Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(1): 189-206, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32220311

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection persists despite years of antiretroviral therapy (ART). To remove the stigma and burden of chronic infection, approaches to eradicate or cure HIV infection are desired. Attempts to augment ART with therapies that reverse viral latency, paired with immunotherapies to clear infection, have advanced into the clinic, but the field is still in its infancy. We review foundational studies and highlight new insights in HIV cure research. Together with advances in ART delivery and HIV prevention strategies, future therapies that clear HIV infection may relieve society of the affliction of the HIV pandemic.


Subject(s)
Anti-HIV Agents/therapeutic use , Chronic Disease/therapy , HIV Infections/therapy , HIV-1/drug effects , Immunotherapy/methods , Virus Latency/drug effects , Animals , Haplorhini , Humans
2.
Cell ; 163(3): 746-58, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496612

ABSTRACT

A key effector route of the Sugar Code involves lectins that exert crucial regulatory controls by targeting distinct cellular glycans. We demonstrate that a single amino-acid substitution in a banana lectin, replacing histidine 84 with a threonine, significantly reduces its mitogenicity, while preserving its broad-spectrum antiviral potency. X-ray crystallography, NMR spectroscopy, and glycocluster assays reveal that loss of mitogenicity is strongly correlated with loss of pi-pi stacking between aromatic amino acids H84 and Y83, which removes a wall separating two carbohydrate binding sites, thus diminishing multivalent interactions. On the other hand, monovalent interactions and antiviral activity are preserved by retaining other wild-type conformational features and possibly through unique contacts involving the T84 side chain. Through such fine-tuning, target selection and downstream effects of a lectin can be modulated so as to knock down one activity, while preserving another, thus providing tools for therapeutics and for understanding the Sugar Code.


Subject(s)
Plant Lectins/chemistry , Plant Lectins/genetics , Anti-HIV Agents/chemistry , Carbohydrate Sequence , Genetic Engineering , Mitogens/chemistry , Models, Molecular , Molecular Dynamics Simulation , Musa/chemistry
3.
Nature ; 591(7850): 451-457, 2021 03.
Article in English | MEDLINE | ID: mdl-33561864

ABSTRACT

All coronaviruses known to have recently emerged as human pathogens probably originated in bats1. Here we use a single experimental platform based on immunodeficient mice implanted with human lung tissue (hereafter, human lung-only mice (LoM)) to demonstrate the efficient in vivo replication of severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as well as two endogenous SARS-like bat coronaviruses that show potential for emergence as human pathogens. Virus replication in this model occurs in bona fide human lung tissue and does not require any type of adaptation of the virus or the host. Our results indicate that bats contain endogenous coronaviruses that are capable of direct transmission to humans. Our detailed analysis of in vivo infection with SARS-CoV-2 in human lung tissue from LoM showed a predominant infection of human lung epithelial cells, including type-2 pneumocytes that are present in alveoli and ciliated airway cells. Acute infection with SARS-CoV-2 was highly cytopathic and induced a robust and sustained type-I interferon and inflammatory cytokine and chemokine response. Finally, we evaluated a therapeutic and pre-exposure prophylaxis strategy for SARS-CoV-2 infection. Our results show that therapeutic and prophylactic administration of EIDD-2801-an oral broad-spectrum antiviral agent that is currently in phase II/III clinical trials-markedly inhibited SARS-CoV-2 replication in vivo, and thus has considerable potential for the prevention and treatment of COVID-19.


Subject(s)
COVID-19 Drug Treatment , COVID-19/prevention & control , Cytidine/analogs & derivatives , Hydroxylamines/administration & dosage , Hydroxylamines/therapeutic use , Administration, Oral , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/virology , Animals , COVID-19/immunology , Chemoprevention , Chiroptera/virology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Cytidine/administration & dosage , Cytidine/therapeutic use , Cytokines/immunology , Epithelial Cells/virology , Female , Heterografts , Humans , Immunity, Innate , Interferon Type I/immunology , Lung/immunology , Lung/pathology , Lung/virology , Lung Transplantation , Male , Mice , Post-Exposure Prophylaxis , Pre-Exposure Prophylaxis , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Virus Replication
4.
Nature ; 578(7793): 154-159, 2020 02.
Article in English | MEDLINE | ID: mdl-31969705

ABSTRACT

Human immunodeficiency virus (HIV) persists indefinitely in individuals with HIV who receive antiretroviral therapy (ART) owing to a reservoir of latently infected cells that contain replication-competent virus1-4. Here, to better understand the mechanisms responsible for latency persistence and reversal, we used the interleukin-15 superagonist N-803 in conjunction with the depletion of CD8+ lymphocytes in ART-treated macaques infected with simian immunodeficiency virus (SIV). Although N-803 alone did not reactivate virus production, its administration after the depletion of CD8+ lymphocytes in conjunction with ART treatment induced robust and persistent reactivation of the virus in vivo. We found viraemia of more than 60 copies per ml in all macaques (n = 14; 100%) and in 41 out of a total of 56 samples (73.2%) that were collected each week after N-803 administration. Notably, concordant results were obtained in ART-treated HIV-infected humanized mice. In addition, we observed that co-culture with CD8+ T cells blocked the in vitro latency-reversing effect of N-803 on primary human CD4+ T cells that were latently infected with HIV. These results advance our understanding of the mechanisms responsible for latency reversal and lentivirus reactivation during ART-suppressed infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-15/agonists , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , Humans , Interleukin-15/immunology , Lymphocyte Depletion , Macaca mulatta , Mice , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Virus Latency , Virus Replication/immunology
6.
Nature ; 578(7793): 160-165, 2020 02.
Article in English | MEDLINE | ID: mdl-31969707

ABSTRACT

Long-lasting, latently infected resting CD4+ T cells are the greatest obstacle to obtaining a cure for HIV infection, as these cells can persist despite decades of treatment with antiretroviral therapy (ART). Estimates indicate that more than 70 years of continuous, fully suppressive ART are needed to eliminate the HIV reservoir1. Alternatively, induction of HIV from its latent state could accelerate the decrease in the reservoir, thus reducing the time to eradication. Previous attempts to reactivate latent HIV in preclinical animal models and in clinical trials have measured HIV induction in the peripheral blood with minimal focus on tissue reservoirs and have had limited effect2-9. Here we show that activation of the non-canonical NF-κB signalling pathway by AZD5582 results in the induction of HIV and SIV RNA expression in the blood and tissues of ART-suppressed bone-marrow-liver-thymus (BLT) humanized mice and rhesus macaques infected with HIV and SIV, respectively. Analysis of resting CD4+ T cells from tissues after AZD5582 treatment revealed increased SIV RNA expression in the lymph nodes of macaques and robust induction of HIV in almost all tissues analysed in humanized mice, including the lymph nodes, thymus, bone marrow, liver and lung. This promising approach to latency reversal-in combination with appropriate tools for systemic clearance of persistent HIV infection-greatly increases opportunities for HIV eradication.


Subject(s)
HIV Infections/virology , HIV-1/physiology , NF-kappa B/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Virus Latency , Alkynes/pharmacology , Animals , Anti-Retroviral Agents/pharmacology , HIV Infections/metabolism , HIV-1/drug effects , Macaca mulatta , Mice , Oligopeptides/pharmacology , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/drug effects , Virus Latency/drug effects
7.
PLoS Pathog ; 19(12): e1011824, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38055722

ABSTRACT

Lifelong treatment is required for people living with HIV as current antiretroviral therapy (ART) does not eradicate HIV infection. Latently infected cells are essentially indistinguishable from uninfected cells and cannot be depleted by currently available approaches. This study evaluated antibody mediated transient CD4+ T cell depletion as a strategy to reduce the latent HIV reservoir. Anti-CD4 antibodies effectively depleted CD4+ T cells in the peripheral blood and tissues of humanized mice. We then demonstrate that antibody-mediated CD4+ T cell depletion of HIV infected ART-suppressed animals results in substantial reductions in cell-associated viral RNA and DNA levels in peripheral blood cells over the course of anti-CD4 antibody treatment. Recovery of CD4+ T cells was observed in all tissues analyzed except for the lung 26 days after cessation of antibody treatment. After CD4+ T cell recovery, significantly lower levels of cell-associated viral RNA and DNA were detected in the tissues of anti-CD4 antibody-treated animals. Further, an 8.5-fold reduction in the levels of intact HIV proviral DNA and a 3.1-fold reduction in the number of latently infected cells were observed in anti-CD4-antibody-treated animals compared with controls. However, there was no delay in viral rebound when ART was discontinued in anti-CD4 antibody-treated animals following CD4+ T cell recovery compared with controls. Our results suggest that transient CD4+ T cell depletion, a long-standing clinical intervention that might have an acceptable safety profile, during suppressive ART can reduce the size of the HIV reservoir in humanized mice.


Subject(s)
HIV Infections , HIV-1 , Humans , Mice , Animals , CD4-Positive T-Lymphocytes , Virus Latency , Virus Replication , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , RNA, Viral , DNA , Viral Load
8.
Retrovirology ; 21(1): 11, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38945996

ABSTRACT

BACKGROUND: Since the introduction of combination antiretroviral therapy (cART) the brain has become an important human immunodeficiency virus (HIV) reservoir due to the relatively low penetration of many drugs utilized in cART into the central nervous system (CNS). Given the inherent limitations of directly assessing acute HIV infection in the brains of people living with HIV (PLWH), animal models, such as humanized mouse models, offer the most effective means of studying the effects of different viral strains and their impact on HIV infection in the CNS. To evaluate CNS pathology during HIV-1 infection in the humanized bone marrow/liver/thymus (BLT) mouse model, a histological analysis was conducted on five CNS regions, including the frontal cortex, hippocampus, striatum, cerebellum, and spinal cord, to delineate the neuronal (MAP2ab, NeuN) and neuroinflammatory (GFAP, Iba-1) changes induced by two viral strains after 2 weeks and 8 weeks post-infection. RESULTS: Findings reveal HIV-infected human cells in the brain of HIV-infected BLT mice, demonstrating HIV neuroinvasion. Further, both viral strains, HIV-1JR-CSF and HIV-1CH040, induced neuronal injury and astrogliosis across all CNS regions following HIV infection at both time points, as demonstrated by decreases in MAP2ab and increases in GFAP fluorescence signal, respectively. Importantly, infection with HIV-1JR-CSF had more prominent effects on neuronal health in specific CNS regions compared to HIV-1CH040 infection, with decreasing number of NeuN+ neurons, specifically in the frontal cortex. On the other hand, infection with HIV-1CH040 demonstrated more prominent effects on neuroinflammation, assessed by an increase in GFAP signal and/or an increase in number of Iba-1+ microglia, across CNS regions. CONCLUSION: These findings demonstrate that CNS pathology is widespread during acute HIV infection. However, neuronal loss and the magnitude of neuroinflammation in the CNS is strain dependent indicating that strains of HIV cause differential CNS pathologies.


Subject(s)
Disease Models, Animal , HIV Infections , HIV-1 , Neuroinflammatory Diseases , Neurons , Animals , Mice , HIV Infections/virology , HIV Infections/pathology , HIV Infections/complications , Humans , Neurons/virology , Neurons/pathology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , Brain/pathology , Brain/virology , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Microfilament Proteins/metabolism
9.
Antimicrob Agents Chemother ; 64(10)2020 09 21.
Article in English | MEDLINE | ID: mdl-32661005

ABSTRACT

Adequate antiretroviral (ARV) concentrations in lymphoid tissues are critical for optimal antiretroviral therapy (ART). While the spleen contains 25% of the body's lymphocytes, there are minimal data on ARV penetration in this organ. This study quantified total and protein-unbound splenic ARV concentrations and determined whether drug transporters, sex, or infection status were modifiers of these concentrations in animal models and humans. Two humanized mice models (hu-HSC-Rag [n = 36; 18 HIV-positive (HIV+) and 18 HIV-negative (HIV-)] and bone marrow-liver-thymus [n = 13; 7 HIV+ and 6 HIV-]) and one nonhuman primate (NHP) model (rhesus macaque [n = 18; 10 SHIV+ and 8 SHIV-]) were dosed to steady state with ARV combinations. HIV+ human spleens (n = 14) from the National NeuroAIDS Tissue Consortium were analyzed postmortem (up to 24 h postdose). ARV concentrations were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS), drug transporter concentrations were measured with LC-MS proteomics, and protein binding in NHP spleens was determined by rapid equilibrium dialysis. Mice generally had the lowest splenic concentrations of the three species. Protein binding in splenic tissue was 6 to 96%, compared to 76 to 99% in blood plasma. NHPs had quantifiable Mrp4, Bcrp, and Ent1 concentrations, and humans had quantifiable ENT1 concentrations. None significantly correlated with tissue ARV concentrations. There was also no observable influence of infection status or sex. With these dosing strategies, NHP splenic penetration most closely resembled that of humans. These data can inform tissue pharmacokinetic scaling to humans to target HIV reservoirs by identifying important species-related differences.


Subject(s)
Anti-HIV Agents , HIV Infections , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Anti-HIV Agents/therapeutic use , Chromatography, Liquid , HIV Infections/drug therapy , Humans , Macaca mulatta , Mice , Models, Animal , Neoplasm Proteins , Spleen , Tandem Mass Spectrometry
10.
Article in English | MEDLINE | ID: mdl-31611355

ABSTRACT

For HIV cure strategies like "kick and kill" to succeed, antiretroviral (ARV) drugs must reach effective concentrations in putative viral reservoirs. We characterize penetration of six ARVs in three preclinical animal models and humans. We found that standard dosing strategies in preclinical species closely mimicked tissue concentrations in humans for some, but not all, ARVs. These results have implications for interpreting HIV treatment, prevention, or cure interventions between preclinical and clinical models.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , Animals , Anti-HIV Agents/therapeutic use , Atazanavir Sulfate/therapeutic use , Emtricitabine/therapeutic use , Female , Humans , In Vitro Techniques , Maraviroc/therapeutic use , Mice , Raltegravir Potassium/therapeutic use , Tenofovir/therapeutic use
11.
Eur J Immunol ; 48(7): 1255-1257, 2018 07.
Article in English | MEDLINE | ID: mdl-29572809

ABSTRACT

We show the presence of lymphoid tissue-resident PLZF+ CD45RA+ RO+ CD4 T cells in humans. They express HLA-DR, granzyme B, and perforin and are low on CCR7 like terminally differentiated effector memory (Temra) cells and are likely generated from effector T cells (Te) or from central (Tcm) or effector (Tem) memory T (Tcm) cells during immune responses. Tn, Naïve T cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Genotype , Lymphoid Tissue/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , T-Lymphocyte Subsets/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cells, Cultured , Granzymes/metabolism , HLA-DR Antigens/metabolism , Humans , Immunity, Cellular , Immunologic Memory , Perforin/metabolism
12.
J Pharmacol Exp Ther ; 370(3): 360-368, 2019 09.
Article in English | MEDLINE | ID: mdl-31235531

ABSTRACT

In a "kick and kill" strategy for human immunodeficiency virus (HIV) eradication, protective concentrations of antiretrovirals (ARVs) in the lymph node are important to prevent vulnerable cells from further HIV infection. However, the factors responsible for drug distribution and concentration into these tissues are largely unknown. Although humanized mice and nonhuman primates (NHPs) are crucial to HIV research, ARV tissue pharmacology has not been well characterized across species. This study investigated the influence of drug transporter expression, viral infection, and sex on ARV penetration within lymph nodes of animal models and humans. Six ARVs were dosed for 10 days in humanized mice and NHPs. Plasma and lymph nodes were collected at necropsy, 24 hours after the last dose. Human lymph node tissue and plasma from deceased patients were collected from tissue banks. ARV, active metabolite, and endogenous nucleotide concentrations were measured by liquid chromatography-tandem mass spectrometry, and drug transporter expression was measured using quantitative polymerase chain reaction and quantitative targeted absolute proteomics. In NHPs and humans, lymph node ARV concentrations were greater than or equal to plasma, and tenofovir diphosphate/deoxyadenosine triphosphate concentration ratios achieved efficacy targets in lymph nodes from all three species. There was no effect of infection or sex on ARV concentrations. Low drug transporter expression existed in lymph nodes from all species, and no predictive relationships were found between transporter gene/protein expression and ARV penetration. Overall, common preclinical models of HIV infection were well suited to predict human ARV exposure in lymph nodes, and low transporter expression suggests primarily passive drug distribution in these tissues. SIGNIFICANCE STATEMENT: During human immunodeficiency virus (HIV) eradication strategies, protective concentrations of antiretrovirals (ARVs) in the lymph node prevent vulnerable cells from further HIV infection. However, ARV tissue pharmacology has not been well characterized across preclinical species used for HIV eradication research, and the influence of drug transporters, HIV infection, and sex on ARV distribution and concentration into the lymph node is largely unknown. Here we show that two animal models of HIV infection (humanized mice and nonhuman primates) were well suited to predict human ARV exposure in lymph nodes. Additionally, we found that drug transporter expression was minimal and-along with viral infection and sex-did not affect ARV penetration into lymph nodes from any species.


Subject(s)
Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Gene Expression Regulation/drug effects , HIV/physiology , Lymph Nodes/metabolism , Membrane Transport Proteins/metabolism , Sex Characteristics , Animals , Anti-HIV Agents/blood , Female , HIV/drug effects , Humans , Lymph Nodes/drug effects , Macaca mulatta , Male , Mice , Species Specificity
13.
Xenobiotica ; 49(10): 1192-1201, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30346892

ABSTRACT

1. Antiretroviral concentrations in cerebrospinal fluid (CSF) are used as surrogate for brain tissue, although sparse data support this. We quantified antiretrovirals in brain tissue across preclinical models, compared them to CSF, and calculated 90% inhibitory quotients (IQ90) for nonhuman primate (NHP) brain tissue. Spatial distribution of efavirenz was performed by mass-spectrometry imaging (MSI). 2. HIV or RT-SHIV-infected and uninfected animals from two humanized mouse models (hemopoietic-stem cell/RAG2-, n = 36; bone marrow-liver-thymus/BLT, n =13) and an NHP model (rhesus macaque, n =18) were dosed with six antiretrovirals. Brain tissue, CSF (NHPs), and plasma were collected at necropsy. Drug concentrations were measured by LC-MS/MS. Rapid equilibrium dialysis determined protein binding in NHP brain. 3. Brain tissue penetration of most antiretrovirals were >10-fold lower (p < 0.02) in humanized mice than NHPs. NHP CSF concentrations were >13-fold lower (p <0.02) than brain tissue with poor agreement except for efavirenz (r = 0.91, p = 0.001). Despite 97% brain tissue protein binding, efavirenz achieved IQ90>1 in all animals and 2-fold greater white versus gray matter concentration. 4. Brain tissue penetration varied across animal models for all antiretrovirals except raltegravir, and extrapolating brain tissue concentrations between models should be avoided. With the exception of efavirenz, CSF is not a surrogate for brain tissue concentrations.


Subject(s)
Anti-HIV Agents , Benzoxazines , Brain , HIV Infections , HIV-1 , Alkynes , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Benzoxazines/pharmacokinetics , Benzoxazines/pharmacology , Brain/metabolism , Brain/pathology , Brain/virology , Cyclopropanes , Drug Evaluation, Preclinical , Female , HIV Infections/cerebrospinal fluid , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Macaca mulatta , Male , Mice
14.
J Neurovirol ; 24(2): 185-191, 2018 04.
Article in English | MEDLINE | ID: mdl-28831774

ABSTRACT

While the human immunodeficiency virus (HIV) epidemic was initially characterized by a high prevalence of severe and widespread neurological pathologies, the development of better treatments to suppress viremia over years and even decades has mitigated many of the severe neurological pathologies previously observed. Despite effective treatment, mild neurocognitive impairment and premature cognitive aging are observed in HIV-infected individuals, suggesting a changing but ongoing role of HIV infection in the central nervous system (CNS). Although current therapies are effective in suppressing viremia, they are not curative and patients must remain on life-long treatment or risk recrudescence of virus. Important for the development and evaluation of a cure for HIV will be animal models that recapitulate critical aspects of infection in vivo. In the following, we seek to summarize some of the recent developments in humanized mouse models and their usefulness in modeling HIV infection of the CNS and HIV cure strategies.


Subject(s)
AIDS Dementia Complex/drug therapy , Anti-HIV Agents/pharmacology , Cognitive Dysfunction/drug therapy , Disease Models, Animal , HIV-1/drug effects , AIDS Dementia Complex/physiopathology , AIDS Dementia Complex/virology , Animals , Antiretroviral Therapy, Highly Active , Brain/drug effects , Brain/physiopathology , Brain/virology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/virology , HIV-1/pathogenicity , HIV-1/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Liver/cytology , Liver/immunology , Mice , Mice, SCID , Myeloid Cells/transplantation , Myeloid Cells/virology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/transplantation , Transplantation, Heterologous
15.
J Infect Dis ; 215(suppl_3): S142-S151, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28520967

ABSTRACT

Current HIV therapy is not curative regardless of how soon after infection it is initiated or how long it is administered, and therapy interruption almost invariably results in robust viral rebound. Human immunodeficiency virus persistence is therefore the major obstacle to a cure for AIDS. The testing and implementation of novel yet unproven approaches to HIV eradication that could compromise the health status of HIV-infected individuals might not be ethically warranted. Therefore, adequate in vitro and in vivo evidence of efficacy is needed to facilitate the clinical implementation of promising strategies for an HIV cure. Animal models of HIV infection have a strong and well-documented history of bridging the gap between laboratory discoveries and eventual clinical implementation. More recently, animal models have been developed and implemented for the in vivo evaluation of novel HIV cure strategies. In this article, we review the recent progress in this rapidly moving area of research, focusing on the two most promising model systems: humanized mice and nonhuman primates.


Subject(s)
Anti-HIV Agents/therapeutic use , Disease Models, Animal , HIV Infections/drug therapy , HIV Infections/virology , HIV/physiology , Virus Latency/drug effects , Animals , HIV/drug effects , Humans , Primates
16.
J Virol ; 90(9): 4681-4695, 2016 May.
Article in English | MEDLINE | ID: mdl-26912618

ABSTRACT

UNLABELLED: The multifunctional HIV-1 accessory protein Vif counters the antiviral activities of APOBEC3G (A3G) and APOBEC3F (A3F), and some Vifs counter stable alleles of APOBEC3H (A3H). Studies in humanized mice have shown that HIV-1 lacking Vif expression is not viable. Here, we look at the relative contributions of the three APOBEC3s to viral extinction. Inoculation of bone marrow/liver/thymus (BLT) mice with CCR5-tropic HIV-1JRCSF(JRCSF) expressing a vif gene inactive for A3G but not A3F degradation activity (JRCSFvifH42/43D) displayed either no or delayed replication. JRCSF expressing a vif gene mutated to inactivate A3F degradation but not A3G degradation (JRCSFvifW79S) always replicated to high viral loads with variable delays. JRCSF with vif mutated to lack both A3G and A3F degradation activities (JRCSFvifH42/43DW79S) failed to replicate, mimicking JRCSF without Vif expression (JRCSFΔvif). JRCSF and JRCSFvifH42/43D, but not JRCSFvifW79S or JRCSFvifH42/43DW79S, degraded APOBEC3D. With one exception, JRCSFs expressing mutant Vifs that replicated acquired enforced vif mutations. These mutations partially restored A3G or A3F degradation activity and fully replaced JRCSFvifH42/43D or JRCSFvifW79S by 10 weeks. Surprisingly, induced mutations temporally lagged behind high levels of virus in blood. In the exceptional case, JRCSFvifH42/43D replicated after a prolonged delay with no mutations in vif but instead a V27I mutation in the RNase H coding sequence. JRCSFvifH42/43D infections exhibited massive GG/AG mutations in pol viral DNA, but in viral RNA, there were no fixed mutations in the Gag or reverse transcriptase coding sequence. A3H did not contribute to viral extinction but, in combination with A3F, could delay JRCSF replication. A3H was also found to hypermutate viral DNA. IMPORTANCE: Vif degradation of A3G and A3F enhances viral fitness, as virus with even a partially restored capacity for degradation outgrows JRCSFvifH42/43D and JRCSFvifW79S. Unexpectedly, fixation of mutations that replaced H42/43D or W79S in viral RNA lagged behind the appearance of high viral loads. In one exceptional JRCSFvifH42/43D infection, vif was unchanged but replication proceeded after a long delay. These results suggest that Vif binds and inhibits the non-cytosine deaminase activities of intact A3G and intact A3F, allowing JRCSFvifH42/43D and JRCSFvifW79S to replicate with reduced fitness. Subsequently, enhanced Vif function is acquired by enforced mutations. In infected cells, JRCSFΔvif and JRCSFvifH42/43DW79S are exposed to active A3F and A3G and fail to replicate. JRCSFvifH42/43D Vif degrades A3F and, in some cases, overcomes A3G mutagenic activity to replicate. Vif may have evolved to inhibit A3F and A3G by stoichiometric binding and subsequently acquired the ability to target these proteins to proteasomes.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine Deaminase/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Virus Replication , APOBEC-3G Deaminase , Alleles , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Base Sequence , Cell Line , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , DNA Mutational Analysis , DNA, Viral , Disease Models, Animal , Gene Amplification , HIV Infections/immunology , Haplotypes , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Protein Binding , Proteolysis , Sequence Alignment , Viral Load , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
17.
PLoS Pathog ; 11(8): e1005075, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26271040

ABSTRACT

Vaginal HIV transmission accounts for the majority of new infections worldwide. Currently, multiple efforts to prevent HIV transmission are based on pre-exposure prophylaxis with various antiretroviral drugs. Here, we describe two novel nanoformulations of the reverse transcriptase inhibitor rilpivirine for pericoital and coitus-independent HIV prevention. Topically applied rilpivirine, encapsulated in PLGA nanoparticles, was delivered in a thermosensitive gel, which becomes solid at body temperature. PLGA nanoparticles with encapsulated rilpivirine coated the reproductive tract and offered significant protection to BLT humanized mice from a vaginal high-dose HIV-1 challenge. A different nanosuspension of crystalline rilpivirine (RPV LA), administered intramuscularly, protected BLT mice from a single vaginal high-dose HIV-1 challenge one week after drug administration. Using transmitted/founder viruses, which were previously shown to establish de novo infection in humans, we demonstrated that RPV LA offers significant protection from two consecutive high-dose HIV-1 challenges one and four weeks after drug administration. In this experiment, we also showed that, in certain cases, even in the presence of drug, HIV infection could occur without overt or detectable systemic replication until levels of drug were reduced. We also showed that infection in the presence of drug can result in acquisition of multiple viruses after subsequent exposures. These observations have important implications for the implementation of long-acting antiretroviral formulations for HIV prevention. They provide first evidence that occult infections can occur, despite the presence of sustained levels of antiretroviral drugs. Together, our results demonstrate that topically- or systemically administered rilpivirine offers significant coitus-dependent or coitus-independent protection from HIV infection.


Subject(s)
HIV Infections/prevention & control , Rilpivirine/administration & dosage , Animals , Anti-HIV Agents/administration & dosage , Chromatography, High Pressure Liquid , Disease Models, Animal , HIV Infections/transmission , HeLa Cells , Humans , Mice , Nanoparticles/administration & dosage , Vaginal Creams, Foams, and Jellies/pharmacology
18.
Retrovirology ; 13(1): 36, 2016 05 21.
Article in English | MEDLINE | ID: mdl-27206407

ABSTRACT

BACKGROUND: The latent reservoir in resting CD4(+) T cells presents a major barrier to HIV cure. Latency-reversing agents are therefore being developed with the ultimate goal of disrupting the latent state, resulting in induction of HIV expression and clearance of infected cells. Histone deacetylase inhibitors (HDACi) have received a significant amount of attention for their potential as latency-reversing agents. RESULTS: Here, we have investigated the in vitro and systemic in vivo effect of panobinostat, a clinically relevant HDACi, on HIV latency. We showed that panobinostat induces histone acetylation in human PBMCs. Further, we showed that panobinostat induced HIV RNA expression and allowed the outgrowth of replication-competent virus ex vivo from resting CD4(+) T cells of HIV-infected patients on suppressive antiretroviral therapy (ART). Next, we demonstrated that panobinostat induced systemic histone acetylation in vivo in the tissues of BLT humanized mice. Finally, in HIV-infected, ART-suppressed BLT mice, we evaluated the effect of panobinostat on systemic cell-associated HIV RNA and DNA levels and the total frequency of latently infected resting CD4(+) T cells. Our data indicate that panobinostat treatment resulted in systemic increases in cellular levels of histone acetylation, a key biomarker for in vivo activity. However, panobinostat did not affect the levels of cell-associated HIV RNA, HIV DNA, or latently infected resting CD4(+) T cells. CONCLUSION: We have demonstrated robust levels of systemic histone acetylation after panobinostat treatment of BLT humanized mice; and we did not observe a detectable change in the levels of cell-associated HIV RNA, HIV DNA, or latently infected resting CD4(+) T cells in HIV-infected, ART-suppressed BLT mice. These results are consistent with the modest effects noted in vitro and suggest that combination therapies may be necessary to reverse latency and enable clearance. Animal models will contribute to the progress towards an HIV cure.


Subject(s)
Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , DNA, Viral/metabolism , HIV-1/drug effects , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , RNA, Viral/metabolism , Virus Latency/drug effects , Acetylation , Animals , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Transgenic , Panobinostat , RNA, Viral/blood , Virus Activation/drug effects , Virus Replication/drug effects
20.
J Antimicrob Chemother ; 71(6): 1586-96, 2016 06.
Article in English | MEDLINE | ID: mdl-27002074

ABSTRACT

OBJECTIVES: Pre-exposure prophylaxis (PrEP) using antiretroviral drugs (ARVs) has been shown to reduce HIV transmission in people at high risk of HIV infection. Adherence to PrEP strongly correlates with the level of HIV protection. Long-acting injectable ARVs provide sustained systemic drug exposures over many weeks and can improve adherence due to infrequent parenteral administration. Here, we evaluated a new long-acting formulation of raltegravir for prevention of vaginal HIV transmission. METHODS: Long-acting raltegravir was administered subcutaneously to BALB/c, NSG (NOD-scid-gamma) and humanized BLT (bone marrow-liver-thymus) mice and rhesus macaques. Raltegravir concentration in peripheral blood and tissue was analysed. Suppression of HIV replication was assessed in infected BLT mice. Two high-dose HIV vaginal challenges were used to evaluate protection from HIV transmission in BLT mice. RESULTS: Two weeks after a single subcutaneous injection of long-acting raltegravir in BLT mice (7.5 mg) and rhesus macaques (160 mg), the plasma concentration of raltegravir was comparable to 400 mg orally, twice daily in humans. Serum collected from mice 3 weeks post-administration of long-acting raltegravir efficiently blocked HIV infection of TZM-bl indicator cells in vitro. Administration of long-acting raltegravir suppressed viral RNA in plasma and cervico-vaginal fluids of infected BLT mice, demonstrating penetration of active raltegravir into the female reproductive tract. Using transmitted/founder HIV we observed that BLT mice administered a single subcutaneous dose of long-acting raltegravir were protected from two high-dose HIV vaginal challenges 1 week and 4 weeks after drug administration. CONCLUSIONS: These preclinical results demonstrated the efficacy of long-acting raltegravir in preventing vaginal HIV transmission.


Subject(s)
Anti-HIV Agents/administration & dosage , Delayed-Action Preparations/administration & dosage , Disease Transmission, Infectious/prevention & control , HIV Infections/prevention & control , Pre-Exposure Prophylaxis/methods , Raltegravir Potassium/administration & dosage , Vagina/virology , Animals , Chemoprevention/methods , Female , HIV Infections/transmission , Injections, Subcutaneous , Macaca mulatta , Mice, Inbred BALB C , Mice, SCID , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL