1
|
You Z, Masai H. Assembly, Activation, and Helicase Actions of MCM2-7: Transition from Inactive MCM2-7 Double Hexamers to Active Replication Forks. BIOLOGY 2024; 13:629. [PMID: 39194567 DOI: 10.3390/biology13080629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
In this review, we summarize the processes of the assembly of multi-protein replisomes at the origins of replication. Replication licensing, the loading of inactive minichromosome maintenance double hexamers (dhMCM2-7) during the G1 phase, is followed by origin firing triggered by two serine-threonine kinases, Cdc7 (DDK) and CDK, leading to the assembly and activation of Cdc45/MCM2-7/GINS (CMG) helicases at the entry into the S phase and the formation of replisomes for bidirectional DNA synthesis. Biochemical and structural analyses of the recruitment of initiation or firing factors to the dhMCM2-7 for the formation of an active helicase and those of origin melting and DNA unwinding support the steric exclusion unwinding model of the CMG helicase.
Collapse
Affiliation(s)
- Zhiying You
- Genome Dynamics Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Hisao Masai
- Genome Dynamics Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| |
Collapse
|
2
|
Piletska E, Thompson D, Jones R, Cruz AG, Poblocka M, Canfarotta F, Norman R, Macip S, Jones DJL, Piletsky S. Snapshot imprinting as a tool for surface mapping and identification of novel biomarkers of senescent cells. NANOSCALE ADVANCES 2022; 4:5304-5311. [PMID: 36540121 PMCID: PMC9724690 DOI: 10.1039/d2na00424k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/11/2022] [Indexed: 06/17/2023]
Abstract
Cellular senescence has proved to be a strong contributor to ageing and age-related diseases, such as cancer and atherosclerosis. Therefore, the protein content of senescent cells is highly relevant to drug discovery, diagnostics and therapeutic applications. However, current technologies for the analysis of proteins are based on a combination of separation techniques and mass spectrometry, which require handling large sample sizes and a large volume of data and are time-consuming. This limits their application in personalised medicine. An easy, quick and inexpensive procedure is needed for qualitative and quantitative analysis of proteins expressed by a cell or tissue. Here, we describe the use of the "snapshot imprinting" approach for the identification of proteins differentially expressed by senescent cells. Molecularly imprinted polymer nanoparticles (MIPs) were formed in the presence of whole cells. Following trypsinolysis, protein epitopes protected by complex with MIPs were eluted from the nanoparticles and analysed by LC-MS/MS. In this work, "snapshot imprinting" was performed parallel to a standard proteomic "shaving approach", showing similar results. The analysis by "snapshot imprinting" identified three senescent-specific proteins: cell division cycle 7-related protein kinase, partitioning defective three homolog B and putative ATP-dependent RNA helicase DHX57, the abundance of which could potentially make them specific markers of senescence. Identifying biomarkers for the future elimination of senescent cells grants the potential for developing therapeutics for age-related diseases.
Collapse
Affiliation(s)
- Elena Piletska
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Dana Thompson
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Rebecca Jones
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Alvaro Garcia Cruz
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Marta Poblocka
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester Leicester LE1 7RH UK
| | - Francesco Canfarotta
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Rachel Norman
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya 08018 Barcelona Spain
| | - Salvador Macip
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester Leicester LE1 7RH UK
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya 08018 Barcelona Spain
| | - Donald J L Jones
- Department of Cancer Studies, RKCSB, University of Leicester Leicester LE2 7LX UK
| | - Sergey Piletsky
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| |
Collapse
|
3
|
Saleh A, Noguchi Y, Aramayo R, Ivanova ME, Stevens KM, Montoya A, Sunidhi S, Carranza NL, Skwark MJ, Speck C. The structural basis of Cdc7-Dbf4 kinase dependent targeting and phosphorylation of the MCM2-7 double hexamer. Nat Commun 2022; 13:2915. [PMID: 35614055 PMCID: PMC9133112 DOI: 10.1038/s41467-022-30576-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022] Open
Abstract
The controlled assembly of replication forks is critical for genome stability. The Dbf4-dependent Cdc7 kinase (DDK) initiates replisome assembly by phosphorylating the MCM2-7 replicative helicase at the N-terminal tails of Mcm2, Mcm4 and Mcm6. At present, it remains poorly understood how DDK docks onto the helicase and how the kinase targets distal Mcm subunits for phosphorylation. Using cryo-electron microscopy and biochemical analysis we discovered that an interaction between the HBRCT domain of Dbf4 with Mcm2 serves as an anchoring point, which supports binding of DDK across the MCM2-7 double-hexamer interface and phosphorylation of Mcm4 on the opposite hexamer. Moreover, a rotation of DDK along its anchoring point allows phosphorylation of Mcm2 and Mcm6. In summary, our work provides fundamental insights into DDK structure, control and selective activation of the MCM2-7 helicase during DNA replication. Importantly, these insights can be exploited for development of novel DDK inhibitors.
Collapse
Affiliation(s)
- Almutasem Saleh
- DNA Replication Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Yasunori Noguchi
- DNA Replication Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Ricardo Aramayo
- DNA Replication Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Marina E Ivanova
- DNA Replication Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Kathryn M Stevens
- DNA Replication Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
| | - Alex Montoya
- Proteomics and Metabolomics Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - S Sunidhi
- InstaDeep Ltd, 5 Merchant Square, London, W2 1AY, UK
| | | | | | - Christian Speck
- DNA Replication Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK.
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
4
|
Huang D, Chowdhury S, Wang H, Savage SR, Ivey RG, Kennedy JJ, Whiteaker JR, Lin C, Hou X, Oberg AL, Larson MC, Eskandari N, Delisi DA, Gentile S, Huntoon CJ, Voytovich UJ, Shire ZJ, Yu Q, Gygi SP, Hoofnagle AN, Herbert ZT, Lorentzen TD, Calinawan A, Karnitz LM, Weroha SJ, Kaufmann SH, Zhang B, Wang P, Birrer MJ, Paulovich AG. Multiomic analysis identifies CPT1A as a potential therapeutic target in platinum-refractory, high-grade serous ovarian cancer. Cell Rep Med 2021; 2:100471. [PMID: 35028612 PMCID: PMC8714940 DOI: 10.1016/j.xcrm.2021.100471] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/24/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022]
Abstract
Resistance to platinum compounds is a major determinant of patient survival in high-grade serous ovarian cancer (HGSOC). To understand mechanisms of platinum resistance and identify potential therapeutic targets in resistant HGSOC, we generated a data resource composed of dynamic (±carboplatin) protein, post-translational modification, and RNA sequencing (RNA-seq) profiles from intra-patient cell line pairs derived from 3 HGSOC patients before and after acquiring platinum resistance. These profiles reveal extensive responses to carboplatin that differ between sensitive and resistant cells. Higher fatty acid oxidation (FAO) pathway expression is associated with platinum resistance, and both pharmacologic inhibition and CRISPR knockout of carnitine palmitoyltransferase 1A (CPT1A), which represents a rate limiting step of FAO, sensitize HGSOC cells to platinum. The results are further validated in patient-derived xenograft models, indicating that CPT1A is a candidate therapeutic target to overcome platinum resistance. All multiomic data can be queried via an intuitive gene-query user interface (https://sites.google.com/view/ptrc-cell-line).
Collapse
Affiliation(s)
- Dongqing Huang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Shrabanti Chowdhury
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hong Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sara R Savage
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard G Ivey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jacob J Kennedy
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey R Whiteaker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Chenwei Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Melissa C Larson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN 55905, USA
| | - Najmeh Eskandari
- Division of Hematology and Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Davide A Delisi
- Division of Hematology and Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Saverio Gentile
- Division of Hematology and Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | - Uliana J Voytovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Zahra J Shire
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Qing Yu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew N Hoofnagle
- Department of Lab Medicine, University of Washington, Seattle, WA 98195, USA
| | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Travis D Lorentzen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Anna Calinawan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Larry M Karnitz
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - S John Weroha
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J Birrer
- University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Amanda G Paulovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
5
|
He W, Rao HBDP, Tang S, Bhagwat N, Kulkarni DS, Ma Y, Chang MAW, Hall C, Bragg JW, Manasca HS, Baker C, Verhees GF, Ranjha L, Chen X, Hollingsworth NM, Cejka P, Hunter N. Regulated Proteolysis of MutSγ Controls Meiotic Crossing Over. Mol Cell 2020; 78:168-183.e5. [PMID: 32130890 DOI: 10.1016/j.molcel.2020.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/03/2020] [Accepted: 01/31/2020] [Indexed: 01/04/2023]
Abstract
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.
Collapse
Affiliation(s)
- Wei He
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - H B D Prasada Rao
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Shangming Tang
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Nikhil Bhagwat
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Dhananjaya S Kulkarni
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Yunmei Ma
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Maria A W Chang
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Christie Hall
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Junxi Wang Bragg
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Harrison S Manasca
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Christa Baker
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Gerrik F Verhees
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA
| | - Lepakshi Ranjha
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Xiangyu Chen
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Nancy M Hollingsworth
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Petr Cejka
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Neil Hunter
- Howard Hughes Medical Institute, University of California, Davis, Davis, California, USA; Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, California, USA; Department of Molecular & Cellular Biology, University of California, Davis, Davis, California, USA; Department of Cell Biology & Human Anatomy, University of California, Davis, Davis, California, USA.
| |
Collapse
|
6
|
Abstract
The conserved serine-threonine kinase, Cdc7, plays a crucial role in initiation of DNA replication by facilitating the assembly of an initiation complex. Cdc7 is expressed at a high level and exhibits significant kinase activity not only during S-phase but also during G2/M-phases. A conserved mitotic kinase, Aurora B, is activated during M-phase by association with INCENP, forming the chromosome passenger complex with Borealin and Survivin. We show that Cdc7 phosphorylates and stimulates Aurora B kinase activity in vitro. We identified threonine-236 as a critical phosphorylation site on Aurora B that could be a target of Cdc7 or could be an autophosphorylation site stimulated by Cdc7-mediated phosphorylation elsewhere. We found that threonines at both 232 (that has been identified as an autophosphorylation site) and 236 are essential for the kinase activity of Aurora B. Cdc7 down regulation or inhibition reduced Aurora B activity in vivo and led to retarded M-phase progression. SAC imposed by paclitaxel was dramatically reversed by Cdc7 inhibition, similar to the effect of Aurora B inhibition under the similar situation. Our data show that Cdc7 contributes to M-phase progression and to spindle assembly checkpoint most likely through Aurora B activation.
Collapse
|
7
|
Rowlands H, Shaban K, Cheng A, Foster B, Yankulov K. Dysfunctional CAF-I reveals its role in cell cycle progression and differential regulation of gene silencing. Cell Cycle 2019; 18:3223-3236. [PMID: 31564230 DOI: 10.1080/15384101.2019.1673100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chromatin Assembly Factor I (CAF-I) plays a central role in the reassembly of H3/H4 histones during DNA replication. In S. cerevisiae CAF-I is not essential and its loss is associated with reduced gene silencing at telomeres and increased sensitivity to DNA damage. Two kinases, Cyclin Dependent Kinase (CDK) and Dbf4-Dependent Kinase (DDK), are known to phosphorylate the Cac1p subunit of CAF-I, but their role in the regulation of CAF-I activity is not well understood. In this study we systematically mutated the phosphorylation target sites of these kinases. We show that concomitant mutations of the CDK and DDK target sites of Cac1p lead to growth retardation and significant cell cycle defects, altered cell morphology and increased sensitivity to DNA damage. Surprisingly, some mutations also produced flocculation, a phenotype that is lost in most laboratory strains, and displayed elevated expression of FLO genes. None of these effects is observed upon the destruction of CAF-I. In contrast, the mutations that caused flocculation did not affect gene silencing at the mating type and subtelomeric loci. We conclude that dysfunctional CAF-I produces severe phenotypes, which reveal a possible role of CAF-I in the coordination of DNA replication, chromatin reassembly and cell cycle progression. Our study highlights the role of phosphorylation of Cac1p by CDK and a putative role for DDK in the transmission and re-assembly of chromatin during DNA replication.
Collapse
Affiliation(s)
- Hollie Rowlands
- Department of Molecular and Cellular Biology, University of Guelph , Guelph , Canada
| | - Kholoud Shaban
- Department of Molecular and Cellular Biology, University of Guelph , Guelph , Canada
| | - Ashley Cheng
- Department of Molecular and Cellular Biology, University of Guelph , Guelph , Canada
| | - Barret Foster
- Department of Molecular and Cellular Biology, University of Guelph , Guelph , Canada
| | - Krassimir Yankulov
- Department of Molecular and Cellular Biology, University of Guelph , Guelph , Canada
| |
Collapse
|
8
|
Furanone derivatives as new inhibitors of CDC7 kinase: development of structure activity relationship model using 3D QSAR, molecular docking, and in silico ADMET. Struct Chem 2018. [DOI: 10.1007/s11224-018-1086-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
O Cdc7 kinase where art thou? Curr Genet 2017; 64:677-680. [PMID: 29134273 DOI: 10.1007/s00294-017-0782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 10/18/2022]
Abstract
Although Cdc7 protein kinase is important for regulating DNA replication in all eukaryotes and is a target for cancer therapy, it has never been localized in cells. Recently, a novel molecular genomic method used by our laboratory to localize Cdc7 to regions of chromosomes. Originally, mutations in the CDC7 gene were found in the classic cdc mutant collection of Hartwell et al. (Genetics 74:267-286, 1973). The CDC7 gene was found to encode a protein kinase called DDK that has been studied for many years, establishing its precise role in the initiation of DNA replication at origins. Recently, clinical studies are underway with DDK inhibitors against DDK in cancer patients. However, the conundrum is that Cdc7 has never been detected at origins of replication even though many studies have suggested it should be there. We used "Calling Card" system in which DNA binding proteins are localized to the genome via retrotransposon insertion and deep-sequencing methods. We have shown that Cdc7 localizes at many regions of the genome and was enriched at functional origins of replication. These results are consistent with DDK's role in many additional genomic processes including mutagenesis, chromatid cohesion, and meiotic recombination. Thus, the main conclusion from our studies is that Cdc7 kinase is found at many locations in the genome, but is enriched at functional origins of replication. Furthermore, we propose that application of the Calling Card system to other eukaryotes should be useful in identification of functional origins in other eukaryotic cells.
Collapse
|
10
|
Forsburg SL, Shen KF. Centromere Stability: The Replication Connection. Genes (Basel) 2017; 8:genes8010037. [PMID: 28106789 PMCID: PMC5295031 DOI: 10.3390/genes8010037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 11/16/2022] Open
Abstract
The fission yeast centromere, which is similar to metazoan centromeres, contains highly repetitive pericentromere sequences that are assembled into heterochromatin. This is required for the recruitment of cohesin and proper chromosome segregation. Surprisingly, the pericentromere replicates early in the S phase. Loss of heterochromatin causes this domain to become very sensitive to replication fork defects, leading to gross chromosome rearrangements. This review examines the interplay between components of DNA replication, heterochromatin assembly, and cohesin dynamics that ensures maintenance of genome stability and proper chromosome segregation.
Collapse
Affiliation(s)
- Susan L Forsburg
- Program in Molecular & Computational Biology, University of Southern California, Los Angeles, CA 90089-2910, USA.
| | - Kuo-Fang Shen
- Program in Molecular & Computational Biology, University of Southern California, Los Angeles, CA 90089-2910, USA.
| |
Collapse
|
11
|
Lai WC, Chang TW, Wu CH, Yang SY, Lee TL, Li WC, Chien T, Cheng YC, Shieh JC. Candida albicans Dbf4-dependent Cdc7 kinase plays a novel role in the inhibition of hyphal development. Sci Rep 2016; 6:33716. [PMID: 27644158 PMCID: PMC5028767 DOI: 10.1038/srep33716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/01/2016] [Indexed: 11/26/2022] Open
Abstract
Candida albicans is an opportunistic human fungal pathogen. The ability to switch among multiple cellular forms is key to its pathogenesis. The Dbf4-dependent protein kinase gene CDC7 is conserved due to its role in initiating DNA replication. Because a C. albicans Cdc7 (Cacdc7) homozygous null was not viable, we generated a C. albicans strain with a deleted C. albicans CDC7 (CaCDC7) allele and an expression-repressible allele. Surprisingly, cells of the strain grew as hyphae under the repressed conditions. The in vitro kinase assays confirmed that CaCdc7 (K232) and CaCdc7 (T437) are critical for catalytic and phosphoacceptor of activation activity, respectively. C. albicans cells formed hyphae when expressing either the catalytically inactive CaCdc7 (K232R) or the phosphoacceptor-deficient CaCdc7 (T437A). While CaCdc7 interacted with CaDbf4, cells of the strain in which CaCDC7 was repressed were not rescued by constitutively expressing C. albicans DBF4 or vice versa. We conclude that CaDBF4-dependent CaCDC7 is an essential gene suppressing the hyphal development.
Collapse
Affiliation(s)
- Wei-Chung Lai
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Tschen-Wei Chang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Chang Hao Wu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Shu-Ya Yang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Tai-Lin Lee
- Department of Molecular Biotechnology, Da-Yah University, Changhua County, Taiwan, Republic of China
| | - Wan Chen Li
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Ting Chien
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Yu-Che Cheng
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China
| | - Jia-Ching Shieh
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung City, Taiwan, Republic of China
| |
Collapse
|
12
|
Chen X, Suhandynata RT, Sandhu R, Rockmill B, Mohibullah N, Niu H, Liang J, Lo HC, Miller DE, Zhou H, Börner GV, Hollingsworth NM. Phosphorylation of the Synaptonemal Complex Protein Zip1 Regulates the Crossover/Noncrossover Decision during Yeast Meiosis. PLoS Biol 2015; 13:e1002329. [PMID: 26682552 PMCID: PMC4684282 DOI: 10.1371/journal.pbio.1002329] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/16/2015] [Indexed: 12/02/2022] Open
Abstract
Interhomolog crossovers promote proper chromosome segregation during meiosis and are formed by the regulated repair of programmed double-strand breaks. This regulation requires components of the synaptonemal complex (SC), a proteinaceous structure formed between homologous chromosomes. In yeast, SC formation requires the "ZMM" genes, which encode a functionally diverse set of proteins, including the transverse filament protein, Zip1. In wild-type meiosis, Zmm proteins promote the biased resolution of recombination intermediates into crossovers that are distributed throughout the genome by interference. In contrast, noncrossovers are formed primarily through synthesis-dependent strand annealing mediated by the Sgs1 helicase. This work identifies a conserved region on the C terminus of Zip1 (called Zip1 4S), whose phosphorylation is required for the ZMM pathway of crossover formation. Zip1 4S phosphorylation is promoted both by double-strand breaks (DSBs) and the meiosis-specific kinase, MEK1/MRE4, demonstrating a role for MEK1 in the regulation of interhomolog crossover formation, as well as interhomolog bias. Failure to phosphorylate Zip1 4S results in meiotic prophase arrest, specifically in the absence of SGS1. This gain of function meiotic arrest phenotype is suppressed by spo11Δ, suggesting that it is due to unrepaired breaks triggering the meiotic recombination checkpoint. Epistasis experiments combining deletions of individual ZMM genes with sgs1-md zip1-4A indicate that Zip1 4S phosphorylation functions prior to the other ZMMs. These results suggest that phosphorylation of Zip1 at DSBs commits those breaks to repair via the ZMM pathway and provides a mechanism by which the crossover/noncrossover decision can be dynamically regulated during yeast meiosis.
Collapse
Affiliation(s)
- Xiangyu Chen
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Ray T. Suhandynata
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Rima Sandhu
- Center for Gene Regulation in Health and Disease and Department of Biological Sciences, Cleveland State University, Cleveland, Ohio, United States of America
| | - Beth Rockmill
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Neeman Mohibullah
- Molecular Biology Program, Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York City, New York, United States of America
- Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York City, New York, United States of America
| | - Hengyao Niu
- Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana, United States of America
| | - Jason Liang
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, California, United States of America
- Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, United States of America
| | - Hsiao-Chi Lo
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Danny E. Miller
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Huilin Zhou
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, California, United States of America
- Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - G. Valentin Börner
- Center for Gene Regulation in Health and Disease and Department of Biological Sciences, Cleveland State University, Cleveland, Ohio, United States of America
| | - Nancy M. Hollingsworth
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
13
|
Jeffery DCB, Kakusho N, You Z, Gharib M, Wyse B, Drury E, Weinreich M, Thibault P, Verreault A, Masai H, Yankulov K. CDC28 phosphorylates Cac1p and regulates the association of chromatin assembly factor I with chromatin. Cell Cycle 2015; 14:74-85. [PMID: 25602519 DOI: 10.4161/15384101.2014.973745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Chromatin Assembly Factor I (CAF-I) plays a key role in the replication-coupled assembly of nucleosomes. It is expected that its function is linked to the regulation of the cell cycle, but little detail is available. Current models suggest that CAF-I is recruited to replication forks and to chromatin via an interaction between its Cac1p subunit and the replication sliding clamp, PCNA, and that this interaction is stimulated by the kinase CDC7. Here we show that another kinase, CDC28, phosphorylates Cac1p on serines 94 and 515 in early S phase and regulates its association with chromatin, but not its association with PCNA. Mutations in the Cac1p-phosphorylation sites of CDC28 but not of CDC7 substantially reduce the in vivo phosphorylation of Cac1p. However, mutations in the putative CDC7 target sites on Cac1p reduce its stability. The association of CAF-I with chromatin is impaired in a cdc28-1 mutant and to a lesser extent in a cdc7-1 mutant. In addition, mutations in the Cac1p-phosphorylation sites by both CDC28 and CDC7 reduce gene silencing at the telomeres. We propose that this phosphorylation represents a regulatory step in the recruitment of CAF-I to chromatin in early S phase that is distinct from the association of CAF-I with PCNA. Hence, we implicate CDC28 in the regulation of chromatin reassembly during DNA replication. These findings provide novel mechanistic insights on the links between cell-cycle regulation, DNA replication and chromatin reassembly.
Collapse
Affiliation(s)
- Daniel C B Jeffery
- a Department of Molecular and Cellular Biology ; University of Guelph ; Guelph , Ontario , Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Balasubramanian PK, Balupuri A, Cho SJ. Molecular Modeling Studies of Trisubstituted Thiazoles as Cdc7 Kinase Inhibitors through 3D-QSAR and Molecular Docking Simulation. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Pavithra K. Balasubramanian
- Department of Bio-New Drug Development, College of Medicine; Chosun University; Gwangju 501-759 Republic of Korea
| | - Anand Balupuri
- Department of Bio-New Drug Development, College of Medicine; Chosun University; Gwangju 501-759 Republic of Korea
| | - Seung Joo Cho
- Department of Bio-New Drug Development, College of Medicine; Chosun University; Gwangju 501-759 Republic of Korea
- Department of Cellular Molecular Medicine, College of Medicine; Chosun University; Gwangju 501-759 Republic of Korea
| |
Collapse
|
15
|
Abstract
Initiation of meiotic recombination by DNA double-strand break formation is temporally coordinated with replication. Murakami and Keeney show that this coordination requires recruitment of the Dbf4-dependent kinase to the replication fork by the conserved TIM-TIPIN complex. The same mechanism may regulate other important replication-associated processes.
Collapse
|
16
|
Yamada M, Masai H, Bartek J. Regulation and roles of Cdc7 kinase under replication stress. Cell Cycle 2014; 13:1859-66. [PMID: 24841992 DOI: 10.4161/cc.29251] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cdc7 (cell division cycle 7) kinase together with its activation subunit ASK (also known as Dbf4) play pivotal roles in DNA replication and contribute also to other aspects of DNA metabolism such as DNA repair and recombination. While the biological significance of Cdc7 is widely appreciated, the molecular mechanisms through which Cdc7 kinase regulates these various DNA transactions remain largely obscure, including the role of Cdc7-ASK/Dbf4 under replication stress, a condition associated with diverse (patho)physiological scenarios. In this review, we first highlight the recent findings on a novel pathway that regulates the stability of the human Cdc7-ASK/Dbf4 complex under replication stress, its interplay with ATR-Chk1 signaling, and significance in the RAD18-dependent DNA damage bypass pathway. We also consider Cdc7 function in a broader context, considering both physiological conditions and pathologies associated with enhanced replication stress, particularly oncogenic transformation and tumorigenesis. Furthermore, we integrate the emerging evidence and propose a concept of Cdc7-ASK/Dbf4 contributing to genome integrity maintenance, through interplay with RAD18 that can serve as a molecular switch to dictate DNA repair pathway choice. Finally, we discuss the possibility of targeting Cdc7, particularly in the context of the Cdc7/RAD18-dependent translesion synthesis, as a potential innovative strategy for treatment of cancer.
Collapse
Affiliation(s)
- Masayuki Yamada
- Institute of Molecular and Translational Medicine; Faculty of Medicine and Dentistry; Palacky University; Olomouc, Czech Republic
| | - Hisao Masai
- Genome Dynamics Project; Department of Genome Medicine; Tokyo Metropolitan Institute of Medical Science; Tokyo, Japan
| | - Jiri Bartek
- Institute of Molecular and Translational Medicine; Faculty of Medicine and Dentistry; Palacky University; Olomouc, Czech Republic; Danish Cancer Society Research Center; Copenhagen, Denmark
| |
Collapse
|