Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 592
Filter
1.
Virol J ; 21(1): 47, 2024 02 23.
Article in English | MEDLINE | ID: mdl-38395987

ABSTRACT

HIV infection compromises both the peripheral and central immune systems due to its pathogenic and neuropathogenic features. The mechanisms driving HIV-1 pathogenesis and neuropathogenesis involve a series of events, including metabolic dysregulation. Furthermore, HIV-subtype-specific variations, particularly alterations in the amino acid sequences of key viral proteins, are known to influence the severity of clinical outcomes in people living with HIV. However, the impact of amino acid sequence variations in specific viral proteins, such as Viral protein R (Vpr), on metabolites within the Tryptophan (Trp)-kynurenine (Kyn) pathway in people living with HIV remains unclear. Our research aimed to explore the relationship between variations in the Vpr amino acid sequence (specifically at positions 22, 41, 45, and 55, as these have been previously linked to neurocognitive function) and peripheral Trp-Kyn metabolites. Additionally, we sought to clarify the systems biology of Vpr sequence variation by examining the link between Trp-Kyn metabolism and peripheral inflammation, as a neuropathogenic mechanism. In this preliminary study, we analyzed a unique cohort of thirty-two (n = 32) South African cART naïve people living with HIV. We employed Sanger sequencing to ascertain blood-derived Vpr amino acid sequence variations and a targeted LC-MS/MS metabolomics platform to assess Trp-Kyn metabolites, such as Trp, Kyn, kynurenic acid (KA), and quinolinic acid (QUIN). Particle-enhanced turbidimetric assay and Enzyme-linked immunosorbent assays were used to measure immune markers, hsCRP, IL-6, suPAR, NGAL and sCD163. After applying Bonferroni corrections (p =.05/3) and adjusting for covariates (age and sex), only the Vpr G41 and A55 groups was nearing significance for higher levels of QUIN compared to the Vpr S41 and T55 groups, respectively (all p =.023). Multiple regression results revealed that Vpr amino acid variations at position 41 (adj R2 = 0.049, ß = 0.505; p =.023), and 55 (adj R2 = 0.126, ß = 0.444; p =.023) displayed significant associations with QUIN after adjusting for age and sex. Lastly, the higher QUIN levels observed in the Vpr G41 group were found to be correlated with suPAR (r =.588, p =.005). These results collectively underscore the importance of specific Vpr amino acid substitutions in influencing QUIN and inflammation (specifically suPAR levels), potentially contributing to our understanding of their roles in the pathogenesis and neuropathogenesis of HIV-1.


Subject(s)
Gene Products, vpr , HIV Infections , HIV Seropositivity , HIV-1 , Humans , Tryptophan/metabolism , Kynurenine/metabolism , HIV-1/genetics , HIV-1/metabolism , Amino Acid Sequence , HIV Infections/complications , Chromatography, Liquid , Pilot Projects , Receptors, Urokinase Plasminogen Activator , Tandem Mass Spectrometry , Inflammation
2.
Kidney Int ; 103(3): 529-543, 2023 03.
Article in English | MEDLINE | ID: mdl-36565808

ABSTRACT

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.


Subject(s)
AIDS-Associated Nephropathy , Gene Products, vpr , HIV Infections , HIV-1 , Renal Insufficiency, Chronic , Animals , Humans , Mice , AIDS-Associated Nephropathy/genetics , Disease Models, Animal , Gene Products, vpr/genetics , Gene Products, vpr/metabolism , Gene Products, vpr/pharmacology , HIV Infections/complications , HIV-1/genetics , HIV-1/metabolism , Human Immunodeficiency Virus Proteins , Mice, Transgenic , Renal Insufficiency, Chronic/complications
3.
PLoS Pathog ; 17(8): e1009775, 2021 08.
Article in English | MEDLINE | ID: mdl-34339457

ABSTRACT

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.


Subject(s)
Cullin Proteins/chemistry , Gene Products, vpr/metabolism , Proteasome Endopeptidase Complex/chemistry , SAM Domain and HD Domain-Containing Protein 1/chemistry , Ubiquitination , Virus Replication , Amino Acid Sequence , Animals , Cryoelectron Microscopy , Cullin Proteins/metabolism , Gene Products, vpr/genetics , NEDD8 Protein/chemistry , NEDD8 Protein/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , SAM Domain and HD Domain-Containing Protein 1/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
4.
Nat Immunol ; 12(10): 975-83, 2011 Aug 28.
Article in English | MEDLINE | ID: mdl-21874023

ABSTRACT

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


Subject(s)
Cytidine Deaminase/physiology , HIV/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/virology , APOBEC-3G Deaminase , Cells, Cultured , Cytotoxicity, Immunologic , DNA Damage , Gene Products, vpr/physiology , Humans , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Uridine/metabolism
5.
J Gen Virol ; 103(10)2022 10.
Article in English | MEDLINE | ID: mdl-36205476

ABSTRACT

Macaque-tropic HIV-1 (HIV-1mt) variants have been developed to establish preferable primate models that are advantageous in understanding HIV-1 infection pathogenesis and in assessing the preclinical efficacy of novel prevention/treatment strategies. We previously reported that a CXCR4-tropic HIV-1mt, MN4Rh-3, efficiently replicates in peripheral blood mononuclear cells (PBMCs) of cynomolgus macaques homozygous for TRIMCyp (CMsTC). However, the CMsTC challenged with MN4Rh-3 displayed low viral loads during the acute infection phase and subsequently exhibited short-term viremia. These virological phenotypes in vivo differed from those observed in most HIV-1-infected people. Therefore, further development of the HIV-1mt variant was needed. In this study, we first reconstructed the MN4Rh-3 clone to produce a CCR5-tropic HIV-1mt, AS38. In addition, serial in vivo passages allowed us to produce a highly adapted AS38-derived virus that exhibits high viral loads (up to approximately 106 copies ml-1) during the acute infection phase and prolonged periods of persistent viremia (lasting approximately 16 weeks postinfection) upon infection of CMsTC. Whole-genome sequencing of the viral genomes demonstrated that the emergence of a unique 15-nt deletion within the vif gene was associated with in vivo adaptation. The deletion resulted in a significant increase in Vpr protein expression but did not affect Vif-mediated antagonism of antiretroviral APOBEC3s, suggesting that Vpr is important for HIV-1mt adaptation to CMsTC. In summary, we developed a novel CCR5-tropic HIV-1mt that can induce high peak viral loads and long-term viremia and exhibits increased Vpr expression in CMsTC.


Subject(s)
Gene Products, vpr , HIV Infections , HIV Seropositivity , HIV-1 , Simian Immunodeficiency Virus , Animals , HIV-1/genetics , Leukocytes, Mononuclear , Macaca fascicularis , Simian Immunodeficiency Virus/genetics , Viremia , Virus Replication
6.
J Virol ; 94(4)2020 01 31.
Article in English | MEDLINE | ID: mdl-31776272

ABSTRACT

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


Subject(s)
Antiviral Agents/metabolism , HIV-1/metabolism , Virus Replication/physiology , vpr Gene Products, Human Immunodeficiency Virus/physiology , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Products, vpr/metabolism , Gene Products, vpr/physiology , HEK293 Cells , HIV Infections/virology , HIV-1/physiology , HeLa Cells , Host-Pathogen Interactions , Humans , Macrophages/metabolism , Primary Cell Culture , Ubiquitin-Protein Ligases/metabolism , Virion/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism
7.
Chem Biodivers ; 18(10): e2100401, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34415099

ABSTRACT

A new menthane-type monoterpene, alpigalanol (1), together with four known terpenes (2-5) were isolated from the ethyl acetate soluble fraction of the 70 % ethanol extract of the Alpinia galanga rhizomes. The structure of 1 was determined by spectroscopic analyses, including 1D- and 2D-NMR. The extract of the A. galanga rhizomes and all isolated compounds (1-5) possessed Vpr inhibitory activities against the TREx-HeLa-Vpr cells at a concentration of 1.25 µM without showing any cytotoxicity.


Subject(s)
Alpinia/chemistry , Gene Products, vpr/antagonists & inhibitors , Monoterpenes/pharmacology , Rhizome/chemistry , Density Functional Theory , Dose-Response Relationship, Drug , HeLa Cells , Humans , Molecular Conformation , Monoterpenes/chemistry , Monoterpenes/isolation & purification , Tumor Cells, Cultured
8.
J Infect Dis ; 218(9): 1447-1452, 2018 09 22.
Article in English | MEDLINE | ID: mdl-29878133

ABSTRACT

A 48-year-old woman was infected with a vpr-defective human immunodeficiency virus (HIV)-1 molecular clone. Seroconversion was markedly delayed, and without treatment she had durably suppressed viremia and normal T-cell levels. Neutralizing antibody and CD8+ T-cell immune responses against HIV-1 were unremarkable. Viral sequences confirmed the source but evolved defective nef, suggesting an unknown mechanistic link to vpr. There were subtle qualitative defects in T and B cells. To our knowledge, this is the only case of human infection with a characterized defective HIV-1 molecular clone, which furthermore recapitulated live-attenuated vaccination in macaque models of HIV-1 vaccine research.


Subject(s)
AIDS Vaccines/immunology , Gene Products, vpr/immunology , HIV Infections/immunology , HIV-1/immunology , Vaccines, Attenuated/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cloning, Molecular , Female , Humans , Middle Aged , Vaccination/methods
9.
Molecules ; 23(8)2018 Jul 26.
Article in English | MEDLINE | ID: mdl-30049955

ABSTRACT

HIV-1 integrase (IN) inhibitors represent a new class of highly effective anti-AIDS therapeutics. Current FDA-approved IN strand transfer inhibitors (INSTIs) share a common mechanism of action that involves chelation of catalytic divalent metal ions. However, the emergence of IN mutants having reduced sensitivity to these inhibitors underlies efforts to derive agents that antagonize IN function by alternate mechanisms. Integrase along with the 96-residue multifunctional accessory protein, viral protein R (Vpr), are both components of the HIV-1 pre-integration complex (PIC). Coordinated interactions within the PIC are important for viral replication. Herein, we report a 7-mer peptide based on the shortened Vpr (69⁻75) sequence containing a biotin group and a photo-reactive benzoylphenylalanyl residue, and which exhibits low micromolar IN inhibitory potency. Photo-crosslinking experiments have indicated that the peptide directly binds IN. The peptide does not interfere with IN-DNA interactions or induce higher-order, aberrant IN multimerization, suggesting a mode of action for the peptide that is distinct from clinically used INSTIs and developmental allosteric IN inhibitors. This compact Vpr-derived peptide may serve as a valuable pharmacological tool to identify a potential new pharmacologic site.


Subject(s)
Gene Products, vpr/chemistry , Gene Products, vpr/metabolism , HIV Infections/virology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/physiology , Peptides/pharmacology , Amino Acid Sequence , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization
10.
J Virol ; 89(1): 230-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25320300

ABSTRACT

UNLABELLED: The accessory gene vpr, common to all primate lentiviruses, induces potent G2/M arrest in cycling cells. A recent study showed that human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) mediates this through activation of the SLX4/MUS81/EME1 exonuclease complex that forms part of the Fanconi anemia DNA repair pathway. To confirm these observations, we have examined the G2/M arrest phenotypes of a panel of simian immunodeficiency virus (SIV) Vpr proteins. We show that SIV Vpr proteins differ in their ability to promote cell cycle arrest in human cells. While this is dependent on the DCAF1/DDB1/CUL4 ubiquitin ligase complex, interaction with human DCAF1 does not predict G2/M arrest activity of SIV Vpr in human cells. In all cases, SIV Vpr-mediated cell cycle arrest in human cells correlated with interaction with human SLX4 (huSLX4) and could be abolished by small interfering RNA (siRNA) depletion of any member of the SLX4 complex. In contrast, all but one of the HIV/SIV Vpr proteins tested, including those that lacked activity in human cells, were competent for G2/M arrest in grivet cells. Correspondingly, here cell cycle arrest correlated with interaction with the grivet orthologues of the SLX4 complex, suggesting a level of host adaptation in these interactions. Phylogenetic analyses strongly suggest that G2/M arrest/SLX4 interactions are ancestral activities of primate lentiviral Vpr proteins and that the ability to dysregulate the Fanconi anemia DNA repair pathway is an essential function of Vpr in vivo. IMPORTANCE: The Vpr protein of HIV-1 and related viruses is essential for the virus in vivo. The ability of Vpr to block the cell cycle at mitotic entry is well known, but the importance of this function for viral replication is unclear. Recent data have shown that HIV-1 Vpr targets the Fanconi anemia DNA repair pathway by interacting with and activating an endonuclease complex, SLX4/MUS81/EME1, that processes interstrand DNA cross-links. Here we show that the ability of a panel of SIV Vpr proteins to mediate cell cycle arrest correlates with species-specific interactions with the SLX4 complex in human and primate cells. The results of these studies suggest that the SLX4 complex is a conserved target of primate lentiviral Vpr proteins and that the ability to dysregulate members of the Fanconi anemia DNA repair pathway is essential for HIV/SIV replication in vivo.


Subject(s)
Cell Cycle Checkpoints , Gene Products, vpr/metabolism , Host-Pathogen Interactions , Recombinases/metabolism , Simian Immunodeficiency Virus/physiology , Animals , Cell Line , Cercopithecinae , Humans
11.
Bioorg Med Chem Lett ; 26(19): 4620-4624, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27575477

ABSTRACT

Viral protein R (Vpr) is an accessory protein that plays important roles in the viral pathogenesis of Human Immunodeficiency Virus-1 (HIV-1). An assay for anti-Vpr activity, using TREx-HeLa-Vpr cells, is a promising strategy to discover Vpr inhibitors. The anti-Vpr assay revealed that the CHCl3-soluble extract of Picrasma javanica bark possesses potent anti-Vpr activity. Furthermore, studies of quassinoids (1-15) previously isolated from the extract demonstrated that all of the tested quassinoids exhibit anti-Vpr activity. Among the tested compounds, javanicin I (15) exhibited the most potent anti-Vpr activity ((***)p <0.001) in comparing with that of the positive control, damnacanthal. The structure-activity relationships of the active quassinoids suggested that the presence of a methyl group at C-13 in the 2,12,14-triene-1,11,16-trione-2,12-dimethoxy-18-norpicrasane quassinoids is the important factor for the potent inhibitory effect in TREx-HeLa-Vpr cells.


Subject(s)
Anti-HIV Agents/therapeutic use , Gene Products, vpr/antagonists & inhibitors , HIV Infections/drug therapy , Picrasma/chemistry , Plant Bark/chemistry , Quassins/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HeLa Cells , Humans , Myanmar , Quassins/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 26(7): 1789-93, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26916438

ABSTRACT

Viral protein R (Vpr), an accessory gene of HIV-1, plays important roles in viral pathogenesis. Screening of Myanmar medicinal plants that are popular as primary treatments for HIV/AIDS and for HIV-related problems revealed the potent anti-Vpr activity of the CHCl3-soluble extract of Kaempferia pulchra rhizomes, in comparison with that of the positive control, damnacanthal. Fractionation of the active CHCl3-soluble extract led to the identification of 30 isopimarane diterpenoids, including kaempulchraols A-W (1-23). All isolates were assayed for anti-Vpr activity against TREx-HeLa-Vpr cells, in which Vpr expression is tightly regulated by tetracycline. Kaempulchraols B (2), D (4), G (7), Q (17), T (20), U (21), and W (23) exhibited potent anti-Vpr activity, at concentrations ranging from 1.56 to 6.25µM. The structure-activity relationships of the active kaempulchraols suggested that the presence of a hydroxy group at C-14 in an isopimara-8(9),15-diene skeleton and the presence of an acetoxy group at C-1 or C-7 in an isopimara-8(14),15-diene skeleton are the critical factors for the inhibitory effects against TREx-HeLa-Vpr cells.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Diterpenes/chemistry , Diterpenes/pharmacology , Gene Products, vpr/antagonists & inhibitors , HIV-1/drug effects , Zingiberaceae/chemistry , Anti-HIV Agents/isolation & purification , Diterpenes/isolation & purification , Gene Products, vpr/metabolism , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/metabolism , HeLa Cells , Humans , Rhizome/chemistry , Structure-Activity Relationship
13.
Retrovirology ; 11: 45, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24912525

ABSTRACT

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.


Subject(s)
Gene Products, vpr/metabolism , HIV-1/physiology , MAP Kinase Kinase Kinases/genetics , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Adaptor Proteins, Signal Transducing , Cell Line , Cell Line, Tumor , Gene Products, vpr/genetics , HEK293 Cells , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/genetics , Phosphorylation , Promoter Regions, Genetic , Signal Transduction , Transcription Factor AP-1/genetics , Ubiquitination , Virus Replication
14.
Retrovirology ; 11: 72, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25169827

ABSTRACT

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.


Subject(s)
Cytidine Deaminase/metabolism , HIV Infections/virology , HIV-1/genetics , Host Specificity/genetics , Introns , APOBEC-3G Deaminase , Cell Line , Cell Line, Tumor , Cytidine Deaminase/genetics , Gene Products, vpr/genetics , HEK293 Cells , HIV Infections/metabolism , HeLa Cells , Humans , Mutation/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Replication/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/genetics
15.
Retrovirology ; 10: 83, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23915234

ABSTRACT

BACKGROUND: Viral protein R (Vpr), a protein of human immunodeficiency virus type-1 (HIV-1) with various biological functions, was shown to be present in the blood of HIV-1-positive patients. However, it remained unclear whether circulating Vpr in patients' blood is biologically active. Here, we examined the activity of blood Vpr using an assay system by which retrotransposition of long interspersed element-1 (L1-RTP) was detected. We also investigated the in vivo effects of recombinant Vpr (rVpr) by administrating it to transgenic mice harboring human L1 as a transgene (hL1-Tg mice). Based on our data, we discuss the involvement of blood Vpr in the clinical symptoms of acquired immunodeficiency syndrome (AIDS). RESULTS: We first discovered that rVpr was active in induction of L1-RTP. Biochemical analyses revealed that rVpr-induced L1-RTP depended on the aryl hydrocarbon receptor, mitogen-activated protein kinases, and CCAAT/enhancer-binding protein ß. By using a sensitive L1-RTP assay system, we showed that 6 of the 15 blood samples from HIV-1 patients examined were positive for induction of L1-RTP. Of note, the L1-RTP-inducing activity was blocked by a monoclonal antibody specific for Vpr. Moreover, L1-RTP was reproducibly induced in various organs, including the kidney, when rVpr was administered to hL1-Tg mice. CONCLUSIONS: Blood Vpr is biologically active, suggesting that its monitoring is worthwhile for clarification of the roles of Vpr in the pathogenesis of AIDS. This is the first report to demonstrate a soluble factor in patients' blood active for L1-RTP activity, and implies the involvement of L1-RTP in the development of human diseases.


Subject(s)
Gene Products, vpr/blood , Gene Products, vpr/metabolism , HIV-1/enzymology , Long Interspersed Nucleotide Elements , Recombination, Genetic , Adult , Animals , Humans , Male , Mice , Mice, Transgenic , Young Adult
16.
Nat Cell Biol ; 8(2): 170-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16429131

ABSTRACT

The Vpr protein of HIV-1 functions as a vital accessory gene by regulating various cellular functions, including cell differentiation, apoptosis, nuclear factor of kappaB (NF-kappaB) suppression and cell-cycle arrest of the host cell. Several reports have indicated that Vpr complexes with the glucocorticoid receptor (GR), but it remains unclear whether the GR pathway is required for Vpr to function. Here, we report that Vpr uses the GR pathway as a recruitment vehicle for the NF-kappaB co-activating protein, poly(ADP-ribose) polymerase-1 (PARP-1). The GR interaction with Vpr is both necessary and sufficient to facilitate this interaction by potentiating the formation of a Vpr-GR-PARP-1 complex. The recruitment of PARP-1 by the Vpr-GR complex prevents its nuclear localization, which is necessary for Vpr to suppress NF-kappaB. The association of GR with PARP-1 is not observed with steroid (glucocorticoid) treatment, indicating that the GR association with PARP-1 is a gain of function that is solely attributed to HIV-1 Vpr. These data provide important insights into Vpr biology and its role in HIV pathogenesis.


Subject(s)
Cell Nucleus/metabolism , Gene Products, vpr/physiology , Poly(ADP-ribose) Polymerases/metabolism , Receptors, Glucocorticoid/metabolism , Active Transport, Cell Nucleus , Animals , Antigens, Bacterial/pharmacology , Cell Line , Chlorocebus aethiops , Enterotoxins/pharmacology , Female , Gene Expression/drug effects , Gene Expression/genetics , Gene Products, vpr/metabolism , Gene Products, vpr/pharmacology , HIV Infections/metabolism , HIV Infections/physiopathology , HeLa Cells , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Interleukin-1/blood , Interleukin-12/blood , Jurkat Cells , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Mutation/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding/drug effects , Protein Interaction Mapping , RNA, Small Interfering/genetics , Receptors, Glucocorticoid/genetics , Transcription Factor RelA/metabolism , Transfection , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells , vpr Gene Products, Human Immunodeficiency Virus
17.
Cell Microbiol ; 14(11): 1745-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22776683

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle , Gene Products, vpr/metabolism , Monomeric GTP-Binding Proteins/metabolism , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , Amino Acid Motifs , Animals , Binding Sites , Cell Line , Conserved Sequence , Gene Products, vpr/genetics , Humans , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins/genetics , vpr Gene Products, Human Immunodeficiency Virus/metabolism
18.
PLoS One ; 17(9): e0273313, 2022.
Article in English | MEDLINE | ID: mdl-36129874

ABSTRACT

HIV-associated nephropathy (HIVAN) impairs functions of both glomeruli and tubules. Attention has been previously focused on the HIVAN glomerulopathy. Tubular injury has drawn increased attention because sodium wasting is common in hospitalized HIV/AIDS patients. We used viral protein R (Vpr)-transgenic mice to investigate the mechanisms whereby Vpr contributes to urinary sodium wasting. In phosphoenolpyruvate carboxykinase promoter-driven Vpr-transgenic mice, in situ hybridization showed that Vpr mRNA was expressed in all nephron segments, including the distal convoluted tubule. Vpr-transgenic mice, compared with wild-type littermates, markedly increased urinary sodium excretion, despite similar plasma renin activity and aldosterone levels. Kidneys from Vpr-transgenic mice also markedly reduced protein abundance of the Na+-Cl- cotransporter (NCC), while mineralocorticoid receptor (MR) protein expression level was unchanged. In African green monkey kidney cells, Vpr abrogated the aldosterone-mediated stimulation of MR transcriptional activity. Gene expression of Slc12a3 (NCC) in Vpr-transgenic mice was significantly lower compared with wild-type mice, assessed by both qRT-PCR and RNAScope in situ hybridization analysis. Chromatin immunoprecipitation assays identified multiple MR response elements (MRE), located from 5 kb upstream of the transcription start site and extending to the third exon of the SLC12A3 gene. Mutation of MRE and SP1 sites in the SLC12A3 promoter region abrogated the transcriptional responses to aldosterone and Vpr, indicating that functional MRE and SP1 are required for the SLC12A3 gene suppression in response to Vpr. Thus, Vpr attenuates MR transcriptional activity and inhibits Slc12a3 transcription in the distal convoluted tubule and contributes to salt wasting in Vpr-transgenic mice.


Subject(s)
Gene Products, vpr , HIV-1 , Aldosterone/metabolism , Aldosterone/pharmacology , Animals , Chlorocebus aethiops , Gene Products, vpr/metabolism , HIV-1/genetics , Kidney Tubules, Distal/metabolism , Mice , Mice, Transgenic , Phosphoenolpyruvate , RNA, Messenger/metabolism , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Renin/metabolism , Sodium/metabolism , Sodium Chloride/metabolism , Sodium Chloride Symporters/metabolism , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Thiazides
19.
J Virol ; 84(3): 1585-96, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923179

ABSTRACT

During HIV-1 assembly, the viral protein R (Vpr) is incorporated into newly made viral particles via an interaction with the C-terminal domain of the Gag polyprotein precursor Pr55(Gag). Vpr has been implicated in the nuclear import of newly made viral DNA and subsequently in its transcription. In addition, Vpr can affect the cell physiology by causing G(2)/M cell cycle arrest and apoptosis. Vpr can form oligomers, but their roles have not yet been investigated. We have developed fluorescence lifetime imaging microscopy-fluorescence resonance energy transfer-based assays to monitor the interaction between Pr55(Gag) and Vpr in HeLa cells. To that end, we used enhanced green fluorescent protein-Vpr that can be incorporated into the virus and tetracysteine (TC)-tagged Pr55(Gag)-TC. This TC motif is tethered to the C terminus of Pr55(Gag) and does not interfere with Pr55(Gag) trafficking and the assembly of virus-like particles (VLPs). Results show that the Pr55(Gag)-Vpr complexes accumulated mainly at the plasma membrane. In addition, results with Pr55(Gag)-TC mutants confirm that the (41)LXXLF domain of Gag-p6 is essential for Pr55(Gag)-Vpr interaction. We also report that Vpr oligomerization is crucial for Pr55(Gag) recognition and its accumulation at the plasma membrane. On the other hand, Pr55(Gag)-Vpr complexes are still formed when Pr55(Gag) carries mutations impairing its multimerization. These findings suggest that Pr55(Gag)-Vpr recognition and complex formation occur early during Pr55(Gag) assembly.


Subject(s)
Gene Products, gag/metabolism , Gene Products, vpr/metabolism , HIV-1/metabolism , Apoptosis , Biopolymers , Cell Division , Cell Membrane/metabolism , G2 Phase , HeLa Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Protein Binding
20.
J Virol ; 84(20): 10907-12, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686015

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Gene Products, rev/immunology , Gene Products, vpr/immunology , Simian Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Gene Products, rev/genetics , Gene Products, vpr/genetics , Host-Pathogen Interactions/immunology , In Vitro Techniques , Kinetics , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL