Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nature ; 563(7733): E33, 2018 11.
Article in English | MEDLINE | ID: mdl-30315222

ABSTRACT

In this Brief Communications Arising Comment, the first three authors (Osuna, Lim and Kublin) should have been listed as equally contributing authors; this has been corrected online.

2.
PLoS Comput Biol ; 16(10): e1008241, 2020 10.
Article in English | MEDLINE | ID: mdl-33001979

ABSTRACT

In order to assess the efficacy of novel HIV-1 treatments leading to a functional cure, the time to viral rebound is frequently used as a surrogate endpoint. The longer the time to viral rebound, the more efficacious the therapy. In support of such an approach, mathematical models serve as a connection between the size of the latent reservoir and the time to HIV-1 rebound after treatment interruption. The simplest of such models assumes that a single successful latent cell reactivation event leads to observable viremia after a period of exponential viral growth. Here we consider a generalization developed by Pinkevych et al. and Hill et al. of this simple model in which multiple reactivation events can occur, each contributing to the exponential growth of the viral load. We formalize and improve the previous derivation of the dynamics predicted by this model, and use the model to estimate relevant biological parameters from SIV rebound data. We confirm a previously described effect of very early antiretroviral therapy (ART) initiation on the rate of recrudescence and the viral load growth rate after treatment interruption. We find that every day ART initiation is delayed results in a 39% increase in the recrudescence rate (95% credible interval: [18%, 62%]), and a 11% decrease of the viral growth rate (95% credible interval: [4%, 20%]). We show that when viral rebound occurs early relative to the viral load doubling time, a model with multiple successful reactivation events fits the data better than a model with only a single successful reactivation event.


Subject(s)
Anti-Retroviral Agents , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Virus Activation , Animals , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , Biomarkers , Computational Biology , Computer Simulation , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology , Viral Load/drug effects , Virus Activation/drug effects , Virus Activation/physiology
3.
Nature ; 512(7512): 74-7, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25042999

ABSTRACT

The viral reservoir represents a critical challenge for human immunodeficiency virus type 1 (HIV-1) eradication strategies. However, it remains unclear when and where the viral reservoir is seeded during acute infection and the extent to which it is susceptible to early antiretroviral therapy (ART). Here we show that the viral reservoir is seeded rapidly after mucosal simian immunodeficiency virus (SIV) infection of rhesus monkeys and before systemic viraemia. We initiated suppressive ART in groups of monkeys on days 3, 7, 10 and 14 after intrarectal SIVMAC251 infection. Treatment with ART on day 3 blocked the emergence of viral RNA and proviral DNA in peripheral blood and also substantially reduced levels of proviral DNA in lymph nodes and gastrointestinal mucosa as compared with treatment at later time points. In addition, treatment on day 3 abrogated the induction of SIV-specific humoral and cellular immune responses. Nevertheless, after discontinuation of ART following 24 weeks of fully suppressive therapy, virus rebounded in all animals, although the monkeys that were treated on day 3 exhibited a delayed viral rebound as compared with those treated on days 7, 10 and 14. The time to viral rebound correlated with total viraemia during acute infection and with proviral DNA at the time of ART discontinuation. These data demonstrate that the viral reservoir is seeded rapidly after intrarectal SIV infection of rhesus monkeys, during the 'eclipse' phase, and before detectable viraemia. This strikingly early seeding of the refractory viral reservoir raises important new challenges for HIV-1 eradication strategies.


Subject(s)
Macaca mulatta/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , Viral Load , Viremia/virology , Animals , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , Carrier State/drug therapy , Carrier State/virology , DNA, Viral/analysis , DNA, Viral/biosynthesis , DNA, Viral/blood , Disease Models, Animal , Female , Kinetics , Macaca mulatta/immunology , Male , Proviruses/genetics , RNA, Viral/blood , Rectum/virology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Time Factors , Treatment Failure , Viral Load/drug effects , Viremia/drug therapy , Virus Replication/drug effects
4.
Proc Natl Acad Sci U S A ; 114(33): 8847-8852, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28765371

ABSTRACT

The recent outbreak of Zika virus (ZIKV) has been associated with fetal abnormalities and neurological complications, prompting global concern. Here we present a mathematical analysis of the within-host dynamics of plasma ZIKV burden in a nonhuman primate model, allowing for characterization of the growth and clearance of ZIKV within individual macaques. We estimate that the eclipse phase for ZIKV, the time between cell infection and viral production, is most likely short (∼4 h), the median within-host basic reproductive number R0 is 10.7, the rate of viral production is rapid (>25,000 virions d-1), and the lifetime of an infected cell while producing virus is ∼5 h. We also estimate that the minimum number of virions produced by an infected cell over its lifetime is ∼5,500. We assess the potential effect of an antiviral treatment that blocks viral replication, showing that the median time to undetectable plasma viral load (VL) can be reduced from ∼5 d to ∼3 d with a drug concentration ∼15 times the drug's EC50 when treatment is given prophylactically starting at the time of infection. In the case of favipiravir, a polymerase inhibitor with activity against ZIKV, we predict a dose of 150 mg/kg given twice a day initiated at the time of infection can reduce the peak median VL by ∼3 logs and shorten the time to undetectable median VL by ∼2 d, whereas treatment given 2 d postinfection is mostly ineffective in accelerating plasma VL loss in macaques.


Subject(s)
Antiviral Agents/pharmacology , Models, Biological , Viral Load , Virus Replication/drug effects , Zika Virus Infection/blood , Zika Virus/physiology , Animals , Macaca mulatta
6.
Nature ; 503(7475): 224-8, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24172905

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1)-specific monoclonal antibodies with extraordinary potency and breadth have recently been described. In humanized mice, combinations of monoclonal antibodies have been shown to suppress viraemia, but the therapeutic potential of these monoclonal antibodies has not yet been evaluated in primates with an intact immune system. Here we show that administration of a cocktail of HIV-1-specific monoclonal antibodies, as well as the single glycan-dependent monoclonal antibody PGT121, resulted in a rapid and precipitous decline of plasma viraemia to undetectable levels in rhesus monkeys chronically infected with the pathogenic simian-human immunodeficiency virus SHIV-SF162P3. A single monoclonal antibody infusion afforded up to a 3.1 log decline of plasma viral RNA in 7 days and also reduced proviral DNA in peripheral blood, gastrointestinal mucosa and lymph nodes without the development of viral resistance. Moreover, after monoclonal antibody administration, host Gag-specific T-lymphocyte responses showed improved functionality. Virus rebounded in most animals after a median of 56 days when serum monoclonal antibody titres had declined to undetectable levels, although, notably, a subset of animals maintained long-term virological control in the absence of further monoclonal antibody infusions. These data demonstrate a profound therapeutic effect of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys as well as an impact on host immune responses. Our findings strongly encourage the investigation of monoclonal antibody therapy for HIV-1 in humans.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/physiology , Animals , DNA, Viral/blood , HIV Antibodies/immunology , Macaca mulatta , T-Lymphocytes/immunology , Viremia/therapy
7.
Immunology ; 155(4): 458-466, 2018 12.
Article in English | MEDLINE | ID: mdl-30014618

ABSTRACT

Antiretroviral therapy (ART) for the treatment of human immunodeficiency virus (HIV) infection represents a major breakthrough in the treatment of HIV/acquired immune-deficiency syndrome. However, it remains unclear how ART influences virus-specific immune responses and understanding this is important for developing novel cure and eradication interventions for HIV-1. In the present study, we evaluate how ART impacts T-cell and antibody responses in simian immunodeficiency virus (SIV) -infected rhesus macaques. We evaluated CD4 and CD8 T-cell responses by multiparameter flow cytometry, viral loads by quantitative RT-PCR by a two-step process using SIV-specific primers and antibody neutralization function by luciferase-based TZM-bl assays. We demonstrate that macaques treated with ART exhibit phenotypic and qualitative effects on T-cell and antibody responses. Macaques on ART exhibited low numbers of virus-specific T-cell responses, and these responses appeared to be partially biased towards central memory subsets. More importantly, there were significantly reduced neutralizing antibody responses in macaques treated with ART. Collectively, these data improve the understanding of how virus-specific immune responses are generated during ART, and suggest the potential importance of therapeutic vaccines to maintain adaptive immunity during treated infection.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Differentiation/immunology , Female , Lymphocyte Activation/immunology , Macaca mulatta , Male , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , T-Lymphocyte Subsets/cytology , Viral Load/immunology
8.
PLoS Pathog ; 12(2): e1005431, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26849216

ABSTRACT

Simian-human immunodeficiency virus (SHIV) challenge stocks are critical for preclinical testing of vaccines, antibodies, and other interventions aimed to prevent HIV-1. A major unmet need for the field has been the lack of a SHIV challenge stock expressing circulating recombinant form 01_AE (CRF01_AE) env sequences. We therefore sought to develop mucosally transmissible SHIV challenge stocks containing HIV-1 CRF01_AE env derived from acutely HIV-1 infected individuals from Thailand. SHIV-AE6, SHIV-AE6RM, and SHIV-AE16 contained env sequences that were >99% identical to the original HIV-1 isolate and did not require in vivo passaging. These viruses exhibited CCR5 tropism and displayed a tier 2 neutralization phenotype. These challenge stocks efficiently infected rhesus monkeys by the intrarectal route, replicated to high levels during acute infection, and established chronic viremia in a subset of animals. SHIV-AE16 was titrated for use in single, high dose as well as repetitive, low dose intrarectal challenge studies. These SHIV challenge stocks should facilitate the preclinical evaluation of vaccines, monoclonal antibodies, and other interventions targeted at preventing HIV-1 CRF01_AE infection.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/immunology , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Animals , Antibodies, Neutralizing/immunology , Cells, Cultured , Female , Humans , Macaca mulatta , Male , Viremia/immunology
9.
PLoS Pathog ; 12(4): e1005545, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27082643

ABSTRACT

Resting CD4+ T-cells harboring inducible HIV proviruses are a critical reservoir in antiretroviral therapy (ART)-treated subjects. These cells express little to no viral protein, and thus neither die by viral cytopathic effects, nor are efficiently cleared by immune effectors. Elimination of this reservoir is theoretically possible by combining latency-reversing agents (LRAs) with immune effectors, such as CD8+ T-cells. However, the relative efficacy of different LRAs in sensitizing latently-infected cells for recognition by HIV-specific CD8+ T-cells has not been determined. To address this, we developed an assay that utilizes HIV-specific CD8+ T-cell clones as biosensors for HIV antigen expression. By testing multiple CD8+ T-cell clones against a primary cell model of HIV latency, we identified several single agents that primed latently-infected cells for CD8+ T-cell recognition, including IL-2, IL-15, two IL-15 superagonists (IL-15SA and ALT-803), prostratin, and the TLR-2 ligand Pam3CSK4. In contrast, we did not observe CD8+ T-cell recognition of target cells following treatment with histone deacetylase inhibitors or with hexamethylene bisacetamide (HMBA). In further experiments we demonstrate that a clinically achievable concentration of the IL-15 superagonist 'ALT-803', an agent presently in clinical trials for solid and hematological tumors, primes the natural ex vivo reservoir for CD8+ T-cell recognition. Thus, our results establish a novel experimental approach for comparative evaluation of LRAs, and highlight ALT-803 as an LRA with the potential to synergize with CD8+ T-cells in HIV eradication strategies.


Subject(s)
Antiviral Agents/pharmacology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , Proteins/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Virus Latency/drug effects , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Humans , Polymerase Chain Reaction , Recombinant Fusion Proteins , Virus Activation/drug effects
10.
Adv Exp Med Biol ; 1075: 241-263, 2018.
Article in English | MEDLINE | ID: mdl-30030796

ABSTRACT

Latently infected cells are very infrequent in CD4+ T cells from antiretroviral (ARV) treated individuals, with only approximately one in a million infected CD4+ T cells in blood. Given the low frequency of infected cells in vivo, multiple in vitro latency models have been developed to facilitate investigations into mechanisms of HIV latency, as well as to enable the evaluation of pharmacological and immunological interventions aimed at depleting latently infected cells. These in vitro models include clones of transformed cell lines with integrated HIV proviruses or primary CD4+ T cells from uninfected donors that have been infected with HIV in particular conditions. This chapter presents a description of these various in vitro models, along with an overview of their advantages and limitations.Preclinical animal models represent a critical bridge between in vitro studies and human clinical trials. Simian immunodeficiency virus (SIV) infection of Indian origin rhesus macaques has been well established as an informative model of HIV infection. Recent years have seen breakthroughs in ARVs that permit the potent suppression of SIV replication, enabling studies of latency and putative curative interventions in this model. Small animal models of HIV infection can be generated by engrafting immunodeficient mice with human immune cells. These "humanized mice" have provided valuable insights into HIV pathogenesis and are under development as models for studying HIV latency. We summarize both the promise of these models and outstanding challenges that remain to be overcome to realize their potential to inform efforts to cure HIV infection.


Subject(s)
HIV-1/physiology , Virus Latency , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , Cell Line, Transformed , Cells, Cultured , Clone Cells/virology , Coculture Techniques , Graft vs Host Disease , HIV-1/drug effects , Heterografts , Humans , In Vitro Techniques , Macaca mulatta , Mice , Mice, Nude , Radiation Chimera , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Virus Latency/drug effects , Virus Replication/drug effects
11.
J Infect Dis ; 216(suppl_10): S928-S934, 2017 12 16.
Article in English | MEDLINE | ID: mdl-29267926

ABSTRACT

Zika virus is a re-emerging flavivirus transmitted primarily by arthropod vectors. The recent devastating outbreak of Zika virus in Brazil was preceded by the slow global encroachment of this virus over many decades. To date, significant research efforts are underway to understand the spread and the unique pathogenesis of this virus; with the intent to rapidly develop vaccines and therapeutics. Several model systems have emerged to study Zika. This review will focus on the use of nonhuman primates to model Zika infection.


Subject(s)
Viral Vaccines , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Disease Models, Animal , Humans , Primates , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/therapy , Zika Virus Infection/virology
12.
J Med Primatol ; 46(4): 137-143, 2017 08.
Article in English | MEDLINE | ID: mdl-28748659

ABSTRACT

BACKGROUND: Understanding natural HIV control may lead to new preventative or therapeutic strategies. Several protective major histocompatibility complex (MHC) genotypes were found in humans and rhesus macaques. Here, we report a simian immunodeficiency virus (SIV) controller MHC genotype in Mauritian cynomolgus macaques (MCMs). METHODS: Twelve MHC-genotyped MCMs were infected with SIVmac251 and monitored for viral loads and CD4+ T-cell counts. RESULTS: Two macaques with M3M4 genotype exhibited the lowest peak viral loads (log plasma SIV RNA copies/mL), nearly 3 logs lower than those in most macaques with other MHC haplotype combinations, and set point viral loads below the level of detection limit by RT-qPCR (<2 log RNA copies/mL). They maintained healthy CD4+ T-cell counts of >500 cells/µL blood, while CD4 counts in the vast majority of other macaques were below this level. CONCLUSIONS: The M3M4 MHC genotype may confer enhanced control of SIV replication in MCMs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Haplotypes , Macaca fascicularis/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Viral Load , Animals , Female , Macaca fascicularis/immunology , Mauritius , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/physiology
13.
J Virol ; 89(4): 1965-74, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25473043

ABSTRACT

UNLABELLED: The development of a panel of mucosally transmissible simian-human immunodeficiency virus (SHIV) challenge stocks from multiple virus clades would facilitate preclinical evaluation of candidate HIV-1 vaccines and therapeutics. The majority of SHIV stocks that have been generated to date have been derived from clade B HIV-1 env sequences from viruses isolated during chronic infection and typically required serial animal-to-animal adaptation for establishing mucosal transmissibility and pathogenicity. To capture essential features of mucosal transmission of clade C viruses, we produced a series of SHIVs with early clade C HIV-1 env sequences from acutely HIV-1-infected individuals from South Africa. SHIV-327c and SHIV-327cRM expressed env sequences that were 99.7 to 100% identical to the original HIV-1 isolate and did not require in vivo passaging for mucosal infectivity. These challenge stocks infected rhesus monkeys efficiently by both intrarectal and intravaginal routes, replicated to high levels during acute infection, and established chronic setpoint viremia in 13 of 17 (76%) infected animals. The SHIV-327cRM challenge stock was also titrated for both single, high-dose intrarectal challenges and repetitive, low-dose intrarectal challenges in rhesus monkeys. These SHIV challenge stocks should facilitate the preclinical evaluation of vaccines and other interventions aimed at preventing clade C HIV-1 infection. IMPORTANCE: We describe the development of two related clade C SHIV challenge stocks. These challenge stocks should prove useful for preclinical testing of vaccines and other interventions aimed at preventing clade C HIV-1 infection.


Subject(s)
HIV-1/growth & development , HIV-1/isolation & purification , Mucous Membrane/virology , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/isolation & purification , Animals , Disease Models, Animal , Female , HIV-1/genetics , HIV-1/pathogenicity , Humans , Macaca mulatta , Male , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Virulence
14.
Curr Opin HIV AIDS ; 19(4): 179-186, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38747727

ABSTRACT

PURPOSE OF REVIEW: HIV-1 infection contributes substantially to global morbidity and mortality, with no immediate promise of an effective prophylactic vaccine. Combination antiretroviral therapy (ART) suppresses HIV replication, but latent viral reservoirs allow the virus to persist and reignite active replication if ART is discontinued. Moreover, inflammation and immune disfunction persist despite ART-mediated suppression of HIV. Immune checkpoint molecules facilitate immune dysregulation and viral persistence. However, their therapeutic modulation may offer an avenue to enhance viral immune control for patients living with HIV-1 (PLWH). RECENT FINDINGS: The success of immune checkpoint inhibitor (ICI) therapy in oncology suggests that targeting these same immune pathways might be an effective therapeutic approach for treating PLWH. Several ICIs have been evaluated for their ability to reinvigorate exhausted T cells, and possibly reverse HIV latency, in both preclinical and clinical HIV-1 studies. SUMMARY: Although there are very encouraging findings showing enhanced CD8 + T-cell function with ICI therapy in HIV infection, it remains uncertain whether ICIs alone could demonstrably impact the HIV reservoir. Moreover, safety concerns and significant clinical adverse events present a hurdle to the development of ICI approaches. This review provides an update on the current knowledge regarding the development of ICIs for the remission of HIV-1 in PWH. We detail recent findings from simian immunodeficiency virus (SIV)-infected rhesus macaque models, clinical trials in PLWH, and the role of soluble immune checkpoint molecules in HIV pathogenesis.


Subject(s)
HIV Infections , Immune Checkpoint Inhibitors , Humans , Immune Checkpoint Inhibitors/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , Animals , HIV-1/drug effects , HIV-1/physiology , HIV-1/immunology
15.
Science ; 383(6687): 1104-1111, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38422185

ABSTRACT

The eradication of the viral reservoir represents the major obstacle to the development of a clinical cure for established HIV-1 infection. Here, we demonstrate that the administration of N-803 (brand name Anktiva) and broadly neutralizing antibodies (bNAbs) results in sustained viral control after discontinuation of antiretroviral therapy (ART) in simian-human AD8 (SHIV-AD8)-infected, ART-suppressed rhesus macaques. N-803+bNAbs treatment induced immune activation and transient viremia but only limited reductions in the SHIV reservoir. Upon ART discontinuation, viral rebound occurred in all animals, which was followed by durable control in approximately 70% of all N-803+bNAb-treated macaques. Viral control was correlated with the reprogramming of CD8+ T cells by N-803+bNAb synergy. Thus, complete eradication of the replication-competent viral reservoir is likely not a prerequisite for the induction of sustained remission after discontinuation of ART.


Subject(s)
Anti-Retroviral Agents , Recombinant Fusion Proteins , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Humans , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/pharmacology , Broadly Neutralizing Antibodies/administration & dosage , CD8-Positive T-Lymphocytes/virology , Immunotherapy , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/therapy , Viral Load , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Remission Induction , Drug Therapy, Combination
16.
PLoS Pathog ; 7(2): e1001293, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21379569

ABSTRACT

We characterized the evolution of simian immunodeficiency virus (SIV) in the male genital tract by examining blood- and semen-associated virus from experimentally and sham vaccinated rhesus monkeys during primary infection. At the time of peak virus replication, SIV sequences were intermixed between the blood and semen supporting a scenario of high-level virus "spillover" into the male genital tract. However, at the time of virus set point, compartmentalization was apparent in 4 of 7 evaluated monkeys, likely as a consequence of restricted virus gene flow between anatomic compartments after the resolution of primary viremia. These findings suggest that SIV replication in the male genital tract evolves to compartmentalization after peak viremia resolves.


Subject(s)
Gene Products, env/genetics , Genitalia, Male/virology , Semen/virology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Animals , DNA, Viral/genetics , Gene Flow , Macaca mulatta , Male , Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/blood , Simian Immunodeficiency Virus/genetics , Vaccination , Viral Load , Viremia/prevention & control , Virus Replication
17.
PLoS Genet ; 6(6): e1000997, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585621

ABSTRACT

CCL3 is a ligand for the HIV-1 co-receptor CCR5. There have recently been conflicting reports in the literature concerning whether CCL3-like gene (CCL3L) copy number variation (CNV) is associated with resistance to HIV-1 acquisition and with both viral load and disease progression following infection with HIV-1. An association has also been reported between CCL3L CNV and clinical sequelae of the simian immunodeficiency virus (SIV) infection in vivo in rhesus monkeys. The present study was initiated to explore the possibility of an association of CCL3L CNV with the control of virus replication and AIDS progression in a carefully defined cohort of SIVmac251-infected, Indian-origin rhesus monkeys. Although we demonstrated extensive variation in copy number of CCL3L in this cohort of monkeys, CCL3L CNV was not significantly associated with either peak or set-point plasma SIV RNA levels in these monkeys when MHC class I allele Mamu-A*01 was included in the models or progression to AIDS in these monkeys. With 66 monkeys in the study, there was adequate power for these tests if the correlation of CCL3L and either peak or set-point plasma SIV RNA levels was 0.34 or 0.36, respectively. These findings call into question the premise that CCL3L CNV is important in HIV/SIV pathogenesis.


Subject(s)
Chemokine CCL3/genetics , DNA Replication , Histocompatibility Antigens Class I/genetics , Macaca mulatta/genetics , Proteins/genetics , Simian Immunodeficiency Virus/physiology , Alleles , Animals , DNA Copy Number Variations , Gene Expression Regulation , India , Macaca mulatta/virology , Ubiquitin-Protein Ligases
18.
J Virol ; 85(19): 10389-98, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775457

ABSTRACT

There is considerable variability in host susceptibility to human immunodeficiency virus type 1 (HIV-1) infection, but the host genetic determinants of that variability are not well understood. In addition to serving as a block for cross-species retroviral infection, TRIM5 was recently shown to play a central role in limiting primate immunodeficiency virus replication. We hypothesized that TRIM5 may also contribute to susceptibility to mucosal acquisition of simian immunodeficiency virus (SIV) in rhesus monkeys. We explored this hypothesis by establishing 3 cohorts of Indian-origin rhesus monkeys with different TRIM5 genotypes: homozygous restrictive, heterozygous permissive, and homozygous permissive. We then evaluated the effect of TRIM5 genotype on the penile transmission of SIVsmE660. We observed a significant effect of TRIM5 genotype on mucosal SIVsmE660 acquisition in that no SIV transmission occurred in monkeys with only restrictive TRIM5 alleles. In contrast, systemic SIV infections were initiated after preputial pocket exposures in monkeys that had at least one permissive TRIM5 allele. These data demonstrate that host genetic factors can play a critical role in restricting mucosal transmission of a primate immunodeficiency virus. In addition, we used our understanding of TRIM5 to establish a novel nonhuman primate penile transmission model for AIDS mucosal pathogenesis and vaccine research.


Subject(s)
Carrier Proteins/immunology , Genetic Predisposition to Disease , Mucous Membrane/immunology , Mucous Membrane/virology , Penis/immunology , Penis/virology , Simian Immunodeficiency Virus/immunology , Animals , Carrier Proteins/genetics , Genotype , Macaca mulatta , Male
19.
PLoS Pathog ; 6(1): e1000738, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20107597

ABSTRACT

The cytoplasmic TRIM5alpha proteins of certain mammalian lineages efficiently recognize the incoming capsids of particular retroviruses and potently restrict infection in a species-specific manner. Successful retroviruses have evolved capsids that are less efficiently recognized by the TRIM5alpha proteins of the natural hosts. To address whether TRIM5alpha contributes to the outcome of retroviral infection in a susceptible host species, we investigated the impact of TRIM5 polymorphisms in rhesus monkeys on the course of a simian immunodeficiency virus (SIV) infection. Full-length TRIM5alpha cDNAs were derived from each of 79 outbred monkeys and sequenced. Associations were explored between the expression of particular TRIM5 alleles and both the permissiveness of cells to SIV infection in vitro and clinical sequelae of SIV infection in vivo. Natural variation in the TRIM5alpha B30.2(SPRY) domain influenced the efficiency of SIVmac capsid binding and the in vitro susceptibility of cells from the monkeys to SIVmac infection. We also show the importance in vivo of the interaction of SIVmac with different allelic forms of TRIM5, demonstrating that particular alleles are associated with as much as 1.3 median log difference in set-point viral loads in SIVmac-infected rhesus monkeys. Moreover, these allelic forms of TRIM5 were associated with the extent of loss of central memory (CM) CD4+ T cells and the rate of progression to AIDS in the infected monkeys. These findings demonstrate a central role for TRIM5alpha in limiting the replication of an immunodeficiency virus infection in a primate host.


Subject(s)
Genetic Predisposition to Disease , Proteins/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Humans , Immunoblotting , Macaca mulatta , Molecular Sequence Data , Polymorphism, Single Nucleotide , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/metabolism , Transduction, Genetic , Ubiquitin-Protein Ligases
20.
J Virol ; 84(19): 9810-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686023

ABSTRACT

Post hoc analysis of the phase 2b Step study evaluating a recombinant adenovirus serotype 5 (rAd5)-based HIV-1 vaccine candidate suggested a potential increased risk of HIV-1 acquisition in subjects who were baseline Ad5 seropositive and uncircumcised. These concerns had a profound impact on the HIV-1 vaccine development field, although the mechanism underlying this observation remains unknown. It has been hypothesized that rAd5 vaccination of baseline Ad5-seropositive individuals may have resulted in anamnestic, vector-specific CD4(+) T lymphocytes that could have trafficked to mucosal sites and served as increased targets for HIV-1 infection. Here we show that Ad5-specific CD4(+) T lymphocyte responses at mucosal sites following rAd5-Gag/Pol/Nef vaccination were comparable in rhesus monkeys with and without baseline Ad5 immunity. Moreover, the total cellular inflammatory infiltrates and the CD3(+), CD4(+), HLA-DR(+), Ki67(+), and langerin(+) cellular subpopulations in colorectal and foreskin mucosa were similar in both groups. Thus, no greater trafficking of Ad5-specific CD4(+) T lymphocytes to mucosal target sites was observed following rAd5 vaccination of rhesus monkeys with baseline Ad5 immunity. These findings from this nonhuman primate model provide evidence against the hypothesis that recruitment of vector-specific target cells to mucosal sites led to increased HIV-1 acquisition in Ad5-seropositive, uncircumcised vaccinees in the Step study.


Subject(s)
Adenoviruses, Human/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Mucous Membrane/immunology , Mucous Membrane/virology , AIDS Vaccines/adverse effects , AIDS Vaccines/genetics , Adenoviruses, Human/genetics , Animals , Foreskin/cytology , Foreskin/immunology , Foreskin/virology , Genetic Vectors , HIV Infections/etiology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1 , Humans , Immunity, Mucosal , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Macaca mulatta , Male , Models, Animal , Models, Immunological , Mucous Membrane/cytology , Vaccination/adverse effects , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL