Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 67(7): e0046223, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37310224

ABSTRACT

HIV-1 integrase-LEDGF allosteric inhibitors (INLAIs) share the binding site on the viral protein with the host factor LEDGF/p75. These small molecules act as molecular glues promoting hyper-multimerization of HIV-1 IN protein to severely perturb maturation of viral particles. Herein, we describe a new series of INLAIs based on a benzene scaffold that display antiviral activity in the single digit nanomolar range. Akin to other compounds of this class, the INLAIs predominantly inhibit the late stages of HIV-1 replication. A series of high-resolution crystal structures revealed how these small molecules engage the catalytic core and the C-terminal domains of HIV-1 IN. No antagonism was observed between our lead INLAI compound BDM-2 and a panel of 16 clinical antiretrovirals. Moreover, we show that compounds retained high antiviral activity against HIV-1 variants resistant to IN strand transfer inhibitors and other classes of antiretroviral drugs. The virologic profile of BDM-2 and the recently completed single ascending dose phase I trial (ClinicalTrials.gov identifier: NCT03634085) warrant further clinical investigation for use in combination with other antiretroviral drugs. Moreover, our results suggest routes for further improvement of this emerging drug class.


Subject(s)
HIV Infections , HIV Integrase Inhibitors , HIV Integrase , Humans , Virus Replication , HIV Integrase Inhibitors/pharmacology , HIV Integrase Inhibitors/therapeutic use , Antiviral Agents/pharmacology , HIV Integrase/metabolism , HIV Infections/drug therapy , Allosteric Regulation
2.
J Med Chem ; 62(9): 4742-4754, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30995398

ABSTRACT

Penicillin-binding proteins (PBPs) are the targets of the ß-lactams, the most successful class of antibiotics ever developed against bacterial infections. Unfortunately, the worldwide and rapid spread of large spectrum ß-lactam resistance genes such as carbapenemases is detrimental to the use of antibiotics in this class. New potent PBP inhibitors are needed, especially compounds that resist ß-lactamase hydrolysis. Here we describe the structure of the E. coli PBP2 in its Apo form and upon its reaction with 2 diazabicyclo derivatives, avibactam and CPD4, a new potent PBP2 inhibitor. Examination of these structures shows that unlike avibactam, CPD4 can perform a hydrophobic stacking on Trp370 in the active site of E. coli PBP2. This result, together with sequence analysis, homology modeling, and SAR, allows us to propose CPD4 as potential starting scaffold to develop molecules active against a broad range of bacterial species at the top of the WHO priority list.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/drug effects , Penicillin-Binding Proteins/antagonists & inhibitors , Amino Acid Sequence , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/metabolism , Catalytic Domain , Drug Design , Escherichia coli/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Ligands , Microbial Sensitivity Tests , Molecular Structure , Penicillin-Binding Proteins/isolation & purification , Penicillin-Binding Proteins/metabolism , Protein Binding , Pseudomonas aeruginosa/drug effects , Sequence Alignment , Structure-Activity Relationship
3.
J Biol Chem ; 293(16): 6172-6186, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29507092

ABSTRACT

Recently, a new class of HIV-1 integrase (IN) inhibitors with a dual mode of action, called IN-LEDGF/p75 allosteric inhibitors (INLAIs), was described. Designed to interfere with the IN-LEDGF/p75 interaction during viral integration, unexpectedly, their major impact was on virus maturation. This activity has been linked to induction of aberrant IN multimerization, whereas inhibition of the IN-LEDGF/p75 interaction accounts for weaker antiretroviral effect at integration. Because these dual activities result from INLAI binding to IN at a single binding site, we expected that these activities co-evolved together, driven by the affinity for IN. Using an original INLAI, MUT-A, and its activity on an Ala-125 (A125) IN variant, we found that these two activities on A125-IN can be fully dissociated: MUT-A-induced IN multimerization and the formation of eccentric condensates in viral particles, which are responsible for inhibition of virus maturation, were lost, whereas inhibition of the IN-LEDGF/p75 interaction and consequently integration was fully retained. Hence, the mere binding of INLAI to A125 IN is insufficient to promote the conformational changes of IN required for aberrant multimerization. By analyzing the X-ray structures of MUT-A bound to the IN catalytic core domain (CCD) with or without the Ala-125 polymorphism, we discovered that the loss of IN multimerization is due to stabilization of the A125-IN variant CCD dimer, highlighting the importance of the CCD dimerization energy for IN multimerization. Our study reveals that affinity for the LEDGF/p75-binding pocket is not sufficient to induce INLAI-dependent IN multimerization and the associated inhibition of viral maturation.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/drug effects , HIV-1/physiology , Virus Assembly/drug effects , Virus Integration/drug effects , Allosteric Regulation , Binding Sites , Cell Line , HIV Integrase Inhibitors/chemistry , Humans , Molecular Structure , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacology
4.
Retrovirology ; 14(1): 50, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29121950

ABSTRACT

BACKGROUND: HIV-1 Integrase (IN) interacts with the cellular co-factor LEDGF/p75 and tethers the HIV preintegration complex to the host genome enabling integration. Recently a new class of IN inhibitors was described, the IN-LEDGF allosteric inhibitors (INLAIs). Designed to interfere with the IN-LEDGF interaction during integration, the major impact of these inhibitors was surprisingly found on virus maturation, causing a reverse transcription defect in target cells. RESULTS: Here we describe the MUT-A compound as a genuine INLAI with an original chemical structure based on a new type of scaffold, a thiophene ring. MUT-A has all characteristics of INLAI compounds such as inhibition of IN-LEDGF/p75 interaction, IN multimerization, dual antiretroviral (ARV) activities, normal packaging of genomic viral RNA and complete Gag protein maturation. MUT-A has more potent ARV activity compared to other INLAIs previously reported, but similar profile of resistance mutations and absence of ARV activity on SIV. HIV-1 virions produced in the presence of MUT-A were non-infectious with the formation of eccentric condensates outside of the core. In studying the immunoreactivity of these non-infectious virions, we found that inactivated HIV-1 particles were captured by anti-HIV-specific neutralizing and non-neutralizing antibodies (b12, 2G12, PGT121, 4D4, 10-1074, 10E8, VRC01) with efficiencies comparable to non-treated virus. Autologous CD4+ T lymphocyte proliferation and cytokine induction by monocyte-derived dendritic cells (MDDC) pulsed either with MUT-A-inactivated HIV or non-treated HIV were also comparable. CONCLUSIONS: Although strongly defective in infectivity, HIV-1 virions produced in the presence of the MUT-A INLAI have a normal protein and genomic RNA content as well as B and T cell immunoreactivities comparable to non-treated HIV-1. These inactivated viruses might form an attractive new approach in vaccine research in an attempt to study if this new type of immunogen could elicit an immune response against HIV-1 in animal models.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , HIV-1/enzymology , Intercellular Signaling Peptides and Proteins/metabolism , Pyridines/pharmacology , Thiophenes/pharmacology , Cell Line , HIV Antibodies/immunology , HIV Integrase Inhibitors/chemistry , HIV-1/immunology , Humans , Pyridines/chemistry , Thiophenes/chemistry , Virus Assembly/drug effects , Virus Integration/drug effects , Virus Replication/drug effects
5.
PLoS One ; 9(7): e103552, 2014.
Article in English | MEDLINE | ID: mdl-25072705

ABSTRACT

The viral integrase (IN) is an essential protein for HIV-1 replication. IN inserts the viral dsDNA into the host chromosome, thereby aided by the cellular co-factor LEDGF/p75. Recently a new class of integrase inhibitors was described: allosteric IN inhibitors (ALLINIs). Although designed to interfere with the IN-LEDGF/p75 interaction to block HIV DNA integration during the early phase of HIV-1 replication, the major impact was surprisingly found on the process of virus maturation during the late phase, causing a reverse transcription defect upon infection of target cells. Virus particles produced in the presence of an ALLINI are misformed with the ribonucleoprotein located outside the virus core. Virus assembly and maturation are highly orchestrated and regulated processes in which several viral proteins and RNA molecules closely interact. It is therefore of interest to study whether ALLINIs have unpredicted pleiotropic effects on these RNA-related processes. We confirm that the ALLINI BI-D inhibits virus replication and that the produced virus is non-infectious. Furthermore, we show that the wild-type level of HIV-1 genomic RNA is packaged in virions and these genomes are in a dimeric state. The tRNAlys3 primer for reverse transcription was properly placed on this genomic RNA and could be extended ex vivo. In addition, the packaged reverse transcriptase enzyme was fully active when extracted from virions. As the RNA and enzyme components for reverse transcription are properly present in virions produced in the presence of BI-D, the inhibition of reverse transcription is likely to reflect the mislocalization of the components in the aberrant virus particle.


Subject(s)
Acetates/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/chemistry , HIV-1/physiology , Quinolines/pharmacology , RNA, Viral/metabolism , Virus Replication/drug effects , Acetates/chemistry , Allosteric Regulation , Cell Line , Dimerization , Genome, Viral , HEK293 Cells , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , Humans , Quinolines/chemistry , RNA, Transfer/metabolism , RNA, Viral/isolation & purification , Virus Assembly/drug effects
6.
Retrovirology ; 10: 144, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24261564

ABSTRACT

BACKGROUND: LEDGF/p75 (LEDGF) is the main cellular cofactor of HIV-1 integrase (IN). It acts as a tethering factor for IN, and targets the integration of HIV in actively transcribed gene regions of chromatin. A recently developed class of IN allosteric inhibitors can inhibit the LEDGF-IN interaction. RESULTS: We describe a new series of IN-LEDGF allosteric inhibitors, the most active of which is Mut101. We determined the crystal structure of Mut101 in complex with IN and showed that the compound binds to the LEDGF-binding pocket, promoting conformational changes of IN which explain at the atomic level the allosteric effect of the IN/LEDGF interaction inhibitor on IN functions. In vitro, Mut101 inhibited both IN-LEDGF interaction and IN strand transfer activity while enhancing IN-IN interaction. Time of addition experiments indicated that Mut101 behaved as an integration inhibitor. Mut101 was fully active on HIV-1 mutants resistant to INSTIs and other classes of anti-HIV drugs, indicative that this compound has a new mode of action. However, we found that Mut101 also displayed a more potent antiretroviral activity at a post-integration step. Infectivity of viral particles produced in presence of Mut101 was severely decreased. This latter effect also required the binding of the compound to the LEDGF-binding pocket. CONCLUSION: Mut101 has dual anti-HIV-1 activity, at integration and post-integration steps of the viral replication cycle, by binding to a unique target on IN (the LEDGF-binding pocket). The post-integration block of HIV-1 replication in virus-producer cells is the mechanism by which Mut101 is most active as an antiretroviral. To explain this difference between Mut101 antiretroviral activity at integration and post-integration stages, we propose the following model: LEDGF is a nuclear, chromatin-bound protein that is absent in the cytoplasm. Therefore, LEDGF can outcompete compound binding to IN in the nucleus of target cells lowering its antiretroviral activity at integration, but not in the cytoplasm where post-integration production of infectious viral particles takes place.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , HIV-1/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Virus Integration/drug effects , Virus Replication/drug effects , Cell Line , Crystallography, X-Ray , HIV Integrase/chemistry , HIV Integrase Inhibitors/chemistry , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Protein Binding , Protein Conformation
7.
Int J Hypertens ; 2013: 513047, 2013.
Article in English | MEDLINE | ID: mdl-23431421

ABSTRACT

A complete renin-angiotensin system (RAS) is locally expressed in the brain and fulfills important functions. Angiotensin II, the major biologically active peptide of the RAS, acts via binding to two main receptor subtypes designated AT1 and AT2. The present paper focuses on AT2 receptors, which have been reported to have neuroprotective effects on stroke, degenerative diseases, and cognitive functions. Our group has identified a family of AT2 receptor interacting proteins (ATIPs) comprising three major members (ATIP1, ATIP3, and ATIP4) with different intracellular localization. Of interest, all ATIP members are expressed in brain tissues and carry a conserved domain able to interact with the AT2 receptor intracellular tail, suggesting a role in AT2-mediated brain functions. We summarize here current knowledge on the ATIP family of proteins, and we present new experimental evidence showing interaction defects between ATIP1 and two mutant forms of the AT2 receptor identified in cases of mental retardation. These studies point to a functional role of the AT2/ATIP1 axis in cognition.

8.
J Virol ; 86(5): 2533-44, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22171270

ABSTRACT

The HIV-1 Vpr protein participates in the early steps of the virus life cycle by influencing the accuracy of reverse transcription. This role of Vpr was related to the recruitment of the nuclear form of the uracil DNA glycosylase (UNG2) enzyme into virus particles, but several conflicting findings have been reported regarding the role of UNG2 encapsidation on viral infectivity. Here, we report that the catalytic activity of UNG2 was not required for influencing HIV-1 mutation, and this function of UNG2 was mapped within a 60-amino-acid domain located in the N-terminal region of the protein required for direct interaction with the p32 subunit of the replication protein A (RPA) complex. Importantly, enforced recruitment of overexpressed UNG2 into virions resulted in a net increase of virus infectivity, and this positive effect on infectivity was also independent of the UNG2 enzymatic activity. In contrast, virus infectivity and replication, as well as the efficiency of the viral DNA synthesis, were significantly reduced when viruses were produced from cells depleted of either endogenous UNG2 or RPA p32. Taken together, these results demonstrate that incorporation of UNG2 into virions has a positive impact on HIV-1 infectivity and replication and positively influences the reverse transcription process through a nonenzymatic mechanism involving the p32 subunit of the RPA complex.


Subject(s)
DNA Glycosylases/metabolism , HIV Infections/enzymology , HIV-1/physiology , Virion/physiology , Cell Line , DNA Glycosylases/genetics , HIV Infections/virology , HIV-1/genetics , Humans , Protein Binding , Virion/genetics , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/metabolism
9.
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
10.
PLoS One ; 4(10): e7514, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19838296

ABSTRACT

Although HIV-1 Vpr displays several functions in vitro, limited information exists concerning their relevance during infection. Here, we characterized Vpr variants isolated from a rapid and a long-term non-progressor (LTNP). Interestingly, vpr alleles isolated from longitudinal samples of the LTNP revealed a dominant sequence that subsequently led to diversity similar to that observed in the progressor patient. Most of primary Vpr proteins accumulated at the nuclear envelope and interacted with host-cell partners of Vpr. They displayed cytostatic and proapoptotic activities, although a LTNP allele, harboring the Q65R substitution, failed to bind the DCAF1 subunit of the Cul4a/DDB1 E3 ligase and was inactive. This Q65R substitution correlated with impairment of Vpr docking at the nuclear envelope, raising the possibility of a functional link between this property and the Vpr cytostatic activity. In contradiction with published results, the R77Q substitution, found in LTNP alleles, did not influence Vpr proapoptotic activity.


Subject(s)
Gene Products, vpr/genetics , Genes, vpr , Terminal Repeat Sequences , vpr Gene Products, Human Immunodeficiency Virus/genetics , Alleles , Amino Acid Sequence , Apoptosis , Genetic Variation , Humans , Leukocytes, Mononuclear/cytology , Models, Genetic , Molecular Sequence Data , Mutation , Protein Binding , Ubiquitin-Protein Ligases/metabolism
11.
J Virol ; 83(23): 12253-65, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19759136

ABSTRACT

Macrophages are major targets of human immunodeficiency virus type 1 (HIV-1). We have previously shown that aggregation of activating immunoglobulin G Fc receptors (FcgammaR) by immune complexes inhibits reverse transcript accumulation and integration of HIV-1 and related lentiviruses in monocyte-derived macrophages. Here, we show that FcgammaR-mediated restriction of HIV-1 is not due to enhanced degradation of incoming viral proteins or cDNA and is associated to the induction of the cyclin-dependent kinase inhibitor p21(Cip1/WAF1) (p21). Small interfering RNA-mediated p21 knockdown rescued viral replication in FcgammaR-activated macrophages and enhanced HIV-1 infection in unstimulated macrophages by increasing reverse transcript and integrated DNA levels. p21 induction by other stimuli, such as phorbol myristate acetate and the histone deacetylase inhibitor MS-275, was also associated with preintegrative blocks of HIV-1 replication in macrophages. Binding of p21 to reverse transcription/preintegration complex-associated HIV-1 proteins was not detected in yeast two-hybrid, pulldown, or coimmunoprecipitation assays, suggesting that p21 may affect viral replication independently of a specific interaction with an HIV-1 component. Consistently, p21 silencing rescued viral replication from the FcgammaR-mediated restriction also in simian immunodeficiency virus SIV(mac)- and HIV-2-infected macrophages. Our results point to a role of p21 as an inhibitory factor of lentiviral infection in macrophages and to its implication in FcgammaR-mediated restriction.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , HIV-1/growth & development , Lentiviruses, Primate/growth & development , Macrophages/immunology , Macrophages/virology , Receptors, IgG/immunology , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Knockdown Techniques , HIV-1/immunology , HIV-2/growth & development , HIV-2/immunology , Humans , Lentiviruses, Primate/immunology , Primates , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/immunology , Virus Integration/immunology , Virus Replication/immunology
12.
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
13.
J Biol Chem ; 283(31): 21686-92, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18524771

ABSTRACT

Many viruses subvert the host ubiquitin-proteasome system to optimize their life cycle. We recently documented such a mechanism for the human immunodeficiency virus type 1 Vpr protein, which promotes cell cycle arrest by recruiting the DCAF1 adaptor of the Cul4A-DDB1 ubiquitin ligase, a finding now confirmed by several groups. Here we examined the impact of Cul4A-DDB1(DCAF1) on Vpr stability. We show that the Vpr(Q65R) mutant, which is defective in DCAF1 binding, undergoes proteasome-mediated degradation at a higher rate than wild-type Vpr. DCAF1 overexpression stabilizes wild-type Vpr and leads to its cytoplasmic accumulation, whereas it has no effect on the Vpr(Q65R) mutant. Conversely, small interfering RNA-mediated silencing of DCAF1 decreases the steady state amount of the viral protein. Stabilization by DCAF1, which is conserved by Vpr species from human immunodeficiency virus type 2 and the SIVmac strain, results in increased G(2) arrest and requires the presence of DDB1, indicating that it occurs through assembly of Vpr with a functional Cul4A-DDB1(DCAF1) complex. Furthermore, in human immunodeficiency virus type 1-infected cells, the Vpr protein, issued from the incoming viral particle, is destabilized under DCAF1 or DDB1 silencing. Together with our previous findings, our data suggest that Cul4A-DDB1(DCAF1) acts at a dual level by providing Vpr with the equipment for the degradation of specific host proteins and by counter-acting its proteasome targeting by another cellular E3 ubiquitin ligase. This protection mechanism may represent an efficient way to optimize the activity of Vpr molecules that are delivered by the incoming virus before neosynthesis takes place. Targeting the Vpr-DCAF1 interaction might therefore present therapeutic interest.


Subject(s)
Cullin Proteins/metabolism , HIV-1/metabolism , Proteasome Endopeptidase Complex/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cell Cycle , Cell Line , DNA-Binding Proteins/metabolism , G2 Phase , Gene Silencing , HeLa Cells , Humans , Models, Biological , Mutation , RNA, Small Interfering/metabolism , Virus Replication
14.
Exp Mol Pathol ; 85(1): 2-10, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18514189

ABSTRACT

Since the first isolation of HIV-1 from a patient with generalized lymphadenopathy in 1983, great progress has been made in understanding the viral life cycle and the functional nuances of each of the nine genes encoded by HIV-1. Considerable attention has been paid to four small HIV-1 open reading frames, vif, vpr, vpu and nef. These genes were originally termed "accessory" because their deletion failed to completely disable viral replication in vitro. More than twenty years after the cloning and sequencing of HIV-1, a great deal of information is available regarding the multiple functions of the accessory proteins and it is well accepted that, collectively, these gene products modulate the host cell biology to favor viral replication, and that they are largely responsible for the pathogenesis of HIV-1. Expression of Vpr, in particular, leads to cell cycle arrest in G(2), followed by apoptosis. Here we summarize our current understanding of Vpr biology with a focus on Vpr-induced G(2) arrest and apoptosis.


Subject(s)
Apoptosis , G2 Phase , HIV Infections/pathology , HIV Infections/virology , HIV-1/physiology , vpr Gene Products, Human Immunodeficiency Virus/physiology , Humans , vpr Gene Products, Human Immunodeficiency Virus/chemistry , vpr Gene Products, Human Immunodeficiency Virus/genetics
15.
Retrovirology ; 4: 84, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-18039376

ABSTRACT

BACKGROUND: HIV-1 Vpr is a dynamic protein that primarily localizes in the nucleus, but a significant fraction is concentrated at the nuclear envelope (NE), supporting an interaction between Vpr and components of the nuclear pore complex, including the nucleoporin hCG1. In the present study, we have explored the contribution of Vpr accumulation at the NE to the Vpr functions, including G2-arrest and pro-apoptotic activities, and virus replication in primary macrophages. RESULTS: In order to define the functional role of Vpr localization at the NE, we have characterized a set of single-point Vpr mutants, and selected two new mutants with substitutions within the first alpha-helix of the protein, Vpr-L23F and Vpr-K27M, that failed to associate with hCG1, but were still able to interact with other known relevant host partners of Vpr. In mammalian cells, these mutants failed to localize at the NE resulting in a diffuse nucleocytoplasmic distribution both in HeLa cells and in primary human monocyte-derived macrophages. Other mutants with substitutions in the first alpha-helix (Vpr-A30L and Vpr-F34I) were similarly distributed between the nucleus and cytoplasm, demonstrating that this helix contains the determinants required for localization of Vpr at the NE. All these mutations also impaired the Vpr-mediated G2-arrest of the cell cycle and the subsequent cell death induction, indicating a functional link between these activities and the Vpr accumulation at the NE. However, this localization is not sufficient, since mutations within the C-terminal basic region of Vpr (Vpr-R80A and Vpr-R90K), disrupted the G2-arrest and apoptotic activities without altering NE localization. Finally, the replication of the Vpr-L23F and Vpr-K27M hCG1-binding deficient mutant viruses was also affected in primary macrophages from some but not all donors. CONCLUSION: These results indicate that the targeting of Vpr to the nuclear pore complex may constitute an early step toward Vpr-induced G2-arrest and subsequent apoptosis; they also suggest that Vpr targeting to the nuclear pore complex is not absolutely required, but can improve HIV-1 replication in macrophages.


Subject(s)
Cell Nucleus/virology , HIV-1/physiology , Macrophages/virology , Nuclear Envelope/metabolism , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus/physiology , Active Transport, Cell Nucleus/physiology , Cell Cycle , HIV-1/chemistry , HIV-1/metabolism , HeLa Cells , Humans , Two-Hybrid System Techniques , vpr Gene Products, Human Immunodeficiency Virus/biosynthesis , vpr Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/metabolism
16.
PLoS Pathog ; 3(7): e104, 2007 Jul 27.
Article in English | MEDLINE | ID: mdl-17676996

ABSTRACT

Viral protein U (Vpu) of HIV-1 has two known functions in replication of the virus: degradation of its cellular receptor CD4 and enhancement of viral particle release. Vpu binds CD4 and simultaneously recruits the betaTrCP subunit of the SCF(betaTrCP) ubiquitin ligase complex through its constitutively phosphorylated DS52GXXS56 motif. In this process, Vpu was found to escape degradation, while inhibiting the degradation of betaTrCP natural targets such as beta-catenin and IkappaBalpha. We further addressed this Vpu inhibitory function with respect to the degradation of Emi1 and Cdc25A, two betaTrCP substrates involved in cell-cycle progression. In the course of these experiments, we underscored the importance of a novel phosphorylation site in Vpu. We show that, especially in cells arrested in early mitosis, Vpu undergoes phosphorylation of the serine 61 residue, which lies adjacent to the betaTrCP-binding motif. This phosphorylation event triggers Vpu degradation by a betaTrCP-independent process. Mutation of Vpu S61 in the HIV-1 provirus extends the half-life of the protein and significantly increases the release of HIV-1 particles from HeLa cells. However, the S61 determinant of regulated Vpu turnover is highly conserved within HIV-1 isolates. Altogether, our results highlight a mechanism where differential phosphorylation of Vpu determines its fate as an adaptor or as a substrate of distinct ubiquitin ligases. Conservation of the Vpu degradation determinant, despite its negative effect on virion release, argues for a role in overall HIV-1 fitness.


Subject(s)
Gene Expression Regulation, Viral , HIV-1/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication/physiology , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Chlorocebus aethiops , F-Box Proteins/metabolism , HIV-1/pathogenicity , Human Immunodeficiency Virus Proteins , Humans , Molecular Sequence Data , Phosphorylation , Ubiquitin/metabolism , cdc25 Phosphatases/metabolism
17.
Cell Cycle ; 6(2): 182-8, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17314515

ABSTRACT

How the HIV1 Vpr protein initiates the host cell response leading to cell cycle arrest in G(2) has remained unknown. Here, we show that recruitment of DCAF1/VprBP by Vpr is essential for its cytostatic activity, which can be abolished either by single mutations of Vpr that impair DCAF1 binding, or by siRNA-mediated silencing of DCAF1. Furthermore, DCAF1 bridges Vpr to DDB1, a core subunit of Cul4 ubiquitin ligases. Altogether these results point to a mechanism where Vpr triggers G(2) arrest by hijacking the Cul4/DDB1(DCAF1) ubiquitin ligase. We further show that, Vpx, a non-cytostatic Vpr-related protein acquired by HIV2 and SIV, also binds DCAF1 through a conserved motif. Thus, Vpr from HIV1 and Vpx from SIV recruit DCAF1 with different physiological outcomes for the host cell. This in turn suggests that both proteins have evolved to preserve interaction with the same Cul4 ubiquitin ligase while diverging in the recognition of host substrates targeted for proteasomal degradation.


Subject(s)
Cell Cycle/physiology , Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Products, vpr/physiology , HIV-1/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cullin Proteins/physiology , Cytotoxins/physiology , Cytotoxins/toxicity , DNA-Binding Proteins/physiology , Gene Products, vpr/toxicity , HeLa Cells , Humans , Molecular Sequence Data , Protein Transport/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Ubiquitin-Protein Ligases/physiology , vpr Gene Products, Human Immunodeficiency Virus
18.
Cancer Res ; 67(3): 1054-61, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17283138

ABSTRACT

The Ras-association domain family 1 (RASSF1) gene has seven different isoforms; isoform A is a tumor-suppressor gene (RASSF1A). The promoter of RASSF1A is inactivated in many cancers, whereas the expression of another major isoform, RASSF1C, is not affected. Here, we show that RASSF1C, but not RASSF1A, interacts with betaTrCP. Binding of RASSF1C to betaTrCP involves serine 18 and serine 19 of the SS(18)GYXS(19) motif present in RASSF1C but not in RASSF1A. This motif is reminiscent of the canonical phosphorylation motif recognized by betaTrCP; however, surprisingly, the association between RASSF1C and betaTrCP does not occur via the betaTrCP substrate binding domain, the WD40 repeats. Overexpression of RASSF1C, but not of RASSF1A, resulted in accumulation and transcriptional activation of the beta-catenin oncogene, due to inhibition of its betaTrCP-mediated degradation. Silencing of RASSF1A by small interfering RNA was sufficient for beta-catenin to accumulate, whereas silencing of both RASSF1A and RASSF1C had no effect. Thus, RASSF1A and RASSF1C have opposite effects on beta-catenin degradation. Our results suggest that RASSF1C expression in the absence of RASSF1A could play a role in tumorigenesis.


Subject(s)
Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Amino Acid Motifs , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Silencing , HeLa Cells , Humans , Protein Binding , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/genetics , beta Catenin/antagonists & inhibitors , beta Catenin/biosynthesis , beta Catenin/genetics
19.
AIDS Res Hum Retroviruses ; 22(7): 630-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16831087

ABSTRACT

HIV-1 Vpr is a 96-amino acid auxiliary protein that performs numerous activities during viral infection. In the present study, 10 antibodies were generated after mice immunization with either the N- or the C-terminus domain of Vpr, respectively, Vpr(1-51) and Vpr(52-96). ELISA and immunoblot experiments using pure synthetic overlapping Vpr peptides suggested that these anti-Vpr antibodies could be classified into five groups and that they recognized conformational or linear Vpr epitopes. Further analysis revealed the effect of C-terminal arginine mutations on the antibody binding. Two of the antibodies precipitated Vpr expressed after transfection of a Vpr-encoding vector in human cells. More importantly, one of them was able to detect Vpr in HIV-1-infected U1 cells and in HIV-1-infected human PBMC. Surface plasmon resonance experiments demonstrated that some of these antibodies prevented the interaction between Vpr and one of its cellular partners, the adenine nucleotide translocator. Thus, these anti-Vpr monoclonal antibodies may be useful to any laboratory working on the molecular mechanism of HIV-1 infection.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Gene Products, vpr/immunology , HIV-1/immunology , Leukocytes, Mononuclear/virology , Animals , Antibodies, Monoclonal/immunology , Epitope Mapping/methods , Female , Humans , Immunoprecipitation/methods , Mice , Mice, Inbred BALB C , Surface Plasmon Resonance , vpr Gene Products, Human Immunodeficiency Virus
20.
Virology ; 339(1): 21-30, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15963546

ABSTRACT

We analyzed the role of human immunodeficiency virus (HIV)-1 matrix protein (MA) during the virus replication afferent phase. Single-round infection of H9 T lymphocytes showed that the combined mutation of MA Lys residues 26-27 in MA reported nuclear localization signal (NLS)-1 impaired infectivity, abrogated 2-LTR-circle formation and significantly reduced integration. However, the mutation did not affect viral DNA docking to chromatin in either interphasic or mitotic cells, indicating that MA N-terminal basic domain should not represent a major determinant of HIV-1 nuclear import in T lymphocytes. These data point to a previously unreported role of MA in the late, post-chromatin-binding, afferent phase of HIV-1 replication cycle.


Subject(s)
Gene Products, gag/physiology , HIV Antigens/physiology , HIV-1/physiology , Viral Proteins/physiology , Cell Cycle , Cell Line , Chromatin/metabolism , DNA, Viral/metabolism , Gene Products, gag/genetics , HIV Antigens/genetics , HIV-1/metabolism , Humans , Mutation , T-Lymphocytes/virology , Viral Proteins/genetics , Virus Integration , Virus Replication , gag Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...