Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cancer Biol Ther ; 24(1): 2219470, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37272701

ABSTRACT

As a catalytic subunit of the positive transcription elongation factor b (P-TEFb), cyclin-dependent kinase 9 (CDK9) has been demonstrated to contribute to carcinogenesis. This review focuses on the development of selective CDK9 inhibitors and proteolysis-targeting chimera (PROTAC) degraders. Twenty selective CDK9 inhibitors and degraders are introduced along with their structures, IC50 values, in vitro and in vivo experiments, mechanisms underlying their inhibitory effects, and combination regimens. NVP-2, MC180295, fadraciclib, KB-0742, LZT-106, and 21e have been developed mainly for treating solid tumors, and most of them work only on certain genotypes of solid tumors. Only VIP152 has been proven to benefit the patients with advanced high-grade lymphoma (HGL) and solid tumors in clinical trials. Continued efforts to explore the molecular mechanisms underlying the inhibitory effects, and to identify suitable tumor genotypes and combination treatment strategies, are crucial to demonstrate the efficacy of selective CDK9 inhibitors and degraders in tumor therapy.


Subject(s)
Cyclin-Dependent Kinase 9 , Neoplasms , Humans , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/genetics , Positive Transcriptional Elongation Factor B , Neoplasms/drug therapy
2.
J Biomol Struct Dyn ; 41(19): 10202-10213, 2023 11.
Article in English | MEDLINE | ID: mdl-36562191

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) is a transcription-associated protein involved in controlling the cell cycle and is often deregulated in stress conditions. CDK9 is being studied as a well-known druggable target for developing effective therapeutics against a wide range of cancer, cardiac dysfunction and inflammatory diseases. Owing to the significance of CDK9 in the etiology of hematological and solid malignancies, its structure, biological activity, regulation and its pharmacological inhibition are being explored for therapeutic management of cancer. We employed a structure-based virtual high-throughput screening of bioactive compounds from the IMPPAT database to discover potential bioactive inhibitors of CDK9. The preliminary results were obtained from the Lipinski criteria, ADMET parameters and sorting compounds without any PAINS patterns. Subsequently, binding affinity and selectivity analyses were used to find effective CDK9 hits. This screening resulted in the identification of two natural compounds, Glabrene and Guggulsterone with high affinity and specificity for the CDK9 binding site. Both compounds exhibit drug-like characteristics, as projected by ADMET analysis, physicochemical data and PASS evaluation. Both compounds preferentially bind to the ATP-binding pocket of CDK9 and interact with functionally important residues. Further, the dynamics and consistency of CDK9 interaction with Glabrene and Guggulsteron were evaluated through all-atom molecular dynamic (MD) simulations which suggested the stability of both complexes. The results might be deployed to introduce novel CDK9 inhibitors that may treat life-threatening diseases, including cancer.Communicated by Ramaswamy H. Sarma.


Subject(s)
Cyclin-Dependent Kinase 9 , Neoplasms , Humans , Molecular Docking Simulation , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Protein Kinase Inhibitors/chemistry , Molecular Dynamics Simulation
3.
Nucleic Acids Res ; 50(2): 750-762, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34935961

ABSTRACT

P-TEFb modulates RNA polymerase II elongation through alternative interaction with negative and positive regulation factors. While inactive P-TEFbs are mainly sequestered in the 7SK snRNP complex in a chromatin-free state, most of its active forms are in complex with its recruitment factors, Brd4 and SEC, in a chromatin-associated state. Thus, switching from inactive 7SK snRNP to active P-TEFb (Brd4/P-TEFb or SEC/P-TEFb) is essential for global gene expression. Although it has been shown that cellular signaling stimulates the disruption of 7SK snRNP, releasing dephosphorylated and catalytically inactive P-TEFb, little is known about how the inactive released P-TEFb is reactivated. Here, we show that the Cdk9/CycT1 heterodimer released from 7SK snRNP is completely dissociated into monomers in response to stress. Brd4 or SEC then recruits monomerized Cdk9 and CycT1 to reassemble the core P-TEFb. Meanwhile, the binding of monomeric dephosphorylated Cdk9 to either Brd4 or SEC induces the autophosphorylation of T186 of Cdk9. Finally, the same mechanism is employed during nocodazole released entry into early G1 phase of cell cycle. Therefore, our studies demonstrate a novel mechanism by which Cdk9 and CycT1 monomers are reassembled on chromatin to form active P-TEFb by its interaction with Brd4 or SEC to regulate transcription.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , DNA-Binding Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Cell Cycle , Cell Line , Cyclin T/chemistry , Cyclin-Dependent Kinase 9/chemistry , Enzyme Activation , Humans , Models, Biological , Phosphorylation , Protein Binding , Protein Multimerization , Recombinant Proteins , Ribonucleoproteins, Small Nuclear/chemistry , Stress, Physiological
4.
J Med Chem ; 64(17): 12548-12571, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34415148

ABSTRACT

Hematologic malignancies (HM) start in blood forming tissue or in the cells of the immune system. Cyclin-dependent kinases (CDKs) regulate cell cycle progression, and some of them control cellular transcription. CDK inhibition can trigger apoptosis and could be particularly useful in hematological malignancies. Herein, we describe our efforts toward the discovery of a novel series of quinazoline derivatives as CDK inhibitors. Intensive structural modifications lead to the identification of compound 37d as the most active inhibitors of CDKs 1, 2, 4, 8 and 9 with balancing potency and selectivity against CDKs. Further biological studies revealed that compound 37d can arrest the cell cycle and induce apoptosis via activating PARP and caspase 3. More importantly, compound 37d showed good antitumor efficacy in multiple HM mice xenograft models with no obvious toxicity. These results indicated that CDK 1, 2, 4, 8, and 9 inhibitors could be potentially used to treat certain hematologic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Area Under Curve , Catalytic Domain , Cell Line, Tumor , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Half-Life , Humans , Mice , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/pharmacokinetics , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
J Med Chem ; 63(22): 13228-13257, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32866383

ABSTRACT

Cyclin-dependent kinase 9 (CDK9), which regulates transcriptional elongation, is an attractive therapeutic target for many cancers, especially for cancers driven by transcriptional dysregulation. In particular, CDK9 promotes RNA polymerase II pause/release, a rate-limiting step in normal transcriptional regulation that is frequently dysregulated in cancers. Emerging evidence indicates that selective CDK9 inhibition or degradation may provide a therapeutic benefit against certain cancers. Indeed, the development of CDK9 modulators (inhibitors and degraders) has attracted great attention, with several molecules currently under clinical development. This review provides an overview of recent advances in CDK9 modulators in general, with special emphasis on compounds under clinical evaluation and new emerging strategies, such as proteolysis targeting chimeras (PROTACs).


Subject(s)
Chemistry, Pharmaceutical/trends , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism , Drug Development/trends , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Chemistry, Pharmaceutical/methods , Cyclin-Dependent Kinase 9/chemistry , Drug Development/methods , Humans , Molecular Docking Simulation/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary
6.
Life Sci ; 258: 118228, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32781071

ABSTRACT

AIMS: Cyclin-dependent kinase 9 (CDK9) is a member of the CDK subfamily and plays a major role in the regulation of transcriptional elongation. It has attracted widespread attention as a therapeutic target for cancer. Here, we aimed to explore novel CDK 9 inhibitors by using a hybrid virtual screening strategy. MAIN METHODS: A hybrid virtual screening strategy was constructed with computer-aided drug design (CADD). First, compounds were filtered in accordance with Lipinski's rule of five and adsorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. Second, a 3D-QSAR pharmacophore model was built and used as a 3D query to screen the obtained hit compounds. Third, the hit compounds were subjected to molecular docking studies. Fourth, molecular dynamics (MD) simulations were performed on CDK9 in complex with the final hits to examine the structural stability. Finally, CDK9 kinase biochemical assay was performed to identify the biological activity of the hit compounds. KEY FINDINGS: Seven hit compounds were screened out. These hit compounds showed drug-like properties in accordance with Lipinski's rule of five and ADMET. Complexes involving the six hit compounds bound to CDK9 exhibited good structural stability in the MD simulation. Furthermore, these six hit compounds had strong inhibitory activity against CDK9 kinase. In particular, hit 3 showed the most promising activity with the percentage of 71%. SIGNIFICANCE: The six hit compounds may be promising novel CDK9 inhibitors, and the hybrid virtual screening strategy designed in this study provides an important reference for the design and synthesis of novel CDK9 inhibitors.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Protein Kinase Inhibitors/metabolism , Cyclin-Dependent Kinase 9/chemistry , Drug Evaluation, Preclinical/methods , Humans , Protein Binding/drug effects , Protein Binding/physiology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary
7.
J Med Chem ; 63(6): 3327-3347, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32129996

ABSTRACT

Cyclin-dependent kinases (CDKs) are promising therapeutic targets for cancer therapy. Herein, we describe our efforts toward the discovery of a series of 5-chloro-N4-phenyl-N2-(pyridin-2-yl)pyrimidine-2,4-diamine derivatives as dual CDK6 and 9 inhibitors. Intensive structural modifications lead to the identification of compound 66 as the most active dual CDK6/9 inhibitor with balancing potency against these two targets and good selectivity over CDK2. Further biological studies revealed that compound 66 was directly bound to CDK6/9, resulting in suppression of their downstream signaling pathway and inhibition of cell proliferation by blocking cell cycle progression and inducing cellular apoptosis. More importantly, compound 66 significantly inhibited tumor growth in a xenograft mouse model with no obvious toxicity, indicating the promising therapeutic potential of CDK6/9 dual inhibitors for cancer treatment. Therefore, the above results are of great importance in the development of dual CDK6/9 inhibitors for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/chemistry , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Humans , Male , Mice, Inbred BALB C , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyridines/administration & dosage , Pyridines/chemical synthesis , Pyridines/metabolism , Pyrimidines/administration & dosage , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Rats, Sprague-Dawley , Structure-Activity Relationship
8.
Nature ; 560(7717): 192-197, 2018 08.
Article in English | MEDLINE | ID: mdl-30046105

ABSTRACT

Visceral leishmaniasis causes considerable mortality and morbidity in many parts of the world. There is an urgent need for the development of new, effective treatments for this disease. Here we describe the development of an anti-leishmanial drug-like chemical series based on a pyrazolopyrimidine scaffold. The leading compound from this series (7, DDD853651/GSK3186899) is efficacious in a mouse model of visceral leishmaniasis, has suitable physicochemical, pharmacokinetic and toxicological properties for further development, and has been declared a preclinical candidate. Detailed mode-of-action studies indicate that compounds from this series act principally by inhibiting the parasite cdc-2-related kinase 12 (CRK12), thus defining a druggable target for visceral leishmaniasis.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Leishmania donovani/drug effects , Leishmania donovani/enzymology , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/parasitology , Molecular Targeted Therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Humans , Mice , Molecular Docking Simulation , Proteome/drug effects , Proteomics , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Reproducibility of Results , Substrate Specificity
9.
Nature ; 558(7710): 460-464, 2018 06.
Article in English | MEDLINE | ID: mdl-29899453

ABSTRACT

The end of the RNA polymerase II (Pol II) transcription cycle is strictly regulated to prevent interference between neighbouring genes and to safeguard transcriptome integrity 1 . The accumulation of Pol II downstream of the cleavage and polyadenylation signal can facilitate the recruitment of factors involved in mRNA 3'-end formation and termination 2 , but how this sequence is initiated remains unclear. In a chemical-genetic screen, human protein phosphatase 1 (PP1) isoforms were identified as substrates of positive transcription elongation factor b (P-TEFb), also known as the cyclin-dependent kinase 9 (Cdk9)-cyclin T1 (CycT1) complex 3 . Here we show that Cdk9 and PP1 govern phosphorylation of the conserved elongation factor Spt5 in the fission yeast Schizosaccharomyces pombe. Cdk9 phosphorylates both Spt5 and a negative regulatory site on the PP1 isoform Dis2 4 . Sites targeted by Cdk9 in the Spt5 carboxy-terminal domain can be dephosphorylated by Dis2 in vitro, and dis2 mutations retard Spt5 dephosphorylation after inhibition of Cdk9 in vivo. Chromatin immunoprecipitation and sequencing analysis indicates that Spt5 is dephosphorylated as transcription complexes traverse the cleavage and polyadenylation signal, concomitant with the accumulation of Pol II phosphorylated at residue Ser2 of the carboxy-terminal domain consensus heptad repeat 5 . A conditionally lethal Dis2-inactivating mutation attenuates the drop in Spt5 phosphorylation on chromatin, promotes transcription beyond the normal termination zone (as detected by precision run-on transcription and sequencing 6 ) and is genetically suppressed by the ablation of Cdk9 target sites in Spt5. These results suggest that the transition of Pol II from elongation to termination coincides with a Dis2-dependent reversal of Cdk9 signalling-a switch that is analogous to a Cdk1-PP1 circuit that controls mitotic progression 4 .


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Phosphoprotein Phosphatases/metabolism , RNA Polymerase II/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Transcription Elongation, Genetic , Transcription Termination, Genetic , Amino Acid Sequence , Cyclin-Dependent Kinase 9/chemistry , Humans , Mitosis , Phosphoprotein Phosphatases/chemistry , Phosphorylation , RNA Polymerase II/chemistry , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/chemistry , Signal Transduction , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism
10.
Retrovirology ; 15(1): 39, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29792216

ABSTRACT

BACKGROUND: HIV-1 transcription activator protein Tat is phosphorylated in vitro by CDK2 and DNA-PK on Ser-16 residue and by PKR on Tat Ser-46 residue. Here we analyzed Tat phosphorylation in cultured cells and its functionality. RESULTS: Mass spectrometry analysis showed primarily Tat Ser-16 phosphorylation in cultured cells. In vitro, CDK2/cyclin E predominantly phosphorylated Tat Ser-16 and PKR-Tat Ser-46. Alanine mutations of either Ser-16 or Ser-46 decreased overall Tat phosphorylation. Phosphorylation of Tat Ser-16 was reduced in cultured cells treated by a small molecule inhibitor of CDK2 and, to a lesser extent, an inhibitor of DNA-PK. Conditional knock-downs of CDK2 and PKR inhibited and induced one round HIV-1 replication respectively. HIV-1 proviral transcription was inhibited by Tat alanine mutants and partially restored by S16E mutation. Pseudotyped HIV-1 with Tat S16E mutation replicated well, and HIV-1 Tat S46E-poorly, but no live viruses were obtained with Tat S16A or Tat S46A mutations. TAR RNA binding was affected by Tat Ser-16 alanine mutation. Binding to cyclin T1 showed decreased binding of all Ser-16 and Ser-46 Tat mutants with S16D and Tat S46D mutationts showing the strongest effect. Molecular modelling and molecular dynamic analysis revealed significant structural changes in Tat/CDK9/cyclin T1 complex with phosphorylated Ser-16 residue, but not with phosphorylated Ser-46 residue. CONCLUSION: Phosphorylation of Tat Ser-16 induces HIV-1 transcription, facilitates binding to TAR RNA and rearranges CDK9/cyclin T1/Tat complex. Thus, phosphorylation of Tat Ser-16 regulates HIV-1 transcription and may serve as target for HIV-1 therapeutics.


Subject(s)
Gene Expression Regulation, Viral , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Serine/metabolism , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Cells, Cultured , Cyclin T/chemistry , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Gene Knockdown Techniques , HIV Infections/genetics , Host-Pathogen Interactions , Humans , Models, Biological , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , RNA, Viral , Ubiquitination , Virus Replication , eIF-2 Kinase/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics
11.
J Biol Chem ; 293(26): 10009-10025, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29743242

ABSTRACT

The HIV trans-activator Tat recruits the host transcription elongation factor P-TEFb to stimulate proviral transcription. Phosphorylation of Thr-186 on the activation loop (T-loop) of cyclin-dependent kinase 9 (CDK9) is essential for its kinase activity and assembly of CDK9 and cyclin T1 (CycT1) to form functional P-TEFb. Phosphorylation of a second highly conserved T-loop site, Ser-175, alters the competitive binding of Tat and the host recruitment factor bromodomain containing 4 (BRD4) to P-TEFb. Here, we investigated the intracellular mechanisms that regulate these key phosphorylation events required for HIV transcription. Molecular dynamics simulations revealed that the CDK9/CycT1 interface is stabilized by intramolecular hydrogen bonding of pThr-186 by an arginine triad and Glu-96 of CycT1. Arginine triad substitutions that disrupted CDK9/CycT1 assembly accumulated Thr-186-dephosphorylated CDK9 associated with the cytoplasmic Hsp90/Cdc37 chaperone. The Hsp90/Cdc37/CDK9 complex was also present in resting T cells, which lack CycT1. Hsp90 inhibition in primary T cells blocked P-TEFb assembly, disrupted Thr-186 phosphorylation, and suppressed proviral reactivation. The selective CDK7 inhibitor THZ1 blocked CDK9 phosphorylation at Ser-175, and in vitro kinase assays confirmed that CDK7 activity is principally responsible for Ser-175 phosphorylation. Mutation of Ser-175 to Lys had no effect on CDK9 kinase activity or P-TEFb assembly but strongly suppressed both HIV expression and BRD4 binding. We conclude that the transfer of CDK9 from the Hsp90/Cdc37 complex induced by Thr-186 phosphorylation is a key step in P-TEFb biogenesis. Furthermore, we demonstrate that CDK7-mediated Ser-175 phosphorylation is a downstream nuclear event essential for facilitating CDK9 T-loop interactions with Tat.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , HIV-1/physiology , Positive Transcriptional Elongation Factor B/metabolism , Virus Activation , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cyclin-Dependent Kinase 9/chemistry , Enzyme Activation , HIV-1/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Jurkat Cells , Phosphorylation , Protein Binding , Serine/metabolism , Threonine/metabolism
12.
Nat Chem Biol ; 14(2): 163-170, 2018 02.
Article in English | MEDLINE | ID: mdl-29251720

ABSTRACT

Cyclin-dependent kinase 9 (CDK9), an important regulator of transcriptional elongation, is a promising target for cancer therapy, particularly for cancers driven by transcriptional dysregulation. We characterized NVP-2, a selective ATP-competitive CDK9 inhibitor, and THAL-SNS-032, a selective CDK9 degrader consisting of a CDK-binding SNS-032 ligand linked to a thalidomide derivative that binds the E3 ubiquitin ligase Cereblon (CRBN). To our surprise, THAL-SNS-032 induced rapid degradation of CDK9 without affecting the levels of other SNS-032 targets. Moreover, the transcriptional changes elicited by THAL-SNS-032 were more like those caused by NVP-2 than those induced by SNS-032. Notably, compound washout did not significantly reduce levels of THAL-SNS-032-induced apoptosis, suggesting that CDK9 degradation had prolonged cytotoxic effects compared with CDK9 inhibition. Thus, our findings suggest that thalidomide conjugation represents a promising strategy for converting multi-targeted inhibitors into selective degraders and reveal that kinase degradation can induce distinct pharmacological effects compared with inhibition.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/chemistry , Peptide Hydrolases/chemistry , Protein Kinase Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing , Apoptosis , Cell Line, Tumor , Cell Proliferation , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Ligands , Oxazoles/pharmacology , Phosphorylation , Protein Binding , Protein Conformation , Proteomics , Thalidomide/pharmacology , Thiazoles/pharmacology , Ubiquitin-Protein Ligases
13.
Proc Natl Acad Sci U S A ; 114(27): E5352-E5361, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28630312

ABSTRACT

The epigenetic reader BRD4 plays a vital role in transcriptional regulation, cellular growth control, and cell-cycle progression. Dysregulation of BRD4 function has been implicated in the pathogenesis of a wide range of cancers. However, how BRD4 is regulated to maintain its normal function in healthy cells and how alteration of this process leads to cancer remain poorly understood. In this study, we discovered that BRD4 is hyperphosphorylated in NUT midline carcinoma and identified CDK9 as a potential kinase mediating BRD4 hyperphosphorylation. Disruption of BRD4 hyperphosphorylation using both chemical and molecular inhibitors led to the repression of BRD4 downstream oncogenes and abrogation of cellular transformation. BRD4 hyperphosphorylation is also observed in other cancers displaying enhanced BRD4 oncogenic activity. Our study revealed a mechanism that may regulate BRD4 biological function through phosphorylation, which, when dysregulated, could lead to oncogenesis. Our finding points to strategies to target the aberrant BRD4 signaling specifically for cancer intervention.


Subject(s)
Carcinoma/genetics , Carcinoma/metabolism , Cyclin-Dependent Kinase 9/chemistry , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/chemistry , A549 Cells , Carcinogenesis , Carcinoma/drug therapy , Cell Cycle Proteins , Cell Transformation, Neoplastic , Drug Screening Assays, Antitumor , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Neoplasm Proteins , Oncogene Proteins, Fusion/genetics , Oncogenes , Phosphorylation , Signal Transduction
14.
PLoS One ; 12(2): e0171727, 2017.
Article in English | MEDLINE | ID: mdl-28178316

ABSTRACT

In this study, we applied molecular dynamics (MD) simulation to analyze the dynamic behavior of the Tat/CycT1/CDK9 tri-molecular complex and revealed the structural changes of P-TEFb upon Tat binding. We found that Tat could deliberately change the local flexibility of CycT1. Although the structural coordinates of the H1 and H2 helices did not substantially change, H1', H2', and H3' exhibited significant changes en masse. Consequently, the CycT1 residues involved in Tat binding, namely Tat-recognition residues (TRRs), lost their flexibility with the addition of Tat to P-TEFb. In addition, we clarified the structural variation of CDK9 in complex with CycT1 in the presence or absence of Tat. Interestingly, Tat addition significantly reduced the structural variability of the T-loop, thus consolidating the structural integrity of P-TEFb. Finally, we deciphered the formation of the hidden catalytic cavity of CDK9 upon Tat binding. MD simulation revealed that the PITALRE signature sequence of CDK9 flips the inactive kinase cavity of CDK9 into the active form by connecting with Thr186, which is crucial for its activity, thus presumably recruiting the substrate peptide such as the C-terminal domain of RNA pol II. These findings provide vital information for the development of effective novel anti-HIV drugs with CDK9 catalytic activity as the target.


Subject(s)
Cyclin T/chemistry , Cyclin-Dependent Kinase 9/chemistry , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry , Binding Sites , Catalysis , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , Multiprotein Complexes/metabolism , Protein Binding , Protein Conformation , Protein Stability , Structure-Activity Relationship , tat Gene Products, Human Immunodeficiency Virus/metabolism
15.
Mol Biosyst ; 13(2): 246-276, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-27833949

ABSTRACT

CDK9 is a prominent member of the transcriptional CDKs subfamily, a group of kinases whose function is to control the primary steps of mRNA synthesis and processing by eukaryotic RNA polymerase II. As a cyclin-dependent kinase, CDK9 activation in vivo depends upon its association with T-type cyclins to assemble the positive transcription elongation factor (P-TEFb). Although CDK9/P-TEFb phosphorylates the C-terminal domain of RNAP II in the same positions targeted by CDK7 (TFIIH) and CDK8 (Mediator), the former does not participate in the transcription initiation, but rather plays a unique role by driving the polymerase to productive elongation. In addition to RNAP II CTD, the negative transcription elongation factors DSIF and NELF also represent major CDK9 substrates, whose phosphorylation is required to overcome the proximal pause of the polymerase. CDK9 is recruited to specific genes through proteins that interact with both P-TEFb and distinct elements in DNA, RNA or chromatin, where it modulates the activity of individual RNAP II transcription complexes. The regulation of CDK9 function is an intricate network that includes post-translational modifications (phosphorylation/dephosphorylation and acetylation/deacetylation of key residues) as well as the association of P-TEFb with various proteins that can stimulate or inhibit its kinase activity. Several cases of CDK9 deregulation have been linked to important human diseases, including various types of cancer and also AIDS (due to its essential role in HIV replication). Not only HIV, but also many other human viruses have been shown to depend strongly on CDK9 activity to be transcribed within host cells. This review summarizes the main advances made on CDK9/P-TEFb field in more than 20 years, introducing the structural, functional and genetic aspects that have been elucidated ever since.


Subject(s)
Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Animals , Cyclin-Dependent Kinase 9/chemistry , Gene Expression Regulation , Gene Expression Regulation, Viral , Humans , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/chemistry , Protein Binding , Transcription, Genetic , Viruses/genetics
16.
Elife ; 52016 10 12.
Article in English | MEDLINE | ID: mdl-27731797

ABSTRACT

HIV-1 Tat hijacks the human superelongation complex (SEC) to promote proviral transcription. Here we report the 5.9 Å structure of HIV-1 TAR in complex with HIV-1 Tat and human AFF4, CDK9, and CycT1. The TAR central loop contacts the CycT1 Tat-TAR recognition motif (TRM) and the second Tat Zn2+-binding loop. Hydrogen-deuterium exchange (HDX) shows that AFF4 helix 2 is stabilized in the TAR complex despite not touching the RNA, explaining how it enhances TAR binding to the SEC 50-fold. RNA SHAPE and SAXS data were used to help model the extended (Tat Arginine-Rich Motif) ARM, which enters the TAR major groove between the bulge and the central loop. The structure and functional assays collectively support an integrative structure and a bipartite binding model, wherein the TAR central loop engages the CycT1 TRM and compact core of Tat, while the TAR major groove interacts with the extended Tat ARM.


Subject(s)
Cyclin T/chemistry , Cyclin-Dependent Kinase 9/chemistry , DNA, Viral/chemistry , HIV Long Terminal Repeat , Repressor Proteins/chemistry , Transcriptional Elongation Factors/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , DNA, Viral/metabolism , Deuterium Exchange Measurement , HIV-1/genetics , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Proviruses/genetics , Repressor Proteins/metabolism , Scattering, Small Angle , Transcription, Genetic , Transcriptional Elongation Factors/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism
17.
J Med Chem ; 59(19): 8667-8684, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27171036

ABSTRACT

Cyclin dependent kinase (CDK) inhibitors have been the topic of intense research for nearly 2 decades due to their widely varied and critical functions within the cell. Recently CDK9 has emerged as a druggable target for the development of cancer therapeutics. CDK9 plays a crucial role in transcription regulation; specifically, CDK9 mediated transcriptional regulation of short-lived antiapoptotic proteins is critical for the survival of transformed cells. Focused chemical libraries based on a plethora of scaffolds have resulted in mixed success with regard to the development of selective CDK9 inhibitors. Here we review the regulation of CDK9, its cellular functions, and common core structures used to target CDK9, along with their selectivity profile and efficacy in vitro and in vivo.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Discovery/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Flavonoids/chemistry , Flavonoids/pharmacology , Humans , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Purines/chemistry , Purines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Triazines/chemistry , Triazines/pharmacology
18.
Methods Mol Biol ; 1336: 13-28, 2016.
Article in English | MEDLINE | ID: mdl-26231705

ABSTRACT

Cyclin-dependent kinases have established roles in the regulation of cell cycle, in gene expression and in cell differentiation. Many of these kinases have been considered as drug targets and numerous efforts have been made to develop specific and potent inhibitors against them. The first step in all of these attempts and in many other biochemical analyses is the production of highly purified and reliable kinase, most frequently in a recombinant form. In this chapter we describe our experience in the cloning, expression, and purification of CDKs using CDK7/CycH, CDK8/CycC, and CDK9/CycT1 as an example.


Subject(s)
Cyclin-Dependent Kinase 8/isolation & purification , Cyclin-Dependent Kinase 9/isolation & purification , Cyclin-Dependent Kinases/isolation & purification , Recombinant Proteins/isolation & purification , Animals , Catalysis , Cell Differentiation , Chromatography , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinases/chemistry , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Gene Expression Regulation , Glutathione Transferase/metabolism , Insecta , Molecular Biology/methods , Nickel/chemistry , Peptides/chemistry , Recombinant Proteins/chemistry
19.
PLoS One ; 10(4): e0124673, 2015.
Article in English | MEDLINE | ID: mdl-25909811

ABSTRACT

The current paradigm of cyclin-dependent kinase (CDK) regulation based on the well-established CDK2 has been recently expanded. The determination of CDK9 crystal structures suggests the requirement of an additional regulatory protein, such as human immunodeficiency virus type 1 (HIV-1) Tat, to exert its physiological functions. In most kinases, the exact number and roles of the cofactor metal ions remain unappreciated, and the repertoire has thus gained increasing attention recently. Here, molecular dynamics (MD) simulations were implemented on CDK9 to explore the functional roles of HIV-1 Tat and the second Mg2+ ion at site 1 (Mg12+). The simulations unveiled that binding of HIV-1 Tat to CDK9 not only stabilized hydrogen bonds (H-bonds) between ATP and hinge residues Asp104 and Cys106, as well as between ATP and invariant Lys48, but also facilitated the salt bridge network pertaining to the phosphorylated Thr186 at the activation loop. By contrast, these H-bonds cannot be formed in CDK9 owing to the absence of HIV-1 Tat. MD simulations further revealed that the Mg12+ ion, coupled with the Mg22+ ion, anchored to the triphosphate moiety of ATP in its catalytic competent conformation. This observation indicates the requirement of the Mg12+ ion for CDK9 to realize its function. Overall, the introduction of HIV-1 Tat and Mg12+ ion resulted in the active site architectural characteristics of phosphorylated CDK9. These data highlighted the functional roles of HIV-1 Tat and Mg12+ ion in the regulation of CDK9 activity, which contributes an important complementary understanding of CDK molecular underpinnings.


Subject(s)
Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , HIV-1/metabolism , Magnesium/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Cyclin T/chemistry , Cyclin-Dependent Kinase 9/chemistry , Humans , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , tat Gene Products, Human Immunodeficiency Virus/chemistry
20.
Cell Cycle ; 13(11): 1788-97, 2014.
Article in English | MEDLINE | ID: mdl-24727379

ABSTRACT

Developing anti-viral therapies targeting HIV-1 transcription has been hampered by the limited structural knowledge of the proteins involved. HIV-1 hijacks the cellular machinery that controls RNA polymerase II elongation through an interaction of HIV-1 Tat with the positive transcription elongation factor P-TEFb, which interacts with an AF4 family member (AFF1/2/3/4) in the super elongation complex (SEC). Because inclusion of Tat•P-TEFb into the SEC is critical for HIV transcription, we have determined the crystal structure of the Tat•AFF4•P-TEFb complex containing HIV-1 Tat (residues 1-48), human Cyclin T1 (1-266), human Cdk9 (7-332), and human AFF4 (27-69). Tat binding to AFF4•P-TEFb causes concerted structural changes in AFF4 via a shift of helix H5' of Cyclin T1 and the α-3 10 helix of AFF4. The interaction between Tat and AFF4 provides structural constraints that explain tolerated Tat mutations. Analysis of the Tat-binding surface of AFF4 coupled with modeling of all other AF4 family members suggests that AFF1 and AFF4 would be preferred over AFF2 or AFF3 for interaction with Tat•P-TEFb. The structure establishes that the Tat-TAR recognition motif (TRM) in Cyclin T1 interacts with both Tat and AFF4, leading to the exposure of arginine side chains for binding to TAR RNA. Furthermore, modeling of Tat Lys28 acetylation suggests that the acetyl group would be in a favorable position for H-bond formation with Asn257 of TRM, thereby stabilizing the TRM in Cyclin T1, and provides a structural basis for the modulation of TAR RNA binding by acetylation of Tat Lys28.


Subject(s)
HIV-1/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Positive Transcriptional Elongation Factor B/chemistry , Repressor Proteins/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry , Acetylation , Crystallization , Cyclin T/chemistry , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Humans , Multiprotein Complexes/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Conformation , Repressor Proteins/metabolism , Transcriptional Elongation Factors , tat Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...