Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Commun ; 12(1): 6864, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824204

ABSTRACT

HIV-1 Vpr is a prototypic member of a large family of structurally related lentiviral virulence factors that antagonize various aspects of innate antiviral immunity. It subverts host cell DNA repair and protein degradation machineries by binding and inhibiting specific post-replication repair enzymes, linking them via the DCAF1 substrate adaptor to the Cullin 4 RING E3 ligase (CRL4DCAF1). HIV-1 Vpr also binds to the multi-domain protein hHR23A, which interacts with the nucleotide excision repair protein XPC and shuttles ubiquitinated proteins to the proteasome. Here, we report the atomic resolution structure of Vpr in complex with the C-terminal half of hHR23A, containing the XPC-binding (XPCB) and ubiquitin-associated (UBA2) domains. The XPCB and UBA2 domains bind to different sides of Vpr's 3-helix-bundle structure, with UBA2 interacting with the α2 and α3 helices of Vpr, while the XPCB domain contacts the opposite side of Vpr's α3 helix. The structure as well as biochemical results reveal that hHR23A and DCAF1 use overlapping binding surfaces on Vpr, even though the two proteins exhibit entirely different three-dimensional structures. Our findings show that Vpr independently targets hHR23A- and DCAF1- dependent pathways and highlight HIV-1 Vpr as a versatile module that interferes with DNA repair and protein degradation pathways.


Subject(s)
DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , HIV-1/chemistry , vpr Gene Products, Human Immunodeficiency Virus/chemistry , Crystallography, X-Ray , DNA Repair , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism
2.
Proc Natl Acad Sci U S A ; 116(19): 9568-9577, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31019079

ABSTRACT

Lentiviruses, including HIV-1, possess the ability to enter the nucleus through nuclear pore complexes and can infect interphase cells, including those actively replicating chromosomal DNA. Viral accessory proteins hijack host cell E3 enzymes to antagonize intrinsic defenses, and thereby provide a more permissive environment for virus replication. The HIV-1 Vpr accessory protein reprograms CRL4DCAF1 E3 to antagonize select postreplication DNA repair enzymes and activates the DNA damage checkpoint in the G2 cell cycle phase. However, little is known about the roles played by these Vpr targets in HIV-1 replication. Here, using a sensitive pairwise replication competition assay, we show that Vpr endows HIV-1 with a strong replication advantage in activated primary CD4+ T cells and established T cell lines. This effect is disabled by a Vpr mutation that abolishes binding to CRL4DCAF1 E3, thereby disrupting Vpr antagonism of helicase-like transcription factor (HLTF) DNA helicase and other DNA repair pathway targets, and by another mutation that prevents induction of the G2 DNA damage checkpoint. Consistent with these findings, we also show that HLTF restricts HIV-1 replication, and that this restriction is antagonized by HIV-1 Vpr. Furthermore, our data imply that HIV-1 Vpr uses additional, yet to be identified mechanisms to facilitate HIV-1 replication in T cells. Overall, we demonstrate that multiple aspects of the cellular DNA repair machinery restrict HIV-1 replication in dividing T cells, the primary target of HIV-1 infection, and describe newly developed approaches to dissect key components.


Subject(s)
CD4-Positive T-Lymphocytes , DNA-Binding Proteins/metabolism , HIV Infections , HIV-1/physiology , Transcription Factors/metabolism , Virus Replication/physiology , vpr Gene Products, Human Immunodeficiency Virus/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , DNA-Binding Proteins/genetics , G2 Phase Cell Cycle Checkpoints , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/pathology , Humans , Transcription Factors/genetics , vpr Gene Products, Human Immunodeficiency Virus/genetics
3.
mBio ; 9(5)2018 10 23.
Article in English | MEDLINE | ID: mdl-30352932

ABSTRACT

Viral accessory proteins hijack host cell E3 ubiquitin ligases to antagonize innate/intrinsic defenses and thereby provide a more permissive environment for virus replication. Human immunodeficiency virus type 1 (HIV-1) accessory protein Vpr reprograms CRL4DCAF1 E3 to antagonize select postreplication DNA repair enzymes, but the significance and role of these Vpr interactions are poorly understood. To gain additional insights, we performed a focused screen for substrates of CRL4DCAF1 E3 reprogrammed by HIV-1 Vpr among known postreplication DNA repair proteins and identified exonuclease 1 (Exo1) as a novel direct HIV-1 Vpr target. We show that HIV-1 Vpr recruits Exo1 to the CRL4DCAF1 E3 complex for ubiquitination and subsequent proteasome-dependent degradation and that Exo1 levels are depleted in HIV-1-infected cells in a Vpr-dependent manner. We also show that Exo1 inhibits HIV-1 replication in T cells. Notably, the antagonism of Exo1 is a conserved function of main group HIV-1 and its ancestor Vpr proteins in the simian immunodeficiency virus from chimpanzee (SIVcpz) lineage, further underscoring the relevance of our findings. Overall, our studies (i) reveal that HIV-1 Vpr extensively remodels the cellular postreplication DNA repair machinery by impinging on multiple repair pathways, (ii) support a model in which Vpr promotes HIV-1 replication by antagonizing select DNA repair enzymes, and (iii) highlight the importance of a new class of restrictions placed on HIV-1 replication in T cells by the cellular DNA repair machinery.IMPORTANCE HIV-1 polymerase reverse transcribes the viral RNA genome into imperfectly double-stranded proviral DNA, containing gaps and flaps, for integration into the host cell chromosome. HIV-1 reverse transcripts share characteristics with cellular DNA replication intermediates and are thought to be converted into fully double-stranded DNA by cellular postreplication DNA repair enzymes. Therefore, the finding that the HIV-1 accessory protein Vpr antagonizes select postreplication DNA repair enzymes that can process HIV-1 reverse transcripts has been surprising. Here, we show that one such Vpr-antagonized enzyme, exonuclease 1, inhibits HIV-1 replication in T cells. We identify exonuclease 1 as a member of a new class of HIV-1 restriction factors in T cells and propose that certain modes of DNA "repair" inhibit HIV-1 infection.


Subject(s)
DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/metabolism , Host-Pathogen Interactions , Ubiquitin-Protein Ligases/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , DNA Repair , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/genetics , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/antagonists & inhibitors , Exodeoxyribonucleases/genetics , HIV Infections/enzymology , HIV Infections/genetics , HIV-1/genetics , HIV-1/physiology , Humans , T-Lymphocytes/virology , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus/genetics
4.
J Biol Chem ; 292(51): 21117-21127, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29079575

ABSTRACT

The viral protein R (Vpr) is an accessory virulence factor of HIV-1 that facilitates infection in immune cells. Cellular functions of Vpr are tied to its interaction with DCAF1, a substrate receptor component of the CRL4 E3 ubiquitin ligase. Recent proteomic approaches suggested that Vpr degrades helicase-like transcription factor (HLTF) DNA helicase in a proteasome-dependent manner by redirecting the CRL4-DCAF1 E3 ligase. However, the precise molecular mechanism of Vpr-dependent HLTF depletion is not known. Here, using in vitro reconstitution assays, we show that Vpr mediates polyubiquitination of HLTF, by directly loading it onto the C-terminal WD40 domain of DCAF1 in complex with the CRL4 E3 ubiquitin ligase. Mutational analyses suggest that Vpr interacts with DNA-binding residues in the N-terminal HIRAN domain of HLTF in a manner similar to the recruitment of another target, uracil DNA glycosylase (UNG2), to the CRL4-DCAF1 E3 by Vpr. Strikingly, Vpr also engages a second, adjacent region, which connects the HIRAN and ATPase/helicase domains. Thus, our findings reveal that Vpr utilizes common as well as distinctive interfaces to recruit multiple postreplication DNA repair proteins to the CRL4-DCAF1 E3 ligase for ubiquitin-dependent proteasomal degradation.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Models, Molecular , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Binding Sites , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Gene Deletion , HEK293 Cells , Humans , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Point Mutation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Serine-Threonine Kinases , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , WD40 Repeats , vpr Gene Products, Human Immunodeficiency Virus/chemistry , vpr Gene Products, Human Immunodeficiency Virus/genetics
5.
Proc Natl Acad Sci U S A ; 113(27): E3921-30, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27335459

ABSTRACT

HIV replication in nondividing host cells occurs in the presence of high concentrations of noncanonical dUTP, apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like 3 (APOBEC3) cytidine deaminases, and SAMHD1 (a cell cycle-regulated dNTP triphosphohydrolase) dNTPase, which maintains low concentrations of canonical dNTPs in these cells. These conditions favor the introduction of marks of DNA damage into viral cDNA, and thereby prime it for processing by DNA repair enzymes. Accessory protein Vpr, found in all primate lentiviruses, and its HIV-2/simian immunodeficiency virus (SIV) SIVsm paralogue Vpx, hijack the CRL4(DCAF1) E3 ubiquitin ligase to alleviate some of these conditions, but the extent of their interactions with DNA repair proteins has not been thoroughly characterized. Here, we identify HLTF, a postreplication DNA repair helicase, as a common target of HIV-1/SIVcpz Vpr proteins. We show that HIV-1 Vpr reprograms CRL4(DCAF1) E3 to direct HLTF for proteasome-dependent degradation independent from previously reported Vpr interactions with base excision repair enzyme uracil DNA glycosylase (UNG2) and crossover junction endonuclease MUS81, which Vpr also directs for degradation via CRL4(DCAF1) E3. Thus, separate functions of HIV-1 Vpr usurp CRL4(DCAF1) E3 to remove key enzymes in three DNA repair pathways. In contrast, we find that HIV-2 Vpr is unable to efficiently program HLTF or UNG2 for degradation. Our findings reveal complex interactions between HIV-1 and the DNA repair machinery, suggesting that DNA repair plays important roles in the HIV-1 life cycle. The divergent interactions of HIV-1 and HIV-2 with DNA repair enzymes and SAMHD1 imply that these viruses use different strategies to guard their genomes and facilitate their replication in the host.


Subject(s)
DNA Glycosylases/metabolism , DNA-Binding Proteins/metabolism , HIV-1/physiology , HIV-2/physiology , Transcription Factors/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Cell Cycle , DNA Helicases/metabolism , Endonucleases/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Monomeric GTP-Binding Proteins/metabolism , Proteomics , SAM Domain and HD Domain-Containing Protein 1 , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
6.
J Biol Chem ; 291(22): 11809-19, 2016 May 27.
Article in English | MEDLINE | ID: mdl-26994143

ABSTRACT

HIV-1 favors integration into active genes and gene-enriched regions of host cell chromosomes, thus maximizing the probability of provirus expression immediately after integration. This requires cleavage and polyadenylation specificity factor 6 (CPSF6), a cellular protein involved in pre-mRNA 3' end processing that binds HIV-1 capsid and connects HIV-1 preintegration complexes to intranuclear trafficking pathways that link integration to transcriptionally active chromatin. CPSF6 together with CPSF5 and CPSF7 are known subunits of the cleavage factor I (CFIm) 3' end processing complex; however, CPSF6 could participate in additional protein complexes. The molecular mechanisms underpinning the role of CPSF6 in HIV-1 infection remain to be defined. Here, we show that a majority of cellular CPSF6 is incorporated into the CFIm complex. HIV-1 capsid recruits CFIm in a CPSF6-dependent manner, which suggests that the CFIm complex mediates the known effects of CPSF6 in HIV-1 infection. To dissect the roles of CPSF6 and other CFIm complex subunits in HIV-1 infection, we analyzed virologic and integration site targeting properties of a CPSF6 variant with mutations that prevent its incorporation into CFIm We show, somewhat surprisingly, that CPSF6 incorporation into CFIm is not required for its ability to direct preferential HIV-1 integration into genes. The CPSF5 and CPSF7 subunits appear to have only a minor, if any, role in this process even though they appear to facilitate CPSF6 binding to capsid. Thus, CPSF6 alone controls the key molecular interactions that specify HIV-1 preintegration complex trafficking to active chromatin.


Subject(s)
Capsid/metabolism , HIV-1/physiology , RNA, Messenger/metabolism , Virus Integration , mRNA Cleavage and Polyadenylation Factors/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , Humans , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , mRNA Cleavage and Polyadenylation Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/genetics
7.
Biomed Res Int ; 2015: 739586, 2015.
Article in English | MEDLINE | ID: mdl-26504826

ABSTRACT

BACKGROUND: To investigate whether one or more SAMHD1 gene mutations are associated with cerebrovascular disease in the general population using a Chinese stroke cohort. METHODS: Patients with a Chinese Han background (N = 300) diagnosed with either cerebral large-artery atherosclerosis (LAA, n = 100), cerebral small vessel disease (SVD, n = 100), or other stroke-free neurological disorders (control, n = 100) were recruited. Genomic DNA from the whole blood of each patient was isolated, and direct sequencing of the SAMHD1 gene was performed. Both wild type and mutant SAMHD1 proteins identified from the patients were expressed in E. coli and purified; then their dNTPase activities and ability to form stable tetramers were analysed in vitro. RESULTS: Three heterozygous mutations, including two missense mutations c.64C>T (P22S) and c.841G>A (p.E281K) and one splice site mutation c.696+2T>A, were identified in the LAA group with a prevalence of 3%. No mutations were found in the patients with SVD or the controls (p = 0.05). The mutant SAMHD1 proteins were functionally impaired in terms of their catalytic activity as a dNTPase and ability to assemble stable tetramers. CONCLUSIONS: Heterozygous SAMHD1 gene mutations might cause genetic predispositions that interact with other risk factors, resulting in increased vulnerability to stroke.


Subject(s)
Atherosclerosis/genetics , Genetic Predisposition to Disease/genetics , Monomeric GTP-Binding Proteins/genetics , Stroke/genetics , Aged , Asian People/genetics , Atherosclerosis/epidemiology , Cerebral Arteries/diagnostic imaging , China/epidemiology , Cohort Studies , DNA Mutational Analysis , Female , Genetic Predisposition to Disease/epidemiology , Humans , Male , Middle Aged , Mutation/genetics , Radiography , SAM Domain and HD Domain-Containing Protein 1 , Stroke/epidemiology
8.
J Biol Chem ; 290(21): 13279-92, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25847232

ABSTRACT

SAMHD1 is a nuclear deoxyribonucleoside triphosphate triphosphohydrolase that contributes to the control of cellular deoxyribonucleoside triphosphate (dNTP) pool sizes through dNTP hydrolysis and modulates the innate immune response to viruses. CyclinA2-CDK1/2 phosphorylates SAMHD1 at Thr-592, but how this modification controls SAMHD1 functions in proliferating cells is not known. Here, we show that SAMHD1 levels remain relatively unchanged during the cell division cycle in primary human T lymphocytes and in monocytic cell lines. Inactivation of the bipartite cyclinA2-CDK-binding site in the SAMHD1 C terminus described herein abolished SAMHD1 phosphorylation on Thr-592 during S and G2 phases thus interfering with DNA replication and progression of cells through S phase. The effects exerted by Thr-592 phosphorylation-defective SAMHD1 mutants were associated with activation of DNA damage checkpoint and depletion of dNTP concentrations to levels lower than those seen upon expression of wild type SAMHD1 protein. These disruptive effects were relieved by either mutation of the catalytic residues of the SAMHD1 phosphohydrolase domain or by a Thr-592 phosphomimetic mutation, thus linking the Thr-592 phosphorylation state to the control of SAMHD1 dNTPase activity. Our findings support a model in which phosphorylation of Thr-592 by cyclinA2-CDK down-modulates, but does not inactivate, SAMHD1 dNTPase in S phase, thereby fine-tuning SAMHD1 control of dNTP levels during DNA replication.


Subject(s)
Cell Cycle/physiology , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/metabolism , Monocytes/metabolism , Monomeric GTP-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Amino Acid Motifs , Amino Acid Sequence , Blotting, Western , CDC2 Protein Kinase , Cells, Cultured , Cyclin A2/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinases/genetics , HEK293 Cells , Humans , Hydrolysis , Immunoenzyme Techniques , Immunoprecipitation , Molecular Sequence Data , Monocytes/cytology , Monomeric GTP-Binding Proteins/genetics , Mutation/genetics , Phosphorylation , Protein Structure, Tertiary , SAM Domain and HD Domain-Containing Protein 1 , Sequence Homology, Amino Acid , T-Lymphocytes/cytology
9.
PLoS One ; 9(1): e84463, 2014.
Article in English | MEDLINE | ID: mdl-24465411

ABSTRACT

Analysis of rhesus macaques infected with a vpx deletion mutant virus of simian immunodeficiency virus mac239 (SIVΔvpx) demonstrates that Vpx is essential for efficient monocyte/macrophage infection in vivo but is not necessary for development of AIDS. To compare myeloid-lineage cell infection in monkeys infected with SIVΔvpx compared to SIVmac239, we analyzed lymphoid and gastrointestinal tissues from SIVΔvpx-infected rhesus (n = 5), SIVmac239-infected rhesus with SIV encephalitis (7 SIV239E), those without encephalitis (4 SIV239noE), and other SIV mutant viruses with low viral loads (4 SIVΔnef, 2 SIVΔ3). SIV+ macrophages and the percentage of total SIV+ cells that were macrophages in spleen and lymph nodes were significantly lower in rhesus infected with SIVΔvpx (2.2%) compared to those infected with SIV239E (22.7%), SIV239noE (8.2%), and SIV mutant viruses (10.1%). In colon, SIVΔvpx monkeys had fewer SIV+ cells, no SIV+ macrophages, and lower percentage of SIV+ cells that were macrophages than the other 3 groups. Only 2 SIVΔvpx monkeys exhibited detectable virus in the colon. We demonstrate that Vpx is essential for efficient macrophage infection in vivo and that simian AIDS and death can occur in the absence of detectable macrophage infection.


Subject(s)
Encephalitis, Viral/virology , Macrophages/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics , Amino Acid Sequence , Animals , Encephalitis, Viral/mortality , Encephalitis, Viral/pathology , Gene Deletion , Gene Expression , Lymph Nodes/pathology , Lymph Nodes/virology , Macaca mulatta , Macrophages/pathology , Molecular Sequence Data , Monocytes/pathology , Monocytes/virology , Simian Acquired Immunodeficiency Syndrome/mortality , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/pathogenicity , Spleen/pathology , Spleen/virology , Survival Analysis , Viral Load , Viral Regulatory and Accessory Proteins/deficiency , Virus Replication
10.
Nat Struct Mol Biol ; 20(11): 1304-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24141705

ABSTRACT

SAMHD1, a dNTP triphosphohydrolase (dNTPase), has a key role in human innate immunity. It inhibits infection of blood cells by retroviruses, including HIV, and prevents the development of the autoinflammatory Aicardi-Goutières syndrome (AGS). The inactive apo-SAMHD1 interconverts between monomers and dimers, and in the presence of dGTP the protein assembles into catalytically active tetramers. Here, we present the crystal structure of the human tetrameric SAMHD1-dGTP complex. The structure reveals an elegant allosteric mechanism of activation through dGTP-induced tetramerization of two inactive dimers. Binding of dGTP to four allosteric sites promotes tetramerization and induces a conformational change in the substrate-binding pocket to yield the catalytically active enzyme. Structure-based biochemical and cell-based biological assays confirmed the proposed mechanism. The SAMHD1 tetramer structure provides the basis for a mechanistic understanding of its function in HIV restriction and the pathogenesis of AGS.


Subject(s)
Allosteric Regulation , Deoxyguanine Nucleotides/chemistry , Deoxyguanine Nucleotides/metabolism , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/metabolism , Protein Multimerization , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , SAM Domain and HD Domain-Containing Protein 1
11.
PLoS Pathog ; 9(7): e1003496, 2013.
Article in English | MEDLINE | ID: mdl-23874202

ABSTRACT

SAMHD1 is a host restriction factor that blocks the ability of lentiviruses such as HIV-1 to undergo reverse transcription in myeloid cells and resting T-cells. This restriction is alleviated by expression of the lentiviral accessory proteins Vpx and Vpr (Vpx/Vpr), which target SAMHD1 for proteasome-mediated degradation. However, the precise determinants within SAMHD1 for recognition by Vpx/Vpr remain unclear. Here we show that evolution of Vpx/Vpr in primate lentiviruses has caused the interface between SAMHD1 and Vpx/Vpr to alter during primate lentiviral evolution. Using multiple HIV-2 and SIV Vpx proteins, we show that Vpx from the HIV-2 and SIVmac lineage, but not Vpx from the SIVmnd2 and SIVrcm lineage, require the C-terminus of SAMHD1 for interaction, ubiquitylation, and degradation. On the other hand, the N-terminus of SAMHD1 governs interactions with Vpx from SIVmnd2 and SIVrcm, but has little effect on Vpx from HIV-2 and SIVmac. Furthermore, we show here that this difference in SAMHD1 recognition is evolutionarily dynamic, with the importance of the N- and C-terminus for interaction of SAMHD1 with Vpx and Vpr toggling during lentiviral evolution. We present a model to explain how the head-to-tail conformation of SAMHD1 proteins favors toggling of the interaction sites by Vpx/Vpr during this virus-host arms race. Such drastic functional divergence within a lentiviral protein highlights a novel plasticity in the evolutionary dynamics of viral antagonists for restriction factors during lentiviral adaptation to its hosts.


Subject(s)
Evolution, Molecular , HIV-2/metabolism , Host-Pathogen Interactions , Models, Biological , Monomeric GTP-Binding Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , Cercopithecinae , Databases, Protein , HEK293 Cells , HIV-2/genetics , Humans , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phylogeny , Proteasome Endopeptidase Complex , Protein Interaction Domains and Motifs , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Ubiquitination , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics , vpr Gene Products, Human Immunodeficiency Virus/chemistry
12.
J Biol Chem ; 288(15): 10406-17, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23426366

ABSTRACT

SAMHD1 is a dGTP-activated dNTPase that has been implicated as a modulator of the innate immune response. In monocytes and their differentiated derivatives, as well as in quiescent cells, SAMHD1 strongly inhibits HIV-1 infection and, to a lesser extent, HIV-2 and simian immunodeficiency virus (SIV) because of their virion-associated virulence factor Vpx, which directs SAMHD1 for proteasomal degradation. Here, we used a combination of biochemical and virologic approaches to gain insights into the functional organization of human SAMHD1. We found that the catalytically active recombinant dNTPase is a dGTP-induced tetramer. Chemical cross-linking studies revealed SAMHD1 tetramers in human monocytic cells, in which it strongly restricts HIV-1 infection. The propensity of SAMHD1 to maintain the tetrameric state in vitro is regulated by its C terminus, located outside of the catalytic domain. Accordingly, we show that the C terminus is required for the full ability of SAMHD1 to deplete dNTP pools and to inhibit HIV-1 infection in U937 monocytes. Interestingly, the human SAMHD1 C terminus contains a docking site for HIV-2/SIVmac Vpx and is known to have evolved under positive selection. This evidence indicates that Vpx targets a functionally important element in SAMHD1. Together, our findings imply that SAMHD1 tetramers are the biologically active form of this dNTPase and provide new insights into the functional organization of SAMHD1.


Subject(s)
HIV Infections/enzymology , HIV-1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Protein Multimerization , HIV Infections/genetics , HIV-1/genetics , HIV-2/genetics , HIV-2/metabolism , Humans , Monomeric GTP-Binding Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Proteolysis , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , U937 Cells
13.
J Biol Chem ; 287(15): 12550-8, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22362772

ABSTRACT

The sterile alpha motif and HD domain-containing protein-1 (SAMHD1) inhibits infection of myeloid cells by human and related primate immunodeficiency viruses (HIV and SIV). This potent inhibition is counteracted by the Vpx accessory virulence factor of HIV-2/SIVsm viruses, which targets SAMHD1 for proteasome-dependent degradation, by reprogramming cellular CRL4(DCAF1) E3 ubiquitin ligase. However, the precise mechanism of Vpx-dependent recruitment of human SAMHD1 onto the ligase, and the molecular interfaces on the respective molecules have not been defined. Here, we show that human SAMHD1 is recruited to the CRL4(DCAF1-Vpx) E3 ubiquitin ligase complex by interacting with the DCAF1 substrate receptor subunit in a Vpx-dependent manner. No stable association is detectable with DCAF1 alone. The SAMHD1 determinant for the interaction is a short peptide located distal to the SAMHD1 catalytic domain and requires the presence of Vpx for stable engagement. This peptide is sufficient to confer Vpx-dependent recruitment to CRL4(DCAF1) and ubiquitination when fused to heterologous proteins. The precise amino acid sequence of the peptide diverges among SAMHD1 proteins from different vertebrate species, explaining selective down-regulation of human SAMHD1 levels by Vpx. Critical amino acid residues of SAMHD1 and Vpx involved in the DCAF1-Vpx-SAMDH1 interaction were identified by mutagenesis. Our findings show that the N terminus of Vpx, bound to DCAF1, recruits SAMHD1 via its C terminus to CRL4, in a species-specific manner for proteasomal degradation.


Subject(s)
Carrier Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Simian Immunodeficiency Virus/physiology , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Conserved Sequence , DNA-Binding Proteins/metabolism , Down-Regulation , HEK293 Cells , Host-Pathogen Interactions , Humans , Molecular Sequence Data , Monomeric GTP-Binding Proteins/genetics , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases , Recombinant Fusion Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1
14.
Nature ; 474(7353): 658-61, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21720370

ABSTRACT

Macrophages and dendritic cells have key roles in viral infections, providing virus reservoirs that frequently resist antiviral therapies and linking innate virus detection to antiviral adaptive immune responses. Human immunodeficiency virus 1 (HIV-1) fails to transduce dendritic cells and has a reduced ability to transduce macrophages, due to an as yet uncharacterized mechanism that inhibits infection by interfering with efficient synthesis of viral complementary DNA. In contrast, HIV-2 and related simian immunodeficiency viruses (SIVsm/mac) transduce myeloid cells efficiently owing to their virion-associated Vpx accessory proteins, which counteract the restrictive mechanism. Here we show that the inhibition of HIV-1 infection in macrophages involves the cellular SAM domain HD domain-containing protein 1 (SAMHD1). Vpx relieves the inhibition of lentivirus infection in macrophages by loading SAMHD1 onto the CRL4(DCAF1) E3 ubiquitin ligase, leading to highly efficient proteasome-dependent degradation of the protein. Mutations in SAMHD1 cause Aicardi-Goutières syndrome, a disease that produces a phenotype that mimics the effects of a congenital viral infection. Failure to dispose of endogenous nucleic acid debris in Aicardi-Goutières syndrome results in inappropriate triggering of innate immune responses via cytosolic nucleic acids sensors. Thus, our findings show that macrophages are defended from HIV-1 infection by a mechanism that prevents an unwanted interferon response triggered by self nucleic acids, and uncover an intricate relationship between innate immune mechanisms that control response to self and to retroviral pathogens.


Subject(s)
HIV Infections/physiopathology , HIV-1/physiology , Macrophages/virology , Monomeric GTP-Binding Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Carrier Proteins/metabolism , Cells, Cultured , HEK293 Cells , HIV Infections/metabolism , Humans , Macrophages/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases , SAM Domain and HD Domain-Containing Protein 1 , Ubiquitin-Protein Ligases/metabolism
15.
PLoS Pathog ; 4(5): e1000059, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18464893

ABSTRACT

Vpx is a small virion-associated adaptor protein encoded by viruses of the HIV-2/SIVsm lineage of primate lentiviruses that enables these viruses to transduce monocyte-derived cells. This probably reflects the ability of Vpx to overcome an as yet uncharacterized block to an early event in the virus life cycle in these cells, but the underlying mechanism has remained elusive. Using biochemical and proteomic approaches, we have found that Vpx protein of the pathogenic SIVmac 239 strain associates with a ternary protein complex comprising DDB1 and VprBP subunits of Cullin 4-based E3 ubiquitin ligase, and DDA1, which has been implicated in the regulation of E3 catalytic activity, and that Vpx participates in the Cullin 4 E3 complex comprising VprBP. We further demonstrate that the ability of SIVmac as well as HIV-2 Vpx to interact with VprBP and its associated Cullin 4 complex is required for efficient reverse transcription of SIVmac RNA genome in primary macrophages. Strikingly, macrophages in which VprBP levels are depleted by RNA interference resist SIVmac infection. Thus, our observations reveal that Vpx interacts with both catalytic and regulatory components of the ubiquitin proteasome system and demonstrate that these interactions are critical for Vpx ability to enable efficient SIVmac replication in primary macrophages. Furthermore, they identify VprBP/DCAF1 substrate receptor for Cullin 4 E3 ubiquitin ligase and its associated protein complex as immediate downstream effector of Vpx for this function. Together, our findings suggest a model in which Vpx usurps VprBP-associated Cullin 4 ubiquitin ligase to enable efficient reverse transcription and thereby overcome a block to lentivirus replication in monocyte-derived cells, and thus provide novel insights into the underlying molecular mechanism.


Subject(s)
Cullin Proteins/antagonists & inhibitors , Fluoresceins/metabolism , Gene Products, vpr/metabolism , Macrophages/virology , Simian Immunodeficiency Virus/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD4-Positive T-Lymphocytes/virology , Cell Line , Cullin Proteins/metabolism , Gene Expression Regulation, Viral , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Signal Transduction , Simian Immunodeficiency Virus/pathogenicity , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/chemistry
16.
J Biol Chem ; 283(6): 3088-3096, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18056264

ABSTRACT

Recognition of cognate Rho GTPases by guanine-nucleotide exchange factors (GEF) is fundamental to Rho GTPase signaling specificity. Two main GEF families use either the Dbl homology (DH) or the DOCK homology region 2 (DHR-2) catalytic domain. How DHR-2-containing GEFs distinguish between the GTPases Rac and Cdc42 is not known. To determine how these GEFs specifically recognize the two Rho GTPases, we studied the amino acid sequences in Rac2 and Cdc42 that are crucial for activation by DOCK2, a Rac-specific GEF, and DOCK9, a distantly related Cdc42-specific GEF. Two elements in the N-terminal regions of Rac2 and Cdc42 were found to be essential for specific interactions with DOCK2 and DOCK9. One element consists of divergent amino acid residues in the switch 1 regions of the GTPases. Significantly, these residues were also found to be important for GTPase recognition by Rac-specific DOCK180, DOCK3, and DOCK4 GEFs. These findings were unexpected because the same residues were shown previously to interact with GTPase effectors rather than GEFs. The other element comprises divergent residues in the beta3 strand that are known to mediate specific recognition by DH domain containing GEFs. Remarkably, Rac2-to-Cdc42 substitutions of four of these residues were sufficient for Rac2 to be specifically activated by DOCK9. Thus, DOCK2 and DOCK9 specifically recognize Rac2 and Cdc42 through their switch 1 as well as beta2-beta3 regions and the mode of recognition via switch 1 appears to be conserved among diverse Rac-specific DHR-2 GEFs.


Subject(s)
Gene Expression Regulation , Guanine Nucleotide Exchange Factors/chemistry , cdc42 GTP-Binding Protein/chemistry , rac GTP-Binding Proteins/chemistry , Amino Acid Sequence , GTPase-Activating Proteins , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Molecular Sequence Data , Point Mutation , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , rac GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
17.
Proc Natl Acad Sci U S A ; 104(28): 11778-83, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17609381

ABSTRACT

The replication of viruses depends on the cell cycle status of the infected cells. Viruses have evolved functions that alleviate restrictions imposed on their replication by the host. Vpr, an accessory factor of primate lentiviruses, arrests cells at the DNA damage checkpoint in G2 phase of the cell cycle, but the mechanism underlying this effect has remained elusive. Here we report that Vpr proteins of both the human (HIV-1) and the distantly related simian (SIVmac) immunodeficiency viruses specifically associate with a protein complex comprising subunits of E3 ubiquitin ligase assembled on Cullin-4 scaffold (Cul4-DDB1[VprBP]). We show that Vpr binding to Cul4-DDB1[VprBP] leads to increased neddylation and elevated intrinsic ubiquitin ligase activity of this E3. This effect is mediated through the VprBP subunit of the complex, which recently has been suggested to function as a substrate receptor for Cul4. We also demonstrate that VprBP regulates G1 phase and is essential for the completion of DNA replication in S phase. Furthermore, the ability of Vpr to arrest cells in G2 phase correlates with its ability to interact with Cul4-DDB1[VprBP] E3 complex. Our studies identify the Cul4-DDB1[VprBP] E3 ubiquitin ligase complex as the downstream effector of lentiviral Vpr for the induction of cell cycle arrest in G2 phase and suggest that Vpr may use this complex to perturb other aspects of the cell cycle and DNA metabolism in infected cells.


Subject(s)
Cell Cycle/physiology , Cullin Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Products, vpr/physiology , HIV-1/physiology , Simian Immunodeficiency Virus/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , COP9 Signalosome Complex , Carrier Proteins/metabolism , Cell Line , Eukaryotic Initiation Factors/metabolism , G2 Phase/physiology , Gene Products, vpr/metabolism , Humans , Protein Binding , Protein Subunits/physiology , vpr Gene Products, Human Immunodeficiency Virus
18.
J Virol ; 80(9): 4469-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16611907

ABSTRACT

Point mutations in SIVmac239 Nef disrupting CD4 downmodulation and enhancement of virion infectivity attenuate viral replication in acutely infected rhesus macaques, but changes selected later in infection fully restore Nef function (A. J. Iafrate et al., J. Virol. 74:9836-9844, 2000). To further evaluate the relevance of these Nef functions for viral persistence and disease progression, we analyzed an SIVmac239 Nef mutant containing a deletion of amino acids Q64 to N67 (delta64-67Nef). This mutation inactivates the N-distal AP-2 clathrin adaptor binding element and disrupts the abilities of Nef to downregulate CD4, CD28 and CXCR4 and to stimulate viral replication in vitro. However, it does not impair the downmodulation of CD3 and class I major histocompatibility complex (MHC-I) or MHC-II and the upregulation of the MHC-II-associated invariant chain, and it has only a moderate effect on the enhancement of virion infectivity. Replication of the delta64-67Nef variant in acutely infected macaques was intermediate between grossly nef-deleted and wild-type SIVmac239. Subsequently, three of six macaques developed moderate to high viral loads and developed disease, whereas the remaining animals efficiently controlled SIV replication and showed a more attenuated clinical course of infection. Sequence analysis revealed that the deletion in nef was not repaired in any of these animals. However, some changes that slightly enhanced the ability of Nef to downmodulate CD4 and moderately increased Nef-mediated enhancement of viral replication and infectivity in vitro were observed in macaques developing high viral loads. Our results imply that both the Nef functions that were disrupted by the delta64-67 mutation and the activities that remained intact contribute to viral pathogenicity.


Subject(s)
Adaptor Protein Complex 2/metabolism , Gene Products, nef/chemistry , Gene Products, nef/metabolism , Macaca mulatta/virology , Simian Immunodeficiency Virus/physiology , Simian Immunodeficiency Virus/pathogenicity , Virus Replication , Acute Disease , Alleles , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Gene Products, nef/genetics , Genetic Variation/genetics , Humans , Lentivirus Infections/genetics , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Macaca mulatta/genetics , Molecular Sequence Data , Protein Binding , Sequence Alignment , Time Factors
19.
Methods Enzymol ; 406: 593-605, 2006.
Article in English | MEDLINE | ID: mdl-16472690

ABSTRACT

The breakthrough discovery that double-stranded RNA of 21 nucleotides in length (referred to as short or small interfering RNA; siRNA) can trigger sequence-specific gene silencing in mammalian cells has led to the development of a powerful new approach to study gene function (Dillon et al., 2005; Dykxhoorn et al., 2003; Elbashir et al., 2001; Hannon et al., 2004). Effective delivery of siRNA molecules into target cells or tissues is critical for successful RNA interference (RNAi) application. Here, we describe the use of human immunodeficiency virus type 1 (HIV-1)-based lentiviral vectors for delivery of short hairpin RNA (shRNA), a precursor of siRNA, into primary neurons to suppress gene expression. Major advantages of lentiviral vectors are their ability to transduce nondividing cells and to confer long-term expression of transgenes. This chapter covers selection of short hairpin sequences, vector design, production of lentiviral supernatants, transduction of dissociated primary hippocampal neurons, and testing the effectiveness of shRNA-mediated silencing.


Subject(s)
Gene Silencing , Genetic Vectors , Neurons/drug effects , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Animals , Cloning, Molecular , Female , Hippocampus/cytology , Hippocampus/embryology , Lentivirus , Rats
20.
J Virol ; 79(16): 10650-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051857

ABSTRACT

Nef proteins of primate lentiviruses promote viral replication, virion infectivity, and evasion of antiviral immune responses by modulating signal transduction pathways and downregulating expression of receptors at the cell surface that are important for efficient antigen-specific responses, such as CD4, CD28, T-cell antigen receptor, and class I and class II major histocompatibility complex. Here we show that Nef proteins from diverse groups of primate lentiviruses which do not require the chemokine receptor CXCR4 for entry into target cells strongly downmodulate the cell surface expression of CXCR4. In contrast, all human immunodeficiency virus type 1 (HIV-1) and the majority of HIV-2 Nef proteins tested did not have such strong effects. SIVmac239 Nef strongly inhibited lymphocyte migration to CXCR4 ligand, the chemokine stromal derived factor 1 (SDF-1). SIVmac239 Nef downregulated CXCR4 by accelerating the rate of its endocytosis. Downmodulation of CXCR4 was abolished by mutations that disrupt the constitutively strong AP-2 clathrin adaptor binding element located in the N-terminal region of the Nef molecule, suggesting that Nef accelerates CXCR4 endocytosis via an AP-2-dependent pathway. Together, these results point to CXCR4 as playing an important role in simian immunodeficiency virus and possibly also HIV-2 persistence in vivo that is unrelated to viral entry into target cells. We speculate that Nef targets CXCR4 to disrupt ordered trafficking of infected leukocytes between local microenvironments in order to facilitate their dissemination and/or impair the antiviral immune response.


Subject(s)
Chemokines, CXC/physiology , Gene Products, nef/physiology , HIV/physiology , Receptors, CXCR4/antagonists & inhibitors , Simian Immunodeficiency Virus/physiology , CD4 Antigens/physiology , Cell Movement , Chemokine CXCL12 , Down-Regulation , Endocytosis , Gene Products, nef/chemistry , Humans , Jurkat Cells , Receptors, CXCR4/physiology , T-Lymphocytes/physiology , nef Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...