Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
FEBS J ; 288(2): 427-433, 2021 01.
Article in English | MEDLINE | ID: mdl-32506843

ABSTRACT

Integrase strand transfer inhibitors (INSTIs) are important components of drug formulations that are used to treat people living with HIV, and second-generation INSTIs dolutegravir and bictegravir impart high barriers to the development of drug resistance. Reported 10 years ago, X-ray crystal structures of prototype foamy virus (PFV) intasome complexes explained how INSTIs bind integrase to inhibit strand transfer activity and provided initial glimpses into mechanisms of drug resistance. However, comparatively low sequence identity between PFV and HIV-1 integrases limited the depth of information that could be gleaned from the surrogate model system. Recent high-resolution structures of HIV-1 intasomes as well as intasomes from a closely related strain of simian immunodeficiency virus (SIV), which were determined using single-particle cryogenic electron microscopy, have overcome this limitation. The new structures reveal the binding modes of several advanced INSTI compounds to the HIV/SIV integrase active site and critically inform the structural basis of drug resistance. These findings will help guide the continued development of this important class of antiretroviral therapeutics.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Oxazines/chemistry , Piperazines/chemistry , Pyridones/chemistry , Simian Immunodeficiency Virus/drug effects , Amides , Animals , Catalytic Domain , Cryoelectron Microscopy , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/chemistry , HIV-1/enzymology , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Oxazines/pharmacology , Piperazines/pharmacology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pyridones/pharmacology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/enzymology , Spumavirus/chemistry , Spumavirus/drug effects , Spumavirus/enzymology
2.
J Biol Chem ; 295(50): 16975-16986, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33008888

ABSTRACT

HIV Type 1 (HIV-1) and simian immunodeficiency virus (SIV) display differential replication kinetics in macrophages. This is because high expression levels of the active host deoxynucleotide triphosphohydrolase sterile α motif domain and histidine-aspartate domain-containing protein 1 (SAMHD1) deplete intracellular dNTPs, which restrict HIV-1 reverse transcription, and result in a restrictive infection in this myeloid cell type. Some SIVs overcome SAMHD1 restriction using viral protein X (Vpx), a viral accessory protein that induces proteasomal degradation of SAMHD1, increasing cellular dNTP concentrations and enabling efficient proviral DNA synthesis. We previously reported that SAMHD1-noncounteracting lentiviruses may have evolved to harbor RT proteins that efficiently polymerize DNA, even at low dNTP concentrations, to circumvent SAMHD1 restriction. Here we investigated whether RTs from SIVmac239 virus lacking a Vpx protein evolve during in vivo infection to more efficiently synthesize DNA at the low dNTP concentrations found in macrophages. Sequence analysis of RTs cloned from Vpx (+) and Vpx (-) SIVmac239-infected animals revealed that Vpx (-) RTs contained more extensive mutations than Vpx (+) RTs. Although the amino acid substitutions were dispersed indiscriminately across the protein, steady-state and pre-steady-state analysis demonstrated that selected SIVmac239 Vpx (-) RTs are characterized by higher catalytic efficiency and incorporation efficiency values than RTs cloned from SIVmac239 Vpx (+) infections. Overall, this study supports the possibility that the loss of Vpx may generate in vivo SIVmac239 RT variants that can counteract the limited availability of dNTP substrate in macrophages.


Subject(s)
Mutation , Nucleotides/metabolism , RNA-Directed DNA Polymerase/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Viral Regulatory and Accessory Proteins/metabolism , Animals , Host-Pathogen Interactions , Kinetics , Macaca mulatta , Macrophages/metabolism , Macrophages/virology , Protein Structure, Tertiary , RNA-Directed DNA Polymerase/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/isolation & purification
3.
Eur J Pharmacol ; 884: 173327, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32726656

ABSTRACT

AIDS, a serious fatal disease caused by the human immunodeficiency virus (HIV), is an epidemic disease for which no effective vaccine has been established. The current therapeutic interventions for AIDS have limited efficacy because they are unable to clear HIV infections and the continuous occurrence of resistant HIV strains. Therefore, the exploitation of new drugs to prevent the spread of AIDS remains a high priority. In this study, the effects of icariin and its metabolite anhydroicaritin on SIV/HIV replication were investigated. In CEM × 174 cells and PBMC cells, both icariin and anhydroicaritin can significantly inhibit HIV-1 or SIVmac251 replication. Furthermore, molecular docking studies revealed that icariin and anhydroicaritin can act on both HIV reverse transcriptase and protease but could not bind to integrase. Reverse transcriptase and protease inhibition biological assays showed that both icariin and anhydroicaritin could significantly inhibit only HIV reverse transcriptase. In summary, the two compounds can significantly inhibit HIV/SIV in vitro and their targets may be mainly involved with HIV reverse transcriptase.


Subject(s)
Anti-HIV Agents/pharmacology , Benzopyrans/pharmacology , Flavonoids/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Simian Immunodeficiency Virus/drug effects , Adult , Anti-HIV Agents/chemistry , Benzopyrans/chemistry , Cell Line , HIV Protease/metabolism , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/growth & development , Humans , Male , Molecular Docking Simulation , Protease Inhibitors/pharmacology , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/growth & development , Structure-Activity Relationship , Virus Replication/drug effects
4.
PLoS One ; 13(8): e0202997, 2018.
Article in English | MEDLINE | ID: mdl-30153293

ABSTRACT

HIV mutates rapidly and infects CD4+ T cells, especially when they are activated. A vaccine targeting conserved, essential viral elements while limiting CD4+ T cell activation could be effective. Learning from natural immunity observed in a group of highly HIV-1 exposed seronegative Kenyan female sex workers, we are testing a novel candidate HIV vaccine targeting the 12 viral protease cleavage sites (PCSs) (the PCS vaccine), in comparison with a vaccine targeting full-length Gag and Env (the Gag/Env vaccine) in a Mauritian cynomolgus macaque/SIV model. In this study we evaluated these vaccines for induction of mucosal antibodies to SIV immunogens at the female genital tract. Bio-Plex and Western blot analyses of cervicovaginal lavage samples showed that both the PCS and Gag/Env vaccines can elicit mucosal IgG antibody responses to SIV immunogens. Significantly higher increase of anti-PCS antibodies was induced by the PCS vaccine than by the Gag/Env vaccine (p<0.0001). The effect of the mucosal antibody responses in protection from repeated low dose pathogenic SIVmac251 challenges is being evaluated.


Subject(s)
Gene Products, env/immunology , Gene Products, gag/immunology , Peptide Hydrolases/metabolism , Proteolysis , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Binding Sites , Cross Reactions , Female , Gene Products, env/chemistry , Gene Products, env/metabolism , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Immunization , Macaca fascicularis
5.
PLoS One ; 13(2): e0190908, 2018.
Article in English | MEDLINE | ID: mdl-29466356

ABSTRACT

Two SIVmac251-infected rhesus macaques received tenofovir/emtricitabine with raltegravir intensification. Viral rebound occurred during treatment and sequencing of reverse transcriptase and integrase genes identified multiple resistance mutations. Similar to HIV infection, antiretroviral-resistance mutations may occur in SIV-infected nonhuman primates receiving nonsuppressive ART. As ART administration to nonhuman primates is currently dramatically expanding, fueled by both cure research and the study of HIV-related comorbidities, viral resistance should be factored in the study design and data interpretation.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Animals , Anti-Retroviral Agents/administration & dosage , Disease Models, Animal , Drug Resistance, Multiple, Viral/genetics , Drug Therapy, Combination , Emtricitabine/administration & dosage , Genes, Viral , Humans , Integrases/genetics , Macaca mulatta , Mutation , RNA-Directed DNA Polymerase/genetics , Raltegravir Potassium/administration & dosage , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/enzymology , Tenofovir/administration & dosage , Viral Load/drug effects
6.
J Acquir Immune Defic Syndr ; 74(3): e67-e74, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27552154

ABSTRACT

The Population Council's microbicide gel MZC (also known as PC-1005) containing MIV-150 and zinc acetate dihydrate (ZA) in carrageenan (CG) has shown promise as a broad-spectrum microbicide against HIV, herpes simplex virus (HSV), and human papillomavirus. Previous data show antiviral activity against these viruses in cell-based assays, prevention of vaginal and rectal simian-human immunodeficiency virus reverse transcriptase (SHIV-RT) infection, and reduction of vaginal HSV shedding in rhesus macaques and also excellent antiviral activity against HSV and human papillomavirus in murine models. Recently, we demonstrated that MZC is safe and effective against SHIV-RT in macaque vaginal explants. Here we established models of ex vivo SHIV-RT/HSV-2 coinfection of vaginal mucosa and SHIV-RT infection of rectal mucosa in macaques (challenge of rectal mucosa with HSV-2 did not result in reproducible tissue infection), evaluated antiviral activity of MZC, and compared quantitative polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay readouts for monitoring SHIV-RT infection. MZC (at nontoxic dilutions) significantly inhibited SHIV-RT in vaginal and rectal mucosas and HSV-2 in vaginal mucosa when present during viral challenge. Analysis of SHIV-RT infection and MZC activity by 1-step simian immunodeficiency virus gag quantitative RT-PCR and p27 enzyme-linked immunosorbent assay demonstrated similar virus growth dynamics and MZC activity by both methods and higher sensitivity of quantitative RT-PCR. Our data provide more evidence that MZC is a promising dual compartment multipurpose prevention technology candidate.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 2, Human/drug effects , Mucous Membrane/virology , Pyridines/pharmacology , RNA-Directed DNA Polymerase/analysis , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/enzymology , Urea/analogs & derivatives , Animals , Female , Gels/pharmacology , Herpesvirus 2, Human/growth & development , Macaca , Microbial Sensitivity Tests , Models, Theoretical , Organ Culture Techniques , Rectum/virology , Simian Immunodeficiency Virus/growth & development , Urea/pharmacology , Vagina/virology
7.
Viruses ; 8(12)2016 12 01.
Article in English | MEDLINE | ID: mdl-27916968

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) protein Vpu is encoded exclusively by HIV-1 and related simian immunodeficiency viruses (SIVs). The transmembrane domain of the protein has dual functions: it counteracts the human restriction factor tetherin and forms a cation channel. Since these two functions are causally unrelated it remains unclear whether the channel activity has any relevance for viral release and replication. Here we examine structure and function correlates of different Vpu homologs from HIV-1 and SIV to understand if ion channel activity is an evolutionary conserved property of Vpu proteins. An electrophysiological testing of Vpus from different HIV-1 groups (N and P) and SIVs from chimpanzees (SIVcpz), and greater spot-nosed monkeys (SIVgsn) showed that they all generate channel activity in HEK293T cells. This implies a robust and evolutionary conserved channel activity and suggests that cation conductance may also have a conserved functional significance.


Subject(s)
HIV-1/enzymology , Human Immunodeficiency Virus Proteins/metabolism , Ion Channels/metabolism , Simian Immunodeficiency Virus/enzymology , Viral Regulatory and Accessory Proteins/metabolism , Cations/metabolism , Cell Line , Electrophysiological Phenomena , Humans
8.
Arch Virol ; 160(12): 3085-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26350979

ABSTRACT

Human integrase interactor 1 (INI1/SMARCB1/SNF5) is a chromatin-remodeling molecule that binds to HIV-1 integrase and enhances proviral DNA integration. INI1 is also known as a tumor suppressor gene and has been found to be mutated in several aggressive tumors such as rhabdoid and lymphoid tumors. To study the function of simian INI1, we screened and cloned simian INI1 cDNA from B lymphoma cells of rhesus monkeys using RT-PCR. Sequence analysis showed 23 single nucleotide differences compared to the human ortholog, which, however, did not result in amino acid changes, and the amino acid sequence is therefore 100% conserved between human and simian INI1. Two alternatively spliced isoforms, INI1a and INI1b, were also found in simian INI1. These two isoforms did not show any functional difference in HIV-1 proviral DNA integration and nuclear localization, suggesting that the specificity of simian INI1 would not be a factor preventing HIV-1 infection of a simian host. Nevertheless, INI1b is expressed only in established cancer cell lines such as Jurkat and COS-7 cells, and not in primary cells, suggesting that INIlb could be an indicator of cell transformation.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HIV Infections/metabolism , HIV Integrase/metabolism , HIV-1/enzymology , Macaca mulatta/genetics , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/enzymology , Transcription Factors/chemistry , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Chromosomal Proteins, Non-Histone/metabolism , Cloning, Molecular , DNA-Binding Proteins/metabolism , HIV Infections/genetics , HIV Infections/virology , HIV Integrase/genetics , HIV-1/genetics , Humans , Macaca mulatta/metabolism , Macaca mulatta/virology , Molecular Sequence Data , Protein Binding , SMARCB1 Protein , Sequence Alignment , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Transcription Factors/metabolism
9.
Antimicrob Agents Chemother ; 59(7): 3829-37, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870063

ABSTRACT

The transmission of both cell-free and cell-associated immunodeficiency viruses has been demonstrated directly in multiple animal species and possibly occurs in humans, as suggested by genotyping of the infecting human immunodeficiency virus (HIV) in acutely infected women and in semen from their partners. Therefore, a microbicide may need to block both mechanisms of HIV transmission to achieve maximum efficacy. To date, most of the preclinical evaluation of candidate microbicides has been performed using cell-free HIV. New models of mucosal transmission of cell-associated HIV are needed to evaluate candidate microbicide performance. The MIV-150/zinc acetate/carrageenan (MZC) gel protects Depo-Provera-treated macaques against cell-free simian-human immunodeficiency virus reverse transcriptase (SHIV-RT) infection when applied vaginally up to 8 h before challenge. We recently demonstrated the potent activity of MZC gel against cell-free SHIV-RT in macaque vaginal explants. In the current study, we established a cell-associated SHIV-RT infection model of macaque vaginal tissues and tested the activity of MZC gel in this model. MZC gel protected tissues against cell-associated SHIV-RT infection when present at the time of viral exposure or when applied up to 4 days prior to viral challenge. These data support clinical testing of the MZC gel. Overall, our ex vivo model of cell-associated SHIV-RT infection in macaque vaginal mucosa complements the cell-free infection models, providing tools for prioritization of products that block both modes of HIV transmission.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , Pyridines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Urea/analogs & derivatives , Zinc Acetate/therapeutic use , Administration, Intravaginal , Animals , Antiviral Agents/therapeutic use , Cervix Uteri/virology , Female , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HIV-1/enzymology , HIV-1/genetics , Macaca mulatta , Monkey Diseases/drug therapy , Monkey Diseases/virology , Mucous Membrane/virology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/genetics , Urea/therapeutic use , Vaginal Creams, Foams, and Jellies/therapeutic use
10.
Retrovirology ; 11: 111, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25524560

ABSTRACT

BACKGROUND: Host SAM domain and HD domain-containing protein 1 (SAMHD1) suppresses reverse transcription kinetics of HIV-1 in nondividing cells such as macrophages by hydrolyzing and nearly depleting cellular dNTPs, which are the substrates of viral reverse transcriptase (RT). However, unlike HIV-1, HIV-2 and SIVsm encode viral protein X (Vpx), which counteracts the dNTPase activity of SAMHD1 and elevates dNTP concentration, allowing the viruses to replicate under abundant dNTP conditions even in nondividing cells. FINDINGS: Here we tested whether RTs of these Vpx coding and noncoding lentiviruses display different enzyme kinetic profiles in response to dNTP concentrations. For this test, we characterized an extensive collection of RTs from 7 HIV-1 strains, 4 HIV-2 strains and 7 SIV strains, and determined their steady-state kinetic parameters. The K m values of all HIV-1 RTs were consistently low and close to the low dNTP concentrations found in macrophages. However, the K m values of SIV and HIV-2 RTs were not only higher than those of HIV-1 RTs but also varied significantly, indicating that HIV-2/SIV RTs require higher dNTP concentrations for efficient DNA synthesis, compared to HIV-1 RT. However, the k cat values of all eighteen lentiviral RTs were very similar. CONCLUSIONS: Our biochemical analysis supports the hypothesis that the enzymological properties, particularly, K m values, of lentivirus RTs, are mechanistically tied with the cellular dNTP availability in nondividing target cells, which is controlled by SAMHD1 and Vpx.


Subject(s)
HIV-1/enzymology , HIV-2/enzymology , RNA-Directed DNA Polymerase/metabolism , Simian Immunodeficiency Virus/enzymology , Kinetics , Nucleotides/metabolism
11.
AIDS Res Hum Retroviruses ; 30(11): 1117-24, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25354024

ABSTRACT

Epidemiological studies suggest that prevalent herpes simplex virus type 2 (HSV-2) infection increases the risk of HIV acquisition, underscoring the need to develop coinfection models to evaluate promising prevention strategies. We previously established a single high-dose vaginal coinfection model of simian human immunodeficiency virus (SHIV)/HSV-2 in Depo-Provera (DP)-treated macaques. However, this model does not appropriately mimic women's exposure. Repeated limiting dose SHIV challenge models are now used routinely to test prevention strategies, yet, at present, there are no reports of a repeated limiting dose cochallenge model in which to evaluate products targeting HIV and HSV-2. Herein, we show that 20 weekly cochallenges with 2-50 TCID50 simian human immunodeficiency virus reverse transcriptase (SHIV-RT) and 10(7) pfu HSV-2 results in infection with both viruses (4/6 SHIV-RT, 6/6 HSV-2). The frequency and level of vaginal HSV-2 shedding were significantly greater in the repeated exposure model compared to the single high-dose model (p<0.0001). We used this new model to test the Council's on-demand microbicide gel, MZC, which is active against SHIV-RT in DP-treated macaques and HSV-2 and human papillomavirus (HPV) in mice. While MZC reduced SHIV and HSV-2 infections in our repeated limiting dose model when cochallenging 8 h after each gel application, a barrier effect of carrageenan (CG) that was not seen in DP-treated animals precluded evaluation of the significance of the antiviral activity of MZC. Both MZC and CG significantly (p<0.0001) reduced the frequency and level of vaginal HSV-2 shedding compared to no gel treatment. This validates the use of this repeated limiting dose cochallenge model for testing products targeting HIV and HSV-2.


Subject(s)
Anti-Infective Agents/administration & dosage , Coinfection/virology , HIV Reverse Transcriptase/metabolism , Herpes Genitalis/virology , Herpesvirus 2, Human/growth & development , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , Animals , Contraceptive Agents, Female/administration & dosage , Disease Models, Animal , Female , Herpes Genitalis/complications , Macaca mulatta , Medroxyprogesterone Acetate/administration & dosage , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus/enzymology , Treatment Outcome , Vagina/virology , Vaginal Creams, Foams, and Jellies/administration & dosage , Virus Shedding
12.
Viral Immunol ; 27(10): 512-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25343523

ABSTRACT

As a prelude to immunization studies in nonhuman primates, we compared in mice the immunogenicity of a simian immunodeficiency virus (SIV)-based integrase (IN)-defective lentiviral vector (IDLV) encoding the model antigen-enhanced green fluorescence protein (eGFP) in the presence or absence of the murine granulocyte-macrophage colony-stimulating factor (mGM-CSF) expressed from an internal ribosomal entry site (IRES) sequence. BALB/c mice were immunized once intramuscularly with IDLV expressing eGFP alone or eGFP and mGM-CSF and immune responses were evaluated up to 90 days from the single intramuscular immunization. Results indicated that the mGM-CSF was unable to improve the magnitude and quality of the immune response against the eGFP transgene in the context of the SIV-based IDLV, as evaluated by enzyme-linked immunosorbent spot (ELISPOT) assays for interferon-γ (IFN-γ) and by intracellular cytokine staining for IFN-γ, interleukin-2 (IL-2), and tumor necrosis factor-alpha (TNF-α). These findings suggest that for vaccination purposes, the presence of mGM-CSF expressed after the IRES in a SIV-based IDLV system does not favor the improvement of the immunological response against the transgene of interest. Further studies should investigate whether the selection of a different cytokine gene might improve the immune response against the transgene.


Subject(s)
Drug Carriers , Gene Expression , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Simian Immunodeficiency Virus/genetics , T-Lymphocytes/immunology , Viral Vaccines/immunology , Animals , Enzyme-Linked Immunospot Assay , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Injections, Intramuscular , Integrases/deficiency , Interferon-gamma/analysis , Interleukin-2/analysis , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Simian Immunodeficiency Virus/enzymology , Staining and Labeling , Tumor Necrosis Factor-alpha/analysis , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
13.
J Virol ; 88(17): 9683-92, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24920794

ABSTRACT

UNLABELLED: Studies on the in vitro susceptibility of SIV to integrase strand transfer inhibitors (INSTIs) have been rare. In order to determine the susceptibility of SIVmac239 to INSTIs and characterize the genetic pathways that might lead to drug resistance, we inserted various integrase (IN) mutations that had been selected with HIV under drug pressure with raltegravir (RAL), elvitegravir (EVG), and dolutegravir (DTG) into the IN gene of SIV. We evaluated the effects of these mutations on SIV susceptibility to INSTIs and on viral infectivity. Sequence alignments of SIVmac239 IN with various HIV-1 isolates showed a high degree of homology and conservation of each of the catalytic triad and the key residues involved in drug resistance. Each of the G118R, Y143R, Q148R, R263K, and G140S/Q148R mutations, when introduced into SIV, impaired infectiousness and replication fitness compared to wild-type virus. Using TZM-bl cells, we demonstrated that the Q148R and N155H mutational pathways conferred resistance to EVG (36- and 62-fold, respectively), whereas R263K also displayed moderate resistance to EVG (12-fold). In contrast, Y143R, Q148R, and N155H all yielded low levels of resistance to RAL. The combination of G140S/Q148R conferred high-level resistance to both RAL and EVG (>300- and 286-fold, respectively). DTG remained fully effective against all site-directed mutants except G118R and R263K. Thus, HIV INSTI mutations, when inserted into SIV, resulted in a similar phenotype. These findings suggest that SIV and HIV may share similar resistance pathways profiles and that SIVmac239 could be a useful nonhuman primate model for studies of HIV resistance to INSTIs. IMPORTANCE: The goal of our project was to establish whether drug resistance against integrase inhibitors in SIV are likely to be the same as those responsible for drug resistance in HIV. Our data answer this question in the affirmative and show that SIV can probably serve as a good animal model for studies of INSTIs and as an early indicator for possible emergent mutations that may cause treatment failure. An SIV-primate model remains an invaluable tool for investigating questions related to the potential role of INSTIs in HIV therapy, transmission, and pathogenesis, and the present study will facilitate each of the above.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , HIV Integrase/genetics , HIV-1/drug effects , HIV-1/enzymology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/enzymology , Amino Acid Substitution , Animals , Cells, Cultured , HIV Integrase/metabolism , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Macaca mulatta , Microbial Sensitivity Tests , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oxazines , Piperazines , Pyridones , Pyrrolidinones/pharmacology , Quinolones/pharmacology , Raltegravir Potassium , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Virus Replication
14.
PLoS One ; 9(4): e94547, 2014.
Article in English | MEDLINE | ID: mdl-24740100

ABSTRACT

Prevalent infection with human herpes simplex 2 (HSV-2) or human papillomavirus (HPV) is associated with increased human immunodeficiency virus (HIV) acquisition. Microbicides that target HIV as well as these sexually transmitted infections (STIs) may more effectively limit HIV incidence. Previously, we showed that a microbicide gel (MZC) containing MIV-150, zinc acetate (ZA) and carrageenan (CG) protected macaques against simian-human immunodeficiency virus (SHIV-RT) infection and that a ZC gel protected mice against HSV-2 infection. Here we evaluated a modified MZC gel (containing different buffers, co-solvents, and preservatives suitable for clinical testing) against both vaginal and rectal challenge of animals with SHIV-RT, HSV-2 or HPV. MZC was stable and safe in vitro (cell viability and monolayer integrity) and in vivo (histology). MZC protected macaques against vaginal (p<0.0001) SHIV-RT infection when applied up to 8 hours (h) prior to challenge. When used close to the time of challenge, MZC prevented rectal SHIV-RT infection of macaques similar to the CG control. MZC significantly reduced vaginal (p<0.0001) and anorectal (p = 0.0187) infection of mice when 10(6) pfu HSV-2 were applied immediately after vaginal challenge and also when 5×10(3) pfu were applied between 8 h before and 4 h after vaginal challenge (p<0.0248). Protection of mice against 8×10(6) HPV16 pseudovirus particles (HPV16 PsV) was significant for MZC applied up to 24 h before and 2 h after vaginal challenge (p<0.0001) and also if applied 2 h before or after anorectal challenge (p<0.0006). MZC provides a durable window of protection against vaginal infection with these three viruses and, against HSV-2 and HPV making it an excellent candidate microbicide for clinical use.


Subject(s)
Alphapapillomavirus/drug effects , Anti-Infective Agents/pharmacology , Herpesvirus 2, Human/drug effects , Simian Immunodeficiency Virus/drug effects , Alphapapillomavirus/physiology , Anal Canal/drug effects , Anal Canal/virology , Animals , Anti-Infective Agents/chemistry , Caco-2 Cells , Carrageenan/chemistry , Carrageenan/pharmacology , Female , Gels , HeLa Cells , Herpes Simplex/prevention & control , Herpes Simplex/virology , Herpesvirus 2, Human/physiology , Host-Pathogen Interactions/drug effects , Humans , Macaca mulatta , Mice, Inbred BALB C , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Pyridines/chemistry , Pyridines/pharmacology , Rectum/drug effects , Rectum/virology , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/physiology , Treatment Outcome , Urea/analogs & derivatives , Urea/chemistry , Urea/pharmacology , Vagina/drug effects , Vagina/virology , Zinc Acetate/chemistry , Zinc Acetate/pharmacology
15.
PLoS One ; 9(2): e88258, 2014.
Article in English | MEDLINE | ID: mdl-24505452

ABSTRACT

Highly active antiretroviral therapy (HAART) significantly reduces HIV-1 replication and prevents progression to AIDS. However, residual low-level viremia (LLV) persists and long-lived viral reservoirs are maintained in anatomical sites. These reservoirs permit a recrudescence of viremia upon cessation of therapy and thus HAART must be maintained indefinitely. HIV-1 reservoirs include latently infected resting memory CD4⁺ T-cells and macrophages which may contribute to residual viremia. It has not been conclusively determined if a component of LLV may also be due to residual replication in cells with sub-therapeutic drug levels and/or long-lived chronically infected cells. In this study, RT-SHIV(mac239) diversity was characterized in five rhesus macaques that received a five-drug HAART regimen [tenofovir, emtricitabine, zidovudine, amdoxovir, (A, C, T, G nucleoside analogs) and the non-nucleoside reverse transcriptase (RT) inhibitor efavirenz]. Before maximal viral load suppression, longitudinal plasma viral RNA RT diversity was analyzed using a 454 sequencer. After suppression, LLV RT diversity (amino acids 65-210) was also assessed. LLV samples had viral levels less than our standard detection limit (50 viral RNA copies/mL) and few transient blips <200 RNA copies/mL. HAART was discontinued in three macaques after 42 weeks of therapy resulting in viral rebound. The level of viral divergence and the prevalence of specific alleles in LLV was similar to pre-suppression viremia. While some LLV sequences contained mutations not observed in the pre-suppression profile, LLV was not characterized by temporal viral evolution or apparent selection of drug resistance mutations. Similarly, resistance mutations were not detected in the viral rebound population. Interestingly, one macaque maintained a putative LLV predominant plasma clone sequence. Together, these results suggest that residual replication did not markedly contribute to LLV and that this model mimics the prevalence and phylogenetic characteristics of LLV during human HAART. Therefore, this model may be ideal for testing HIV-1 eradication strategies.


Subject(s)
Antiretroviral Therapy, Highly Active , Macaca mulatta/virology , Monkey Diseases/drug therapy , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Viremia/drug therapy , Animals , Anti-Retroviral Agents/therapeutic use , Disease Models, Animal , Female , Gene Frequency , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans , Monkey Diseases/blood , Monkey Diseases/genetics , Monkey Diseases/virology , Mutation , Phylogeny , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/genetics , Viral Load/drug effects , Viremia/blood , Viremia/genetics , Viremia/virology
16.
PLoS One ; 9(1): e86997, 2014.
Article in English | MEDLINE | ID: mdl-24498008

ABSTRACT

RT-SHIV is a chimera of simian immunodeficiency virus (SIV) containing the reverse transcriptase (RT)-encoding region of human immunodeficiency virus type 1 (HIV-1) within the backbone of SIVmac239. It has been used in a non-human primate model for studies of non-nucleoside RT inhibitors (NNRTI) and highly active antiretroviral therapy (HAART). We and others have identified several mutations that arise in the "foreign" HIV-1 RT of RT-SHIV during in vivo replication. In this study we catalogued amino acid substitutions in the HIV-1 RT and in regions of the SIV backbone with which RT interacts that emerged 30 weeks post-infection from seven RT-SHIV-infected rhesus macaques. The virus set points varied from relatively high virus load, moderate virus load, to undetectable virus load. The G196R substitution in RT was detected from 6 of 7 animals at week 4 post-infection and remained in virus from 4 of 6 animals at week 30. Virus from four high virus load animals showed several common mutations within RT, including L74V or V75L, G196R, L214F, and K275R. The foreign RT from high virus load isolates exhibited as much variation as that of the highly variable envelope surface glycoprotein, and 10-fold higher than that of the native RT of SIVmac239. Isolates from moderate virus load animals showed much less variation in the foreign RT than the high virus load isolates. No variation was found in SIVmac239 genes known to interact with RT. Our results demonstrate substantial adaptation of the foreign HIV-1 RT in RT-SHIV-infected macaques, which most likely reflects selective pressure upon the foreign RT to attain optimal activity within the context of the chimeric RT-SHIV and the rhesus macaque host.


Subject(s)
HIV Reverse Transcriptase/genetics , Mutation , Recombinant Fusion Proteins/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , Amino Acid Substitution , Animals , Antiretroviral Therapy, Highly Active , Cells, Cultured , Female , Genome, Viral/genetics , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , Host-Pathogen Interactions/genetics , Humans , Macaca mulatta , Male , Models, Molecular , Protein Structure, Tertiary , RNA, Viral/blood , RNA, Viral/genetics , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Inhibitors , Simian Acquired Immunodeficiency Syndrome/enzymology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Viral Load/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
17.
PLoS One ; 8(12): e84275, 2013.
Article in English | MEDLINE | ID: mdl-24367650

ABSTRACT

Although antiretroviral therapy (ART) can suppress HIV-1 replication sufficiently to eliminate measurable plasma viremia, infected cells remain and ensure viral recrudescence after discontinuation of ART. We used a macaque model of HIV-1/AIDS to evaluate the location of infected cells during ART. Twelve macaques were infected with RT-SHIVmne, a SIV containing HIV-1 reverse transcriptase, conferring sensitivity to non-nucleoside reverse transcriptase inhibitors (NNRTIs). Ten to fourteen weeks post-infection, 6 animals were treated with 3 or 4 antiretroviral drugs for 17-20 weeks; 6 control animals remained untreated. Viral DNA (vDNA) and RNA (vRNA) were measured in peripheral blood mononuclear cells (PBMC) and at necropsy in multiple tissues by quantitative PCR and RT-PCR. The majority of virally infected cells were located in lymphoid tissues with variable levels in the gastrointestinal tract of both treated and untreated animals. Tissue viral DNA levels correlated with week 1 plasma viremia, suggesting that tissues that harbor proviral DNA are established within the first week of infection. PBMC vDNA levels did not correlate with plasma viremia or tissue levels of vDNA. vRNA levels were high in lymphoid and gastrointestinal tissues of the untreated animals; animals on ART had little vRNA expressed in tissues and virus could not be cultured from lymph node resting CD4+ cells after 17-20 weeks on ART, indicating little or no ongoing viral replication. Strategies for eradication of HIV-1 will need to target residual virus in ART suppressed individuals, which may not be accurately reflected by frequencies of infected cells in blood.


Subject(s)
Anti-Retroviral Agents/pharmacology , HIV Reverse Transcriptase/metabolism , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/physiology , Virus Latency/drug effects , Animals , DNA, Viral/metabolism , Gene Expression Regulation, Viral/drug effects , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Macaca , Male , RNA, Viral/metabolism , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/genetics , Viremia/virology , Virus Replication/drug effects
18.
PLoS Pathog ; 9(1): e1003135, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23359341

ABSTRACT

Naturally circulating lentiviruses are abundant in African primate species today, yet their origins and history of transmitting between hosts remain obscure. As a means to better understand the age of primate lentiviruses, we analyzed primate genomes for signatures of lentivirus-driven evolution. Specifically, we studied the adaptive evolution of host restriction factor APOBEC3G (A3G) in Old World Monkey (OWM) species. We find recurrent mutation of A3G in multiple primate lineages at sites that determine susceptibility to antagonism by the lentiviral accessory protein Vif. Using a broad panel of SIV Vif isolates, we demonstrate that natural variation in OWM A3G confers resistance to Vif-mediated degradation, suggesting that adaptive variants of the host factor were selected upon exposure to pathogenic lentiviruses at least 5-6 million years ago (MYA). Furthermore, in members of the divergent Colobinae subfamily of OWM, a multi-residue insertion event in A3G that arose at least 12 MYA blocks the activity of Vif, suggesting an even more ancient origin of SIV. Moreover, analysis of the lentiviruses associated with Colobinae monkeys reveal that the interface of the A3G-Vif interaction has shifted and given rise to a second genetic conflict. Our analysis of virus-driven evolution describes an ancient yet ongoing genetic conflict between simian primates and lentiviruses on a million-year time scale.


Subject(s)
Cercopithecidae/virology , Cytidine Deaminase/genetics , Evolution, Molecular , Gene Products, vif/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , Adaptive Immunity/genetics , Animals , Disease Resistance/genetics , Gene Expression Regulation, Viral , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Molecular Sequence Data , Mutation , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Virus Replication
19.
J Virol ; 86(23): 12525-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22933296

ABSTRACT

It has been proposed that most drug-resistant mutants, resulting from a single-nucleotide change, exist at low frequency in human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) populations in vivo prior to the initiation of antiretroviral therapy (ART). To test this hypothesis and to investigate the emergence of resistant mutants with drug selection, we developed a new ultrasensitive allele-specific PCR (UsASP) assay, which can detect drug resistance mutations at a frequency of ≥0.001% of the virus population. We applied this assay to plasma samples obtained from macaques infected with an SIV variant containing HIV-1 reverse transcriptase (RT) (RT-simian-human immunodeficiency [SHIV](mne)), before and after they were exposed to a short course of efavirenz (EFV) monotherapy. We detected RT inhibitor (RTI) resistance mutations K65R and M184I but not K103N in 2 of 2 RT-SHIV-infected macaques prior to EFV exposure. After three doses over 4 days of EFV monotherapy, 103N mutations (AAC and AAT) rapidly emerged and increased in the population to levels of ∼20%, indicating that they were present prior to EFV exposure. The rapid increase of 103N mutations from <0.001% to 20% of the viral population indicates that the replicating virus population size in RT-SHIV-infected macaques must be 10(6) or more infected cells per replication cycle.


Subject(s)
Benzoxazines/pharmacology , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , Macaca nemestrina , Polymerase Chain Reaction/methods , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Alkynes , Alleles , Animals , Benzoxazines/therapeutic use , Cyclopropanes , Mutation/drug effects , Mutation/genetics , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/genetics
20.
Retrovirology ; 9: 57, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22805180

ABSTRACT

BACKGROUND: We reported previously that while prolonged tenofovir monotherapy of macaques infected with virulent simian immunodeficiency virus (SIV) resulted invariably in the emergence of viral mutants with reduced in vitro drug susceptibility and a K65R mutation in reverse transcriptase, some animals controlled virus replication for years. Transient CD8+ cell depletion or short-term tenofovir interruption within 1 to 5 years of treatment demonstrated that a combination of CD8+ cell-mediated immune responses and continued tenofovir therapy was required for sustained suppression of viremia. We report here follow-up data on 5 such animals that received tenofovir for 8 to 14 years. RESULTS: Although one animal had a gradual increase in viremia from 3 years onwards, the other 4 tenofovir-treated animals maintained undetectable viremia with occasional viral blips (≤ 300 RNA copies/ml plasma). When tenofovir was withdrawn after 8 to 10 years from three animals with undetectable viremia, the pattern of occasional episodes of low viremia (≤ 3600 RNA/ml plasma) continued throughout the 10-month follow-up period. These animals had low virus levels in lymphoid tissues, and evidence of multiple SIV-specific immune responses. CONCLUSION: Under certain conditions (i.e., prolonged antiviral therapy initiated early after infection; viral mutants with reduced drug susceptibility) a virus-host balance characterized by strong immunologic control of virus replication can be achieved. Although further research is needed to translate these findings into clinical applications, these observations provide hope for a functional cure of HIV infection via immunotherapeutic strategies that boost antiviral immunity and reduce the need for continuous antiretroviral therapy.


Subject(s)
Adenine/analogs & derivatives , Organophosphonates/pharmacology , RNA-Directed DNA Polymerase/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/pathogenicity , Virus Replication , Adenine/immunology , Adenine/pharmacology , Alleles , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Formation , Antiviral Agents/immunology , Antiviral Agents/pharmacology , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Genes, MHC Class I , Genotyping Techniques , Immunity, Cellular , Lymphocyte Activation , Macaca mulatta , Neutralization Tests , Organophosphonates/immunology , RNA, Viral/blood , RNA-Directed DNA Polymerase/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Tenofovir , Time Factors , Treatment Outcome , Viremia/pathology , Viremia/virology
SELECTION OF CITATIONS
SEARCH DETAIL