Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Protein Sci ; 33(6): e5007, 2024 Jun.
Article En | MEDLINE | ID: mdl-38723187

The identification of an effective inhibitor is an important starting step in drug development. Unfortunately, many issues such as the characterization of protein binding sites, the screening library, materials for assays, etc., make drug screening a difficult proposition. As the size of screening libraries increases, more resources will be inefficiently consumed. Thus, new strategies are needed to preprocess and focus a screening library towards a targeted protein. Herein, we report an ensemble machine learning (ML) model to generate a CDK8-focused screening library. The ensemble model consists of six different algorithms optimized for CDK8 inhibitor classification. The models were trained using a CDK8-specific fragment library along with molecules containing CDK8 activity. The optimized ensemble model processed a commercial library containing 1.6 million molecules. This resulted in a CDK8-focused screening library containing 1,672 molecules, a reduction of more than 99.90%. The CDK8-focused library was then subjected to molecular docking, and 25 candidate compounds were selected. Enzymatic assays confirmed six CDK8 inhibitors, with one compound producing an IC50 value of ≤100 nM. Analysis of the ensemble ML model reveals the role of the CDK8 fragment library during training. Structural analysis of molecules reveals the hit compounds to be structurally novel CDK8 inhibitors. Together, the results highlight a pipeline for curating a focused library for a specific protein target, such as CDK8.


Cyclin-Dependent Kinase 8 , Machine Learning , Molecular Docking Simulation , Protein Kinase Inhibitors , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 8/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Humans , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Drug Evaluation, Preclinical/methods
2.
Int J Mol Sci ; 25(10)2024 May 15.
Article En | MEDLINE | ID: mdl-38791449

Dysregulation of cyclin-dependent kinase 8 (CDK8) activity has been associated with many diseases, including colorectal and breast cancer. As usual in the CDK family, the activity of CDK8 is controlled by a regulatory protein called cyclin C (CycC). But, while human CDK family members are generally activated in two steps, that is, the binding of the cyclin to CDK and the phosphorylation of a residue in the CDK activation loop, CDK8 does not require the phosphorylation step to be active. Another peculiarity of CDK8 is its ability to be associated with CycC while adopting an inactive form. These specificities raise the question of the role of CycC in the complex CDK8-CycC, which appears to be more complex than the other members of the CDK family. Through molecular dynamics (MD) simulations and binding free energy calculations, we investigated the effect of CycC on the structure and dynamics of CDK8. In a second step, we particularly focused our investigation on the structural and molecular basis of the protein-protein interaction between the two partners by finely analyzing the energetic contribution of residues and simulating the transition between the active and the inactive form. We found that CycC has a stabilizing effect on CDK8, and we identified specific interaction hotspots within its interaction surface compared to other human CDK/Cyc pairs. Targeting these specific interaction hotspots could be a promising approach in terms of specificity to effectively disrupt the interaction between CDK8. The simulation of the conformational transition from the inactive to the active form of CDK8 suggests that the residue Glu99 of CycC is involved in the orientation of three conserved arginines of CDK8. Thus, this residue may assume the role of the missing phosphorylation step in the activation mechanism of CDK8. In a more general view, these results point to the importance of keeping the CycC in computational studies when studying the human CDK8 protein in both the active and the inactive form.


Cyclin C , Cyclin-Dependent Kinase 8 , Molecular Dynamics Simulation , Protein Binding , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 8/chemistry , Cyclin C/metabolism , Cyclin C/chemistry , Humans , Phosphorylation , Thermodynamics , Binding Sites
3.
Int J Mol Sci ; 21(20)2020 Oct 12.
Article En | MEDLINE | ID: mdl-33053834

Cyclin-dependent kinase 8 (CDK8) and its regulatory partner Cyclin C (CycC) play conserved roles in modulating RNA polymerase II (Pol II)-dependent gene expression. To understand the structure and function relations of CDK8, we analyzed the structures of human and Drosophila CDK8 proteins using molecular dynamics simulations, combined with functional analyses in Drosophila. Specifically, we evaluated the structural differences between hCDK8 and dCDK8 to predict the effects of the LXXLL motif mutation (AQKAA), the P154L mutations, and drug binding on local structures of the CDK8 proteins. First, we have observed that both the LXXLL motif and the kinase activity of CDK8 are required for the normal larval-to-pupal transition in Drosophila. Second, our molecular dynamic analyses have revealed that hCDK8 has higher hydrogen bond occupation of His149-Asp151 and Asp151-Asn156 than dCDK8. Third, the substructure of Asp282, Phe283, Arg285, Thr287 and Cys291 can distinguish human and Drosophila CDK8 structures. In addition, there are two hydrogen bonds in the LXXLL motif: a lower occupation between L312 and L315, and a relatively higher occupation between L312 and L316. Human CDK8 has higher hydrogen bond occupation between L312 and L316 than dCDK8. Moreover, L312, L315 and L316 in the LXXLL motif of CDK8 have the specific pattern of hydrogen bonds and geometries, which could be crucial for the binding to nuclear receptors. Furthermore, the P154L mutation dramatically decreases the hydrogen bond between L312 and L315 in hCDK8, but not in dCDK8. The mutations of P154L and AQKAA modestly alter the local structures around residues 154. Finally, we identified the inhibitor-induced conformational changes of hCDK8, and our results suggest a structural difference in the drug-binding site between hCDK8 and dCDK8. Taken together, these results provide the structural insights into the roles of the LXXLL motif and the kinase activity of CDK8 in vivo.


Amino Acid Motifs , Binding Sites , Cyclin-Dependent Kinase 8/chemistry , Drosophila Proteins/chemistry , Models, Molecular , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Drosophila Proteins/antagonists & inhibitors , Humans , Hydrogen Bonding , Ligands , Molecular Conformation , Mutation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Species Specificity , Structure-Activity Relationship
4.
Bioorg Chem ; 96: 103624, 2020 03.
Article En | MEDLINE | ID: mdl-32078847

A major goal of medicinal chemists is to identify and validate novel and effective kinase targets for treatment of cancer. Recent studies have shown that cyclin-dependent kinase 8 (CDK8) is a target for treatment of colorectal, breast, melanoma, and prostate cancers. The crystal structure of CDK8 has been reported, and eutectic interactions have been identified for 24 compounds that target CDK8. To more effectively develop CDK8 inhibitors, particularly those with improved selectivity, we summarized the structure, structure-activity relationships, and binding information of typical CDK8 inhibitors, which may serve as a reference for development of novel CDK8 inhibitors.


Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 8/metabolism , Drug Discovery , Humans , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/metabolism , Structure-Activity Relationship
5.
Proc Natl Acad Sci U S A ; 117(6): 2894-2905, 2020 02 11.
Article En | MEDLINE | ID: mdl-31988137

The Mediator kinase module regulates eukaryotic transcription by phosphorylating transcription-related targets and by modulating the association of Mediator and RNA polymerase II. The activity of its catalytic core, cyclin-dependent kinase 8 (CDK8), is controlled by Cyclin C and regulatory subunit MED12, with its deregulation contributing to numerous malignancies. Here, we combine in vitro biochemistry, cross-linking coupled to mass spectrometry, and in vivo studies to describe the binding location of the N-terminal segment of MED12 on the CDK8/Cyclin C complex and to gain mechanistic insights into the activation of CDK8 by MED12. Our data demonstrate that the N-terminal portion of MED12 wraps around CDK8, whereby it positions an "activation helix" close to the T-loop of CDK8 for its activation. Intriguingly, mutations in the activation helix that are frequently found in cancers do not diminish the affinity of MED12 for CDK8, yet likely alter the exact positioning of the activation helix. Furthermore, we find the transcriptome-wide gene-expression changes in human cells that result from a mutation in the MED12 activation helix to correlate with deregulated genes in breast and colon cancer. Finally, functional assays in the presence of kinase inhibitors reveal that binding of MED12 remodels the active site of CDK8 and thereby precludes the inhibition of ternary CDK8 complexes by type II kinase inhibitors. Taken together, our results not only allow us to propose a revised model of how CDK8 activity is regulated by MED12, but also offer a path forward in developing small molecules that target CDK8 in its MED12-bound form.


Cyclin-Dependent Kinase 8/metabolism , Mediator Complex/metabolism , Catalytic Domain , Cyclin C/genetics , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 8/genetics , Enzyme Activation , Humans , Mediator Complex/genetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains
6.
Eur J Med Chem ; 164: 77-91, 2019 Feb 15.
Article En | MEDLINE | ID: mdl-30594029

Cyclin-dependent kinases 8 (CDK8) regulates transcriptional process via associating with the mediator complex or phosphorylating transcription factors (TF). Overexpression of CDK8 has been observed in various cancers. It mediates aberrant activation of Wnt/ß-catenin signaling pathway, which is initially recognized and best studied in colorectal cancer (CRC). CDK8 acts as an oncogene and represents a potential target for developing novel CDK8 inhibitors in cancer therapeutics. However, other study has revealed its contrary role. The function of CDK8 is context dependent. Even so, a variety of potent and selective CDK8 inhibitors have been discovered after crystal structures were resolved in two states (active or inactive). In this review, we summarize co-crystal structures, biological mechanisms, dysregulation in cancers and recent progress in the field of CDK8 inhibitors, trying to offer an outlook of CDK8 inhibitors in cancer therapy in future.


Cyclin-Dependent Kinase 8/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Colorectal Neoplasms/drug therapy , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 8/metabolism , Drug Discovery , Humans , Mediator Complex/metabolism , Molecular Structure , Oncogenes , Wnt Signaling Pathway
7.
J Comput Aided Mol Des ; 32(6): 671-685, 2018 06.
Article En | MEDLINE | ID: mdl-29737445

Abnormal activity of cyclin-dependent kinase 8 (CDK8) along with its partner protein cyclin C (CycC) is a common feature of many diseases including colorectal cancer. Using molecular dynamics (MD) simulations, this study determined the dynamics of the CDK8-CycC system and we obtained detailed breakdowns of binding energy contributions for four type-I and five type-II CDK8 inhibitors. We revealed system motions and conformational changes that will affect ligand binding, confirmed the essentialness of CycC for inclusion in future computational studies, and provide guidance in development of CDK8 binders. We employed unbiased all-atom MD simulations for 500 ns on twelve CDK8-CycC systems, including apoproteins and protein-ligand complexes, then performed principal component analysis (PCA) and measured the RMSF of key regions to identify protein dynamics. Binding pocket volume analysis identified conformational changes that accompany ligand binding. Next, H-bond analysis, residue-wise interaction calculations, and MM/PBSA were performed to characterize protein-ligand interactions and find the binding energy. We discovered that CycC is vital for maintaining a proper conformation of CDK8 to facilitate ligand binding and that the system exhibits motion that should be carefully considered in future computational work. Surprisingly, we found that motion of the activation loop did not affect ligand binding. Type-I and type-II ligand binding is driven by van der Waals interactions, but electrostatic energy and entropic penalties affect type-II binding as well. Binding of both ligand types affects protein flexibility. Based on this we provide suggestions for development of tighter-binding CDK8 inhibitors and offer insight that can aid future computational studies.


Cyclin C/chemistry , Cyclin-Dependent Kinase 8/chemistry , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Allosteric Site , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Drug Discovery/methods , Ligands , Protein Binding , Protein Conformation , Static Electricity , Thermodynamics
8.
J Med Chem ; 61(12): 5073-5092, 2018 06 28.
Article En | MEDLINE | ID: mdl-29266937

Cyclin-dependent kinase 8 (CDK8) plays a vital role in regulating transcription either through its association with the Mediator complex or by phosphorylating transcription factors. Myriads of genetic and biochemical studies have established CDK8 as a key oncogenic driver in many cancers. Specifically, CDK8-mediated activation of oncogenic Wnt-ß-catenin signaling, transcription of estrogen-inducible genes, and suppression of super enhancer-associated genes contributes to oncogenesis in colorectal, breast, and hematological malignancies, respectively. However, while most research supports the role of CDK8 as an oncogene, other work has raised the possibility of its contrary function. The diverse biological functions of CDK8 and its seemingly context-specific roles in different types of cancers have spurred a great amount of interest and perhaps an even greater amount of controversy in the development of CDK8 inhibitors as potential cancer therapeutic agents. Herein, we review the latest landscape of CDK8 biology and its involvement in carcinogenesis. We dissect current efforts in discovering CDK8 inhibitors and attempt to provide an outlook at the future of CDK8-targeted cancer therapies.


Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/metabolism , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 8/genetics , Cytokines/genetics , Cytokines/metabolism , Humans , Male , Neoplasms/enzymology , Phosphorylation , Protein Kinase Inhibitors/chemistry , Transcription Factors/metabolism , Tumor Escape/physiology
9.
Oncotarget ; 8(20): 33779-33795, 2017 May 16.
Article En | MEDLINE | ID: mdl-28422713

Inhibition of oncogenic transcriptional programs is a promising therapeutic strategy. A substituted tricyclic benzimidazole, SEL120-34A, is a novel inhibitor of Cyclin-dependent kinase 8 (CDK8), which regulates transcription by associating with the Mediator complex. X-ray crystallography has shown SEL120-34A to be a type I inhibitor forming halogen bonds with the protein's hinge region and hydrophobic complementarities within its front pocket. SEL120-34A inhibits phosphorylation of STAT1 S727 and STAT5 S726 in cancer cells in vitro. Consistently, regulation of STATs- and NUP98-HOXA9- dependent transcription has been observed as a dominant mechanism of action in vivo. Treatment with the compound resulted in a differential efficacy on AML cells with elevated STAT5 S726 levels and stem cell characteristics. In contrast, resistant cells were negative for activated STAT5 and revealed lineage commitment. In vivo efficacy in xenotransplanted AML models correlated with significant repression of STAT5 S726. Favorable pharmacokinetics, confirmed safety and in vivo efficacy provide a rationale for the further clinical development of SEL120-34A as a personalized therapeutic approach in AML.


Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Protein Interaction Domains and Motifs/drug effects , Protein Kinase Inhibitors/pharmacology , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cyclin-Dependent Kinase 8/chemistry , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Models, Molecular , Molecular Conformation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemistry , STAT1 Transcription Factor/chemistry , STAT5 Transcription Factor/chemistry , Xenograft Model Antitumor Assays
10.
J Chem Inf Model ; 57(3): 413-416, 2017 03 27.
Article En | MEDLINE | ID: mdl-28191946

Aberrant activity of cyclin-dependent kinase (CDK) 8 is implicated in various cancers. While CDK8-targeting anticancer drugs are highly sought-after, no CDK8 inhibitor has yet reached clinical trials. Herein a large library of drug-like molecules was computationally screened using two complementary cascades to identify potential CDK8 inhibitors. Thirty-three hits were identified to inhibit CDK8 and seven of them were active against colorectal cancer cell lines. Finally, the primary target was confirmed using three promising hits.


Cyclin-Dependent Kinase 8/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinase 8/metabolism , Humans , Protein Kinase Inhibitors/metabolism , User-Computer Interface
11.
J Chem Inf Model ; 57(2): 159-169, 2017 02 27.
Article En | MEDLINE | ID: mdl-28080056

The duration of drug efficacy in vivo is a key aspect primarily addressed during the lead optimization phase of drug discovery. Hence, the availability of robust computational approaches that can predict the residence time of a compound at its target would accelerate candidate selection. Nowadays the theoretical prediction of this parameter is still very challenging. Starting from methods reported in the literature, we set up and validated a new metadynamics (META-D)-based protocol that was used to rank the experimental residence times of 10 arylpyrazole cyclin-dependent kinase 8 (CDK8) inhibitors for which target-bound X-ray structures are available. The application of reported methods based on the detection of the escape from the first free energy well gave a poor correlation with the experimental values. Our protocol evaluates the energetics of the whole unbinding process, accounting for multiple intermediates and transition states. Using seven collective variables (CVs) encoding both roto-translational and conformational motions of the ligand, a history-dependent biasing potential is deposited as a sum of constant-height Gaussian functions until the ligand reaches an unbound state. The time required to achieve this state is proportional to the integral of the deposited potential over the CV hyperspace. Average values of this time, for replicated META-D simulations, provided an accurate classification of CDK8 inhibitors spanning short, medium, and long residence times.


Cyclin-Dependent Kinase 8/antagonists & inhibitors , Molecular Dynamics Simulation , Protein Kinase Inhibitors/pharmacology , Cyclin-Dependent Kinase 8/chemistry , Protein Conformation , Protein Kinase Inhibitors/pharmacokinetics , Thermodynamics , Time Factors
13.
Methods Mol Biol ; 1336: 13-28, 2016.
Article En | MEDLINE | ID: mdl-26231705

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.


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
14.
Crit Rev Biochem Mol Biol ; 50(5): 393-426, 2015.
Article En | MEDLINE | ID: mdl-26182352

Mediator is a conserved multi-subunit signal processor through which regulatory informatiosn conveyed by gene-specific transcription factors is transduced to RNA Polymerase II (Pol II). In humans, MED13, MED12, CDK8 and Cyclin C (CycC) comprise a four-subunit "kinase" module that exists in variable association with a 26-subunit Mediator core. Genetic and biochemical studies have established the Mediator kinase module as a major ingress of developmental and oncogenic signaling through Mediator, and much of its function in signal-dependent gene regulation derives from its resident CDK8 kinase activity. For example, CDK8-targeted substrate phosphorylation impacts transcription factor half-life, Pol II activity and chromatin chemistry and functional status. Recent structural and biochemical studies have revealed a precise network of physical and functional subunit interactions required for proper kinase module activity. Accordingly, pathologic change in this activity through altered expression or mutation of constituent kinase module subunits can have profound consequences for altered signaling and tumor formation. Herein, we review the structural organization, biological function and oncogenic potential of the Mediator kinase module. We focus principally on tumor-associated alterations in kinase module subunits for which mechanistic relationships as opposed to strictly correlative associations are established. These considerations point to an emerging picture of the Mediator kinase module as an oncogenic unit, one in which pathogenic activation/deactivation through component change drives tumor formation through perturbation of signal-dependent gene regulation. It follows that therapeutic strategies to combat CDK8-driven tumors will involve targeted modulation of CDK8 activity or pharmacologic manipulation of dysregulated CDK8-dependent signaling pathways.


Carcinogenesis/metabolism , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/metabolism , Gene Expression Regulation, Neoplastic , Mediator Complex/metabolism , Models, Biological , Animals , Cyclin C/chemistry , Cyclin-Dependent Kinase 8/chemistry , Gene Expression Regulation, Developmental , Humans , Mediator Complex/chemistry , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism
15.
Comput Biol Chem ; 51: 1-11, 2014 Aug.
Article En | MEDLINE | ID: mdl-24754906

The Mediator, a conserved multisubunit protein complex in eukaryotic organisms, regulates gene expression by bridging sequence-specific DNA-binding transcription factors to the general RNA polymerase II machinery. In yeast, Mediator complex is organized in three core modules (head, middle and tail) and a separable 'CDK8 submodule' consisting of four subunits including Cyclin-dependent kinase CDK8 (CDK8), Cyclin C (CycC), MED12, and MED13. The 3-D structure of human CDK8-CycC complex has been recently experimentally determined. To take advantage of this structure and the improved theoretical calculation methods, we have performed molecular dynamic simulations to study dynamics of CDK8 and two CDK8 point mutations (D173A and D189N), which have been identified in human cancers, with and without full length of the A-loop, as well as the binding between CDK8 and CycC. We found that CDK8 structure gradually loses two helical structures during the 50-ns molecular dynamic simulation, likely due to the presence of the full-length A-loop. In addition, our studies showed the hydrogen bond occupation of the CDK8 A-loop increases during the first 20-ns MD simulation and stays stable during the later 30-ns MD simulation. Four residues in the A-loop of CDK8 have high hydrogen bond occupation, while the rest residues have low or no hydrogen bond occupation. The hydrogen bond dynamic study of the A-loop residues exhibits three types of changes: increasing, decreasing, and stable. Furthermore, the 3-D structures of CDK8 point mutations D173A, D189N, T196A and T196D have been built by molecular modeling and further investigated by 50-ns molecular dynamic simulations. D173A has the highest average potential energy, while T196D has the lowest average potential energy, indicating that T196D is the most stable structure. Finally, we calculated theoretical binding energy of CDK8 and CycC by MM/PBSA and MM/GBSA methods, and the negative values obtained from both methods demonstrate stability of CDK8-CycC complex. Taken together, these analyses will improve our understanding of the exact functions of CDK8 and the interaction with its partner CycC.


Alanine/chemistry , Asparagine/chemistry , Aspartic Acid/chemistry , Cyclin C/chemistry , Cyclin-Dependent Kinase 8/chemistry , Point Mutation , Amino Acid Sequence , Binding Sites , Cyclin C/genetics , Cyclin-Dependent Kinase 8/genetics , Gene Expression , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Thermodynamics
16.
Protein Cell ; 4(12): 911-20, 2013 Dec.
Article En | MEDLINE | ID: mdl-24043446

Mediator is a highly conserved large protein complex (25 proteins, >1000 kDa) and preeminently responsible for eukaryotic transcription, which contains a dissociable 'Cdk8 module'. Although increasing evidence demonstrates that Cdk8 module plays both positive and negative roles in transcription regulation, the detailed structure, and subunit organization, molecular mechanism how it regulates transcription remain elusive. Here we used single-particle electron microscopy to characterize the structure and subunit organization of the Cdk8 module and illuminated the substantial mobility of the Med13 subunit results in the structural flexibility. The Cdk8 module interaction with core Mediator is concurrent with active transcription in vivo. An interaction with the Cdk8 module induces core Mediator into very extended conformation in vitro, which is presumed to be an active functional state of Mediator. Taken together, our results illuminated the detailed architecture of Cdk8 module, and suggested the Cdk8 module could positively regulate transcription by modulating Mediator conformation.


Cyclin-Dependent Kinase 8/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Humans , Mediator Complex/chemistry , Models, Molecular , Protein Conformation , Protein Subunits/chemistry
17.
Cell ; 153(6): 1327-39, 2013 Jun 06.
Article En | MEDLINE | ID: mdl-23746844

The transcription factor HIF1A is a key mediator of the cellular response to hypoxia. Despite the importance of HIF1A in homeostasis and various pathologies, little is known about how it regulates RNA polymerase II (RNAPII). We report here that HIF1A employs a specific variant of the Mediator complex to stimulate RNAPII elongation. The Mediator-associated kinase CDK8, but not the paralog CDK19, is required for induction of many HIF1A target genes. HIF1A induces binding of CDK8-Mediator and the super elongation complex (SEC), containing AFF4 and CDK9, to alleviate RNAPII pausing. CDK8 is dispensable for HIF1A chromatin binding and histone acetylation, but it is essential for binding of SEC and RNAPII elongation. Global analysis of active RNAPII reveals that hypoxia-inducible genes are paused and active prior to their induction. Our results provide a mechanistic link between HIF1A and CDK8, two potent oncogenes, in the cellular response to hypoxia.


Cell Hypoxia , Cyclin-Dependent Kinase 8/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mediator Complex/metabolism , Neoplasms/metabolism , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Acetylation , Cell Line, Tumor , Cyclin-Dependent Kinase 8/chemistry , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Histones/metabolism , Humans
18.
Proc Natl Acad Sci U S A ; 110(20): 8081-6, 2013 May 14.
Article En | MEDLINE | ID: mdl-23630251

In contrast with the very well explored concept of structure-activity relationship, similar studies are missing for the dependency between binding kinetics and compound structure of a protein ligand complex, the structure-kinetic relationship. Here, we present a structure-kinetic relationship study of the cyclin-dependent kinase 8 (CDK8)/cyclin C (CycC) complex. The scaffold moiety of the compounds is anchored in the kinase deep pocket and extended with diverse functional groups toward the hinge region and the front pocket. These variations can cause the compounds to change from fast to slow binding kinetics, resulting in an improved residence time. The flip of the DFG motif ("DMG" in CDK8) to the inactive DFG-out conformation appears to have relatively little influence on the velocity of binding. Hydrogen bonding with the kinase hinge region contributes to the residence time but has less impact than hydrophobic complementarities within the kinase front pocket.


Cyclin C/chemistry , Cyclin-Dependent Kinase 8/chemistry , Amino Acid Motifs , Catalytic Domain , Crystallography, X-Ray , Drug Design , Humans , Hydrogen Bonding , Kinetics , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Salts/chemistry , Temperature , Time Factors
19.
Nat Struct Mol Biol ; 20(5): 611-9, 2013 May.
Article En | MEDLINE | ID: mdl-23563140

The CDK8 kinase module (CKM) is a conserved, dissociable Mediator subcomplex whose component subunits were genetically linked to the RNA polymerase II (RNAPII) C-terminal domain (CTD) and individually recognized as transcriptional repressors before Mediator was identified as a pre-eminent complex in eukaryotic transcription regulation. We used macromolecular EM and biochemistry to investigate the subunit organization, structure and Mediator interaction of the Saccharomyces cerevisiae CKM. We found that interaction of the CKM with Mediator's middle module interferes with CTD-dependent RNAPII binding to a previously unknown middle-module CTD-binding site and with the holoenzyme formation process. Taken together, our results reveal the basis for CKM repression, clarify the origin of the connection between CKM subunits and the CTD and suggest that a combination of competitive interactions and conformational changes that facilitate holoenzyme formation underlie the mechanism of transcription regulation by Mediator.


Cyclin-Dependent Kinase 8/metabolism , Gene Expression Regulation, Fungal , Mediator Complex/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cyclin-Dependent Kinase 8/chemistry , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Mediator Complex/chemistry , Microscopy, Electron , Protein Binding , RNA Polymerase II/chemistry , Saccharomyces cerevisiae Proteins/chemistry
20.
J Mol Biol ; 412(2): 251-66, 2011 Sep 16.
Article En | MEDLINE | ID: mdl-21806996

Cyclin-dependent kinase (CDK) 8 associates with cyclin C (CycC) and belongs to the CDK module of the Mediator of transcription, together with MED12 and MED13. CDK8 is involved in the regulation of mRNA transcription and was identified as a potent oncogene in colon cancerogenesis. We have solved the 2.2-Å crystal structure of CDK8/CycC in complex with sorafenib, an anti-cancer drug of clinical relevance. The CDK8 structure reveals a unique CycC recognition helix that explains the specificity of the CDK8/CycC pair and discrimination among the highly promiscuous binding in the CDK/cyclin family. In contrast to all CDKs, the CDK8 activation loop appears not to be phosphorylated. Based on the structure, we discuss an alternate mode of CDK8 activation to the general CDK activation by T-loop phosphorylation. Sorafenib binds to the catalytic cleft of CDK8. It displays a deep pocket binding mode and is the first small molecule to induce a DFG-out conformation in the CDK family, which is actually DMG-out in CDK8.


Cyclin C/chemistry , Cyclin-Dependent Kinase 8/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/metabolism , Cyclins/chemistry , Enzyme Activation , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
...