Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
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
2.
J Virol ; 84(10): 5201-11, 2010 May.
Article in English | MEDLINE | ID: mdl-20219919

ABSTRACT

APOBEC3G (A3G) is a host cytidine deaminase that serves as a potent intrinsic inhibitor of retroviral replication. A3G is packaged into human immunodeficiency virus type 1 virions and deaminates deoxycytidine to deoxyuridine on nascent minus-strand retroviral cDNA, leading to hyper-deoxyguanine-to-deoxyadenine mutations on positive-strand cDNA and inhibition of viral replication. The antiviral activity of A3G is suppressed by Vif, a lentiviral accessory protein that prevents encapsidation of A3G. In this study, we identified dominant negative mutants of Vif that interfered with the ability of wild-type Vif to inhibit the encapsidation and antiviral activity of A3G. These mutants were nonfunctional due to mutations in the highly conserved HCCH and/or SOCS box motifs, which are required for assembly of a functional Cul5-E3 ubiquitin ligase complex. Similarly, mutation or deletion of a PPLP motif, which was previously reported to be important for Vif dimerization, induced a dominant negative phenotype. Expression of dominant negative Vif counteracted the Vif-induced reduction of intracellular A3G levels, presumably by preventing Vif-induced A3G degradation. Consequently, dominant negative Vif interfered with wild-type Vif's ability to exclude A3G from viral particles and reduced viral infectivity despite the presence of wild-type Vif. The identification of dominant negative mutants of Vif presents exciting possibilities for the design of novel antiviral strategies.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , HIV-1/immunology , HIV-1/pathogenicity , Mutant Proteins/metabolism , Mutation , Virulence Factors/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , Cell Line , HIV-1/genetics , Humans , Microbial Viability , Mutant Proteins/genetics , Virulence Factors/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics
3.
Retrovirology ; 6: 99, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19886996

ABSTRACT

BACKGROUND: The cellular cytidine deaminase APOBEC3G (A3G), when incorporated into the human immunodeficiency virus type 1 (HIV-1), renders viral particles non-infectious. We previously observed that mutation of a single cysteine residue of A3G (C100S) inhibited A3G packaging. In addition, several recent studies showed that mutation of tryptophan 127 (W127) and tyrosine 124 (Y124) inhibited A3G encapsidation suggesting that the N-terminal CDA constitutes a viral packaging signal in A3G. It was also reported that W127 and Y124 affect A3G oligomerization. RESULTS: Here we studied the mechanistic basis of the packaging defect of A3G W127A and Y124A mutants. Interestingly, cell fractionation studies revealed a strong correlation between encapsidation, lipid raft association, and genomic RNA binding of A3G. Surprisingly, the presence of a C-terminal epitope tag affected lipid raft association and encapsidation of the A3G W127A mutant but had no effect on wt A3G encapsidation, lipid raft association, and interaction with viral genomic RNA. Mutation of Y124 abolished A3G encapsidation irrespective of the presence or absence of an epitope tag. Contrasting a recent report, our co-immunoprecipitation studies failed to reveal a correlation between A3G oligomerization and A3G encapsidation. In fact, our W127A and Y124A mutants both retained the ability to oligomerize. CONCLUSION: Our results confirm that W127 and Y124 residues in A3G are important for encapsidation into HIV-1 virions and our data establish a novel correlation between genomic RNA binding, lipid raft association, and viral packaging of A3G. In contrast, we were unable to confirm a role of W127 and Y124 in A3G oligomerization and we thus failed to confirm a correlation between A3G oligomerization and virus encapsidation.


Subject(s)
Cytidine Deaminase/metabolism , HIV-1/physiology , Membrane Microdomains/metabolism , Virus Assembly , APOBEC-3G Deaminase , Amino Acid Substitution/genetics , Cell Fractionation , Cytidine Deaminase/genetics , Humans , Immunoprecipitation , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Protein Binding , RNA, Viral/metabolism
4.
J Virol ; 83(2): 1156-60, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004939

ABSTRACT

HIV-1 Vif counteracts the antiviral activity of APOBEC3G by inhibiting its encapsidation into virions. Here, we compared the relative sensitivity to Vif of APOBEC3G in stable HeLa cells containing APOBEC3G (HeLa-A3G cells) versus that of newly synthesized APOBEC3G. We observed that newly synthesized APOBEC3G was more sensitive to degradation than preexisting APOBEC3G. Nevertheless, preexisting and transiently expressed APOBEC3G were packaged with similar efficiencies into vif-deficient human immunodeficiency virus type 1 (HIV-1) virions, and Vif inhibited the encapsidation of both forms of APOBEC3G into HIV particles equally well. Our results suggest that HIV-1 Vif preferentially induces degradation of newly synthesized APOBEC3G but indiscriminately inhibits encapsidation of "old" and "new" APOBEC3G.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , HIV-1/physiology , HeLa Cells , Humans
5.
Retrovirology ; 5: 51, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18577210

ABSTRACT

Members of the APOBEC family of cellular cytidine deaminases represent a recently identified group of proteins that provide immunity to infection by retroviruses and protect the cell from endogenous mobile retroelements. Yet, HIV-1 is largely immune to the intrinsic antiviral effects of APOBEC proteins because it encodes Vif (viral infectivity factor), an accessory protein that is critical for in vivo replication of HIV-1. In the absence of Vif, APOBEC proteins are encapsidated by budding virus particles and either cause extensive cytidine to uridine editing of negative sense single-stranded DNA during reverse transcription or restrict virus replication through deaminase-independent mechanisms. Thus, the primary function of Vif is to prevent encapsidation of APOBEC proteins into viral particles. This is in part accomplished by the ability of Vif to induce the ubiquitin-dependent degradation of some of the APOBEC proteins. However, Vif is also able to prevent encapsidation of APOBEC3G and APOBEC3F through degradation-independent mechanism(s). The goal of this review is to recapitulate current knowledge of the functional interaction of HIV-1 and its Vif protein with the APOBEC3 subfamily of proteins and to summarize our present understanding of the mechanism of APOBEC3-dependent retrovirus restriction.


Subject(s)
Cytosine Deaminase/antagonists & inhibitors , Cytosine Deaminase/immunology , HIV Infections/immunology , HIV-1/physiology , vif Gene Products, Human Immunodeficiency Virus/metabolism , Humans
6.
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
7.
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
8.
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
9.
Retrovirology ; 4: 61, 2007 Aug 29.
Article in English | MEDLINE | ID: mdl-17727729

ABSTRACT

BACKGROUND: APOBEC3 (A3) proteins constitute a family of cytidine deaminases that provide intracellular resistance to retrovirus replication and to transposition of endogenous retroelements. A3A has significant homology to the C-terminus of A3G but has only a single cytidine deaminase active site (CDA), unlike A3G, which has a second N-terminal CDA previously found to be important for Vif sensitivity and virus encapsidation. A3A is packaged into HIV-1 virions but, unlike A3G, does not have antiviral properties. Here, we investigated the reason for the lack of A3A antiviral activity. RESULTS: Sequence alignment of A3G and A3A revealed significant homology of A3A to the C-terminal region of A3G. However, while A3G co-purified with detergent-resistant viral nucleoprotein complexes (NPC), virus-associated A3A was highly detergent-sensitive leading us to speculate that the ability to assemble into NPC may be a property conveyed by the A3G N-terminus. To test this model, we constructed an A3G-3A chimeric protein, in which the N-terminal half of A3G was fused to A3A. Interestingly, the A3G-3A chimera was packaged into HIV-1 particles and, unlike A3A, associated with the viral NPC. Furthermore, the A3G-3A chimera displayed strong antiviral activity against HIV-1 and was sensitive to inhibition by HIV-1 Vif. CONCLUSION: Our results suggest that the A3G N-terminal domain carries determinants important for targeting the protein to viral NPCs. Transfer of this domain to A3A results in A3A targeting to viral NPCs and confers antiviral activity.


Subject(s)
Cytidine Deaminase/metabolism , Gene Products, vif/metabolism , HIV Infections/enzymology , HIV-1/physiology , Nucleoproteins/metabolism , Proteins/metabolism , Amino Acid Sequence , Gene Products, vif/genetics , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , HIV-1/pathogenicity , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Subcellular Fractions/metabolism , Transfection , Virus Replication , vif Gene Products, Human Immunodeficiency Virus
10.
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
11.
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
12.
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
13.
J Virol ; 80(22): 10957-71, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16956950

ABSTRACT

3-O-(3',3'-dimethylsuccinyl)betulinic acid (PA-457 or bevirimat) potently inhibits human immunodeficiency virus type 1 (HIV-1) maturation by blocking a late step in the Gag processing pathway, specifically the cleavage of SP1 from the C terminus of capsid (CA). To gain insights into the mechanism(s) by which HIV-1 could evolve resistance to PA-457 and to evaluate the likelihood of such resistance arising in PA-457-treated patients, we sought to identify and characterize a broad spectrum of HIV-1 variants capable of conferring resistance to this compound. Numerous independent rounds of selection repeatedly identified six single-amino-acid substitutions that independently confer PA-457 resistance: three at or near the C terminus of CA (CA-H226Y, -L231F, and -L231M) and three at the first and third residues of SP1 (SP1-A1V, -A3T, and -A3V). We determined that mutations CA-H226Y, CA-L231F, CA-L231M, and SP1-A1V do not impose a significant replication defect on HIV-1 in culture. In contrast, mutations SP1-A3V and -A3T severely impaired virus replication and inhibited virion core condensation. The replication defect imposed by SP1-A3V was reversed by a second-site compensatory mutation in CA (CA-G225S). Intriguingly, high concentrations of PA-457 enhanced the maturation of SP1 residue 3 mutants. The different phenotypes associated with mutations that confer PA-457 resistance suggest the existence of multiple mechanisms by which HIV-1 can evolve resistance to this maturation inhibitor. These findings have implications for the ongoing development of PA-457 to treat HIV-1 infection in vivo.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV-1/drug effects , Succinates/pharmacology , Triterpenes/pharmacology , Amino Acid Substitution/genetics , Capsid Proteins/genetics , DNA Mutational Analysis , HIV-1/genetics , HIV-1/ultrastructure , Humans , Jurkat Cells , Microscopy, Electron, Transmission , Mutation, Missense , Suppression, Genetic , Viral Proteins/analysis , Viral Proteins/isolation & purification , Virion/ultrastructure , Virus Assembly/drug effects , Virus Replication/drug effects
14.
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
15.
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
16.
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
17.
J Virol ; 77(11): 6507-19, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12743307

ABSTRACT

Retrovirus budding is greatly stimulated by the presence of Gag sequences known as late or L domains. The L domain of human immunodeficiency virus type 1 (HIV-1) maps to a highly conserved Pro-Thr-Ala-Pro (PTAP) sequence in the p6 domain of Gag. We and others recently observed that the p6 PTAP motif interacts with the cellular endosomal sorting protein TSG101. Consistent with a role for TSG101 in virus release, we demonstrated that overexpressing the N-terminal, Gag-binding domain of TSG101 (TSG-5') suppresses HIV-1 budding by blocking L domain function. To elucidate the role of TSG101 in HIV-1 budding, we evaluated the significance of the binding between Gag and TSG-5' on the inhibition of HIV-1 release. We observed that a mutation in TSG-5' that disrupts the Gag/TSG101 interaction suppresses the ability of TSG-5' to inhibit HIV-1 release. We also determined the effect of overexpressing a panel of truncated TSG101 derivatives and full-length TSG101 (TSG-F) on virus budding. Overexpressing TSG-F inhibits HIV-1 budding; however, the effect of TSG-F on virus release does not require Gag binding. Furthermore, overexpression of the C-terminal portion of TSG101 (TSG-3') potently inhibits budding of not only HIV-1 but also murine leukemia virus. Confocal microscopy data indicate that TSG-F and TSG-3' overexpression induces an aberrant endosome phenotype; this defect is dependent upon the C-terminal, Vps-28-binding domain of TSG101. We propose that TSG-5' suppresses HIV-1 release by binding PTAP and blocking HIV-1 L domain function, whereas overexpressing TSG-F or TSG-3' globally inhibits virus release by disrupting the cellular endosomal sorting machinery. These results highlight the importance of TSG101 and the endosomal sorting pathway in virus budding and suggest that inhibitors can be developed that, like TSG-5', target HIV-1 without disrupting endosomal sorting.


Subject(s)
DNA-Binding Proteins/metabolism , Endosomes/metabolism , Gene Expression Regulation, Viral , HIV-1/growth & development , HIV-1/physiology , Transcription Factors/metabolism , Amino Acid Motifs , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Endosomal Sorting Complexes Required for Transport , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Mutation , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...