Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Biochem J ; 478(8): 1525-1545, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33787846

ABSTRACT

The Nef protein of human and simian immunodeficiency viruses boosts viral pathogenicity through its interactions with host cell proteins. By combining the polyvalency of its large unstructured regions with the binding selectivity and strength of its folded core domain, Nef can associate with many different host cell proteins, thereby disrupting their functions. For example, the combination of a linear proline-rich motif and hydrophobic core domain surface allows Nef to bind tightly and specifically to SH3 domains of Src family kinases. We investigated whether the interplay between Nef's flexible regions and its core domain could allosterically influence ligand selection. We found that the flexible regions can associate with the core domain in different ways, producing distinct conformational states that alter the way in which Nef selects for SH3 domains and exposes some of its binding motifs. The ensuing crosstalk between ligands might promote functionally coherent Nef-bound protein ensembles by synergizing certain subsets of ligands while excluding others. We also combined proteomic and bioinformatics analyses to identify human proteins that select SH3 domains in the same way as Nef. We found that only 3% of clones from a whole-human fetal library displayed Nef-like SH3 selectivity. However, in most cases, this selectivity appears to be achieved by a canonical linear interaction rather than by a Nef-like 'tertiary' interaction. Our analysis supports the contention that Nef's mode of hijacking SH3 domains is a virus-specific adaptation with no or very few cellular counterparts. Thus, the Nef tertiary binding surface is a promising virus-specific drug target.


Subject(s)
HIV-1/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/chemistry , Nuclear Proteins/chemistry , Proto-Oncogene Proteins c-fyn/chemistry , nef Gene Products, Human Immunodeficiency Virus/chemistry , Allosteric Site , Amino Acid Sequence , Cloning, Molecular , Computational Biology/methods , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fetus , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HIV-1/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Host-Pathogen Interactions/genetics , Humans , Ligands , Molecular Dynamics Simulation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism
3.
FEBS Lett ; 586(13): 1759-64, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22641034

ABSTRACT

The functions of Src family kinases are tightly regulated through Src homology (SH) domain-mediated protein-protein interactions. We previously reported the biophysical characteristics of the apoptosis-linked gene 2-interacting protein X (Alix) in complex with the haemopoietic cell kinase (Hck) SH3 domain. In the current study, we have combined ITC, NMR, SAXS and molecular modeling to determine a 3D model of the complex. We demonstrate that Hck SH3 recognizes an extended linear proline-rich region of Alix. This particular binding mode enables Hck SH3 to sense a specific non-canonical residue situated in the SH3 RT-loop of the kinase. The resulting model helps clarify the mechanistic insights of Alix-Hck interaction.


Subject(s)
Calcium-Binding Proteins/chemistry , Cell Cycle Proteins/chemistry , Endosomal Sorting Complexes Required for Transport/chemistry , Proto-Oncogene Proteins c-hck/chemistry , src Homology Domains , Binding Sites , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Proline/genetics , Proline/metabolism , Protein Conformation , Proto-Oncogene Proteins c-hck/metabolism , Scattering, Small Angle , nef Gene Products, Human Immunodeficiency Virus/chemistry , nef Gene Products, Human Immunodeficiency Virus/metabolism
4.
Bioorg Med Chem ; 19(24): 7401-6, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22061824

ABSTRACT

The HIV-1 auxiliary protein Nef is required for the onset and progression of AIDS in HIV-1-infected persons. Here, we have deciphered the mode of action of a second-generation inhibitor of Nef, DLC27-14, presenting a competitive IC(50) of ∼16 µM measured by MALDI-TOF experiments. Thermal protein denaturation experiments revealed a negative effect on stability of Nef in the presence of a saturating concentration of the inhibitor. The destabilizing action of DLC27-14 was confirmed by a HIV protease-based experiment, in which the protease sensitivity of DLC27-14-bound Nef was three times as high as that of apo Nef. The only compatible docking modes of action for DLC27-14 suggest that DLC27-14 promotes an opening of two α-helices that would destabilize the Nef core domain. DLC27-14 thus acts as a specific protein disorder catalyzer that destabilizes the folded conformation of the protein. Our results open novel avenues toward the development of next-generation Nef inhibitors.


Subject(s)
Acquired Immunodeficiency Syndrome/metabolism , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV-1/metabolism , nef Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , nef Gene Products, Human Immunodeficiency Virus/metabolism , Acquired Immunodeficiency Syndrome/drug therapy , HIV Protease/metabolism , HIV-1/chemistry , HIV-1/drug effects , Humans , Models, Molecular , Protein Denaturation/drug effects , Protein Structure, Tertiary/drug effects , nef Gene Products, Human Immunodeficiency Virus/chemistry
5.
J Virol ; 84(21): 11067-75, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702622

ABSTRACT

APOBEC3F (A3F) is a member of the family of cytidine deaminases that is often coexpressed with APOBEC3G (A3G) in cells susceptible to HIV infection. A3F has been shown to have strong antiviral activity in transient-expression studies, and together with A3G, it is considered the most potent cytidine deaminase targeting HIV. Previous analyses suggested that the antiviral properties of A3F can be dissociated from its catalytic deaminase activity. We were able to confirm the deaminase-independent antiviral activity of exogenously expressed A3F; however, we also noted that exogenous expression was associated with very high A3F mRNA and protein levels. In analogy to our previous study of A3G, we produced stable HeLa cell lines constitutively expressing wild-type or deaminase-defective A3F at levels that were more in line with the levels of endogenous A3F in H9 cells. A3F expressed in stable HeLa cells was packaged into Vif-deficient viral particles with an efficiency similar to that of A3G and was properly targeted to the viral nucleoprotein complex. Surprisingly, however, neither wild-type nor deaminase-defective A3F inhibited HIV-1 infectivity. These results imply that the antiviral activity of endogenous A3F is negligible compared to that of A3G.


Subject(s)
Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , APOBEC-3G Deaminase , Cytosine Deaminase/analysis , Cytosine Deaminase/genetics , HIV Infections , HIV-1/pathogenicity , HeLa Cells , Humans , RNA, Messenger/analysis , Virion , vif Gene Products, Human Immunodeficiency Virus
6.
Biochem J ; 431(1): 93-102, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20670214

ABSTRACT

SFKs (Src family kinases) are central regulators of many signalling pathways. Their functions are tightly regulated through SH (Src homology) domain-mediated protein-protein interactions. A yeast two-hybrid screen using SH3 domains as bait identified Alix [ALG-2 (apoptosis-linked gene 2)-interacting protein X] as a novel Hck (haemopoietic cell kinase) SH3 domain interactor. The Alix-Hck-SH3 interaction was confirmed in vitro by a GST (glutathione transferase) pull-down assay and in intact cells by a mammalian two-hybrid assay. Furthermore, the interaction was demonstrated to be biologically relevant in cells. Through biophysical experiments, we then identified the PRR (proline-rich region) motif of Alix that binds Hck-SH3 and determined a dissociation constant of 34.5 µM. Heteronuclear NMR spectroscopy experiments were used to map the Hck-SH3 residues that interact with an ALIX construct containing the V and PRR domains or with the minimum identified interacting motif. Finally, SAXS (small-angle X-ray scattering) analysis showed that the N-terminal PRR of Alix is unfolded, at least before Hck-SH3 recognition. These results indicate that residues outside the canonical PxxP motif of Alix enhance its affinity and selectivity towards Hck-SH3. The structural framework of the Hck-Alix interaction will help to clarify how Hck and Alix assist during virus budding and cell-surface receptor regulation.


Subject(s)
Calcium-Binding Proteins/chemistry , Cell Cycle Proteins/chemistry , Endosomal Sorting Complexes Required for Transport/chemistry , Proto-Oncogene Proteins c-hck/chemistry , src Homology Domains , Amino Acid Sequence , Binding Sites , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Proto-Oncogene Proteins c-hck/metabolism , Scattering, Small Angle , Two-Hybrid System Techniques , Virus Release
7.
Virology ; 372(1): 136-46, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18023836

ABSTRACT

HIV-1 Vif inhibits the antiviral activity of APOBEC3G (APO3G) by inducing proteasomal degradation. Here, we studied the effects of Vif on APO3G in vitro. In this system, Vif did not cause APO3G degradation. Instead, Vif induced changes in APO3G that affected immunoprecipitation of the native protein. This effect required wt Vif and was reversed by heat denaturation of APO3G. Sucrose gradient analysis demonstrated that wt Vif induced the gradual transition of APO3G translated in vitro or expressed in HeLa cells from a low molecular mass conformation to puromycin-sensitive high molecular mass (HMM) complexes. In the absence of Vif or the presence of biologically inactive Vif APO3G failed to form HMM complexes. Our results expose a novel function of Vif that promotes the assembly of APO3G into presumably packaging-incompetent HMM complexes and may explain how Vif can overcome the APO3G-imposed block to HIV replication under conditions of no or inefficient APO3G degradation.


Subject(s)
Cytidine Deaminase/metabolism , HIV-1/pathogenicity , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , Centrifugation, Density Gradient , Cytidine Deaminase/genetics , HIV-1/metabolism , HeLa Cells , Hot Temperature , Humans , Immunoblotting , Immunoprecipitation , Mutation , Protein Biosynthesis , Protein Denaturation , Transcription, Genetic , vif Gene Products, Human Immunodeficiency Virus/genetics
8.
Proc Natl Acad Sci U S A ; 104(49): 19256-61, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18042718

ABSTRACT

Protein-protein recognition is the cornerstone of multiple cellular and pathological functions. Therefore, protein-protein interaction inhibition (2P2I) is endowed with great therapeutic potential despite the initial belief that 2P2I was refractory to small-molecule intervention. Improved knowledge of complex molecular binding surfaces has recently stimulated renewed interest for 2P2I, especially after identification of "hot spots" and first inhibitory compounds. However, the combination of target complexity and lack of starting compound has thwarted experimental results and created intellectual barriers. Here we combined virtual and experimental screening when no previously known inhibitors can be used as starting point in a structure-based research program that targets an SH3 binding surface of the HIV type I Nef protein. High-throughput docking and application of a pharmacophoric filter on one hand and search for analogy on the other hand identified drug-like compounds that were further confirmed to bind Nef in the micromolar range (isothermal titration calorimetry), to target the Nef SH3 binding surface (NMR experiments), and to efficiently compete for Nef-SH3 interactions (cell-based assay, GST pull-down). Initial identification of these compounds by virtual screening was validated by screening of the very same library of compounds in the cell-based assay, demonstrating that a significant enrichment factor was attained by the in silico screening. To our knowledge, our results identify the first set of drug-like compounds that functionally target the HIV-1 Nef SH3 binding surface and provide the basis for a powerful discovery process that should help to speed up 2P2I strategies and open avenues for new class of antiviral molecules.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Drug Design , nef Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , src Homology Domains/drug effects , Animals , Anti-HIV Agents/pharmacology , COS Cells , Chlorocebus aethiops , Computer Simulation , Drug Evaluation, Preclinical/methods , Protein Conformation , Two-Hybrid System Techniques , nef Gene Products, Human Immunodeficiency Virus/chemistry , nef Gene Products, Human Immunodeficiency Virus/metabolism
9.
Vaccine ; 25(50): 8441-7, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17997200

ABSTRACT

Humoral responses against extra-cellular HIV-1 Tat may be beneficial as Tat has been implicated in the viral pathogenesis associated with HIV-1 disease progression. We determined the levels of anti-Tat IgG in sera of HIV-1 seropositive individuals from the Rural Clinical Cohort in Uganda using nine different Tat proteins representative of the major subtypes presently accounting for 97% of infections worldwide. We observed the presence of anti-Tat IgG able to react against the various subtypes tested, although none cross-reacted against all nine variants. We show that 46.25% of seropositive patients were able to recognise at least one Tat variant with 1:1000 sera dilution. We also show that the C terminus of Tat is the most variable region and an important epitope that might explain the limitation of cross-recognition of Tat antibodies regarding Tat variants. This study shows in seropositive patients that Tat can tolerate mutations without modification of its primary function but with changes in its immunogenic properties. These findings should be considered when designing Tat-based HIV-1 vaccines.


Subject(s)
HIV Antibodies/blood , HIV-1/immunology , Mutation , Transcriptional Activation , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Circular Dichroism , Cohort Studies , Cross Reactions , Disease Progression , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , HIV-1/genetics , Humans , Immunoglobulin G/blood , Models, Molecular , Molecular Sequence Data , Rabbits , Rural Population , Uganda , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/metabolism
10.
J Virol ; 81(24): 13346-53, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17928335

ABSTRACT

APOBEC3G (APO3G) is a cellular cytidine deaminase with potent antiviral activity. Initial studies of the function of APO3G demonstrated extensive mutation of the viral genome, suggesting a model in which APO3G's antiviral activity is due to hypermutation of the viral genome. Recent studies, however, found that deaminase-defective APO3G mutants transiently expressed in virus-producing cells exhibited significant antiviral activity, suggesting that the antiviral activity of APO3G could be dissociated from its deaminase activity. To directly compare the antiviral activities of wild-type (wt) and deaminase-defective APO3G, we used two approaches: (i) we titrated wt and deaminase-defective APO3G in transient-transfection studies to achieve similar levels of virus-associated APO3G and (ii) we constructed stable cell lines and selected clones expressing comparable amounts of wt and deaminase-defective APO3G. Viruses produced under these conditions were tested for viral infectivity. The results from the two approaches were consistent and suggested that the antiviral activity of deaminase-defective APO3G was significantly lower than that of wt APO3G. We conclude that efficient inhibition of vif-defective human immunodeficiency virus type 1 requires catalytically active APO3G.


Subject(s)
Antiviral Agents/pharmacology , Cytidine Deaminase/pharmacology , HIV-1/drug effects , HIV-1/pathogenicity , APOBEC-3G Deaminase , Animals , Antiviral Agents/metabolism , Cell Line , Cytidine Deaminase/chemistry , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Genes, vif , HIV-1/genetics , HeLa Cells , Humans , Mutation , Rabbits , Transfection
11.
Virology ; 369(2): 329-39, 2007 Dec 20.
Article in English | MEDLINE | ID: mdl-17825339

ABSTRACT

HIV-1 Vif regulates viral infectivity by inhibiting the encapsidation of APOBEC3G (APO3G) through proteasomal degradation of the protein. Here we compared various Vif proteins for their ability to induce APO3G degradation and rescue viral infectivity. We found that Vif expressed from proviral vectors caused relatively inefficient degradation of APO3G in HeLa cells yet was very effective in inhibiting APO3G's antiviral activity. On the other hand, Vif expressed autonomously from a codon-optimized vector caused very efficient APO3G degradation and also effectively inhibited APO3G's antiviral effects. In contrast, a Vif chimera containing an N-terminal fluorescent tag efficiently induced APO3G degradation but was unable to restore viral infectivity. The lack of a direct correlation between APO3G degradation and rescue of viral infectivity suggests that these two properties of Vif are functionally separable. Our data imply that intracellular degradation of APO3G may not be the sole activity of Vif required for the production of infectious virions from APO3G-expressing cells.


Subject(s)
Cytidine Deaminase/metabolism , HIV-1/physiology , vif Gene Products, Human Immunodeficiency Virus/physiology , APOBEC-3G Deaminase , Cell Line , Cytidine Deaminase/genetics , Genes, vif , HIV-1/genetics , HIV-1/pathogenicity , HeLa Cells , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/physiology , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics
12.
Retrovirology ; 4: 48, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17631688

ABSTRACT

BACKGROUND: Efficient incorporation of the cellular cytidine deaminase APOBEC3G (APO3G) into HIV-1 virions is necessary for its antiviral activity. Even though cellular RNAs are known to be non-specifically incorporated into virus particles, we have previously found that encapsidation of APO3G into HIV-1 virions is specifically enhanced by viral genomic RNA. Intracellularly, APO3G was found to form large RNA-protein complexes involving a variety of cellular RNAs. The goal of this study was to investigate the possible contribution of host RNAs recently identified in intracellular APO3G ribonucleoprotein complexes to APO3G's encapsidation into HIV-1 virions. RESULTS: Our results show that 7SL RNA, a component of signal recognition particles, and hY1, hY3, hY4, hY5 RNAs were present in intracellular APO3G complexes and were packaged into HIV-1 particles lacking viral genomic RNA unlike APO3G, which was not packaged in significant amounts into genomic RNA-deficient particles. These results indicate that packaging of 7SL or hY RNAs is not sufficient for the packaging of APO3G into HIV-1 virions. We also tested the encapsidation of several other cellular RNAs including beta-actin, GAPDH, alpha-tubulin, and small nuclear RNAs and determined their effect on the packaging of APO3G into nascent virions. Again, we were unable to observe any correlation between APO3G encapsidation and the packaging of any of these cellular RNAs. CONCLUSION: The results from this study support our previous conclusion that viral genomic RNA is a critical determinant for APO3G incorporation into HIV-1 virions. While most cellular RNAs tested in this study were packaged into viruses or virus-like particles we failed to identify a correlation between APO3G encapsidation and the packaging of these cellular RNAs.


Subject(s)
HIV-1/physiology , Nucleoside Deaminases/genetics , RNA, Viral/genetics , Repressor Proteins/genetics , Virion/physiology , APOBEC-3G Deaminase , Cytidine Deaminase , DNA Primers , Genes, myc , HeLa Cells , Humans , Nucleoside Deaminases/metabolism , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Viral/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
J Virol ; 81(15): 8236-46, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17522211

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Vif counteracts the antiviral activity of the human cytidine deaminase APOBEC3G (APO3G) by inhibiting its incorporation into virions. This has been attributed to the Vif-induced degradation of APO3G by cytoplasmic proteasomes. We recently demonstrated that although APO3G has a natural tendency to form RNA-dependent homo-multimers, multimerization was not essential for encapsidation into HIV-1 virions or antiviral activity. We now demonstrate that a multimerization-defective APO3G variant (APO3G C97A) is able to assemble into RNase-sensitive high-molecular-mass (HMM) complexes, suggesting that homo-multimerization of APO3G and assembly into HMM complexes are unrelated RNA-dependent processes. Interestingly, APO3G C97A was highly resistant to Vif-induced degradation even though the two proteins were found to interact in coimmunoprecipitation experiments and exhibited partial colocalization in transfected HeLa cells. Surprisingly, encapsidation and antiviral activity of APO3G C97A were both inhibited by Vif despite resistance to degradation. These results demonstrate that targeting of APO3G to proteasome degradation and interference with viral encapsidation are distinct functional properties of Vif.


Subject(s)
Gene Products, vif/metabolism , HIV-1/metabolism , Nucleoside Deaminases/metabolism , Repressor Proteins/metabolism , APOBEC-3G Deaminase , Animals , Cytidine Deaminase , Gene Products, vif/genetics , HIV-1/pathogenicity , HeLa Cells , Humans , Mutation , Nucleoside Deaminases/chemistry , Nucleoside Deaminases/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/genetics , Virion , vif Gene Products, Human Immunodeficiency Virus
14.
J Virol ; 81(15): 8080-90, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17522232

ABSTRACT

Vif is a primate lentiviral accessory protein that is crucial for viral infectivity. Vif counteracts the antiviral activity of host deaminases such as APOBEC3G and APOBEC3F. We now report a novel function of African green monkey simian immunodeficiency virus (SIVagm) Vif that promotes replication of SIVagm in human cells lacking detectable deaminase activity. We found that cyclophilin A (CypA) was excluded from wild-type SIV particles but was efficiently packaged into vif-deficient SIVagm virions. The presence of CypA in vif-defective SIVagm was correlated with reduced viral replication. Infection of CypA knockout Jurkat cells or treatment of Jurkat cells with cyclosporine A eliminated the Vif-sensitive inhibition and resulted in replication profiles that were similar for wild-type and vif-deficient SIVagm. Importantly, the inhibitory effect of CypA was restricted to virus-producing cells and was TRIM5alpha independent. The abilities of SIVagm Vif to inhibit encapsidation of CypA and to increase viral infectivity were shared by rhesus macaque SIV Vif and thus seem to be general properties of SIV Vif proteins. Exclusion of CypA from SIVagm particles was not associated with intracellular degradation, suggesting a mode of Vif action distinct from that proposed for APOBEC3G. This is the first report of a novel vif-sensitive antiviral activity of human CypA that may limit zoonotic transmission of SIV and the first demonstration of CypA encapsidation into a virus other than human immunodeficiency virus type 1.


Subject(s)
Cyclophilin A/metabolism , Gene Products, vif/metabolism , Simian Immunodeficiency Virus/metabolism , Animals , Antiviral Restriction Factors , Carrier Proteins/metabolism , Cell Line , Cyclophilin A/genetics , Cyclosporine/metabolism , Gene Products, vif/genetics , Humans , Immunosuppressive Agents/metabolism , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Virion/metabolism , Virus Replication
15.
J Virol ; 80(10): 4673-82, 2006 May.
Article in English | MEDLINE | ID: mdl-16641260

ABSTRACT

APOBEC3G (APO3G) is a cytidine deaminase that restricts replication of vif-defective human immunodeficiency virus type 1 (HIV-1). Like other members of the cellular deaminase family, APO3G has the propensity to form homo-multimers. In the current study, we investigated the functional determinants for multimerization of human APO3G and studied the role of APO3G multimerization for catalytic activity, virus encapsidation, and antiviral activity. We found that human APO3G is capable of forming multimeric complexes in transfected HeLa cells. Interestingly, multimerization of APO3G was exquisitely sensitive to RNase treatment, suggesting that interaction of APO3G subunits is facilitated or stabilized by an RNA bridge. Mutation of a conserved cysteine residue (C97) that is part of an N-terminal zinc-finger motif in APO3G abolished multimerization of APO3G; however, the C97 mutation inhibited neither in vitro deaminase activity nor antiviral function of APO3G. These results suggest that monomeric APO3G is both catalytically active and has antiviral activity. Interference studies employing either catalytically inactive or packaging-incompetent APO3G variants suggest that wild-type APO3G is packaged into HIV-1 particles in monomeric form. These results provide novel insights into the catalytic function and antiviral property of APO3G and demonstrate an important role for C97 in the RNA-dependent multimerization of this protein.


Subject(s)
Antiviral Agents/pharmacology , Catalytic Domain , HIV-1/physiology , Nucleoside Deaminases/chemistry , Nucleoside Deaminases/physiology , Repressor Proteins/chemistry , Repressor Proteins/physiology , Virus Assembly/physiology , APOBEC-3G Deaminase , Amino Acid Substitution/genetics , Antiviral Agents/metabolism , Catalytic Domain/genetics , Cysteine/genetics , Cytidine Deaminase , HeLa Cells , Humans , Nucleoside Deaminases/metabolism , Repressor Proteins/metabolism , Ribonucleases/pharmacology
16.
Retrovirology ; 3: 8, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16441880

ABSTRACT

BACKGROUND: Extra-cellular roles of Tat might be the main cause of maintenance of HIV-1 infected CD4 T cells or reservoir cells. We developed a synthetic vaccine based on a Tat variant of 101 residues called Tat Oyi, which was identified in HIV infected patients in Africa who did not progress to AIDS. We compared, using rabbits, different adjuvants authorized for human use to test on ELISA the recognition of Tat variants from the five main HIV-1 subtypes. A formulation was tested on macaques followed by a SHIV challenge with a European strain. RESULTS: Tat Oyi with Montanide or Calcium Phosphate gave rabbit sera able to recognize all Tat variants. Five on seven Tat Oyi vaccinated macaques showed a better control of viremia compared to control macaques and an increase of CD8 T cells was observed only on Tat Oyi vaccinated macaques. Reservoir cells were not detectable at 56 days post-challenge in all Tat Oyi vaccinated macaques but not in the controls. CONCLUSION: The Tat Oyi vaccine should be efficient worldwide. No toxicity was observed on rabbits and macaques. We show in vivo that antibodies against Tat could restore the cellular immunity and make it possible the elimination of reservoir cells.


Subject(s)
AIDS Vaccines/immunology , Gene Products, tat/immunology , HIV-1/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, Synthetic , Animals , Enzyme-Linked Immunosorbent Assay , Gene Products, tat/genetics , Genetic Variation , Immunization/methods , Macaca mulatta/immunology , Rabbits/immunology , Viral Vaccines/immunology , tat Gene Products, Human Immunodeficiency Virus
17.
J Virol ; 79(9): 5870-4, 2005 May.
Article in English | MEDLINE | ID: mdl-15827203

ABSTRACT

APOBEC3G (APO3G) is a host cytidine deaminase that is incorporated into human immunodeficiency virus type 1 (HIV-1) particles. We report here that viral RNA promotes stable association of APO3G with HIV-1 nucleoprotein complexes (NPC). A target sequence located within the 5'-untranslated region of the HIV-1 RNA was identified to be necessary and sufficient for efficient APO3G packaging. Fine mapping revealed a sequence normally involved in viral genomic RNA dimerization and Gag binding to be important for APO3G packaging and association with viral NPC. Our data suggest that packaging of APO3G into HIV-1 NPC is enhanced by viral RNA.


Subject(s)
HIV-1/physiology , Nucleoproteins/metabolism , Proteins/metabolism , RNA, Viral/biosynthesis , 5' Untranslated Regions/metabolism , APOBEC-3G Deaminase , Cytidine Deaminase , HIV-1/metabolism , HeLa Cells , Humans , Nucleoside Deaminases , Repressor Proteins , Virus Replication
18.
Retrovirology ; 2: 5, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15691386

ABSTRACT

BACKGROUND: HIV infection and progression to AIDS is characterized by the depletion of T cells, which could be due, in part, to apoptosis mediated by the extra-cellular HIV-encoded Tat protein as a consequence of Tat binding to tubulin. Microtubules are tubulin polymers that are essential for cell structure and division. Molecules that target microtubules induce apoptosis and are potent anti-cancer drugs. We studied the effect on tubulin polymerization of three Tat variants: Tat HxB2 and Tat Eli from patients who are rapid progressors (RP) and Tat Oyi from highly exposed but persistently seronegative (HEPS) patients. We compared the effect on tubulin polymerization of these Tat variants and peptides corresponding to different parts of the Tat sequence, with paclitaxel, an anti-cancer drug that targets microtubules. RESULTS: We show that Tat, and specifically, residues 38-72, directly enhance tubulin polymerization. We demonstrate that Tat could also directly trigger the mitochondrial pathway to induce T cell apoptosis, as shown in vitro by the release of cytochrome c from isolated mitochondria. CONCLUSIONS: These results show that Tat directly acts on microtubule polymerization and provide insights into the mechanism of T cell apoptosis mediated by extra-cellular Tat.


Subject(s)
Gene Products, tat/metabolism , Microtubules/metabolism , Polymers/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cytochromes c/metabolism , Gene Products, tat/pharmacology , HIV-1/metabolism , Humans , Jurkat Cells , Mitochondria/physiology , Paclitaxel/pharmacology , T-Lymphocytes , Tubulin/metabolism , tat Gene Products, Human Immunodeficiency Virus
19.
Retrovirology ; 1: 27, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15373943

ABSTRACT

BACKGROUND: The human immunodeficiency virus Vif protein overcomes the inhibitory activity of the APOBEC3G cytidine deaminase by prohibiting its packaging into virions. Inhibition of APOBEC3G encapsidation is paralleled by a reduction of its intracellular level presumably caused by the Vif-induced proteasome-dependent degradation of APOBEC3G. RESULTS: In this report we employed confocal microscopy to study the effects of Vif on the expression of APOBEC3G on a single cell level. HeLa cells dually transfected with Vif and APOBEC3G expression vectors revealed efficient co-expression of the two proteins. Under optimal staining conditions approximately 80% of the transfected cells scored double-positive for Vif and APOBEC3G. However, the proportion of double-positive cells observed in identical cultures varied dependent on the fixation protocol and on the choice of antibodies used ranging from as low as 40% to as high as 80% of transfected cells. Importantly, single-positive cells expressing either Vif or APOBEC3G were observed both with wild type Vif and a biologically inactive Vif variant. Thus, the lack of APOBEC3G in some Vif-expressing cells cannot be attributed to Vif-induced degradation of APOBEC3G. These findings are consistent with our results from immunoblot analyses that revealed only moderate effects of Vif on the APOBEC3G steady state levels. Of note, viruses produced under such conditions were fully infectious demonstrating that the Vif protein used in our analyses was both functional and expressed at saturating levels. CONCLUSIONS: Our results suggest that Vif and APOBEC3G can be efficiently co-expressed. Thus, depletion of APOBEC3G from Vif expressing cells as suggested previously is not a universal property of Vif and thus is not imperative for the production of infectious virions.


Subject(s)
HIV-1/genetics , HIV-1/pathogenicity , Nucleoside Deaminases/genetics , Repressor Proteins/genetics , APOBEC-3G Deaminase , Cloning, Molecular , Cytidine Deaminase/genetics , Gene Products, vif/immunology , Gene Products, vif/physiology , HIV Seropositivity/virology , HeLa Cells , Humans , Microscopy, Confocal , Promoter Regions, Genetic , Sequence Deletion , Virion/physiology , vif Gene Products, Human Immunodeficiency Virus
20.
Vaccine ; 22(23-24): 3105-11, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15297062

ABSTRACT

AIDS vaccines now use a truncated version of 86 residues of the Tat protein related to the HIV-1 HXB2 strain predominant in Europe and North America. We compared antibodies raised in rabbits using a B subtype short Tat HXB2(86) and a full-length Tat HXB2(100). Serum against HXB2(86) recognizes only B and D subtypes while serum against HXB2(100) recognizes B, D, and C subtype variants. Conformational epitopes appear to be involved in the capacity of anti-Tat HXB2 sera to recognized non-homologous Tat variants. A linear B-epitope identified in sequence 71-81 in HXB2(86) disappears in HXB2(100), which has a new linear B-epitope identified at the C-terminus. Anti-HXB2(100) serum has a higher titer in neutralizing antibody against homologous and non-homologous variants compared to anti-HXB2(86) serum. We suggest that a Tat vaccine should contain a Tat variant with regular size, up to 99-101 residues now found in the field.


Subject(s)
AIDS Vaccines/immunology , Gene Products, tat/immunology , HIV-1/immunology , AIDS Vaccines/chemistry , Amino Acid Sequence , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Gene Products, tat/chemical synthesis , Gene Products, tat/chemistry , HIV-1/chemistry , Molecular Sequence Data , Neutralization Tests , Protein Conformation , Rabbits , Transcriptional Activation , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...