Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38328062

ABSTRACT

Gene therapy-based HIV cure strategies typically aim to excise the HIV provirus directly, or target host dependency factors (HDFs) that support viral persistence. Cure approaches will likely require simultaneous co-targeting of multiple sites within the HIV genome to prevent evolution of resistance, and/or co-targeting of multiple HDFs to fully render host cells refractory to HIV infection. Bulk cell-based methods do not enable inference of co-editing within individual viral or target cell genomes, and do not discriminate between monoallelic and biallelic gene disruption. Here, we describe a targeted single-cell DNA sequencing (scDNA-seq) platform characterizing the near full-length HIV genome and 50 established HDF genes, designed to evaluate anti-HIV gene therapy strategies. We implemented the platform to investigate the capacity of multiplexed CRISPR-Cas9 ribonucleoprotein complexes (Cas9-RNPs) to simultaneously 1) inactivate the HIV provirus, and 2) knockout the CCR5 and CXCR4 HDF (entry co-receptor) genes in microglia and primary monocyte-derived macrophages (MDMs). Our scDNA-seq pipeline revealed that antiviral gene editing is rarely observed at multiple loci (or both alleles of a locus) within an individual cell, and editing probabilities across sites are linked. Our results demonstrate that single-cell sequencing is critical to evaluate the true efficacy and therapeutic potential of HIV gene therapy.

2.
PLoS Pathog ; 15(1): e1007498, 2019 01.
Article in English | MEDLINE | ID: mdl-30645648

ABSTRACT

The establishment of HIV-1 latency gives rise to persistent chronic infection that requires life-long treatment. To reverse latency for viral eradiation, the HIV-1 Tat protein and its associated ELL2-containing Super Elongation Complexes (ELL2-SECs) are essential to activate HIV-1 transcription. Despite efforts to identify effective latency-reversing agents (LRA), avenues for exposing latent HIV-1 remain inadequate, prompting the need to identify novel LRA targets. Here, by conducting a CRISPR interference-based screen to reiteratively enrich loss-of-function genotypes that increase HIV-1 transcription in latently infected CD4+ T cells, we have discovered a key role of the proteasome in maintaining viral latency. Downregulating or inhibiting the proteasome promotes Tat-transactivation in cell line models. Furthermore, the FDA-approved proteasome inhibitors bortezomib and carfilzomib strongly synergize with existing LRAs to reactivate HIV-1 in CD4+ T cells from antiretroviral therapy-suppressed individuals without inducing cell activation or proliferation. Mechanistically, downregulating/inhibiting the proteasome elevates the levels of ELL2 and ELL2-SECs to enable Tat-transactivation, indicating the proteasome-ELL2 axis as a key regulator of HIV-1 latency and promising target for therapeutic intervention.


Subject(s)
HIV-1/metabolism , Proteasome Endopeptidase Complex/metabolism , Virus Latency/drug effects , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , CRISPR-Cas Systems , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Seropositivity , HIV-1/pathogenicity , Humans , Jurkat Cells , Proteasome Endopeptidase Complex/physiology , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/pharmacology , Transcriptional Elongation Factors , Virus Activation/drug effects , Virus Latency/physiology
3.
PLoS Pathog ; 12(6): e1005677, 2016 06.
Article in English | MEDLINE | ID: mdl-27253379

ABSTRACT

Identifying host immune determinants governing HIV transcription, latency and infectivity in vivo is critical to developing an HIV cure. Based on our recent finding that the host factor p21 regulates HIV transcription during antiretroviral therapy (ART), and published data demonstrating that the human carbohydrate-binding immunomodulatory protein galectin-9 regulates p21, we hypothesized that galectin-9 modulates HIV transcription. We report that the administration of a recombinant, stable form of galectin-9 (rGal-9) potently reverses HIV latency in vitro in the J-Lat HIV latency model. Furthermore, rGal-9 reverses HIV latency ex vivo in primary CD4+ T cells from HIV-infected, ART-suppressed individuals (p = 0.002), more potently than vorinostat (p = 0.02). rGal-9 co-administration with the latency reversal agent "JQ1", a bromodomain inhibitor, exhibits synergistic activity (p<0.05). rGal-9 signals through N-linked oligosaccharides and O-linked hexasaccharides on the T cell surface, modulating the gene expression levels of key transcription initiation, promoter proximal-pausing, and chromatin remodeling factors that regulate HIV latency. Beyond latent viral reactivation, rGal-9 induces robust expression of the host antiviral deaminase APOBEC3G in vitro and ex vivo (FDR<0.006) and significantly reduces infectivity of progeny virus, decreasing the probability that the HIV reservoir will be replenished when latency is reversed therapeutically. Lastly, endogenous levels of soluble galectin-9 in the plasma of 72 HIV-infected ART-suppressed individuals were associated with levels of HIV RNA in CD4+ T cells (p<0.02) and with the quantity and binding avidity of circulating anti-HIV antibodies (p<0.009), suggesting a role of galectin-9 in regulating HIV transcription and viral production in vivo during therapy. Our data suggest that galectin-9 and the host glycosylation machinery should be explored as foundations for novel HIV cure strategies.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Galectins/metabolism , HIV Infections/metabolism , Virus Activation/physiology , Virus Latency/physiology , Anti-HIV Agents/therapeutic use , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/physiology , Humans , Polymerase Chain Reaction , Transcription, Genetic/physiology , Transcriptome
4.
PLoS Pathog ; 11(6): e1004955, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26067822

ABSTRACT

Highly active antiretroviral therapy (HAART) suppresses human immunodeficiency virus (HIV) replication to undetectable levels but cannot fully eradicate the virus because a small reservoir of CD4+ T cells remains latently infected. Since HIV efficiently infects only activated CD4+ T cells and since latent HIV primarily resides in resting CD4+ T cells, it is generally assumed that latency is established when a productively infected cell recycles to a resting state, trapping the virus in a latent state. In this study, we use a dual reporter virus--HIV Duo-Fluo I, which identifies latently infected cells immediately after infection--to investigate how T cell activation affects the establishment of HIV latency. We show that HIV latency can arise from the direct infection of both resting and activated CD4+ T cells. Importantly, returning productively infected cells to a resting state is not associated with a significant silencing of the integrated HIV. We further show that resting CD4+ T cells from human lymphoid tissue (tonsil, spleen) show increased latency after infection when compared to peripheral blood. Our findings raise significant questions regarding the most commonly accepted model for the establishment of latent HIV and suggest that infection of both resting and activated primary CD4+ T cells produce latency.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , Lymphocyte Activation/immunology , Virus Latency/physiology , Cell Separation , Cells, Cultured , Flow Cytometry , Humans , Virus Replication/immunology
5.
J Biol Chem ; 290(22): 13736-48, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25873391

ABSTRACT

The presence of a small number of infected but transcriptionally dormant cells currently thwarts a cure for the more than 35 million individuals infected with HIV. Reactivation of these latently infected cells may result in three fates: 1) cell death due to a viral cytopathic effect, 2) cell death due to immune clearance, or 3) a retreat into latency. Uncovering the dynamics of HIV gene expression and silencing in the latent reservoir will be crucial for developing an HIV-1 cure. Here we identify and characterize an intracellular circuit involving TRIM32, an HIV activator, and miR-155, a microRNA that may promote a return to latency in these transiently activated reservoir cells. Notably, we demonstrate that TRIM32, an E3 ubiquitin ligase, promotes reactivation from latency by directly modifying IκBα, leading to a novel mechanism of NF-κB induction not involving IκB kinase activation.


Subject(s)
HIV-1/physiology , MicroRNAs/metabolism , Transcription Factors/metabolism , Virus Latency , 3' Untranslated Regions , Amino Acid Motifs , Base Sequence , CD4-Positive T-Lymphocytes/virology , Cell Death , Gene Silencing , Genes, Reporter , Humans , I-kappa B Proteins/metabolism , Lentivirus/metabolism , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Sequence Homology, Nucleic Acid , Tripartite Motif Proteins , Ubiquitin/chemistry , Ubiquitin-Protein Ligases/metabolism , Virus Replication
6.
Virology ; 446(1-2): 283-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074592

ABSTRACT

HIV latency constitutes the main barrier for clearing HIV infection from patients. Our inability to recognize and isolate latently infected cells hinders the study of latent HIV. We engineered two HIV-based viral reporters expressing different fluorescent markers: one HIV promoter-dependent marker for productive HIV infection, and a second marker under a constitutive promoter independent of HIV promoter activity. Infection of cells with these viruses allows the identification and separation of latently infected cells from uninfected and productively infected cells. These reporters are sufficiently sensitive and robust for high-throughput screening to identify drugs that reactivate latent HIV. These reporters can be used in primary CD4 T lymphocytes and reveal a rare population of latently infected cells responsive to physiological stimuli. In summary, our HIV-1 reporters enable visualization and purification of latent-cell populations and open up new perspectives for studies of latent HIV infection.


Subject(s)
HIV Infections/pathology , HIV Infections/virology , HIV-1/isolation & purification , HIV-1/physiology , Staining and Labeling/methods , Virology/methods , Virus Latency , CD4-Positive T-Lymphocytes/virology , HIV-1/genetics , Humans , Virus Activation/drug effects
7.
Trends Microbiol ; 21(6): 277-85, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23517573

ABSTRACT

Latent HIV persists in CD4(+) T cells in infected patients under antiretroviral therapy (ART). Latency is associated with transcriptional silencing of the integrated provirus and driven, at least in part, by histone deacetylases (HDACs), a family of chromatin-associated proteins that regulate histone acetylation and the accessibility of DNA to transcription factors. Remarkably, inhibition of HDACs is sufficient to reactivate a fraction of latent HIV in a variety of experimental systems. This basic observation led to the shock and kill idea that forcing the transcriptional activation of HIV might lead to virus expression, to virus- or host-induced cell death of the reactivated cells, and to the eradication of the pool of latently infected cells. Such intervention might possibly lead to a cure for HIV-infected patients. Here, we review the basic biology of HDACs and their inhibitors, the role of HDACs in HIV latency, and recent efforts to use HDAC inhibitors to reactivate latent HIV in vitro and in vivo.


Subject(s)
HIV-1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Virus Activation/drug effects , Virus Latency/drug effects , HIV Infections/virology , HIV-1/enzymology , HIV-1/physiology , Histone Deacetylase Inhibitors/chemistry , Humans , T-Lymphocytes/virology
8.
FEMS Microbiol Rev ; 36(3): 706-16, 2012 May.
Article in English | MEDLINE | ID: mdl-22372374

ABSTRACT

Highly active antiretroviral therapy (HAART) has shown great efficacy in increasing the survival of HIV infected individuals. However, HAART does not lead to the full eradication of infection and therefore has to be continued for life. HIV persists in a transcriptionally inactive form in resting T cells in HAART-treated patients and can be reactivated following T-cell activation. These latently infected cells allow the virus to persist in the presence of HAART. Here, we review recent advances in the study of the molecular mechanisms of HIV latency. We also review experimental models in which latency is currently studied. We focus on the epigenetic mechanisms controlling HIV transcription and on the role of chromatin and its post-translational modifications. We discuss how small molecule inhibitors that target epigenetic regulators, such as HDAC (histone deacetylase) inhibitors, are being tested for their ability to reactivate latent HIV. Finally, we discuss the clinical potential of these drugs to flush out latently infected cells from HIV-infected patients and to eradicate the virus.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV/immunology , HIV/pathogenicity , Host-Pathogen Interactions , T-Lymphocytes/virology , Virus Latency , Gene Expression Regulation, Viral , Humans , Models, Molecular , Virus Activation
9.
Curr Opin HIV AIDS ; 6(1): 19-24, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21242889

ABSTRACT

PURPOSE OF REVIEW: A reservoir of latently infected cells remains in HIV-infected patients treated with highly active antiretroviral therapy treatment. Persistence of HIV in this latent reservoir has prevented full viral eradication. In order to understand and develop rational therapeutics to flush out HIV latency, the molecular mechanisms governing the phenomena of HIV latency need to be understood. Several mechanisms have been proposed to explain HIV latency. RECENT FINDINGS: Epigenetic regulation of the HIV promoter in the 5' long terminal repeat of HIV-1 via histone protein modifications and the presence of inhibitory nucleosomes play a critical role in the establishment, maintenance, and reactivation of HIV latency. Recent reports have shed further light on how HIV latency might be epigenetically regulated. In this review, we discuss how these recent reports broaden our understanding of how HIV latency is regulated. Here, we review how histone modifications and chromatin remodeling affect the transcriptional activity of the HIV promoter in the context of HIV latency. SUMMARY: These new epigenetic regulators of HIV latency pose as potential interesting candidates for therapeutics against HIV latency.


Subject(s)
Epigenesis, Genetic , HIV Infections/virology , HIV-1/pathogenicity , Host-Pathogen Interactions , Virus Latency , Gene Expression Regulation, Viral , Humans
10.
Methods ; 53(1): 47-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20670606

ABSTRACT

HIV-1 latency is a barrier to overcome in the effort to fully eradicate the virus from infected individuals using highly active anti-retroviral therapy (HAART). Therefore, the study of the mechanisms underlying the establishment and maintenance of HIV-1 latency are vital to achieving a cure. Transcriptional repression of the viral promoter is the major cause of HIV-1 latency. DNA methylation of genomic regions known as CpG islands (CpGIs) is a well-established transcriptional regulatory mechanism, and the HIV-1 provirus contains several conserved CpGIs including two that are located within the viral promoter region. The study of these CpGIs in both in vitro and in vivo models of HIV-1 latency using the technique of bisulfite-mediated methylcytosine mapping has led to their identification as factors that contribute to the maintenance of HIV-1 latency. Here, we discuss the identification of CpGIs within the HIV-1 provirus and the study of their differential methylation patterns in several HIV-1 latency models using bisulfite-mediated methylcytosine mapping.


Subject(s)
Genome, Viral , HIV Infections/genetics , HIV-1/physiology , Proviruses/physiology , 5-Methylcytosine , Base Sequence , Cloning, Molecular , Computer Simulation , CpG Islands , DNA Methylation , DNA, Viral/metabolism , HIV-1/genetics , Humans , Models, Genetic , Proviruses/genetics , Sulfites , Virus Integration , Virus Latency
11.
Virology ; 351(1): 188-95, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16647096

ABSTRACT

We have solved the NMR solution structure of domain III from the Omsk hemorrhagic fever virus envelope protein and report the first sequencing of the Guriev strain of this virus. Important structural differences between tick-borne flaviviruses, such as OHFV and TBE, and mosquito-borne flaviviruses, such as West Nile virus, are discussed.


Subject(s)
Encephalitis Viruses, Tick-Borne/chemistry , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary
12.
J Virol ; 79(12): 7396-401, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15919895

ABSTRACT

The Kaposi's sarcoma-associated herpesvirus (KSHV) SOX protein, encoded by ORF37, promotes shutoff of host cell gene expression during lytic viral replication by dramatically impairing mRNA accumulation. SOX is the KSHV homolog of the alkaline exonuclease of other herpesviruses, which has been shown to function as a DNase involved in processing and packaging the viral genome. Although the exonuclease activity of these proteins is widely conserved across all herpesviruses, the host shutoff activity observed for KSHV SOX is not. We show here that SOX expression sharply reduces the half-life of target mRNAs. Extensive mutational analysis reveals that the DNase and host shutoff activities of SOX are genetically separable. Lesions affecting the DNase activity cluster in conserved regions of the protein, but residues critical for mRNA degradation are not conserved across the viral family. Additionally, we present evidence suggesting that the two different functions of SOX occur within distinct cellular compartments-DNase activity in the nucleus and host shutoff activity in the cytoplasm.


Subject(s)
Exonucleases , Herpesvirus 8, Human/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Cell Line , Exonucleases/genetics , Exonucleases/metabolism , Gene Expression Regulation , Herpesvirus 8, Human/genetics , Humans , Mutation , Open Reading Frames/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleases , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...