187 results on '"cdc7"'
Search Results
52. Increased Cdc7 expression is a marker of oral squamous cell carcinoma and overexpression of Cdc7 contributes to the resistance to DNA-damaging agents.
- Author
-
Cheng, An Ning, Jiang, Shih Sheng, Fan, Chi-Chen, Lo, Yu-Kang, Kuo, Chan-Yen, Chen, Chung-Hsing, Liu, Ying-Lan, Lee, Chun-Chung, Chen, Wei-Shone, Huang, Tze-Sing, Wang, Tao-Yeuan, and Lee, Alan Yueh-Luen
- Subjects
- *
CELL division , *CANCER treatment , *SQUAMOUS cell carcinoma , *ORAL cancer , *CANCER-related mortality , *CANCER chemotherapy , *DNA damage , *PROGNOSIS - Abstract
Highlights: [•] This report first mentions that Cdc7-Dbf4 is overexpressed in oral cancer. [•] Cdc7 expression is significantly associated with poor outcome of OSCC patients. [•] Cdc7 expression is an independent prognosis factor for overall survival of OSCC patients. [•] Overexpressed Cdc7 enhances the resistance to chemotherapy using DNA-damaging agents. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
53. Cdc7-dependent and -independent phosphorylation of Claspin in the induction of the DNA replication checkpoint.
- Author
-
Rainey, Michael D., Harhen, Brendan, Guan-Nan Wang, Murphy, Paul V., and Santocanale, Corrado
- Published
- 2013
- Full Text
- View/download PDF
54. CDC7
- Author
-
Schwab, Manfred, editor
- Published
- 2009
- Full Text
- View/download PDF
55. High expression of cell division cycle 7 protein correlates with poor prognosis in patients with diffuse large B-cell lymphoma.
- Author
-
Hou, Yun, Wang, Hua-Qing, and Ba, Yi
- Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common form of lymphoma. According to the clinical risk factors and biological heterogeneity, clinical outcome of DLBCL is extremely various, with 5-year survival rates between 30 and 80 %. Although the International Prognostic Index (IPI) remains the primary clinical tool used to predict outcome for patients with DLBCL, notable variability in outcome is still observed within the same IPI score. The cell division cycle 7 (CDC7) is a serine-threonine kinase, which is required to initiate DNA replication. Study showed that high expression of CDC7 was correlated with poor prognosis in patients with DLBCL. Whether CDC7 is an independent prognostic factor for DLBCL remains debatable. We studied the expression of CDC7 protein in 60 Chinese DLBCL patients with immunohistochemical analysis, 34 patients (56.7 %) categorized as low CDC7 expression and 26 patients (43.3 %) as high CDC7 expression. The median survival time of patients with low CDC7 expression was not achieved and that of high expression was 9 months ( P = 0.027). A multivariate analysis showed that IPI score and Ann Arbor stage were independent prognostic factors in relation to patients' OS ( P < 0.05). Pearson correlation analysis showed that CDC7 expression level was positively correlated with IPI score and Ann Arbor stage ( P < 0.001). The results suggest that CDC7 expression level in combination with IPI score and Ann Arbor stage can be specific prognostic factors for DLBCL patients. CDC7 could also be a potential therapeutic target in DLBCL, especially for ABC-DLBCL. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
56. Effects of CDC7 gene silencing and Rituximab on apoptosis in diffuse large B cell lymphoma cells.
- Author
-
Hou, Yun, Wang, Hua-qing, and Ba, Yi
- Subjects
- *
GENE silencing , *RITUXIMAB , *APOPTOSIS , *B cell lymphoma , *SERINE/THREONINE kinases , *DNA replication , *SMALL interfering RNA - Abstract
Background: The cell division cycle 7 (CDC7) is a serine-threonine kinase, which is required for DNA replication and is high expressed in diffuse large B cell lymphoma (DLBCL). Methods: In this study, we targeted CDC7 in human DLBCL-ABC subtype cells (ly3) and examined the subsequent alterations in cellular apoptosis. The expression of CDC7 was silenced with small interfering RNA (siRNA)-expressing plasmid. CDC7 gene silencing cells were then incubated with or without rituximab for 24 h. Following treatment, Annexin V/propidium iodide staining followed by flow cytometry was used to examine cellular apoptosis. Furthermore, the expression of caspase 3, Bax, and Bcl-2 protein was analyzed by Western blotting. The expression of Bax and Bcl-2 mRNA was analyzed by quantitative real-time PCR. Results: Compared to non-treated or control siRNA-transfected cells, significantly higher levels of apoptosis were detected in siCDC7-transfected cells and rituximab-treated cells ( P < 0.05), which was further enhanced in CDC7-targeted cells with rituximab treatment ( P < 0.05). The pro-apoptotic effects were accompanied with up-regulation of caspase 3 and Bax, meanwhile down-regulation of Bcl-2. Conclusion: Combined treatments using rituximab and CDC7 gene silencing significantly increases apoptosis in ly3 cells and plays synergistic effect. CDC7 is a novel therapeutic target in DLBCL patients. CDC7 inhibitors combined with rituximab will be the new therapy for the ABC-DLBCL patients. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
57. "The octet": eight protein kinases that control mammalian DNA replication.
- Author
-
DePamphilis, Melvin L., de Renty, Christelle M., Ullah, Zakir, and Lee, ChrissieY.
- Subjects
PROTEIN kinases ,DNA replication ,HUMAN genome ,CYTOKINESIS ,CELL cycle - Abstract
Development of a fertilized human egg into an average sized adult requires about 29 trillion cell divisions, thereby producing enough DNA to stretch to the Sun and back 200 times (DePamphilis and Bell, 2011)! Even more amazing is the fact that throughout these mitotic cell cycles, the human genome is duplicated once and only once each time a cell divides. If a cell accidentally begins to re-replicate its nuclear DNA prior to cell division, checkpoint pathways trigger apoptosis. And yet, some cells are developmentally programmed to respond to environmental cues by switching from mitotic cell cycles to endocycles, a process in which multiple S phases occur in the absence of either mitosis or cytokinesis. Endocycles allow production of viable, differentiated, polyploid cells that no longer proliferate. What is surprising is that among the 516 (Manning et al., 2002) to 557 (BioMart web site) protein kinases encoded by the human genome, only eight regulate nuclear DNA replication directly. These are Cdk1, Cdk2, Cdk4, Cdk6, Cdk7, Cdc7, Checkpoint kinase-1 (Chk1), and Checkpoint kinase-2. Even more remarkable is the fact that only four of these enzymes (Cdk1, Cdk7, Cdc7, and Chk1) are essential for mammalian development. Here we describe how these protein kinases determine when DNA replication occurs during mitotic cell cycles, how mammalian cells switch from mitotic cell cycles to endocycles, and how cancer cells can be selectively targeted for destruction by inducing them to begin a second S phase before mitosis is complete. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
58. Aminopyrimidinone Cdc7 Kinase Inhibitors
- Author
-
Woods, Keith W., Lai, Chunqiu, Miyashiro, Julie M., Tong, Yunsong, Florjancic, Alan S., Han, Edward K., Soni, Niru, Shi, Yan, Lasko, Loren, Leverson, Joel D., Johnson, Eric F., Shoemaker, Alexander R., and Penning, Thomas D.
- Subjects
- *
ZALCITABINE (Drug) , *ENZYME inhibitors , *DRUG synergism , *CELL-mediated cytotoxicity , *PHOSPHORYLATION , *DRUG design - Abstract
Abstract: We have investigated the SAR of a series of pyrimidinone-containing Cdc7 kinase inhibitors. A wide range of amine substitutions give potent compounds with activities (K i) less than 1nM. Kinase selectivity is reasonable and cytotoxicity corresponds to inhibition of MCM2 phosphorylation. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
59. Integrating DNA replication with trans-lesion synthesis via Cdc7.
- Author
-
Vaziri, Cyrus and Masai, Hisao
- Published
- 2010
- Full Text
- View/download PDF
60. Hsk1 kinase and Cdc45 regulate replication stress-induced checkpoint responses in fission yeast.
- Author
-
Matsumoto, Seiji, Shimmoto, Michie, Kakusho, Naoko, Yokoyama, Mika, Kanoh, Yutaka, Hayano, Motoshi, Russell, Paul, and Masai, Hisao
- Published
- 2010
- Full Text
- View/download PDF
61. Chk1 promotes replication fork progression by controlling replication initiation.
- Author
-
Petermann, Eva, Woodcock, Mick, and Helleday, Thomas
- Subjects
- *
DNA replication , *FOCAL adhesion kinase , *CYCLINS , *RNA , *PROTEIN kinases - Abstract
DNA replication starts at initiation sites termed replication origins. Metazoan cells contain many more potential origins than are activated (fired) during each S phase. Origin activation is controlled by the ATR checkpoint kinase and its downstream effector kinase Chk1, which suppresses origin firing in response to replication blocks and during normal S phase by inhibiting the cyclin-dependent kinase Cdk2. In addition to increased origin activation, cells deficient in Chk1 activity display reduced rates of replication fork progression. Here we investigate the causal relationship between increased origin firing and reduced replication fork progression. We use the Cdk inhibitor roscovitine or RNAi depletion of Cdc7 to inhibit origin firing in Chk1-inhibited or RNAi-depleted cells. We report that Cdk inhibition and depletion of Cdc7 can alleviate the slow replication fork speeds in Chk1-deficient cells. Our data suggest that increased replication initiation leads to slow replication fork progression and that Chk1 promotes replication fork progression during normal S phase by controlling replication origin activity. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
62. Overexpression of cell division cycle 7 homolog is associated with gene amplification frequency in breast cancer.
- Author
-
Choschzick, Matthias, Lebeau, Annette, Marx, Andreas H., Tharun, Lars, Terracciano, Luigi, Heilenkötter, Uwe, Jaenicke, Fritz, Bokemeyer, Carsten, Simon, Ronald, Sauter, Guido, and Schwarz, Jörg
- Subjects
GENE expression ,CELL division ,CELL cycle ,HOMOLOGY (Biology) ,GENE amplification ,GENETICS of breast cancer ,PROTEIN kinases ,PHENOTYPES - Abstract
Summary: Cell division cycle 7 is a widely expressed protein kinase implicated in cell division, cell cycle checkpoint mechanisms, and cancer progression. To determine the relationship of cell division cycle 7 protein expression with tumor phenotype, molecular features and prognosis, 2197 highly characterized breast carcinomas were analyzed on a tissue microarray. Detectable cell division cycle 7 expression was found in 1088 (57%) of breast cancer specimens and 228 (11.9%) exhibited a moderate or strong expression. High levels of cell division cycle 7 expression were significantly related to medullary histotype (P < .0001); high tumor grade (P < .0001); negative estrogen receptor status (P < .0001); high Ki67 expression level (P < .0001); p53 and p16 overexpression (P < .0001); and amplification of HER2 (P < .0001), c-myc (P < .0001), MDM2 (P = .043), CCND1 (P = .0084), and ESR1 (P = .0012) as well as with the number of amplified genes (P < .0001). There was also a tendency towards worse prognosis in cell division cycle 7 positive as compared to negative breast cancers. The relationship between cell division cycle 7 and number of amplifications was independent from tumor proliferation raising the possibility of a direct influence of cell division cycle 7 expression for amplification development. In conclusion, cell division cycle 7 is a replication associated protein with relationships to gene amplification and genomic instability in breast carcinomas. These data support the potential utility of newly developed small molecule cell division cycle 7 inhibitors as a therapeutic alternative in at least a subset of breast carcinomas. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
63. CDC7
- Author
-
Rédei, George P.
- Published
- 2008
- Full Text
- View/download PDF
64. Growth inhibition of hepatocellular carcinoma cells in vitro and in vivo by the 8-methoxy analog of WMC79.
- Author
-
Kosakowska-Cholody, Teresa, Cholody, Wieslaw M., Hariprakasha, Humcha K., Monks, Anne, Kar, Siddhartha, Wang, Meifang, Michejda, Christopher J., and Carr, Brian I.
- Subjects
- *
LIVER cancer , *CANCER cells , *ANTINEOPLASTIC agents , *CELL growth , *CELLULAR pathology - Abstract
HKH40A (RTA 502), an optimized 8-methoxy analog of the unsymmetrical bifunctional antitumor agent WMC79, was found to be potently active against liver cancer cell growth in vitro and in vivo. Studies on selected human hepatocellular carcinoma (HCC) cell lines with differing p53 status (HepG2, Hep3B, and PLC/PRF/5), revealed that drug-mediated growth inhibition was independent of p53 status. FACS analysis showed an accumulation of cells in S-phase within 24 h of treatment with 100 nM HKH40A. Subsequent incubation of cells, either in the presence of drug or without, caused cell cycle block at the S and G2/M checkpoints, which was consistent with the observed up-regulation of p21, cyclin A, cyclin B1, sustained phosphorylation of Cdk1, and down-regulation of Cdc6, Cdc7, and RRM2. This irreversible growth arrest eventually led to apoptosis. HKH40A completely suppressed growth of the rat transplantable HCC cell line (JM-1) in an orthotopic model in Fisher 344 rats in vivo, without evidence of toxicity. HKH40A may be a useful agent for new therapeutic strategies focusing on inhibition of HCC cell proliferation. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
65. 4-(1H-Indazol-5-yl)-6-phenylpyrimidin-2(1H)-one analogs as potent CDC7 inhibitors
- Author
-
Shafer, Cynthia M., Lindvall, Mika, Bellamacina, Cornelia, Gesner, Thomas G., Yabannavar, Asha, Jia, Weiping, Lin, Song, and Walter, Annette
- Subjects
- *
PHARMACEUTICAL chemistry , *COMBINATORIAL chemistry , *STRUCTURE-activity relationship in pharmacology , *CHEMICAL inhibitors - Abstract
Abstract: A series of 4-(4-hydroxyphenyl)-6-phenylpyrimidin-2(1H)-ones were identified by HTS as inhibitors of CDC7. Molecular modeling and medicinal chemistry techniques were employed to explore the SAR for this series with a focus on removing potential metabolic liabilities and improving cellular potency. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
66. CR/periphilin is a transcriptional co-repressor involved in cell cycle progression
- Author
-
Kurita, Megumi, Suzuki, Hiroaki, Kawano, Yuko, Aiso, Sadakazu, and Matsuoka, Masaaki
- Subjects
- *
CELL cycle regulation , *NUCLEIC acids , *DNA replication , *TRANSCRIPTION factors - Abstract
Abstract: CR/periphilin (CR) retards cell cycle progression mainly at the S-phase in part by transcriptionally repressing expression of Cdc7, the key regulator of DNA replication, and in part by unknown mechanisms. In this study, we show that enforced expression of CR inhibits Cdc7 promoter activity. The attachment of the DNA-binding domain of the yeast GAL4 transcription factor to CR that appears without DNA-binding sequences, enables CR to repress GAL4 promoter-mediated transcription in a histone deacetylase (HDAC) activity-dependent manner. CR forms a complex with mSin3A, a common component in transcriptional repressor complexes, as well as with HDAC1, suggesting that CR may behave as a co-repressor by functional interaction with the Sin3/HDAC co-repressor complex. We also demonstrate that an alternatively spliced variant of CR, CR-S, which is without a region encoded by exon 4 of the CR gene and is a weak interactor with HDAC1, shows a suppressing effect on CR activity. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
67. High levels of Cdc7 and Dbf4 proteins can arrest cell-cycle progression
- Author
-
Guo, Baoqing, Romero, Julia, Kim, Byung-Ju, and Lee, Hoyun
- Subjects
- *
NUCLEIC acids , *DNA replication , *CELLS , *AMINO acids - Abstract
Abstract: Cdc7-Dbf4 serine/threonine kinase is essential for initiation of DNA replication. It was previously found that overexpression of certain replication proteins such as Cdc6 and Cdt1 in fission yeast resulted in multiple rounds of DNA replication in the absence of mitosis. Since this phenomenon is dependent upon the presence of wild-type Cdc7/Hsk1, we hypothesized that high levels of Cdc7 and/or Dbf4 could also cause multiple rounds of DNA replication, or could facilitate entry into S phase. To test this hypothesis, we transiently overexpressed hamster Cdc7, Dbf4 or both in CHO cells. Direct observations of individual cells by fluorescence microscopy and flow cytometric analysis on cell populations suggest that overexpression of Cdc7 and/or Dbf4 does not result in multiple rounds of DNA replication or facilitating entry into S phase. In contrast, moderately increased levels of Dbf4, but not Cdc7, cause cell-cycle arrest in G2/M. This G2/M arrest coincides with hyperphosphorylation of Cdc2/Cdk1 at Tyr-15, raising the possibility that high levels of Dbf4 may activate a G2/M cell-cycle checkpoint. Further increase in Cdc7 and/or Dbf4 by 2–4 fold can arrest cells in G1 and significantly slow down S-phase progression for the cells already in S phase. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
68. Cdc7-Drf1 is a developmentally regulated protein kinase required for the initiation of vertebrate DNA replication.
- Author
-
Takahashi, Tatsuro S. and Walter, Johannes C.
- Subjects
- *
PROTEIN kinases , *DNA , *EUKARYOTIC cells , *PROTISTA , *PHOSPHORYLATION , *CHEMICAL reactions - Abstract
Cdc7, a protein kinase required for the initiation of eukaryotic DNA replication, is activated by a regulatory subunit, Dbf4. A second activator of Cdc7 called Drf1 exists in vertebrates, but its function is unknown. Here, we report that in Xenopus egg extracts, Cdc7-Drf1 is far more abundant than Cdc7-Dbf4, and removal of Drf1 but not Dbf4 severely inhibits phosphorylation of Mcm4 and DNA replication. After gastrulation, when the cell cycle acquires somatic characteristics, Drf1 levels decline sharply and Cdc7-Dbf4 becomes the more abundant kinase. These results identify Drf1 as a developmentally regulated, essential activator of Cdc7 in Xenopus. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
69. Overexpression of CR/periphilin downregulates Cdc7 expression and induces S-phase arrest
- Author
-
Kurita, Megumi, Suzuki, Hiroaki, Masai, Hisao, Mizumoto, Kiyohisa, Ogata, Etsuro, Nishimoto, Ikuo, Aiso, Sadakazu, and Matsuoka, Masaaki
- Subjects
- *
DNA synthesis , *MESSENGER RNA , *NUCLEAR matrix , *GENE expression - Abstract
Cdc7 expression repressor (CR)/periphilin has been originally cloned as an interactor with periplakin, a precursor of the cornified cell envelope, and suggested to constitute a new type of nuclear matrix. We here show that CR/periphilin is a ubiquitously expressed nuclear protein with speckled distribution. Overexpression of CR/periphilin induces S-phase arrest. Analysis of expression of regulators involved in DNA replication has revealed that both mRNA and protein expression of Cdc7, a regulator of the initiation and continuation of DNA replication, are markedly downregulated by overexpression of CR/periphilin. However, co-expression of Cdc7 only marginally rescues S-phase arrest induced by CR, indicating that CR retards S-phase progression by modifying expression of some genes including Cdc7, which are involved in progression of DNA replication or coordination of DNA replication and S-phase progression. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
70. Ibp1p, a novel Cdc25-related phosphatase, suppresses Schizosaccharomyces pombe hsk1 (cdc7).
- Author
-
Snaith, Hilary A., Marlett, John, and Forsburg, Susan L.
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *SCHIZOSACCHAROMYCES pombe , *PROTEIN kinases , *DNA replication , *DNA polymerases , *PROTEOMICS - Abstract
We report the identification of a novel Cdc25-like protein phosphatase, Ibp1, in the fission yeast Schizosaccharomyces pombe. Ibp1 is closely related to the catalytic subunit of the Cdc25 dual-specificity phosphatases and has phosphatase activity in vitro. Over-production of catalytically active Ibp1 robustly suppresses a mutation in the replication initiation kinase Hsk1p, a member of the Cdc7 family of protein kinases and weakly suppresses mutation of Rad4/Cut5, a DNA polymerase ε-associated factor. Ibp1 is not required for viability, suggesting it may be a non-essential regulator of DNA replication or chromosome structure during S phase. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
71. Drf1, a novel regulatory subunit for human Cdc7 kinase.
- Author
-
Montagnoli, A., Bosotti, R., Villa, F., Rialland, M., Brotherton, D., Mercurio, C., Berthelsen, J., and Santocanale, C
- Subjects
- *
CELL cycle regulation , *DNA replication , *HUMAN genome , *HUMAN DNA , *SERINE/THREONINE kinases , *CYCLIN-dependent kinases - Abstract
Studies in model organisms have contributed to elucidate multiple levels at which regulation of eukaryotic DNA replication occurs. Cdc7, an evolutionarily conserved serine–threonine kinase, plays a pivotal role in linking cell cycle regulation to genome duplication, being essential for the firing of DNA replication origins. Binding of the cell cycle‐regulated subunit Dbf4 to Cdc7 is necessary for in vitro kinase activity. This binding is also thought to be the key regulatory event that controls Cdc7 activity in cells. Here, we describe a novel human protein, Drf1, related to both human and yeast Dbf4. Drf1 is a nuclear cell cycle‐regulated protein, it binds to Cdc7 and activates the kinase. Therefore, human Cdc7, like cyclin‐dependent kinases, can be activated by alternative regulatory subunits. Since the Drf1 gene is either absent or not yet identified in the genome of model organisms such as yeast and Drosophila, these findings introduce a new level of complexity in the regulation of DNA replication of the human genome. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
72. The protein kinases AtMAP3Kε1 and BnMAP3Kε1 are functional homologues of S. pombe cdc7p and may be involved in cell division.
- Author
-
Jouannic, Stefan, Champion, Anthony, Segui-Simarro, José-Maria, Salimova, Ekaterina, Picaud, Alain, Tregear, James, Testillano, Pilar, Risueño, Maria-Carmen, Simanis, Viesturs, Kreis, Martin, and Henry, Yves
- Subjects
- *
ARABIDOPSIS thaliana , *CELL cycle , *PROTEIN kinases , *GENETICS - Abstract
Summary We identified an Arabidopsis thaliana gene, AtMAP3Kε1, and a Brassica napus cDNA, BnMAP3Kε1, encoding functional protein serine/threonine kinases closely related to cdc7p and Cdc15p from Schizosaccharomyces pombe and Saccharomyces cerevisiae, respectively. This is the first report of cdc7-related genes in non-fungal eukaryotes; no such genes have as yet been identified in Metazoans. The B. napus protein is able to partially complement a cdc7 loss of function mutation in S. pombe. RT–PCR and in situ hybridisation revealed that the A. thaliana and B. napus genes are expressed in both the sporophytic and the gametophytic tissues of the respective plant species and revealed further that expression is highest in dividing cells. Moreover, AtMAP3Kε1 gene expression is cell cycle-regulated, with higher expression in G2-M phases. Our results strongly suggest that the plant cdc7p-related protein kinases are involved in a signal transduction pathway similar to the SIN pathway, which positively regulates cytokinesis in S. pombe. [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
73. The role of the Sid1p kinase and Cdc14p in regulating the onset of cytokinesis in fission yeast.
- Author
-
Guertin, David A., Chang, Louise, Irshad, Farid, Gould, Kathleen L., and McCollum, Dannel
- Subjects
- *
SCHIZOSACCHAROMYCES pombe , *SACCHAROMYCETACEAE , *YEAST fungi , *PROTEIN kinases , *CYTOKINESIS , *GENES , *EPISTASIS (Genetics) , *MICROBIAL genomics - Abstract
Coordination of mitosis and cytokinesis is crucial for ensuring proper chromosome segregation and genomic stability. In Schizosaccharomyces pombe, the sid genes (cdc7, cdc11, cdcl4, spgl, sid1, sid2 and sid4) define a signaling pathway that regulates septation and cytokinesis. Here we describe the characterization of a novel protein kinase, Sid1p. Sid1p localizes asymmetrically to one spindle pole body (SPB) in anaphase. Sid1p localization is maintained during medial ring constriction and septum synthesis and disappears prior to cell separation. Additionally, we found that Cdcl4p is in a complex with Sid1p. Epistasis analysis places Sid1p-Cdcl4p downstream of Spg1p-Cdc7p but upstream of Sid2p. Finally, we show that cyclin proteolysis during mitosis is unaffected by inactivating the sid pathway; in fact, loss of Cdc2-cyclin activity promotes Sid1p-Cdcl4p association with the SPB, possibly providing a mechanism that couples cytokinesis with mitotic exit. [ABSTRACT FROM AUTHOR]
- Published
- 2000
- Full Text
- View/download PDF
74. Overexpression of CDC7 in malignant salivary gland tumors correlates with tumor differentiation
- Author
-
Jaafari-Ashkavandi, Zohreh, Ashraf, Mohammad Javad, and Abbaspoorfard, Ali Asghar
- Subjects
Salivary gland ,Adenoma pleomórfico ,Pleomorphic adenoma ,Carcinoma adenoide cístico ,Glândula salivar ,Adenoid cystic carcinoma ,Mucoepidermoid carcinoma ,CDC7 ,Carcinoma mucoepidermoide - Abstract
Introduction: Cell division cycle-7 protein is a serine/threonine kinase that has a basic role in cell cycle regulation and is a potential prognostic or therapeutic target in some human cancers. Objectives: This study investigated the expression of cell division cycle-7 protein in benign and malignant salivary gland tumors and also its correlation with clinicopathologic factors. Methods: Immunohistochemical expression of cell division cycle-7 was evaluated in 46 cases, including 15 adenoid cystic carcinoma, 12 mucoepidermoid carcinoma, 14 pleomorphic adenoma, and 5 normal salivary glands. Cell division cycle-7 expression rate and intensity were compared statistically. Results: The protein was expressed in almost all tumors. The intensity and mean of cell division cycle-7 expression were higher in malignant tumors in comparison with pleomorphic adenomas (p = 0.000). The protein expression was correlated with tumor grades (p = 0.000). Conclusions: The present study demonstrated cell division cycle-7 overexpression in malignant salivary gland tumors in comparison with pleomorphic adenomas, and also a correlation with tumor differentiation. Therefore, this protein might be a potential prognostic and therapeutic target for salivary gland tumors. Resumo Introdução: A cell division cycle-7 é uma serina/treonina quinase que tem um papel básico na regulação do ciclo celular e é um potencial marcador prognóstico ou terapêutico em alguns tipos de câncer humano. Objetivos: Este estudo investigou a expressão de cell division cycle-7 em tumores de glândulas salivares benignos e malignos e também sua correlação com fatores clínico-patológicos. Método: A expressão imuno-histoquímica de cell division cycle-7 foi avaliada em 46 casos, incluindo 15 carcinomas adenoide císticos, 12 carcinomas mucoepidermoides, 14 adenomas pleomórficos e 5 glândulas salivares normais. A taxa de expressão e a intensidade da proteína cell division cycle-7 foram comparadas estatisticamente. Resultados: A proteína foi expressa em quase todos os tumores. A intensidade e a média da expressão de cell division cycle-7 foram maiores em tumores malignos em comparação com adenoma pleomórfico (p = 0,000). A expressão da proteína foi correlacionada com os graus do tumor (p = 0,000). Conclusões: O presente estudo demonstrou a superexpressão de cell division cycle-7 em tumores malignos de glândulas salivares quando comparada com o adenoma pleomórfico, além de uma correlação com a diferenciação de tumores. Portanto, essa proteína pode ser um potencial marcador prognóstico e terapêutico para tumores de glândulas salivares.
- Published
- 2019
75. WEE1 inhibition augments CDC7 (DDK) inhibitor-induced cell death in Ewing sarcoma by forcing premature mitotic entry and mitotic catastrophe.
- Author
-
Martin JC, Sims JR, Gupta A, Bakin AV, and Ohm JE
- Subjects
- Humans, Child, Cell Cycle Proteins, Protein-Tyrosine Kinases, Pyrimidinones pharmacology, Cell Line, Tumor, Cell Death, Protein Serine-Threonine Kinases, Sarcoma, Ewing drug therapy, Antineoplastic Agents pharmacology
- Abstract
Ewing sarcoma is an aggressive childhood cancer for which treatment options remain limited and toxic. There is an urgent need for the identification of novel therapeutic strategies. Our group has recently shown that Ewing cells rely on the S-phase kinase CDC7 (DDK) to maintain replication rates and cell viability and that DDK inhibition causes an increase in the phosphorylation of CDK1 and a significant delay in mitotic entry. Here, we expand on our previous findings and show that DDK inhibitor-induced mitotic entry delay is dependent upon WEE1 kinase. Specifically, WEE1 phosphorylates CDK1 and prevents mitotic entry upon DDK inhibition due to the presence of under-replicated DNA, potentially limiting the cytotoxic effects of DDK inhibition. To overcome this, we combined the inhibition of DDK with the inhibition of WEE1 and found that this results in elevated levels of premature mitotic entry, mitotic catastrophe, and apoptosis. Importantly, we have found that DDK and WEE1 inhibitors display a synergistic relationship with regards to reducing cell viability of Ewing sarcoma cells. Interestingly, the cytotoxic nature of this combination can be suppressed by the inhibition of CDK1 or microtubule polymerization, indicating that mitotic progression is required to elicit the cytotoxic effects. This is the first study to display the potential of utilizing the combined inhibition of DDK and WEE1 for the treatment of cancer. We believe this will offer a potential therapeutic strategy for the treatment of Ewing sarcoma as well as other tumor types that display sensitivity to DDK inhibitors., Competing Interests: Conflict of Interest: The authors declare no potential conflicts of interest.
- Published
- 2022
- Full Text
- View/download PDF
76. CDC7
- Author
-
Schwab, Manfred, editor
- Published
- 2001
- Full Text
- View/download PDF
77. Population Pharmacokinetics of TAK-931, a Cell Division Cycle 7 Kinase Inhibitor, in Patients With Advanced Solid Tumors.
- Author
-
Zhou X, Ouerdani A, Diderichsen PM, and Gupta N
- Subjects
- Adult, Aged, Area Under Curve, Biological Transport, Computer Simulation, Creatinine blood, Dose-Response Relationship, Drug, Female, Humans, Liver Failure metabolism, Male, Metabolic Clearance Rate, Middle Aged, Racial Groups, Antineoplastic Agents pharmacokinetics, Cell Cycle Proteins antagonists & inhibitors, Neoplasms drug therapy, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrazolones pharmacokinetics, Pyrimidines pharmacokinetics
- Abstract
A population pharmacokinetic (PK) analysis was conducted to characterize sources of interpatient variability on the PK of TAK-931, a cell division cycle 7 kinase inhibitor, in adult patients with advanced solid tumors using data from 198 patients who received oral TAK-931 over the range of 30 to 150 mg once daily in multiple dosing schedules in 2 phase 1 and 1 phase 2 clinical studies. A 2-compartment model with 2 transit compartments describing the absorption and first-order linear elimination adequately described the PK of TAK-931. The apparent oral clearance (CL/F) of TAK-931 was estimated to be 38 L/h, and the terminal half-life was estimated to be approximately 6 hours. Creatinine clearance (CrCL) was identified as a covariate on CL/F, and body weight as a covariate on CL/F, apparent central volume of distribution, and apparent intercompartmental clearance. Simulations using the final model indicated that the effect of CrCL (≥35 mL/min) or body weight (29.8-127 kg) on TAK-931 systemic exposures was not considered clinically meaningful, suggesting that no dose adjustments were necessary to account for body weight or renal function (CrCL ≥35 mL/min). Sex, age (36-88 years), race, and mild hepatic impairment had no impact on the CL/F of TAK-931. Taken together, the population PK analysis supports the same starting dose of TAK-931 in Asian and Western cancer patients in a global setting., (© 2021 Millennium Pharmaceuticals, Inc. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.)
- Published
- 2022
- Full Text
- View/download PDF
78. p53 controls CDC7 levels to reinforce G1 cell cycle arrest upon genotoxic stress
- Author
-
Slavica Tudzarova, Andrei L. Okorokov, Ayona Dey, Gareth H. Williams, Kai Stoeber, and Paul Mulholland
- Subjects
0301 basic medicine ,DNA re-replication ,CDK2 ,p53 ,Cell cycle checkpoint ,DNA repair ,Genotoxic Stress ,DNA replication ,CDC7 ,S Phase ,03 medical and health sciences ,0302 clinical medicine ,Report ,Fbxw7β ,CHEK1 ,Molecular Biology ,biology ,p21 ,Cyclin-dependent kinase 2 ,GSK3β ,Cell Biology ,Cell cycle ,miRNA-192/215 ,G1 Phase Cell Cycle Checkpoints ,3. Good health ,Ubiquitin ligase ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,protein degradation ,DNA damage ,cell cycle ,Tumor Suppressor Protein p53 ,Developmental Biology - Abstract
DNA replication initiation is a key event in the cell cycle, which is dependent on 2 kinases - CDK2 and CDC7. Here we report a novel mechanism in which p53 induces G1 checkpoint and cell cycle arrest by downregulating CDC7 kinase in response to genotoxic stress. We demonstrate that p53 controls CDC7 stability post-transcriptionally via miR-192/215 and post-translationally via Fbxw7β E3 ubiquitin ligase. The p53-dependent pathway of CDC7 downregulation is interlinked with the p53-p21-CDK2 pathway, as p21-mediated inhibition of CDK2-dependent phosphorylation of CDC7 on Thr376 is required for GSK3ß-phosphorylation and Fbxw7ß-dependent degradation of CDC7. Notably, sustained oncogenic high levels of active CDC7 exert a negative feedback onto p53, leading to unrestrained S-phase progression and accumulation of DNA damage. Thus, p53-dependent control of CDC7 levels is essential for blocking G1/S cell-cycle transition upon genotoxic stress, thereby safeguarding the genome from instability and thus representing a novel general stress response.
- Published
- 2016
79. Cdc7 is a potent anti-cancer target in pancreatic cancer due to abrogation of the DNA origin activation checkpoint
- Author
-
Marco Loddo, Stephen P. Pereira, Gareth H. Williams, Matthew T. Huggett, Kai Stoeber, Margaret G. Keane, Ian Proctor, and Slavica Tudzarova
- Subjects
0301 basic medicine ,DNA Replication ,Pathology ,medicine.medical_specialty ,pancreatic cancer ,Cell Cycle Proteins ,Adenocarcinoma ,Protein Serine-Threonine Kinases ,Transfection ,Cdc7 ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Annexin ,Pancreatic cancer ,Cell Line, Tumor ,Medicine ,Humans ,Pyrroles ,Molecular Targeted Therapy ,Piperidones ,business.industry ,Kinase ,Cell cycle ,medicine.disease ,G1 Phase Cell Cycle Checkpoints ,3. Good health ,Blot ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,cell cycle ,business ,Research Paper - Abstract
// Matthew T. Huggett 1, 2 , Slavica Tudzarova 2 , Ian Proctor 2 , Marco Loddo 2, 3 , Margaret G. Keane 1 , Kai Stoeber 2 , Gareth H. Williams 2, 3 , Stephen P. Pereira 1 1 UCL Institute for Liver and Digestive Health and UCL Cancer Institute, University College London, London, UK 2 The Research Department of Pathology, UCL Cancer Institute, University College London, London, UK 3 Oncologica Ltd, The Science Village, Chesterford Research Park, Cambridge, UK Correspondence to: Gareth Williams, e-mail: gareth.williams@oncologica.com Keywords: pancreatic cancer, cell cycle, DNA replication, Cdc7 Received: July 22, 2015 Accepted: January 23, 2016 Published: February 23, 2016 ABSTRACT Purpose: Cdc7 is a serine/threonine kinase which is responsible for the ‘firing’ of replication origins leading to initiation of DNA replication. Inhibition or depletion of Cdc7 in normal cells triggers a DNA origin activation checkpoint causing a reversible G1 arrest. Here we investigate Cdc7 as a novel therapeutic target in pancreatic cancer. Experimental design: Cdc7 target validation was performed by immunoexpression profiling in a cohort of 73 patients with pancreatic adenocarcinoma including 24 controls. Secondly Cdc7 kinase was targeted in Capan-1 and PANC-1 pancreatic cancer cell line models using either an siRNA against Cdc7 or alternatively a small molecule inhibitor (SMI) of Cdc7 (PHA-767491). Results: Cdc7 was significantly overexpressed in pancreatic adenocarcinoma compared to benign pancreatic tissue (median LI 34.3% vs. 1.3%; P
- Published
- 2016
80. Physical interactions between MCM and Rad51 facilitate replication fork lesion bypass and ssDNA gap filling by non-recombinogenic functions.
- Author
-
Cabello-Lobato, María J., González-Garrido, Cristina, Cano-Linares, María I., Wong, Ronald P., Yáñez-Vílchez, Aurora, Morillo-Huesca, Macarena, Roldán-Romero, Juan M., Vicioso, Marta, González-Prieto, Román, Ulrich, Helle D., and Prado, Félix
- Abstract
The minichromosome maintenance (MCM) helicase physically interacts with the recombination proteins Rad51 and Rad52 from yeast to human cells. We show, in Saccharomyces cerevisiae , that these interactions occur within a nuclease-insoluble scaffold enriched in replication/repair factors. Rad51 accumulates in a MCM- and DNA-binding-independent manner and interacts with MCM helicases located outside of the replication origins and forks. MCM, Rad51, and Rad52 accumulate in this scaffold in G1 and are released during the S phase. In the presence of replication-blocking lesions, Cdc7 prevents their release from the scaffold, thus maintaining the interactions. We identify a rad51 mutant that is impaired in its ability to bind to MCM but not to the scaffold. This mutant is proficient in recombination but partially defective in single-stranded DNA (ssDNA) gap filling and replication fork progression through damaged DNA. Therefore, cells accumulate MCM/Rad51/Rad52 complexes at specific nuclear scaffolds in G1 to assist stressed forks through non-recombinogenic functions. [Display omitted] • Rad51 and Rad52 interact with MCM in a nuclease-insoluble nucleoprotein scaffold • MCM/Rad51/Rad52 accumulation is regulated by cell cycle and replicative DNA damage • Cdc7 prevents Rad51/Rad52 release from the scaffold under replicative DNA damage • MCM/Rad51 promotes MMS-induced gap filling and fork progression by non-HR processes Cabello-Lobato et al. find that MCM interacts dynamically with Rad51 and Rad52 within a nuclease-insoluble nucleoprotein scaffold. These interactions are established in G1 and maintained in the S phase by Cdc7 in the presence of replication-blocking lesions to assist stressed forks through non-recombinogenic functions. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
81. Structural Basis for the Activation and Target Site Specificity of CDC7 Kinase
- Author
-
Siobhan M. Hughes, Stefania Federico, Valerie E. Pye, Nicola O’Reilly, Samual D. Dick, and Peter Cherepanov
- Subjects
DBF4 ,Cell Cycle Proteins ,Substrate Specificity ,INITIATION ,Structural Biology ,Catalytic Domain ,BINDING ,Phosphorylation ,0303 health sciences ,biology ,Kinase ,Chemistry ,030302 biochemistry & molecular biology ,Zinc Fingers ,Cell cycle ,DNA-DAMAGE CHECKPOINT ,Molecular Docking Simulation ,PHOSPHORYLATION SITES ,bisubstrate ,S-PHASE ,Target site ,cell cycle ,03 Chemical Sciences ,Life Sciences & Biomedicine ,BISUBSTRATE INHIBITORS ,Protein Binding ,Biochemistry & Molecular Biology ,crystal structure ,kinase ,kinase inhibitor ,DDK ,Biophysics ,Protein Serine-Threonine Kinases ,Article ,CDC7 ,XL413 ,03 medical and health sciences ,zinc-binding domain ,Humans ,Molecular Biology ,S phase ,030304 developmental biology ,Science & Technology ,Helicase ,Active site ,Cell Biology ,PROTEIN-KINASE ,06 Biological Sciences ,MOLECULAR-MECHANISM ,Activation loop ,REPLICATION ,biology.protein ,CDC7-DBF4 ,08 Information and Computing Sciences - Abstract
Summary CDC7 is an essential Ser/Thr kinase that acts upon the replicative helicase throughout the S phase of the cell cycle and is activated by DBF4. Here, we present crystal structures of a highly active human CDC7-DBF4 construct. The structures reveal a zinc-finger domain at the end of the kinase insert 2 that pins the CDC7 activation loop to motif M of DBF4 and the C lobe of CDC7. These interactions lead to ordering of the substrate-binding platform and full opening of the kinase active site. In a co-crystal structure with a mimic of MCM2 Ser40 phosphorylation target, the invariant CDC7 residues Arg373 and Arg380 engage phospho-Ser41 at substrate P+1 position, explaining the selectivity of the S-phase kinase for Ser/Thr residues followed by a pre-phosphorylated or an acidic residue. Our results clarify the role of DBF4 in activation of CDC7 and elucidate the structural basis for recognition of its preferred substrates., Graphical Abstract, Highlights • DBF4 activates CDC7 kinase via a two-step mechanism • Zinc-finger domain in CDC7 KI2 interacts with DBF4 motif M • Invariant CDC7 residues Arg373 and Arg380 engage P+1 substrate site, CDC7 is a protein kinase that is essential for cell division. Using X-ray crystallography, Cherepanov and colleagues explain the two-step mechanism of CDC7 activation by its dedicated regulator protein DBF4. They also identify amino acid residues in CDC7 that are crucial for the recognition of its preferred substrates.
- Published
- 2020
- Full Text
- View/download PDF
82. Regulated Proteolysis of MutSγ Controls Meiotic Crossing Over
- Author
-
Petr Cejka, Xiangyu Chen, Junxi Wang Bragg, Harrison S. Manasca, Shangming Tang, Yunmei Ma, Dhananjaya S. Kulkarni, Wei He, Gerrik F. Verhees, Nancy M. Hollingsworth, Lepakshi Ranjha, Christa Baker, Nikhil R. Bhagwat, Maria A.W. Chang, Christie Hall, Neil Hunter, and H.B.D. Prasada Rao
- Subjects
Proteasome Endopeptidase Complex ,Saccharomyces cerevisiae Proteins ,1.1 Normal biological development and functioning ,MutS ,crossing over ,homologous recombination ,Cell Cycle Proteins ,Context (language use) ,Protein Serine-Threonine Kinases ,Protein degradation ,Biology ,Medical and Health Sciences ,Article ,Cdc7 ,Chromosomal crossover ,Chromosome segregation ,03 medical and health sciences ,0302 clinical medicine ,Genetic ,Underpinning research ,Genetics ,Holliday junction ,meiosis ,aneuploidy ,chromosome ,Crossing Over, Genetic ,Phosphorylation ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Holliday Junction ,Cell Biology ,Biological Sciences ,degron ,Cell biology ,DNA-Binding Proteins ,Chromosome Pairing ,Meiosis ,proteasome ,Proteolysis ,Generic health relevance ,Degron ,Homologous recombination ,030217 neurology & neurosurgery ,Biotechnology ,Developmental Biology - Abstract
Summary Crossover recombination is essential for accurate chromosome segregation during meiosis. The MutSγ complex, Msh4-Msh5, facilitates crossing over by binding and stabilizing nascent recombination intermediates. We show that these activities are governed by regulated proteolysis. MutSγ is initially inactive for crossing over due to an N-terminal degron on Msh4 that renders it unstable by directly targeting proteasomal degradation. Activation of MutSγ requires the Dbf4-dependent kinase Cdc7 (DDK), which directly phosphorylates and thereby neutralizes the Msh4 degron. Genetic requirements for Msh4 phosphorylation indicate that DDK targets MutSγ only after it has bound to nascent joint molecules (JMs) in the context of synapsing chromosomes. Overexpression studies confirm that the steady-state level of Msh4, not phosphorylation per se, is the critical determinant for crossing over. At the DNA level, Msh4 phosphorylation enables the formation and crossover-biased resolution of double-Holliday Junction intermediates. Our study establishes regulated protein degradation as a fundamental mechanism underlying meiotic crossing over.
- Published
- 2020
- Full Text
- View/download PDF
83. Human and Xenopus cDNAs encoding budding yeast Cdc7-related kinases: in vitro phosphorylation of MCM subunits by a putative human homologue of Cdc7.
- Author
-
Sato, Noriko, Arai, Ken-ichi, and Masai, Hisao
- Subjects
- *
SACCHAROMYCES cerevisiae , *DNA replication , *SCHIZOSACCHAROMYCES pombe , *FISSION (Asexual reproduction) , *YEAST , *CELLS , *AMINO acids , *CELL cycle , *CYTOPLASM , *MITOSIS - Abstract
Saccharomyces cerevisiae Cdc7 kinase is essential for initiation of DNA replication, and Hsk1, a related kinase of Schizosaccharomyces pombe, is also required for DNA replication of fission yeast cells. We report here cDNAs encoding Cdc7-related kinases from human and Xenopus (huCdc7 and xeCdc7, respectively). The cloned cDNA for huCdc7 contains an open reading frame consisting of 574 amino acids with a predicted molecular weight of 63 847 that possesses overall amino acid identity of 32% (54% including similar residues) to Cdc7 and Hsk1. huCDC7 is transcribed in the various tissues examined, but most abundantly in testis. Three transcripts of 4.4, 3.5 and 2.4 kb in length are detected. The 3.5 kb transcript is the most predominant and is expressed in all the tissues examined. A cDNA containing a 91 nucleotide insertion at the N-terminal region of huCDC7 is also detected, suggesting the presence of multiple splicing variants. The huCdc7 protein is expressed at a constant level during the mitotic cell cycle and is localized primarily in nuclei in interphase and distributed diffusibly in cytoplasm in the mitotic phase. The wild-type huCdc7 protein expressed in COS7 cells phosphorylates MCM2 and MCM3 proteins in vitro, suggesting that huCdc7 may regulate processes of DNA replication by modulating MCM functions. [ABSTRACT FROM AUTHOR]
- Published
- 1997
- Full Text
- View/download PDF
84. Regulation of DNA Synthesis: The Identification of New Drosophila melanogaster Cdc7 Regulatory Subunits
- Author
-
Anderson, Benjamin D, Zelada, Eliana Torres, Stephenson, Robert E, and Weake, Vikki M
- Subjects
Drosophila melanogaster ,cell cycle ,regulation ,protein interactions ,Yeast 2-Hybrid ,Cdc7 - Abstract
Cell division cycle 7 (Cdc7) is an enzyme required for the initiation of DNA replication. Cdc7 cannot act alone requiring the binding of its regulatory subunit Dbf4 to perform its enzymatic function. Previous studies show that Dbf4 and Cdc7 are well conserved across eukaryotic organisms. Humans and Xenopus have multiple Cdc7 regulatory subunits, and recent studies suggest that Drosophila melanogaster might as well. Human Dbf4 was discovered because of its similarity to yeast Dbf4. It is possible that finding additional Cdc7 regulatory subunits in D. melanogaster could reveal related proteins in humans. As cancer is a disease caused by improper cell cycling, furthering our understanding of Cdc7 and the cell cycle regulation could lead to advances in cancer treatment. This study seeks to identify possible Cdc7 regulatory subunits by screening for D. melanogaster proteins that directly interact with Cdc7. The first goal was to use a Yeast 2-Hybrid assay to repeat results that indicated an interaction between Cdc7 and Drosophila Dbf4, known as Chiffon. This allowed for testing media and the effectiveness of the assay. While not preformed yet, screening will be completed using a Yeast 2-Hybrid assay to determine interactions between Cdc7 and proteins from a D. melanogaster cDNA library. Further testing will remove false positives. Any remaining plasmids be sequenced and identified by the sequence comparison software, BLAST. Our study will test for D. melanogaster proteins that interact with Cdc7, but once these proteins are found further experimentation will be required to confirm interaction and function with Cdc7.
- Published
- 2017
85. Structural Basis for the Activation and Target Site Specificity of CDC7 Kinase.
- Author
-
Dick, Samual D., Federico, Stefania, Hughes, Siobhan M., Pye, Valerie E., O'Reilly, Nicola, and Cherepanov, Peter
- Subjects
- *
AMINO acid residues , *CELL division , *DNA helicases , *X-ray crystallography , *PROTEIN kinases , *CELL cycle , *DNA replication - Abstract
CDC7 is an essential Ser/Thr kinase that acts upon the replicative helicase throughout the S phase of the cell cycle and is activated by DBF4. Here, we present crystal structures of a highly active human CDC7-DBF4 construct. The structures reveal a zinc-finger domain at the end of the kinase insert 2 that pins the CDC7 activation loop to motif M of DBF4 and the C lobe of CDC7. These interactions lead to ordering of the substrate-binding platform and full opening of the kinase active site. In a co-crystal structure with a mimic of MCM2 Ser40 phosphorylation target, the invariant CDC7 residues Arg373 and Arg380 engage phospho-Ser41 at substrate P+1 position, explaining the selectivity of the S-phase kinase for Ser/Thr residues followed by a pre-phosphorylated or an acidic residue. Our results clarify the role of DBF4 in activation of CDC7 and elucidate the structural basis for recognition of its preferred substrates. • DBF4 activates CDC7 kinase via a two-step mechanism • Zinc-finger domain in CDC7 KI2 interacts with DBF4 motif M • Invariant CDC7 residues Arg373 and Arg380 engage P+1 substrate site CDC7 is a protein kinase that is essential for cell division. Using X-ray crystallography, Cherepanov and colleagues explain the two-step mechanism of CDC7 activation by its dedicated regulator protein DBF4. They also identify amino acid residues in CDC7 that are crucial for the recognition of its preferred substrates. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
86. Human DDK rescues stalled forks and counteracts checkpoint inhibition at unfired origins to complete DNA replication.
- Author
-
Jones MJK, Gelot C, Munk S, Koren A, Kawasoe Y, George KA, Santos RE, Olsen JV, McCarroll SA, Frattini MG, Takahashi TS, and Jallepalli PV
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Checkpoint Kinase 1 genetics, Checkpoint Kinase 1 metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Female, HCT116 Cells, HEK293 Cells, HeLa Cells, Humans, Phosphorylation, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, S Phase Cell Cycle Checkpoints, Substrate Specificity, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Xenopus laevis, DNA Replication drug effects, Replication Origin
- Abstract
Eukaryotic genomes replicate via spatially and temporally regulated origin firing. Cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK) promote origin firing, whereas the S phase checkpoint limits firing to prevent nucleotide and RPA exhaustion. We used chemical genetics to interrogate human DDK with maximum precision, dissect its relationship with the S phase checkpoint, and identify DDK substrates. We show that DDK inhibition (DDKi) leads to graded suppression of origin firing and fork arrest. S phase checkpoint inhibition rescued origin firing in DDKi cells and DDK-depleted Xenopus egg extracts. DDKi also impairs RPA loading, nascent-strand protection, and fork restart. Via quantitative phosphoproteomics, we identify the BRCA1-associated (BRCA1-A) complex subunit MERIT40 and the cohesin accessory subunit PDS5B as DDK effectors in fork protection and restart. Phosphorylation neutralizes autoinhibition mediated by intrinsically disordered regions in both substrates. Our results reveal mechanisms through which DDK controls the duplication of large vertebrate genomes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
87. The C-terminus of S. pombe DDK subunit Dfp1 is required for meiosis-specific transcription and cohesin cleavage
- Author
-
Susan L. Forsburg, Anh Huy Phan Le, and Tara L. Mastro
- Subjects
QH301-705.5 ,Protein subunit ,Science ,Dbf4 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cdc7 ,Chromosome segregation ,03 medical and health sciences ,0302 clinical medicine ,Meiosis ,Transcription (biology) ,Biology (General) ,Gene ,030304 developmental biology ,Zinc finger ,Genetics ,0303 health sciences ,Cohesin ,fungi ,DNA replication ,Rec8 ,Fission yeast ,Hsk1 ,General Agricultural and Biological Sciences ,030217 neurology & neurosurgery ,Research Article - Abstract
Summary The DDK complex is a conserved kinase complex, consisting of a catalytic subunit, Hsk1 (Cdc7), and its regulatory subunit Dfp1 (Dbf4). This kinase is essential for DNA replication. In this work, we show that dfp1-r35, which truncates the Dfp1 C-terminus zinc finger, causes severe meiotic defects, including reduced spore viability, reduced formation of programmed double strand breaks, altered expression of meiotic genes, and disrupted chromosome segregation. There is a high frequency of dyad formation. Mutants are also defective in the phosphorylation and degradation of the meiotic cohesion, Rec8, resulting in a failure to proceed through the MII division. These defects are more pronounced in a haploid meiosis model than in a normal diploid meiosis. Thus, several critical meiotic functions are linked specifically to the C-terminus of Dfp1, which may target specific substrates for phosphorylation by Hsk1.
- Published
- 2013
88. Cdc7-dependent and -independent phosphorylation of Claspin in the induction of the DNA replication checkpoint
- Author
-
Corrado Santocanale, Guan-Nan Wang, Michael D. Rainey, Brendan Harhen, and Paul V. Murphy
- Subjects
Phosphopeptides ,damage checkpoint ,claspin ,repeated phosphopeptide motifs ,mammalian-cells ,Cell Cycle Proteins ,environment and public health ,Mass Spectrometry ,Substrate Specificity ,checkpoint ,atr ,chk1 ,human cdc7 kinase ,Hydroxyurea ,Protein Interaction Maps ,s-phase ,Phosphorylation ,RNA, Small Interfering ,cdc7 ,Chromatography, High Pressure Liquid ,biology ,Kinase ,Cell biology ,Chromatin ,DNA replication checkpoint ,Biochemistry ,RNA Interference ,complex ,DNA Replication ,kinase ,Molecular Sequence Data ,Protein Serine-Threonine Kinases ,Report ,Humans ,Pyrroles ,Amino Acid Sequence ,Kinase activity ,Molecular Biology ,Piperidones ,Adaptor Proteins, Signal Transducing ,HEK 293 cells ,DNA replication ,Helicase ,Minichromosome Maintenance Complex Component 2 ,Cell Biology ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,Checkpoint Kinase 1 ,biology.protein ,stress-response ,protein ,Protein Kinases ,HeLa Cells ,Developmental Biology - Abstract
Claspin is a critical mediator protein in the DNA replication checkpoint, responsible for ATR-dependent activation of the effector kinase Chk1. Cdc7, an essential kinase required for the initiation of DNA replication, can also interact with and phosphorylate Claspin. In this study we use small-molecule inhibitors of Cdc7 kinase to further understand the relationship between Cdc7, Claspin and Chk1 activation. We demonstrate that inhibition of Cdc7 kinase delays HU-induced phosphorylation of Chk1 but does not affect the maintenance of the replication checkpoint once it is established. We find that while chromatin association of Claspin is not affected by Cdc7 inhibition, Claspin phosphorylation is attenuated following HU treatment, which may be responsible for the altered kinetics of HU-induced Chk1 phosphorylation. We demonstrate that Claspin is an in vitro substrate of Cdc7 kinase, and using mass-spectrometry, we identify multiple phosphorylation sites that help to define a Cdc7 phosphorylation motif. Finally, we show that the interaction between Claspin and Cdc7 is not dependent on Cdc7 kinase activity, but Claspin interaction with the DNA helicase subunit Mcm2 is lost upon Cdc7 inhibition. We propose Cdc7-dependent phosphorylation regulates critical protein-protein interactions and modulates Claspin's function in the DNA replication checkpoint.
- Published
- 2013
- Full Text
- View/download PDF
89. Maintenance of the spindle assembly checkpoint by PLK1 and CDC7 kinases and characterisation of cell lines carrying mutations in the genes coding for the CDC7 regulatory subunits
- Author
-
O'Connor, Aisling, Santocanale, Corrado, Cancer Care West Hardiman Scholarship, and Mitosis
- Subjects
Medicine ,Aurora B ,Cell cycle ,DNA replication ,PLK1 ,Spindle assembly checkpoint ,CDC7 - Abstract
The Spindle Assembly Checkpoint (SAC) ensures that accurate chromosome segregation occurs, by preventing progression into anaphase until all kinetochore-microtubule attachments are stable and bi-polar. During a mitotic arrest, induced by microtubule targeting drugs, the weakening of the SAC allows cells to progress through the cell cycle without chromosome segregation occurring. PLK1 plays a major role in mitosis and has recently emerged as a player in establishing and maintaining a robust SAC. However, mechanistically, the role of PLK1 in the SAC is not fully understood. CDC7 kinase is essential for the initiation of DNA replication in eukaryotic organisms. In humans, it requires the binding of a regulatory subunit, either DBF4 or DRF1, for its activity, which persists from S-phase until anaphase. To date, there is limited knowledge about the distinct functions of DBF4 and DRF1. Furthermore, the role for CDC7 kinase in mitosis in humans has not been established. In this thesis, the role of PLK1 in SAC maintenance was assessed. PLK1 was found to be required for maintenance of Aurora B and its activity at the kinetochores. PLK1 and Aurora B cooperate to maintain a robust SAC but PLK1 does not cooperate with MPS1 or Haspin during checkpoint maintenance. It was found that CDC7 is an active kinase during mitosis where it cooperates with PLK1 in SAC maintenance. Using genetically edited cell lines, this study has revealed that a deletion in DBF4 causes an accumulation of cells in S-phase, and defective phosphorylation of the replicative DNA helicase subunit, MCM2. These results indicate that PLK1 is directly involved in maintaining efficient SAC signalling and provide evidence for a novel role of CDC7 kinase in mitosis. Preliminary evidence suggests that DBF4 is the main CDC7 kinase regulatory subunit that is responsible for efficient S-phase progression and for phosphorylating MCM2. 2020-11-11
- Published
- 2016
90. Funktsionaalse Saccharomyces cerevisiae Cdc7/Dbf4 kompleksi puhastamine rekombinantse bakuloviiruse ekspressioonisüsteemist
- Author
-
Puss, Kait Kaarel, Valk, Ervin, Tartu Ülikool. Loodus- ja tehnoloogiateaduskond, and Tartu Ülikool. Molekulaar- ja rakubioloogia instituut
- Subjects
bakalaureusetööd ,Dbf4 ,G1/S üleminek ,kasvaja ,vähiteraapia ,Cdc7 - Abstract
Rakutsükli korrektne kontroll võimaldab rakkudel edukalt jaguneda, ilma DNA kahjustusteta või patoloogiate tekkimiseta. Rakutsükli neli faasi on G1, S, G2 ja M ning kolm kontrollpunkti, mis asuvad G1, G2 ja metafaasis. Cdc7/Dbf4 kompleks on oluline G1/S üleminekul ning aktivatsioon tuleneb G1 faasis kontrollpunkti otsustusest jätkata rakutsükliga. Valgu kompleksi üleekspressioon põhjustab inimestes vähiteket. Töös puhastatakse S.cerevisiae Cdc7/Dbf4 valgukompleks rekombinantse bakuloviiruse abil putukarakkudest.
- Published
- 2016
91. Cdc7 kinase mediates Claspin phosphorylation in DNA replication checkpoint
- Author
-
Kim, J M, Kakusho, N, Yamada, M, Kanoh, Y, Takemoto, N, and Masai, H
- Published
- 2008
- Full Text
- View/download PDF
92. The Xenopus Xmus101 protein is required for the recruitment of Cdc45 to origins of DNA replication
- Author
-
Antonin V. Tutter, W. Matthew Michael, Ruth A. Van Hatten, Johannes C. Walter, Alyssa M. Khederian, and Antonia H. Holway
- Subjects
DNA Replication ,Male ,Saccharomyces cerevisiae Proteins ,Cell Cycle Proteins ,Replication Origin ,Eukaryotic DNA replication ,Xenopus Proteins ,Biology ,Pre-replication complex ,DNA replication factor CDT1 ,Xenopus laevis ,03 medical and health sciences ,0302 clinical medicine ,Minichromosome maintenance ,Report ,Animals ,Drosophila Proteins ,Humans ,Cloning, Molecular ,030304 developmental biology ,Genetics ,0303 health sciences ,DNA replication ,Nuclear Proteins ,Cell Biology ,Spermatozoa ,Chromatin ,MCM10 ,Cdc7 ,ORC ,TopBP1 ,cell cycle ,3. Good health ,Cell biology ,DNA-Binding Proteins ,030220 oncology & carcinogenesis ,biology.protein ,Origin recognition complex ,Carrier Proteins - Abstract
The initiation of eukaryotic DNA replication involves origin recruitment and activation of the MCM2-7 complex, the putative replicative helicase. Mini-chromosome maintenance (MCM)2-7 recruitment to origins in G1 requires origin recognition complex (ORC), Cdt1, and Cdc6, and activation at G1/S requires MCM10 and the protein kinases Cdc7 and S-Cdk, which together recruit Cdc45, a putative MCM2-7 cofactor required for origin unwinding. Here, we show that the Xenopus BRCA1 COOH terminus repeat–containing Xmus101 protein is required for loading of Cdc45 onto the origin. Xmus101 chromatin association is dependent on ORC, and independent of S-Cdk and MCM2-7. These results define a new factor that is required for Cdc45 loading. Additionally, these findings indicate that the initiation complex assembly pathway bifurcates early, after ORC association with the origin, and that two parallel pathways, one controlled by MCM2-7, and the other by Xmus101, cooperate to load Cdc45 onto the origin.
- Published
- 2002
- Full Text
- View/download PDF
93. A High Through-Put Screen for Small Molecules Modulating MCM2 Phosphorylation Identifies Ryuvidine as an Inducer of the DNA Damage Response
- Author
-
Sandra Healy, Laura Murillo, Aisling O'Connor, Guan-Nan Wang, Michael D. Rainey, Alessandro Natoni, Corrado Santocanale, Kevin Z. L. Wu, Michael O'Dwyer, Enda O'Connell, Jennifer FitzGerald, and Gemma O'Brien
- Subjects
Phosphorylation ,DNA Damage ,MCM2 ,CDC7 ,lcsh:Medicine ,DNA helicase complex ,Eukaryotic DNA replication ,Cell Cycle Proteins ,Biochemistry ,Cell Signaling ,cdc7 kinase inhibitors ,Nucleic Acids ,topoisomerase-ii ,Molecular Cell Biology ,Drug Discovery ,Medicine and Health Sciences ,Cell Cycle and Cell Division ,s-phase ,Enzyme Inhibitors ,lcsh:Science ,Cellular Stress Responses ,double-strand breaks ,Multidisciplinary ,Protein Kinase Signaling Cascade ,Kinase ,Signaling Cascades ,3. Good health ,Cell biology ,Cell Processes ,Topoisomerase inhibitor ,Research Article ,Signal Transduction ,DNA Replication ,Drug Research and Development ,replication stress ,DNA damage ,medicine.drug_class ,DNA repair ,Biology ,Protein Serine-Threonine Kinases ,ionizing-radiation ,Small Molecule Libraries ,antitumor-activity ,medicine ,Humans ,Pharmacology ,cell-cycle regulation ,DNA synthesis ,Biology and life sciences ,lcsh:R ,DNA replication ,Reproducibility of Results ,Minichromosome Maintenance Complex Component 2 ,DNA ,Cell Biology ,Molecular biology ,High-Throughput Screening Assays ,Enzyme Activation ,lcsh:Q ,protein ,cancer-therapy ,HeLa Cells - Abstract
DNA replication is an essential process for cell division and as such it is a process that is directly targeted by several anticancer drugs. CDC7 plays an essential role in the activation of replication origins and has recently been proposed as a novel target for drug discovery. The MCM DNA helicase complex (MCM2-7) is a key target of the CDC7 kinase, and MCM phosphorylation status at specific sites is a reliable biomarker of CDC7 cellular activity. In this work we describe a cell-based assay that utilizes the "In Cell Western Technique'' (ICW) to identify compounds that affect cellular CDC7 activity. By screening a library of approved drugs and kinase inhibitors we found several compounds that can affect CDC7-dependent phosphorylation of MCM2 in HeLa cells. Among these, Mitoxantrone, a topoisomerase inhibitor, and Ryuvidine, previously described as a CDK4 inhibitor, cause a reduction in phosphorylated MCM2 levels and a sudden blockade of DNA synthesis that is accompanied by an ATM-dependent checkpoint response. This study sheds light on the previously observed cytotoxity of Ryuvidine, strongly suggesting that it is related to its effect of causing DNA damage.
- Published
- 2014
94. The Role of CAF-1 in Epigenetic Conversions at the Telomeres of Saccharomyces cerevisiae and its Control by Protein Kinases
- Author
-
Jeffery, Daniel Charles Barrie and Yankulov, Krassimir
- Subjects
telomere ,CDC28 ,epigenetics ,chromatin ,PCNA ,CAF-1 ,CDC7 - Abstract
Epigenetics is the study of heritable changes in gene expression that are not caused by changes in the DNA sequence. The expression profile of a given cell is regulated by the complex interactions of histone post-translational modifications, DNA methylation, nuclear organization and ATP-dependent histone exchange, as well as a multitude of interacting factors that come along with them. The inheritance of these epigenetic marks that are associated with silenced or active DNA are regulated by multiple mechanisms to ensure that they are faithfully copied after DNA replication. CAF-1 is a central figure in this process that reassembles nucleosomes at the replication fork. However, a mechanism for epigenetic conversions is necessary for cells to undergo differentiation and alter the epigenetic states of certain genes. This mechanism is currently unknown. In this thesis, I use the telomeres of S. cerevisiae as a model to study the mechanism of epigenetic conversions. I developed an assay for the quantitative assessment of the frequency of epigenetic conversions in telomere position effect (TPE) variegation. Then, using this assay, I demonstrated that the deletion of CAC1, the largest subunit of the CAF-1 complex, causes a major reduction in the frequency of telomeric epigenetic conversions, indicating that CAC1 is a key factor in this process. I also present evidence that the cell cycle regulator CDK (Cdc28p) is a major regulator of CAF-1. I showed that CDK phosphorylates Cac1p at the S94 and S515 residues to recruit it to chromatin. Finally, I demonstrated the role of sub-telomeric elements in the control of TPE, showing that Core X and Y’ elements suppress extreme variations in gene silencing at telomeres. With these data, I propose a model for the control of epigenetic conversions at regions of position effect variegation (PEV). This research represents the first comprehensive analysis of epigenetic conversions in a model organism and links epigenetic maintenance and conversions to cell cycle. NSERC, JSPS, OGSST, University of Guelph
- Published
- 2014
95. Xenopus Cdc7 executes its essential function early in S phase and is counteracted by checkpoint-regulated protein phosphatase 1
- Author
-
Peter J. Gillespie, Wei Theng Poh, Philipp Kaldis, J. Julian Blow, and Gaganmeet Singh Chadha
- Subjects
Immunology ,Cell Cycle Proteins ,CHO Cells ,Biology ,Protein Serine-Threonine Kinases ,Xenopus Proteins ,General Biochemistry, Genetics and Molecular Biology ,Replication factor C ,Cricetulus ,Control of chromosome duplication ,Cyclin-dependent kinase ,Cricetinae ,Protein Phosphatase 1 ,Animals ,Pyrroles ,Enzyme Inhibitors ,Phosphorylation ,lcsh:QH301-705.5 ,S phase ,Piperidones ,cdc7 ,Etoposide ,Ovum ,pha-767491 ,Replication timing ,dna replication ,General Neuroscience ,Research ,DNA replication ,pp1 ,Chromatin ,xenopus ,3. Good health ,Biochemistry ,lcsh:Biology (General) ,S Phase Cell Cycle Checkpoints ,biology.protein ,Origin recognition complex ,Research Article - Abstract
The initiation of DNA replication requires two protein kinases: cyclin-dependent kinase (Cdk) and Cdc7. Although S phase Cdk activity has been intensively studied, relatively little is known about how Cdc7 regulates progression through S phase. We have used a Cdc7 inhibitor, PHA-767491, to dissect the role of Cdc7 inXenopusegg extracts. We show that hyperphosphorylation of mini-chromosome maintenance (MCM) proteins by Cdc7 is required for the initiation, but not for the elongation, of replication forks. Unlike Cdks, we demonstrate that Cdc7 executes its essential functions by phosphorylating MCM proteins at virtually all replication origins early in S phase and is not limiting for progression through theXenopusreplication timing programme. We demonstrate that protein phosphatase 1 (PP1) is recruited to chromatin and rapidly reverses Cdc7-mediated MCM hyperphosphorylation. Checkpoint kinases induced by DNA damage or replication inhibition promote the association of PP1 with chromatin and increase the rate of MCM dephosphorylation, thereby counteracting the previously completed Cdc7 functions and inhibiting replication initiation. This novel mechanism for regulating Cdc7 function provides an explanation for previous contradictory results concerning the control of Cdc7 by checkpoint kinases and has implications for the use of Cdc7 inhibitors as anti-cancer agents.
- Published
- 2014
- Full Text
- View/download PDF
96. Overexpression of CDC7 in malignant salivary gland tumors correlates with tumor differentiation.
- Author
-
Jaafari-Ashkavandi Z, Ashraf MJ, and Abbaspoorfard AA
- Subjects
- Adult, Aged, Biomarkers, Tumor analysis, Case-Control Studies, Cell Differentiation, Cross-Sectional Studies, Female, Humans, Immunohistochemistry, Male, Middle Aged, Prognosis, Reference Values, Retrospective Studies, Adenoma, Pleomorphic pathology, Carcinoma, Adenoid Cystic pathology, Carcinoma, Mucoepidermoid pathology, Cell Cycle Proteins analysis, Protein Serine-Threonine Kinases analysis, Salivary Gland Neoplasms pathology
- Abstract
Introduction: Cell division cycle-7 protein is a serine/threonine kinase that has a basic role in cell cycle regulation and is a potential prognostic or therapeutic target in some human cancers., Objectives: This study investigated the expression of cell division cycle-7 protein in benign and malignant salivary gland tumors and also its correlation with clinicopathologic factors., Methods: Immunohistochemical expression of cell division cycle-7 was evaluated in 46 cases, including 15 adenoid cystic carcinoma, 12 mucoepidermoid carcinoma, 14 pleomorphic adenoma, and 5 normal salivary glands. Cell division cycle-7 expression rate and intensity were compared statistically., Results: The protein was expressed in almost all tumors. The intensity and mean of cell division cycle-7 expression were higher in malignant tumors in comparison with pleomorphic adenomas (p=0.000). The protein expression was correlated with tumor grades (p=0.000)., Conclusions: The present study demonstrated cell division cycle-7 overexpression in malignant salivary gland tumors in comparison with pleomorphic adenomas, and also a correlation with tumor differentiation. Therefore, this protein might be a potential prognostic and therapeutic target for salivary gland tumors., (Copyright © 2017 Associação Brasileira de Otorrinolaringologia e Cirurgia Cérvico-Facial. Published by Elsevier Editora Ltda. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
97. Nuclear PGK1 Alleviates ADP-Dependent Inhibition of CDC7 to Promote DNA Replication.
- Author
-
Li, Xinjian, Qian, Xu, Jiang, Hongfei, Xia, Yan, Zheng, Yanhua, Li, Jing, Huang, Bi-Jun, Fang, Jing, Qian, Chao-Nan, Jiang, Tao, Zeng, Yi-Xin, and Lu, Zhimin
- Subjects
- *
DNA replication , *DNA helicases , *PHOSPHORYLATION , *CELL proliferation , *ALLOSTERIC regulation - Abstract
Summary DNA replication is initiated by assembly of the kinase cell division cycle 7 (CDC7) with its regulatory activation subunit, activator of S-phase kinase (ASK), to activate DNA helicase. However, the mechanism underlying regulation of CDC7-ASK complex is unclear. Here, we show that ADP generated from CDC7-mediated MCM phosphorylation binds to an allosteric region of CDC7, disrupts CDC7-ASK interaction, and inhibits CDC7-ASK activity in a feedback way. EGFR- and ERK-activated casein kinase 2α (CK2α) phosphorylates nuclear phosphoglycerate kinase (PGK) 1 at S256, resulting in interaction of PGK1 with CDC7. CDC7-bound PGK1 converts ADP to ATP, thereby abrogating the inhibitory effect of ADP on CDC7-ASK activity, promoting the recruitment of DNA helicase to replication origins, DNA replication, cell proliferation, and brain tumorigenesis. These findings reveal an instrumental self-regulatory mechanism of CDC7-ASK activity by its kinase reaction product ADP and a nonglycolytic role for PGK1 in abrogating this negative feedback in promoting tumor development. Graphical Abstract Highlights • ADP generated from CDC7-mediated phosphorylation inhibits CDC7-ASK activity • EGFR- and ERK1/2-dependent activation of CK2α phosphorylates PGK1 at S256 • Phosphorylated PGK1 binds to CDC7 and converts ADP to ATP • PGK1 releases ADP's inhibition on CDC7 to promote DNA replication and tumorigenesis Li et al. demonstrate that ADP generated from CDC7-mediated MCM phosphorylation binds to an allosteric region of CDC7, disrupts CDC7-ASK interaction, and inhibits CDC7-ASK activity in a feedback way. CDC7-bound PGK1 under EGFR activation condition converts ADP to ATP, thereby abrogating ADP's inhibition on CDC7-ASK activity and promoting DNA replication. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
98. Characterization of a Dual CDC7/CDK9 Inhibitor in Multiple Myeloma Cellular Models
- Author
-
Michael O'Dwyer, Gemma O'Brien, Alessandro Natoni, Sandra Healy, Corrado Santocanale, Alan Jacobsen, Alessia Montagnoli, Jürgen Moll, Mark R.E. Coyne, and Michael D. Rainey
- Subjects
Melphalan ,Cancer Research ,Stromal cell ,apoptosis ,BCL2-family ,DNA replication ,multiple myeloma ,kinase inhibitor ,cell cycle ,CDK9 ,Pharmacology ,lcsh:RC254-282 ,Article ,CDC7 ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Doxorubicin ,Multiple myeloma ,Bortezomib ,business.industry ,Cancer ,Multiple Myeloma ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Oncology ,Cell culture ,business ,medicine.drug - Abstract
Two key features of myeloma cells are the deregulation of the cell cycle and the dependency on the expression of the BCL2 family of anti-apoptotic proteins. The cell division cycle 7 (CDC7) is an essential S-phase kinase and emerging CDC7 inhibitors are effective in a variety of preclinical cancer models. These compounds also inhibit CDK9 which is relevant for MCL-1 expression. The activity and mechanism of action of the dual CDC7/CDK9 inhibitor PHA-767491 was assessed in a panel of multiple myeloma cell lines, in primary samples from patients, in the presence of stromal cells and in combination with drugs used in current chemotherapeutic regimens. We report that in all conditions myeloma cells undergo cell death upon PHA-767491 treatment and we report an overall additive effect with melphalan, bortezomib and doxorubicin, thus supporting further assessment of targeting CDC7 and CDK9 in multiple myeloma.
- Published
- 2013
99. Mechanisms Governing DDK Regulation of the Initiation of DNA Replication
- Author
-
Bernard P. Duncker and Larasati
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,lcsh:QH426-470 ,Protein subunit ,DDK ,Phosphatase ,Dbf4 ,Review ,DNA replication ,Biology ,Cdc7 ,03 medical and health sciences ,Rif1 ,Minichromosome maintenance ,Genetics ,Genetics (clinical) ,Kinase ,MCM6 ,Sld3 ,MCM ,lcsh:Genetics ,030104 developmental biology ,Biochemistry ,Rad53 ,biology.protein ,Phosphorylation ,cell cycle checkpoint - Abstract
The budding yeast Dbf4-dependent kinase (DDK) complex—comprised of cell division cycle (Cdc7) kinase and its regulatory subunit dumbbell former 4 (Dbf4)—is required to trigger the initiation of DNA replication through the phosphorylation of multiple minichromosome maintenance complex subunits 2-7 (Mcm2-7). DDK is also a target of the radiation sensitive 53 (Rad53) checkpoint kinase in response to replication stress. Numerous investigations have determined mechanistic details, including the regions of Mcm2, Mcm4, and Mcm6 phosphorylated by DDK, and a number of DDK docking sites. Similarly, the way in which the Rad53 forkhead-associated 1 (FHA1) domain binds to DDK—involving both canonical and non-canonical interactions—has been elucidated. Recent work has revealed mutual promotion of DDK and synthetic lethal with dpb11-1 3 (Sld3) roles. While DDK phosphorylation of Mcm2-7 subunits facilitates their interaction with Sld3 at origins, Sld3 in turn stimulates DDK phosphorylation of Mcm2. Details of a mutually antagonistic relationship between DDK and Rap1-interacting factor 1 (Rif1) have also recently come to light. While Rif1 is able to reverse DDK-mediated Mcm2-7 complex phosphorylation by targeting the protein phosphatase glycogen 7 (Glc7) to origins, there is evidence to suggest that DDK can counteract this activity by binding to and phosphorylating Rif1.
- Published
- 2016
- Full Text
- View/download PDF
100. ‘The octet’: eight protein kinases that control mammalian DNA replication
- Author
-
Chrissie Y. Lee, Zakir Ullah, Melvin L. DePamphilis, and Christelle de Renty
- Subjects
DNA re-replication ,endoreplication ,checkpoint kinases ,lcsh:QP1-981 ,Physiology ,Dbf4 ,Eukaryotic DNA replication ,mitotic cell cycle ,Review Article ,Polo-like kinase ,Biology ,Cell cycle ,Cdc7-Dbf4 ,Cyclin-Dependent Kinases ,lcsh:Physiology ,Cell biology ,Cdc7 ,endocycle ,Mitotic cell cycle ,Mitotic exit ,Physiology (medical) ,Cyclins ,Endoreduplication ,Mitosis - Abstract
Development of a fertilized human egg into an average sized adult requires about 29 trillion cell divisions, thereby producing enough DNA to stretch to the Sun and back 200 times (DePamphilis and Bell, 2011)! Even more amazing is the fact that throughout these mitotic cell cycles, the human genome is duplicated once and only once each time a cell divides. If a cell accidentally begins to re-replicate its nuclear DNA prior to cell division, checkpoint pathways trigger apoptosis. And yet, some cells are developmentally programmed to respond to environmental cues by switching from mitotic cell cycles to endocycles, a process in which multiple S phases occur in the absence of either mitosis or cytokinesis. Endocycles allow production of viable, differentiated, polyploid cells that no longer proliferate. What is surprising is that among the 516 (Manning et al., 2002) to 557 (BioMart web site) protein kinases encoded by the human genome, only eight regulate nuclear DNA replication directly. These are Cdk1, Cdk2, Cdk4, Cdk6, Cdk7, Cdc7, Checkpoint kinase-1 (Chk1), and Checkpoint kinase-2. Even more remarkable is the fact that only four of these enzymes (Cdk1, Cdk7, Cdc7, and Chk1) are essential for mammalian development. Here we describe how these protein kinases determine when DNA replication occurs during mitotic cell cycles, how mammalian cells switch from mitotic cell cycles to endocycles, and how cancer cells can be selectively targeted for destruction by inducing them to begin a second S phase before mitosis is complete.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.