), MPHOSPH8 (MPP8), PPHLN1 (PERIPHILIN) and MORC29-13. Vpx associated with the HUSH complex and decreased steady-state level of these proteins in a DCAF1/CUL4A/B/proteasome-dependent manner14,15. Replication kinetics of HIV-1 and SIVMAC was accelerated to a similar extent by vpx or FAM208A knockdown. Finally, vpx increased steady-state levels of LINE-1 ORF1p, as previously described for FAM208A disruption11. These results demonstrate that the HUSH complex represses primate immunodeficiency virus transcription, and that, to counteract this restriction, viral Vpx or Vpr proteins degrade the HUSH complex.


Asunto(s)
Productos del Gen vpr/metabolismo , Lentivirus de los Primates/metabolismo , Provirus/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Antígenos de Neoplasias , Proteínas Portadoras , Proteínas Cullin , Productos del Gen vpr/genética , Células HEK293 , Infecciones por VIH/virología , VIH-1/genética , Humanos , Lentivirus de los Primates/genética , Proteínas Nucleares , Fosfoproteínas , Proteínas Serina-Treonina Quinasas , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas , Proteínas Reguladoras y Accesorias Virales/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
6.
PLoS One ; 10(8): e0135633, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26270987

RESUMEN

Neurocognitive impairments affect a substantial population of HIV-1 infected individuals despite the success of anti-retroviral therapy in controlling viral replication. Astrocytes are emerging as a crucial cell type that might be playing a very important role in the persistence of neuroinflammation seen in patients suffering from HIV-1 associated neurocognitive disorders. HIV-1 viral proteins including Vpr exert neurotoxicity through direct and indirect mechanisms. Induction of IL-8 in microglial cells has been shown as one of the indirect mechanism through which Vpr reduces neuronal survival. We show that HIV-1 Vpr induces IL-6 and IL-8 in astrocytes in a time-dependent manner. Additional experiments utilizing chemical inhibitors and siRNA revealed that HIV-1 Vpr activates transcription factors NF-κB, AP-1 and C/EBP-δ via upstream protein kinases PI3K/Akt, p38-MAPK and Jnk-MAPK leading to the induction of IL-6 and IL-8 in astrocytes. We demonstrate that one of the mechanism for neuroinflammation seen in HIV-1 infected individuals involves induction of IL-6 and IL-8 by Vpr in astrocytes. Understanding the molecular pathways involved in the HIV-1 neuroinflammation would be helpful in the design of adjunct therapy to ameliorate some of the symptoms associated with HIV-1 neuropathogenesis.


Asunto(s)
Astrocitos/enzimología , Regulación de la Expresión Génica/fisiología , Productos del Gen vpr/metabolismo , VIH-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular , Células Cultivadas , Productos del Gen vpr/genética , Humanos , Inmunohistoquímica , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
7.
Virology ; 477: 10-17, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25618414

RESUMEN

Vpr and Vpx are a group of highly related accessory proteins from primate lentiviruses. Despite the high degree of amino acid homology within this group, these proteins can be highly divergent in their functions. In this work, we constructed chimeric and mutant proteins between HIV-1 and SIVagm Vpr in order to better understand the structure-function relationships. We tested these constructs for their abilities to induce G2 arrest in human cells and to degrade agmSAMHD1 and Mus81. We found that the C-terminus of HIV-1 Vpr, when transferred onto SIVagm Vpr, provides the latter with the de novo ability to induce G2 arrest in human cells. We confirmed that HIV-1 Vpr induces degradation of Mus81 although, surprisingly, degradation is independent and genetically separable from Vpr׳s ability to induce G2 arrest.


Asunto(s)
Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Productos del Gen vpr/metabolismo , VIH-1/fisiología , Interacciones Huésped-Patógeno , Proteínas de Unión al GTP Monoméricas/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Productos del Gen vpr/genética , Células HeLa , Humanos , Proteolisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína 1 que Contiene Dominios SAM y HD
8.
Retrovirology ; 11: 72, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25169827

RESUMEN

BACKGROUND: The HIV-1 accessory proteins, Viral Infectivity Factor (Vif) and the pleiotropic Viral Protein R (Vpr) are important for efficient virus replication. While in non-permissive cells an appropriate amount of Vif is critical to counteract APOBEC3G-mediated host restriction, the Vpr-induced G2 arrest sets the stage for highest transcriptional activity of the HIV-1 long terminal repeat. RESULTS: We identified a G run localized deep in the vpr AUG containing intron 3 (GI3-2), which was critical for balanced splicing of both vif and vpr non-coding leader exons. Inactivation of GI3-2 resulted in excessive exon 3 splicing as well as exon-definition mediated vpr mRNA formation. However, in an apparently mutually exclusive manner this was incompatible with recognition of upstream exon 2 and vif mRNA processing. As a consequence, inactivation of GI3-2 led to accumulation of Vpr protein with a concomitant reduction in Vif protein. We further demonstrate that preventing hnRNP binding to intron 3 by GI3-2 mutation diminished levels of vif mRNA. In APOBEC3G-expressing but not in APOBEC3G-deficient T cell lines, mutation of GI3-2 led to a considerable replication defect. Moreover, in HIV-1 isolates carrying an inactivating mutation in GI3-2, we identified an adjacent G-rich sequence (GI3-1), which was able to substitute for the inactivated GI3-2. CONCLUSIONS: The functionally conserved intronic G run in HIV-1 intron 3 plays a major role in the apparently mutually exclusive exon selection of vif and vpr leader exons and hence in vif and vpr mRNA formation. The competition between these exons determines the ability to evade APOBEC3G-mediated antiviral effects due to optimal vif expression.


Asunto(s)
Citidina Desaminasa/metabolismo , Infecciones por VIH/virología , VIH-1/genética , Especificidad del Huésped/genética , Intrones , Desaminasa APOBEC-3G , Línea Celular , Línea Celular Tumoral , Citidina Desaminasa/genética , Productos del Gen vpr/genética , Células HEK293 , Infecciones por VIH/metabolismo , Células HeLa , Humanos , Mutación/genética , Empalme del ARN/genética , ARN Mensajero/genética , Linfocitos T/metabolismo , Linfocitos T/virología , Replicación Viral/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética
9.
Retrovirology ; 11: 45, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24912525

RESUMEN

BACKGROUND: The Vpr protein of human immunodeficiency virus type 1 (HIV-1) plays an important role in viral replication. It has been reported that Vpr stimulates the nuclear factor-κB (NF-κB) and activator protein 1 (AP-1) signaling pathways, and thereby regulates viral and host cell gene expression. However, the molecular mechanism behind this function of Vpr is not fully understood. RESULTS: Here, we have identified transforming growth factor-ß-activated kinase 1 (TAK1) as the important upstream signaling molecule that Vpr associates with in order to activate NF-κB and AP-1 signaling. HIV-1 virion-associated Vpr is able to stimulate phosphorylation of TAK1. This activity of Vpr depends on its association with TAK1, since the S79A Vpr mutant lost interaction with TAK1 and was unable to activate TAK1. This association allows Vpr to promote the interaction of TAB3 with TAK1 and increase the polyubiquitination of TAK1, which renders TAK1 phosphorylation. In further support of the key role of TAK1 in this function of Vpr, knockdown of endogenous TAK1 significantly attenuated the ability of Vpr to activate NF-κB and AP-1 as well as the ability to stimulate HIV-1 LTR promoter. CONCLUSIONS: HIV-1 Vpr enhances the phosphorylation and polyubiquitination of TAK1, and as a result, activates NF-κB and AP-1 signaling pathways and stimulates HIV-1 LTR promoter.


Asunto(s)
Productos del Gen vpr/metabolismo , VIH-1/fisiología , Quinasas Quinasa Quinasa PAM/genética , FN-kappa B/metabolismo , Factor de Transcripción AP-1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Línea Celular , Línea Celular Tumoral , Productos del Gen vpr/genética , Células HEK293 , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Jurkat , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/genética , Fosforilación , Regiones Promotoras Genéticas , Transducción de Señal , Factor de Transcripción AP-1/genética , Ubiquitinación , Replicación Viral
10.
PLoS One ; 8(12): e82128, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367500

RESUMEN

BACKGROUND: Since HIV-1 Tat and Vpr genes are involved in promoter transactivation, apoptosis, etc, we carried out studies to find out nature and extent of natural variation in the two genes from seropositive patients from Northern India and determined their functional implications. METHODS: HIV-1 tat exon 1 and vpr were amplified from the genomic DNA isolated from the blood of HIV-1 infected individuals using specific primers by Polymerase Chain reaction (PCR) and subjected to extensive genetic analysis (CLUSTAL W, Simplot etc). Their expression was monitored by generating myc fusion clones. Tat exon 1 and Vpr variants were co-transfected with the reporter gene construct (LTR-luc) and their transactivation potential was monitored by measuring luciferase activity. Apoptosis and cell cycle analysis was done by Propidium Iodide (PI) staining followed by FACS. RESULTS: Exon 1 of tat was amplified from 21 samples and vpr was amplified from 16 samples. One of the Tat exon 1 variants showed phylogenetic relatedness to subtype B & C and turned out to be a unique recombinant. Two of the Vpr variants were B/C/D recombinants. These natural variations were found to have no impact on the stability of Tat and Vpr. These variants differed in their ability to transactivate B LTR and C LTR promoters. B/C recombinant Tat showed better co-operative interaction with Vpr. B/C/D recombination in Vpr was found to have no effect on its co-operativity with Tat. Recombinant Tat (B/C) induced more apoptosis than wild type B and C Tat. The B/C/D recombination in Vpr did not affect its G2 arrest induction potential but reduced its apoptosis induction ability. CONCLUSIONS: Extensive sequence and region-specific variations were observed in Tat and Vpr genes from HIV-1 infected individuals from Northern India. These variations have functional implications & therefore important for the pathogenicity of virus.


Asunto(s)
Apoptosis/genética , Regulación Viral de la Expresión Génica/genética , Productos del Gen vpr/genética , VIH-1/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Adolescente , Adulto , Western Blotting , Electroforesis en Gel de Poliacrilamida , Exones/genética , Femenino , Citometría de Flujo , Células HeLa , Humanos , India , Masculino , Persona de Mediana Edad , Adulto Joven
11.
Sci Transl Med ; 5(213): 213ra164, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24285483

RESUMEN

Viral infections, such as HIV, have been linked to obesity, but mechanistic evidence that they cause adipose dysfunction in vivo is lacking. We investigated a pathogenic role for the HIV-1 accessory protein viral protein R (Vpr), which can coactivate the glucocorticoid receptor (GR) and co-repress peroxisome proliferator-activated receptor γ (PPARγ) in vitro, in HIV-associated adipose dysfunction. Vpr circulated in the blood of most HIV-infected patients tested, including those on antiretroviral therapy (ART) with undetectable viral load. Vpr-mediated mechanisms were dissected in vivo using mouse models expressing the Vpr transgene in adipose tissues and liver (Vpr-Tg) or infused with synthetic Vpr. Both models demonstrated accelerated whole-body lipolysis, hyperglycemia and hypertriglyceridemia, and tissue-specific findings. Fat depots in these mice had diminished mass, macrophage infiltration, and blunted PPARγ target gene expression but increased GR target gene expression. In liver, we observed blunted PPARα target gene expression, steatosis with decreased adenosine monophosphate-activated protein kinase activity, and insulin resistance. Similar to human HIV-infected patients, Vpr circulated in the serum of Vpr-Tg mice. Vpr blocked differentiation in preadipocytes through cell cycle arrest, whereas in mature adipocytes, it increased lipolysis with reciprocally altered association of PPARγ and GR with their target promoters. These results delineate a distinct pathogenic sequence: Vpr, released from HIV-1 in tissue reservoirs after ART, can disrupt PPAR/GR co-regulation and cell cycle control to produce adipose dysfunction and hepatosteatosis. Confirmation of these mechanisms in HIV patients could lead to targeted treatment of the metabolic complications with Vpr inhibitors, GR antagonists, or PPARγ/PPARα agonists.


Asunto(s)
Productos del Gen vpr/metabolismo , VIH-1/metabolismo , Receptores de Glucocorticoides/metabolismo , Células 3T3-L1 , Animales , Cromatografía en Capa Delgada , Ensayo de Inmunoadsorción Enzimática , Productos del Gen vpr/genética , VIH-1/genética , Humanos , Immunoblotting , Masculino , Ratones , Ratones Transgénicos , PPAR alfa/agonistas , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Receptores de Glucocorticoides/agonistas
12.
Virology ; 446(1-2): 144-51, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24074576

RESUMEN

Viral protein R (Vpr) is an accessory protein of HIV and SIV involved in the pathogenesis of viral infection. In this study, we monitored SIV evolution in the central nervous system and other organs from morphine-dependent and control animals by sequencing vpr in an attempt to understand the relationship between drug abuse, disease progression, and compartmentalization of viral evolution. Animals in the morphine group developed accelerated disease and died within twenty weeks post-infection. A unique mutation, R50G, was identified in the macaques that survived regardless of morphine exposure. Functional studies revealed that the R50G mutation exhibited altered cellular localization and decreased the expression levels of both IL-6 and IL-8. Our results, therefore, suggest that sequence changes within the SIV/17E-Fr vpr occur regardless of drug abuse but correlate with survival, and that they alter disease progression rates by affecting Vpr functions.


Asunto(s)
Productos del Gen vpr/genética , Morfina/administración & dosificación , Mutación Missense , Trastornos Relacionados con Opioides/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Factores de Virulencia/genética , Animales , Progresión de la Enfermedad , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Análisis de Supervivencia
13.
Cell Host Microbe ; 14(1): 85-92, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23870316

RESUMEN

HIV-1 resulted from cross-species transmission of SIVcpz, a simian immunodeficiency virus that naturally infects chimpanzees. SIVcpz, in turn, is a recombinant between two SIV lineages from Old World monkeys. Lentiviral interspecies transmissions are partly driven by the evolution and capacity of viral accessory genes, such as vpx, vpr, and vif, to antagonize host antiviral factors, such as SAMHD1 and the APOBEC3 proteins. We show that vpx, which in other lentiviruses antagonizes SAMHD1, was deleted during the creation of SIVcpz. This genomic deletion resulted in the reconstruction of the overlapping vif gene by "overprinting," creating a unique vif that overlaps in its 3' end with the vpr gene and can antagonize hominid APOBEC3s. Moreover, passage of SIVs through chimpanzees facilitated the subsequent adaptation of HIV-1 to humans. Thus, HIV-1 originated through a series of gene loss and adaptation events that generated its chimpanzee precursor and lowered the species barrier to human infection.


Asunto(s)
Evolución Molecular , Eliminación de Gen , Infecciones por VIH/virología , VIH-1/genética , Hominidae/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Secuencia de Aminoácidos , Animales , Productos del Gen vif/química , Productos del Gen vif/genética , Productos del Gen vif/metabolismo , Productos del Gen vpr/química , Productos del Gen vpr/genética , Productos del Gen vpr/metabolismo , VIH-1/química , VIH-1/clasificación , VIH-1/metabolismo , Haplorrinos , Humanos , Datos de Secuencia Molecular , Pan troglodytes , Filogenia , Alineación de Secuencia , Virus de la Inmunodeficiencia de los Simios/química , Virus de la Inmunodeficiencia de los Simios/clasificación , Virus de la Inmunodeficiencia de los Simios/metabolismo
14.
Cell Microbiol ; 14(11): 1745-56, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22776683

RESUMEN

HIV-2 and closely related SIV Vpx proteins are essential for viral replication in macrophages and dendritic cells. Vpx hijacks DCAF1-DDB1-Cul4 E3 ubiquitin ligase to promote viral replication. DCAF1 is essential for cell proliferation and embryonic development and is responsible for the polyubiquitination of poorly defined cellular proteins. How substrate receptors recruit the DCAF1-containing E3 ubiquitin ligase to induce protein degradation is still poorly understood. Here we identify a highly conserved motif (Wx4Φx2Φx3AΦxH) that is present in diverse Vpx and Vpr proteins of primate lentiviruses. We demonstrate that the Wx4Φx2Φx3AΦxH motif in SIVmac Vpx is required for both the Vpx-DCAF1 interaction and/or Vpx-mediated degradation of SAMHD1. DCAF1-binding defective Vpx mutants also have impaired ability to promote SIVΔVpx virus infection of myeloid cells. Critical amino acids in the Wx4Φx2Φx3AΦxH motif of SIV Vpx that are important for DCAF1 interaction maintained the ability to bind SAMHD1, indicating that the DCAF1 and SAMHD1 interactions involve distinctive interfaces in Vpx. Surprisingly, VpxW24A mutant proteins that were still capable of binding DCAF1 and SAMHD1 lost the ability to induce SAMHD1 degradation, suggesting that Vpx is not a simple linker between the DCAF1-DDB1-Cul4 E3 ubiquitin ligase and its substrate, SAMHD1.VpxW24A maintained the ability to accumulate in the nucleus despite the fact that nuclear, but not cytoplasmic, mutant forms of SAMHD1 were more sensitive to Vpx-mediated degradation. The Wx4Φx2Φx3AΦxH motif in HIV-1 Vpr is also required for the Vpr-DCAF1 interaction and Vpr-induced G2 cell cycle arrest. Thus, our data reveal previously unrecognized functional interactions involved in the assembly of virally hijacked DCAF1-DDB1-based E3 ubiquitin ligase complex.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclo Celular , Productos del Gen vpr/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral , Secuencias de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Secuencia Conservada , Productos del Gen vpr/genética , Humanos , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas Reguladoras y Accesorias Virales/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-21710851

RESUMEN

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


Asunto(s)
Complejo 2 de Proteína Adaptadora/fisiología , Subunidades alfa de Complejo de Proteína Adaptadora/fisiología , Productos del Gen gag/fisiología , Productos del Gen vpr/fisiología , VIH-1/fisiología , Integrasas/fisiología , Replicación Viral/genética , Replicación Viral/fisiología , Complejo 2 de Proteína Adaptadora/genética , Subunidades alfa de Complejo de Proteína Adaptadora/genética , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Productos del Gen gag/genética , Productos del Gen vpr/genética , VIH-1/genética , Proteínas del Virus de la Inmunodeficiencia Humana/fisiología , Humanos , Integrasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/fisiología
16.
Zhonghua Zhong Liu Za Zhi ; 32(10): 725-8, 2010 Oct.
Artículo en Chino | MEDLINE | ID: mdl-21163059

RESUMEN

OBJECTIVE: To study the anti-glioma effect of recombinant adenovirus mediated combined gene therapy of bFGF-siRNA and HIV1-Vpr in vivo. METHODS: Mouse glioma model was established by injecting 5 × 10(6) LN229 cells into BALB/c-nu nude mice. 30 nude mice were randomly divided into 5 groups: the negative control group, mock group, bFGF-siRNA group, Vpr group and combined therapy group, which at regular intervals were injected with PBS, rAd5-null, rAd5-bFGF-siRNA, rAd5-Vpr, rAd5-bFGF-siRNA plus rAd5-Vpr, respectively. The tumor volume was recorded every third day to draw a growth curve. After four weeks treatment, the mice were killed and specimens were taken. HE, immunohistochemical and TUNEL staining were performed to observe the cell morphology, detect the changes of relevant target proteins and cell apoptosis, respectively. Also the ultrastructural changes were observed by electron microscopy. RESULTS: The tumor growth inhibition rates were 36.9%, 37.2% and 58.6% in the bFGF-siRNA group, Vpr group and combined therapy group, respectively, and the combined therapy group showed the most significant effect (P < 0.05). Also the results of HE, immunohistochemical and TUNEL staining revealed that the combined therapy group had the best effects on proliferation inhibition and apoptosis induced in glioma cells (P < 0.05). The most significant ultrastructural changes were observed in the combined therapy group. CONCLUSION: The combined gene therapy of bFGF-siRNA with Vpr shows a prominent and synergistic anti-glioma effect compared with that of mono-gene therapy in nude mice.


Asunto(s)
Apoptosis , Factor 2 de Crecimiento de Fibroblastos/genética , Productos del Gen vpr/genética , Glioma/terapia , ARN Interferente Pequeño/genética , Adenoviridae/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Proliferación Celular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Productos del Gen vpr/metabolismo , Terapia Genética , Glioma/metabolismo , Glioma/patología , VIH-1/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Distribución Aleatoria , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
J Virol ; 84(20): 10907-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20686015

RESUMEN

The kinetics of CD8(+) T cell epitope presentation contribute to the antiviral efficacy of these cells yet remain poorly defined. Here, we demonstrate presentation of virion-derived Vpr peptide epitopes early after viral penetration and prior to presentation of Vif-derived epitopes, which required de novo Vif synthesis. Two Rev epitopes exhibited differential presentation kinetics, with one Rev epitope presented within 1 h of infection. We also demonstrate that cytolytic activity mirrors the recognition kinetics of infected cells. These studies show for the first time that Vpr- and Rev-specific CD8(+) T cells recognize and kill simian immunodeficiency virus (SIV)-infected CD4(+) T cells early after SIV infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Productos del Gen rev/inmunología , Productos del Gen vpr/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Citotoxicidad Inmunológica , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Productos del Gen rev/genética , Productos del Gen vpr/genética , Interacciones Huésped-Patógeno/inmunología , Técnicas In Vitro , Cinética , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/enzimología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Virus de la Inmunodeficiencia de los Simios/fisiología
18.
FASEB J ; 24(11): 4343-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20628092

RESUMEN

Painful peripheral neuropathy has become the principal neurological disorder in HIV/AIDS patients. Herein, we investigated the effects of a cytotoxic HIV-1 accessory protein, viral protein R (Vpr), on the peripheral nervous system (PNS). Host and viral gene expression was investigated in peripheral nerves from HIV-infected individuals and in HIV-infected human dorsal root ganglion (DRG) cultures by RT-PCR and immunocytochemistry. Cytosolic calcium ([Ca(2+)]) fluxes and neuronal membrane responses were analyzed in cultured DRGs. Neurobehavioral responses and cytokine levels were assessed in a transgenic mouse model in which the vpr transgene was expressed in an immunodeficient background (vpr/RAG1(-/-)). Vpr transcripts and proteins were detected in peripheral nerves and DRGs from HIV-infected patients. Exposure of rat or human cultured DRG neurons to Vpr rapidly increased [Ca(2+)] and action potential frequency while increasing input resistance. HIV infection of human DRG cultures caused neurite retraction (P<0.05), accompanied by induction of interferon-α (IFN-α) transcripts (P<0.05). vpr/RAG1(-/-) mice expressed Vpr together with increased IFN-α (P<0.05) in the PNS and also exhibited mechanical allodynia, unlike their vpr/RAG1(-/-) littermates (P<0.05). Herein, Vpr caused DRG neuronal damage, likely through cytosolic calcium activation and cytokine perturbation, highlighting Vpr's contribution to HIV-associated peripheral neuropathy and ensuing neuropathic pain.


Asunto(s)
Productos del Gen vpr/metabolismo , VIH-1 , Neuralgia/complicaciones , Enfermedades del Sistema Nervioso Periférico/complicaciones , Traumatismos del Sistema Nervioso/complicaciones , Animales , Células Cultivadas , Ganglios Espinales/fisiopatología , Ganglios Espinales/virología , Regulación de la Expresión Génica , Productos del Gen vpr/genética , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
AIDS ; 24(8): 1107-19, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20404718

RESUMEN

OBJECTIVE: HIV-associated nephropathy (HIVAN) is the most common cause of end-stage renal disease in persons with HIV/AIDS and is characterized by focal glomerulosclerosis and dysregulated renal tubular epithelial cell (RTEC) proliferation and apoptosis. HIV-1 viral protein r (Vpr) has been implicated in HIV-induced RTEC apoptosis but the mechanisms of Vpr-induced RTEC apoptosis are unknown. The aim of this study was therefore to determine the mechanisms of Vpr-induced apoptosis in RTEC. METHODS: Apoptosis and caspase activation were analyzed in human RTEC (HK2) after transduction with Vpr-expressing and control lentiviral vectors. Bax and BID were inhibited with lentiviral shRNA, and ERK activation was blocked with the MEK1,2 inhibitor, U0126. RESULTS: Vpr induced apoptosis as indicated by caspase 3/7 activation, PARP-1 cleavage and mitochondrial injury. Vpr activated both caspases-8 and 9. Inhibition of Bax reduced Vpr-induced apoptosis, as reported in other cell types. Additionally, Vpr-induced cleavage of BID to tBID and suppression of BID expression prevented Vpr-induced apoptosis. Since sustained ERK activation can activate caspase-8 in some cell types, we studied the role of ERK in Vpr-induced caspase-8 activation. Vpr induced sustained ERK activation in HK2 cells and incubation with U0126 reduced Vpr-induced caspase-8 activation, BID cleavage and apoptosis. We detected phosphorylated ERK in RTEC in HIVAN biopsy specimens by immunohistochemistry. CONCLUSIONS: These studies delineate a novel pathway of Vpr-induced apoptosis in RTEC, which is mediated by sustained ERK activation, resulting in caspase 8-mediated cleavage of BID to tBID, thereby facilitating Bax-mediated mitochondrial injury and apoptosis.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , Apoptosis/fisiología , Caspasa 8/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Productos del Gen vpr/metabolismo , VIH-1 , Fallo Renal Crónico/metabolismo , Nefropatía Asociada a SIDA/genética , Nefropatía Asociada a SIDA/virología , Apoptosis/genética , Caspasa 8/genética , Proliferación Celular , Regulación Viral de la Expresión Génica , Productos del Gen vpr/genética , Humanos , Fallo Renal Crónico/genética , Fallo Renal Crónico/virología , Túbulos Renales/virología , ARN Viral , Replicación Viral
20.
PLoS Pathog ; 5(10): e1000613, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19798433

RESUMEN

Natural killer (NK) cells are stimulated by ligands on virus-infected cells. We have recently demonstrated that NK cells respond to human immunodeficiency virus type-1 (HIV-1)-infected autologous T-cells, in part, through the recognition of ligands for the NK cell activating receptor NKG2D on the surface of the infected cells. Uninfected primary CD4(pos) T-cell blasts express little, if any, NKG2D ligands. In the present study we determined the mechanism through which ligands for NKG2D are induced on HIV-1-infected cells. Our studies reveal that expression of vpr is necessary and sufficient to elicit the expression of NKG2D ligands in the context of HIV-1 infection. Vpr specifically induces surface expression of the unique-long 16 binding proteins (ULBP)-1 and ULBP-2, but not ULBP-3, MHC class I-related chain molecules (MIC)-A or MIC-B. In these studies we also demonstrated that Vpr increases the level of ULBP-1 and ULBP-2 mRNA in primary CD4(pos) T-cell blasts. The presence of ULBP-1 and ULBP-2 on HIV-1 infected cells is dependent on the ability of Vpr to associate with a protein complex know as Cullin 4a (Cul4a)/damaged DNA binding protein 1 (DDB1) and Cul4a-associated factor-1(DCAF-1) E3 ubiquitin ligase (Cul4a(DCAF-1)). ULBP-1 and -2 expression by Vpr is also dependent on activation of the DNA damage sensor, ataxia telangiectasia and rad-3-related kinase (ATR). When T-cell blasts are infected with a vpr-deficient HIV-1, NK cells are impaired in killing the infected cells. Thus, HIV-1 Vpr actively triggers the expression of the ligands to the NK cell activation receptor.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Productos del Gen vpr/metabolismo , Infecciones por VIH/inmunología , Células Asesinas Naturales/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos T CD4-Positivos/virología , Proteínas Cullin/biosíntesis , Daño del ADN , Citometría de Flujo , Proteínas Ligadas a GPI , Expresión Génica , Regulación Viral de la Expresión Génica , Productos del Gen vpr/genética , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/biosíntesis , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/virología , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA