1
|
Cevaal PM, Kan S, Fisher BM, Moso MA, Tan A, Liu H, Ali A, Tanaka K, Shepherd RA, Kim Y, Ong J, Furtado DL, Holz M, Purcell DFJ, Casan JML, Payne T, Zhao W, Fareh M, McMahon JH, Deeks SG, Hoh R, Telwatte S, Pouton CW, Johnston APR, Caruso F, Symons J, Lewin SR, Roche M. Efficient mRNA delivery to resting T cells to reverse HIV latency. Nat Commun 2025; 16:4979. [PMID: 40442114 PMCID: PMC12122926 DOI: 10.1038/s41467-025-60001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 05/12/2025] [Indexed: 06/02/2025] Open
Abstract
A major hurdle to curing HIV is the persistence of integrated proviruses in resting CD4+ T cells that remain in a transcriptionally silent, latent state. One strategy to eradicate latent HIV is to activate viral transcription, followed by elimination of infected cells through virus-mediated cytotoxicity or immune-mediated clearance. We hypothesised that mRNA-lipid nanoparticle (LNP) technology would provide an opportunity to deliver mRNA encoding proteins able to reverse HIV latency in resting CD4+ T cells. Here we develop an LNP formulation (LNP X) with unprecedented potency to deliver mRNA to hard-to-transfect resting CD4+ T cells in the absence of cellular toxicity or activation. Encapsulating an mRNA encoding the HIV Tat protein, an activator of HIV transcription, LNP X enhances HIV transcription in ex vivo CD4+ T cells from people living with HIV. LNP X further enables the delivery of clustered regularly interspaced short palindromic repeats (CRISPR) activation machinery to modulate both viral and host gene transcription. These findings offer potential for the development of a range of nucleic acid-based T cell therapeutics.
Collapse
Affiliation(s)
- Paula M Cevaal
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Stanislav Kan
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Bridget M Fisher
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Michael A Moso
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Abigail Tan
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Haiyin Liu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Abdalla Ali
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kiho Tanaka
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Rory A Shepherd
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Youry Kim
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jesslyn Ong
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Denzil L Furtado
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Marvin Holz
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Damian F J Purcell
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Joshua M L Casan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Thomas Payne
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Wei Zhao
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Mohamed Fareh
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - James H McMahon
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Sushama Telwatte
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Jori Symons
- Translational Virology, Department of Medical Microbiology, University Medical Center, Utrecht, the Netherlands
| | - Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, Australia.
| | - Michael Roche
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Viox EG, Richard J, Grandea AG, Nguyen K, Harper J, Auger J, Ding S, Gasser R, Prévost J, Marchitto L, Medjahed H, Bourassa C, Gaudette F, Pagliuzza A, Trifone CA, Gavegnano C, Hurwitz SJ, Park J, Clark NM, Hammad I, Capuano S, Martin MA, Schinazi RF, Silvestri G, Kulpa DA, Kumar P, Chomont N, Pazgier M, Smith AB, Sodroski J, Evans DT, Finzi A, Paiardini M. Safety, pharmacokinetics, and biological activity of CD4-mimetic BNM-III-170 in SHIV-infected rhesus macaques. J Virol 2025; 99:e0006225. [PMID: 40192306 PMCID: PMC12090809 DOI: 10.1128/jvi.00062-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 05/21/2025] Open
Abstract
Anti-HIV-1 antibodies capable of mediating ADCC are elicited by the majority of people with HIV-1 and preferentially target the "open," CD4-bound conformation of HIV-1 envelope glycoproteins (Env). However, due to the "closed" conformation sampled by unliganded HIV-1-Envs, these antibodies are ineffective at eliminating infected cells. BNM-III-170 is a small-molecule CD4-mimetic compound that binds the Phe43 cavity of the gp120 subunit of Env, forcing Env to "open up," thus exposing epitopes targeted by CD4-induced (CD4i), ADCC-mediating antibodies. Here, we assessed the safety, pharmacokinetics, and biological activity of BNM-III-170 in uninfected and SHIV-AD8-EO-infected rhesus macaques (RMs). In uninfected RMs, single subcutaneous administrations of 3-36 mg/kg BNM-III-170 were well-tolerated, with serum half-lives ranging from 3 to 6 h. In SHIV-infected RMs, four different regimens were evaluated: 2 × 36 mg/kg daily, 1 × 24 mg/kg, 3 × 36 mg/kg every 7 days, and 3 × 36 mg/kg every 3 days. While toxicity was observed with daily doses, all other regimens demonstrated reasonable safety profiles. No changes in plasma viral loads were observed in SHIV-infected RMs following any of the evaluated BNM-III-170 dosing regimens. However, plasma collected following BNM-III-170 administration was shown to have increased binding to infected cells and to sensitize SHIV AD8-EO virions to neutralization by otherwise non-neutralizing antibodies. In addition, the plasma of treated animals mediated ADCC in the presence of BNM-III-170. These results establish a well-tolerated BNM-III-170 dosing regimen in SHIV-infected RMs and serve as proof of concept for its biological activity in promoting the targeting of infected cells by CD4i ADCC-mediating antibodies. Thus, they inform future studies evaluating CD4mc treatment in ART-treated animals.IMPORTANCEA therapeutic regimen able to eradicate or functionally cure HIV-1 remains elusive and may require a "shock-and-kill" approach to reactivate and then purge the latent HIV-1 reservoir. The small-molecule CD4-mimetic compound BNM-III-170 has previously been shown to (i) sensitize HIV-1-infected cells to ADCC mediated by plasma from people with HIV-1 (PWH) in vitro and (ii) significantly delay the time to viral rebound following ART interruption when combined with anti-CoRBS + anti-cluster A Abs or plasma from PWH in humanized mice. To evaluate the use of BNM-III-170 as part of a kill approach, we characterized the safety, pharmacokinetics, and biological activity of BNM-III-170 in uninfected and SHIV-infected RMs. Our study identifies a tolerable BNM-III-170 dosing regimen in SHIV-infected RMs and provides insights into its antiviral activities; as such, it informs future studies evaluating the efficacy of BNM-III-170 in reducing the viral reservoir.
Collapse
Affiliation(s)
- Elise G. Viox
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jonathan Richard
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Andres G. Grandea
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kevin Nguyen
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Justin Harper
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - James Auger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Shilei Ding
- Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Lorie Marchitto
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | | | | | | | | | - Cesar Ariel Trifone
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Christina Gavegnano
- Department of Pediatrics, Laboratory of Biochemical Pharmacology, Emory Center for AIDS Research, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Selwyn J. Hurwitz
- Department of Pediatrics, Laboratory of Biochemical Pharmacology, Emory Center for AIDS Research, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jun Park
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Natasha M. Clark
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Iman Hammad
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Saverio Capuano
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Malcolm A. Martin
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Raymond F. Schinazi
- Department of Pediatrics, Laboratory of Biochemical Pharmacology, Emory Center for AIDS Research, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Deanna A. Kulpa
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nicolas Chomont
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Marzena Pazgier
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amos B. Smith
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph Sodroski
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - David T. Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montréal, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Tompkins LAR, Khoei A, Kapeljushnik O, Dumond J, Kashuba ADM, Tropsha A, Hubal R, Cottrell ML. HIV Pharmacology Data Repository: Setting the New Information-Sharing Standard for Clinical and Preclinical Pharmacokinetic Studies. Clin Pharmacol Ther 2025. [PMID: 40354181 DOI: 10.1002/cpt.3699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
Drug development approaches increasingly harness computational modeling to predict drug behavior. These in silico approaches, collectively termed "pharmacometrics", have significant value in deriving biological meaning from the analysis of pooled drug concentration vs. time (CvT) datasets. However, the field lacks standardization for pharmacokinetic data description, requiring expert annotation to enable aggregate mining and sharing. These limitations impede data sharing and preservation as mandated by current NIH policies. To this end, we propose a minimum information standard for pharmacokinetic studies composed of three categories (Intervention, System, and Concentration). We implement this standard in the development of a web-based database: the HIV Pharmacology Data Repository (HIV PDR). We describe our technical approach for creating the HIV PDR, the protocols we established for standardized data deposition, and the current content of the database. We also demonstrate the utility of the HIV PDR for pharmacometrics research through computational modeling of CvT data extracted from this new database. Based on these efforts, we propose the HIV PDR as a standard to preserve and share pharmacokinetic data generated through preclinical and clinical HIV research.
Collapse
Affiliation(s)
- Lauren A R Tompkins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adrian Khoei
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Oleg Kapeljushnik
- Renaissance Computing Institute at UNC (RENCI), Chapel Hill, North Carolina, USA
| | - Julie Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Angela D M Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexander Tropsha
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Renaissance Computing Institute at UNC (RENCI), Chapel Hill, North Carolina, USA
| | - Robert Hubal
- Renaissance Computing Institute at UNC (RENCI), Chapel Hill, North Carolina, USA
| | - Mackenzie L Cottrell
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
4
|
Pellaers E, Janssens J, Wils L, Denis A, Bhat A, Van Belle S, Feng D, Christ F, Zhan P, Debyser Z. BRD4 modulator ZL0580 and LEDGINs additively block and lock HIV-1 transcription. Nat Commun 2025; 16:4226. [PMID: 40335477 PMCID: PMC12059001 DOI: 10.1038/s41467-025-59398-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/22/2025] [Indexed: 05/09/2025] Open
Abstract
The persistence of HIV-1 in a latent state within long-lived immune cells remains a major barrier to a cure for HIV-1 infection. The "block-and-lock" strategy aims to silence the HIV-1 provirus permanently using latency promoting agents (LPAs). LEDGINs, a well-known class of LPAs, inhibit the interaction between viral integrase and LEDGF/p75, reducing viral integration and retargeting the provirus to regions resistant to reactivation. However, proximity to enhancers may still permit residual transcription. Given BRD4's central role in the enhancer biology, we now test two BRD4 modulators, JQ1 and ZL0580. Mechanistic studies reveal that JQ1 and ZL0580 have contrasting effects on Tat-dependent HIV-1 transcription, resulting in JQ1 promoting viral reactivation and ZL0580 inducing transcriptional silencing. Combining ZL0580 with LEDGINs has an additive effect in blocking HIV-1 transcription and reactivation, in both cell lines and primary cells. These findings demonstrate the potential of ZL0580 to enhance the block-and-lock cure strategy.
Collapse
Affiliation(s)
- Eline Pellaers
- Laboratory for Advanced Disease Modelling, Targeted Drug Discovery and Gene Therapy (ADVANTAGE), Herestraat 49, Leuven, Flanders, Belgium
| | - Julie Janssens
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Lore Wils
- Laboratory for Advanced Disease Modelling, Targeted Drug Discovery and Gene Therapy (ADVANTAGE), Herestraat 49, Leuven, Flanders, Belgium
| | - Alexe Denis
- Laboratory for Advanced Disease Modelling, Targeted Drug Discovery and Gene Therapy (ADVANTAGE), Herestraat 49, Leuven, Flanders, Belgium
| | - Anayat Bhat
- Department of Microbiology, Washington University (WashU), Saint Louis, MI, USA
| | - Siska Van Belle
- Laboratory for Advanced Disease Modelling, Targeted Drug Discovery and Gene Therapy (ADVANTAGE), Herestraat 49, Leuven, Flanders, Belgium
| | - Da Feng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Frauke Christ
- Laboratory for Advanced Disease Modelling, Targeted Drug Discovery and Gene Therapy (ADVANTAGE), Herestraat 49, Leuven, Flanders, Belgium
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zeger Debyser
- Laboratory for Advanced Disease Modelling, Targeted Drug Discovery and Gene Therapy (ADVANTAGE), Herestraat 49, Leuven, Flanders, Belgium.
| |
Collapse
|
5
|
Ali H, Wadas J, Bendoumou M, Chen HC, Maiuri P, Dutilleul A, Selberg S, Nestola L, Lalik K, Avettand-Fenoël V, Necsoi C, Marcello A, Kankuri E, Karelson M, De Wit S, Pyrc K, Pasternak AO, Van Lint C, Kula-Pacurar A. Inhibition of ALKBH5 demethylase of m 6A pathway potentiates HIV-1 reactivation from latency. Virol J 2025; 22:124. [PMID: 40296171 PMCID: PMC12039216 DOI: 10.1186/s12985-025-02744-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Current latency-reversing agents (LRAs) employed in the "shock-and-kill" strategy primarily focus on relieving epigenetic and transcriptional blocks to reactivate the latent HIV-1. However, their clinical efficacy is limited, partly due to their inability to fully reverse latency and the lack of LRAs specifically targeting post-transcriptional mechanisms. N6-methyladenosine (m6A) modification in HIV-1 RNA is emerging as an important post-transcriptional regulator of HIV-1 gene expression, yet its role in latency and reactivation remains largely unrecognized. Here, we explored the potential of small chemical compounds targeting the m6A pathway, specifically investigating the inhibition of ALKBH5 and its effect on latent HIV-1 reactivation mediated by the LRA romidepsin. METHODS We used four in vitro cellular models of latency, primary model of CD4+ T cells HIV-1 infection and ex vivo cultures of CD8+-depleted PMBCs from ART-treated HIV+ patients. We measured latent viral reactivation by evaluating the expression of reporter protein GFP by flow cytometry, viral production by CA-p24 ELISA, and viral transcripts by RT-qPCR. CRISPR/Cas9 method was used to deplete ALKBH5. MeRIP and immuno-RNA FISH were used to address the m6A methylation levels on HIV-1 RNA upon ALKBH5 inhibition. RESULTS We showed that ALKBH5 inhibitor 3 (ALKi-3) potentiated romidepsin-mediated viral reactivation in in vitro models of latency, primary model of CD4+ T cells infected with HIV-1 as well as in ex vivo cultures of CD8+-depleted PBMCs from ART-treated HIV+ patients. CRISPR/Cas9-mediated depletion of ALKBH5 mimicked the effects of ALKi-3. ALKi-3 increased levels of m6A-methylated HIV-1 RNA as shown by meRIP and immuno-RNA FISH. CONCLUSION Our study provides a proof-of-concept for the modulation of the m6A pathway in enhancing HIV-1 reactivation. This approach represents a promising adjunct to existing reactivation protocols and provides a concept of "dual-kick", aiming to target transcriptional and post-transcriptional steps in HIV-1 reactivation from latency.
Collapse
Affiliation(s)
- Haider Ali
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Jakub Wadas
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Maryam Bendoumou
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Heng-Chang Chen
- Quantitative Virology Research Group, Population Diagnostics Center, Lukasiewicz Research Network- PORT Polish Center for Technology Development, Wroclaw, Poland
- The Laboratory of Quantitative Virology, Centre for Advanced Materials and Technologies, Warsaw University of Technology, 19 Poleczki St, Warsaw, 02-822, Poland
| | - Paolo Maiuri
- Dept of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Antoine Dutilleul
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Simona Selberg
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Lorena Nestola
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Kamil Lalik
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Veronique Avettand-Fenoël
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
- CHU d'Orléans, Orléans, France
| | - Coca Necsoi
- Service des Maladies Infectieuses, CHU St-Pierre, Université Libre de Bruxelles (ULB), Brussels, 1000, Belgium
| | - Alessandro Marcello
- Laboratory of Molecular Virology, The International Centre of Genetic Engineering and Biotechnology, Trieste, Italy
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Stéphane De Wit
- Service des Maladies Infectieuses, CHU St-Pierre, Université Libre de Bruxelles (ULB), Brussels, 1000, Belgium
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Alexander O Pasternak
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Anna Kula-Pacurar
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
6
|
Bontempo A, Heidari A, Pastore MR, Madonia R, Sadik A, Schweizer M, Cayabyab M. Yoda1, a Piezo1 agonist, induced latent HIV reactivation associated with upregulation of CD3/TCR complex and HLA genes. RESEARCH SQUARE 2025:rs.3.rs-6208371. [PMID: 40297703 PMCID: PMC12036472 DOI: 10.21203/rs.3.rs-6208371/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
There is currently no cure for HIV because of the presence of latent viral reservoirs in people with HIV (PWH) on antiretroviral therapy (ART). Latency-reversing agents (LRAs) that can effectively reactivate and destroy latent HIV are being developed as a possible cure for HIV. Here, we identify Yoda1, a Piezo1 agonist, as a novel LRA. Yoda1 reactivated latent HIV in vitro ACH2 cells and ex vivo PBMCs from an HIV patient on ART. Yoda1 induced infectious virus production and HIV gene expression via Piezo1 activation and calcium signaling. Transcriptomic and proteomic analyses revealed a unique latent HIV reactivation pathway involving T cell activation, upregulation of TCR/CD3 and HLA genes, as well as modulation of host and viral transcription and translation that favors viral gene expression. These findings suggest further testing and development of Yoda1 as an effective LRA to reactivate latent HIV and destroy latent reservoirs for the cure of HIV.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mark Cayabyab
- Nova Southeastern University College of Dental Medicine
| |
Collapse
|
7
|
Gray CN, Ashokkumar M, Janssens DH, Kirchherr JL, Allard B, Hsieh E, Hafer TL, Archin NM, Browne EP, Emerman M. Integrator complex subunit 12 knockout overcomes a transcriptional block to HIV latency reversal. eLife 2025; 13:RP103064. [PMID: 40207620 PMCID: PMC11984954 DOI: 10.7554/elife.103064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
The latent HIV reservoir is a major barrier to HIV cure. Combining latency reversal agents (LRAs) with differing mechanisms of action such as AZD5582, a non-canonical NF-kB activator, and I-BET151, a bromodomain inhibitor is appealing toward inducing HIV-1 reactivation. However, even this LRA combination needs improvement as it is inefficient at activating proviruses in cells of people living with HIV (PLWH). We performed a CRISPR screen in conjunction with AZD5582 & I-BET151 and identified a member of the Integrator complex as a target to improve this LRA combination, specifically Integrator complex subunit 12 (INTS12). Integrator functions as a genome-wide attenuator of transcription that acts on elongation through its RNA cleavage and phosphatase modules. Knockout of INTS12 improved latency reactivation at the transcriptional level and is more specific to the HIV-1 provirus than AZD5582 & I-BET151 treatment alone. We found that INTS12 is present on chromatin at the promoter of HIV and therefore its effect on HIV may be direct. Additionally, we observed more RNAPII in the gene body of HIV only with the combination of INTS12 knockout with AZD5582 & I-BET151, indicating that INTS12 induces a transcriptional elongation block to viral reactivation. Moreover, knockout of INTS12 increased HIV-1 reactivation in CD4 T cells from virally suppressed PLWH ex vivo, and we detected viral RNA in the supernatant from CD4 T cells of all three virally suppressed PLWH tested upon INTS12 knockout, suggesting that INTS12 prevents full-length HIV RNA production in primary T cells. Finally, we found that INTS12 more generally limits the efficacy of a variety of LRAs with different mechanisms of action.
Collapse
Affiliation(s)
- Carley N Gray
- Department of Microbiology, University of WashingtonSeattleUnited States
| | - Manickam Ashokkumar
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel HillChapel HillUnited States
- UNC HIV Cure Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Derek H Janssens
- Division of Basic Sciences, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Jennifer L Kirchherr
- UNC HIV Cure Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Brigitte Allard
- UNC HIV Cure Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Emily Hsieh
- Molecular and Cellular Biology Graduate Program, University of WashingtonSeattleUnited States
| | - Terry L Hafer
- Division of Basic Sciences, Fred Hutchinson Cancer CenterSeattleUnited States
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Nancie M Archin
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel HillChapel HillUnited States
- UNC HIV Cure Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Edward P Browne
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel HillChapel HillUnited States
- UNC HIV Cure Center, University of North Carolina at Chapel HillChapel HillUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Michael Emerman
- Division of Basic Sciences, Fred Hutchinson Cancer CenterSeattleUnited States
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
| |
Collapse
|
8
|
Chang LC, Yin MT, Laird GM, Ritter KD, Shah JG, Debnath AK. A First-in-Class Dual Degrader of Bcl-2/Bcl-xL Reverses HIV Latency and Minimizes Ex Vivo Reservoirs from Patients. Int J Mol Sci 2025; 26:2772. [PMID: 40141414 PMCID: PMC11942780 DOI: 10.3390/ijms26062772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
The persistence of latent HIV-1 proviruses in CD4+ T cells is a major obstacle to curing HIV. The "shock and kill" strategy involves reversing latency with latency-reversing agents (LRAs) and selectively inducing cell death in infected cells. However, current LRAs have shown limited efficacy in eliminating the ex vivo HIV reservoir and thus failed in clinical study. In this study, we repurposed PZ703b, a pro-apoptotic protein degrader initially developed for anti-leukemia therapy, to target HIV eradication. PZ703b induced the degradation of Bcl-2 and Bcl-xL, activating the non-canonical NF-kB pathway and caspases cascade, resulting in latency reversal and the selective apoptosis of infected cells. The treatment of ex vivo CD4+ T cells from ART-suppressed HIV-1 patients led to approximately a 50% reduction in the replication-competent reservoir. While this result does not reach the threshold required for a complete cure, it demonstrates the potential of a dual degrader of Bcl-2/Bcl-xL in reversing HIV latency and inducing selective cell death. Our study provides a proof-of-concept for using dual degraders of Bcl-2/Bcl-xL as a novel category of LRAs in therapeutic strategies aimed at reducing HIV reservoirs. This approach may pave the way for the further exploration of targeted interventions to eliminate the HIV-inducible reservoir.
Collapse
Affiliation(s)
- Lin-Chun Chang
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Michael T. Yin
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.T.Y.)
| | - Gregory M. Laird
- Accelevir Diagnostics, Baltimore, MD 21202, USA; (G.M.L.); (K.D.R.)
| | | | - Jayesh G. Shah
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.T.Y.)
| | - Asim K. Debnath
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| |
Collapse
|
9
|
Wadas J, Ali H, Osiecka A, Dorman A, Pyrc K, Kula-Pacurar A. Development and characterization of a double-fluorescent HIV-1 reporter cellular model to tackle the Rev-dependent export pathway. Microbiol Spectr 2025; 13:e0190324. [PMID: 39902983 PMCID: PMC11878058 DOI: 10.1128/spectrum.01903-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/09/2025] [Indexed: 02/06/2025] Open
Abstract
The Rev-dependent nuclear export of unspliced and singly-spliced transcripts of human immunodeficiency virus type 1 (HIV-1) constitutes a critical yet poorly characterized post-transcriptional event essential for effective viral replication. In this study, we engineered a dual-fluorescent HIV-1-based cellular reporter system to elucidate the mechanisms underpinning Rev-dependent export. By generating multiple stably integrated inducible cellular clones, we ensured the expression of two distinct fluorescent proteins, mKO2, and ECFP, from unspliced (Rev dependent) and multiply spliced (Rev independent) HIV-1 transcripts, respectively. Utilizing flow cytometry, we performed quantitative analyses of dual-fluorescent cell populations. The developed tool enables precise assessment of the Rev-dependent export, and we validated it using known inhibitors of this pathway (leptomycin D), as well as targeted depletion of MATR3, an essential cofactor of Rev, and CRNKL1, a repressor of unspliced HIV-1 RNA export.IMPORTANCEThe developed dual-fluorescent reporter system represents a powerful and handy tool for the identification and characterization of novel molecular players involved in the Rev-dependent export pathway. This system not only holds promise for advancing our understanding of human immunodeficiency virus type 1 (HIV-1) biology but also serves as an invaluable platform for high-throughput drug screening aimed at targeting post-transcriptional HIV-1 RNA processes, particularly nuclear export. Consequently, this study offers significant implications for the development of novel therapeutic strategies to eradicate the virus.
Collapse
Affiliation(s)
- Jakub Wadas
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Haider Ali
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Aleksandra Osiecka
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Dorman
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Krzysztof Pyrc
- Laboratory of Virology–Virogenetics, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Kula-Pacurar
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
10
|
Cai J, Zhang J, Wang K, Dai Z, Hu Z, Dong Y, Peng Z. Evaluating the long-term effects of combination antiretroviral therapy of HIV infection: a modeling study. J Math Biol 2025; 90:36. [PMID: 40025191 PMCID: PMC11872777 DOI: 10.1007/s00285-025-02196-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/04/2025] [Accepted: 02/08/2025] [Indexed: 03/04/2025]
Abstract
Current HIV/AIDS treatments effectively reduce viral loads to undetectable levels as measured by conventional clinical assays, but immune recovery remains highly variable among patients. To assess the long-term treatment efficacy, we propose a mathematical model that incorporates latently infected CD4+ T cells and the homeostatic proliferation of CD4+ T cells. We investigate the dynamics of this model both theoretically and numerically, demonstrating that homeostatic proliferation can induce bistability, which implies that steady-state CD4+ T cell count is sensitively affected by initial conditions. The model exhibits rich dynamics, including saddle node bifurcations, Hopf bifurcations, and saddle node bifurcations related to periodic orbits. The interplay between homeostatic proliferation and latent HIV infection significantly influences the model's dynamic behavior. Additionally, we integrate combination antiretroviral therapy (cART) into the model and fit the revised model to clinical data on long-term CD4+ T cell counts before and after treatment. Quantitative analysis estimates the effects of long-term cART, revealing an increasing sensitivity of steady-state CD4+ T cell count to drug efficacy. Correlation analysis indicates that the heightened activation of latently infected cells helps enhance treatment efficacy. These findings underscore the critical roles of CD4+ T cell homeostatic proliferation and latently infected cell production in HIV persistence despite treatment, providing valuable insights for understanding disease progression and developing more effective therapies, potentially towards eradication.
Collapse
Affiliation(s)
- Jing Cai
- School of Public Health, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Jun Zhang
- School of Mathematics and Statistics, and Key Laboratory of Nonlinear Analysis and Applications (Ministry of Education), Central China Normal University, 152 Luoyu Road, Wuhan, 430079, Hubei, China
| | - Kai Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, No. 2 East Cherry Garden Street, Beijing, 100029, China
| | - Zhixiang Dai
- School of Public Health, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Zhiliang Hu
- Nanjing Infectious Disease Center, The Second Hospital of Nanjing, Tangshan Street, Nanjing, 211113, Jiangsu, China
| | - Yueping Dong
- School of Mathematics and Statistics, and Key Laboratory of Nonlinear Analysis and Applications (Ministry of Education), Central China Normal University, 152 Luoyu Road, Wuhan, 430079, Hubei, China.
| | - Zhihang Peng
- School of Public Health, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China.
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Beijing, 102206, China.
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Beijing, 102206, China.
| |
Collapse
|
11
|
Dorman A, Bendoumou M, Valaitienė A, Wadas J, Ali H, Dutilleul A, Maiuri P, Nestola L, Bociaga-Jasik M, Mchantaf G, Necsoi C, De Wit S, Avettand-Fenoël V, Marcello A, Pyrc K, Pasternak AO, Van Lint C, Kula-Pacurar A. Nuclear retention of unspliced HIV-1 RNA as a reversible post-transcriptional block in latency. Nat Commun 2025; 16:2078. [PMID: 40021667 PMCID: PMC11871326 DOI: 10.1038/s41467-025-57290-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
HIV-1 latency is mainly characterized at transcriptional level, and little is known about post-transcriptional mechanisms and their contribution to reactivation. The viral protein Rev controls the nucleocytoplasmic export of unspliced and singly-spliced RNA that is central to proviral replication-competence and is therefore a prerequisite for efficient viral reactivation during the "shock-and-kill" cure therapy. Here we show that during infection and reactivation, unspliced HIV-1 RNA is a subject to complex and dynamic regulation by the Rev cofactor MATR3 and the MTR4 cofactor of the nuclear exosome. MATR3 and MTR4 coexist in the same ribonucleoprotein complex functioning to either maintain or degrade the RNA, respectively, with Rev orchestrating this regulatory switch. Moreover, we provide evidence of nuclear retention of unspliced HIV-1 RNA in ex vivo cultures from 22 ART-treated people with HIV, highlighting a reversible post-transcriptional block to viral RNA nucleocytoplasmic export that is relevant to the design of curative interventions.
Collapse
Affiliation(s)
- Agnieszka Dorman
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Maryam Bendoumou
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Aurelija Valaitienė
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jakub Wadas
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Haider Ali
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Antoine Dutilleul
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Paolo Maiuri
- Dept of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Lorena Nestola
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Monika Bociaga-Jasik
- Department of Infectious Diseases and Tropical Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Gilbert Mchantaf
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
- CHU d'Orléans, Orléans, France
- Université d'Orléans, LI²RSO, Orléans, France
| | - Coca Necsoi
- Service des Maladies Infectieuses, CHU St-Pierre, Université Libre de Bruxelles (ULB), Brussels, 1000, Belgium
| | - Stéphane De Wit
- Service des Maladies Infectieuses, CHU St-Pierre, Université Libre de Bruxelles (ULB), Brussels, 1000, Belgium
| | - Véronique Avettand-Fenoël
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
- CHU d'Orléans, Orléans, France
- Université d'Orléans, LI²RSO, Orléans, France
| | - Alessandro Marcello
- Laboratory of Molecular Virology, The International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Alexander O Pasternak
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium.
| | - Anna Kula-Pacurar
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
12
|
Tioka L, Diez RC, Sönnerborg A, van de Klundert MAA. Latency Reversing Agents and the Road to an HIV Cure. Pathogens 2025; 14:232. [PMID: 40137717 PMCID: PMC11944434 DOI: 10.3390/pathogens14030232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
HIV-1 infection cannot be cured due to the presence of HIV-1 latently infected cells. These cells do not produce the virus, but they can resume virus production at any time in the absence of antiretroviral therapy. Therefore, people living with HIV (PLWH) need to take lifelong therapy. Strategies have been coined to eradicate the viral reservoir by reactivating HIV-1 latently infected cells and subsequently killing them. Various latency reversing agents (LRAs) that can reactivate HIV-1 in vitro and ex vivo have been identified. The most potent LRAs also strongly activate T cells and therefore cannot be applied in vivo. Many LRAs that reactivate HIV in the absence of general T cell activation have been identified and have been tested in clinical trials. Although some LRAs could reduce the reservoir size in clinical trials, so far, they have failed to eradicate the reservoir. More recently, immune modulators have been applied in PLWH, and the first results seem to indicate that these may reduce the reservoir and possibly improve immunological control after therapy interruption. Potentially, combinations of LRAs and immune modulators could reduce the reservoir size, and in the future, immunological control may enable PLWH to live without developing HIV-related disease in the absence of therapy.
Collapse
Affiliation(s)
- Louis Tioka
- Faculty of Medicine, Erlangen-Nürnberg, Friedrich-Alexander-Universität, 91054 Erlangen, Germany;
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden; (R.C.D.); (A.S.)
| | - Rafael Ceña Diez
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden; (R.C.D.); (A.S.)
| | - Anders Sönnerborg
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden; (R.C.D.); (A.S.)
- Department of Infectious Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Maarten A. A. van de Klundert
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden; (R.C.D.); (A.S.)
| |
Collapse
|
13
|
Council OD, Tyers L, Moeser M, Sondgeroth A, Spielvogel E, Richardson BD, Doolabh D, Zhou S, Emery A, Archin NM, Shook-Sa B, Margolis DM, Abdool Karim SS, Kosakovsky Pond S, Garrett N, Abrahams MR, Joseph SB, Williamson C, Swanstrom R. The persistent pool of HIV-1-infected cells is formed episodically during untreated infection. J Virol 2025; 99:e0097924. [PMID: 39723838 PMCID: PMC11852786 DOI: 10.1128/jvi.00979-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Previous studies have shown that the majority of long-lived cells harboring persistent HIV-1 proviral genomes originates from viruses circulating in the year prior to antiretroviral therapy (ART) initiation, but a smaller proportion originates from viruses circulating much earlier in untreated infection. These observations suggest that discrete biological factors influence the entry and persistence of viruses into the persistent proviral pool, and there may be periods earlier in untreated infection with increased seeding. Therefore, we examined the timing of formation of the long-lived pool of infected cells that persists during ART in seven women (after a median of 5.1 years of suppressive ART) by comparing the phylogenetic distance between unique 3' half genome on-ART proviral sequences and longitudinally sampled pre-ART viral RNA sequences, focusing on the period >1 year prior to ART initiation (i.e., the "early" proviral pool). We constructed models of continuous entry into the persistent proviral pool prior to ART initiation and analyzed the fit of our experimentally derived data to these models. We found that the pattern of persistent proviral pool formation in five of seven participants is incongruent with a model of continuous entry, implying that persistent proviral pool formation can occur episodically during untreated infection. Notably, increased entry into the persistent proviral pool was not universally observed during acute infection, and the timing of enhanced early entry differed across the participants.IMPORTANCECells harboring HIV-1 proviruses that persist on antiretroviral therapy (ART) constitute the main barrier to an HIV-1 cure. Recent work has elucidated that the majority of persisting proviruses harbor HIV-1 variants circulating near the time of ART initiation, whether the proviruses are intact or defective, though a portion forms earlier in untreated infection. We examined the formation of the "early-forming" persistent proviral pool and found that in 5/7 participants, persistent proviral pool formation was episodic, rather than continuous, suggesting that there are host/biological factors that periodically enhance the formation of the persistent proviral pool. Further characterization of these factors will aid in the development of methods to abrogate their effect, thereby reducing the size of the persistent proviral pool.
Collapse
Affiliation(s)
- Olivia D. Council
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lynn Tyers
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch, Western Cape, South Africa
| | - Matthew Moeser
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Amy Sondgeroth
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ean Spielvogel
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brian D. Richardson
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Deelan Doolabh
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch, Western Cape, South Africa
| | - Shuntai Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ann Emery
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nancie M. Archin
- UNC HIV Cure Center and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Bonnie Shook-Sa
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David M. Margolis
- UNC HIV Cure Center and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Salim S. Abdool Karim
- Center for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Sergei Kosakovsky Pond
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Nigel Garrett
- Center for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
- Division of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Melissa-Rose Abrahams
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch, Western Cape, South Africa
- Center for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Sarah B. Joseph
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Carolyn Williamson
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch, Western Cape, South Africa
- Center for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
- National Health Laboratory Services of South Africa, Johannesburg, Gauteng, South Africa
| | - Ronald Swanstrom
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
14
|
Gray CN, Ashokkumar M, Janssens DH, Kirchherr J, Allard B, Hsieh E, Hafer TL, Archin NM, Browne EP, Emerman M. Integrator complex subunit 12 knockout overcomes a transcriptional block to HIV latency reversal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.30.610517. [PMID: 39257755 PMCID: PMC11383676 DOI: 10.1101/2024.08.30.610517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The latent HIV reservoir is a major barrier to HIV cure. Combining latency reversal agents (LRAs) with differing mechanisms of action such as AZD5582, a non-canonical NF-kB activator, and I-BET151, a bromodomain inhibitor is appealing towards inducing HIV-1 reactivation. However, even this LRA combination needs improvement as it is inefficient at activating proviruses in cells from people living with HIV (PLWH). We performed a CRISPR screen in conjunction with AZD5582 & I-BET151 and identified a member of the Integrator complex as a target to improve this LRA combination, specifically Integrator complex subunit 12 (INTS12). Integrator functions as a genome-wide attenuator of transcription that acts on elongation through its RNA cleavage and phosphatase modules. Knockout of INTS12 improved latency reactivation at the transcriptional level and is more specific to the HIV-1 provirus than AZD5582 & I-BET151 treatment alone. We found that INTS12 is present on chromatin at the promoter of HIV and therefore its effect on HIV may be direct. Additionally, we observed more RNAPII in the gene body of HIV only with the combination of INTS12 knockout with AZD5582 & I-BET151, indicating that INTS12 induces a transcriptional elongation block to viral reactivation. Moreover, knockout of INTS12 increased HIV-1 reactivation in CD4 T cells from virally suppressed PLWH ex vivo, and we detected viral RNA in the supernatant from CD4 T cells of all three virally suppressed PLWH tested upon INTS12 knockout suggesting that INTS12 prevents full-length HIV RNA production in primary T cells. Finally, we found that INTS12 more generally limits the efficacy of a variety of LRAs with different mechanisms of action.
Collapse
Affiliation(s)
- Carley N. Gray
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Manickam Ashokkumar
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Derek H. Janssens
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jennifer Kirchherr
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brigitte Allard
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily Hsieh
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Terry L. Hafer
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nancie M. Archin
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edward P. Browne
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael Emerman
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
15
|
Hajikhezri Z, Zygouras I, Sönnerborg A, van Domselaar R. Pan-caspase inhibitors induce secretion of HIV-1 latency reversal agent lymphotoxin-alpha from cytokine-primed NK cells. Cell Death Discov 2025; 11:44. [PMID: 39905001 PMCID: PMC11794648 DOI: 10.1038/s41420-025-02330-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
The persistence of HIV-1 latency reservoirs in CD4+ T cells is a significant obstacle for curing HIV-1. Shock-and-kill strategies, which aim to reactivate latent HIV-1 followed by cytotoxic clearance, have shown limited success in vivo due to insufficient efficacy of latency reversal agents (LRAs) and off-target effects. Natural killer (NK) cells, with their ability to mediate cytotoxicity independent of antigen specificity, offer a promising avenue for enhancing the shock-and-kill approach. Previously, we observed that pan-caspase inhibitors induce NK cells to secrete an LRA in vitro. Here, we aimed to identify this LRA using a targeted proteomic approach. We identified lymphotoxin-α (LTα) as the key LRA secreted by NK cells following pan-caspase inhibitor treatment. LTα was shown to significantly induce HIV-1 LTR promoter activity, a hallmark of viral reactivation. Neutralization of LTα effectively abolished the observed LRA activity, confirming its central role. Moreover, cytokine-primed but not resting human primary NK cells exhibited LRA activity that could be neutralized with LTα neutralizing antibodies. Finally, pan-caspase inhibitor treatment did not decrease the ability of the cytokine-primed NK cells to kill target cells. These findings demonstrate that cytokine-primed NK cells, through LTα secretion, can effectively reactivate latent HIV-1 following pan-caspase inhibitor treatment, without compromising NK cell cytotoxicity. This highlights a potential enhancement strategy utilizing NK cells for shock-and-kill approaches in HIV-1 cure research.
Collapse
Affiliation(s)
- Zamaneh Hajikhezri
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Zygouras
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Robert van Domselaar
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
16
|
Wang J, Xiao N, Zhu Z, Qiao H, Zhao F, Zhang L, Gou J, Lu M, He Y, Lu H, Li Q. Comparing acute versus AIDS ART initiation on HIV-1 integration sites and clonal expansion. Signal Transduct Target Ther 2025; 10:23. [PMID: 39788938 PMCID: PMC11718275 DOI: 10.1038/s41392-024-02113-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
Early antiretroviral therapy (ART) initiation is known to limit the establishment of the HIV reservoir, with studies suggesting benefits such as a reduced number of infected cells and a smaller latent reservoir. However, the long-term impact of early ART initiation on the dynamics of the infected cell pool remains unclear, and clinical evidence directly comparing proviral integration site counts between early and late ART initiation is limited. In this study, we used Linear Target Amplification-PCR (LTA-PCR) and Next Generation Sequencing to compare unique integration site (UIS) clonal counts between individuals who initiated ART during acute HIV infection stage (Acute-ART group) and those in the AIDS stage (AIDS-ART group). Our analysis revealed distinct clonal distribution patterns, with greater UIS heterogeneity in Acute-ART group and more homogeneity in AIDS-ART group. Monoclonal UIS accumulation, predominantly in-gene regions, was influenced by ART timing and duration, with early treatment delaying this process. Host cell genes integrated by HIV provirus as monoclonal types were enriched in cell cycle and lymphocyte activation pathways. Tumor suppressor genes (TSGs) were more frequently integrated as monoclonal types in AIDS-ART group, suggesting potential risk factors. Overall, we introduced a sequencing method to assess provirus size in human peripheral blood and identified the widespread presence of monoclonal distribution of UIS in AIDS-ART group after long-term treatment. The early intervention helps slow the progress of clonal expansion of infected cells, reducing the formation of stable and persistent reservoirs, and ultimately posing fewer barriers to achieving a functional cure.
Collapse
Affiliation(s)
- Jun Wang
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
- Clinical Research Center, The Fifth People's Hospital of Wuxi, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Nan Xiao
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
| | - Zhengnong Zhu
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
| | - Haiyan Qiao
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
| | - Fang Zhao
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
| | - Lukun Zhang
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
| | - Jizhou Gou
- Department of Pathology, Shenzhen Third People's Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Mengji Lu
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
- Institute of virology, Essen University Hospital, University of Duisburg-Essen, Essen, 45147, Germany
| | - Yun He
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China.
| | - Hongzhou Lu
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China.
| | - Qian Li
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China.
| |
Collapse
|
17
|
Chhabra L, Pandey RK, Kumar R, Sundar S, Mehrotra S. Navigating the Roadblocks: Progress and Challenges in Cell-Based Therapies for Human Immunodeficiency Virus. J Cell Biochem 2025; 126:e30669. [PMID: 39485037 DOI: 10.1002/jcb.30669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024]
Abstract
Cell-based therapies represent a major advancement in the treatment and management of HIV/AIDS, with a goal to overcome the limitations of traditional antiretroviral therapy (ART). These innovative approaches not only promise a functional cure by reconstructing the immune landscape but also address the persistent viral reservoirs. For example, stem cell therapies have emerged from the foundational success of allogeneic hematopoietic stem cell transplantation in curing HIV infection in a limited number of cases. B cell therapies make use of genetically modified B cells constitutively expressing broadly neutralizing antibodies (bNAbs) against target viral particles and infected cells. Adoptive cell transfer (ACT), including TCR-T therapy, CAR-T cells, NK-CAR cells, and DC-based therapy, is adapted from cancer immunotherapy and repurposed for HIV eradication. In this review, we summarize the mechanisms through which these engineered cells recognize and destroy HIV-infected cells, the modification strategies, and their role in sustaining remission in the absence of ART. The review also addresses the challenges to cell-based therapies against HIV and discusses the recent advancements aimed at overcoming them.
Collapse
Affiliation(s)
- Lakshay Chhabra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
18
|
Hetrick B, Siddiqui S, Spear M, Guo J, Liang H, Fu Y, Yang Z, Doyle-Meyers L, Pahar B, Veazey RS, Dufour J, Andalibi A, Ling B, Wu Y. Suppression of viral rebound by a Rev-dependent lentiviral particle in SIV-infected rhesus macaques. Gene Ther 2025; 32:16-24. [PMID: 39025983 PMCID: PMC11785524 DOI: 10.1038/s41434-024-00467-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Persistence of human immunodeficiency virus (HIV) reservoirs prevents viral eradication, and consequently HIV-infected patients require lifetime treatment with antiretroviral therapy (ART) [1-5]. Currently, there are no effective therapeutics to prevent HIV rebound upon ART cessation. Here we describe an HIV/SIV Rev-dependent lentiviral particle that can be administered to inhibit viral rebound [6-9]. Using simian immunodeficiency virus (SIV)-infected rhesus macaques as a model, we demonstrate that the administration of pre-assembled SIV Rev-dependent lentiviral particles into SIVmac239-infected Indian rhesus macaques can lead to reduction of viral rebound upon ART termination. One of the injected animals, KC50, controlled plasma and CNS viremia to an undetectable level most of the time for over two years after ART termination. Surprisingly, detailed molecular and immunological characterization revealed that viremia control was concomitant with the induction of neutralizing antibodies (nAbs) following the administration of the Rev-dependent vectors. This study emphasizes the importance of neutralizing antibodies (nAbs) for viremia control [10-15], and also provides proof of concept that the Rev-dependent vector can be used to target viral reservoirs, including the CNS reservoirs, in vivo. However, future large-scale in vivo studies are needed to understand the potential mechanisms of viremia control induced by the Rev-dependent vector.
Collapse
Affiliation(s)
- Brian Hetrick
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Summer Siddiqui
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Mark Spear
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Jia Guo
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Huizhi Liang
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Yajing Fu
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Zhijun Yang
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Lara Doyle-Meyers
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Bapi Pahar
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Jason Dufour
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Ali Andalibi
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Binhua Ling
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W Military Dr., San Antonio, TX, 78227, USA
| | - Yuntao Wu
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA.
| |
Collapse
|
19
|
Dong B, Qi W, Chen Y, Zhang Y, Gu S, Zhao J, Zhou Q, Shen J, Xie L. Stabilized Carbon Radical-Mediated Assembly of Arylthianthrenium Salts, Alkenes and Amino Acid/Peptide Derivatives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411579. [PMID: 39573977 PMCID: PMC11727398 DOI: 10.1002/advs.202411579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/07/2024] [Indexed: 01/14/2025]
Abstract
Efficiently assembling amino acids and peptides with bioactive molecules facilitates the modular and streamlined synthesis of a diverse library of peptide-related compounds. Particularly notable is their application in pharmaceutical development, leveraging site-selective late-stage functionalization. Here, a visible light-induced three-component reaction involving arylthianthrenium salts, amino acid/peptide derivatives, and alkenes are introduced. This approach utilizes captodatively-stabilized carbon radicals to enable radical-radical C─C coupling, effectively constructing complex bioactive molecules. This method offers a promising alternative route for modular synthesis of peptide-derived bio-relevant compounds.
Collapse
Affiliation(s)
- Bo Dong
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
| | - Weiguan Qi
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
| | - Yifeng Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
| | - Yufei Zhang
- State Key Laboratory of Natural MedicinesDepartment of Organic ChemistryChina Pharmaceutical UniversityNanjing210009P. R. China
| | - Shiyu Gu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
| | - Jianlin Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
| | - Qingfa Zhou
- State Key Laboratory of Natural MedicinesDepartment of Organic ChemistryChina Pharmaceutical UniversityNanjing210009P. R. China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
- Jiangsu Engineering Research Center of Interfacial ChemistryNanjing UniversityNanjing210023P. R. China
| | - Lan‐Gui Xie
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, School of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023P. R. China
| |
Collapse
|
20
|
Li Y, Luo H, Pang H, Qin B. Epigenetic Targeting for Controlling Persistent Neurotropic Infections Caused by Borna Virus and HIV. Rev Med Virol 2025; 35:e70000. [PMID: 39643925 DOI: 10.1002/rmv.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 12/09/2024]
Abstract
Long-lasting persistence within infected cells is a major challenge for viral pathogens, as it necessitates an exact regulation of viral replication to reduce viral cytopathic effects. This is particularly challenging for viruses that persistently infect cells with limited renewal capabilities, such as neurons. Accordingly, neurotropic viruses have evolved various specific mechanisms to promote a long-lasting persistent infection in the host cells without inducing an exacerbated cytopathic effect. Borna disease virus (BDV) and Human immunodeficiency virus (HIV) are two neurotropic RNA viruses that, in contrast to other RNA viruses, can establish long-lasting intranuclear infections within the nervous system. These viruses interact with different cellular processes such as epigenetic modifications to develop a successful persistence infection. Studies show that cellular epigenetic mechanisms play a significant role in the pathogenesis of BDV and HIV and their neurological disorders. Hence, targeting these mechanisms by epigenetic modulator agents can be regarded as a novel therapeutic strategy to manage BDV- and HIV-associated neurological diseases. This review provides an overview of different epigenetic modulator compounds as a potential therapeutic target for controlling persistent neurotropic intranuclear infections caused by BDV and HIV.
Collapse
Affiliation(s)
- Yadi Li
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huating Luo
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Pang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Qin
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Moskovljevic M, Dragoni F, Board NL, Wu F, Lai J, Zhang H, White JR, Hoh R, Lynn K, Tebas P, Mounzer K, Deeks SG, Montaner LJ, Siliciano JD, Siliciano RF, Simonetti FR. Cognate antigen engagement induces HIV-1 expression in latently infected CD4 + T cells from people on long-term antiretroviral therapy. Immunity 2024; 57:2928-2944.e6. [PMID: 39612916 PMCID: PMC11896817 DOI: 10.1016/j.immuni.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Despite antiretroviral therapy (ART), HIV-1 persists in latently infected CD4+ T cells, preventing a cure. Antigens drive the proliferation of infected cells, precluding latent reservoir decay. However, the relationship between antigen recognition and HIV-1 gene expression is poorly understood because most studies of latency reversal use agents that induce non-specific global T cell activation. Here, we isolated rare CD4+ T cells responding to cytomegalovirus (CMV) or HIV-1 Gag antigens from people living with HIV-1 on long-term ART and assessed T cell activation and HIV-1 RNA expression upon coculture with autologous dendritic cells (DCs) presenting cognate antigens. Presentation of cognate antigens ex vivo induced broad T cell activation (median 42-fold increase in CD154+CD69+ cells) and significantly increased HIV-1 transcription (median 4-fold), mostly through the induction of rare cells with higher viral expression. Thus, despite low proviral inducibility, antigen recognition can promote HIV-1 expression, potentially contributing to spontaneous reservoir activity and viral rebound upon ART interruption.
Collapse
Affiliation(s)
- Milica Moskovljevic
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Filippo Dragoni
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nathan L Board
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Fengting Wu
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jun Lai
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Rebecca Hoh
- Division of HIV, School of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Kenneth Lynn
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pablo Tebas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karam Mounzer
- Jonathan Lax Treatment Center, Philadelphia FIGHT, Philadelphia, PA 19107, USA
| | - Steven G Deeks
- Division of HIV, School of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | | | - Janet D Siliciano
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert F Siliciano
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Baltimore, MD 21205, USA.
| | - Francesco R Simonetti
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
22
|
Maikoo S, Palstra RJ, Dong KL, Mahmoudi T, Ndung'u T, Madlala P. Development of a latency model for HIV-1 subtype C and the impact of long terminal repeat element genetic variation on latency reversal. J Virus Erad 2024; 10:100575. [PMID: 39811575 PMCID: PMC11730875 DOI: 10.1016/j.jve.2024.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Sub-Saharan Africa accounts for almost 70 % of people living with HIV (PLWH) worldwide, with the greatest numbers centred in South Africa where 98 % of infections are caused by subtype C (HIV-1C). However, HIV-1 subtype B (HIV-1B), prevalent in Europe and North America, has been the focus of most cure research and testing despite making up only 12 % of HIV-1 infections globally. Development of latency models for non-subtype B viruses is a necessary step to address this disproportionate focus. Furthermore, the impact of genetic variation between viral subtypes, specifically within the long terminal repeat (LTR) element of the viral transcriptional promoter on latency reversal, remains unclear. To address this scientific gap, we constructed a minimal genome retroviral vector expressing HIV-1C consensus transactivator of transcription protein (Tat) and green fluorescent protein (GFP) under the control of either HIV-1C consensus LTR (C731CC) or the transmitted/founder (T/F) LTRs derived from PLWH (CT/F731CC), produced corresponding LTR pseudotyped viruses using a vesicular stomatitis virus (VSV-G) pseudotyped Envelope vector and the pCMVΔR8.91 packaging vector containing HIV-1 accessory and rev genes. Viruses produced in this way were used to infect Jurkat E6 and primary CD4+ T cells in vitro. By enriching for latently infected cells, and treating them with different latency reversing agents, we developed an HIV-1C latency model that demonstrated that the HIV-1C consensus LTR has lower reactivation potential compared to its HIV-1B counterpart. Furthermore, HIV-1C T/F LTR pseudotyped proviral genetic variants exhibited a heterogenous reactivation response which was modulated by host cell (genetic) variation. Our data suggests that genetic variation both within and between HIV-1 subtypes influences latency reversal. Future studies should investigate the specific role of variation in host cellular environment on reactivation differences.
Collapse
Affiliation(s)
- Shreyal Maikoo
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Robert-Jan Palstra
- Department of Biochemistry, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, the Netherlands
- Department of Pathology, Erasmus University Medical Center, the Netherlands
- Department of Urology, Erasmus University Medical Center, the Netherlands
| | - Krista L. Dong
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital, Infectious Disease Division, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, the Netherlands
- Department of Pathology, Erasmus University Medical Center, the Netherlands
- Department of Urology, Erasmus University Medical Center, the Netherlands
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
- Africa Health Research Institute, Durban, KwaZulu-Natal, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Paradise Madlala
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
23
|
Ngo MH, Pankrac J, Ho RCY, Ndashimye E, Pawa R, Ceccacci R, Biru T, Olabode AS, Klein K, Li Y, Kovacs C, Assad R, Jacobson JM, Canaday DH, Tomusange S, Jamiru S, Anok A, Kityamuweesi T, Buule P, Galiwango RM, Reynolds SJ, Quinn TC, Redd AD, Prodger JL, Mann JFS, Arts EJ. Effective and targeted latency reversal in CD4 + T cells from individuals on long term combined antiretroviral therapy initiated during chronic HIV-1 infection. Emerg Microbes Infect 2024; 13:2327371. [PMID: 38444369 PMCID: PMC10967673 DOI: 10.1080/22221751.2024.2327371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
To date, an affordable, effective treatment for an HIV-1 cure remains only a concept with most "latency reversal" agents (LRAs) lacking specificity for the latent HIV-1 reservoir and failing in early clinical trials. We assessed HIV-1 latency reversal using a multivalent HIV-1-derived virus-like particle (HLP) to treat samples from 32 people living with HIV-1 (PLWH) in Uganda, US and Canada who initiated combined antiretroviral therapy (cART) during chronic infection. Even after 5-20 years on stable cART, HLP could target CD4+ T cells harbouring latent HIV-1 reservoir resulting in 100-fold more HIV-1 release into culture supernatant than by common recall antigens, and 1000-fold more than by chemotherapeutic LRAs. HLP induced release of a divergent and replication-competent HIV-1 population from PLWH on cART. These findings suggest HLP provides a targeted approach to reactivate the majority of latent HIV-1 proviruses among individuals infected with HIV-1.
Collapse
Affiliation(s)
- Minh Ha Ngo
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- College of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Joshua Pankrac
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Ryan C. Y. Ho
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Emmanuel Ndashimye
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Rahul Pawa
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Renata Ceccacci
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Tsigereda Biru
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Abayomi S. Olabode
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Katja Klein
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | - Yue Li
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Colin Kovacs
- Maple Leaf Medical Clinic and Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Canada
| | - Robert Assad
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey M. Jacobson
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David H. Canaday
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Aggrey Anok
- Rakai Health Sciences Program, Kalisizo, Uganda
| | | | - Paul Buule
- Rakai Health Sciences Program, Kalisizo, Uganda
| | | | - Steven J. Reynolds
- Rakai Health Sciences Program, Kalisizo, Uganda
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas C. Quinn
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew D. Redd
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jessica L. Prodger
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Jamie F. S. Mann
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | - Eric J. Arts
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| |
Collapse
|
24
|
Lilie T, Bouzy J, Asundi A, Taylor J, Roche S, Olson A, Coxen K, Corry H, Jordan H, Clayton K, Lin N, Tsibris A. HIV-1 latency reversal agent boosting is not limited by opioid use. JCI Insight 2024; 9:e185480. [PMID: 39470739 PMCID: PMC11601940 DOI: 10.1172/jci.insight.185480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/04/2024] [Indexed: 11/01/2024] Open
Abstract
Opioid use may affect the HIV-1 reservoir and its reversal from latency. We studied 47 virally suppressed people with HIV (PWH) and observed that lower concentration of HIV-1 latency reversal agents (LRAs), used with small molecules that did not reverse latency, synergistically increased the magnitude of HIV-1 reactivation ex vivo, regardless of opioid use. This LRA boosting, which combined a second mitochondria-derived activator of caspases mimetic or low-dose PKC agonist with histone deacetylase inhibitors, generated more unspliced HIV-1 transcription than PMA with ionomycin (PMAi), the maximal known HIV-1 reactivator. LRA boosting associated with greater histone acetylation, modulated surface activation-induced markers, and altered T cell production of TNF-α, IL-2, and IFN-γ. HIV-1 reservoirs in PWH contained unspliced and polyadenylated virus mRNA, the ratios of which were greater in resting than total CD4+ T cells and corrected to 1:1 with PMAi exposure. We characterized treated suppressed HIV-1 infection as a period of inefficient, not absent, virus transcription. Multiply spliced HIV-1 transcripts and virion production did not consistently increase with LRA boosting, suggesting the presence of a persistent posttranscriptional block. LRA boosting can be leveraged to probe mechanisms of an effective cellular HIV-1 latency reversal program.
Collapse
Affiliation(s)
- Tyler Lilie
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Bouzy
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Archana Asundi
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jessica Taylor
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Grayken Center for Addiction, Boston Medical Center, Boston, Massachusetts, USA
| | - Samantha Roche
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Alex Olson
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kendyll Coxen
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Heather Corry
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hannah Jordan
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kiera Clayton
- Department of Pathology, University of Massachusetts T.H. Chan School of Medicine, Worcester, Massachusetts, USA
| | - Nina Lin
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Athe Tsibris
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Barbehenn A, Shi L, Shao J, Hoh R, Hartig HM, Pae V, Sarvadhavabhatla S, Donaire S, Sheikhzadeh C, Milush J, Laird GM, Mathias M, Ritter K, Peluso MJ, Martin J, Hecht F, Pilcher C, Cohen SE, Buchbinder S, Havlir D, Gandhi M, Henrich TJ, Hatano H, Wang J, Deeks SG, Lee SA. Rapid biphasic decay of intact and defective HIV DNA reservoir during acute treated HIV disease. Nat Commun 2024; 15:9966. [PMID: 39557853 PMCID: PMC11574060 DOI: 10.1038/s41467-024-54116-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
Despite antiretroviral therapy (ART), HIV persists in latently-infected cells (the HIV reservoir) which decay slowly over time. Here, leveraging >500 longitudinal samples from 67 people living with HIV (PLWH) treated during acute infection, we developed a mathematical model to predict reservoir decay from peripheral CD4 + T cells. Nonlinear generalized additive models demonstrated rapid biphasic decay of intact DNA (week 0-5: t1/2 ~ 2.83 weeks; week 5-24: t1/2 ~ 15.4 weeks) that extended out to 1 year. These estimates were ~5-fold faster than prior decay estimates among chronic treated PLWH. Defective DNA had a similar biphasic pattern, but data were more variable. Predicted intact and defective decay rates were faster for PLWH with earlier timing of ART initiation, higher initial CD4 + T cell count, and lower pre-ART viral load. In this study, we advanced our limited understanding of HIV reservoir decay at the time of ART initiation, informing future curative strategies targeting this critical time.
Collapse
Affiliation(s)
- Alton Barbehenn
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Lei Shi
- Department of Biostatistics, University of California Berkeley, Berkeley, CA, USA
| | - Junzhe Shao
- Department of Biostatistics, University of California Berkeley, Berkeley, CA, USA
| | - Rebecca Hoh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Heather M Hartig
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Vivian Pae
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sannidhi Sarvadhavabhatla
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sophia Donaire
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Caroline Sheikhzadeh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey Milush
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Michael J Peluso
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey Martin
- Department of Biostatistics & Epidemiology, University of California San Francisco, San Francisco, CA, USA
| | - Frederick Hecht
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christopher Pilcher
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Stephanie E Cohen
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Susan Buchbinder
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Diane Havlir
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Monica Gandhi
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Timothy J Henrich
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Hiroyu Hatano
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jingshen Wang
- Department of Biostatistics, University of California Berkeley, Berkeley, CA, USA
| | - Steven G Deeks
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sulggi A Lee
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
26
|
Sun W, Gao C, Gladkov GT, Roseto I, Carrere L, Parsons EM, Gasca-Capote C, Frater J, Fidler S, Yu XG, Lichterfeld M. Footprints of innate immune activity during HIV-1 reservoir cell evolution in early-treated infection. J Exp Med 2024; 221:e20241091. [PMID: 39466203 PMCID: PMC11519379 DOI: 10.1084/jem.20241091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/28/2024] [Accepted: 09/27/2024] [Indexed: 10/29/2024] Open
Abstract
Antiretroviral treatment (ART) initiation during the early stages of HIV-1 infection is associated with a higher probability of maintaining drug-free viral control during subsequent treatment interruptions, for reasons that remain unclear. Using samples from a randomized-controlled human clinical trial evaluating therapeutic HIV-1 vaccines, we here show that early ART commencement is frequently associated with accelerated and efficient selection of genome-intact HIV-1 proviruses in repressive chromatin locations during the first year after treatment initiation. This selection process was unaffected by vaccine-induced HIV-1-specific T cell responses. Single-cell proteogenomic profiling demonstrated that cells harboring intact HIV-1 displayed a discrete phenotypic signature of immune selection by innate immune responses, characterized by a slight but significant upregulation of HLA-C, HLA-G, the IL-10 receptor, and other markers involved in innate immune regulation. Together, these results suggest an accelerated immune selection of viral reservoir cells during early-treated HIV-1 infection that seems at least partially driven by innate immune responses.
Collapse
Affiliation(s)
- Weiwei Sun
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Gregory Takashi Gladkov
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Isabelle Roseto
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Leah Carrere
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth M. Parsons
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Carmen Gasca-Capote
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College and Imperial College NIHR Biomedical Research Centre, London, UK
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
27
|
Mustafa M, Musselman D, Jayaweera D, da Fonseca Ferreira A, Marzouka G, Dong C. HIV-Associated Neurocognitive Disorder (HAND) and Alzheimer's Disease Pathogenesis: Future Directions for Diagnosis and Treatment. Int J Mol Sci 2024; 25:11170. [PMID: 39456951 PMCID: PMC11508543 DOI: 10.3390/ijms252011170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
HIV-associated neurocognitive disorder (HAND) and Alzheimer's disease (AD) are two neurocognitive disorders with overlapping clinical presentations and pathophysiology. The two have been thought to be two separate entities. However, the introduction and widespread use of antiretroviral therapy (ART) has altered the clinical manifestations of HAND, shifting from a pattern of subcortical dementia to one more akin to cortical dementia, resembling AD. Thus, the line between the two disease entities is not clear-cut. In this review, we discuss the concept of Alzheimer's disease-like dementia (ADLD) in HIV, which describes this phenomenon. While the mechanisms of HIV-associated ADLD remain to be elucidated, potential mechanisms include HIV-specific pathways, including epigenetic imprinting from initial viral infection, persistent and low viral load (which can only be detected by ultra-sensitive PCR), HIV-related inflammation, and putative pathways underlying traditional AD risk factors. Importantly, we have shown that HIV-specific microRNAs (miRs) encapsulated in extracellular vesicles (EV-miRs) play an important role in mediating the detrimental effects in the cardiovascular system. A useful preclinical model to study ADLD would be to expose AD mice to HIV-positive EVs to identify candidate EV-miRs that mediate the HIV-specific effects underlying ADLD. Characterization of the candidate EV-miRs may provide novel therapeutic armamentaria for ADLD.
Collapse
Affiliation(s)
- Mohammed Mustafa
- Department of Medicine, Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.M.); (D.J.)
| | - Dominique Musselman
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Dushyantha Jayaweera
- Department of Medicine, Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.M.); (D.J.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Andrea da Fonseca Ferreira
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - George Marzouka
- Department of Medicine, Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.M.); (D.J.)
- Division of Cardiovascular Disease, Department of Medicine, Miami VA Health System, University of Miami, Miami, FL 33136, USA
| | - Chunming Dong
- Department of Medicine, Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.M.); (D.J.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Division of Cardiovascular Disease, Department of Medicine, Miami VA Health System, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
28
|
Hiner CR, Mueller AL, Su H, Goldstein H. Interventions during Early Infection: Opening a Window for an HIV Cure? Viruses 2024; 16:1588. [PMID: 39459922 PMCID: PMC11512236 DOI: 10.3390/v16101588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Although combination antiretroviral therapy (ART) has been a landmark achievement for the treatment of human immunodeficiency virus (HIV), an HIV cure has remained elusive. Elimination of latent HIV reservoirs that persist throughout HIV infection is the most challenging barrier to an HIV cure. The progressive HIV infection is marked by the increasing size and diversity of latent HIV reservoirs until an effective immune response is mobilized, which can control but not eliminate HIV infection. The stalemate between HIV replication and the immune response is manifested by the establishment of a viral set point. ART initiation during the early stage limits HIV reservoir development, preserves immune function, improves the quality of life, and may lead to ART-free viral remission in a few people living with HIV (PLWH). However, for the overwhelming majority of PLWH, early ART initiation alone does not cure HIV, and lifelong ART is needed to sustain viral suppression. A critical area of research is focused on determining whether HIV could be functionally cured if additional treatments are provided alongside early ART. Several HIV interventions including Block and Lock, Shock and Kill, broadly neutralizing antibody (bNAb) therapy, adoptive CD8+ T cell therapy, and gene therapy have demonstrated delayed viral rebound and/or viral remission in animal models and/or some PLWH. Whether or not their application during early infection can improve the success of HIV remission is less studied. Herein, we review the current state of clinical and investigative HIV interventions and discuss their potential to improve the likelihood of post-treatment remission if initiated during early infection.
Collapse
Affiliation(s)
- Christopher R. Hiner
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - April L. Mueller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - Hang Su
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - Harris Goldstein
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
29
|
Shin Y, Park CM, Kim DE, Kim S, Lee SY, Lee JY, Jeon WH, Kim HG, Bae S, Yoon CH. Discovery of new acetamide derivatives of 5-indole-1,3,4-oxadiazol-2-thiol as inhibitors of HIV-1 Tat-mediated viral transcription. Antimicrob Agents Chemother 2024; 68:e0064324. [PMID: 39230310 PMCID: PMC11459959 DOI: 10.1128/aac.00643-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) encodes a transcriptional factor called Tat, which is critical for viral transcription. Tat-mediated transcription is highly ordered apart from the cellular manner; therefore, it is considered a target for developing anti-HIV-1 drugs. However, drugs targeting Tat-mediated viral transcription are not yet available. Our high-throughput screen of a compound library employing a dual-reporter assay identified a 1,3,4-oxadiazole scaffold against Tat and HIV-1 infection. Furthermore, a serial structure-activity relation (SAR) study performed with biological assays found 1,3,4-oxadiazole derivatives (9 and 13) containing indole and acetamide that exhibited potent inhibitory effects on HIV-1 infectivity, with half-maximal effective concentrations (EC50) of 0.17 (9) and 0.24 µM (13), respectively. The prominent derivatives specifically interfered with the viral transcriptional step without targeting other infection step(s) and efficiently inhibited the HIV-1 replication cycle in the T cell lines and peripheral blood mononuclear cells infected with HIV-1. Additionally, compared to the wild type, the compounds exhibited similar potency against anti-retroviral drug-resistant HIV-1 strains. In a series of mode-of-action studies, the compounds inhibited the ejection of histone H3 for facilitating viral transcription on the long-terminal repeat (LTR) promoter. Furthermore, SAHA (a histone deacetylase inhibitor) treatment abolished the compound potency, revealing that the compounds can possibly target Tat-regulated epigenetic modulation of LTR to inhibit viral transcription. Overall, our screening identified novel 1,3,4-oxadiazole compounds that inhibited HIV-1 Tat, and subsequent SAR-based optimization led to the derivatives 9 and 13 development that could be a promising scaffold for developing a new class of therapeutic agents for HIV-1 infection.
Collapse
Affiliation(s)
- YoungHyun Shin
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Chul Min Park
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Dong-Eun Kim
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Sungmin Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sang-Yeop Lee
- Research Center for Bioconvergence Analysis, Ochang Center, Korea Basic Science Institute, Cheongju-si, Republic of Korea
| | - Jun Young Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Won-Hui Jeon
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Hong Gi Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Songmee Bae
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Cheol-Hee Yoon
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, Korea National Institute of Health, Cheongju, Republic of Korea
| |
Collapse
|
30
|
Ling L, Kim M, Soper A, Kovarova M, Spagnuolo RA, Begum N, Kirchherr J, Archin N, Battaglia D, Cleveland D, Wahl A, Margolis DM, Browne EP, Garcia JV. Analysis of the effect of HDAC inhibitors on the formation of the HIV reservoir. mBio 2024; 15:e0163224. [PMID: 39136440 PMCID: PMC11389399 DOI: 10.1128/mbio.01632-24] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
The HIV reservoir is more dynamic than previously thought with around 70% of the latent reservoir originating from viruses circulating within 1 year of the initiation of antiretroviral therapy (ART). In an ex vivo model system of HIV latency, it was reported that early exposure to class I histone deacetylase (HDAC) inhibitors might prevent these more recently infected cells from entering a state of stable viral latency. This finding raises the possibility that co-administration of HDAC inhibitors at the time of ART initiation may prevent the establishment of much of the HIV reservoir. Here, we tested the effects of the HDAC inhibitors suberoylanilide hydroxamic acid (SAHA) and panobinostat co-administered at the time of ART initiation on the formation of the viral reservoir in HIV-infected humanized mice. As previously shown, SAHA and panobinostat were well tolerated in humanized mice. Unexpectedly, co-administration of SAHA resulted in an increase in the frequency of CD4+ cells carrying HIV DNA but did not alter the frequency of cell-associated HIV RNA in HIV-infected, ART-treated humanized mice. Co-administration of panobinostat did not alter levels of cell-associated HIV DNA or RNA. Our in vivo findings indicate that co-administration of HDAC inhibitors initiated at the same time of ART treatment does not prevent recently infected cells from entering latency.IMPORTANCECurrent antiretroviral therapy (ART) does not eradicate cells harboring replication-competent HIV reservoir. Withdrawal of ART inevitably results in a rapid viremia rebound. The HIV reservoir is more dynamic than previously thought. Early exposure to class I histone deacetylase (HDAC) inhibitors inhibit these more recently infected cells from entering a state of stable viral latency in an ex vivo model of latency, raising the possibility that co-administration of HDAC inhibitors at the time of ART initiation may reduce much of the HIV reservoir. Here, we tested the effects of the HDAC inhibitors suberoylanilide hydroxamic acid or panobinostat during ART initiation on the formation of the viral reservoir in HIV-infected humanized mice. Our in vivo study indicates that in contrast to in vitro observations, the co-administration of HDAC inhibitors at the same time of ART initiation does not prevent recently infected cells from entering latency.
Collapse
Affiliation(s)
- Lijun Ling
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manse Kim
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew Soper
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Martina Kovarova
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rae Ann Spagnuolo
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nurjahan Begum
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer Kirchherr
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nancie Archin
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Diana Battaglia
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dave Cleveland
- Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Angela Wahl
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David M. Margolis
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Edward P. Browne
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J. Victor Garcia
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
31
|
Margolis DM. Advancing Toward a Human Immunodeficiency Virus Cure: Initial Progress on a Difficult Path. Infect Dis Clin North Am 2024; 38:487-497. [PMID: 38969530 PMCID: PMC11410351 DOI: 10.1016/j.idc.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
Therapies to eradicate human immunodeficiency virus (HIV) infection, sparing lifelong antiviral therapy, are a still-distant goal. But significant advances have been made to reverse HIV latency while antiretroviral therapy (ART) is maintained to allow targeting of the persistent viral reservoir, to test interventions that could clear cells emerging from latent infection, and to improve HIV cure research assays and infrastructure. Steady progress gives hope that future therapies to clear HIV infection may relieve individuals and society of the burden of HIV.
Collapse
Affiliation(s)
- David M Margolis
- Medicine, Microbiology & Immunology, Epidemiology; UNC HIV Cure Center; University of North Carolina at Chapel Hill, 2016 Genetic Medicine Building, 120 Mason Farm Road, CB 7042, Chapel Hill, NC 27599-7042, USA.
| |
Collapse
|
32
|
Tarasova O, Petrou A, Ivanov SM, Geronikaki A, Poroikov V. Viral Factors in Modulation of Host Immune Response: A Route to Novel Antiviral Agents and New Therapeutic Approaches. Int J Mol Sci 2024; 25:9408. [PMID: 39273355 PMCID: PMC11395507 DOI: 10.3390/ijms25179408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Viruses utilize host cells at all stages of their life cycle, from the transcription of genes and translation of viral proteins to the release of viral copies. The human immune system counteracts viruses through a variety of complex mechanisms, including both innate and adaptive components. Viruses have an ability to evade different components of the immune system and affect them, leading to disruption. This review covers contemporary knowledge about the virus-induced complex interplay of molecular interactions, including regulation of transcription and translation in host cells resulting in the modulation of immune system functions. Thorough investigation of molecular mechanisms and signaling pathways that are involved in modulating of host immune response to viral infections can help to develop novel approaches for antiviral therapy. In this review, we consider new therapeutic approaches for antiviral treatment. Modern therapeutic strategies for the treatment and cure of human immunodeficiency virus (HIV) are considered in detail because HIV is a unique example of a virus that leads to host T lymphocyte deregulation and significant modulation of the host immune response. Furthermore, peculiarities of some promising novel agents for the treatment of various viral infections are described.
Collapse
Affiliation(s)
- Olga Tarasova
- Institute of Biomedical Chemistry, Moscow 119121, Russia
| | - Anthi Petrou
- School of Pharmacy, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | |
Collapse
|
33
|
Barbehenn A, Shi L, Shao J, Hoh R, Hartig HM, Pae V, Sarvadhavabhatla S, Donaire S, Sheikhzadeh C, Milush J, Laird GM, Mathias M, Ritter K, Peluso MJ, Martin J, Hecht F, Pilcher C, Cohen SE, Buchbinder S, Havlir D, Gandhi M, Henrich TJ, Hatano H, Wang J, Deeks SG, Lee SA. Rapid Biphasic Decay of Intact and Defective HIV DNA Reservoir During Acute Treated HIV Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.27.24304867. [PMID: 38585951 PMCID: PMC10996734 DOI: 10.1101/2024.03.27.24304867] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Despite antiretroviral therapy (ART), HIV persists in latently-infected cells ("the reservoir") which decay slowly over time. Here, leveraging >500 longitudinal samples from 67 people with HIV (PWH) treated during acute infection, we developed a novel mathematical model to predict reservoir decay from peripheral CD4+ T cells. Nonlinear generalized additive models demonstrated rapid biphasic decay of intact DNA (week 0-5: t1/2~2.83 weeks; week 5-24: t1/2~15.4 weeks) that extended out to 1 year. These estimates were ~5-fold faster than prior decay estimates among chronic treated PWH. Defective DNA had a similar biphasic pattern, but data were more variable. Predicted intact and defective decay rates were faster for PWH with earlier timing of ART initiation, higher initial CD4+ T cell count, and lower pre-ART viral load. These data add to our limited understanding of HIV reservoir decay at the time of ART initiation, informing future curative strategies targeting this critical time.
Collapse
Affiliation(s)
- Alton Barbehenn
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Lei Shi
- Department of Biostatistics, University of California Berkeley, Berkeley, CA 94110, USA
| | - Junzhe Shao
- Department of Biostatistics, University of California Berkeley, Berkeley, CA 94110, USA
| | - Rebecca Hoh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Heather M. Hartig
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Vivian Pae
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sannidhi Sarvadhavabhatla
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sophia Donaire
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Caroline Sheikhzadeh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jeffrey Milush
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | | | | | | | - Michael J. Peluso
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jeffrey Martin
- Department of Biostatistics & Epidemiology, University of California San Francisco, CA 94158, USA
| | - Frederick Hecht
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Christopher Pilcher
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Stephanie E. Cohen
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
- San Francisco Department of Public Health, San Francisco, CA 94102, USA
| | - Susan Buchbinder
- San Francisco Department of Public Health, San Francisco, CA 94102, USA
| | - Diane Havlir
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Monica Gandhi
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Timothy J. Henrich
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Hiroyu Hatano
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jingshen Wang
- Department of Biostatistics, University of California Berkeley, Berkeley, CA 94110, USA
| | - Steven G. Deeks
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sulggi A. Lee
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
34
|
Vasukutty A, Pillarisetti S, Choi J, Kang SH, Park IK. CXCR4 Targeting Nanoplatform for Transcriptional Activation of Latent HIV-1 Infected T Cells. ACS APPLIED BIO MATERIALS 2024; 7:4831-4842. [PMID: 37586084 DOI: 10.1021/acsabm.3c00456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Antiretroviral drugs are limited in their ability to target latent retroviral reservoirs in CD4+ T cells, highlighting the need for a T cell-targeted drug delivery system that activates the transcription of inactivated viral DNA in infected cells. Histone deacetylase inhibitors (HDACi) disrupt chromatin-mediated silencing of the viral genome and are explored in HIV latency reversal. But single drug formulations of HDACi are insufficient to elicit therapeutic efficacy, warranting combination therapy. Furthermore, protein kinase C activators (PKC) have shown latency reversal activity in HIV by activating the NF-κB signaling pathway. Combining HDACi (SAHA) with PKC (PMA) activators enhances HIV reservoir activation by promoting chromatin decondensation and subsequent transcriptional activation. In this study, we developed a mixed nanomicelle (PD-CR4) drug delivery system for simultaneous targeting of HIV-infected CD4+ T cells with two drugs, suberoylanilide hydroxamic acid (SAHA) and phorbol 12-myristate 13-acetate (PMA). SAHA is a HDACi that promotes chromatin decondensation, while PMA is a PKC agonist that enhances transcriptional activation. The physicochemical properties of the formulated PD-CR4 nanoparticles were characterized by NMR, CMC, DLS, and TEM analyses. Further, we investigated in vitro safety profiles, targeting efficacy, and transcriptional activation of inactivated HIV reservoir cells. Our results suggest that we successfully prepared a targeted PD system with dual drug loading. We have compared latency reversal efficacy of a single drug nanoformulation and combination drug nanoformulation. Final PD-SP-CR4 successfully activated infected CD4+ T cell reservoirs and showed enhanced antigen release from HIV reservoir T cells, compared with the single drug treatment group as expected. To summarize, our data shows PD-SP-CR4 has potential T cell targeting efficiency and efficiently activated dormant CD4+ T cells. Our data indicate that a dual drug-loaded particle has better therapeutic efficacy than a single loaded particle as expected. Hence, PD-CR4 can be further explored for HIV therapeutic drug delivery studies.
Collapse
Affiliation(s)
- Arathy Vasukutty
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Shameer Pillarisetti
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Shin Hyuk Kang
- Departments of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
35
|
Li TW, Park Y, Watters EG, Wang X, Zhou D, Fiches GN, Wu Z, Badley AD, Sacha JB, Ho WZ, Santoso NG, Qi J, Zhu J. KDM5A/B contribute to HIV-1 latent infection and survival of HIV-1 infected cells. Antiviral Res 2024; 228:105947. [PMID: 38925368 PMCID: PMC11927087 DOI: 10.1016/j.antiviral.2024.105947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
Combinational antiretroviral therapy (cART) suppresses human immunodeficiency virus type 1 (HIV-1) viral replication and pathogenesis in acquired immunodeficiency syndrome (AIDS) patients. However, HIV-1 remains in the latent stage of infection by suppressing viral transcription, which hinders an HIV-1 cure. One approach for an HIV-1 cure is the "shock and kill" strategy. The strategy focuses on reactivating latent HIV-1, inducing the viral cytopathic effect and facilitating the immune clearance for the elimination of latent HIV-1 reservoirs. Here, we reported that the H3K4 trimethylation (H3K4me3)-specific demethylase KDM5A/B play a role in suppressing HIV-1 Tat/LTR-mediated viral transcription in HIV-1 latent cells. Furthermore, we evaluated the potential of KDM5-specific inhibitor JQKD82 as an HIV-1 "shock and kill" agent. Our results showed that JQKD82 increases the H3K4me3 level at HIV-1 5' LTR promoter regions, HIV-1 reactivation, and the cytopathic effects in an HIV-1-latent T cell model. In addition, we identified that the combination of JQKD82 and AZD5582, a non-canonical NF-κB activator, generates a synergistic impact on inducing HIV-1 lytic reactivation and cell death in the T cell. The latency-reversing potency of the JQKD82 and AZD5582 pair was also confirmed in peripheral blood mononuclear cells (PBMCs) isolated from HIV-1 aviremic patients and in an HIV-1 latent monocyte. In latently infected microglia (HC69) of the brain, either deletion or inhibition of KDM5A/B results in a reversal of the HIV-1 latency. Overall, we concluded that KDM5A/B function as a host repressor of the HIV-1 lytic reactivation and thus promote the latency and the survival of HIV-1 infected reservoirs.
Collapse
Affiliation(s)
- Tai-Wei Li
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Youngmin Park
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Emily G Watters
- Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Dawei Zhou
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Guillaume N Fiches
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhenyu Wu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN, 55902, USA
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Netty G Santoso
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jun Qi
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Jian Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
36
|
Chou TC, Maggirwar NS, Marsden MD. HIV Persistence, Latency, and Cure Approaches: Where Are We Now? Viruses 2024; 16:1163. [PMID: 39066325 PMCID: PMC11281696 DOI: 10.3390/v16071163] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The latent reservoir remains a major roadblock to curing human immunodeficiency virus (HIV) infection. Currently available antiretroviral therapy (ART) can suppress active HIV replication, reduce viral loads to undetectable levels, and halt disease progression. However, antiretroviral drugs are unable to target cells that are latently infected with HIV, which can seed viral rebound if ART is stopped. Consequently, a major focus of the field is to study the latent viral reservoir and develop safe and effective methods to eliminate it. Here, we provide an overview of the major mechanisms governing the establishment and maintenance of HIV latency, the key challenges posed by latent reservoirs, small animal models utilized to study HIV latency, and contemporary cure approaches. We also discuss ongoing efforts to apply these approaches in combination, with the goal of achieving a safe, effective, and scalable cure for HIV that can be extended to the tens of millions of people with HIV worldwide.
Collapse
Affiliation(s)
- Tessa C. Chou
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
| | - Nishad S. Maggirwar
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
| | - Matthew D. Marsden
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
- Department of Medicine, Division of Infectious Disease, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
37
|
Kadiyala GN, Telwatte S, Wedrychowski A, Janssens J, Kim SJ, Kim P, Deeks S, Wong JK, Yukl SA. Differential susceptibility of cells infected with defective and intact HIV proviruses to killing by obatoclax and other small molecules. AIDS 2024; 38:1281-1291. [PMID: 38626436 PMCID: PMC11216394 DOI: 10.1097/qad.0000000000003908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/18/2024]
Abstract
OBJECTIVES Some drugs that augment cell-intrinsic defenses or modulate cell death/survival pathways have been reported to selectively kill cells infected with HIV or Simian Immunodeficiency Virus (SIV), but comparative studies are lacking. We hypothesized that these drugs may differ in their ability to kill cells infected with intact and defective proviruses. DESIGN To investigate this hypothesis, drugs were tested ex vivo on peripheral blood mononuclear cells (PBMC) from nine antiretroviral therapy (ART)-suppressed individuals. METHODS We tested drugs currently in clinical use or human trials, including auranofin (p53 modulator), interferon alpha2A, interferon gamma, acitretin (RIG-I inducer), GS-9620/vesatolimod (TLR7 agonist), nivolumab (PD-1 blocker), obatoclax (Bcl-2 inhibitor), birinapant [inhibitor of apoptosis proteins (IAP) inhibitor], bortezomib (proteasome inhibitor), and INK128/sapanisertib [mammalian target of rapamycin mTOR] [c]1/2 inhibitor). After 6 days of treatment, we measured cell counts/viabilities and quantified levels of total, intact, and defective HIV DNA by droplet digital PCR (Intact Proviral DNA Assay). RESULTS Obatoclax reduced intact HIV DNA [median = 27-30% of dimethyl sulfoxide control (DMSO)] but not defective or total HIV DNA. Other drugs showed no statistically significant effects. CONCLUSION Obatoclax and other Bcl-2 inhibitors deserve further study in combination therapies aimed at reducing the intact HIV reservoir in order to achieve a functional cure and/or reduce HIV-associated immune activation.
Collapse
Affiliation(s)
- Gayatri Nikhila Kadiyala
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Sushama Telwatte
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Adam Wedrychowski
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Julie Janssens
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Sun Jin Kim
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Peggy Kim
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Steven Deeks
- Department of Medicine, University of California, San Francisco
| | - Joseph K. Wong
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Steven A. Yukl
- Department of Medicine, University of California, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| |
Collapse
|
38
|
Raines SLM, Falcinelli SD, Peterson JJ, Van Gulck E, Allard B, Kirchherr J, Vega J, Najera I, Boden D, Archin NM, Margolis DM. Nanoparticle delivery of Tat synergizes with classical latency reversal agents to express HIV antigen targets. Antimicrob Agents Chemother 2024; 68:e0020124. [PMID: 38829049 PMCID: PMC11232404 DOI: 10.1128/aac.00201-24] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
Limited cellular levels of the HIV transcriptional activator Tat are one contributor to proviral latency that might be targeted in HIV cure strategies. We recently demonstrated that lipid nanoparticles containing HIV tat mRNA induce HIV expression in primary CD4 T cells. Here, we sought to further characterize tat mRNA in the context of several benchmark latency reversal agents (LRAs), including inhibitor of apoptosis protein antagonists (IAPi), bromodomain and extra-Terminal motif inhibitors (BETi), and histone deacetylase inhibitors (HDACi). tat mRNA reversed latency across several different cell line models of HIV latency, an effect dependent on the TAR hairpin loop. Synergistic enhancement of tat mRNA activity was observed with IAPi, HDACi, and BETi, albeit to variable degrees. In primary CD4 T cells from durably suppressed people with HIV, tat mRNA profoundly increased the frequencies of elongated, multiply-spliced, and polyadenylated HIV transcripts, while having a lesser impact on TAR transcript frequencies. tat mRNAs alone resulted in variable HIV p24 protein induction across donors. However, tat mRNA in combination with IAPi, BETi, or HDACi markedly enhanced HIV RNA and protein expression without overt cytotoxicity or cellular activation. Notably, combination regimens approached or in some cases exceeded the latency reversal activity of maximal mitogenic T cell stimulation. Higher levels of tat mRNA-driven HIV p24 induction were observed in donors with larger mitogen-inducible HIV reservoirs, and expression increased with prolonged exposure time. Combination LRA strategies employing both small molecule inhibitors and Tat delivered to CD4 T cells are a promising approach to effectively target the HIV reservoir.
Collapse
Affiliation(s)
- Samuel L. M. Raines
- Department of Medicine and UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shane D. Falcinelli
- Department of Medicine and UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jackson J. Peterson
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ellen Van Gulck
- Janssen Infectious Diseases, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Brigitte Allard
- Department of Medicine and UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer Kirchherr
- Department of Medicine and UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jerel Vega
- Arcturus Therapeutics, Science Center Drive, San Diego, California, USA
| | - Isabel Najera
- Janssen Infectious Diseases, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Daniel Boden
- Janssen Infectious Diseases, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Nancie M. Archin
- Department of Medicine and UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David M. Margolis
- Department of Medicine and UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
39
|
Jalan A, Jayasree PJ, Karemore P, Narayan KP, Khandelia P. Decoding the 'Fifth' Nucleotide: Impact of RNA Pseudouridylation on Gene Expression and Human Disease. Mol Biotechnol 2024; 66:1581-1598. [PMID: 37341888 DOI: 10.1007/s12033-023-00792-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023]
Abstract
Cellular RNAs, both coding and noncoding are adorned by > 100 chemical modifications, which impact various facets of RNA metabolism and gene expression. Very often derailments in these modifications are associated with a plethora of human diseases. One of the most oldest of such modification is pseudouridylation of RNA, wherein uridine is converted to a pseudouridine (Ψ) via an isomerization reaction. When discovered, Ψ was referred to as the 'fifth nucleotide' and is chemically distinct from uridine and any other known nucleotides. Experimental evidence accumulated over the past six decades, coupled together with the recent technological advances in pseudouridine detection, suggest the presence of pseudouridine on messenger RNA, as well as on diverse classes of non-coding RNA in human cells. RNA pseudouridylation has widespread effects on cellular RNA metabolism and gene expression, primarily via stabilizing RNA conformations and destabilizing interactions with RNA-binding proteins. However, much remains to be understood about the RNA targets and their recognition by the pseudouridylation machinery, the regulation of RNA pseudouridylation, and its crosstalk with other RNA modifications and gene regulatory processes. In this review, we summarize the mechanism and molecular machinery involved in depositing pseudouridine on target RNAs, molecular functions of RNA pseudouridylation, tools to detect pseudouridines, the role of RNA pseudouridylation in human diseases like cancer, and finally, the potential of pseudouridine to serve as a biomarker and as an attractive therapeutic target.
Collapse
Affiliation(s)
- Abhishek Jalan
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - P J Jayasree
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - Pragati Karemore
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - Piyush Khandelia
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India.
| |
Collapse
|
40
|
De La Torre Tarazona E, Passaes C, Moreno S, Sáez-Cirión A, Alcamí J. High concentrations of Maraviroc do not alter immunological and metabolic parameters of CD4 T cells. Sci Rep 2024; 14:13980. [PMID: 38886484 PMCID: PMC11183235 DOI: 10.1038/s41598-024-64902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Maraviroc (MVC) is an antiretroviral drug capable of binding to CCR5 receptors and block HIV entry into target cells. Moreover, MVC can activate NF-kB pathway and induce viral transcription in HIV-infected cells, being proposed as a latency reversal agent (LRA) in HIV cure strategies. However, the evaluation of immunological and metabolic parameters induced by MVC concentrations capable of inducing HIV transcription have not been explored in depth. We cultured isolated CD4 T cells in the absence or presence of MVC, and evaluated the frequency of CD4 T cell subpopulations and activation markers levels by flow cytometry, and the oxidative and glycolytic metabolic rates of CD4 T cells using a Seahorse Analyzer. Our results indicate that a high concentration of MVC did not increase the levels of activation markers, as well as glycolytic or oxidative metabolic rates in CD4 T cells. Furthermore, MVC did not induce significant changes in the frequency and activation levels of memory cell subpopulations. Our data support a safety profile of MVC as a promising LRA candidate since it does not induce alterations of the immunological and metabolic parameters that could affect the functionality of these immune cells.
Collapse
Affiliation(s)
- Erick De La Torre Tarazona
- Infectious Diseases Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), University Hospital Ramón y Cajal, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Caroline Passaes
- HIV, Inflammation and Persistence Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Viral Reservoirs and Immune Control Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Santiago Moreno
- Infectious Diseases Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), University Hospital Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Alcalá University, Madrid, Spain
| | - Asier Sáez-Cirión
- HIV, Inflammation and Persistence Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Viral Reservoirs and Immune Control Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - José Alcamí
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
41
|
Ishii T, Kobayakawa T, Matsuda K, Nigorikawa K, Bolah P, Noborio A, Tsuji K, Ohashi N, Yoshimura K, Nomura W, Mitsuya H, Maeda K, Tamamura H. Discovery of Potent DAG-Lactone Derivatives as HIV Latency Reversing Agents. ACS Infect Dis 2024; 10:2250-2261. [PMID: 38771724 DOI: 10.1021/acsinfecdis.4c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Toward human immunodeficiency virus type-1 (HIV-1) cure, cells latently infected with HIV-1 must be eliminated from people living with HIV-1. We previously developed a protein kinase C (PKC) activator, diacylglycerol (DAG)-lactone derivative 3, with high HIV-1 latency-reversing activity, based on YSE028 (2) as a lead compound and found that the activity was correlated with binding affinity for PKC and stability against esterase-mediated hydrolysis. Here, we synthesized new DAG-lactone derivatives not only containing a tertiary ester group or an isoxazole surrogate but also several symmetric alkylidene moieties to improve HIV-1 latency reversing activity. Compound 9a, with a dimethyl group at the α-position of the ester group, exerted twice higher HIV-1 latency reversing activity than compound 3, and compound 26, with the isoxazole moiety, was significantly active. In addition, DAG-lactone derivatives with moderate hydrophobicity and potent biostability showed high biological activity.
Collapse
Affiliation(s)
- Takahiro Ishii
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan
| | - Takuya Kobayakawa
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan
| | - Kouki Matsuda
- Division of Antiviral Therapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Kagoshima 890-8544, Japan
| | - Kiyomi Nigorikawa
- Department of Genome and Biomolecular Engineering for Drug Discovery, School of Pharmaceutical Sciences and Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Hiroshima 734-8553, Japan
| | - Peter Bolah
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan
| | - Airi Noborio
- Division of Antiviral Therapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Kagoshima 890-8544, Japan
| | - Kohei Tsuji
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan
| | - Nami Ohashi
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Kazuhisa Yoshimura
- Institute of Public Health, Bureau of Social Welfare and Public Health, Tokyo Metropolitan Government, Shinjuku-ku, Tokyo 169-0073, Japan
| | - Wataru Nomura
- Department of Genome and Biomolecular Engineering for Drug Discovery, School of Pharmaceutical Sciences and Graduate School of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Hiroshima 734-8553, Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Shinjuku-ku, Tokyo 162-8655, Japan
- Experimental Retrovirology Section, HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Department of Clinical Sciences, Kumamoto University Hospital, Chuo-ku, Kumamoto 860-8556, Japan
| | - Kenji Maeda
- Division of Antiviral Therapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Kagoshima 890-8544, Japan
| | - Hirokazu Tamamura
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan
| |
Collapse
|
42
|
Lilie T, Bouzy J, Asundi A, Taylor J, Roche S, Olson A, Coxen K, Corry H, Jordan H, Clayton K, Lin N, Tsibris A. HIV-1 latency reversal agent boosting is not limited by opioid use. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.05.26.23290576. [PMID: 37398278 PMCID: PMC10312897 DOI: 10.1101/2023.05.26.23290576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The opioid epidemic may impact the HIV-1 reservoir and its reversal from latency in virally suppressed people with HIV (PWH). We studied forty-seven PWH and observed that lowering the concentration of HIV-1 latency reversal agents (LRA), used in combination with small molecules that do not reverse latency, synergistically increases the magnitude of HIV-1 re-activation ex vivo, regardless of opioid use. This LRA boosting, which combines a Smac mimetic or low-dose protein kinase C agonist with histone deacetylase inhibitors, can generate significantly more unspliced HIV-1 transcription than phorbol 12-myristate 13-acetate (PMA) with ionomycin (PMAi), the maximal known HIV-1 reactivator. LRA boosting associated with greater histone acetylation in CD4+ T cells and modulated T cell activation-induced markers and intracellular cytokine production; Smac mimetic-based boosting was less likely to induce immune activation. We found that HIV-1 reservoirs in PWH contain unspliced and polyadenylated (polyA) virus mRNA, the ratios of which are greater in resting than total CD4+ T cells and can correct to 1:1 with PMAi exposure. Latency reversal results in greater fold-change increases to HIV-1 poly(A) mRNA than unspliced message. Multiply spliced HIV-1 transcripts and virion production did not consistently increase with LRA boosting, suggesting the presence of a persistent post-transcriptional block. LRA boosting can be leveraged to probe the mechanisms of an effective cellular HIV-1 latency reversal program.
Collapse
Affiliation(s)
- Tyler Lilie
- Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Archana Asundi
- Department of Medicine, Boston University School of Medicine & Boston Medical Center, Boston, MA USA
| | - Jessica Taylor
- Department of Medicine, Boston University School of Medicine & Boston Medical Center, Boston, MA USA
- Grayken Center for Addiction, Boston Medical Center, Boston, MA USA
| | - Samantha Roche
- Department of Medicine, Boston University School of Medicine & Boston Medical Center, Boston, MA USA
| | - Alex Olson
- Department of Medicine, Boston University School of Medicine & Boston Medical Center, Boston, MA USA
| | | | | | | | - Kiera Clayton
- Department of Pathology, University of Massachusetts T.H. Chan School of Medicine, Worcester, MA, USA
| | - Nina Lin
- Department of Medicine, Boston University School of Medicine & Boston Medical Center, Boston, MA USA
| | - Athe Tsibris
- Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Armani-Tourret M, Bone B, Tan TS, Sun W, Bellefroid M, Struyve T, Louella M, Yu XG, Lichterfeld M. Immune targeting of HIV-1 reservoir cells: a path to elimination strategies and cure. Nat Rev Microbiol 2024; 22:328-344. [PMID: 38337034 PMCID: PMC11131351 DOI: 10.1038/s41579-024-01010-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Successful approaches for eradication or cure of HIV-1 infection are likely to include immunological mechanisms, but remarkably little is known about how human immune responses can recognize and interact with the few HIV-1-infected cells that harbour genome-intact viral DNA, persist long term despite antiretroviral therapy and represent the main barrier to a cure. For a long time regarded as being completely shielded from host immune responses due to viral latency, these cells do, on closer examination with single-cell analytic techniques, display discrete footprints of immune selection, implying that human immune responses may be able to effectively engage and target at least some of these cells. The failure to eliminate rebound-competent virally infected cells in the majority of persons likely reflects the evolution of a highly selected pool of reservoir cells that are effectively camouflaged from immune recognition or rely on sophisticated approaches for resisting immune-mediated killing. Understanding the fine-tuned interplay between host immune responses and viral reservoir cells will help to design improved interventions that exploit the immunological vulnerabilities of HIV-1 reservoir cells.
Collapse
Affiliation(s)
- Marie Armani-Tourret
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Bone
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Toong Seng Tan
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Weiwei Sun
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Maxime Bellefroid
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Tine Struyve
- HIV Cure Research Center, Ghent University, Ghent, Belgium
| | - Michael Louella
- Community Advisory Board, Delaney AIDS Research Enterprise (DARE), San Francisco, CA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
44
|
Bussey-Sutton CR, Ward A, Fox JA, Turner AMW, Peterson JJ, Emery A, Longoria AR, Gomez-Martinez I, Jones C, Hepperla A, Margolis DM, Strahl BD, Browne EP. The histone methyltransferase SETD2 regulates HIV expression and latency. PLoS Pathog 2024; 20:e1012281. [PMID: 38848441 PMCID: PMC11189200 DOI: 10.1371/journal.ppat.1012281] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/20/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Understanding the mechanisms that drive HIV expression and latency is a key goal for achieving an HIV cure. Here we investigate the role of the SETD2 histone methyltransferase, which deposits H3K36 trimethylation (H3K36me3), in HIV infection. We show that prevention of H3K36me3 by a potent and selective inhibitor of SETD2 (EPZ-719) leads to reduced post-integration viral gene expression and accelerated emergence of latently infected cells. CRISPR/Cas9-mediated knockout of SETD2 in primary CD4 T cells confirmed the role of SETD2 in HIV expression. Transcriptomic profiling of EPZ-719-exposed HIV-infected cells identified numerous pathways impacted by EPZ-719. Notably, depletion of H3K36me3 prior to infection did not prevent HIV integration but resulted in a shift of integration sites from highly transcribed genes to quiescent chromatin regions and to polycomb repressed regions. We also observed that SETD2 inhibition did not apparently affect HIV RNA levels, indicating a post-transcriptional mechanism affecting HIV expression. Viral RNA splicing was modestly reduced in the presence of EPZ-719. Intriguingly, EPZ-719 exposure enhanced responsiveness of latent HIV to the HDAC inhibitor vorinostat, suggesting that H3K36me3 can contribute to a repressive chromatin state at the HIV locus. These results identify SETD2 and H3K36me3 as novel regulators of HIV integration, expression and latency.
Collapse
Affiliation(s)
- Cameron R. Bussey-Sutton
- Department of Biochemistry, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Airlie Ward
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joshua A. Fox
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Anne-Marie W. Turner
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jackson J. Peterson
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ann Emery
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Arturo R. Longoria
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ismael Gomez-Martinez
- Department of Genetics, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Corbin Jones
- Department of Genetics, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Austin Hepperla
- Department of Genetics, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David M. Margolis
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Brian D. Strahl
- Department of Biochemistry, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Edward P. Browne
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
45
|
Roy S, Majee P, Sudhakar S, Mishra S, Kalia J, Pradeepkumar PI, Srivatsan SG. Structural elucidation of HIV-1 G-quadruplexes in a cellular environment and their ligand binding using responsive 19F-labeled nucleoside probes. Chem Sci 2024; 15:7982-7991. [PMID: 38817587 PMCID: PMC11134374 DOI: 10.1039/d4sc01755b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Understanding the structure and recognition of highly conserved regulatory segments of the integrated viral DNA genome that forms unique topologies can greatly aid in devising novel therapeutic strategies to counter chronic infections. In this study, we configured a probe system using highly environment-sensitive nucleoside analogs, 5-fluoro-2'-deoxyuridine (FdU) and 5-fluorobenzofuran-2'-deoxyuridine (FBFdU), to investigate the structural polymorphism of HIV-1 long terminal repeat (LTR) G-quadruplexes (GQs) by fluorescence and 19F NMR. FdU and FBFdU, serving as hairpin and GQ sensors, produced distinct spectral signatures for different GQ topologies adopted by LTR G-rich oligonucleotides. Importantly, systematic 19F NMR analysis in Xenopus laevis oocytes gave unprecedented information on the structure adopted by the LTR G-rich region in the cellular environment. The results indicate that it forms a unique GQ-hairpin hybrid architecture, a potent hotspot for selective targeting. Furthermore, structural models generated using MD simulations provided insights on how the probe system senses different GQs. Using the responsiveness of the probes and Taq DNA polymerase stop assay, we monitored GQ- and hairpin-specific ligand interactions and their synergistic inhibitory effect on the replication process. Our findings suggest that targeting GQ and hairpin motifs simultaneously using bimodal ligands could be a new strategy to selectively block the viral replication.
Collapse
Affiliation(s)
- Sarupa Roy
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr Homi Bhabha Road Pune 411008 India
| | - Priyasha Majee
- Department of Chemistry, Indian Institute of Technology Bombay Mumbai 400076 India
| | - Sruthi Sudhakar
- Department of Chemistry, Indian Institute of Technology Bombay Mumbai 400076 India
| | - Satyajit Mishra
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal Bhopal Bypass Road, Bhauri Bhopal 462066 India
| | - Jeet Kalia
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal Bhopal Bypass Road, Bhauri Bhopal 462066 India
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Bhopal Bhopal Bypass Road, Bhauri Bhopal 462066 India
| | - P I Pradeepkumar
- Department of Chemistry, Indian Institute of Technology Bombay Mumbai 400076 India
| | - Seergazhi G Srivatsan
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr Homi Bhabha Road Pune 411008 India
| |
Collapse
|
46
|
Mao Y, Liao Q, Zhu Y, Bi M, Zou J, Zheng N, Zhu L, Zhao C, Liu Q, Liu L, Chen J, Gu L, Liu Z, Pan X, Xue Y, Feng M, Ying T, Zhou P, Wu Z, Xiao J, Zhang R, Leng J, Sun Y, Zhang X, Xu J. Efficacy and safety of novel multifunctional M10 CAR-T cells in HIV-1-infected patients: a phase I, multicenter, single-arm, open-label study. Cell Discov 2024; 10:49. [PMID: 38740803 DOI: 10.1038/s41421-024-00658-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 02/02/2024] [Indexed: 05/16/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells have been proposed for HIV-1 treatment but have not yet demonstrated desirable therapeutic efficacy. Here, we report newly developed anti-HIV-1 CAR-T cells armed with endogenic broadly neutralizing antibodies (bNAbs) and the follicle-homing receptor CXCR5, termed M10 cells. M10 cells were designed to exercise three-fold biological functions, including broad cytotoxic effects on HIV-infected cells, neutralization of cell-free viruses produced after latency reversal, and B-cell follicle homing. After demonstrating the three-fold biological activities, M10 cells were administered to treat 18 HIV-1 patients via a regimen of two allogenic M10 cell infusions with an interval of 30 days, with each M10 cell infusion followed by two chidamide stimulations for HIV-1 reservoir activation. Consequently, 74.3% of M10 cell infusions resulted in significant suppression of viral rebound, with viral loads declining by an average of 67.1%, and 10 patients showed persistently reduced cell-associated HIV-1 RNA levels (average decrease of 1.15 log10) over the 150-day observation period. M10 cells were also found to impose selective pressure on the latent viral reservoir. No significant treatment-related adverse effects were observed. Overall, our study supported the potential of M10 CAR-T cells as a novel, safe, and effective therapeutic option for the functional cure of HIV-1/AIDS.
Collapse
Affiliation(s)
- Yunyu Mao
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qibin Liao
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Youwei Zhu
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Mingyuan Bi
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Jun Zou
- AIDS Clinical Treatment Center, The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Nairong Zheng
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lingyan Zhu
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Chen Zhao
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qing Liu
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Li Liu
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jun Chen
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ling Gu
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhuoqun Liu
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xinghao Pan
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ying Xue
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Meiqi Feng
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tianlei Ying
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Pingyu Zhou
- Shanghai Skin Disease Hospital, Tongji University, Shanghai, China
| | - Zhanshuai Wu
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Department of Medical Immunology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jian Xiao
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Department of Medical Immunology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Renfang Zhang
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| | - Jing Leng
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Department of Medical Immunology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| | - Yongtao Sun
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China.
| | - Xiaoyan Zhang
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| | - Jianqing Xu
- Clinical Center of Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
47
|
Ashokkumar M, Mei W, Peterson JJ, Harigaya Y, Murdoch DM, Margolis DM, Kornfein C, Oesterling A, Guo Z, Rudin CD, Jiang Y, Browne EP. Integrated Single-cell Multiomic Analysis of HIV Latency Reversal Reveals Novel Regulators of Viral Reactivation. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae003. [PMID: 38902848 PMCID: PMC11189801 DOI: 10.1093/gpbjnl/qzae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 06/22/2024]
Abstract
Despite the success of antiretroviral therapy, human immunodeficiency virus (HIV) cannot be cured because of a reservoir of latently infected cells that evades therapy. To understand the mechanisms of HIV latency, we employed an integrated single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin with sequencing (scATAC-seq) approach to simultaneously profile the transcriptomic and epigenomic characteristics of ∼ 125,000 latently infected primary CD4+ T cells after reactivation using three different latency reversing agents. Differentially expressed genes and differentially accessible motifs were used to examine transcriptional pathways and transcription factor (TF) activities across the cell population. We identified cellular transcripts and TFs whose expression/activity was correlated with viral reactivation and demonstrated that a machine learning model trained on these data was 75%-79% accurate at predicting viral reactivation. Finally, we validated the role of two candidate HIV-regulating factors, FOXP1 and GATA3, in viral transcription. These data demonstrate the power of integrated multimodal single-cell analysis to uncover novel relationships between host cell factors and HIV latency.
Collapse
Affiliation(s)
- Manickam Ashokkumar
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wenwen Mei
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jackson J Peterson
- HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuriko Harigaya
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David M Murdoch
- Department of Medicine, Duke University, Durham, NC 27708, USA
| | - David M Margolis
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Caleb Kornfein
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Alex Oesterling
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Zhicheng Guo
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Cynthia D Rudin
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Yuchao Jiang
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Edward P Browne
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
48
|
Joshi VR, Altfeld M. Harnessing natural killer cells to target HIV-1 persistence. Curr Opin HIV AIDS 2024; 19:141-149. [PMID: 38457230 DOI: 10.1097/coh.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review recent advances in the role of natural killer (NK) cells in approaches aimed at reducing the latent HIV-1 reservoir. RECENT FINDINGS Multiple approaches to eliminate cells harboring latent HIV-1 are being explored, but have been met with limited success so far. Recent studies have highlighted the role of NK cells and their potential in HIV-1 cure efforts. Anti-HIV-1 NK cell function can be optimized by enhancing NK cell activation, antibody dependent cellular cytotoxicity, reversing inhibition of NK cells as well as by employing immunotherapeutic complexes to enable HIV-1 specificity of NK cells. While NK cells alone do not eliminate the HIV-1 reservoir, boosting NK cell function might complement other strategies involving T cell and B cell immunity towards an HIV-1 functional cure. SUMMARY Numerous studies focusing on targeting latently HIV-1-infected cells have emphasized a potential role of NK cells in these strategies. Our review highlights recent advances in harnessing NK cells in conjunction with latency reversal agents and other immunomodulatory therapeutics to target HIV-1 persistence.
Collapse
Affiliation(s)
- Vinita R Joshi
- Department of Virus Immunology, Leibniz Institute of Virology
| | - Marcus Altfeld
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
49
|
Kobayashi-Ishihara M, Tsunetsugu-Yokota Y. Post-Transcriptional HIV-1 Latency: A Promising Target for Therapy? Viruses 2024; 16:666. [PMID: 38793548 PMCID: PMC11125802 DOI: 10.3390/v16050666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Human Immunodeficiency Virus type 1 (HIV-1) latency represents a significant hurdle in finding a cure for HIV-1 infections, despite tireless research efforts. This challenge is partly attributed to the intricate nature of HIV-1 latency, wherein various host and viral factors participate in multiple physiological processes. While substantial progress has been made in discovering therapeutic targets for HIV-1 transcription, targets for the post-transcriptional regulation of HIV-1 infections have received less attention. However, cumulative evidence now suggests the pivotal contribution of post-transcriptional regulation to the viral latency in both in vitro models and infected individuals. In this review, we explore recent insights on post-transcriptional latency in HIV-1 and discuss the potential of its therapeutic targets, illustrating some host factors that restrict HIV-1 at the post-transcriptional level.
Collapse
Affiliation(s)
- Mie Kobayashi-Ishihara
- Department of Molecular Biology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | | |
Collapse
|
50
|
LaPorte A, Pathak R, Eliscovich C, Martins L, Nell R, Spivak A, Suzuki M, Planelles V, Singer R, Kalpana G. Single-molecule RNA-FISH analysis reveals stochasticity in reactivation of latent HIV-1 regulated by Nuclear Orphan Receptors NR4A and cMYC. RESEARCH SQUARE 2024:rs.3.rs-4166090. [PMID: 38699331 PMCID: PMC11065080 DOI: 10.21203/rs.3.rs-4166090/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
HIV-1 eradication strategies require complete reactivation of HIV-1 latent cells by Latency Reversing Agents (LRA). Current methods lack effectiveness due to incomplete proviral reactivation. We employed a single-molecule RNA-FISH (smRNA-FISH) and FISH-Quant analysis and found that proviral reactivation is highly variable from cell-to-cell, stochastic, and occurs in bursts and waves, with different kinetics in response to diverse LRAs. Approximately 1-5% of latent cells exhibited stochastic reactivation without LRAs. Through single-cell RNA-seq analysis, we identified NR4A3 and cMYC as extrinsic factors associated with stochastic HIV-1 reactivation. Concomitant with HIV-1 reactivation cMYC was downregulated and NR4A3 was upregulated in both latent cell lines and primary CD4+ T-cells from aviremic patients. By inhibiting cMYC using SN-38, an active metabolite of irinotecan, we induced NR4A3 and HIV-1 expression. Our results suggest that inherent stochasticity in proviral reactivation contributes to cell-to-cell variability, which could potentially be modulated by drugs targeting cMYC and NR4A3.
Collapse
|