Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 05.
Article in English | MEDLINE | ID: mdl-38746145

ABSTRACT

Signal-induced transcriptional programs regulate critical biological processes through the precise spatiotemporal activation of Immediate Early Genes (IEGs); however, the mechanisms of transcription induction remain poorly understood. By combining an acute depletion system with high resolution genomics approaches to interrogate synchronized, temporal transcription, we reveal that KAP1/TRIM28 is a first responder that fulfills the temporal and heightened transcriptional demand of IEGs. Unexpectedly, acute KAP1 loss triggers an increase in RNA polymerase II elongation kinetics during early stimulation time points. This elongation defect derails the normal progression through the transcriptional cycle during late stimulation time points, ultimately leading to decreased recruitment of the transcription apparatus for re-initiation thereby dampening IEGs transcriptional output. Collectively, KAP1 plays a counterintuitive role by negatively regulating transcription elongation to support full activation across multiple transcription cycles of genes critical for cell physiology and organismal functions.

2.
Viruses ; 16(2)2024 02 04.
Article in English | MEDLINE | ID: mdl-38400024

ABSTRACT

At every integrated HIV-1 genome, there is a transcriptional cycle that ultimately shapes proviral fate [...].


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , HIV-1/genetics , Virus Latency/genetics , Proviruses/genetics , Transcription, Genetic
3.
Viruses ; 16(2)2024 02 13.
Article in English | MEDLINE | ID: mdl-38400062

ABSTRACT

HIV-1 latency remains a barrier to a functional cure because of the ability of virtually silent yet inducible proviruses within reservoir cells to transcriptionally reactivate upon cell stimulation. HIV-1 reactivation occurs through the sequential action of host transcription factors (TFs) during the "host phase" and the viral TF Tat during the "viral phase", which together facilitate the positive feedback loop required for exponential transcription, replication, and pathogenesis. The sequential action of these TFs poses a challenge to precisely delineate the contributions of the host and viral phases of the transcriptional program to guide future mechanistic and therapeutic studies. To address this limitation, we devised a genome engineering approach to mutate tat and create a genetically matched pair of Jurkat T cell clones harboring HIV-1 at the same integration site with and without Tat expression. By comparing the transcriptional profile of both clones, the transition point between the host and viral phases was defined, providing a system that enables the temporal mechanistic interrogation of HIV-1 transcription prior to and after Tat synthesis. Importantly, this CRISPR method is broadly applicable to knockout individual viral proteins or genomic regulatory elements to delineate their contributions to various aspects of the viral life cycle and ultimately may facilitate therapeutic approaches in our race towards achieving a functional cure.


Subject(s)
HIV Infections , HIV-1 , Humans , Proviruses/genetics , CRISPR-Cas Systems , HIV-1/genetics , Virus Latency/genetics
4.
Viruses ; 16(1)2024 01 13.
Article in English | MEDLINE | ID: mdl-38257816

ABSTRACT

HIV-1 latency maintenance and reactivation are regulated by several viral and host factors. One such factor is Krüppel-associated box (KRAB)-associated protein 1 (KAP1: also named TRIM28 or TIF1ß). While initial studies have revealed KAP1 to be a positive regulator of latency reversal in transformed and primary CD4+ T cells, subsequent studies have proposed KAP1 to be a repressor required for latency maintenance. Given this discrepancy, in this study, we re-examine KAP1 transcription regulatory functions using a chemical genetics strategy to acutely deplete KAP1 expression to avoid the accumulation of indirect effects. Notably, KAP1 acute loss partially decreased HIV-1 promoter activity in response to activating signals, a function that can be restored upon complementation with exogenous KAP1, thus revealing that KAP1-mediated activation is on target. By combining comprehensive KAP1 domain deletion and mutagenesis in a cell-based reporter assay, we genetically defined the RING finger domain and an Intrinsically Disordered Region as key activating features. Together, our study solidifies the notion that KAP1 activates HIV-1 transcription by exploiting its multi-domain protein arrangement via previously unknown domains and functions.


Subject(s)
HIV-1 , Transcriptional Activation , Tripartite Motif-Containing Protein 28 , Humans , HIV-1/genetics , Mutagenesis , RING Finger Domains , Tripartite Motif-Containing Protein 28/genetics
5.
Viruses ; 15(10)2023 10 19.
Article in English | MEDLINE | ID: mdl-37896896

ABSTRACT

HIV-1 latency is a major barrier to curing infections with antiretroviral therapy and, consequently, to eliminating the disease globally. The establishment, maintenance, and potential clearance of latent infection are complex dynamic processes and can be best described with the help of mathematical models followed by experimental validation. Here, we review the use of viral dynamics models for HIV-1, with a focus on applications to the latent reservoir. Such models have been used to explain the multi-phasic decay of viral load during antiretroviral therapy, the early seeding of the latent reservoir during acute infection and the limited inflow during treatment, the dynamics of viral blips, and the phenomenon of post-treatment control. Finally, we discuss that mathematical models have been used to predict the efficacy of potential HIV-1 cure strategies, such as latency-reversing agents, early treatment initiation, or gene therapies, and to provide guidance for designing trials of these novel interventions.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , HIV-1/genetics , Virus Latency , Models, Biological , Models, Theoretical , HIV Infections/drug therapy , CD4-Positive T-Lymphocytes
6.
Nat Commun ; 13(1): 7447, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36460671

ABSTRACT

Control of RNA Polymerase II (pol II) elongation is a critical component of gene expression in mammalian cells. The PNUTS-PP1 complex controls elongation rates, slowing pol II after polyadenylation sites to promote termination. The Kaposi's sarcoma-associated herpesvirus (KSHV) co-opts pol II to express its genes, but little is known about its regulation of pol II elongation. We identified PNUTS as a suppressor of a KSHV reporter gene in a genome-wide CRISPR screen. PNUTS depletion enhances global KSHV gene expression and overall viral replication. Mechanistically, PNUTS requires PP1 interaction, binds viral RNAs downstream of polyadenylation sites, and restricts transcription readthrough of viral genes. Surprisingly, PNUTS also represses productive elongation at the 5´ ends of the KSHV reporter and the KSHV T1.4 RNA. From these data, we conclude that PNUTS' activity constitutes an intrinsic barrier to KSHV replication likely by suppressing pol II elongation at promoter-proximal regions.


Subject(s)
Herpesviridae , Herpesvirus 8, Human , Multiple Myeloma , Sarcoma, Kaposi , Animals , Herpesvirus 8, Human/genetics , Nucleotidyltransferases , RNA, Viral/genetics , Transcription, Genetic , Mammals
7.
Viruses ; 14(10)2022 10 18.
Article in English | MEDLINE | ID: mdl-36298843

ABSTRACT

Immune cell state alterations rewire HIV-1 gene expression, thereby influencing viral latency and reactivation, but the mechanisms are still unfolding. Here, using a screen approach on CD4+ T cell models of HIV-1 latency, we revealed Small Molecule Reactivators (SMOREs) with unique chemistries altering the CD4+ T cell state and consequently promoting latent HIV-1 transcription and reactivation through an unprecedented mechanism of action. SMOREs triggered rapid oxidative stress and activated a redox-responsive program composed of cell-signaling kinases (MEK-ERK axis) and atypical transcription factor (AP-1 and HIF-1α) cooperativity. SMOREs induced an unusual AP-1 phosphorylation signature to promote AP-1/HIF-1α binding to the latent HIV-1 proviral genome for its activation. Consistently, latent HIV-1 reactivation was compromised with pharmacologic inhibition of oxidative stress sensing or of cell-signaling kinases, and transcription factor's loss of expression, thus functionally linking the host redox-responsive program to viral transcriptional rewiring. Notably, SMOREs induced the redox program in primary CD4+ T cells and reactivated latent HIV-1 in aviremic patient samples alone and in combination with known latency-reversing agents, thus providing physiological relevance. Our findings suggest that manipulation of redox-sensitive pathways could be exploited to alter the course of HIV-1 latency, thus rendering host cells responsive to help achieve a sterilizing cure.


Subject(s)
HIV Infections , HIV-1 , Transcription Factor AP-1 , Virus Activation , Virus Latency , Humans , CD4-Positive T-Lymphocytes , HIV Infections/genetics , HIV Infections/immunology , HIV Seropositivity/genetics , HIV Seropositivity/immunology , HIV-1/genetics , HIV-1/immunology , Jurkat Cells , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/immunology , Oxidation-Reduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Virus Activation/genetics , Virus Activation/immunology , Virus Latency/genetics , Virus Latency/immunology
8.
Nat Commun ; 13(1): 1109, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35232997

ABSTRACT

Immune stimulation fuels cell signaling-transcriptional programs inducing biological responses to eliminate virus-infected cells. Yet, retroviruses that integrate into host cell chromatin, such as HIV-1, co-opt these programs to switch between latent and reactivated states; however, the regulatory mechanisms are still unfolding. Here, we implemented a functional screen leveraging HIV-1's dependence on CD4+ T cell signaling-transcriptional programs and discovered ADAP1 is an undescribed modulator of HIV-1 proviral fate. Specifically, we report ADAP1 (ArfGAP with dual PH domain-containing protein 1), a previously thought neuronal-restricted factor, is an amplifier of select T cell signaling programs. Using complementary biochemical and cellular assays, we demonstrate ADAP1 inducibly interacts with the immune signalosome to directly stimulate KRAS GTPase activity thereby augmenting T cell signaling through targeted activation of the ERK-AP-1 axis. Single cell transcriptomics analysis revealed loss of ADAP1 function blunts gene programs upon T cell stimulation consequently dampening latent HIV-1 reactivation. Our combined experimental approach defines ADAP1 as an unexpected tuner of T cell programs facilitating HIV-1 latency escape.


Subject(s)
Adaptor Proteins, Signal Transducing , HIV Infections , HIV-1 , MAP Kinase Signaling System , Nerve Tissue Proteins , Proto-Oncogene Proteins p21(ras) , T-Lymphocytes , Transcription Factor AP-1 , Adaptor Proteins, Signal Transducing/metabolism , CD4-Positive T-Lymphocytes , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Humans , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Transcription Factor AP-1/metabolism , Virus Activation , Virus Latency
9.
Bioinform Biol Insights ; 16: 11779322211072333, 2022.
Article in English | MEDLINE | ID: mdl-35250265

ABSTRACT

Fundamental principles of HIV-1 integration into the human genome have been revealed in the past 2 decades. However, the impact of the integration site on proviral transcription and expression remains poorly understood. Solving this problem requires the analysis of multiple genomic datasets for thousands of proviral integration sites. Here, we generated and combined large-scale datasets, including epigenetics, transcriptome, and 3-dimensional genome architecture to interrogate the chromatin states, transcription activity, and nuclear sub-compartments around HIV-1 integrations in Jurkat CD4+ T cells to decipher human genome regulatory features shaping the transcription of proviral classes based on their position and orientation in the genome. Through a Hidden Markov Model and ranked informative values prior to a machine learning logistic regression model, we defined nuclear sub-compartments and chromatin states contributing to genomic architecture, transcriptional activity, and nucleosome density of regions neighboring the integration site, as additive features influencing HIV-1 expression. Our integrated genomics approach also allows for a robust experimental design, in which HIV-1 can be genetically introduced into precise genomic locations with known regulatory features to assess the relationship of integration positions to viral transcription and fate.

10.
Front Cell Infect Microbiol ; 12: 834636, 2022.
Article in English | MEDLINE | ID: mdl-35281453

ABSTRACT

Several transcriptional and epigenetic regulators have been functionally linked to the control of viral and cellular gene expression programs. One such regulator is Krüppel-associated box (KRAB)-associated protein 1 (KAP1: also named TRIM28 or TIF1ß), which has been extensively studied in the past three decades. Here we offer an up-to date review of its various functions in a diversity of contexts. We first summarize the discovery of KAP1 repression of endogenous retroviruses during development. We then deliberate evidence in the literature suggesting KAP1 is both an activator and repressor of HIV-1 transcription and discuss experimental differences and limitations of previous studies. Finally, we discuss KAP1 regulation of DNA and RNA viruses, and then expand on KAP1 control of cellular responses and immune functions. While KAP1 positive and negative regulation of viral and cellular transcriptional programs is vastly documented, our mechanistic understanding remains narrow. We thus propose that precision genetic tools to reveal direct KAP1 functions in gene regulation will be required to not only illuminate new biology but also provide the foundation to translate the basic discoveries from the bench to the clinics.


Subject(s)
Repressor Proteins , Transcription Factors , Cell Line , Gene Expression Regulation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
11.
Mol Cell ; 81(14): 2871-2872, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34270942

ABSTRACT

In a tour de force, Skalska et al. (2021) discover transcription inhibition and RNA degradation elicit recruitment of chromatin modifiers and transcriptional regulators to chromatin, suggesting a broad role for nascent RNA as factor-chromatin antagonizer.


Subject(s)
Chromatin , RNA , Chromatin/genetics , RNA/genetics , Transcription, Genetic
12.
mBio ; 12(3): e0109821, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34154414

ABSTRACT

The HIV-1 latent reservoir is the major barrier to an HIV cure. Due to low levels or lack of transcriptional activity, HIV-1 latent proviruses in vivo are not easily detectable and cannot be targeted by either natural immune mechanisms or molecular therapies based on protein expression. To target the latent reservoir, further understanding of HIV-1 proviral transcription is required. In this study, we demonstrate a novel role for cleavage and polyadenylation specificity factor 6 (CPSF6) in HIV-1 transcription. We show that knockout of CPSF6 hinders reactivation of latent HIV-1 proviruses by PMA in primary CD4+ cells. CPSF6 knockout reduced HIV-1 transcription, concomitant with a drastic reduction in the phosphorylation levels of Pol II and CDK9. Knockout of CPSF6 led to abnormal stabilization of protein phosphatase 2A (PP2A) subunit A, which then acted to dephosphorylate CDK9, downmodulating CDK9's ability to phosphorylate the Pol II carboxy-terminal domain. In agreement with this mechanism, incubation with the PP2A inhibitor, LB100, restored HIV-1 transcription in the CPSF6 knockout cells. Destabilization of PP2A subunit A occurs in the ubiquitin proteasome pathway, wherein CPSF6 acts as a substrate adaptor for the ITCH ubiquitin ligase. Our observations reveal a novel role of CPSF6 in HIV-1 transcription, which appears to be independent of its known roles in cleavage and polyadenylation and the targeting of preintegration complexes to the chromatin for viral DNA integration. IMPORTANCE CPSF6 is a cellular factor that regulates cleavage and polyadenylation of mRNAs and participates in HIV-1 infection by facilitating targeting of preintegration complexes to the chromatin. Our observations reveal a second role of CPSF6 in the HIV-1 life cycle that involves regulation of viral transcription through controlling the stability of protein phosphatase 2A, which in turn regulates the phosphorylation/dephosphorylation status of critical residues in CDK9 and Pol II.


Subject(s)
HIV-1/genetics , HIV-1/metabolism , Virus Latency/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Disease Reservoirs/virology , HEK293 Cells , Humans , Monocytes/virology , Phosphorylation , Proviruses/genetics , Proviruses/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Virus Integration , Virus Latency/physiology , Virus Replication
13.
Proc Natl Acad Sci U S A ; 117(51): 32594-32605, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288725

ABSTRACT

Inducible transcriptional programs mediate the regulation of key biological processes and organismal functions. Despite their complexity, cells have evolved mechanisms to precisely control gene programs in response to environmental cues to regulate cell fate and maintain normal homeostasis. Upon stimulation with proinflammatory cytokines such as tumor necrosis factor-α (TNF), the master transcriptional regulator nuclear factor (NF)-κB utilizes the PPM1G/PP2Cγ phosphatase as a coactivator to normally induce inflammatory and cell survival programs. However, how PPM1G activity is precisely regulated to control NF-κB transcription magnitude and kinetics remains unknown. Here, we describe a mechanism by which the ARF tumor suppressor binds PPM1G to negatively regulate its coactivator function in the NF-κB circuit thereby promoting insult resolution. ARF becomes stabilized upon binding to PPM1G and forms a ternary protein complex with PPM1G and NF-κB at target gene promoters in a stimuli-dependent manner to provide tunable control of the NF-κB transcriptional program. Consistently, loss of ARF in colon epithelial cells leads to up-regulation of NF-κB antiapoptotic genes upon TNF stimulation and renders cells partially resistant to TNF-induced apoptosis in the presence of agents blocking the antiapoptotic program. Notably, patient tumor data analysis validates these findings by revealing that loss of ARF strongly correlates with sustained expression of inflammatory and cell survival programs. Collectively, we propose that PPM1G emerges as a therapeutic target in a variety of cancers arising from ARF epigenetic silencing, to loss of ARF function, as well as tumors bearing oncogenic NF-κB activation.


Subject(s)
Inflammation/metabolism , NF-kappa B/genetics , Neoplasms/metabolism , Protein Phosphatase 2C/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Epithelial Cells/pathology , Humans , Inflammation/genetics , Multiprotein Complexes , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/pathology , Promoter Regions, Genetic , Protein Domains , Protein Interaction Maps , Protein Phosphatase 2C/chemistry , Protein Phosphatase 2C/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Protein p14ARF/genetics
14.
Mol Cell ; 78(6): 1133-1151.e14, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32402252

ABSTRACT

Precise control of the RNA polymerase II (RNA Pol II) cycle, including pausing and pause release, maintains transcriptional homeostasis and organismal functions. Despite previous work to understand individual transcription steps, we reveal a mechanism that integrates RNA Pol II cycle transitions. Surprisingly, KAP1/TRIM28 uses a previously uncharacterized chromatin reader cassette to bind hypo-acetylated histone 4 tails at promoters, guaranteeing continuous progression of RNA Pol II entry to and exit from the pause state. Upon chromatin docking, KAP1 first associates with RNA Pol II and then recruits a pathway-specific transcription factor (SMAD2) in response to cognate ligands, enabling gene-selective CDK9-dependent pause release. This coupling mechanism is exploited by tumor cells to aberrantly sustain transcriptional programs commonly dysregulated in cancer patients. The discovery of a factor integrating transcription steps expands the functional repertoire by which chromatin readers operate and provides mechanistic understanding of transcription regulation, offering alternative therapeutic opportunities to target transcriptional dysregulation.


Subject(s)
RNA Polymerase II/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Acetylation , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation/genetics , Histones/metabolism , Humans , Oncogenes/genetics , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/genetics , RNA Polymerase II/genetics , Smad2 Protein/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Tripartite Motif-Containing Protein 28/genetics
15.
Viruses ; 12(5)2020 05 18.
Article in English | MEDLINE | ID: mdl-32443452

ABSTRACT

Three decades of extensive work in the HIV field have revealed key viral and host cell factors controlling proviral transcription. Various models of transcriptional regulation have emerged based on the collective information from in vitro assays and work in both immortalized and primary cell-based models. Here, we provide a recount of the past and current literature, highlight key regulatory aspects, and further describe potential limitations of previous studies. We particularly delve into critical steps of HIV gene expression including the role of the integration site, nucleosome positioning and epigenomics, and the transition from initiation to pausing and pause release. We also discuss open questions in the field concerning the generality of previous regulatory models to the control of HIV transcription in patients under suppressive therapy, including the role of the heterogeneous integration landscape, clonal expansion, and bottlenecks to eradicate viral persistence. Finally, we propose that building upon previous discoveries and improved or yet-to-be discovered technologies will unravel molecular mechanisms of latency establishment and reactivation in a "new era".


Subject(s)
HIV Infections/virology , HIV-1/genetics , Proviruses/genetics , Transcription, Genetic/genetics , Virus Latency/genetics , Anti-Retroviral Agents/therapeutic use , Epigenesis, Genetic , Gene Expression Regulation, Viral , HIV Infections/drug therapy , HIV-1/physiology , Humans , Virus Integration
16.
Cell Rep ; 27(1): 154-171.e9, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943398

ABSTRACT

Transcriptional circuit architectures in several organisms have been evolutionarily selected to dictate precise given responses. Unlike these cellular systems, HIV is regulated through a complex circuit composed of two successive phases (host and viral), which create a positive feedback loop facilitating viral replication. However, it has long remained unclear whether both phases operate identically and to what extent the host phase influences the entire circuit. Here, we report that, although the host phase is regulated by a checkpoint whereby KAP1 mediates transcription activation, the virus evolved a minimalist system bypassing KAP1. Given the complex circuit's architecture, cell-to-cell KAP1 fluctuations impart heterogeneity in the host transcriptional responses, thus affecting the feedback loop. Mathematical modeling of a complete circuit reveals how these oscillations ultimately influence homogeneous reactivation potential of a latent virus. Thus, although HIV drives molecular innovation to fuel robust gene activation, it experiences transcriptional fragility, thereby influencing viral fate and cure efforts.


Subject(s)
Gene Regulatory Networks/physiology , HIV Infections/virology , HIV-1/genetics , Proviruses , Virus Activation/genetics , Virus Latency/genetics , Cells, Cultured , Gene Expression Regulation, Viral , Genome, Viral , Genomic Instability/physiology , HEK293 Cells , HIV Infections/genetics , HIV-1/physiology , Humans , Jurkat Cells , Proviruses/genetics , Proviruses/physiology , Transcription, Genetic , Virus Replication/genetics
17.
Transcription ; 10(2): 57-75, 2019 04.
Article in English | MEDLINE | ID: mdl-30227759

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) is critical for RNA Polymerase II (Pol II) transcription initiation, elongation, and termination in several key biological processes including development, differentiation, and cell fate responses. A broad range of diseases are characterized by CDK9 malfunction, illustrating its importance in maintaining transcriptional homeostasis in basal- and signal-regulated conditions. Here we provide a historical recount of CDK9 discovery and the current models suggesting CDK9 is a central hub necessary for proper execution of different steps in the transcription cycle. Finally, we discuss the current therapeutic strategies to treat CDK9 malfunction in several disease states. Abbreviations: CDK: Cyclin-dependent kinase; Pol II: RNA Polymerase II; PIC: Pre-initiation Complex; TFIIH: Transcription Factor-II H; snoRNA: small nucleolar RNA; CycT: CyclinT1/T2; P-TEFb: Positive Transcription Elongation Factor Complex; snRNP: small nuclear ribonucleo-protein; HEXIM: Hexamethylene Bis-acetamide-inducible Protein 1/2; LARP7: La-related Protein 7; MePCE: Methylphosphate Capping Enzyme; HIV: human immunodeficiency virus; TAT: trans-activator of transcription; TAR: Trans-activation response element; Hsp70: Heat Shock Protein 70; Hsp90/Cdc37: Hsp90- Hsp90 co-chaperone Cdc37; DSIF: DRB Sensitivity Inducing Factor; NELF: Negative Elongation Factor; CPSF: cleavage and polyadenylation-specific factor; CSTF: cleavage-stimulatory factor; eRNA: enhancer RNA; BRD4: Bromodomain-containing protein 4; JMJD6: Jumonji C-domain-containing protein 6; SEC: Super Elongation Complex; ELL: eleven-nineteen Lys-rich leukemia; ENL: eleven-nineteen leukemia; MLL: mixed lineage leukemia; BEC: BRD4-containing Elongation Complex; SEC-L2/L3: SEC-like complexes; KAP1: Kruppel-associated box-protein 1; KEC: KAP1-7SK Elongation Complex; DRB: Dichloro-1-ß-D-Ribofuranosylbenzimidazole; H2Bub1: H2B mono-ubiquitination; KM: KM05382; PP1: Protein Phosphatase 1; CDK9i: CDK9 inhibitor; SHAPE: Selective 2'-hydroxyl acylation analyzed by primer extension; TE: Typical enhancer; SE : Super enhancer.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Signal Transduction/genetics , Transcription, Genetic/genetics , Humans
18.
Elife ; 72018 05 30.
Article in English | MEDLINE | ID: mdl-29845934

ABSTRACT

The HIV-1 Tat protein hijacks P-TEFb kinase to activate paused RNA polymerase II (RNAP II) at the viral promoter. Tat binds additional host factors, but it is unclear how they regulate RNAP II elongation. Here, we identify the cytoplasmic ubiquitin ligase UBE2O as critical for Tat transcriptional activity. Tat hijacks UBE2O to ubiquitinate the P-TEFb kinase inhibitor HEXIM1 of the 7SK snRNP, a fraction of which also resides in the cytoplasm bound to P-TEFb. HEXIM1 ubiquitination sequesters it in the cytoplasm and releases P-TEFb from the inhibitory 7SK complex. Free P-TEFb then becomes enriched in chromatin, a process that is also stimulated by treating cells with a CDK9 inhibitor. Finally, we demonstrate that UBE2O is critical for P-TEFb recruitment to the HIV-1 promoter. Together, the data support a unique model of elongation control where non-degradative ubiquitination of nuclear and cytoplasmic 7SK snRNP pools increases P-TEFb levels for transcriptional activation.


Subject(s)
HIV-1/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Transcriptional Activation/genetics , Ubiquitin-Conjugating Enzymes/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin/metabolism , Dichlororibofuranosylbenzimidazole/pharmacology , HEK293 Cells , HeLa Cells , Humans , Models, Biological , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , RNA Interference , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Transcription Factors , Transcriptional Activation/drug effects , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitination/drug effects
19.
BMC Bioinformatics ; 18(1): 363, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28789639

ABSTRACT

BACKGROUND: Next-generation sequencing (NGS) approaches are commonly used to identify key regulatory networks that drive transcriptional programs. Although these technologies are frequently used in biological studies, NGS data analysis remains a challenging, time-consuming, and often irreproducible process. Therefore, there is a need for a comprehensive and flexible workflow platform that can accelerate data processing and analysis so more time can be spent on functional studies. RESULTS: We have developed an integrative, stand-alone workflow platform, named CIPHER, for the systematic analysis of several commonly used NGS datasets including ChIP-seq, RNA-seq, MNase-seq, DNase-seq, GRO-seq, and ATAC-seq data. CIPHER implements various open source software packages, in-house scripts, and Docker containers to analyze and process single-ended and pair-ended datasets. CIPHER's pipelines conduct extensive quality and contamination control checks, as well as comprehensive downstream analysis. A typical CIPHER workflow includes: (1) raw sequence evaluation, (2) read trimming and adapter removal, (3) read mapping and quality filtering, (4) visualization track generation, and (5) extensive quality control assessment. Furthermore, CIPHER conducts downstream analysis such as: narrow and broad peak calling, peak annotation, and motif identification for ChIP-seq, differential gene expression analysis for RNA-seq, nucleosome positioning for MNase-seq, DNase hypersensitive site mapping, site annotation and motif identification for DNase-seq, analysis of nascent transcription from Global-Run On (GRO-seq) data, and characterization of chromatin accessibility from ATAC-seq datasets. In addition, CIPHER contains an "analysis" mode that completes complex bioinformatics tasks such as enhancer discovery and provides functions to integrate various datasets together. CONCLUSIONS: Using public and simulated data, we demonstrate that CIPHER is an efficient and comprehensive workflow platform that can analyze several NGS datasets commonly used in genome biology studies. Additionally, CIPHER's integrative "analysis" mode allows researchers to elicit important biological information from the combined dataset analysis.


Subject(s)
High-Throughput Nucleotide Sequencing , Regulatory Sequences, Nucleic Acid/genetics , Software , Chromatin Immunoprecipitation , Chromosome Mapping , Databases, Genetic , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Sequence Analysis, RNA
20.
J Vis Exp ; (119)2017 01 26.
Article in English | MEDLINE | ID: mdl-28190026

ABSTRACT

The purification of active protein-protein and protein-nucleic acid complexes is crucial for the characterization of enzymatic activities and de novo identification of novel subunits and post-translational modifications. Bacterial systems allow for the expression and purification of a wide variety of single polypeptides and protein complexes. However, this system does not enable the purification of protein subunits that contain post-translational modifications (e.g., phosphorylation and acetylation), and the identification of novel regulatory subunits that are only present/expressed in the eukaryotic system. Here, we provide a detailed description of a novel, robust, and efficient tandem affinity purification (TAP) method using STREP- and FLAG-tagged proteins that facilitates the purification of protein complexes with transiently or stably expressed epitope-tagged proteins from eukaryotic cells. This protocol can be applied to characterize protein complex functionality, to discover post-translational modifications on complex subunits, and to identify novel regulatory complex components by mass spectrometry. Notably, this TAP method can be applied to study protein complexes formed by eukaryotic or pathogenic (viral and bacterial) components, thus yielding a wide array of downstream experimental opportunities. We propose that researchers working with protein complexes could utilize this approach in many different ways.


Subject(s)
Eukaryotic Cells/chemistry , Multiprotein Complexes/isolation & purification , Proteomics/methods , Chromatography, Affinity/methods , Cyclin T/genetics , Cyclin T/isolation & purification , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/isolation & purification , Epitopes/chemistry , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Immunoprecipitation/methods , Mass Spectrometry/methods , Multiprotein Complexes/chemistry , Positive Transcriptional Elongation Factor B/isolation & purification , Protein Interaction Maps , Protein Processing, Post-Translational , Proteins/chemistry , Proteins/isolation & purification , Proteomics/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...