Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nat Immunol ; 13(3): 223-228, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327569

ABSTRACT

SAMHD1 restricts the infection of dendritic and other myeloid cells by human immunodeficiency virus type 1 (HIV-1), but in lentiviruses of the simian immunodeficiency virus of sooty mangabey (SIVsm)-HIV-2 lineage, SAMHD1 is counteracted by the virion-packaged accessory protein Vpx. Here we found that SAMHD1 restricted infection by hydrolyzing intracellular deoxynucleoside triphosphates (dNTPs), lowering their concentrations to below those required for the synthesis of the viral DNA by reverse transcriptase (RT). SAMHD1-mediated restriction was alleviated by the addition of exogenous deoxynucleosides. An HIV-1 with a mutant RT with low affinity for dNTPs was particularly sensitive to SAMHD1-mediated restriction. Vpx prevented the SAMHD1-mediated decrease in dNTP concentration and induced the degradation of human and rhesus macaque SAMHD1 but had no effect on mouse SAMHD1. Nucleotide-pool depletion could be a general mechanism for protecting cells from infectious agents that replicate through a DNA intermediate.


Subject(s)
HIV-1/physiology , Monomeric GTP-Binding Proteins/metabolism , Nucleotides/metabolism , Virus Replication , Animals , Cell Line , Humans , Intracellular Space/metabolism , Macaca mulatta , Macrophages/immunology , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/immunology , SAM Domain and HD Domain-Containing Protein 1
2.
PLoS Pathog ; 17(10): e1009609, 2021 10.
Article in English | MEDLINE | ID: mdl-34699574

ABSTRACT

Human Immunodeficiency viruses type 1 and 2 (HIV-1 and HIV-2) succeed to evade host immune defenses by using their viral auxiliary proteins to antagonize host restriction factors. HIV-2/SIVsmm Vpx is known for degrading SAMHD1, a factor impeding the reverse transcription. More recently, Vpx was also shown to counteract HUSH, a complex constituted of TASOR, MPP8 and periphilin, which blocks viral expression from the integrated viral DNA. In a classical ubiquitin ligase hijacking model, Vpx bridges the DCAF1 ubiquitin ligase substrate adaptor to SAMHD1, for subsequent ubiquitination and degradation. Here, we investigated whether the same mechanism is at stake for Vpx-mediated HUSH degradation. While we confirm that Vpx bridges SAMHD1 to DCAF1, we show that TASOR can interact with DCAF1 in the absence of Vpx. Nonetheless, this association was stabilized in the presence of Vpx, suggesting the existence of a ternary complex. The N-terminal PARP-like domain of TASOR is involved in DCAF1 binding, but not in Vpx binding. We also characterized a series of HIV-2 Vpx point mutants impaired in TASOR degradation, while still degrading SAMHD1. Vpx mutants ability to degrade TASOR correlated with their capacity to enhance HIV-1 minigenome expression as expected. Strikingly, several Vpx mutants impaired for TASOR degradation, but not for SAMHD1 degradation, had a reduced binding affinity for DCAF1, but not for TASOR. In macrophages, Vpx R34A-R42A and Vpx R42A-Q47A-V48A, strongly impaired in DCAF1, but not in TASOR binding, could not degrade TASOR, while being efficient in degrading SAMHD1. Altogether, our results highlight the central role of a robust Vpx-DCAF1 association to trigger TASOR degradation. We then propose a model in which Vpx interacts with both TASOR and DCAF1 to stabilize a TASOR-DCAF1 complex. Furthermore, our work identifies Vpx mutants enabling the study of HUSH restriction independently from SAMHD1 restriction in primary myeloid cells.


Subject(s)
HIV Infections/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Cell Line , HIV-2 , Humans
3.
Retrovirology ; 19(1): 23, 2022 10 29.
Article in English | MEDLINE | ID: mdl-36309692

ABSTRACT

BACKGROUND: TASOR, a component of the HUSH repressor epigenetic complex, and SAMHD1, a cellular triphosphohydrolase (dNTPase), are both anti-HIV proteins antagonized by HIV-2/SIVsmm Viral protein X. As a result, the same viral protein is able to relieve two different blocks along the viral life cell cycle, one at the level of reverse transcription, by degrading SAMHD1, the other one at the level of proviral expression, by degrading TASOR. Phosphorylation of SAMHD1 at T592 has been shown to downregulate its antiviral activity. The discovery that T819 in TASOR was lying within a SAMHD1 T592-like motif led us to ask whether TASOR is phosphorylated on this residue and whether this post-translational modification could regulate its repressive activity. RESULTS: Using a specific anti-phospho-antibody, we found that TASOR is phosphorylated at T819, especially in cells arrested in early mitosis by nocodazole. We provide evidence that the phosphorylation is conducted by a Cyclin/CDK1 complex, like that of SAMHD1 at T592. While we could not detect TASOR in quiescent CD4 + T cells, TASOR and its phosphorylated form are present in activated primary CD4 + T lymphocytes. In addition, TASOR phosphorylation appears to be independent from TASOR repressive activity. Indeed, on the one hand, nocodazole barely reactivates HIV-1 in the J-Lat A1 HIV-1 latency model despite TASOR T819 phosphorylation. On the other hand, etoposide, a second cell cycle arresting drug, reactivates latent HIV-1, without concomitant TASOR phosphorylation. Furthermore, overexpression of wt TASOR or T819A or T819E similarly represses gene expression driven by an HIV-1-derived LTR promoter. Finally, while TASOR is degraded by HIV-2 Vpx, TASOR phosphorylation is prevented by HIV-1 Vpr, likely as a consequence of HIV-1 Vpr-mediated-G2 arrest. CONCLUSIONS: Altogether, we show that TASOR phosphorylation occurs in vivo on T819. This event does not appear to correlate with TASOR-mediated HIV-1 silencing. We speculate that TASOR phosphorylation is related to a role of TASOR during cell cycle progression.


Subject(s)
HIV Infections , HIV-1 , Monomeric GTP-Binding Proteins , Humans , SAM Domain and HD Domain-Containing Protein 1/metabolism , HIV-1/physiology , Phosphorylation , Threonine , Nocodazole/metabolism , Virus Latency , Viral Regulatory and Accessory Proteins/metabolism , Nuclear Proteins/metabolism
4.
J Virol ; 94(8)2020 03 31.
Article in English | MEDLINE | ID: mdl-32024775

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) Tax oncoprotein is required for viral gene expression. Tax transactivates the viral promoter by recruiting specific transcription factors but also by interfering with general transcription factors involved in the preinitiation step, such as TFIIA and TFIID. However, data are lacking regarding Tax interplay with TFIIH, which intervenes during the last step of preinitiation. We previously reported that XPB, the TFIIH subunit responsible for promoter opening and promoter escape, is required for Tat-induced human-immunodeficiency virus promoter transactivation. Here, we investigated whether XPB may also play a role in HTLV-1 transcription. We report that Tax and XPB directly interact in vitro and that endogenous XPB produced by HTLV-1-infected T cells binds to Tax and is recruited on proviral LTRs. In contrast, XPB recruitment at the LTR is not detected in Tax-negative HTLV-1-infected T cells and is strongly reduced when Tax-induced HTLV-1 LTR transactivation is blocked. XPB overexpression does not affect basal HTLV-1 promoter activation but enhances Tax-mediated transactivation in T cells. Conversely, downregulating XPB strongly reduces Tax-mediated transactivation. Importantly, spironolactone (SP)-mediated inhibition of LTR activation can be rescued by overexpressing XPB but not XPD, another TFIIH subunit. Furthermore, an XPB mutant defective for the ATPase activity responsible for promoter opening does not show rescue of the effect of SP. Finally, XPB downregulation reduces viability of Tax-positive but not Tax-negative HTLV-1-transformed T cell lines. These findings reveal that XPB is a novel cellular cofactor hijacked by Tax to facilitate HTLV-1 transcription.IMPORTANCE HTLV-1 is considered the most potent human oncovirus and is also responsible for severe inflammatory disorders. HTLV-1 transcription is undertaken by RNA polymerase II and is controlled by the viral oncoprotein Tax. Tax transactivates the viral promoter first via the recruitment of CREB and its cofactors to the long terminal repeat (LTR). However, how Tax controls subsequent steps of the transcription process remains unclear. In this study, we explore the link between Tax and the XPB subunit of TFIIH that governs, via its ATPase activity, the promoter-opening step of transcription. We demonstrate that XPB is a novel physical and functional partner of Tax, recruited on HTLV-1 LTR, and required for viral transcription. These findings extend the mechanism of Tax transactivation to the recruitment of TFIIH and reinforce the link between XPB and transactivator-induced viral transcription.


Subject(s)
Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/physiology , Trans-Activators/metabolism , Transcription Factor TFIIH/metabolism , Gene Expression Regulation, Viral , Gene Products, tax/metabolism , HEK293 Cells , HTLV-I Infections/virology , Humans , Promoter Regions, Genetic , Terminal Repeat Sequences , Transcription Factors/metabolism , Transcription, Genetic , Virus Replication
5.
PLoS Pathog ; 15(5): e1007669, 2019 05.
Article in English | MEDLINE | ID: mdl-31042779

ABSTRACT

HIV-1 is dependent on the host cell for providing the metabolic resources for completion of its viral replication cycle. Thus, HIV-1 replicates efficiently only in activated CD4+ T cells. Barriers preventing HIV-1 replication in resting CD4+ T cells include a block that limits reverse transcription and also the lack of activity of several inducible transcription factors, such as NF-κB and NFAT. Because FOXO1 is a master regulator of T cell functions, we studied the effect of its inhibition on T cell/HIV-1 interactions. By using AS1842856, a FOXO1 pharmacologic inhibitor, we observe that FOXO1 inhibition induces a metabolic activation of T cells with a G0/G1 transition in the absence of any stimulatory signal. One parallel outcome of this change is the inhibition of the activity of the HIV restriction factor SAMHD1 and the activation of the NFAT pathway. FOXO1 inhibition by AS1842856 makes resting T cells permissive to HIV-1 infection. In addition, we found that FOXO1 inhibition by either AS1842856 treatment or upon FOXO1 knockdown induces the reactivation of HIV-1 latent proviruses in T cells. We conclude that FOXO1 has a central role in the HIV-1/T cell interaction and that inhibiting FOXO1 with drugs such as AS1842856 may be a new therapeutic shock-and-kill strategy to eliminate the HIV-1 reservoir in human T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Box Protein O1/antagonists & inhibitors , Gene Expression Regulation , HIV Infections/virology , HIV-1/immunology , Virus Activation/immunology , Virus Replication , Animals , CD4-Positive T-Lymphocytes/virology , Cell Cycle , Forkhead Box Protein O1/genetics , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/metabolism , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Macaca fascicularis , Male , Virus Latency
6.
Proc Natl Acad Sci U S A ; 113(19): 5311-6, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114546

ABSTRACT

Viruses often interfere with the DNA damage response to better replicate in their hosts. The human immunodeficiency virus 1 (HIV-1) viral protein R (Vpr) protein has been reported to modulate the activity of the DNA repair structure-specific endonuclease subunit (SLX4) complex and to promote cell cycle arrest. Vpr also interferes with the base-excision repair pathway by antagonizing the uracil DNA glycosylase (Ung2) enzyme. Using an unbiased quantitative proteomic screen, we report that Vpr down-regulates helicase-like transcription factor (HLTF), a DNA translocase involved in the repair of damaged replication forks. Vpr subverts the DDB1-cullin4-associated-factor 1 (DCAF1) adaptor of the Cul4A ubiquitin ligase to trigger proteasomal degradation of HLTF. This event takes place rapidly after Vpr delivery to cells, before and independently of Vpr-mediated G2 arrest. HLTF is degraded in lymphocytic cells and macrophages infected with Vpr-expressing HIV-1. Our results reveal a previously unidentified strategy for HIV-1 to antagonize DNA repair in host cells.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Macrophages/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Cells, Cultured , HeLa Cells , Humans , vpr Gene Products, Human Immunodeficiency Virus
7.
J Virol ; 90(23): 10972-10980, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27681137

ABSTRACT

Tat protein, the HIV transactivator, regulates transcription of the HIV genome by the host transcription machinery. Efficient inhibitors of HIV transcription that target Tat or the cellular cofactor NF-κB are well known. However, inhibition of HIV Tat-dependent transcription by targeting the general transcription and DNA repair factor II human (TFIIH) has not been reported. Here, we show that spironolactone (SP), an aldosterone antagonist approved for clinical use, inhibits HIV-1 and HIV-2 infection of permissive T cells by blocking viral Tat-dependent transcription from the long terminal repeat (LTR). We found that treatment of Jurkat and primary CD4+ T cells with SP induces degradation of the XPB cellular helicase, a component of the TFIIH complex, without affecting cellular mRNA levels, T cell viability, or T cell proliferation. We further demonstrate that the effect of SP on HIV infection is independent of its aldosterone antagonist function, since the structural analogue, eplerenone, does not induce XPB degradation and does not inhibit HIV infection. Rescue experiments showed that the SP-induced block of HIV infection relies, at least partially, on XPB degradation. In addition, we demonstrate that SP specifically inhibits Tat-dependent transcription, since basal transcription from the LTR is not affected. Our results demonstrate that SP is a specific inhibitor of HIV Tat-dependent transcription in T cells, which additionally suggests that XPB is a cofactor required for HIV infection. Targeting a cellular cofactor of HIV transcription constitutes an alternative strategy to inhibit HIV infection, together with the existing antiretroviral therapy. IMPORTANCE: Transcription from the HIV promoter is regulated by the combined activities of the host transcription machinery and the viral transactivator Tat protein. Here, we report that the drug spironolactone-an antagonist of aldosterone-blocks viral Tat-dependent transcription, thereby inhibiting both HIV-1 and HIV-2 infection of permissive T cells. This inhibition relies on the degradation of the cellular helicase XPB, a component of the TFIIH transcription factor complex. Consequently, XPB appears to be a novel HIV cofactor. Our discovery of the HIV-inhibitory activity of spironolactone opens the way for the development of novel anti-HIV strategies targeting a cellular cofactor without the limitations of antiretroviral therapy of drug resistance and high cost.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/prevention & control , Spironolactone/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-2/drug effects , Humans , Jurkat Cells , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors
8.
Proc Natl Acad Sci U S A ; 110(42): E3997-4006, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082141

ABSTRACT

Macrophages are a major target cell for HIV-1, and their infection contributes to HIV pathogenesis. We have previously shown that the cyclin-dependent kinase inhibitor p21 inhibits the replication of HIV-1 and other primate lentiviruses in human monocyte-derived macrophages by impairing reverse transcription of the viral genome. In the attempt to understand the p21-mediated restriction mechanisms, we found that p21 impairs HIV-1 and simian immunodeficiency virus (SIV)mac reverse transcription in macrophages by reducing the intracellular deoxyribonucleotide (dNTP) pool to levels below those required for viral cDNA synthesis by a SAM domain and HD domain-containing protein 1 (SAMHD1)-independent pathway. We found that p21 blocks dNTP biosynthesis by down-regulating the expression of the RNR2 subunit of ribonucleotide reductase, an enzyme essential for the reduction of ribonucleotides to dNTP. p21 inhibits RNR2 transcription by repressing E2F1 transcription factor, its transcriptional activator. Our findings unravel a cellular pathway that restricts HIV-1 and other primate lentiviruses by affecting dNTP synthesis, thereby pointing to new potential cellular targets for anti-HIV therapeutic strategies.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Deoxyribonucleotides/biosynthesis , Gene Expression Regulation, Enzymologic , HIV Infections/metabolism , HIV-1/physiology , Macrophages/metabolism , Ribonucleotide Reductases/biosynthesis , Virus Replication/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , DNA, Viral/biosynthesis , DNA, Viral/genetics , Deoxyribonucleotides/genetics , Down-Regulation/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , HIV Infections/therapy , HIV Infections/virology , Macrophages/virology , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Ribonucleotide Reductases/genetics , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/physiology , Transcription, Genetic/genetics
9.
Virol J ; 12: 201, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26606981

ABSTRACT

BACKGROUND: SAMHD1 counteracts HIV-1 or HIV-2/SIVsmm that lacks Vpx by depleting the intracellular pool of nucleotides in myeloid cells and CD4+ quiescent T cells, thereby inhibiting the synthesis of retroviral DNA by reverse transcriptase. Depletion of nucleotides has been shown to underline the establishment of quiescence in certain cellular systems. These observations led us to investigate whether SAMHD1 could control the transition between proliferation and quiescence using the THP-1 cell model. FINDINGS: The entry of dividing THP-1 myeloid cells into a non-dividing differentiated state was monitored after addition of phorbol-12-myristate-13-acetate (PMA), an inducer of differentiation. Under PMA treatment, cells overexpressing SAMHD1 display stronger and faster adhesion to their support, compared to cells expressing a catalytically inactive form of SAMHD1, or cells depleted of SAMHD1, which appear less differentiated. After PMA removal, cells overexpressing SAMHD1 maintain low levels of cyclin A, in contrast to other cell lines. Interestingly, SAMHD1 overexpression slightly increases cell adhesion even in the absence of the differentiation inducer PMA. Finally, we found that levels of SAMHD1 are reduced in proliferating primary CD4+ T cells after T cell receptor activation, suggesting that SAMHD1 may also be involved in the transition from a quiescent state to a dividing state in primary T cells. CONCLUSIONS: Altogether, we provide evidence that SAMHD1 may facilitate some aspects of THP-1 cell differentiation. Restriction of HIV-1 by SAMHD1 may rely upon its ability to modify cell cycle parameters, in addition to the direct inhibition of reverse transcription.


Subject(s)
Cell Differentiation , Cell Proliferation , Monocytes/physiology , Monomeric GTP-Binding Proteins/metabolism , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , Cell Adhesion/drug effects , Cell Line , HIV-1/immunology , HIV-1/physiology , Humans , Monocytes/drug effects , SAM Domain and HD Domain-Containing Protein 1 , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/metabolism , Virus Replication
10.
Retrovirology ; 11: 117, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25496524

ABSTRACT

Vpr is one of the most enigmatic viral auxiliary proteins of HIV. During the past twenty years, several activities have been ascribed to this viral protein, but one, its ability to mediate cell cycle arrest at the G2 to M transition has been the most extensively studied. Nonetheless, the genuine role of Vpr and its pathophysiological relevance in the viral life cycle have remained mysterious. Recent work by Laguette et al. (Cell 156:134-145, 2014) provides important insight into the molecular mechanism of Vpr-mediated G2 arrest. This study highlights for the first time how Vpr recruits the SLX4 endonuclease complex and how Vpr-induced inappropriate activation of this complex leads to G2 arrest. Here, we will discuss these findings in the light of previous work to show how they change the view of Vpr's mechanism of action. We will also discuss how these findings open new questions towards the understanding of the biological function of Vpr regarding innate immune sensing.


Subject(s)
Cell Cycle Checkpoints , HIV-1/physiology , Host-Pathogen Interactions , Recombinases/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Humans
11.
Retrovirology ; 10: 30, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23497353

ABSTRACT

BACKGROUND: Interferon-α (IFN-α) is an essential mediator of the antiviral response, which potently inhibits both early and late phases of HIV replication. The SAMHD1 deoxynucleoside triphosphate (dNTP) hydrolase represents the prototype of a new antiviral strategy we referred to as "nucleotide depletion". SAMHD1 depletes dNTP levels in myeloid cells below those required for optimal synthesis of HIV viral DNA. HIV-2 and its SIVsm and SIVmac close relatives encode a protein termed Vpx, which counteracts SAMHD1. The potentiality of IFN-α to cooperate with nucleotide depletion has been poorly investigated so far. Here we wondered whether IFN-α affects SAMHD1 expression, Vpx-induced SAMHD1 degradation, Vpx-mediated rescue of HIV-1 transduction and the dNTP supply in monocyte-derived macrophages (MDMs). RESULTS: IFN-α inhibited HIV-1 transduction in monocytes and in MDMs while SAMHD1 expression was not up-regulated. Vpx triggered SAMHD1 degradation in IFN-α treated cells, and weakly restored HIV-1 transduction from the IFN-α block. Vpx helper effect towards HIV-1 transduction was gradually inhibited with increasing doses of IFN-α. dNTP levels were not significantly affected in MDMs and CD4+ primary activated T lymphocytes by IFN-α and, in correlation with SAMHD1 degradation, restoration of dNTP levels by Vpx was efficient in MDMs treated with the cytokine. In contrast, IFN-α inhibited Vpx-mediated SAMHD1 degradation in THP-1 cells, where, accordingly, Vpx could not rescue HIV-1 transduction. CONCLUSION: Our results suggest that the early antiviral effect of IFN-α results from a mechanism independent of nucleotide depletion in MDMs. In addition, they indicate that the macrophage-like THP-1 cell line may provide a system to characterize an IFN-α-induced cell response that inhibits Vpx-mediated SAMHD1 degradation.


Subject(s)
HIV-1/genetics , Interferon-alpha/immunology , Macrophages/immunology , Macrophages/virology , Monomeric GTP-Binding Proteins/immunology , Transduction, Genetic , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Humans , Monomeric GTP-Binding Proteins/metabolism , Nucleotides/metabolism , Proteolysis , SAM Domain and HD Domain-Containing Protein 1 , Viral Regulatory and Accessory Proteins/metabolism
12.
J Biol Chem ; 286(27): 23742-52, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21566118

ABSTRACT

Vpr, a small HIV auxiliary protein, hijacks the CUL4 ubiquitin ligase through DCAF1 to inactivate an unknown cellular target, leading to cell cycle arrest at the G(2) phase and cell death. Here we first sought to delineate the Vpr determinants involved in the binding to DCAF1 and to the target. On the one hand, the three α-helices of Vpr are necessary and sufficient for binding to DCAF1; on the other hand, nonlinear determinants in Vpr are required for binding to the target, as shown by using protein chimeras. We also underscore that a SRIG motif conserved in the C-terminal tail of Vpr proteins from HIV-1/SIVcpz and HIV-2/SIVsmm lineages is critical for G(2) arrest. Our results suggest that this motif may be predictive of the ability of Vpr proteins from other SIV lineages to mediate G(2) arrest. We took advantage of the characterization of a subset of G(2) arrest-defective, but DCAF1 binding-proficient mutants, to investigate whether Vpr interferes with cell viability independently of its ability to induce G(2) arrest. These mutants inhibited cell colony formation in HeLa cells and are cytotoxic in lymphocytes, unmasking a G(2) arrest-independent cytopathic effect of Vpr. Furthermore these mutants do not block cell cycle progression at the G(1) or S phases but trigger apoptosis through caspase 3. Disruption of DCAF1 binding restored efficiency of colony formation. However, DCAF1 binding per se is not sufficient to confer cytopathicity. These data support a model in which Vpr recruits DCAF1 to induce the degradation of two host proteins independently required for proper cell growth.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle , HIV-1/metabolism , Models, Biological , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Motifs , Carrier Proteins/genetics , Cell Death/genetics , Cullin Proteins/genetics , Cullin Proteins/metabolism , HEK293 Cells , HIV-1/genetics , HeLa Cells , Humans , Mutation , Protein Serine-Threonine Kinases , Ubiquitin-Protein Ligases , vpr Gene Products, Human Immunodeficiency Virus/genetics
14.
Nat Commun ; 13(1): 66, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013187

ABSTRACT

The Human Silencing Hub (HUSH) complex constituted of TASOR, MPP8 and Periphilin recruits the histone methyl-transferase SETDB1 to spread H3K9me3 repressive marks across genes and transgenes in an integration site-dependent manner. The deposition of these repressive marks leads to heterochromatin formation and inhibits gene expression, but the underlying mechanism is not fully understood. Here, we show that TASOR silencing or HIV-2 Vpx expression, which induces TASOR degradation, increases the accumulation of transcripts derived from the HIV-1 LTR promoter at a post-transcriptional level. Furthermore, using a yeast 2-hybrid screen, we identify new TASOR partners involved in RNA metabolism including the RNA deadenylase CCR4-NOT complex scaffold CNOT1. TASOR and CNOT1 synergistically repress HIV expression from its LTR. Similar to the RNA-induced transcriptional silencing complex found in fission yeast, we show that TASOR interacts with the RNA exosome and RNA Polymerase II, predominantly under its elongating state. Finally, we show that TASOR facilitates the association of RNA degradation proteins with RNA polymerase II and is detected at transcriptional centers. Altogether, we propose that HUSH operates at the transcriptional and post-transcriptional levels to repress HIV proviral expression.


Subject(s)
Epigenetic Repression , HIV-2/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Stability , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin Assembly and Disassembly , Gene Expression , Gene Silencing , HIV Infections/virology , HIV Long Terminal Repeat , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Phosphoproteins , Proviruses/genetics , RNA Polymerase II/metabolism , Schizosaccharomyces
15.
J Biol Chem ; 285(7): 4273-7, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20028970

ABSTRACT

Clusterin (CLU) is a ubiquitous protein that has been implicated in tumorigenesis, apoptosis, inflammation, and cell proliferation. We and others have previously shown that CLU is an inhibitor of the NF-kappaB pathway. However, the exact form of CLU and the region(s) of CLU involved in this effect were unknown. Using newly generated molecular constructs encoding for CLU and various regions of the molecule, we demonstrated that the presecretory form of CLU (psCLU) form bears the NF-kappaB regulatory activity. Sequence comparison analysis showed sequence motif identity between CLU and beta-transducin repeat-containing protein (beta-TrCP), a main E3 ubiquitin ligase involved in IkappaB-alpha degradation. These homologies were localized in the disulfide constraint region of CLU. We generated a specific molecular construct of this region, named DeltaCLU, and showed that it has the same NF-kappaB regulatory activity as CLU. Neither the alpha-chain nor the beta-chain of CLU had any NF-kappaB regulatory activity. Furthermore, we showed that following tumor necrosis factor-alpha stimulation of transfected cells, we could co-immunoprecipitate phospho-IkappaB-alpha with DeltaCLU. Moreover, we showed that DeltaCLU could localize both in the cytoplasm and in the nucleus. These results demonstrate the identification of a new CLU activity site involved in NF-kappaB pathway regulation.


Subject(s)
Clusterin/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Nucleus/metabolism , Clusterin/genetics , Cytoplasm/metabolism , Gene Expression/drug effects , HeLa Cells , Humans , I-kappa B Proteins/metabolism , Immunoprecipitation , Microscopy, Confocal , NF-KappaB Inhibitor alpha , Protein Binding , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/pharmacology
16.
J Biol Chem ; 285(23): 17390-7, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20378537

ABSTRACT

The COP9 signalosome (CSN) is an essential multisubunit complex that regulates the activity of cullin-RING ubiquitin ligases by removing the ubiquitin-like peptide NEDD8 from cullins. Here, we demonstrate that the CSN can affect other components of the ubiquitination cascade. Down-regulation of human CSN4 or CSN5 induced proteasome-mediated degradation of the ubiquitin-conjugating enzyme UBC3/Cdc34. UBC3 was targeted for ubiquitination by the cullin-RING ubiquitin ligase SCF(betaTrCP). This interaction required the acidic C-terminal extension of UBC3, which is absent in ubiquitin-conjugating enzymes of the UBCH5 family. Conversely, the UBC3 acidic domain was sufficient to impart sensitivity to SCF(betaTrCP)-mediated ubiquitination to UBCH5 enzymes. Our work indicates that the CSN is necessary to ensure the stability of selected ubiquitin-conjugating enzymes and uncovers a novel pathway of regulation of ubiquitination processes.


Subject(s)
Multiprotein Complexes/metabolism , Peptide Hydrolases/metabolism , Transducin/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , COP9 Signalosome Complex , Cell Line , Down-Regulation , HeLa Cells , Humans , Lentivirus/metabolism , Oligonucleotides/chemistry , Peptide Hydrolases/chemistry , Plasmids/metabolism , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity , Ubiquitin/chemistry , Ubiquitin-Conjugating Enzymes
17.
PLoS Pathog ; 5(5): e1000450, 2009 May.
Article in English | MEDLINE | ID: mdl-19478868

ABSTRACT

The interferon-induced transmembrane protein BST-2/CD317 (tetherin) restricts the release of diverse enveloped viruses from infected cells. The HIV-1 accessory protein Vpu antagonizes this restriction by an unknown mechanism that likely involves the down-regulation of BST-2 from the cell surface. Here, we show that the optimal removal of BST-2 from the plasma membrane by Vpu requires the cellular protein beta-TrCP, a substrate adaptor for a multi-subunit SCF E3 ubiquitin ligase complex and a known Vpu-interacting protein. beta-TrCP is also required for the optimal enhancement of virion-release by Vpu. Mutations in the DSGxxS beta-TrCP binding-motif of Vpu impair both the down-regulation of BST-2 and the enhancement of virion-release. Such mutations also confer dominant-negative activity, consistent with a model in which Vpu links BST-2 to beta-TrCP. Optimal down-regulation of BST-2 from the cell surface by Vpu also requires the endocytic clathrin adaptor AP-2, although the rate of endocytosis is not increased; these data suggest that Vpu induces post-endocytic membrane trafficking events whose net effect is the removal of BST-2 from the cell surface. In addition to its marked effect on cell-surface levels, Vpu modestly decreases the total cellular levels of BST-2. The decreases in cell-surface and intracellular BST-2 are inhibited by bafilomycin A1, an inhibitor of endosomal acidification; these data suggest that Vpu induces late endosomal targeting and partial degradation of BST-2 in lysosomes. The Vpu-mediated decrease in surface expression is associated with reduced co-localization of BST-2 and the virion protein Gag along the plasma membrane. Together, the data support a model in which Vpu co-opts the beta-TrCP/SCF E3 ubiquitin ligase complex to induce endosomal trafficking events that remove BST-2 from its site of action as a virion-tethering factor.


Subject(s)
HIV-1/pathogenicity , Human Immunodeficiency Virus Proteins/physiology , Lysosomes/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/physiology , beta-Transducin Repeat-Containing Proteins/metabolism , Adaptor Protein Complex 2 , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line , Down-Regulation , Endosomes/metabolism , GPI-Linked Proteins , HIV-1/physiology , Human Immunodeficiency Virus Proteins/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mutation , Protein Transport , Viral Regulatory and Accessory Proteins/genetics , beta-Transducin Repeat-Containing Proteins/genetics
18.
Nat Commun ; 12(1): 4582, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321470

ABSTRACT

SAMHD1 is a cellular triphosphohydrolase (dNTPase) proposed to inhibit HIV-1 reverse transcription in non-cycling immune cells by limiting the supply of the dNTP substrates. Yet, phosphorylation of T592 downregulates SAMHD1 antiviral activity, but not its dNTPase function, implying that additional mechanisms contribute to viral restriction. Here, we show that SAMHD1 is SUMOylated on residue K595, a modification that relies on the presence of a proximal SUMO-interacting motif (SIM). Loss of K595 SUMOylation suppresses the restriction activity of SAMHD1, even in the context of the constitutively active phospho-ablative T592A mutant but has no impact on dNTP depletion. Conversely, the artificial fusion of SUMO2 to a non-SUMOylatable inactive SAMHD1 variant restores its antiviral function, a phenotype that is reversed by the phosphomimetic T592E mutation. Collectively, our observations clearly establish that lack of T592 phosphorylation cannot fully account for the restriction activity of SAMHD1. We find that SUMOylation of K595 is required to stimulate a dNTPase-independent antiviral activity in non-cycling immune cells, an effect that is antagonized by cyclin/CDK-dependent phosphorylation of T592 in cycling cells.


Subject(s)
Cell Cycle/physiology , HIV-1/physiology , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism , Sumoylation/physiology , Amino Acid Substitution , HEK293 Cells , HIV Infections/virology , Humans , Lysine , Mutation , Phosphorylation , SAM Domain and HD Domain-Containing Protein 1/chemistry , U937 Cells
19.
Retrovirology ; 7: 35, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20380700

ABSTRACT

HIV viruses encode a set of accessory proteins, which are important determinants of virulence due to their ability to manipulate the host cell physiology for the benefit of the virus. Although these viral proteins are dispensable for viral growth in many in vitro cell culture systems, they influence the efficiency of viral replication in certain cell types. Macrophages are early targets of HIV infection which play a major role in viral dissemination and persistence in the organism. This review focuses on two HIV accessory proteins whose functions might be more specifically related to macrophage infection: Vpr, which is conserved across primate lentiviruses including HIV-1 and HIV-2, and Vpx, a protein genetically related to Vpr, which is unique to HIV-2 and a subset of simian lentiviruses. Recent studies suggest that both Vpr and Vpx exploit the host ubiquitination machinery in order to inactivate specific cellular proteins. We review here why it remains difficult to decipher the role of Vpr in macrophage infection by HIV-1 and how recent data underscore the ability of Vpx to antagonize a restriction factor which counteracts synthesis of viral DNA in monocytic cells.


Subject(s)
HIV/pathogenicity , Macrophages/immunology , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/physiology , Virulence Factors/physiology , vpr Gene Products, Human Immunodeficiency Virus/physiology , Animals , HIV/immunology , Host-Pathogen Interactions , Humans , Macrophages/virology , Primates , Ubiquitination , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/immunology , vpr Gene Products, Human Immunodeficiency Virus/immunology
20.
J Virol ; 83(10): 4854-60, 2009 May.
Article in English | MEDLINE | ID: mdl-19264781

ABSTRACT

The human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) genomes encode several auxiliary proteins that have increasingly shown their importance in the virus-host relationship. One of these proteins, Vpx, is unique to the HIV-2/SIVsm lineage and is critical for viral replication in macrophages. The functional basis for this requirement, as well as the Vpx mode of action, has remained unexplained, and it is all the more enigmatic that HIV type 1 (HIV-1), which has no Vpx counterpart, can infect macrophages. Here, we underscore DCAF1 as a critical host effector of Vpx in its ability to mediate infection and long-term replication of HIV-2 in human macrophages. Vpx assembles with the CUL4A-DDB1 ubiquitin ligase through DCAF1 recruitment. Precluding Vpx present in the incoming virions from recruiting DCAF1 in target macrophages leads to a postentry block characterized by defective accumulation of HIV-2 reverse transcripts. In addition, Vpx from SIVsm functionally complements Vpx-defective HIV-2 in a DCAF1-binding-dependent manner. Altogether, our data point to a mechanism in which Vpx diverts the Cul4A-DDB1(DCAF1) ligase to inactivate an evolutionarily conserved factor, which restricts macrophage infection by HIV-2 and closely related simian viruses.


Subject(s)
HIV Infections/virology , HIV-2/metabolism , Macrophages/virology , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/metabolism , CD4-Positive T-Lymphocytes/virology , Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Gene Silencing , HIV-2/genetics , HIV-2/physiology , HeLa Cells , Humans , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL