scispace - formally typeset
Search or ask a question

Showing papers on "Cyclin-dependent kinase 8 published in 2019"


Journal ArticleDOI
15 Mar 2019
TL;DR: This review discusses cellular roles of the Mediator kinases and mechanisms that enable their biological functions, and how CDK8 or CDK19 may enable metabolic and transcriptional reprogramming through enhancers and chromatin looping.
Abstract: The Mediator-associated kinases CDK8 and CDK19 function in the context of three additional proteins: CCNC and MED12, which activate CDK8/CDK19 kinase function, and MED13, which enables their association with the Mediator complex. The Mediator kinases affect RNA polymerase II (pol II) transcription indirectly, through phosphorylation of transcription factors and by controlling Mediator structure and function. In this review, we discuss cellular roles of the Mediator kinases and mechanisms that enable their biological functions. We focus on sequence-specific, DNA-binding transcription factors and other Mediator kinase substrates, and how CDK8 or CDK19 may enable metabolic and transcriptional reprogramming through enhancers and chromatin looping. We also summarize Mediator kinase inhibitors and their therapeutic potential. Throughout, we note conserved and divergent functions between yeast and mammalian CDK8, and highlight many aspects of kinase module function that remain enigmatic, ranging from potential roles in pol II promoter-proximal pausing to liquid-liquid phase separation.

76 citations


Journal ArticleDOI
TL;DR: These studies reveal a mechanism in which NPR1 recruits CDK8, WRKY18, and TGA transcription factors along with RNA polymerase II in the presence of SA and thereby facilitates its own and target gene expression for the establishment of plant immunity.
Abstract: NPR1 (NONEXPRESSER OF PR GENES1) functions as a master regulator of the plant hormone salicylic acid (SA) signaling and plays an essential role in plant immunity. In the nucleus, NPR1 interacts with transcription factors to induce the expression of PR (PATHOGENESIS-RELATED) genes and thereby promote defense responses. However, the underlying molecular mechanism of PR gene activation is poorly understood. Furthermore, despite the importance of NPR1 in plant immunity, the regulation of NPR1 expression has not been extensively studied. Here, we show that SA promotes the interaction of NPR1 with both CDK8 (CYCLIN-DEPENDENT KINASE8) and WRKY18 (WRKY DNA-BINDING PROTEIN18) in Arabidopsis (Arabidopsis thaliana). NPR1 recruits CDK8 and WRKY18 to the NPR1 promoter, facilitating its own expression. Intriguingly, CDK8 and its associated Mediator subunits positively regulate NPR1 and PR1 expression and play a pivotal role in local and systemic immunity. Moreover, CDK8 interacts with WRKY6, WRKY18, and TGA transcription factors and brings RNA polymerase II to NPR1 and PR1 promoters and coding regions to facilitate their expression. Our studies reveal a mechanism in which NPR1 recruits CDK8, WRKY18, and TGA transcription factors along with RNA polymerase II in the presence of SA and thereby facilitates its own and target gene expression for the establishment of plant immunity.

62 citations


Journal ArticleDOI
TL;DR: Context and function of the Mediator kinases are reviewed at a molecular, cellular and animal level to illuminate emerging concepts underpinning Mediator Kinase biology and highlight certain aspects that remain unsolved.
Abstract: CDK8 and its paralog, CDK19, collectively termed 'Mediator Kinase,' are cyclin-dependent kinases that have been implicated as key rheostats in cellular homeostasis and developmental programming. CDK8 and CDK19 are incorporated, in a mutually exclusive manner, as part of a 4-protein complex called the Mediator kinase module. This module reversibly associates with the Mediator, a 26 subunit protein complex that regulates RNA Polymerase II mediated gene expression. As part of this complex, the Mediator kinases have been implicated in diverse process such as developmental signaling, metabolic homeostasis and in innate immunity. In recent years, dysregulation of Mediator kinase module proteins, including CDK8/19, has been implicated in the development of different human diseases, and in particular cancer. This has led to intense efforts to understand how CDK8/19 regulate diverse biological outputs and develop Mediator kinase inhibitors that can be exploited therapeutically. Herein, we review both context and function of the Mediator kinases at a molecular, cellular and animal level. In so doing, we illuminate emerging concepts underpinning Mediator kinase biology and highlight certain aspects that remain unsolved.

50 citations


Journal ArticleDOI
TL;DR: CDK8 kinase activity promotes RNA polymerase II pause release in response to interferon-γ (IFN-γ), a universal cytokine involved in immunity and tumor surveillance and implicate CDK8 and CDK19 as essential for this transcriptional reprogramming.

46 citations


Journal ArticleDOI
TL;DR: CDK8, independent of its kinase activity, regulates mTOR signalling for the maintenance of BCR-ABL1+ leukemia, and that the dual inhibition of CDK8 and m TOR signalling induces apoptosis in these cells.
Abstract: Cyclin-dependent kinases (CDKs) are frequently deregulated in cancer and represent promising drug targets. We provide evidence that CDK8 has a key role in B-ALL. Loss of CDK8 in leukemia mouse models significantly enhances disease latency and prevents disease maintenance. Loss of CDK8 is associated with pronounced transcriptional changes, whereas inhibiting CDK8 kinase activity has minimal effects. Gene set enrichment analysis suggests that the mTOR signaling pathway is deregulated in CDK8-deficient cells and, accordingly, these cells are highly sensitive to mTOR inhibitors. Analysis of large cohorts of human ALL and AML patients reveals a significant correlation between the level of CDK8 and of mTOR pathway members. We have synthesized a small molecule YKL-06-101 that combines mTOR inhibition and degradation of CDK8, and induces cell death in human leukemic cells. We propose that simultaneous CDK8 degradation and mTOR inhibition might represent a potential therapeutic strategy for the treatment of ALL patients.

32 citations


Journal ArticleDOI
TL;DR: A mutation in Cyclin-Dependent Kinase 8 (CDK8) partially suppresses the dwarfism and constitutive resistance phenotypes of camta1/2/3, suggesting that the whole Mediator kinase module contributes to the transcriptional regulation of SA levels and SAR.
Abstract: In plants, the calmodulin-binding transcription activators (CAMTAs) are required for transcriptional regulation of abiotic and biotic stress responses. Among them, CAMTA3 in Arabidopsis has been intensively studied and shown to function redundantly with CAMTA1 and CAMTA2 to negatively regulate plant immunity. The camta1/2/3 triple mutant accordingly exhibits severe dwarfism due to autoimmunity. Here, through a suppressor screen using camta1/2/3 triple mutant, we found that a mutation in Cyclin-Dependent Kinase 8 (CDK8) partially suppresses the dwarfism and constitutive resistance phenotypes of camta1/2/3. CDK8 positively regulates steady-state salicylic acid (SA) levels and systemic required resistance (SAR). The expression of SA biosynthesis genes such as ICS1 and EDS5 is down-regulated in cdk8 mutants under uninfected conditions, suggesting that CDK8 contributes to the transcriptional regulation of these SA pathway genes. Knocking out another Mediator kinase module member MED12 yielded similar defects including decreased steady-state SA level and compromised SAR, suggesting that the whole Mediator kinase module contributes to the transcriptional regulation of SA levels and SAR.

22 citations


Journal ArticleDOI
15 Mar 2019
TL;DR: Five Cyclin-dependent kinases involved in transcription and processing of HIV-1 RNA are discussed, which may lead to strategies to selectively reactivate latent viruses in patients in whom viral replication is suppressed by antiviral drugs.
Abstract: Studies of RNA Polymerase II (Pol II) transcription of the HIV-1 genome are of clinical interest, as the insight gained may lead to strategies to selectively reactivate latent viruses in patients in whom viral replication is suppressed by antiviral drugs. Such a targeted reactivation may contribute to a functional cure of infection. This review discusses five Cyclin-dependent kinases - CDK7, CDK9, CDK11, CDK2, and CDK8 - involved in transcription and processing of HIV-1 RNA. CDK7 is required for Pol II promoter clearance of reactivated viruses; CDK7 also functions as an activating kinase for CDK9 when resting CD4+ T cells harboring latent HIV-1 are activated. CDK9 is targeted by the viral Tat protein and is essential for productive Pol II elongation of the HIV-1 genome. CDK11 is associated with the TREX/THOC complex and it functions in the 3' end processing and polyadenylation of HIV-1 transcripts. CDK2 phosphorylates Tat and CDK9 and this stimulates Tat activation of Pol II transcription. CDK8 may stimulate Pol II transcription of the HIV-1 genome through co-recruitment with NF-κB to the viral promoter. Some notable open questions are discussed concerning the roles of these CDKs in HIV-1 replication and viral latency.

20 citations


Journal ArticleDOI
04 Jan 2019-Biology
TL;DR: Cyclin C-induced mitochondrial fission was found to increase sensitivity of both mammalian and yeast cells to apoptosis, and its transcriptional and non-transcriptional roles in tumor promotion or suppression are reviewed.
Abstract: The class I cyclin family is a well-studied group of structurally conserved proteins that interact with their associated cyclin-dependent kinases (Cdks) to regulate different stages of cell cycle progression depending on their oscillating expression levels. However, the role of class II cyclins, which primarily act as transcription factors and whose expression remains constant throughout the cell cycle, is less well understood. As a classic example of a transcriptional cyclin, cyclin C forms a regulatory sub-complex with its partner kinase Cdk8 and two accessory subunits Med12 and Med13 called the Cdk8-dependent kinase module (CKM). The CKM reversibly associates with the multi-subunit transcriptional coactivator complex, the Mediator, to modulate RNA polymerase II-dependent transcription. Apart from its transcriptional regulatory function, recent research has revealed a novel signaling role for cyclin C at the mitochondria. Upon oxidative stress, cyclin C leaves the nucleus and directly activates the guanosine 5’-triphosphatase (GTPase) Drp1, or Dnm1 in yeast, to induce mitochondrial fragmentation. Importantly, cyclin C-induced mitochondrial fission was found to increase sensitivity of both mammalian and yeast cells to apoptosis. Here, we review and discuss the biology of cyclin C, focusing mainly on its transcriptional and non-transcriptional roles in tumor promotion or suppression.

20 citations


Journal ArticleDOI
TL;DR: The study reveals genetic interactions between Mediator tail and kinase module subunits and enhances the understanding of dwarfing in phenylpropanoid pathway mutants.
Abstract: The Mediator complex functions as a hub for transcriptional regulation. MED5, an Arabidopsis Mediator tail subunit, is required for maintaining phenylpropanoid homeostasis. A semidominant mutation (ref4-3) that causes a single amino acid substitution in MED5b functions as a strong suppressor of the pathway, leading to decreased soluble phenylpropanoid accumulation, reduced lignin content and dwarfism. By contrast, loss of MED5 results in increased concentrations of phenylpropanoids. We used a reverse genetic approach to identify suppressors of ref4-3 and found that ref4-3 requires CDK8, a kinase module subunit of Mediator, to repress plant growth. The genetic interaction between MED5 and CDK8 was further characterized using mRNA-sequencing (RNA-seq) and metabolite analysis. Growth inhibition and suppression of phenylpropanoid metabolism can be genetically separated in ref4-3 by elimination of CDK8 kinase activity; however, the stunted growth of ref4-3 is not dependent on the phosphorylation event introduced by the G383S mutation. In addition, rather than perturbation of lignin biosynthesis, misregulation of DJC66, a gene encoding a DNAJ protein, is involved in the dwarfism of the med5 mutants. Together, our study reveals genetic interactions between Mediator tail and kinase module subunits and enhances our understanding of dwarfing in phenylpropanoid pathway mutants.

17 citations


Journal ArticleDOI
TL;DR: It is found that 37% Cyclin C-dependent genes were downregulated following stress, consistent with a model that cyclin C relocalization to the cytoplasm, and corresponding inactivation of Cdk8, represents a regulatory mechanism to repress and stimulate transcription of stress-responsive genes.
Abstract: The transcriptional changes that occur in response to oxidative stress help direct the decision to maintain cell viability or enter a cell death pathway. Cyclin C-Cdk8 is a conserved kinase that associates with the RNA polymerase II Mediator complex that stimulates or represses transcription depending on the locus. In response to oxidative stress, cyclin C, but not Cdk8, displays partial translocation into the cytoplasm. These findings open the possibility that cyclin C relocalization is a regulatory mechanism governing oxidative stress-induced transcriptional changes. In the present study, the cyclin C-dependent transcriptome was determined and compared to transcriptional changes occurring in oxidatively stressed Mus musculus embryonic fibroblasts. We observed a similar number (∼2000) of genes up or downregulated in oxidatively stressed cells. Induced genes include cellular repair/survival factors while repressed loci were generally involved in proliferation or differentiation. Depleting cyclin C in unstressed cells produced an approximately equal number of genes (∼2400) that were repressed by, or whose transcription required, cyclin C. Consistent with the possibility that cyclin C nuclear release contributes to transcriptional remodeling in response to oxidative stress, we found that 37% cyclin C-dependent genes were downregulated following stress. Moreover, 20% of cyclin C- repressed genes were induced in response to stress. These findings are consistent with a model that cyclin C relocalization to the cytoplasm, and corresponding inactivation of Cdk8, represents a regulatory mechanism to repress and stimulate transcription of stress-responsive genes.

12 citations


Journal ArticleDOI
TL;DR: It is demonstrated that the assembly state of the Mediator complex undergoes physiologic regulation during normal cycles of fasting and feeding in the mouse liver and is dysregulated in states of obesity and insulin resistance.

Journal ArticleDOI
TL;DR: This work suggested that loss of CDK8 could overcome transcriptional and/or posttranscriptional alterations in the fcp1 mutant through its regulatory effect on Skn7, and implicated FCP1 andCDK8 in the broader response to environmental stressors in yeast.

Journal ArticleDOI
TL;DR: A systematic analysis of the effects of CDK8 knockdown or kinase inhibition on colon cancer cell growth in culture, at different primary tumor sites, as well as in the liver, the primary site of metastasis and the leading cause of colon cancer mortality.
Abstract: While the best-known members of the cyclindependent kinase (CDK) family, such as CDK1, CDK2, CDK4/6, regulate cell cycle progression, many other CDKs play no direct role in cell cycle transitions and instead are involved in transcription or RNA splicing. One of these transcriptional kinases, CDK8 (expressed nearly universally), together with its closely related paralog CDK19 (expressed in a tissue-specific pattern), has a unique function of preferentially regulating transcription of newly activated genes, acting in conjunction with various transcription-initiating factors, such as Wnt/βcatenin [1], serum response network, HIF1A [2], TGFβ/ Smad [3], ERα [4], STAT1 and NFκB [5]. As a result, CDK8/19 inhibition has only minor effects on basal transcription but attenuates signal-induced activation of genes that are not expressed in the absence of the signal. CDK8 has been first linked to cancer when it was identified as an oncogene that is frequently amplified or overexpressed in colon carcinoma [1]. Subsequently, CDK8 has been implicated as a tumor-promoting factor in breast, pancreatic and prostate cancers, melanoma and leukemia [6,7]. CDK8 inhibition was also found to stimulate natural killer cells and to increase innate immunity [8]. Importantly, CDK8 was identified as a mediator of chemotherapyor radiation-induced expression of genes implicated in cancer metastasis and drug resistance [9]. CDK8/19 has become an actively pursued drug target [6] and the first selective CDK8/19 inhibitor, Senexin B, has recently entered clinical trials. These trials were designed on the basis of preclinical studies where this compound suppressed the growth of estrogen-receptor-positive breast cancers when combined with hormone therapy [4]. Despite the initial reports that CDK8 knockdown by shRNA in colon cancer cells with CDK8 amplification or overexpression inhibited cell proliferation [1], several groups found that CDK8/19 kinase inhibitors have no significant effect on colon cancer cell growth [8,9]. Nevertheless, CDK8 is one of the most frequently amplified genes in clinical colon cancers and elevated CDK8 expression is associated with shorter patient survival. To reconcile these paradoxical observations, we have recently carried out a systematic analysis of the effects of CDK8 knockdown or kinase inhibition on colon cancer cell growth in culture, at different primary tumor sites, as well as in the liver, the primary site of metastasis and the leading cause of colon cancer mortality [10]. In cell culture assays, CDK8/19 kinase inhibition by Senexin B had no effect on short-term growth in three different CDK8-overexpressing human colon cancer cell lines, in two of which CDK8 gene was amplified, whereas only one of the three cell lines showed a significant response in the long-term clonogenic assay. Furthermore, in contrast to previous reports comparing the growth of CDK8-overexpressing colon cancer cells with their derivatives with stable CDK8 knockdown [1], CDK8 knockdown by inducible shRNA expression had no effect on cell growth or colony formation. In subsequent studies, which were conducted primarily in a transplantable murine CT26 colon cancer model, CDK8 knockdown or kinase inhibition were found to have no significant effect on primary tumor growth in mice when the tumor cells were implanted subcutaneously, orthotopically (in the cecum) or in the spleen. In contrast, the growth of tumors that arise in the liver following splenic injection of colon cancer cells, was strongly suppressed by CDK8 knockdown in tumor cells or by treating mice with Senexin B. Selective inhibition of tumor growth in the liver (but not at the primary injection site) was also observed in the CDK8overexpressing human HCT116 colon cancer cells. Importantly, CDK8/19 inhibitor treatment, when started after liver metastases have been already established, Editorial

Journal ArticleDOI
TL;DR: Results indicate that MDKs are not required for HIV transcription, as neither CA nor SnxA inhibited viral reactivation in Jurkat cell models of HIV latency.
Abstract: While the roles in HIV transcription of many cyclin-dependent kinases (CDKs) have been well defined, little is known about the impact of mediator kinases (MDKs), CDK8 and CDK19, in this pr...

Posted ContentDOI
01 Oct 2019-bioRxiv
TL;DR: It is found that CDK8 and its paralogue CDK19 are significantly overexpressed in HCC patients, where high levels correlate with poor prognosis and support important and non-redundant roles for mediator kinases in liver carcinogenesis.
Abstract: Hepatocellular carcinoma (HCC) is a common cancer with high mortality. The limited therapeutic options for advanced disease include treatment with Sorafenib, a multi-kinase inhibitor whose targets include the Mediator kinase CDK8. Since CDK8 has reported oncogenic activity in Wnt-dependent colorectal cancer, we investigated whether it is also involved in HCC. We find that CDK8 and its paralogue CDK19 are significantly overexpressed in HCC patients, where high levels correlate with poor prognosis. Liver-specific genetic deletion of CDK8 in mice is well supported and protects against chemical carcinogenesis. Deletion of either CDK8 or CDK19 in hepatic precursors had little effect on gene expression in exponential cell growth but prevented oncogene-induced transformation. This phenotype was reversed by concomitant deletion of TP53. These data support important and non-redundant roles for mediator kinases in liver carcinogenesis, where they genetically interact with the TP53 tumor suppressor.