Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Drug Dev Res ; 82(8): 1131-1143, 2021 12.
Article in English | MEDLINE | ID: mdl-33818788

ABSTRACT

Circular RNA FAT atypical cadherin 1 (circFAT1) has been reported to play vital roles in the progression of some cancers. However, the regulatory role and underlying mechanisms of circFAT1 in cervical cancer (CC) remain largely unknown. The expression of circFAT1, microRNA (miR)-409-3p and cyclin-dependent kinase 8 (CDK8) was detected using qRT-PCR and Western blot assays. Cell proliferation, apoptosis, migration and invasion in vitro were investigated using cell counting kit-8, colony formation, flow cytometry, and transwell assays, respectively. Western blot assay was used to determine the activation of ERK1/2 and p38 MAPK pathway. The interaction miR-409-3p and circFAT1 or CDK8 was confirmed by dual-luciferase reporter, pull-down or RIP assays. The effects of circFAT1 in vivo were determined using xenograft models. CircFAT1 was highly expressed in CC, and closely associated with poor prognosis. CircFAT1 knockdown resulted in the suppression of proliferation, migration and invasion, and promotion of apoptosis in CC cells via the inactivation of ERK1/2 and p38 MAPK pathway; also, circFAT1 silencing could inactivate this pathway and repressed CC tumor growth in vivo. Mechanistic analysis showed that circFAT1 directly sponged miR-409-3p and then relieved the repressive effect of miR-409-3p on its target CDK8. Furthermore, miR-409-3p inhibition reversed the effects of circFAT1 silencing on CC cells. Whereas, miR-409-3p overexpression impeded CC cell growth and motility, which was attenuated by CDK8. CircFAT1 promoted CC progression via activating ERK1/2 and p38 MAPK pathway through the miR-409-3p/CDK8 axis, suggesting a promising prognostic biomarker and therapeutic target for CC.


Subject(s)
Cadherins/genetics , Cyclin-Dependent Kinase 8/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , MicroRNAs/physiology , RNA, Circular/physiology , Uterine Cervical Neoplasms/etiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 8/genetics , Disease Progression , Female , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred BALB C
2.
Sci Rep ; 10(1): 17575, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33067521

ABSTRACT

Cyclin-dependent kinase 8 (CDK8) is a member of the CDK/Cyclin module of the mediator complex. A recent study reported that heterozygous missense CDK8 mutations cause a neurodevelopmental disorder in humans. The mechanistic basis of CDK8-related disorder has yet to be delineated. Here, we report 2 patients with de novo missense mutations within the kinase domain of CDK8 along with the results of in vitro and in vivo functional analyses using a zebrafish model. Patient 1 and Patient 2 had intellectual disabilities and congenital anomalies. Exome analyses showed that patient 1 had a heterozygous de novo missense p.G28A variant in the CDK8 (NM_001260.3) gene and patient 2 had a heterozygous de novo missense p.N156S variant in the CDK8 gene. We assessed the pathogenicity of these two variants using cultured-cells and zebrafish model. An in vitro kinase assay of human CDK8 showed that enzymes with a p.G28A or p.N156S substitution showed decreased kinase activity. An in vivo assays of zebrafish overexpression analyses also showed that the p.G28A and p.N156S alleles were hypomorphic alleles. Importantly, the inhibition of CDK8 kinase activity in zebrafish embryos using a specific chemical inhibitor induced craniofacial and heart defects similar to the patients' phenotype. Taken together, zebrafish studies showed that non-synonymous variants in the kinase domain of CDK8 act as hypomorphic alleles causing human congenital disorder.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Mutation, Missense , Neurodevelopmental Disorders/genetics , Point Mutation , Abnormalities, Multiple/genetics , Animals , Child , Craniofacial Abnormalities/genetics , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/deficiency , Cyclin-Dependent Kinase 8/physiology , Cyclin-Dependent Kinases/physiology , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/enzymology , Female , Heart Defects, Congenital/genetics , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Infant , Intellectual Disability/genetics , Loss of Function Mutation , Male , Protein Domains , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Zebrafish/embryology , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/physiology
3.
Cells ; 8(8)2019 08 03.
Article in English | MEDLINE | ID: mdl-31382571

ABSTRACT

CDK8 and CDK19 Mediator kinases are transcriptional co-regulators implicated in several types of cancer. Small-molecule CDK8/19 inhibitors have recently entered or are entering clinical trials, starting with breast cancer and acute myeloid leukemia (AML). To identify other cancers where these novel drugs may provide benefit, we queried genomic and transcriptomic databases for potential impact of CDK8, CDK19, or their binding partner CCNC. sgRNA analysis of a panel of tumor cell lines showed that most tumor types represented in the panel, except for some central nervous system tumors, were not dependent on these genes. In contrast, analysis of clinical samples for alterations in these genes revealed a high frequency of gene amplification in two highly aggressive subtypes of prostate cancer and in some cancers of the GI tract, breast, bladder, and sarcomas. Analysis of survival correlations identified a group of cancers where CDK8 expression correlated with shorter survival (notably breast, prostate, cervical cancers, and esophageal adenocarcinoma). In some cancers (AML, melanoma, ovarian, and others), such correlations were limited to samples with a below-median tumor mutation burden. These results suggest that Mediator kinases are especially important in cancers that are driven primarily by transcriptional rather than mutational changes and warrant an investigation of their role in additional cancer types.


Subject(s)
Cyclin C/physiology , Cyclin-Dependent Kinase 8/physiology , Cyclin-Dependent Kinases/physiology , Neoplasms/metabolism , Cell Line, Tumor , Cyclin C/antagonists & inhibitors , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/drug therapy , Neoplasms/genetics
4.
PLoS Biol ; 17(1): e2006767, 2019 01.
Article in English | MEDLINE | ID: mdl-30695077

ABSTRACT

Accurate genome duplication underlies genetic homeostasis. Metazoan Mdm2 binding protein (MTBP) forms a main regulatory platform for origin firing together with Treslin/TICRR and TopBP1 (Topoisomerase II binding protein 1 (TopBP1)-interacting replication stimulating protein/TopBP1-interacting checkpoint and replication regulator). We report the first comprehensive analysis of MTBP and reveal conserved and metazoa-specific MTBP functions in replication. This suggests that metazoa have evolved specific molecular mechanisms to adapt replication principles conserved with yeast to the specific requirements of the more complex metazoan cells. We uncover one such metazoa-specific process: a new replication factor, cyclin-dependent kinase 8/19-cyclinC (Cdk8/19-cyclin C), binds to a central domain of MTBP. This interaction is required for complete genome duplication in human cells. In the absence of MTBP binding to Cdk8/19-cyclin C, cells enter mitosis with incompletely duplicated chromosomes, and subsequent chromosome segregation occurs inaccurately. Using remote homology searches, we identified MTBP as the metazoan orthologue of yeast synthetic lethal with Dpb11 7 (Sld7). This homology finally demonstrates that the set of yeast core factors sufficient for replication initiation in vitro is conserved in metazoa. MTBP and Sld7 contain two homologous domains that are present in no other protein, one each in the N and C termini. In MTBP the conserved termini flank the metazoa-specific Cdk8/19-cyclin C binding region and are required for normal origin firing in human cells. The N termini of MTBP and Sld7 share an essential origin firing function, the interaction with Treslin/TICRR or its yeast orthologue Sld3, respectively. The C termini may function as homodimerisation domains. Our characterisation of broadly conserved and metazoa-specific initiation processes sets the basis for further mechanistic dissection of replication initiation in vertebrates. It is a first step in understanding the distinctions of origin firing in higher eukaryotes.


Subject(s)
Carrier Proteins/metabolism , Carrier Proteins/physiology , Saccharomyces cerevisiae Proteins/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , Computational Biology/methods , Cyclin C/genetics , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 8/physiology , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/physiology , DNA Replication/physiology , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mitosis , Protein Binding , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
5.
Dev Biol ; 444(2): 62-70, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30352217

ABSTRACT

The complex interplay between genetic and environmental factors, such as diet and lifestyle, defines the initiation and progression of multifactorial diseases, including cancer, cardiovascular and metabolic diseases, and neurological disorders. Given that most of the studies have been performed in controlled experimental settings to ensure the consistency and reproducibility, the impacts of environmental factors, such as dietary perturbation, on the development of animals with different genotypes and the pathogenesis of these diseases remain poorly understood. By analyzing the cdk8 and cyclin C (cycC) mutant larvae in Drosophila, we have previously reported that the CDK8-CycC complex coordinately regulates lipogenesis by repressing dSREBP (sterol regulatory element-binding protein)-activated transcription and developmental timing by activating EcR (ecdysone receptor)-dependent gene expression. Here we report that dietary nutrients, particularly proteins and carbohydrates, modulate the developmental timing through the CDK8/CycC/EcR pathway. We observed that cdk8 and cycC mutants are sensitive to the levels of dietary proteins and seven amino acids (arginine, glutamine, isoleucine, leucine, methionine, threonine, and valine). Those mutants are also sensitive to dietary carbohydrates, and they are more sensitive to monosaccharides than disaccharides. These results suggest that CDK8-CycC mediates the dietary effects on lipid metabolism and developmental timing in Drosophila larvae.


Subject(s)
Cyclin-Dependent Kinase 8/physiology , Drosophila Proteins/physiology , Larva/metabolism , Nutritional Requirements/physiology , Animals , Cyclin C/metabolism , Cyclin C/physiology , Cyclin-Dependent Kinase 8/metabolism , Diet , Dietary Proteins/metabolism , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/metabolism , Gene Expression , Reproducibility of Results
6.
Nucleic Acids Res ; 45(17): 10056-10067, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973445

ABSTRACT

ADP-ribosyltransferases promote repair of DNA single strand breaks and disruption of this pathway by Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) is toxic to cells with defects in homologous recombination (HR). Here, we show that this relationship is conserved in the simple eukaryote Dictyostelium and exploit this organism to define mechanisms that drive resistance of the HR-deficient cells to PARPi. Dictyostelium cells disrupted in exonuclease I, a critical factor for HR, are sensitive to PARPi. Deletion of exo1 prevents the accumulation of Rad51 in chromatin induced by PARPi, resulting in DNA damage being channelled through repair by non-homologous end-joining (NHEJ). Inactivation of NHEJ supresses the sensitivity of exo1- cells to PARPi, indicating this pathway drives synthetic lethality and that in its absence alternative repair mechanisms promote cell survival. This resistance is independent of alternate-NHEJ and is instead achieved by re-activation of HR. Moreover, inhibitors of Mre11 restore sensitivity of dnapkcs-exo1- cells to PARPi, indicating redundancy between nucleases that initiate HR can drive PARPi resistance. These data inform on mechanism of PARPi resistance in HR-deficient cells and present Dictyostelium as a convenient genetic model to characterize these pathways.


Subject(s)
ADP Ribose Transferases/physiology , Dictyostelium/enzymology , Drug Resistance/physiology , Homologous Recombination/physiology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/physiology , Protozoan Proteins/physiology , Benzamides/pharmacology , Clone Cells , Cyclin-Dependent Kinase 8/deficiency , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/physiology , DNA Damage , Dictyostelium/drug effects , Dictyostelium/genetics , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/physiology , Gene Deletion , Indoles/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Quinazolines/pharmacology , Rad51 Recombinase/deficiency , Rad51 Recombinase/physiology , Recombinant Proteins/metabolism
7.
Genes Cells ; 22(3): 265-276, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28151579

ABSTRACT

In eukaryotes, the Mediator complex has important roles in regulation of transcription by RNA polymerase II. Mediator is a large complex with more than 20 subunits that form head, middle, tail and CDK/cyclin modules. Among them, CDK8 and/or CDK19 (CDK8/19), and their counterpart cyclin C, form the CDK/cyclin module together with Mediator subunits MED12 and MED13. Despite evidences of both activation and repression, the precise functional roles of CDK8/19 in transcription are still elusive. Our previous results indicate that CDK8/19 recruits epigenetic regulators to repress immunoresponse genes. Here, this study focused on Toll-like receptors (TLRs), which exert innate immune responses through recognition of pathogen-associated molecular patterns and examined the functional roles of CDK8/19. As a result, CDK8/19 regulated transcription of inflammatory genes on stimulation of TLR9 in myeloma-derived RPMI8226 cells, which led to expression of inflammation-associated genes such as IL8, IL10, PTX3 and CCL2. Mediator subunits CDK8/19 and MED1, inflammation-related transcriptional activator NF-κB and C/EBPß, and general transcription factors TFIIE and TFIIB colocalized at the promoter regions of these genes under this condition. Our results show that CDK8/19 positively regulates inflammatory gene transcription in cooperation with NF-κB and C/EBPß on stimulation of TLR9.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclin-Dependent Kinase 8/physiology , Cyclin-Dependent Kinases/physiology , NF-kappa B/metabolism , Toll-Like Receptor 9/physiology , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Transcriptional Activation
8.
Biochim Biophys Acta ; 1845(1): 66-83, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24342527

ABSTRACT

Mediator complex (MED) is an evolutionarily conserved multiprotein, fundamental for growth and survival of all cells. In eukaryotes, the mRNA transcription is dependent on RNA polymerase II that is associated to various molecules like general transcription factors, MED subunits and chromatin regulators. To date, transcriptional machinery dysfunction has been shown to elicit broad effects on cell proliferation, development, differentiation, and pathologic disease induction, including cancer. Indeed, in malignant cells, the improper activation of specific genes is usually ascribed to aberrant transcription machinery. Here, we focus our attention on the correlation of MED subunits with carcinogenesis. To date, many subunits are mutated or display altered expression in human cancers. Particularly, the role of MED1, MED28, MED12, CDK8 and Cyclin C in cancer is well documented, although several studies have recently reported a possible association of other subunits with malignancy. Definitely, a major comprehension of the involvement of the whole complex in cancer may lead to the identification of MED subunits as novel diagnostic/prognostic tumour markers to be used in combination with imaging technique in clinical oncology, and to develop novel anti-cancer targets for molecular-targeted therapy.


Subject(s)
Mediator Complex/physiology , Neoplasms/etiology , Cyclin C/physiology , Cyclin-Dependent Kinase 8/physiology , Humans , Mediator Complex Subunit 1/physiology
9.
Biochim Biophys Acta ; 1829(9): 916-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23643644

ABSTRACT

Cyclin dependent kinase 8 (Cdk8) is a component of Mediator, an evolutionary conserved multiprotein complex that regulates RNA polymerase II-dependent transcription. Cdk8 has been implicated as a regulator of multiple steps in cell cycle progression. We here discuss recent advances in our understanding of Cdk8 function and a possible role for Mediator as a hub for integrating transcription regulation with cell cycle progression.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase 8/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/cytology , RNA Polymerase II/metabolism
10.
Oncogene ; 32(30): 3520-30, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-22945643

ABSTRACT

Activation of the Wnt/ß-catenin pathway is a critical step in the development of colorectal cancers. A key mediator of this activation is the recently described oncogene CDK8, which is amplified in a large number of colorectal tumors. CDK8 affects ß-catenin activation by interaction of the CDK8 submodule of the mediator complex with ß-catenin/TCF transcriptional complex, and by CDK8 interacting with and phosphorylating E2F1, which acts as a repressor of ß-catenin/TCF transcriptional activity. The amino-acid residue in E2F1 that CDK8 phosphorylates and how this phosphorylation impacts E2F1 activity in general is not known. Here, we describe that CDK8 phosphorylates serine 375 in E2F1 both in vitro and in cells, and that phosphorylation of this residue is required for E2F1 interaction with CDK8, and that the phosphorylation is dependent on CDK8 kinase activity. The phosphorylation of S375 by CDK8 regulates E2F1 ability to repress transcription of ß-catenin/TCF-dependent genes, as well as activation of E2F1-dependent genes. This regulation is due to inactivation of E2F1 transcriptional activation, and not to the interference of E2F1's ability to bind to E2F1-binding sites in various promoters or to interact with DP1.


Subject(s)
Cyclin-Dependent Kinase 8/physiology , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/physiology , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , E2F1 Transcription Factor/chemistry , Humans , Phosphorylation/genetics , Serine/metabolism , Transcriptional Activation/genetics
11.
Proc Natl Acad Sci U S A ; 109(34): 13799-804, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22869755

ABSTRACT

Conventional chemotherapy not only kills tumor cells but also changes gene expression in treatment-damaged tissues, inducing production of multiple tumor-supporting secreted factors. This secretory phenotype was found here to be mediated in part by a damage-inducible cell-cycle inhibitor p21 (CDKN1A). We developed small-molecule compounds that inhibit damage-induced transcription downstream of p21. These compounds were identified as selective inhibitors of a transcription-regulating kinase CDK8 and its isoform CDK19. Remarkably, p21 was found to bind to CDK8 and stimulate its kinase activity. p21 and CDK8 also cooperate in the formation of internucleolar bodies, where both proteins accumulate. A CDK8 inhibitor suppresses damage-induced tumor-promoting paracrine activities of tumor cells and normal fibroblasts and reverses the increase in tumor engraftment and serum mitogenic activity in mice pretreated with a chemotherapeutic drug. The inhibitor also increases the efficacy of chemotherapy against xenografts formed by tumor cell/fibroblast mixtures. Microarray data analysis revealed striking correlations between CDK8 expression and poor survival in breast and ovarian cancers. CDK8 inhibition offers a promising approach to increasing the efficacy of cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 8/physiology , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Nucleolus/metabolism , Cellular Senescence , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Genomics , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , S-Phase Kinase-Associated Proteins/metabolism , Transcription, Genetic , Treatment Outcome
12.
J Clin Invest ; 122(7): 2417-27, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22684109

ABSTRACT

Altered lipid metabolism underlies several major human diseases, including obesity and type 2 diabetes. However, lipid metabolism pathophysiology remains poorly understood at the molecular level. Insulin is the primary stimulator of hepatic lipogenesis through activation of the SREBP-1c transcription factor. Here we identified cyclin-dependent kinase 8 (CDK8) and its regulatory partner cyclin C (CycC) as negative regulators of the lipogenic pathway in Drosophila, mammalian hepatocytes, and mouse liver. The inhibitory effect of CDK8 and CycC on de novo lipogenesis was mediated through CDK8 phosphorylation of nuclear SREBP-1c at a conserved threonine residue. Phosphorylation by CDK8 enhanced SREBP-1c ubiquitination and protein degradation. Importantly, consistent with the physiologic regulation of lipid biosynthesis, CDK8 and CycC proteins were rapidly downregulated by feeding and insulin, resulting in decreased SREBP-1c phosphorylation. Moreover, overexpression of CycC efficiently suppressed insulin and feeding-induced lipogenic gene expression. Taken together, these results demonstrate that CDK8 and CycC function as evolutionarily conserved components of the insulin signaling pathway in regulating lipid homeostasis.


Subject(s)
Cyclin-Dependent Kinase 8/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Lipogenesis , Sterol Regulatory Element Binding Protein 1/metabolism , Amino Acid Motifs , Animals , Cell Line , Cell Nucleus/metabolism , Cyclin C/genetics , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Fasting/metabolism , Fat Body/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Hepatocytes/metabolism , Humans , Larva/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phosphorylation , Primary Cell Culture , Protein Processing, Post-Translational , Protein Stability , RNA Interference , Rats
13.
Int J Oncol ; 38(5): 1375-83, 2011 May.
Article in English | MEDLINE | ID: mdl-21344156

ABSTRACT

Gastric adenocarcinoma is a common cause of cancer-related death. The Wnt/ß-catenin pathway plays an important role in various cancers. However, relatively little is known about the regulatory mechanism of ß-catenin in stomach cancer. To determine the patterns of cyclin-dependent kinase (CDK) 8 and ß-catenin expression and the relationship between CDK8 and ß-catenin, we conducted a study of immuno-histochemical staining of tumor tissues (12 adenomas, 24 early gastric carcinomas, 24 advanced gastric carcinomas and 21 metastatic lymph nodes), together with Western blot analysis and CDK8 interference studies using gastric cancer cell lines. Gastric adenocarcinomas with CDK8 expression had distinct clinical, prognostic and molecular attributes. CDK8 expression and the delocalization of ß-catenin expression showed a significant positive correlation with carcinogenesis and tumor progression, especially lymph node metastasis. Immunohisto-chemically, CDK8 expression in gastric adenocarcinoma was independently associated with ß-catenin activation (p<0.05). ß-catenin expression was suppressed by CDK8 interference in the gastric adenocarcinoma cell lines, SNU-601 and SNU-638. These data support the potential link between CDK8 and ß-catenin, and suggest that CDK8 detection and ß-catenin delocalization could be related to a poor prognosis. Moreover, the interference of CDK8 could be a promising therapeutic modality for gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/etiology , Cyclin-Dependent Kinase 8/physiology , Stomach Neoplasms/etiology , beta Catenin/physiology , Adult , Aged , Aged, 80 and over , Cyclin-Dependent Kinase 8/analysis , Disease Progression , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , beta Catenin/analysis
14.
Oncol Rep ; 24(1): 285-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20514474

ABSTRACT

Colorectal adenocarcinoma is a major cause of morbidity and mortality. The Wnt/beta-catenin pathway plays an important role in colon cancers. However, relatively little is known about the regulatory mechanism of beta-catenin in colon cancers. CDK8 is a cyclin-dependent kinase (CDK) member of the mediator complex that couples transcriptional regulators to the basal transcriptional machinery, and is implicated in the transcriptional regulation of key pathways involved in colon cancers. To determine the relationship between CDK8 and beta-catenin expressions, a population-based study was conducted for immunohistochemical staining analysis of tumor tissues, and Western blot analysis and CDK8 interference studies of colon cancer cell lines. The hypothesis that colorectal cancers with CDK8 expression have distinct clinical, prognostic and molecular attributes was tested. Among 127 colorectal cancers, CDK8 expression was detected in 96 (76%) tumors by immunohistochemistry. CDK8 and beta-catenin expression had significant positive correlation with carcinogenesis, tumor progression and patient survival. Immunohistochemically, CDK8 expression in colorectal cancer was independently associated with beta-catenin activation (P=0.0002). However, beta-catenin expression was not completely suppressed by CDK8 interference in the colon cancer cell lines HCT-116, HT-29 and SNU-C5. These data support a potential link between CDK8 and beta-catenin, and suggest that CDK8 may identify a subset of colon cancer patients with a poor prognosis. However, control of CDK8 is not an effective therapeutic strategy through beta-catenin regulation of general colon cancer.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Cyclin-Dependent Kinase 8/physiology , beta Catenin/physiology , Adenocarcinoma/diagnosis , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Case-Control Studies , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/metabolism , Drug Evaluation, Preclinical , Female , HCT116 Cells , HT29 Cells , Humans , Male , Prognosis , RNA, Small Interfering/pharmacology , Survival Analysis , Tumor Cells, Cultured , beta Catenin/metabolism
16.
Nat Struct Mol Biol ; 17(2): 194-201, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20098423

ABSTRACT

The Mediator complex allows communication between transcription factors and RNA polymerase II (RNAPII). Cyclin-dependent kinase 8 (CDK8), the kinase found in some variants of Mediator, has been characterized mostly as a transcriptional repressor. Recently, CDK8 was demonstrated to be a potent oncoprotein. Here we show, using a human tumor cell line, that CDK8 is a positive regulator of genes within the serum response network, including several members of the activator protein 1 and early growth response family of oncogenic transcription factors. Mechanistic studies show that CDK8 is not required for RNAPII recruitment or promoter escape. Instead, CDK8 depletion leads to the appearance of slower elongation complexes carrying hypophosphorylated RNAPII. CDK8-Mediator regulates precise steps in the assembly of the RNAPII elongation complex, including the recruitment of positive transcription elongation factor b and BRD4. Furthermore, CDK8-Mediator specifically interacts with positive transcription elongation factor b. Thus, we have uncovered a role for CDK8 in transcriptional regulation that may contribute to its oncogenic effects.


Subject(s)
Cyclin-Dependent Kinase 8/physiology , Serum Response Factor/metabolism , Trans-Activators/physiology , Transcription, Genetic , Cell Cycle Proteins , Cell Line, Tumor , Cyclin-Dependent Kinase 8/genetics , Gene Knockdown Techniques , Humans , Nuclear Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding , Protein Interaction Mapping , RNA Polymerase II/metabolism , Serum Response Element , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism
17.
Cancer Res ; 69(20): 7899-901, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19808961

ABSTRACT

The Wnt/beta-catenin pathway plays an important role in initiation in most, if not all, colon cancers. Prior work has provided important insights into the regulation of beta-catenin stability in the cytoplasm; however, relatively little is known about the mechanism by which beta-catenin activates gene transcription in the nucleus. Using genetic approaches, studies in human colon cancers and Drosophila have identified CDK8 as a colon cancer oncogene that regulates beta-catenin transcriptional activity. These convergent observations provide new insights into the regulation of nuclear beta-catenin activity and identify a novel therapeutic target for beta-catenin-driven malignancies.


Subject(s)
Cyclin-Dependent Kinase 8/physiology , Gene Expression Regulation/physiology , Neoplasms/genetics , Oncogenes/physiology , beta Catenin/genetics , Animals , Humans , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL