Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Biochem Pharmacol ; 195: 114844, 2022 01.
Article in English | MEDLINE | ID: mdl-34801521

ABSTRACT

Latency reversing agents (LRAs), such as protein kinase C (PKC) agonists, constitute a promising strategy for exposing and eliminating the HIV-1 latent reservoir. PKC agonists activate NF-κB and induce deleterious pro-inflammatory cytokine production. Adjuvant pharmacological agents, such as ruxolitinib, a JAK inhibitor, have previously been combined with LRAs to reduce deleterious pro-inflammatory cytokine secretion without inhibiting HIV-1 reactivation in vitro. Histone deacetylase inhibitors (HDACi) are known to dampen pro-inflammatory cytokine secretion in the context of other diseases and synergize with LRAs to reactivate latent HIV-1. This study investigates whether a panel of epigenetic modifiers, including HDACi, could dampen PKC-induced pro-inflammatory cytokine secretion during latency reversal. We screened an epigenetic modifier library for compounds that reduced intracellular IL-6 production induced by the PKC agonist Ingenol-3,20-dibenzoate. We further tested the most promising epigenetic inhibitor class, HDACi, for their ability to reduce pro-inflammatory cytokines and reactivate latent HIV-1 ex vivo. We identified nine epigenetic modulators that reduced PKC-induced intracellular IL-6. In cells from aviremic individuals living with HIV-1, the HDAC1-3 inhibitor, suberohydroxamic acid (SBHA), reduced secretion of pro-inflammatory cytokines TNF-α, IL-5, IL-2r, and IL-17 but did not significantly reactivate latent HIV-1 when combined with Ingenol-3,20-dibenzoate. Combining SBHA and Ingenol-3,20-dibenzoate reduces deleterious cytokine production during latency reversal but does not induce significant viral reactivation in aviremic donor PBMCs. The ability of SBHA to reduce PKC-induced pro-inflammatory cytokines when combined with Ingenol-3,20-dibenzoate suggests SBHA can be used to reduced PKC induced pro-inflammatory cytokines but not to achieve latency reversal in the context of HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cytokines/metabolism , Diterpenes/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Inflammation Mediators/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Humans , Protein Kinase C/metabolism , Virus Activation/drug effects , Virus Latency/drug effects
2.
PLoS Pathog ; 17(9): e1009581, 2021 09.
Article in English | MEDLINE | ID: mdl-34529720

ABSTRACT

The switch between HIV latency and productive transcription is regulated by an auto-feedback mechanism initiated by the viral trans-activator Tat, which functions to recruit the host transcription elongation factor P-TEFb to proviral HIV. A heterodimeric complex of CDK9 and one of three cyclin T subunits, P-TEFb is expressed at vanishingly low levels in resting memory CD4+ T cells and cellular mechanisms controlling its availability are central to regulation of the emergence of HIV from latency. Using a well-characterized primary T-cell model of HIV latency alongside healthy donor memory CD4+ T cells, we characterized specific T-cell receptor (TCR) signaling pathways that regulate the generation of transcriptionally active P-TEFb, defined as the coordinate expression of cyclin T1 and phospho-Ser175 CDK9. Protein kinase C (PKC) agonists, such as ingenol and prostratin, stimulated active P-TEFb expression and reactivated latent HIV with minimal cytotoxicity, even in the absence of intracellular calcium mobilization with an ionophore. Unexpectedly, inhibition-based experiments demonstrated that PKC agonists and TCR-mobilized diacylglycerol signal through MAP kinases ERK1/2 rather than through PKC to effect the reactivation of both P-TEFb and latent HIV. Single-cell and bulk RNA-seq analyses revealed that of the four known isoforms of the Ras guanine nucleotide exchange factor RasGRP, RasGRP1 is by far the predominantly expressed diacylglycerol-dependent isoform in CD4+ T cells. RasGRP1 should therefore mediate the activation of ERK1/2 via Ras-Raf signaling upon TCR co-stimulation or PKC agonist challenge. Combined inhibition of the PI3K-mTORC2-AKT-mTORC1 pathway and the ERK1/2 activator MEK prior to TCR co-stimulation abrogated active P-TEFb expression and substantially suppressed latent HIV reactivation. Therefore, contrary to prevailing models, the coordinate reactivation of P-TEFb and latent HIV in primary T cells following either TCR co-stimulation or PKC agonist challenge is independent of PKC but rather involves two complementary signaling arms of the TCR cascade, namely, RasGRP1-Ras-Raf-MEK-ERK1/2 and PI3K-mTORC2-AKT-mTORC1.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , HIV/physiology , Positive Transcriptional Elongation Factor B/metabolism , Protein Kinase C/metabolism , Virus Latency/physiology , CD4-Positive T-Lymphocytes/virology , HIV Infections/metabolism , HIV Infections/virology , Humans , Signal Transduction/physiology , Virus Activation/physiology
3.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33400687

ABSTRACT

Antiretroviral therapies (ARTs) abrogate HIV replication; however, infection persists as long-lived reservoirs of infected cells with integrated proviruses, which reseed replication if ART is interrupted. A central tenet of our current understanding of this persistence is that infected cells are shielded from immune recognition and elimination through a lack of antigen expression from proviruses. Efforts to cure HIV infection have therefore focused on reactivating latent proviruses to enable immune-mediated clearance, but these have yet to succeed in reducing viral reservoirs. Here, we revisited the question of whether HIV reservoirs are predominately immunologically silent from a new angle: by querying the dynamics of HIV-specific T cell responses over long-term ART for evidence of ongoing recognition of HIV-infected cells. In longitudinal assessments, we show that the rates of change in persisting HIV Nef-specific responses, but not responses to other HIV gene products, were associated with residual frequencies of infected cells. These Nef-specific responses were highly stable over time and disproportionately exhibited a cytotoxic, effector functional profile, indicative of recent in vivo recognition of HIV antigens. These results indicate substantial visibility of the HIV-infected cells to T cells on stable ART, presenting both opportunities and challenges for the development of therapeutic approaches to curing infection.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Antigens/immunology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Adult , Aged , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cohort Studies , Female , Granzymes/metabolism , HIV Infections/virology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Immune Evasion , Interferon-gamma/metabolism , Longitudinal Studies , Male , Middle Aged , T-Lymphocytes/drug effects , Viral Load , Young Adult
4.
J Med Virol ; 93(2): 726-732, 2021 02.
Article in English | MEDLINE | ID: mdl-32692406

ABSTRACT

Since its first appearance in Wuhan, China, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread throughout the world and has become a global pandemic. Several medical comorbidities have been identified as risk factors for coronavirus disease 2019 (COVID-19). However, it remains unclear whether people living with human immunodefeciency virus (PLWH) are at an increased risk of COVID-19 and severe disease manifestation, with controversial suggestion that HIV-infected individuals could be protected from severe COVID-19 by means of antiretroviral therapy or HIV-related immunosuppression. Several cases of coinfection with HIV and SARS-CoV-2 have been reported from different parts of the globe. This review seeks to provide a holistic overview of SARS-CoV-2 infection in PLWH.


Subject(s)
Anti-HIV Agents/therapeutic use , COVID-19/epidemiology , HIV Infections/epidemiology , Immunocompromised Host , Pandemics , SARS-CoV-2/pathogenicity , Adult , Antiretroviral Therapy, Highly Active/statistics & numerical data , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Coinfection , Comorbidity , Diabetes Mellitus/epidemiology , Diabetes Mellitus/physiopathology , Female , HIV/drug effects , HIV/growth & development , HIV/pathogenicity , HIV Infections/drug therapy , HIV Infections/mortality , HIV Infections/virology , Humans , Hypertension/epidemiology , Hypertension/physiopathology , Male , Middle Aged , SARS-CoV-2/immunology , Survival Analysis , Treatment Outcome
5.
Proc Natl Acad Sci U S A ; 117(50): 32066-32077, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33239444

ABSTRACT

In untreated HIV-1 infection, rapid viral evolution allows escape from immune responses. Viral replication can be blocked by antiretroviral therapy. However, HIV-1 persists in a latent reservoir in resting CD4+ T cells, and rebound viremia occurs following treatment interruption. The reservoir, which is maintained in part by clonal expansion, can be measured using quantitative viral outgrowth assays (QVOAs) in which latency is reversed with T cell activation to allow viral outgrowth. Recent studies have shown that viruses detected in QVOAs prior to treatment interruption often differ from rebound viruses. We hypothesized that autologous neutralizing antibodies directed at the HIV-1 envelope (Env) protein might block outgrowth of some reservoir viruses. We modified the QVOA to reflect pressure from low concentrations of autologous antibodies and showed that outgrowth of a substantial but variable fraction of reservoir viruses is blocked by autologous contemporaneous immunoglobulin G (IgG). A reduction in outgrowth of >80% was seen in 6 of 15 individuals. This effect was due to direct neutralization. We established a phylogenetic relationship between rebound viruses and viruses growing out in vitro in the presence of autologous antibodies. Some large infected cell clones detected by QVOA carried neutralization-sensitive viruses, providing a cogent explanation for differences between rebound virus and viruses detected in standard QVOAs. Measurement of the frequency of reservoir viruses capable of outgrowth in the presence of autologous IgG might allow more accurate prediction of time to viral rebound. Ultimately, therapeutic immunization targeting the subset of variants resistant to autologous IgG might contribute to a functional cure.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/therapy , HIV-1/immunology , Virus Replication/immunology , Adult , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Blood Transfusion, Autologous/methods , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Combined Modality Therapy/methods , Female , HIV Antibodies/blood , HIV Antibodies/isolation & purification , HIV Antibodies/therapeutic use , HIV Infections/blood , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Immunoglobulin G/therapeutic use , Leukapheresis , Male , Middle Aged , Primary Cell Culture , Virus Latency/drug effects , Virus Latency/immunology , Virus Replication/drug effects , env Gene Products, Human Immunodeficiency Virus/immunology
7.
PLoS Pathog ; 15(7): e1007869, 2019 07.
Article in English | MEDLINE | ID: mdl-31291371

ABSTRACT

Clonal expansion of HIV infected cells plays an important role in the formation and persistence of the reservoir that allows the virus to persist, in DNA form, despite effective antiretroviral therapy. We used integration site analysis to ask if there is a similar clonal expansion of SIV infected cells in macaques. We show that the distribution of HIV and SIV integration sites in vitro is similar and that both viruses preferentially integrate in many of the same genes. We obtained approximately 8000 integration sites from blood samples taken from SIV-infected macaques prior to the initiation of ART, and from blood, spleen, and lymph node samples taken at necropsy. Seven clones were identified in the pre-ART samples; one persisted for a year on ART. An additional 100 clones were found only in on-ART samples; a number of these clones were found in more than one tissue. The timing and extent of clonal expansion of SIV-infected cells in macaques and HIV-infected cells in humans is quite similar. This suggests that SIV-infected macaques represent a useful model of the clonal expansion of HIV infected cells in humans that can be used to evaluate strategies intended to control or eradicate the viral reservoir.


Subject(s)
HIV Infections/drug therapy , HIV Infections/virology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , Disease Reservoirs/virology , HIV Infections/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , In Vitro Techniques , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/pathogenicity , Viral Load/drug effects , Virus Integration/genetics , Virus Integration/physiology , Virus Replication/drug effects
8.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282829

ABSTRACT

The presence of a reservoir of latently infected cells in HIV-infected patients is a major barrier towards finding a cure. One active cure strategy is to find latency-reversing agents that induce viral reactivation, thus leading to immune cell recognition and elimination of latently infected cells, known as the shock-and-kill strategy. Therefore, the identification of molecules that reactivate latent HIV and increase immune activation has the potential to further these strategies into the clinic. Here, we characterized synthetic molecules composed of a TLR2 and a TLR7 agonist (dual TLR2/7 agonists) as latency-reversing agents and compared their activity with that of the TLR2 agonist Pam2CSK4 and the TLR7 agonist GS-9620. We found that these dual TLR2/7 agonists reactivate latency by 2 complementary mechanisms. The TLR2 component reactivates HIV by inducing NF-κB activation in memory CD4+ T cells, while the TLR7 component induces the secretion of TNF-α by monocytes and plasmacytoid dendritic cells, promoting viral reactivation in CD4+ T cells. Furthermore, the TLR2 component induces the secretion of IL-22, which promotes an antiviral state and blocks HIV infection in CD4+ T cells. Our study provides insight into the use of these agonists as a multipronged approach targeting eradication of latent HIV.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , HIV-1/physiology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 7/agonists , Virus Activation/drug effects , Adolescent , Adult , Aged , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Evaluation, Preclinical , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , Humans , Interleukins/immunology , Interleukins/metabolism , Jurkat Cells , Lipopeptides/pharmacology , Lipopeptides/therapeutic use , Male , Middle Aged , Primary Cell Culture , Pteridines/pharmacology , Pteridines/therapeutic use , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Virus Activation/immunology , Virus Latency/drug effects , Virus Latency/immunology , Young Adult , Interleukin-22
9.
Curr HIV Res ; 16(2): 143-150, 2018.
Article in English | MEDLINE | ID: mdl-29968539

ABSTRACT

BACKGROUND: The role and mechanism of drug use or abuse in Antiretroviral Therapy (ART)-treated HIV disease are not completely known. METHODS: To investigate the impact of drug use on HIV pathogenesis without confounding by HIV replication and ART adherence, we first analyzed the data from our clinical database in 103 HIV+ subjects with viral-suppressed ART treatment by a multiple regression test. RESULTS: We found that HIV+ drug users had lower CD4+ T cell counts but higher CD8+ T cell counts compared to HIV+ non-drug users, and both drug use and nadir CD4+ T cell counts was independently associated with CD4+ T cell recovery after controlling for sex and age. Next, we enrolled individuals from four study groups, HIV-negative and HIV+ subjects without any substance use, HIV-negative and HIV+ subjects with current illicit drug use (either non-injection cocaine or cannabis). All HIV+ subjects were viral-suppressed with ART treatment (≥ 2 years). Notably, HIV+ drug users had increased plasma anti-CD4 IgG levels compared to the other three study groups which were inversely correlated with decreased CD4+ T cell counts only in HIV+ drug users. There was a significant increase in CD4+ T cell recovery following ART in HIV+ non-drug users but not in HIV+ drug users. Anti-CD4 IgGs purified from plasma of HIV+ drug users induced CD4+ T cell death in vitro through Antibody-Dependent Cytotoxicity (ADCC). CONCLUSION: These results suggest that drug use prevents immune reconstitution in HIV-infected individuals despite long-term ART treatment and viral suppression.


Subject(s)
Anti-HIV Agents/pharmacology , CD4 Antigens/antagonists & inhibitors , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , HIV Antibodies/pharmacology , HIV Infections/virology , Immunoglobulin G/pharmacology , Anti-HIV Agents/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Female , HIV Antibodies/immunology , HIV Antibodies/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Male
10.
mBio ; 8(3)2017 05 02.
Article in English | MEDLINE | ID: mdl-28465428

ABSTRACT

Antiviral innate host defenses against acute viral infections include suppression of host protein synthesis to restrict viral protein production. Less is known about mechanisms by which viral pathogens subvert host antiviral innate responses for establishing their replication and dissemination. We investigated early innate defense against human immunodeficiency virus (HIV) infection and viral evasion by utilizing human CD4+ T cell cultures in vitro and a simian immunodeficiency virus (SIV) model of AIDS in vivo Our data showed that early host innate defense against the viral infection involves GCN2-ATF4 signaling-mediated suppression of global protein synthesis, which is exploited by the virus for supporting its own replication during early viral infection and dissemination in the gut mucosa. Suppression of protein synthesis and induction of protein kinase GCN2-ATF4 signaling were detected in the gut during acute SIV infection. These changes diminished during chronic viral infection. HIV replication induced by serum deprivation in CD4+ T cells was linked to the induction of ATF4 that was recruited to the HIV long terminal repeat (LTR) to promote viral transcription. Experimental inhibition of GCN2-ATF4 signaling either by a specific inhibitor or by amino acid supplementation suppressed the induction of HIV expression. Enhancing ATF4 expression through selenium administration resulted in reactivation of latent HIV in vitro as well as ex vivo in the primary CD4+ T cells isolated from patients receiving suppressive antiretroviral therapy (ART). In summary, HIV/SIV exploits the early host antiviral response through GCN2-ATF4 signaling by utilizing ATF4 for activating the viral LTR transcription to establish initial viral replication and is a potential target for HIV prevention and therapy.IMPORTANCE Understanding how HIV overcomes host antiviral innate defense response in order to establish infection and dissemination is critical for developing prevention and treatment strategies. Most investigations focused on the viral pathogenic mechanisms leading to immune dysfunction following robust viral infection and dissemination. Less is known about mechanisms that enable HIV to establish its presence despite rapid onset of host antiviral innate response. Our novel findings provide insights into the viral strategy that hijacks the host innate response of the suppression of protein biosynthesis to restrict the virus production. The virus leverages transcription factor ATF4 expression during the GCN2-ATF4 signaling response and utilizes it to activate viral transcription through the LTR to support viral transcription and production in both HIV and SIV infections. This unique viral strategy is exploiting the innate response and is distinct from the mechanisms of immune dysfunction after the critical mass of viral loads is generated.


Subject(s)
Activating Transcription Factor 4/metabolism , CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , Host-Pathogen Interactions , Immunity, Innate , Protein Serine-Threonine Kinases/metabolism , Virus Replication , Activating Transcription Factor 4/genetics , Animals , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Disease Models, Animal , Gastrointestinal Tract/virology , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Humans , Immune Evasion , Macaca mulatta , Protein Serine-Threonine Kinases/genetics , Selenium/pharmacology , Signal Transduction , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus , Viral Load , Virus Latency
11.
J Neurovirol ; 23(3): 441-450, 2017 06.
Article in English | MEDLINE | ID: mdl-28251596

ABSTRACT

This study investigated the association of HIV infection and cocaine dependence with cerebral white matter integrity using diffusion tensor imaging (DTI). One hundred thirty-five participants stratified by HIV and cocaine status (26 HIV+/COC+, 37 HIV+/COC-, 37 HIV-/COC+, and 35 HIV-/COC-) completed a comprehensive substance abuse assessment, neuropsychological testing, and MRI with DTI. Among HIV+ participants, all were receiving HIV care and 46% had an AIDS diagnosis. All COC+ participants were current users and met criteria for cocaine use disorder. We used tract-based spatial statistics (TBSS) to assess the relation of HIV and cocaine to fractional anisotropy (FA) and mean diffusivity (MD). In whole-brain analyses, HIV+ participants had significantly reduced FA and increased MD compared to HIV- participants. The relation of HIV and FA was widespread throughout the brain, whereas the HIV-related MD effects were restricted to the corpus callosum and thalamus. There were no significant cocaine or HIV-by-cocaine effects. These DTI metrics correlated significantly with duration of HIV disease, nadir CD4+ cell count, and AIDS diagnosis, as well as some measures of neuropsychological functioning. These results suggest that HIV is related to white matter integrity throughout the brain, and that HIV-related effects are more pronounced with increasing duration of infection and greater immune compromise. We found no evidence for independent effects of cocaine dependence on white matter integrity, and cocaine dependence did not appear to exacerbate the effects of HIV.


Subject(s)
Cerebral Cortex/pathology , Cocaine-Related Disorders/pathology , Corpus Callosum/pathology , HIV Infections/pathology , Thalamus/pathology , White Matter/pathology , Adult , Anisotropy , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/virology , Cocaine-Related Disorders/complications , Cocaine-Related Disorders/diagnostic imaging , Cocaine-Related Disorders/immunology , Corpus Callosum/diagnostic imaging , Corpus Callosum/virology , Diffusion Tensor Imaging , Female , HIV Infections/complications , HIV Infections/diagnostic imaging , HIV Infections/immunology , Humans , Male , Middle Aged , Neuropsychological Tests , Thalamus/diagnostic imaging , Thalamus/virology , White Matter/diagnostic imaging , White Matter/virology
12.
J Gen Virol ; 98(4): 799-809, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28113052

ABSTRACT

Antiretroviral therapy (ART) can control human immunodeficiency virus-1 (HIV-1) replication in infected individuals. Unfortunately, patients remain persistently infected owing to the establishment of latent infection requiring that ART be maintained indefinitely. One strategy being pursued involves the development of latency-reversing agents (LRAs) to eliminate the latent arm of the infection. One class of molecules that has been tested for LRA activity is the epigenetic modulating compounds histone deacetylases inhibitors (HDACis). Previously, initial screening of these molecules typically commenced using established cell models of viral latency, and although certain drugs such as the HDACi suberoylanilide hydroxamic acid demonstrated strong activity in these models, it did not translate to comparable activity with patient samples. Here we developed a primary cell model of viral latency using primary resting CD4+ T cells infected with Vpx-complemented HIV-1 and found that the activation profile using previously described LRAs mimicked that obtained with patient samples. This primary cell model was used to evaluate 94 epigenetic compounds. Not surprisingly, HDACis were found to be the strongest activators. However, within the HDACi class, the most active LRAs with the least pronounced toxicity contained a benzamide functional moiety with a pyridyl cap group, as exemplified by the HDACi chidamide. The results indicate that HDACis with a benzamide moiety and pyridyl cap group should be considered for further drug development in the pursuit of a successful viral clearance strategy.


Subject(s)
Benzamides/metabolism , CD4-Positive T-Lymphocytes/virology , Drug Evaluation, Preclinical/methods , HIV-1/physiology , Histone Deacetylase Inhibitors/metabolism , Virus Activation/drug effects , Virus Latency/drug effects , Aminopyridines/chemistry , Aminopyridines/metabolism , Benzamides/chemistry , Cells, Cultured , Histone Deacetylase Inhibitors/chemistry , Humans
13.
Retrovirology ; 13(1): 88, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27998278

ABSTRACT

BACKGROUND: Despite the durable viral suppression afforded by antiretroviral therapy, HIV-1 eradication will require strategies to target latently infected cells that persist in infected individuals. Protein kinase C (PKC) activation is a promising strategy to reactivate latent proviruses and allow for subsequent recognition and clearance of infected cells by the immune system. Ingenol derivatives are PKC agonists that induce latency reversal but also lead to T cell activation and the release of pro-inflammatory cytokines, which would be undesirable in vivo. In this work, we sought to identify compounds that would suppress pro-inflammatory cytokine production in the context of PKC activation. DESIGN AND METHODS: We performed an in vitro screen to identify compounds that could dampen pro-inflammatory cytokine release associated with T cell activation, using IL-6 as a model cytokine. We then tested the ability of the most promising screening hit, the FDA-approved Janus Kinase (JAK) inhibitor ruxolitinib, to diminish release of multiple cytokines and its effect on latency reversal using cells from HIV-1-positive, aviremic participants. RESULTS: We demonstrate that co-administration of ruxolitinib with ingenol-3,20-dibenzoate significantly reduces pro-inflammatory cytokine release without impairing latency reversal ex vivo. CONCLUSION: The combination of ingenol compounds and JAK inhibition represents a novel strategy for HIV-1 eradication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cytokines/metabolism , Diterpenes/pharmacology , HIV-1/physiology , Janus Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Virus Latency , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Drug Evaluation, Preclinical , HIV Infections/drug therapy , HIV-1/drug effects , High-Throughput Screening Assays , Humans , Interleukin-6/analysis , Lymphocyte Activation , Nitriles , Protein Kinase C/metabolism , Pyrimidines , Virus Activation
14.
Virol J ; 13(1): 177, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27769267

ABSTRACT

BACKGROUND: HIV-1 latency is a major obstacle for HIV-1 eradication. Extensive efforts are being directed toward the reactivation of latent HIV reservoirs with the aim of eliminating latently infected cells via the host immune system and/or virus-mediated cell lysis. RESULTS: We screened over 1,500 small molecules and kinase inhibitors and found that a small molecule, PKC412 (midostaurin, a broad-spectrum kinase inhibitor), can stimulate viral transcription and expression from the HIV-1 latently infected ACH2 cell line and primary resting CD4+ T cells. PKC412 reactivated HIV-1 expression in ACH2 cells in a dose- and time-dependent manner. Our results also suggest that the nuclear factor κB (NF-κB) signaling could be one of cellular pathways activated during PKC412-mediated activation of latent HIV-1 expression. Additionally, combining PKC412 with the HDAC inhibitor vorinostat (VOR) had an additive effect on HIV-1 reactivation in both ACH2 cells and infected resting CD4+ T cells. CONCLUSIONS: These studies provide evidence that PKC412 is a new compound with the potential for optimization as a latency-reactivator to eradicate HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , HIV-1/drug effects , Protein Kinase Inhibitors/metabolism , Staurosporine/analogs & derivatives , Virus Activation/drug effects , Virus Latency/drug effects , Drug Evaluation, Preclinical , HIV-1/physiology , Humans , Staurosporine/metabolism
16.
AIDS Res Hum Retroviruses ; 32(12): 1237-1247, 2016 12.
Article in English | MEDLINE | ID: mdl-27356854

ABSTRACT

Failure of combination antiretroviral (ARV) therapy in HIV-infected patients is often associated with the emergence of drug resistance-associated mutations (RAMs). To facilitate analysis of the barrier to resistance at therapeutically relevant ARV concentrations, we performed fixed-dose in vitro HIV-1 drug resistance selection assays using the immortalized MT-2 T-cell line and primary human CD4+ T cells with a panel of FDA-approved ARVs, each at their respective cell culture equivalent clinical trough concentration (CCE Cmin). At high multiples of its CCE Cmin, emtricitabine (FTC) selected for the rapid emergence of M184I/V, a result consistent with resistance emergence in vivo. While the rate of viral breakthrough in the presence of rilpivirine or efavirenz was delayed relative to FTC, both inhibitors selected for virus with known clinically relevant RAMs. No viral breakthrough was observed for the protease inhibitor atazanavir even at subtherapeutic drug concentrations, which is consistent with its previously characterized high in vivo barrier to resistance. Depending on assay conditions, treatment with integrase inhibitors elvitegravir and raltegravir resulted in breakthrough of both resistant and wild-type virus. The RAMs observed in drug selections were not detected above a 2% threshold by deep sequencing in the in vitro virus inoculum, and only rarely in isolates from treatment-naive HIV+ patients. These new viral breakthrough assays facilitate the analysis of multiple experimental replicates and conditions in parallel and provide a rapid quantitative means to evaluate drug resistance emergence at therapeutically relevant drug concentrations, which should facilitate the identification of new ARVs with a high barrier to resistance.


Subject(s)
Anti-Retroviral Agents/pharmacology , Drug Resistance, Viral , HIV-1/drug effects , Microbial Sensitivity Tests/methods , Selection, Genetic , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Humans
17.
Antimicrob Agents Chemother ; 59(12): 7197-204, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26324276

ABSTRACT

Cryptococcal antigen screening is recommended among people living with AIDS when entering HIV care with a CD4 count of <100 cells/µl, and preemptive fluconazole monotherapy treatment is recommended for those with subclinical cryptococcal antigenemia. Yet, knowledge is limited of current antimicrobial resistance in Africa. We examined antifungal drug susceptibility in 198 clinical isolates collected from Kampala, Uganda, between 2010 and 2014 using the CLSI broth microdilution assay. In comparison with two previous studies from 1998 to 1999 that reported an MIC50 of 4 µg/ml and an MIC90 of 8 µg/ml prior to widespread human fluconazole and agricultural azole fungicide usage, we report an upward shift in the fluconazole MIC50 to 8 µg/ml and an MIC90 value of 32 µg/ml, with 31% of isolates with a fluconazole MIC of ≥ 16 µg/ml. We observed an amphotericin B MIC50 of 0.5 µg/ml and an MIC90 of 1 µg/ml, of which 99.5% of isolates (197 of 198 isolates) were still susceptible. No correlation between MIC and clinical outcome was observed in the context of amphotericin B and fluconazole combination induction therapy. We also analyzed Cryptococcus susceptibility to sertraline, with an MIC50 of 4 µg/ml, suggesting that sertraline is a promising oral, low-cost, available, novel medication and a possible alternative to fluconazole. Although the CLSI broth microdilution assay is ideal to standardize results, limit human bias, and increase assay capacity, such assays are often inaccessible in low-income countries. Thus, we also developed and validated an assay that could easily be implemented in a resource-limited setting, with similar susceptibility results (P = 0.52).


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Cryptococcus neoformans/drug effects , Drug Resistance, Fungal , Fluconazole/therapeutic use , Meningitis, Cryptococcal/drug therapy , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Coinfection , Cryptococcus neoformans/genetics , Cryptococcus neoformans/growth & development , Drug Therapy, Combination , Female , HIV/isolation & purification , HIV Infections/diagnosis , HIV Infections/immunology , HIV Infections/virology , Humans , Male , Meningitis, Cryptococcal/diagnosis , Meningitis, Cryptococcal/immunology , Meningitis, Cryptococcal/microbiology , Microbial Sensitivity Tests , Multilocus Sequence Typing , Treatment Outcome , Uganda
18.
PLoS One ; 10(4): e0121099, 2015.
Article in English | MEDLINE | ID: mdl-25830320

ABSTRACT

The continued burden of HIV in resource-limited regions such as parts of sub-Saharan Africa, combined with adverse effects and potential risks of resistance to existing antiretroviral therapies, emphasize the need to identify new HIV inhibitors. Here we performed a virtual screen of molecules from the pan-African Natural Product Library, the largest collection of medicinal plant-derived pure compounds on the African continent. We identified eight molecules with structural similarity to reported interactors of Vpu, an HIV-1 accessory protein with reported ion channel activity. Using in vitro HIV-1 replication assays with a CD4+ T cell line and peripheral blood mononuclear cells, we confirmed antiviral activity and minimal cytotoxicity for two compounds, ixoratannin A-2 and boldine. Notably, ixoratannin A-2 retained inhibitory activity against recombinant HIV-1 strains encoding patient-derived mutations that confer resistance to protease, non-nucleoside reverse transcriptase, or integrase inhibitors. Moreover, ixoratannin A-2 was less effective at inhibiting replication of HIV-1 lacking Vpu, supporting this protein as a possible direct or indirect target. In contrast, boldine was less effective against a protease inhibitor-resistant HIV-1 strain. Both ixoratannin A-2 and boldine also inhibited in vitro replication of hepatitis C virus (HCV). However, BIT-225, a previously-reported Vpu inhibitor, demonstrated antiviral activity but also cytotoxicity in HIV-1 and HCV replication assays. Our work identifies pure compounds derived from African plants with potential novel activities against viruses that disproportionately afflict resource-limited regions of the world.


Subject(s)
Aporphines/pharmacology , Biological Products/chemistry , HIV-1/drug effects , Proanthocyanidins/pharmacology , Aporphines/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Line , Drug Resistance, Viral , Guanidines/pharmacology , HIV-1/physiology , Hepacivirus/drug effects , Hepacivirus/physiology , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Molecular Docking Simulation , Proanthocyanidins/chemistry , Pyrazoles/pharmacology , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication/drug effects
19.
Biochem Pharmacol ; 94(4): 241-56, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25732195

ABSTRACT

HIV-1 infection cannot be cured due to reservoirs formed early after infection. Decreasing the massive CD4+ T cell activation that occurs at the beginning of the disease would delay reservoir seeding, providing a better prognosis for patients. CD4+ T cell activation is mediated by protein kinase C (PKC) theta (θ), which is involved in T-cell proliferation, as well as NF-κB, NF-AT, and AP-1 activation. We found that PKCθ activity increased viral replication, but also that HIV-1 induced higher activation of PKCθ in infected CD4+ T cells, creating a feedback loop. Therefore, specific inhibition of PKCθ activity could contribute to control HIV-1 replication. We tested the efficacy of seven PKCθ specific inhibitors to control HIV-1 replication in CD4+ T cells and selected two of the more potent and safer: CGX1079 and CGX0471. They reduced PKCθ phosphorylation at T538 and its translocation to the plasma membrane, which correlated with decreased HIV-1 retrotranscription through partial inhibition of SAMHD1 antiviral activity, rendering lower proviral integration. CGX1079 and CGX0471 also interfered with viral transcription, which would reduce the production of new virions, as well as the subsequent spread and infection of new targets that would increase the reservoir size. CGX1079 and CGX0471 did not completely abrogate T-cell functions such as proliferation and CD8-mediated release of IFN-γ in PBMCs from HIV-infected patients, thereby avoiding general immunosuppresion. Consequently, using PKCθ inhibitors as adjuvant of antiretroviral therapy in recently infected patients would decrease the pool of activated CD4+ T cells, thwarting proviral integration and reducing the reservoir size.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , HIV-1/drug effects , Isoenzymes/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Retroelements , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , HIV-1/genetics , HIV-1/physiology , Humans , Jurkat Cells , Monomeric GTP-Binding Proteins/metabolism , Phosphorylation , Protein Kinase C-theta , Protein Transport , SAM Domain and HD Domain-Containing Protein 1 , Transcription, Genetic , Virus Integration/drug effects , Virus Internalization , Virus Replication
20.
J Clin Invest ; 125(5): 1901-12, 2015 May.
Article in English | MEDLINE | ID: mdl-25822022

ABSTRACT

Reversal of HIV-1 latency by small molecules is a potential cure strategy. This approach will likely require effective drug combinations to achieve high levels of latency reversal. Using resting CD4+ T cells (rCD4s) from infected individuals, we developed an experimental and theoretical framework to identify effective latency-reversing agent (LRA) combinations. Utilizing ex vivo assays for intracellular HIV-1 mRNA and virion production, we compared 2-drug combinations of leading candidate LRAs and identified multiple combinations that effectively reverse latency. We showed that protein kinase C agonists in combination with bromodomain inhibitor JQ1 or histone deacetylase inhibitors robustly induce HIV-1 transcription and virus production when directly compared with maximum reactivation by T cell activation. Using the Bliss independence model to quantitate combined drug effects, we demonstrated that these combinations synergize to induce HIV-1 transcription. This robust latency reversal occurred without release of proinflammatory cytokines by rCD4s. To extend the clinical utility of our findings, we applied a mathematical model that estimates in vivo changes in plasma HIV-1 RNA from ex vivo measurements of virus production. Our study reconciles diverse findings from previous studies, establishes a quantitative experimental approach to evaluate combinatorial LRA efficacy, and presents a model to predict in vivo responses to LRAs.


Subject(s)
Anti-HIV Agents/pharmacology , Azepines/pharmacology , Bryostatins/pharmacology , CD4-Positive T-Lymphocytes/virology , Disulfiram/pharmacology , HIV-1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Triazoles/pharmacology , Virus Latency/drug effects , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Drug Evaluation, Preclinical , Drug Synergism , Female , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/physiology , Humans , Lymphocyte Activation , Lymphokines/metabolism , Male , Middle Aged , Phorbol Esters/pharmacology , Protein Kinase C/drug effects , RNA, Messenger/analysis , RNA, Viral/analysis , Transcription, Genetic/drug effects , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL