Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(12): 3143-3162.e32, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34004147

ABSTRACT

Gene expression by RNA polymerase II (RNAPII) is tightly controlled by cyclin-dependent kinases (CDKs) at discrete checkpoints during the transcription cycle. The pausing checkpoint following transcription initiation is primarily controlled by CDK9. We discovered that CDK9-mediated, RNAPII-driven transcription is functionally opposed by a protein phosphatase 2A (PP2A) complex that is recruited to transcription sites by the Integrator complex subunit INTS6. PP2A dynamically antagonizes phosphorylation of key CDK9 substrates including DSIF and RNAPII-CTD. Loss of INTS6 results in resistance to tumor cell death mediated by CDK9 inhibition, decreased turnover of CDK9 phospho-substrates, and amplification of acute oncogenic transcriptional responses. Pharmacological PP2A activation synergizes with CDK9 inhibition to kill both leukemic and solid tumor cells, providing therapeutic benefit in vivo. These data demonstrate that fine control of gene expression relies on the balance between kinase and phosphatase activity throughout the transcription cycle, a process dysregulated in cancer that can be exploited therapeutically.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Protein Phosphatase 2/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Phosphorylation , Protein Binding , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Substrate Specificity
2.
Cell ; 175(5): 1244-1258.e26, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30454645

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) promotes transcriptional elongation through RNAPII pause release. We now report that CDK9 is also essential for maintaining gene silencing at heterochromatic loci. Through a live cell drug screen with genetic confirmation, we discovered that CDK9 inhibition reactivates epigenetically silenced genes in cancer, leading to restored tumor suppressor gene expression, cell differentiation, and activation of endogenous retrovirus genes. CDK9 inhibition dephosphorylates the SWI/SNF protein BRG1, which contributes to gene reactivation. By optimization through gene expression, we developed a highly selective CDK9 inhibitor (MC180295, IC50 = 5 nM) that has broad anti-cancer activity in vitro and is effective in in vivo cancer models. Additionally, CDK9 inhibition sensitizes to the immune checkpoint inhibitor α-PD-1 in vivo, making it an excellent target for epigenetic therapy of cancer.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Methylation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Mol Cell ; 83(3): 393-403, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36599353

ABSTRACT

The positive transcription elongation factor b (P-TEFb) is composed of cyclins T1 or T2 and cyclin-dependent kinase 9 that regulate the elongation phase of transcription by RNA polymerase II. By antagonizing negative elongation factors and phosphorylating the C-terminal domain of RNA polymerase II, P-TEFb facilitates the elongation and co-transcriptional processing of nascent transcripts. This step is critical for the expression of most eukaryotic genes. In growing cells, P-TEFb is regulated negatively by its reversible associations with HEXIM1/2 in the 7SK snRNP and positively by a number of transcription factors, as well as the super elongation complex. In resting cells, P-TEFb falls apart, and cyclin T1 is degraded by the proteasome. This complex regulation of P-TEFb has evolved for the precise temporal and spatial regulation of gene expression in the organism. Its dysregulation contributes to inflammatory and neoplastic conditions.


Subject(s)
Positive Transcriptional Elongation Factor B , RNA Polymerase II , Humans , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , HeLa Cells , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin T/genetics , Cyclin T/metabolism , Transcription, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Mol Cell ; 81(5): 1013-1026.e11, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33548202

ABSTRACT

In response to stress, human cells coordinately downregulate transcription and translation of housekeeping genes. To downregulate transcription, the negative elongation factor (NELF) is recruited to gene promoters impairing RNA polymerase II elongation. Here we report that NELF rapidly forms nuclear condensates upon stress in human cells. Condensate formation requires NELF dephosphorylation and SUMOylation induced by stress. The intrinsically disordered region (IDR) in NELFA is necessary for nuclear NELF condensation and can be functionally replaced by the IDR of FUS or EWSR1 protein. We find that biomolecular condensation facilitates enhanced recruitment of NELF to promoters upon stress to drive transcriptional downregulation. Importantly, NELF condensation is required for cellular viability under stressful conditions. We propose that stress-induced NELF condensates reported here are nuclear counterparts of cytosolic stress granules. These two stress-inducible condensates may drive the coordinated downregulation of transcription and translation, likely forming a critical node of the stress survival strategy.


Subject(s)
Heat-Shock Response/genetics , Intrinsically Disordered Proteins/genetics , Protein Processing, Post-Translational , RNA Polymerase II/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Chromatin/chemistry , Chromatin/metabolism , Clone Cells , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Signal Transduction , Stress, Physiological , Sumoylation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism , Red Fluorescent Protein
5.
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
6.
Proc Natl Acad Sci U S A ; 121(7): e2307150121, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38315842

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) plays a critical role in transcription initiation and is essential for maintaining gene silencing at heterochromatic loci. Inhibition of CDK9 increases sensitivity to immunotherapy, but the underlying mechanism remains unclear. We now report that RNF20 stabilizes LSD1 via K29-mediated ubiquitination, which is dependent on CDK9-mediated phosphorylation. This CDK9- and RNF20-dependent LSD1 stabilization is necessary for the demethylation of histone H3K4, then subsequent repression of endogenous retrovirus, and an interferon response, leading to epigenetic immunosuppression. Moreover, we found that loss of RNF20 sensitizes cancer cells to the immune checkpoint inhibitor anti-PD-1 in vivo and that this effect can be rescued by the expression of ectopic LSD1. Our findings are supported by the observation that RNF20 levels correlate with LSD1 levels in human breast cancer specimens. This study sheds light on the role of RNF20 in CDK9-dependent LSD1 stabilization, which is crucial for epigenetic silencing and immunosuppression. Our findings explore the potential importance of targeting the CDK9-RNF20-LSD1 axis in the development of new cancer therapies.


Subject(s)
Cyclin-Dependent Kinase 9 , Histone Demethylases , Immune Tolerance , Ubiquitin-Protein Ligases , Humans , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Epigenesis, Genetic , Histone Demethylases/metabolism , Histones/metabolism , Ubiquitin-Protein Ligases/genetics
7.
Proc Natl Acad Sci U S A ; 121(15): e2321502121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38564636

ABSTRACT

The release of paused RNA polymerase II (RNAPII) from promoter-proximal regions is tightly controlled to ensure proper regulation of gene expression. The elongation factor PTEF-b is known to release paused RNAPII via phosphorylation of the RNAPII C-terminal domain by its cyclin-dependent kinase component, CDK9. However, the signal and stress-specific roles of the various RNAPII-associated macromolecular complexes containing PTEF-b/CDK9 are not yet clear. Here, we identify and characterize the CDK9 complex required for transcriptional response to hypoxia. Contrary to previous reports, our data indicate that a CDK9 complex containing BRD4 but not AFF1/4 is essential for this hypoxic stress response. We demonstrate that BRD4 bromodomains (BET) are dispensable for the release of paused RNAPII at hypoxia-activated genes and that BET inhibition by JQ1 is insufficient to impair hypoxic gene response. Mechanistically, we demonstrate that the C-terminal region of BRD4 is required for Polymerase-Associated Factor-1 Complex (PAF1C) recruitment to establish an elongation-competent RNAPII complex at hypoxia-responsive genes. PAF1C disruption using a small-molecule inhibitor (iPAF1C) impairs hypoxia-induced, BRD4-mediated RNAPII release. Together, our results provide insight into potentially targetable mechanisms that control the hypoxia-responsive transcriptional elongation.


Subject(s)
Nuclear Proteins , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Gene Expression Regulation , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Phosphorylation , Hypoxia , Transcription, Genetic , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Bromodomain Containing Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
8.
PLoS Genet ; 20(6): e1011308, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829886

ABSTRACT

Acute myeloid leukemia (AML) accounts for greater than twenty thousand new cases of leukemia annually in the United States. The average five-year survival rate is approximately 30%, pointing to the need for developing novel model systems for drug discovery. In particular, patients with chromosomal rearrangements in the mixed lineage leukemia (MLL) gene have higher relapse rates with poor outcomes. In this study we investigated the expression of human MLL-ENL and MLL-AF9 in the myeloid lineage of zebrafish embryos. We observed an expansion of MLL positive cells and determined these cells colocalized with the myeloid markers spi1b, mpx, and mpeg. In addition, expression of MLL-ENL and MLL-AF9 induced the expression of endogenous bcl2 and cdk9, genes that are often dysregulated in MLL-r-AML. Co-treatment of lyz: MLL-ENL or lyz:MLL-AF9 expressing embryos with the BCL2 inhibitor, Venetoclax, and the CDK9 inhibitor, Flavopiridol, significantly reduced the number of MLL positive cells compared to embryos treated with vehicle or either drug alone. In addition, cotreatment with Venetoclax and Flavopiridol significantly reduced the expression of endogenous mcl1a compared to vehicle, consistent with AML. This new model of MLL-r-AML provides a novel tool to understand the molecular mechanisms underlying disease progression and a platform for drug discovery.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Cyclin-Dependent Kinase 9 , Leukemia, Myeloid, Acute , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion , Proto-Oncogene Proteins c-bcl-2 , Zebrafish , Zebrafish/genetics , Zebrafish/embryology , Animals , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Humans , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Sulfonamides/pharmacology , Piperidines/pharmacology , Embryo, Nonmammalian , Flavonoids/pharmacology , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
J Biol Chem ; 300(3): 105698, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301887

ABSTRACT

Nuclear actin has been demonstrated to be essential for optimal transcription, but the molecular mechanisms and direct binding partner for actin in the RNA polymerase complex have remained unknown. By using purified proteins in a variety of biochemical assays, we demonstrate a direct and specific interaction between monomeric actin and Cdk9, the kinase subunit of the positive transcription elongation factor b required for RNA polymerase II pause-release. This interaction efficiently prevents actin polymerization, is not dependent on kinase activity of Cdk9, and is not involved with releasing positive transcription elongation factor b from its inhibitor 7SK snRNP complex. Supporting the specific role for actin in the elongation phase of transcription, chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) reveals that actin interacts with genes only upon their active transcription elongation. This study therefore provides novel insights into the mechanisms by which actin facilitates the transcription process.


Subject(s)
Actins , Cyclin-Dependent Kinase 9 , Positive Transcriptional Elongation Factor B , Humans , Actins/genetics , Actins/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic
10.
J Biol Chem ; 300(8): 107566, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002676

ABSTRACT

Mixed lineage leukemia-fusion proteins (MLL-FPs) are believed to maintain gene activation and induce MLL through aberrantly stimulating transcriptional elongation, but the underlying mechanisms are incompletely understood. Here, we show that both MLL1 and AF9, one of the major fusion partners of MLL1, mainly occupy promoters and distal intergenic regions, exhibiting chromatin occupancy patterns resembling that of RNA polymerase II in HEL, a human erythroleukemia cell line without MLL1 rearrangement. MLL1 and AF9 only coregulate over a dozen genes despite of their co-occupancy on thousands of genes. They do not interact with each other, and their chromatin occupancy is also independent of each other. Moreover, AF9 deficiency in HEL cells decreases global TBP occupancy while decreases CDK9 occupancy on a small number of genes, suggesting an accessory role of AF9 in CDK9 recruitment and a possible major role in transcriptional initiation via initiation factor recruitment. Importantly, MLL1 and MLL-AF9 occupy promoters and distal intergenic regions, exhibiting identical chromatin occupancy patterns in MLL cells, and MLL-AF9 deficiency decreased occupancy of TBP and TFIIE on major target genes of MLL-AF9 in iMA9, a murine acute myeloid leukemia cell line inducibly expressing MLL-AF9, suggesting that it can also regulate initiation. These results suggest that there is no difference between MLL1 and MLL-AF9 with respect to location and size of occupancy sites, contrary to what people have believed, and that MLL-AF9 may also regulate transcriptional initiation in addition to widely believed elongation.


Subject(s)
Cyclin-Dependent Kinase 9 , Histone-Lysine N-Methyltransferase , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion , Humans , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinase 9/genetics , Animals , Mice , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/genetics , Gene Expression Regulation, Leukemic , Cell Line, Tumor , Chromatin/metabolism , Chromatin/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Promoter Regions, Genetic , Transcription Initiation, Genetic , Transcriptional Elongation Factors
11.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-34523672

ABSTRACT

Sustained neutrophilic inflammation is detrimental for cardiac repair and associated with adverse outcomes following myocardial infarction (MI). An attractive therapeutic strategy to treat MI is to reduce or remove infiltrating neutrophils to promote downstream reparative mechanisms. CDK9 inhibitor compounds enhance the resolution of neutrophilic inflammation; however, their effects on cardiac repair/regeneration are unknown. We have devised a cardiac injury model to investigate inflammatory and regenerative responses in larval zebrafish using heartbeat-synchronised light-sheet fluorescence microscopy. We used this model to test two clinically approved CDK9 inhibitors, AT7519 and flavopiridol, examining their effects on neutrophils, macrophages and cardiomyocyte regeneration. We found that AT7519 and flavopiridol resolve neutrophil infiltration by inducing reverse migration from the cardiac lesion. Although continuous exposure to AT7519 or flavopiridol caused adverse phenotypes, transient treatment accelerated neutrophil resolution while avoiding these effects. Transient treatment with AT7519, but not flavopiridol, augmented wound-associated macrophage polarisation, which enhanced macrophage-dependent cardiomyocyte number expansion and the rate of myocardial wound closure. Using cdk9-/- knockout mutants, we showed that AT7519 is a selective CDK9 inhibitor, revealing the potential of such treatments to promote cardiac repair/regeneration.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Flavonoids/pharmacology , Myocardium/enzymology , Neutrophils/enzymology , Piperidines/pharmacology , Pyrazoles/pharmacology , Regeneration/drug effects , Zebrafish Proteins/antagonists & inhibitors , Animals , Cyclin-Dependent Kinase 9/metabolism , Inflammation/drug therapy , Inflammation/enzymology , Zebrafish , Zebrafish Proteins/metabolism
12.
FASEB J ; 38(8): e23628, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38661032

ABSTRACT

Cancer cells frequently exhibit hyperactivation of transcription, which can lead to increased sensitivity to compounds targeting the transcriptional kinases, in particular CDK9. However, mechanistic details of CDK9 inhibition-induced cancer cell-selective anti-proliferative effects remain largely unknown. Here, we discover that CDK9 inhibition activates the innate immune response through viral mimicry in cancer cells. In MYC over-expressing prostate cancer cells, CDK9 inhibition leads to the gross accumulation of mis-spliced RNA. Double-stranded RNA (dsRNA)-activated kinase can recognize these mis-spliced RNAs, and we show that the activity of this kinase is required for the CDK9 inhibitor-induced anti-proliferative effects. Using time-resolved transcriptional profiling (SLAM-seq), targeted proteomics, and ChIP-seq, we show that, similar to viral infection, CDK9 inhibition significantly suppresses transcription of most genes but allows selective transcription and translation of cytokines related to the innate immune response. In particular, CDK9 inhibition activates NFκB-driven cytokine signaling at the transcriptional and secretome levels. The transcriptional signature induced by CDK9 inhibition identifies prostate cancers with a high level of genome instability. We propose that it is possible to induce similar effects in patients using CDK9 inhibition, which, we show, causes DNA damage in vitro. In the future, it is important to establish whether CDK9 inhibitors can potentiate the effects of immunotherapy against late-stage prostate cancer, a currently lethal disease.


Subject(s)
Cyclin-Dependent Kinase 9 , Immunity, Innate , Humans , Male , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Prostatic Neoplasms/immunology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism
13.
Mol Cell ; 65(6): 1068-1080.e5, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28262505

ABSTRACT

The BET proteins are major transcriptional regulators and have emerged as new drug targets, but their functional distinction has remained elusive. In this study, we report that the BET family members Brd2 and Brd4 exert distinct genomic functions at genes whose transcription they co-regulate during mouse T helper 17 (Th17) cell differentiation. Brd2 is associated with the chromatin insulator CTCF and the cohesin complex to support cis-regulatory enhancer assembly for gene transcriptional activation. In this context, Brd2 binds the transcription factor Stat3 in an acetylation-sensitive manner and facilitates Stat3 recruitment to active enhancers occupied with transcription factors Irf4 and Batf. In parallel, Brd4 temporally controls RNA polymerase II (Pol II) processivity during transcription elongation through cyclin T1 and Cdk9 recruitment and Pol II Ser2 phosphorylation. Collectively, our study uncovers both separate and interdependent Brd2 and Brd4 functions in potentiating the genetic program required for Th17 cell development and adaptive immunity.


Subject(s)
Adaptive Immunity , Cell Differentiation , Chromatin/enzymology , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Th17 Cells/enzymology , Transcription Factors/metabolism , Transcription, Genetic , Acetylation , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin/genetics , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice, Inbred C57BL , Models, Molecular , Nuclear Proteins/genetics , Phenotype , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , RNA Polymerase II/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Structure-Activity Relationship , Th17 Cells/immunology , Transcription Factors/genetics , Transfection , Cohesins
14.
Mol Cell ; 67(1): 5-18.e19, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28673542

ABSTRACT

Processive elongation of RNA Polymerase II from a proximal promoter paused state is a rate-limiting event in human gene control. A small number of regulatory factors influence transcription elongation on a global scale. Prior research using small-molecule BET bromodomain inhibitors, such as JQ1, linked BRD4 to context-specific elongation at a limited number of genes associated with massive enhancer regions. Here, the mechanistic characterization of an optimized chemical degrader of BET bromodomain proteins, dBET6, led to the unexpected identification of BET proteins as master regulators of global transcription elongation. In contrast to the selective effect of bromodomain inhibition on transcription, BET degradation prompts a collapse of global elongation that phenocopies CDK9 inhibition. Notably, BRD4 loss does not directly affect CDK9 localization. These studies, performed in translational models of T cell leukemia, establish a mechanism-based rationale for the development of BET bromodomain degradation as cancer therapy.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Nuclear Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Elongation, Genetic , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Proteins , Cyclin-Dependent Kinase 9/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Leukemic , HCT116 Cells , HEK293 Cells , Humans , Jurkat Cells , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Multiprotein Complexes , Nuclear Proteins/genetics , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Stability , Proteolysis , RNA Polymerase II/metabolism , Time Factors , Transcription Elongation, Genetic/drug effects , Transcription Factors/genetics , Transfection , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
15.
Proc Natl Acad Sci U S A ; 119(17): e2110557119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35442775

ABSTRACT

Anticancer drug development campaigns often fail due to an incomplete understanding of the therapeutic index differentiating the efficacy of the agent against the cancer and its on-target toxicities to the host. To address this issue, we established a versatile preclinical platform in which genetically defined cancers are produced using somatic tissue engineering in transgenic mice harboring a doxycycline-inducible short hairpin RNA against the target of interest. In this system, target inhibition is achieved by the addition of doxycycline, enabling simultaneous assessment of efficacy and toxicity in the same animal. As proof of concept, we focused on CDK9­a cancer target whose clinical development has been hampered by compounds with poorly understood target specificity and unacceptable toxicities. We systematically compared phenotypes produced by genetic Cdk9 inhibition to those achieved using a recently developed highly specific small molecule CDK9 inhibitor and found that both perturbations led to robust antitumor responses. Remarkably, nontoxic levels of CDK9 inhibition could achieve significant treatment efficacy, and dose-dependent toxicities produced by prolonged CDK9 suppression were largely reversible upon Cdk9 restoration or drug withdrawal. Overall, these results establish a versatile in vivo target validation platform that can be employed for rapid triaging of therapeutic targets and lend support to efforts aimed at advancing CDK9 inhibitors for cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/genetics , RNA Interference
16.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Article in English | MEDLINE | ID: mdl-35082156

ABSTRACT

Enzyme-assisted posttranslational modifications (PTMs) constitute a major means of signaling across different cellular compartments. However, how nonenzymatic PTMs-despite their direct relevance to covalent drug development-impinge on cross-compartment signaling remains inaccessible as current target-identification (target-ID) technologies offer limited spatiotemporal resolution, and proximity mapping tools are also not guided by specific, biologically-relevant, ligand chemotypes. Here we establish a quantitative and direct profiling platform (Localis-rex) that ranks responsivity of compartmentalized subproteomes to nonenzymatic PTMs. In a setup that contrasts nucleus- vs. cytoplasm-specific responsivity to reactive-metabolite modification (hydroxynonenylation), ∼40% of the top-enriched protein sensors investigated respond in compartments of nonprimary origin or where the canonical activity of the protein sensor is inoperative. CDK9-a primarily nuclear-localized kinase-was hydroxynonenylated only in the cytoplasm. Site-specific CDK9 hydroxynonenylation-which we identified in untreated cells-drives its nuclear translocation, downregulating RNA-polymerase-II activity, through a mechanism distinct from that of commonly used CDK9 inhibitors. Taken together, this work documents an unmet approach to quantitatively profile and decode localized and context-specific signaling/signal-propagation programs orchestrated by reactive covalent ligands.


Subject(s)
Proteins/genetics , Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , RAW 264.7 Cells , Signal Transduction/physiology , Transcription, Genetic/genetics
17.
J Cell Mol Med ; 28(8): e18327, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38661437

ABSTRACT

Cartilage defects in the knee are often associated with the progression of degenerative osteoarthritis (OA), and cartilage repair is a useful strategy for managing this disease. However, cartilage repair is challenging because of the unique environment within the tissue. Recently, stem cell-based therapies have shed new light on this issue. In this study, we prepared exosomes (EXOs) from cartilage stem/progenitor cells (CSPCs) and found that treatment with EXOs increased the viability, migration, and proliferation of cultured primary chondrocytes. In a subacute OA rat model, the application of EXOs facilitated cartilage regeneration as evidenced by histological staining. Exosomal protein analysis together with bioinformatics suggested that cyclin-dependent kinase 9 (CDK9) is a key factor for chondrocyte growth and migration. Functional studies confirmed this prediction, that is, inhibiting CDK9 reduced the beneficial effects induced by EXOs in primary chondrocytes; while overexpression of CDK9 recapitulated the EXOs-induced phenotypes. RNA-Seq data showed that a set of genes involved in cell growth and migration were up-regulated by EXOs in chondrocytes. These changes could be partially reproduced by CDK9 overexpression. Overall, our data suggest that EXOs derived from primary CSPCs hold great therapeutic potential for treating cartilage defect-associated disorders such as degenerative OA, and that CDK9 is a key factor in this process.


Subject(s)
Cartilage, Articular , Cell Proliferation , Chondrocytes , Disease Models, Animal , Exosomes , Animals , Exosomes/metabolism , Rats , Chondrocytes/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Stem Cells/metabolism , Stem Cells/cytology , Cell Movement , Rats, Sprague-Dawley , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinase 9/genetics , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/therapy , Male , Cells, Cultured , Regeneration , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/therapy
18.
Antimicrob Agents Chemother ; 68(3): e0107223, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38319085

ABSTRACT

Current antiretroviral therapy for HIV-1 infection does not represent a cure for infection as viral rebound inevitably occurs following discontinuation of treatment. The "block and lock" therapeutic strategy is intended to enforce proviral latency and durably suppress viremic reemergence in the absence of other intervention. The transcription-associated cyclin-dependent protein kinases (tCDKs) are required for expression from the 5´ HIV-1 long-terminal repeat, but the therapeutic potential of inhibiting these kinases for enforcing HIV-1 latency has not been characterized. Here, we expanded previous observations to directly compare the effect of highly selective small molecule inhibitors of CDK7 (YKL-5-124), CDK9 (LDC000067), and CDK8/19 (Senexin A), and found each of these prevented HIV-1 provirus expression at concentrations that did not cause cell toxicity. Inhibition of CDK7 caused cell cycle arrest, whereas CDK9 and CDK8/19 inhibitors did not, and could be continuously administered to establish proviral latency. Upon discontinuation of drug administration, HIV immediately rebounded in cells that had been treated with the CDK9 inhibitor, while proviral latency persisted for several days in cells that had been treated with CDK8/19 inhibitors. These results identify the mediator kinases CDK8/CDK19 as potential "block and lock" targets for therapeutic suppression of HIV-1 provirus expression.


Subject(s)
HIV-1 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/pharmacology , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclins/metabolism , Cyclins/pharmacology
19.
Bioorg Med Chem Lett ; 112: 129941, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39222890

ABSTRACT

Emerging clinical evidence indicates that selective CDK9 inhibition may provide clinical benefits in the management of certain cancers. Many CDK9 selective inhibitors have entered clinical developments, and are being investigated. No clear winner has emerged because of unforeseen toxicity often observed in clinic with these agents. Therefore, a novel agent with differentiated profiles is still desirable. Herein, we report our design, syntheses of a novel azaindole series of selective CDK9 inhibitors. SAR studies led to a preclinical candidate YK-2168. YK2168 exhibited improved CDK9 selectivity over AZD4573 and BAY1251152; also showed differentiated intravenous PK profile and remarkable solid tumor efficacy in a mouse gastric cancer SNU16 CDX model in preclinical studies. YK-2168 is currently in clinical development in China (CTR20212900).


Subject(s)
Cyclin-Dependent Kinase 9 , Protein Kinase Inhibitors , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism , Animals , Humans , Structure-Activity Relationship , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Molecular Structure , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Discovery , Dose-Response Relationship, Drug , Cell Line, Tumor , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects
20.
Cell ; 138(6): 1122-36, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19766566

ABSTRACT

The phosphorylation of the serine 10 at histone H3 has been shown to be important for transcriptional activation. Here, we report the molecular mechanism through which H3S10ph triggers transcript elongation of the FOSL1 gene. Serum stimulation induces the PIM1 kinase to phosphorylate the preacetylated histone H3 at the FOSL1 enhancer. The adaptor protein 14-3-3 binds the phosphorylated nucleosome and recruits the histone acetyltransferase MOF, which triggers the acetylation of histone H4 at lysine 16 (H4K16ac). This histone crosstalk generates the nucleosomal recognition code composed of H3K9acS10ph/H4K16ac determining a nucleosome platform for the bromodomain protein BRD4 binding. The recruitment of the positive transcription elongation factor b (P-TEFb) via BRD4 induces the release of the promoter-proximal paused RNA polymerase II and the increase of its processivity. Thus, the single phosphorylation H3S10ph at the FOSL1 enhancer triggers a cascade of events which activate transcriptional elongation.


Subject(s)
Histone Code , Histones/metabolism , Transcription, Genetic , Animals , Cell Line , Cyclin-Dependent Kinase 9/metabolism , Drosophila , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , Humans , Mice , Nucleosomes/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factors/metabolism , Yeasts
SELECTION OF CITATIONS
SEARCH DETAIL