Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
mBio ; 11(4)2020 07 14.
Article in English | MEDLINE | ID: mdl-32665270

ABSTRACT

HLA-C-mediated antigen presentation induces the killing of human immunodeficiency virus (HIV)-infected CD4+ T cells by cytotoxic T lymphocytes (CTLs). To evade killing, many HIV-1 group M strains decrease HLA-C surface levels using their accessory protein Vpu. However, some HIV-1 group M isolates lack this activity, possibly to prevent the activation of natural killer (NK) cells. Analyzing diverse primate lentiviruses, we found that Vpu-mediated HLA-C downregulation is not limited to pandemic group M but is also found in HIV-1 groups O and P as well as several simian immunodeficiency viruses (SIVs). We show that Vpu targets HLA-C primarily at the protein level, independently of its ability to suppress NF-κB-driven gene expression, and that in some viral lineages, HLA-C downregulation may come at the cost of efficient counteraction of the restriction factor tetherin. Remarkably, HIV-2, which does not carry a vpu gene, uses its accessory protein Vif to decrease HLA-C surface expression. This Vif activity requires intact binding sites for the Cullin5/Elongin ubiquitin ligase complex but is separable from its ability to counteract APOBEC3G. Similar to HIV-1 Vpu, the degree of HIV-2 Vif-mediated HLA-C downregulation varies considerably among different virus isolates. In agreement with opposing selection pressures in vivo, we show that the reduction of HLA-C surface levels by HIV-2 Vif is accompanied by increased NK cell-mediated killing. In summary, our results highlight the complex role of HLA-C in lentiviral infections and demonstrate that HIV-1 and HIV-2 have evolved at least two independent mechanisms to decrease HLA-C levels on infected cells.IMPORTANCE Genome-wide association studies suggest that HLA-C expression is a major determinant of viral load set points and CD4+ T cell counts in HIV-infected individuals. On the one hand, efficient HLA-C expression enables the killing of infected cells by cytotoxic T lymphocytes (CTLs). On the other hand, HLA-C sends inhibitory signals to natural killer (NK) cells and enhances the infectivity of newly produced HIV particles. HIV-1 group M viruses modulate HLA-C expression using the accessory protein Vpu, possibly to balance CTL- and NK cell-mediated immune responses. Here, we show that the second human immunodeficiency virus, HIV-2, can use its accessory protein Vif to evade HLA-C-mediated restriction. Furthermore, our mutational analyses provide insights into the underlying molecular mechanisms. In summary, our results reveal how the two human AIDS viruses modulate HLA-C, a key component of the antiviral immune response.


Subject(s)
Evolution, Molecular , HIV-1/genetics , HIV-2/genetics , HLA-C Antigens/genetics , Human Immunodeficiency Virus Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HEK293 Cells , HIV Infections/immunology , HIV Infections/virology , HIV-1/classification , HIV-1/immunology , HIV-2/immunology , Host-Pathogen Interactions/immunology , Humans , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology
2.
J Virol ; 94(8)2020 03 31.
Article in English | MEDLINE | ID: mdl-32024773

ABSTRACT

Optimization of immunogen is crucial for induction of effective T-cell responses in the development of a human immunodeficiency virus (HIV) vaccine. Conventional T-cell-based vaccines have been designed to induce virus-specific CD4+ T as well as CD8+ T cells. However, it has been indicated that induction of HIV-specific CD4+ T cells, preferential targets for HIV infection, by vaccination may be detrimental and accelerate viral replication after HIV exposure. In the present study, we present a novel immunogen to selectively induce CD8+ T cells but not CD4+ T cells targeting viral antigens. The immunogen, CaV11, was constructed by tandem connection of overlapping 11-mer peptides spanning simian immunodeficiency virus (SIV) Gag capsid (CA) and Vif. Prime-boost immunization with DNA and Sendai virus (SeV) vectors expressing CaV11 efficiently induced Gag/Vif-specific CD8+ T-cell responses with inefficient Gag/Vif-specific CD4+ T-cell induction in rhesus macaques (n = 6). None of the macaques exhibited the enhancement of acute viral replication after an intravenous high-dose SIV challenge, which was observed in those immunized with DNA and SeV expressing the whole Gag protein in our previous study. Set point viral control postinfection was associated with SeV-specific CD4+ T-cell responses postimmunization, suggesting contribution of SeV-specific helper responses to effective Gag/Vif-specific CD8+ T-cell induction by vaccination. This immunogen design could be a promising method for selective induction of effective anti-HIV CD8+ T-cell responses.IMPORTANCE Induction of effective CD8+ T-cell responses is an important HIV vaccine strategy. Several promising vaccine delivery tools have been developed, and immunogen optimization is now crucial for effective T-cell induction. Conventional immunogens have been designed to induce virus-specific CD4+ T cells as well as CD8+ T cells, but induction of virus-specific CD4+ T cells that are preferential targets for HIV infection could enhance acute HIV proliferation. Here, we designed a novel immunogen to induce HIV-specific CD8+ T cells without HIV-specific CD4+ T-cell induction but with non-HIV antigen-specific CD4+ T-cell help. Our analysis in a macaque AIDS model showed that our immunogen can efficiently elicit effective CD8+ T but not CD4+ T cells targeting viral antigens, resulting in no enhancement of acute viral replication after virus exposure. This immunogen design, also applicable for other currently developed immunogens, could be a promising method for selective induction of effective anti-HIV CD8+ T-cell responses.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , AIDS Vaccines/immunology , Amino Acid Sequence , Animals , Disease Models, Animal , Immunization , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccination , Viral Load , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology
3.
Nat Commun ; 9(1): 4137, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297863

ABSTRACT

Human APOBEC3H (A3H) is a single-stranded DNA cytosine deaminase that inhibits HIV-1. Seven haplotypes (I-VII) and four splice variants (SV154/182/183/200) with differing antiviral activities and geographic distributions have been described, but the genetic and mechanistic basis for variant expression and function remains unclear. Using a combined bioinformatic/experimental analysis, we find that SV200 expression is specific to haplotype II, which is primarily found in sub-Saharan Africa. The underlying genetic mechanism for differential mRNA splicing is an ancient intronic deletion [del(ctc)] within A3H haplotype II sequence. We show that SV200 is at least fourfold more HIV-1 restrictive than other A3H splice variants. To counteract this elevated antiviral activity, HIV-1 protease cleaves SV200 into a shorter, less restrictive isoform. Our analyses indicate that, in addition to Vif-mediated degradation, HIV-1 may use protease as a  counter-defense mechanism against A3H in >80% of sub-Saharan African populations.


Subject(s)
Alternative Splicing/immunology , Aminohydrolases/immunology , HIV Protease/immunology , HIV-1/immunology , Haplotypes/immunology , Alternative Splicing/genetics , Amino Acid Sequence , Aminohydrolases/genetics , Aminohydrolases/metabolism , Base Sequence , HEK293 Cells , HIV Protease/metabolism , HIV-1/metabolism , Haplotypes/genetics , Humans , Isoenzymes/genetics , Isoenzymes/immunology , Isoenzymes/metabolism , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/immunology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Virus Replication/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/metabolism
4.
PLoS One ; 13(5): e0196942, 2018.
Article in English | MEDLINE | ID: mdl-29758076

ABSTRACT

Traditional restriction endonuclease-based cloning has been routinely used to generate replication-competent simian-human immunodeficiency viruses (SHIV) and simian tropic HIV (stHIV). This approach requires the existence of suitable restriction sites or the introduction of nucleotide changes to create them. Here, using an In-Fusion cloning technique that involves homologous recombination, we generated SHIVs and stHIVs based on epidemiologically linked clade C transmitted/founder HIV molecular clones from Zambia. Replacing vif from these HIV molecular clones with vif of SIVmac239 resulted in chimeric genomes used to generate infectious stHIV viruses. Likewise, exchanging HIV env genes and introducing N375 mutations to enhance macaque CD4 binding site and cloned into a SHIVAD8-EO backbone. The generated SHIVs and stHIV were infectious in TZMbl and ZB5 cells, as well as macaque PBMCs. Therefore, this method can replace traditional methods and be a valuable tool for the rapid generation and testing of molecular clones of stHIV and SHIV based on primary clinical isolates will be valuable to generate rapid novel challenge viruses for HIV vaccine/cure studies.


Subject(s)
HIV-1 , Mutation, Missense , Organisms, Genetically Modified , Simian Immunodeficiency Virus , vif Gene Products, Human Immunodeficiency Virus , Amino Acid Substitution , Animals , Cell Line , HIV Envelope Protein gp160/genetics , HIV Envelope Protein gp160/immunology , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/transmission , HIV-1/genetics , HIV-1/immunology , HIV-1/pathogenicity , Humans , Macaca mulatta , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/immunology , Organisms, Genetically Modified/pathogenicity , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Zambia , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/immunology
5.
Trends Mol Med ; 24(5): 507-520, 2018 05.
Article in English | MEDLINE | ID: mdl-29609878

ABSTRACT

The infectivity of HIV depends on overcoming APOBEC3 (A3) innate immunity, predominantly through the expression of the viral protein Vif, which induces A3 degradation in the proteasome. Disruption of the functional interactions of Vif enables A3 mutagenesis of the HIV genome during viral replication, which can result in a broadly neutralizing antiviral effect. Vif function requires self-association along with interactions with A3 proteins, protein chaperones, and factors of the ubiquitination machinery and these are described here as a potential platform for novel antiviral drug discovery. This Review will examine the current state of development of Vif inhibitors that we believe to have therapeutic and functional cure potential.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Cytosine Deaminase/immunology , HIV Infections/drug therapy , HIV-1/drug effects , Immunity, Innate/drug effects , vif Gene Products, Human Immunodeficiency Virus/immunology , APOBEC Deaminases , Cytidine Deaminase , Cytosine Deaminase/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Protein Binding/drug effects , Proteolysis/drug effects , vif Gene Products, Human Immunodeficiency Virus/metabolism
6.
PLoS Pathog ; 14(1): e1006830, 2018 01.
Article in English | MEDLINE | ID: mdl-29304101

ABSTRACT

The lentiviral protein Viral Infectivity Factor (Vif) counteracts the antiviral effects of host APOBEC3 (A3) proteins and contributes to persistent HIV infection. Vif targets A3 restriction factors for ubiquitination and proteasomal degradation by recruiting them to a multi-protein ubiquitin E3 ligase complex. Here, we describe a degradation-independent mechanism of Vif-mediated antagonism that was revealed through detailed structure-function studies of antibody antigen-binding fragments (Fabs) to the Vif complex. Two Fabs were found to inhibit Vif-mediated A3 neutralization through distinct mechanisms: shielding A3 from ubiquitin transfer and blocking Vif E3 assembly. Combined biochemical, cell biological and structural studies reveal that disruption of Vif E3 assembly inhibited A3 ubiquitination but was not sufficient to restore its packaging into viral particles and antiviral activity. These observations establish that Vif can neutralize A3 family members in a degradation-independent manner. Additionally, this work highlights the potential of Fabs as functional probes, and illuminates how Vif uses a multi-pronged approach involving both degradation dependent and independent mechanisms to suppress A3 innate immunity.


Subject(s)
Antiviral Agents/pharmacology , Cytosine Deaminase/metabolism , Immunoglobulin Fab Fragments/chemistry , vif Gene Products, Human Immunodeficiency Virus/immunology , APOBEC Deaminases , Antiviral Agents/chemistry , Cullin Proteins/chemistry , Cullin Proteins/metabolism , Cytidine Deaminase , HEK293 Cells , HIV Infections/immunology , HIV Infections/therapy , HIV Infections/virology , HIV-1/immunology , HIV-1/metabolism , Humans , Ubiquitin/metabolism , Ubiquitination , Virus Assembly , vif Gene Products, Human Immunodeficiency Virus/chemistry
7.
Adv Exp Med Biol ; 962: 415-431, 2017.
Article in English | MEDLINE | ID: mdl-28299671

ABSTRACT

In order to achieve a persistent infection, viruses must overcome the host immune system. Host restriction factors dominantly block virus transmission, but are subject to down regulation by viral accessory proteins. HIV encodes several accessory factors that overcome different cellular restriction factors. For example, the HIV-1 protein Vif down regulates the human APOBEC3 family of restriction factors by targeting them for proteolysis by the ubiquitin-proteasome pathway. Recently, this function was shown to require the transcription cofactor CBFß, which acts as a template to assist in Vif folding and allow for assembly of an APOBEC3-targeting E3 ligase complex. In uninfected cells, CBFß is an essential binding partner of RUNX transcription factors. By binding CBFß, Vif has also been shown to perturb transcription of genes regulated by the RUNX proteins, including restrictive APOBEC3 family members. Here we review how the link between CBFß and Vif supports transcriptional and post-transcriptional repression of innate immunity. The ability of a single viral protein to coopt multiple host pathways is an economical strategy for a pathogen with limited protein coding capacity to achieve a productive infection.


Subject(s)
Core Binding Factor beta Subunit/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Core Binding Factor beta Subunit/immunology , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , HIV Infections/immunology , HIV-1/immunology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Immunity, Innate/immunology , Transcription, Genetic/immunology , Transcription, Genetic/physiology , vif Gene Products, Human Immunodeficiency Virus/immunology
8.
J Virol ; 88(24): 14380-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25275135

ABSTRACT

UNLABELLED: The APOBEC3 deoxycytidine deaminases can restrict the replication of HIV-1 in cell culture to differing degrees. The effects of APOBEC3 enzymes are largely suppressed by HIV-1 Vif that interacts with host proteins to form a Cullin5-Ring E3 ubiquitin ligase that induces (48)K-linked polyubiquitination (poly-Ub) and proteasomal degradation of APOBEC3 enzymes. Vif variants have differing abilities to induce degradation of APOBEC3 enzymes and the underlying biochemical mechanisms for these differences is not fully understood. We hypothesized that by characterizing the interaction of multiple APOBEC3 enzymes and Vif variants we could identify common features that resulted in Vif-mediated degradation and further define the determinants required for efficient Vif-mediated degradation of APOBEC3 enzymes. We used Vifs from HIV-1 NL4-3 (IIIB) and HXB2 to characterize their induced degradation of and interaction with APOBEC3G, APOBEC3G D128K, APOBEC3H, and APOBEC3B in 293T cells. We quantified the APOBEC3G-Vif and APOBEC3H-Vif interaction strengths in vitro using rotational anisotropy. Our biochemical and cellular analyses of the interactions support a model in which the degradation efficiency of VifIIIB and VifHXB2 correlated with both the binding strength of the APOBEC3-Vif interaction and the APOBEC3-Vif interface, which differs for APOBEC3G and APOBEC3H. Notably, Vif bound to APOBEC3H and APOBEC3B in the natural absence of Vif-induced degradation and the interaction resulted in (63)K-linked poly-Ub of APOBEC3H and APOBEC3B, demonstrating additional functionality of the APOBEC3-Vif interaction apart from induction of proteasomal degradation. IMPORTANCE: APOBEC3 enzymes can potently restrict the replication of HIV-1 in the absence of HIV-1 Vif. Vif suppresses APOBEC3 action by inducing their degradation through a direct interaction with APOBEC3 enzymes and other host proteins. Vif variants from different HIV-1 strains have different effects on APOBEC3 enzymes. We used differing Vif degradation capacities of two Vif variants and various APOBEC3 enzymes with differential sensitivities to Vif to delineate determinants of the APOBEC3-Vif interaction that are required for inducing efficient degradation. Using a combined biochemical and cellular approach we identified that the strength of the APOBEC3-Vif binding interaction and the APOBEC3-Vif interface are determinants for degradation efficiency. Our results highlight the importance of using Vif variants with different degradation potential when delineating mechanisms of Vif-induced APOBEC3 degradation and identify features important for consideration in the development of HIV-1 therapies that disrupt the APOBEC3-Vif interaction.


Subject(s)
Cytosine Deaminase/antagonists & inhibitors , HIV-1/physiology , Host-Pathogen Interactions , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Cell Line , Cytidine Deaminase , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , HIV-1/immunology , Humans , Protein Binding , Proteolysis , vif Gene Products, Human Immunodeficiency Virus/immunology
9.
Curr Opin Virol ; 3(6): 692-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24246762

ABSTRACT

Primate immunodeficiency viruses, including HIV-1, are characterized by the presence of accessory genes such as vif, vpr, vpx, vpu, and nef. Current knowledge indicates that none of the primate lentiviral accessory proteins has enzymatic activity. Instead, these proteins interact with cellular ligands to either act as adapter molecules to redirect the normal function of host factors for virus-specific purposes or to inhibit a normal host function by mediating degradation or causing intracellular mislocalization/sequestration of the factors involved. This review aims at providing an update of our current understanding of how Vif, Vpu, and Vpx control the cellular restriction factors APOBEC3G, BST-2, and SAMHD1, respectively.


Subject(s)
HIV/immunology , HIV/physiology , Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/metabolism , Immune Evasion , Viral Regulatory and Accessory Proteins/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , Antigens, CD/metabolism , Cytidine Deaminase/metabolism , GPI-Linked Proteins/metabolism , Human Immunodeficiency Virus Proteins/immunology , Humans , Lentiviruses, Primate , Monomeric GTP-Binding Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Viral Regulatory and Accessory Proteins/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology
10.
Viral Immunol ; 26(1): 68-74, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23330678

ABSTRACT

DNA vaccine can induce the prolonged immune responses against the encoded antigen with the appropriate adjuvant. To study the immunogenicity of the HIV-1 vif DNA vaccine in inducing the humoral and cellular immune responses and the immunoadjuvant effect of LIGHT, which is a member of TNF superfamily and can stimulate the proliferation of naïve T cells as a co-stimulatory molecule, DNA vaccine plasmid pcDNA-Vif was constructed by inserting HIV-1 vif gene into the downstream of CMV promoter in eukaryotic expression vector pcDNA3.1(+). In vitro expression of HIV-1 Vif in pcDNA-Vif-transfected HeLa cells was confirmed in transcriptional and protein level by RT-PCR and Western blot, respectively. After BALB/c mice were injected muscularly with DNA vaccines for three times, the specific immune responses were analyzed. The data showed that anti-Vif antibody response, Vif-specific T cell proliferation, and CTL activities were induced in the mice that were inoculated with HIV-1 vif DNA vaccine plasmid. Interestingly, stronger humoral and cellular immune responses were detected in mice that were immunized with plasmid pcDNA-Vif and pcDNA-LIGHT together compared to the single immunization with plasmid pcDNA-Vif alone. Together, the results of the study suggest that candidate HIV-1 DNA vaccine can elicit HIV-1 Vif-specific immune responses in mice and that LIGHT plays the role of immunoadjuvant in co-immunization with DNA vaccine.


Subject(s)
Adjuvants, Immunologic/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Vaccines, DNA/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , Adjuvants, Immunologic/genetics , Animals , Female , HIV Antibodies/blood , HeLa Cells , Humans , Injections, Intramuscular , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , vif Gene Products, Human Immunodeficiency Virus/genetics
11.
J Biosci Bioeng ; 111(5): 597-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21277253

ABSTRACT

Immune phage antibody libraries are an attractive technology for isolating antigen-specific monoclonal antibodies (mAbs). Here we show that the immunization schedule affects the immune phage antibody library properties. We subcutaneously (s.c.) administered HIV-1 Nef and Vif antigens with different schedules (25 µg × 2 s.c. and 10 µg × 3 s.c.). The variety of isolated mAbs in 25 µg × 2 s.c. groups (Nef: 11 clones, Vif: 9 clones) was superior to that in the 10 µg × 3 s.c. groups (Nef: 2 clones, Vif: 1 clone). This finding suggests that it is important to optimize the immunization schedule for isolating a wide variety of mAbs.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Immunization Schedule , Peptide Library , nef Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Cloning, Molecular , Female , HIV Antibodies/biosynthesis , HIV Antibodies/blood , HIV Antibodies/immunology , HIV-1/immunology , Mice , Mice, Inbred BALB C , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/immunology
12.
PLoS One ; 5(7): e11436, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20625436

ABSTRACT

BACKGROUND: HIV-1-infected individuals who spontaneously control viral replication represent an example of successful containment of the AIDS virus. Understanding the anti-viral immune responses in these individuals may help in vaccine design. However, immune responses against HIV-1 are normally analyzed using HIV-1 consensus B 15-mers that overlap by 11 amino acids. Unfortunately, this method may underestimate the real breadth of the cellular immune responses against the autologous sequence of the infecting virus. METHODOLOGY AND PRINCIPAL FINDINGS: Here we compared cellular immune responses against nef and vif-encoded consensus B 15-mer peptides to responses against HLA class I-predicted minimal optimal epitopes from consensus B and autologous sequences in six patients who have controlled HIV-1 replication. Interestingly, our analysis revealed that three of our patients had broader cellular immune responses against HLA class I-predicted minimal optimal epitopes from either autologous viruses or from the HIV-1 consensus B sequence, when compared to responses against the 15-mer HIV-1 type B consensus peptides. CONCLUSION AND SIGNIFICANCE: This suggests that the cellular immune responses against HIV-1 in controller patients may be broader than we had previously anticipated.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , Immunity, Cellular/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , CD4-Positive T-Lymphocytes , Cohort Studies , Epitopes/genetics , Epitopes/immunology , Genetic Variation/genetics , Genome, Viral/genetics , HIV-1/classification , HIV-1/genetics , HIV-1/immunology , HLA-B Antigens/genetics , Immunity, Cellular/genetics , Likelihood Functions , Phylogeny , Polymerase Chain Reaction
13.
J Exp Med ; 207(1): 39-49, 2010 Jan 18.
Article in English | MEDLINE | ID: mdl-20038599

ABSTRACT

The cytidine deaminase APOBEC3G (A3G) enzyme exerts an intrinsic anti-human immunodeficiency virus (HIV) defense by introducing lethal G-to-A hypermutations in the viral genome. The HIV-1 viral infectivity factor (Vif) protein triggers degradation of A3G and counteracts this antiviral effect. The impact of A3G on the adaptive cellular immune response has not been characterized. We examined whether A3G-edited defective viruses, which are known to express truncated or misfolded viral proteins, activate HIV-1-specific (HS) CD8+ cytotoxic T lymphocytes (CTLs). To this end, we compared the immunogenicity of cells infected with wild-type or Vif-deleted viruses in the presence or absence of the cytidine deaminase. The inhibitory effect of A3G on HIV replication was associated with a strong activation of cocultivated HS-CTLs. CTL activation was particularly marked with Vif-deleted HIV and with viruses harboring A3G. Enzymatically inactive A3G mutants failed to enhance CTL activation. We also engineered proviruses bearing premature stop codons in their genome as scars of A3G editing. These viruses were not infectious but potently activated HS-CTLs. Therefore, the pool of defective viruses generated by A3G represents an underestimated source of viral antigens. Our results reveal a novel function for A3G, acting not only as an intrinsic antiviral factor but also as an inducer of the adaptive immune system.


Subject(s)
Adaptive Immunity , CD8-Positive T-Lymphocytes/immunology , Cytidine Deaminase/immunology , HIV Infections/immunology , HIV-1/immunology , Lymphocyte Activation/immunology , APOBEC-3G Deaminase , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Codon, Terminator/genetics , Codon, Terminator/immunology , Codon, Terminator/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Gene Deletion , Genes, vif/genetics , Genes, vif/immunology , HIV Infections/enzymology , HIV Infections/genetics , HIV-1/genetics , HIV-1/metabolism , Humans , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Mutation , Proviruses/genetics , Proviruses/immunology , Proviruses/metabolism , RNA Editing/genetics , RNA Editing/immunology , RNA, Viral/genetics , RNA, Viral/immunology , RNA, Viral/metabolism , Virus Replication/genetics , Virus Replication/immunology , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/immunology
14.
J Interferon Cytokine Res ; 29(9): 569-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19694548

ABSTRACT

Recent discoveries have revealed previously unappreciated complexity with which retroviruses interact with their hosts. In particular, we have become aware that many mammals, including humans, are equipped with genes encoding so-called "restriction factors," that provide considerable resistance to retroviral infection. Such antiretroviral genes are sometimes constitutively expressed, and sometimes interferon-induced. Thus they can be viewed as comprising an intrinsic immune system that provides a pre-mobilized defense against retroviral infection or, alternatively, as a specialized extension of conventional innate immunity. Antiretroviral restriction factors have evolved at an unusually rapid pace, particularly in primates, and some startling examples of evolutionary change are present in genes encoding restriction factors. Our understanding of the mechanisms by which restriction factors interfere with retroviral replication, and how their effects are avoided by certain retroviruses, is accruing, but far from complete. Such knowledge could allow for novel forms of therapeutic intervention in pathogenic retroviral infections, as well as the development of animal models of human disease.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV/physiology , Interferons/immunology , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antiviral Restriction Factors , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cytidine Deaminase , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , Evolution, Molecular , GPI-Linked Proteins , HIV/pathogenicity , HIV Infections/genetics , Humans , Immunity, Innate , Interferons/genetics , Interferons/metabolism , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Transcriptional Activation , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Virulence , Virus Assembly , vif Gene Products, Human Immunodeficiency Virus/immunology
15.
Philos Trans R Soc Lond B Biol Sci ; 364(1517): 689-703, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19008196

ABSTRACT

The intrinsic antiretroviral factor APOBEC3G (A3G) is highly active against HIV-1 and other retroviruses. In different cell types, A3G is expressed in high-molecular-mass (HMM) RNA- protein complexes or low-molecular-mass (LMM) forms displaying different biological activities. In resting CD4 T cells, a LMM form of A3G potently restricts HIV-1 infection soon after virion entry. However, when T cells are activated, LMM A3G is recruited into HMM complexes that include Staufen-containing RNA granules. These complexes are probably nucleated by the induced expression of Alu/hY retroelement RNAs that accompany T-cell activation. HMM A3G sequesters these retroelement RNAs away from the nuclear long interspersed nuclear element-derived enzymes required for Alu/hY retrotransposition. Human immunodeficiency virus (HIV) exploits this 'window of opportunity' provided by the loss of LMM A3G in activated CD4 T cells to productively infect these cells. During HIV virion formation, newly synthesized LMM A3G is preferentially encapsidated but only under conditions where Vif is absent and thus not able to target A3G for proteasome-mediated degradation. Together, these findings highlight the discrete functions of the different forms of A3G. LMM A3G opposes the external threat posed by exogenous retroviruses, while HMM A3G complexes oppose the internal threat posed by the retrotransposition of select types of retroelements.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cytidine Deaminase/immunology , HIV Infections/immunology , HIV-1/immunology , APOBEC-3G Deaminase , CD4-Positive T-Lymphocytes/immunology , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Cytoskeletal Proteins/metabolism , HIV Infections/genetics , Humans , Models, Biological , RNA-Binding Proteins/metabolism , Retroelements/genetics , Retroelements/immunology , Ribonucleoproteins/metabolism , Virion/metabolism , vif Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/metabolism
17.
Clin Vaccine Immunol ; 15(11): 1745-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18815234

ABSTRACT

We measured CD8(+) T-cell responses in 12 potentially exposed but uninfected men who have sex with men by using cytokine flow cytometry. Four of the individuals screened exhibited polyfunctional immune responses to human immunodeficiency virus type 1 Gag or Vif. The minimum cytotoxic T lymphocyte epitope was mapped in one Gag responder.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HIV Infections/immunology , HIV-1/immunology , Cytokines/biosynthesis , Epitope Mapping , Homosexuality, Male , Humans , Male , gag Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology
18.
Nat Biotechnol ; 26(10): 1187-92, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18806783

ABSTRACT

The HIV-1 protein Vif, essential for in vivo viral replication, targets the human DNA-editing enzyme, APOBEC3G (A3G), which inhibits replication of retroviruses and hepatitis B virus. As Vif has no known cellular homologs, it is an attractive, yet unrealized, target for antiviral intervention. Although zinc chelation inhibits Vif and enhances viral sensitivity to A3G, this effect is unrelated to the interaction of Vif with A3G. We identify a small molecule, RN-18, that antagonizes Vif function and inhibits HIV-1 replication only in the presence of A3G. RN-18 increases cellular A3G levels in a Vif-dependent manner and increases A3G incorporation into virions without inhibiting general proteasome-mediated protein degradation. RN-18 enhances Vif degradation only in the presence of A3G, reduces viral infectivity by increasing A3G incorporation into virions and enhances cytidine deamination of the viral genome. These results demonstrate that the HIV-1 Vif-A3G axis is a valid target for developing small molecule-based new therapies for HIV infection or for enhancing innate immunity against viruses.


Subject(s)
Anti-HIV Agents/therapeutic use , Cytidine Deaminase/immunology , Cytosine Deaminase/antagonists & inhibitors , Cytosine Deaminase/immunology , HIV Infections/immunology , HIV Infections/prevention & control , Immunity, Innate/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , APOBEC-3G Deaminase , Anti-HIV Agents/immunology , Cytidine Deaminase/genetics , HIV-1/drug effects , HIV-1/physiology , Humans , Immunity, Innate/drug effects
19.
Biosens Bioelectron ; 23(3): 384-92, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17574408

ABSTRACT

In this paper recombinant single-chain fragments (scFv-4BL), and single domain antibodies (4BL-V(H)) and (4BL-V(H)D) generated against HIV1 virion infectivity factor (Vif) are used to develop piezoimmunosensors for HIV1 recognition. Mixed self assembled monolayers were generated at the surface of gold coated crystal sensors to which scFv-4BL, 4BL-V(H), or 4BL-V(H)D were immobilized. Impedance analysis was used to discriminate interfering signals from frequency variation data and to increase the sensor sensitivity. The elimination of interfering signals enabled the quantification of the amount of immobilized protein and gave some indication on the viscoelasticity of immobilized biofilms. All the modified sensors were able to specifically recognize HIV1 Vif in liquid samples. The results indicate that lower sensitivities are obtained with 4BL-V(H) single domain antibodies, possibly due to its higher hydrophobic character. The sensitivity obtained when using scFv-4BL was reestablished when using the more hydrophilic 4BL-V(H)D single domain. 4BL-V(H)D piezoimunosensors were effective in recognizing HIV1 Vif from protein mixtures and from cell extracts of human embryonic kidney cells expressing HIV1 Vif. The results presented in this paper demonstrate the potential applicability of the developed piezoimmunosensors to monitor HIV1 infection evolution.


Subject(s)
Biosensing Techniques/methods , HIV Antibodies/immunology , Immunoassay/methods , Immunoglobulin Variable Region/immunology , vif Gene Products, Human Immunodeficiency Virus/analysis , Cell Line , Humans , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , vif Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/isolation & purification
20.
J Biotechnol ; 132(2): 142-8, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17566584

ABSTRACT

In this work three piezoelectric sensors modified with anti-HIV-1 Vif (virion infectivity factor) single fragment antibodies (4BL scFV), single domains (VH) and camelized single domains (VHD) were constructed and used to detect HIV1 Vif in liquid samples. Dithio-bis-succinimidyl-undecanoate (DSU) and 11-hydroxy-1-undecanethiol (HUT) mixed self assembled monolayers (SAM) were generated at the sensors surface onto which the antibodies were immobilized. All sensors detected specifically the target HIV1-Vif antigen in solution and no unspecific binding was monitored. Impedance analysis was performed to quantify electroacoustic and viscoelastic interferences during antibody immobilization and antigen recognition. The elimination of such interferences enabled the quantitative use of the piezoelectric immunosensors to estimate the antibody surface density as well as antigen binding and equilibrium constants. In spite of the possible limitation regarding mass transport and other related molecular phenomena, which were not considered in the binding model used, this work demonstrates the usefulness of piezoelectric biosensors in biorecognition analysis and evidences the advantages on using simultaneous impedance analysis to bring analytical significance to measured data, and thus to improve piezoelectric sensors sensitivity and applicability.


Subject(s)
Biosensing Techniques/instrumentation , HIV Antibodies , HIV-1/immunology , Recombinant Proteins , vif Gene Products, Human Immunodeficiency Virus/analysis , Electrochemistry/instrumentation , Humans , Molecular Probe Techniques/instrumentation , Quartz/chemistry , Recombinant Proteins/metabolism , vif Gene Products, Human Immunodeficiency Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL