51
|
Blickwedehl J, Olejniczak S, Cummings R, Sarvaiya N, Mantilla A, Chanan-Khan A, Pandita TK, Schmidt M, Thompson CB, Bangia N. The proteasome activator PA200 regulates tumor cell responsiveness to glutamine and resistance to ionizing radiation. Mol Cancer Res 2012; 10:937-44. [PMID: 22550082 DOI: 10.1158/1541-7786.mcr-11-0493-t] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The cellular response to ionizing radiation (IR) involves a variety of mechanisms to repair damage and maintain cell survival. We previously reported that the proteasome activator PA200 promotes long-term cell survival after IR exposure. The molecular function of PA200 is to enhance proteasome-mediated cleavage after glutamate; however, it is not known how this molecular function promotes survival after IR exposure. Here, we report that upon IR exposure, cellular demand for exogenous glutamine is increased. Cells containing PA200 are capable of surviving this IR-induced glutamine demand, whereas PA200-deficient cells show impaired long-term survival. Additional glutamine supplementation reverses the radiosensitivity of PA200-knockdown cells suggesting impaired glutamine homeostasis in these cells. Indeed, PA200-knockdown cells are unable to maintain intracellular glutamine levels. Furthermore, when extracellular glutamine is limiting, cells that contain PA200 respond by slowing growth, but PA200-knockdown cells and cells in which post-glutamyl proteasome activity is inhibited are nonresponsive and continue rapid growth. This cellular unresponsiveness to nutrient depletion is also reflected at the level of the mTOR substrate ribosomal S6 kinase (S6K). Thus, inability to restrict growth causes PA200-deficient cells to continue growing and eventually die due to lack of available glutamine. Together, these data indicate an important role for PA200 and post-glutamyl proteasome activity in maintaining glutamine homeostasis, which appears to be especially important for long-term survival of tumor cells after radiation exposure.
Collapse
|
52
|
Finn K, Lowndes NF, Grenon M. Eukaryotic DNA damage checkpoint activation in response to double-strand breaks. Cell Mol Life Sci 2012; 69:1447-73. [PMID: 22083606 PMCID: PMC11115150 DOI: 10.1007/s00018-011-0875-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 10/19/2011] [Accepted: 10/20/2011] [Indexed: 02/07/2023]
Abstract
Double-strand breaks (DSBs) are the most detrimental form of DNA damage. Failure to repair these cytotoxic lesions can result in genome rearrangements conducive to the development of many diseases, including cancer. The DNA damage response (DDR) ensures the rapid detection and repair of DSBs in order to maintain genome integrity. Central to the DDR are the DNA damage checkpoints. When activated by DNA damage, these sophisticated surveillance mechanisms induce transient cell cycle arrests, allowing sufficient time for DNA repair. Since the term "checkpoint" was coined over 20 years ago, our understanding of the molecular mechanisms governing the DNA damage checkpoint has advanced significantly. These pathways are highly conserved from yeast to humans. Thus, significant findings in yeast may be extrapolated to vertebrates, greatly facilitating the molecular dissection of these complex regulatory networks. This review focuses on the cellular response to DSBs in Saccharomyces cerevisiae, providing a comprehensive overview of how these signalling pathways function to orchestrate the cellular response to DNA damage and preserve genome stability in eukaryotic cells.
Collapse
Affiliation(s)
- Karen Finn
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | | | | |
Collapse
|
53
|
Xu J, Li Y, Wang F, Wang X, Cheng B, Ye F, Xie X, Zhou C, Lu W. Suppressed miR-424 expression via upregulation of target gene Chk1 contributes to the progression of cervical cancer. Oncogene 2012; 32:976-87. [PMID: 22469983 DOI: 10.1038/onc.2012.121] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
MicroRNAs (miRNAs) act as important gene regulators in human genomes and their aberrant expression links to many malignancies. We previously identified a different characteristic miRNA expression profile in cervical cancer from that in cervical normal tissues, including the downregulated miR-424. However, the role and mechanism of miR-424 in cervical cancer still remain unknown. Here, we focused on identifying the tumor-suppressive function and clinical significance of miR-424 and exploring the mechanistic relevance by characterizing its target. We showed a significantly decreased expression of miR-424 in 147 cervical cancer tissues versus 74 cervical normal tissues by performing quantitative RT-PCR. In 147 cervical cancer tissue samples, low-level expression of miR-424 was positively correlated with poor tumor differentiation, advanced clinical stage, lymph node metastasis and other poor prognostic clinicopathological parameters. Further in vitro observations showed that enforced expression of miR-424 inhibited cell growth by both enhancing apoptosis and blocking G1/S transition, and suppressed cell migration and invasion in two human cervical cancer cell lines, SiHa and CaSki, implying that miR-424 functions as a tumor suppressor in the progression of cervical cancer. Interestingly, overexpression of miR-424 inhibited the expression of protein checkpoint kinase 1 (Chk1) and phosphorylated Chk1 (p-Chk1) at residues Ser345 and decreased the activity of luciferase-reporter containing the 3'-untranslated region (UTR) of Chk1 with predicted miR-424-binding site. Moreover, miR-424 expression levels were inversely correlated with Chk1 and p-Chk1 protein levels in both cervical cancer and normal tissues. Furthermore, RNAi-mediated knockdown of Chk1 decreased matrix metalloproteinase 9 expression and phenocopied the tumor suppressive effects of miR-424 in cell models. Taken together, our results identify a crucial tumor suppressive role of miR-424 in the progression of cervical cancer at least partly via upreglating the expression of Chk1 and p-Chk1, and suggest that miR-424 might be a candidate of prognostic predictor or an anticancer therapeutic target for cervical cancer patients.
Collapse
Affiliation(s)
- J Xu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Akt: a double-edged sword in cell proliferation and genome stability. JOURNAL OF ONCOLOGY 2012; 2012:951724. [PMID: 22481935 PMCID: PMC3317191 DOI: 10.1155/2012/951724] [Citation(s) in RCA: 216] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 12/29/2011] [Indexed: 01/31/2023]
Abstract
The Akt family of serine/threonine protein kinases are key regulators of multiple aspects of cell behaviour, including proliferation, survival, metabolism, and tumorigenesis. Growth-factor-activated Akt signalling promotes progression through normal, unperturbed cell cycles by acting on diverse downstream factors involved in controlling the G1/S and G2/M transitions. Remarkably, several recent studies have also implicated Akt in modulating DNA damage responses and genome stability. High Akt activity can suppress ATR/Chk1 signalling and homologous recombination repair (HRR) via direct phosphorylation of Chk1 or TopBP1 or, indirectly, by inhibiting recruitment of double-strand break (DSB) resection factors, such as RPA, Brca1, and Rad51, to sites of damage. Loss of checkpoint and/or HRR proficiency is therefore a potential cause of genomic instability in tumor cells with high Akt. Conversely, Akt is activated by DNA double-strand breaks (DSBs) in a DNA-PK- or ATM/ATR-dependent manner and in some circumstances can contribute to radioresistance by stimulating DNA repair by nonhomologous end joining (NHEJ). Akt therefore modifies both the response to and repair of genotoxic damage in complex ways that are likely to have important consequences for the therapy of tumors with deregulation of the PI3K-Akt-PTEN pathway.
Collapse
|
55
|
|
56
|
Xu LH, Huang M, Fang SG, Liu DX. Coronavirus infection induces DNA replication stress partly through interaction of its nonstructural protein 13 with the p125 subunit of DNA polymerase δ. J Biol Chem 2011; 286:39546-59. [PMID: 21918226 PMCID: PMC3234778 DOI: 10.1074/jbc.m111.242206] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 08/29/2011] [Indexed: 12/20/2022] Open
Abstract
Perturbation of cell cycle regulation is a characteristic feature of infection by many DNA and RNA viruses, including Coronavirus infectious bronchitis virus (IBV). IBV infection was shown to induce cell cycle arrest at both S and G(2)/M phases for the enhancement of viral replication and progeny production. However, the underlying mechanisms are not well explored. In this study we show that activation of cellular DNA damage response is one of the mechanisms exploited by Coronavirus to induce cell cycle arrest. An ATR-dependent cellular DNA damage response was shown to be activated by IBV infection. Suppression of the ATR kinase activity by chemical inhibitors and siRNA-mediated knockdown of ATR reduced the IBV-induced ATR signaling and inhibited the replication of IBV. Furthermore, yeast two-hybrid screens and subsequent biochemical and functional studies demonstrated that interaction between Coronavirus nsp13 and DNA polymerase δ induced DNA replication stress in IBV-infected cells. These findings indicate that the ATR signaling activated by IBV replication contributes to the IBV-induced S-phase arrest and is required for efficient IBV replication and progeny production.
Collapse
Affiliation(s)
- Ling Hui Xu
- From the School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Mei Huang
- From the School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Shou Guo Fang
- From the School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Ding Xiang Liu
- From the School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| |
Collapse
|
57
|
Abstract
The deubiquitylating enzyme USP7 (HAUSP) sits at a critical node regulating the activities of numerous proteins broadly characterized as tumor suppressors, DNA repair proteins, immune responders, viral proteins, and epigenetic modulators. Aberrant USP7 activity may promote oncogenesis and viral disease making it a compelling target for therapeutic intervention. Disclosed drug discovery programs have identified inhibitors of USP7 such as P005091 with cellular proof of concept and anti-proliferative activity in cancer models. Taken together, USP7 inhibitors hold promise as a new strategy for the treatment of disease.
Collapse
|
58
|
C-terminal domain of Chk1 regulates its subcellular location and kinase activity for DNA repair. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/s11434-011-4538-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
59
|
Sinha S, Singh RK, Bhattacharya N, Mukherjee N, Ghosh S, Alam N, Roy A, Roychoudhury S, Panda CK. Frequent alterations of LOH11CR2A, PIG8 and CHEK1 genes at chromosomal 11q24.1-24.2 region in breast carcinoma: clinical and prognostic implications. Mol Oncol 2011; 5:454-64. [PMID: 21803008 PMCID: PMC5528301 DOI: 10.1016/j.molonc.2011.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 06/28/2011] [Indexed: 11/24/2022] Open
Abstract
To understand the importance of frequent deletions at chromosome 11q24.1-24.2 region in breast carcinoma, alterations (deletion/methylation) of the candidate genes LOH11CR2A, ROBO3, ROBO4, HEPACAM, PIG8 and CHEK1 located in this region were analyzed in 106 breast carcinoma samples. Among these genes, LOH11CR2A showed highest frequency of deletion (56%), followed by PIG8 (35%), CHEK1 (31%) and ROBO3/ROBO4/HEPACAM loci (28%). Comparable frequency of promoter methylation (26-35%) was observed for LOH11CR2A, CHEK1 and PIG8. Overall alterations (deletion/methylation) of these genes were in the following order: LOH11CR2A (60%) > PIG8 (46%) > CHEK1 (41%) and showed significant association with each other. Breast carcinoma samples that were estrogen/progesterone receptor negative showed significantly high deletion and overall alterations than estrogen/progesterone receptor positive samples for LOH11CR2A, CHEK1 and PIG8. The methylation and overall alteration of LOH11CR2A were significantly associated with tumor stages in breast carcinoma. However, in early/late onset and estrogen/progesterone receptor positive/negative breast carcinoma, the overall alterations of LOH11CR2A, PIG8 and CHEK1 were differentially associated with advanced stages, tumor grade and lymph node metastasis. Alterations of PIG8 and CHEK1 were significantly associated with poor prognosis in patients with early age of onset of the disease indicating significant prognostic importance. Quantitative mRNA expression analysis detected reduced expression of the genes in the order LOH11CR2A > CHEK1 > PIG8. Immunohistochemical analysis showed reduced protein expression of PIG8 and CHEK1 that was concordant with their molecular alterations. Thus, our study suggests that LOH11CR2A, PIG8 and CHEK1 are candidate tumor suppressor genes associated with breast carcinoma and have significant clinical as well as prognostic importance.
Collapse
MESH Headings
- Adult
- Aged
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms, Male/genetics
- Breast Neoplasms, Male/pathology
- Cell Line, Tumor
- Checkpoint Kinase 1
- Chromosome Deletion
- Chromosomes, Human, Pair 11/genetics
- DNA Methylation/genetics
- Female
- Genes, Neoplasm/genetics
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Male
- Middle Aged
- Mutation/genetics
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Polymorphism, Single Nucleotide/genetics
- Prognosis
- Promoter Regions, Genetic
- Protein Kinases/genetics
- Protein Kinases/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Young Adult
Collapse
Affiliation(s)
- Satyabrata Sinha
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Ratnesh K. Singh
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nilanjana Bhattacharya
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nupur Mukherjee
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Susmita Ghosh
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anup Roy
- Department of Pathology, Midnapur Medical College and Hospital, Midnapur, India
| | - Susanta Roychoudhury
- Molecular and Human Genetics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
60
|
Feng Z, Zhang J. A dual role of BRCA1 in two distinct homologous recombination mediated repair in response to replication arrest. Nucleic Acids Res 2011; 40:726-38. [PMID: 21954437 PMCID: PMC3258139 DOI: 10.1093/nar/gkr748] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Homologous recombination (HR) is a major mechanism utilized to repair blockage of DNA replication forks. Here, we report that a sister chromatid exchange (SCE) generated by crossover-associated HR efficiently occurs in response to replication fork stalling before any measurable DNA double-strand breaks (DSBs). Interestingly, SCE produced by replication fork collapse following DNA DSBs creation is specifically suppressed by ATR, a central regulator of the replication checkpoint. BRCA1 depletion leads to decreased RPA2 phosphorylation (RPA2-P) following replication fork stalling but has no obvious effect on RPA2-P following replication fork collapse. Importantly, we found that BRCA1 promotes RAD51 recruitment and SCE induced by replication fork stalling independent of ATR. In contrast, BRCA1 depletion leads to a more profound defect in RAD51 recruitment and SCE induced by replication fork collapse when ATR is depleted. We concluded that BRCA1 plays a dual role in two distinct HR-mediated repair upon replication fork stalling and collapse. Our data established a molecular basis for the observation that defective BRCA1 leads to a high sensitivity to agents that cause replication blocks without being associated with DSBs, and also implicate a novel mechanism by which loss of cell cycle checkpoints promotes BRCA1-associated tumorigenesis via enhancing HR defect resulting from BRCA1 deficiency.
Collapse
Affiliation(s)
- Zhihui Feng
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Boulevard, St. Louis, Missouri 63108, USA
| | | |
Collapse
|
61
|
Xu N, Libertini S, Zhang Y, Gillespie DA. Cdk phosphorylation of Chk1 regulates efficient Chk1 activation and multiple checkpoint proficiency. Biochem Biophys Res Commun 2011; 413:465-70. [PMID: 21907702 DOI: 10.1016/j.bbrc.2011.08.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 08/26/2011] [Indexed: 10/17/2022]
Abstract
It has been reported previously that both Cdk1 and Cdk2 phosphorylate Chk1 in a cell-cycle dependent manner. Cdk-mediated phosphorylation is required for efficient activation of Chk1 and checkpoint proficiency in response to DNA damage. Here, we demonstrate that Cdk-mediated phosphorylation is also required for replication stress induced Chk1 activation and S/M checkpoint proficiency. Re-introduction of Chk1 mutant (S286A/S301A) into Chk1 deficient cells is capable of restraining mitosis in cells with completely unreplicated DNA, but the mitotic delay at later stage of the cell cycle is largely impaired. The mutation strongly attenuates aphidicolin induced Chk1 activation without altering the S-phase dependent Chk1 activation. These data indicate that Cdk-mediated phosphorytion is required for efficient Chk1 activation and multiple checkpoint proficiency.
Collapse
Affiliation(s)
- Naihan Xu
- Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China.
| | | | | | | |
Collapse
|
62
|
Structure-specific DNA endonuclease Mus81/Eme1 generates DNA damage caused by Chk1 inactivation. PLoS One 2011; 6:e23517. [PMID: 21858151 PMCID: PMC3157403 DOI: 10.1371/journal.pone.0023517] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 07/20/2011] [Indexed: 01/17/2023] Open
Abstract
The DNA-damage checkpoint kinase Chk1 is essential in higher eukaryotes due to its role in maintaining genome stability in proliferating cells. CHK1 gene deletion is embryonically lethal, and Chk1 inhibition in replicating cells causes cell-cycle defects that eventually lead to perturbed replication and replication-fork collapse, thus generating endogenous DNA damage. What is the cause of replication-fork collapse when Chk1 is inactivated, however, remains poorly understood. Here, we show that generation of DNA double-strand breaks at replication forks when Chk1 activity is compromised relies on the DNA endonuclease complex Mus81/Eme1. Importantly, we show that Mus81/Eme1-dependent DNA damage—rather than a global increase in replication-fork stalling—is the cause of incomplete replication in Chk1-deficient cells. Consequently, Mus81/Eme1 depletion alleviates the S-phase progression defects associated with Chk1 deficiency, thereby increasing cell survival. Chk1-mediated protection of replication forks from Mus81/Eme1 even under otherwise unchallenged conditions is therefore vital to prevent uncontrolled fork collapse and ensure proper S-phase progression in human cells.
Collapse
|
63
|
Ye W, Blain SW. Chk1 has an essential role in the survival of differentiated cortical neurons in the absence of DNA damage. Apoptosis 2011; 16:449-59. [PMID: 21336968 DOI: 10.1007/s10495-011-0579-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Neuronal death in the central nervous system contributes to the development of age-related neurodegeneration. The ATR/Chk1 pathway appears to function neuroprotectively to prevent DNA damage induced by cytotoxic agents. Here, we examine the function of Chk1 on cell viability of cortical neurons in the absence of additional DNA damaging stimuli. The Chk1-specific inhibitor, UCN-01, and the ATR inhibitor, Caffeine, cause neuronal apoptosis in differentiated neurons in the absence of additional treatment, whereas inhibition of ATM or Chk2, does not. UCN-01 treatment increased the detection of γ-H2AX phosphorylation, DNA strand breaks, and an activated p53-dependent DNA damage response (DDR), suggesting that Chk1 normally helps to maintain genomic stability. UCN-01 treatment also enhanced the apoptosis seen in neurons treated with DNA damaging agents, such as camptothecin (CPT). Our results indicate that Chk1 is essential for neuronal survival, and perturbation of this pathway increases a cell's sensitivity to naturally accumulating DNA damage.
Collapse
Affiliation(s)
- Weizhen Ye
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, 12003, USA
| | | |
Collapse
|
64
|
Xu N, Libertini S, Black EJ, Lao Y, Hegarat N, Walker M, Gillespie DA. Cdk-mediated phosphorylation of Chk1 is required for efficient activation and full checkpoint proficiency in response to DNA damage. Oncogene 2011; 31:1086-94. [PMID: 21765472 DOI: 10.1038/onc.2011.310] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Here, we show that activation of the checkpoint effector kinase Chk1 in response to irradiation-induced DNA damage is minimal in G1, maximal during S-phase and diminishes as cells enter G2. In addition, formation of irradiation-induced replication protein A (RPA)-coated single-stranded DNA (RPA-ssDNA), a structure required for ATM and Rad3-related (ATR)-Chk1 activation, occurs in a broadly similar pattern. Cyclin-dependent kinase (Cdk) activity is thought to promote RPA-ssDNA formation by stimulating DNA strand resection at double-strand breaks (DSBs), providing one possible mechanism of imposing cell cycle dependence on DNA damage signaling. However, it has recently been shown that Chk1 itself is also subject to Cdk-mediated phosphorylation at serines 286 and 301 (S286 and 301). We show that Chk1 S301 phosphorylation increases as cells progress through S and G2 and that both Cdk1 and Cdk2 are likely to contribute to this modification in vivo. We also find that substitution of S286 and S301 with non-phosphorylatable alanine residues strongly attenuates DNA damage-induced Chk1 activation and G2 checkpoint proficiency, but does not eliminate the underlying cell cycle dependence of Chk1 regulation. Taken together, these data indicate that Cdk activity regulates multiple steps in the DNA damage response pathway including full activation of Chk1 and checkpoint proficiency.
Collapse
Affiliation(s)
- N Xu
- Beatson Institute for Cancer Research, Garscube Estate, Glasgow, UK.
| | | | | | | | | | | | | |
Collapse
|
65
|
How dormant origins promote complete genome replication. Trends Biochem Sci 2011; 36:405-14. [PMID: 21641805 DOI: 10.1016/j.tibs.2011.05.002] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/26/2011] [Accepted: 05/04/2011] [Indexed: 01/21/2023]
Abstract
Many replication origins that are licensed by loading MCM2-7 complexes in G1 are not normally used. Activation of these dormant origins during S phase provides a first line of defence for the genome if replication is inhibited. When replication forks fail, dormant origins are activated within regions of the genome currently engaged in replication. At the same time, DNA damage-response kinases activated by the stalled forks preferentially suppress the assembly of new replication factories, thereby ensuring that chromosomal regions experiencing replicative stress complete synthesis before new regions of the genome are replicated. Mice expressing reduced levels of MCM2-7 have fewer dormant origins, are cancer-prone and are genetically unstable, demonstrating the importance of dormant origins for preserving genome integrity. We review the function of dormant origins, the molecular mechanism of their regulation and their physiological implications.
Collapse
|
66
|
Spadafora ND, Doonan JH, Herbert RJ, Bitonti MB, Wallace E, Rogers HJ, Francis D. Arabidopsis T-DNA insertional lines for CDC25 are hypersensitive to hydroxyurea but not to zeocin or salt stress. ANNALS OF BOTANY 2011; 107:1183-92. [PMID: 20647223 PMCID: PMC3091795 DOI: 10.1093/aob/mcq142] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 06/08/2010] [Accepted: 06/09/2010] [Indexed: 05/21/2023]
Abstract
BACKGROUND AND AIMS In yeasts and animals, cyclin-dependent kinases are key regulators of cell cycle progression and are negatively and positively regulated by WEE1 kinase and CDC25 phosphatase, respectively. In higher plants a full-length orthologue of CDC25 has not been isolated but a shorter gene with homology only to the C-terminal catalytic domain is present. The Arabidopis thaliana;CDC25 can act as a phosphatase in vitro. Since in arabidopsis, WEE1 plays an important role in the DNA damage/DNA replication checkpoints, the role of Arath;CDC25 in conditions that induce these checkpoints or induce abiotic stress was tested. Methods arath;cdc25 T-DNA insertion lines, Arath;CDC25 over-expressing lines and wild type were challenged with hydroxyurea (HU) and zeocin, substances that stall DNA replication and damage DNA, respectively, together with an abiotic stressor, NaCl. A molecular and phenotypic assessment was made of all genotypes Key RESULTS There was a null phenotypic response to perturbation of Arath;CDC25 expression under control conditions. However, compared with wild type, the arath;cdc25 T-DNA insertion lines were hypersensitive to HU, whereas the Arath;CDC25 over-expressing lines were relatively insensitive. In particular, the over-expressing lines consistently outgrew the T-DNA insertion lines and wild type when challenged with HU. All genotypes were equally sensitive to zeocin and NaCl. CONCLUSIONS Arath;CDC25 plays a role in overcoming stress imposed by HU, an agent know to induce the DNA replication checkpoint in arabidopsis. However, it could not enhance tolerance to either a zeocin treatment, known to induce DNA damage, or salinity stress.
Collapse
Affiliation(s)
- Natasha D. Spadafora
- School of Biosciences, Cardiff University, Main College, Cardiff CF10 3AT, UK
- Institute of Science and the Environment, University of Worcester, Henwick Grove, Worcester WR2 6AJ, UK
| | - John H. Doonan
- Department of Cell Biology, John Innes Centre, Colney Lane, Norwich NR4 7UH, UK
| | - Robert J. Herbert
- Institute of Science and the Environment, University of Worcester, Henwick Grove, Worcester WR2 6AJ, UK
| | - M. Beatrice Bitonti
- Dipartimento di Ecologia, Università della Calabria, Arcavacata di Rende, I-87030 Cosenza, Italy
| | - Emily Wallace
- School of Biosciences, Cardiff University, Main College, Cardiff CF10 3AT, UK
| | - Hilary J. Rogers
- School of Biosciences, Cardiff University, Main College, Cardiff CF10 3AT, UK
| | - Dennis Francis
- School of Biosciences, Cardiff University, Main College, Cardiff CF10 3AT, UK
| |
Collapse
|
67
|
Smith J, Tho LM, Xu N, Gillespie DA. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res 2011; 108:73-112. [PMID: 21034966 DOI: 10.1016/b978-0-12-380888-2.00003-0] [Citation(s) in RCA: 916] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
DNA damage is a key factor both in the evolution and treatment of cancer. Genomic instability is a common feature of cancer cells, fuelling accumulation of oncogenic mutations, while radiation and diverse genotoxic agents remain important, if imperfect, therapeutic modalities. Cellular responses to DNA damage are coordinated primarily by two distinct kinase signaling cascades, the ATM-Chk2 and ATR-Chk1 pathways, which are activated by DNA double-strand breaks (DSBs) and single-stranded DNA respectively. Historically, these pathways were thought to act in parallel with overlapping functions; however, more recently it has become apparent that their relationship is more complex. In response to DSBs, ATM is required both for ATR-Chk1 activation and to initiate DNA repair via homologous recombination (HRR) by promoting formation of single-stranded DNA at sites of damage through nucleolytic resection. Interestingly, cells and organisms survive with mutations in ATM or other components required for HRR, such as BRCA1 and BRCA2, but at the cost of genomic instability and cancer predisposition. By contrast, the ATR-Chk1 pathway is the principal direct effector of the DNA damage and replication checkpoints and, as such, is essential for the survival of many, although not all, cell types. Remarkably, deficiency for HRR in BRCA1- and BRCA2-deficient tumors confers sensitivity to cisplatin and inhibitors of poly(ADP-ribose) polymerase (PARP), an enzyme required for repair of endogenous DNA damage. In addition, suppressing DNA damage and replication checkpoint responses by inhibiting Chk1 can enhance tumor cell killing by diverse genotoxic agents. Here, we review current understanding of the organization and functions of the ATM-Chk2 and ATR-Chk1 pathways and the prospects for targeting DNA damage signaling processes for therapeutic purposes.
Collapse
Affiliation(s)
- Joanne Smith
- Beatson Institute for Cancer Research, Garscube Estate, Glasgow, UK
| | | | | | | |
Collapse
|
68
|
Guzi TJ, Paruch K, Dwyer MP, Labroli M, Shanahan F, Davis N, Taricani L, Wiswell D, Seghezzi W, Penaflor E, Bhagwat B, Wang W, Gu D, Hsieh Y, Lee S, Liu M, Parry D. Targeting the replication checkpoint using SCH 900776, a potent and functionally selective CHK1 inhibitor identified via high content screening. Mol Cancer Ther 2011; 10:591-602. [PMID: 21321066 DOI: 10.1158/1535-7163.mct-10-0928] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Checkpoint kinase 1 (CHK1) is an essential serine/threonine kinase that responds to DNA damage and stalled DNA replication. CHK1 is essential for maintenance of replication fork viability during exposure to DNA antimetabolites. In human tumor cell lines, ablation of CHK1 function during antimetabolite exposure led to accumulation of double-strand DNA breaks and cell death. Here, we extend these observations and confirm ablation of CHK2 does not contribute to these phenotypes and may diminish them. Furthermore, concomitant suppression of cyclin-dependent kinase (CDK) activity is sufficient to completely antagonize the desired CHK1 ablation phenotypes. These mechanism-based observations prompted the development of a high-content, cell-based screen for γ-H2AX induction, a surrogate marker for double-strand DNA breaks. This mechanism-based functional approach was used to optimize small molecule inhibitors of CHK1. Specifically, the assay was used to mechanistically define the optimal in-cell profile with compounds exhibiting varying degrees of CHK1, CHK2, and CDK selectivity. Using this approach, SCH 900776 was identified as a highly potent and functionally optimal CHK1 inhibitor with minimal intrinsic antagonistic properties. SCH 900776 exposure phenocopies short interfering RNA-mediated CHK1 ablation and interacts synergistically with DNA antimetabolite agents in vitro and in vivo to selectively induce dsDNA breaks and cell death in tumor cell backgrounds.
Collapse
|
69
|
Ge XQ, Blow JJ. Chk1 inhibits replication factory activation but allows dormant origin firing in existing factories. J Cell Biol 2010; 191:1285-97. [PMID: 21173116 PMCID: PMC3010067 DOI: 10.1083/jcb.201007074] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 11/15/2010] [Indexed: 01/09/2023] Open
Abstract
Replication origins are licensed by loading MCM2-7 hexamers before entry into S phase. However, only ∼10% of licensed origins are normally used in S phase, with the others remaining dormant. When fork progression is inhibited, dormant origins initiate nearby to ensure that all of the DNA is eventually replicated. In apparent contrast, replicative stress activates ataxia telangiectasia and rad-3-related (ATR) and Chk1 checkpoint kinases that inhibit origin firing. In this study, we show that at low levels of replication stress, ATR/Chk1 predominantly suppresses origin initiation by inhibiting the activation of new replication factories, thereby reducing the number of active factories. At the same time, inhibition of replication fork progression allows dormant origins to initiate within existing replication factories. The inhibition of new factory activation by ATR/Chk1 therefore redirects replication toward active factories where forks are inhibited and away from regions that have yet to start replication. This minimizes the deleterious consequences of fork stalling and prevents similar problems from arising in unreplicated regions of the genome.
Collapse
Affiliation(s)
- Xin Quan Ge
- Wellcome Trust Centre for Gene Regulation and Expression, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | | |
Collapse
|
70
|
Petermann E, Woodcock M, Helleday T. Chk1 promotes replication fork progression by controlling replication initiation. Proc Natl Acad Sci U S A 2010; 107:16090-5. [PMID: 20805465 PMCID: PMC2941317 DOI: 10.1073/pnas.1005031107] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
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.
Collapse
Affiliation(s)
- Eva Petermann
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Mick Woodcock
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Thomas Helleday
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
- Department of Genetics, Microbiology and Toxicology, Stockholm University, S-106 91 Stockholm, Sweden; and
- Science for Life Laboratory, Stockholm University, Box 1031, SE-171 21 Solna, Sweden
| |
Collapse
|
71
|
Abstract
The role of Chk1 in the cellular response to DNA replication stress is well established. However recent work indicates a novel role for Chk1 in the suppression of apoptosis following the disruption of DNA replication or DNA damage. This review will consider these findings in the context of known pathways of Chk1 signalling and potential applications of therapies that target Chk1.
Collapse
Affiliation(s)
- Mark Meuth
- Institute for Cancer Studies, University of Sheffield, School of Medicine and Biomedical Sciences, Sheffield S10 2RX, UK.
| |
Collapse
|
72
|
Malzer E, Daly ML, Moloney A, Sendall TJ, Thomas SE, Ryder E, Ryoo HD, Crowther DC, Lomas DA, Marciniak SJ. Impaired tissue growth is mediated by checkpoint kinase 1 (CHK1) in the integrated stress response. J Cell Sci 2010; 123:2892-900. [PMID: 20682638 PMCID: PMC2923568 DOI: 10.1242/jcs.070078] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2010] [Indexed: 12/21/2022] Open
Abstract
The integrated stress response (ISR) protects cells from numerous forms of stress and is involved in the growth of solid tumours; however, it is unclear how the ISR acts on cellular proliferation. We have developed a model of ISR signalling with which to study its effects on tissue growth. Overexpression of the ISR kinase PERK resulted in a striking atrophic eye phenotype in Drosophila melanogaster that could be rescued by co-expressing the eIF2alpha phosphatase GADD34. A genetic screen of 3000 transposon insertions identified grapes, the gene that encodes the Drosophila orthologue of checkpoint kinase 1 (CHK1). Knockdown of grapes by RNAi rescued eye development despite ongoing PERK activation. In mammalian cells, CHK1 was activated by agents that induce ER stress, which resulted in a G2 cell cycle delay. PERK was both necessary and sufficient for CHK1 activation. These findings indicate that non-genotoxic misfolded protein stress accesses DNA-damage-induced cell cycle checkpoints to couple the ISR to cell cycle arrest.
Collapse
Affiliation(s)
- Elke Malzer
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
- Department of Genetics, University of Cambridge, Downing Site, Cambridge, CB2 3EH, UK
| | - Marie-Louise Daly
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Aileen Moloney
- Department of Genetics, University of Cambridge, Downing Site, Cambridge, CB2 3EH, UK
| | - Timothy J. Sendall
- Department of Genetics, University of Cambridge, Downing Site, Cambridge, CB2 3EH, UK
| | - Sally E. Thomas
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Edward Ryder
- Department of Genetics, University of Cambridge, Downing Site, Cambridge, CB2 3EH, UK
| | - Hyung Don Ryoo
- Department of Cell Biology, NYU Langone Medical Center, 550 First Avenue, New York, NY 1106, USA
| | - Damian C. Crowther
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
- Department of Genetics, University of Cambridge, Downing Site, Cambridge, CB2 3EH, UK
| | - David A. Lomas
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Stefan J. Marciniak
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| |
Collapse
|
73
|
Xu N, Hegarat N, Black EJ, Scott MT, Hochegger H, Gillespie DA. Akt/PKB suppresses DNA damage processing and checkpoint activation in late G2. ACTA ACUST UNITED AC 2010; 190:297-305. [PMID: 20679434 PMCID: PMC2922641 DOI: 10.1083/jcb.201003004] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activated Akt suppresses checkpoint activation by cells in late G2: although they are able to detect DNA damage, the repair pathway is put on hold until mitosis is complete. Using chemical genetics to reversibly inhibit Cdk1, we find that cells arrested in late G2 are unable to delay mitotic entry after irradiation. Late G2 cells detect DNA damage lesions and form γ-H2AX foci but fail to activate Chk1. This reflects a lack of DNA double-strand break processing because late G2 cells fail to recruit RPA (replication protein A), ATR (ataxia telangiectasia and Rad3 related), Rad51, or CtIP (C-terminal interacting protein) to sites of radiation-induced damage, events essential for both checkpoint activation and initiation of DNA repair by homologous recombination. Remarkably, inhibition of Akt/PKB (protein kinase B) restores DNA damage processing and Chk1 activation after irradiation in late G2. These data demonstrate a previously unrecognized role for Akt in cell cycle regulation of DNA repair and checkpoint activation. Because Akt/PKB is frequently activated in many tumor types, these findings have important implications for the evolution and therapy of such cancers.
Collapse
Affiliation(s)
- Naihan Xu
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, Scotland, UK
| | | | | | | | | | | |
Collapse
|
74
|
Mocciaro A, Berdougo E, Zeng K, Black E, Vagnarelli P, Earnshaw W, Gillespie D, Jallepalli P, Schiebel E. Vertebrate cells genetically deficient for Cdc14A or Cdc14B retain DNA damage checkpoint proficiency but are impaired in DNA repair. ACTA ACUST UNITED AC 2010; 189:631-9. [PMID: 20479464 PMCID: PMC2872905 DOI: 10.1083/jcb.200910057] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cdc14A and Cdc14B knockout cells with double-strand breaks still arrest in G2, but they fail to repair the damage. A recent study suggested that human Cdc14B phosphatase has a central function in the G2 DNA damage checkpoint. In this study, we show that chicken DT40, human HCT116, and human telomerase reverse transcription–immortalized retinal pigment epithelial cells deleted for the Cdc14A or Cdc14B gene are DNA damage checkpoint proficient and arrest efficiently in G2 in response to irradiation. Cdc14A knockout (KO) or Cdc14B-KO cells also maintain normal levels of Chk1 and Chk2 activation after irradiation. Surprisingly, however, irradiation-induced γ-H2A.X foci and DNA double-strand breaks persist longer in Cdc14A-KO or Cdc14B-KO cells than controls, suggesting that Cdc14 phosphatases are required for efficient DNA repair.
Collapse
Affiliation(s)
- Annamaria Mocciaro
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Allianz, 69117 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Li DQ, Ohshiro K, Khan MN, Kumar R. Requirement of MTA1 in ATR-mediated DNA damage checkpoint function. J Biol Chem 2010; 285:19802-12. [PMID: 20427275 PMCID: PMC2888391 DOI: 10.1074/jbc.m109.085258] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Revised: 04/27/2010] [Indexed: 11/06/2022] Open
Abstract
MTA1 (metastasis-associated protein 1), an integral component of the nucleosome remodeling and deacetylase complex, has recently been implicated in the ionizing radiation-induced DNA damage response. However, whether MTA1 also participates in the UV-induced DNA damage checkpoint pathway remains unknown. In response to UV radiation, ATR (ataxia teleangiectasia- and Rad3-related) is the major kinase activated that orchestrates cell cycle progression with DNA repair machinery by phosphorylating and activating a number of downstream substrates, such as Chk1 (checkpoint kinase 1) and H2AX (histone 2A variant X). Here, we report that UV radiation stabilizes MTA1 in an ATR-dependent manner and increases MTA1 binding to ATR. On the other hand, depletion of MTA1 compromises the ATR-mediated Chk1 activation following UV treatment, accompanied by a marked down-regulation of Chk1 and its interacting partner Claspin, an adaptor protein that is required for the phosphorylation and activation of Chk1 by ATR. Furthermore, MTA1 deficiency decreases the induction of phosphorylated H2AX (referred to as gamma-H2AX) and gamma-H2AX focus formation after UV treatment. Consequently, depletion of MTA1 results in a defect in the G(2)-M checkpoint and increases cellular sensitivity to UV-induced DNA damage. Thus, MTA1 is required for the activation of the ATR-Claspin-Chk1 and ATR-H2AX pathways following UV treatment, and the noted abrogation of the DNA damage checkpoint in the MTA1-depleted cells may be, at least in part, a consequence of dysregulation of the expression of these two pathways. These findings suggest that, in addition to its role in the repair of double strand breaks caused by ionizing radiation, MTA1 also participates in the UV-induced ATR-mediated DNA damage checkpoint pathway.
Collapse
Affiliation(s)
- Da-Qiang Li
- From the Department of Biochemistry and Molecular Biology, George Washington University Medical Center, Washington, D. C. 20037 and
| | - Kazufumi Ohshiro
- From the Department of Biochemistry and Molecular Biology, George Washington University Medical Center, Washington, D. C. 20037 and
| | - Mudassar N. Khan
- From the Department of Biochemistry and Molecular Biology, George Washington University Medical Center, Washington, D. C. 20037 and
| | - Rakesh Kumar
- From the Department of Biochemistry and Molecular Biology, George Washington University Medical Center, Washington, D. C. 20037 and
- the Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
76
|
Barr AR, Kilmartin JV, Gergely F. CDK5RAP2 functions in centrosome to spindle pole attachment and DNA damage response. ACTA ACUST UNITED AC 2010; 189:23-39. [PMID: 20368616 PMCID: PMC2854379 DOI: 10.1083/jcb.200912163] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Two domains of centrosomal protein CDK5RAP2, CNN1 and CNN2, link centrosomes to mitotic spindle poles. CNN1 lacking centrosomes are unable to recruit pericentriolar matrix components that mediate attachment to spindle poles. The centrosomal protein, CDK5RAP2, is mutated in primary microcephaly, a neurodevelopmental disorder characterized by reduced brain size. The Drosophila melanogaster homologue of CDK5RAP2, centrosomin (Cnn), maintains the pericentriolar matrix (PCM) around centrioles during mitosis. In this study, we demonstrate a similar role for CDK5RAP2 in vertebrate cells. By disrupting two evolutionarily conserved domains of CDK5RAP2, CNN1 and CNN2, in the avian B cell line DT40, we find that both domains are essential for linking centrosomes to mitotic spindle poles. Although structurally intact, centrosomes lacking the CNN1 domain fail to recruit specific PCM components that mediate attachment to spindle poles. Furthermore, we show that the CNN1 domain enforces cohesion between parental centrioles during interphase and promotes efficient DNA damage–induced G2 cell cycle arrest. Because mitotic spindle positioning, asymmetric centrosome inheritance, and DNA damage signaling have all been implicated in cell fate determination during neurogenesis, our findings provide novel insight into how impaired CDK5RAP2 function could cause premature depletion of neural stem cells and thereby microcephaly.
Collapse
Affiliation(s)
- Alexis R Barr
- Cancer Research UK Cambridge Research Institute, Cambridge CB2 0RE, England, UK
| | | | | |
Collapse
|
77
|
Carrassa L, Montelatici E, Lazzari L, Zangrossi S, Simone M, Broggini M, Damia G. Role of Chk1 in the differentiation program of hematopoietic stem cells. Cell Mol Life Sci 2010; 67:1713-22. [PMID: 20146081 PMCID: PMC11115872 DOI: 10.1007/s00018-010-0274-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 12/23/2009] [Accepted: 01/15/2010] [Indexed: 02/05/2023]
Abstract
Hematopoietic stem cells (HSC) isolated from umbilical cord blood (UCB) were treated with ionizing radiation (IR) and sensitivity and IR induced checkpoints activation were investigated. No difference in the sensitivity and in the activation of DNA damage pathways was observed between CD133+ HSC and cells derived from them after ex vivo expansion. Chk1 protein was very low in freshly isolated CD133+ cells, and undetectable in ex vivo expanded UCB CD133+ cells. Chk1 was expressed only on day 3 of the ex vivo expansion. This pattern of Chk1 expression was corroborated in CD133+ cells isolated from peripheral blood apheresis collected from an healthy donor. Treatment with a specific Chk1 inhibitor resulted in a strong reduction in the percentage of myeloid precursors (CD33+) and an increase in the percentage of lymphoid precursors (CD38+) compared to untreated cells, suggesting a possible role for Chk1 in the differentiation program of UCB CD133+ HSC.
Collapse
Affiliation(s)
- Laura Carrassa
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
78
|
Yoshida K, Sugimoto N, Iwahori S, Yugawa T, Narisawa-Saito M, Kiyono T, Fujita M. CDC6 interaction with ATR regulates activation of a replication checkpoint in higher eukaryotic cells. J Cell Sci 2010; 123:225-35. [PMID: 20048340 DOI: 10.1242/jcs.058693] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
CDC6, a replication licensing protein, is partially exported to the cytoplasm in human cells through phosphorylation by Cdk during S phase, but a significant proportion remains in the nucleus. We report here that human CDC6 physically interacts with ATR, a crucial checkpoint kinase, in a manner that is stimulated by phosphorylation by Cdk. CDC6 silencing by siRNAs affected ATR-dependent inhibition of mitotic entry elicited by modest replication stress. Whereas a Cdk-phosphorylation-mimicking CDC6 mutant could rescue the checkpoint defect by CDC6 silencing, a phosphorylation-deficient mutant could not. Furthermore, we found that the CDC6-ATR interaction is conserved in Xenopus. We show that the presence of Xenopus CDC6 during S phase is essential for Xenopus ATR to bind to chromatin in response to replication inhibition. In addition, when human CDC6 amino acid fragment 180-220, which can bind to both human and Xenopus ATR, was added to Xenopus egg extracts after assembly of the pre-replication complex, Xenopus Chk1 phosphorylation was significantly reduced without lowering replication, probably through a sequestration of CDC6-mediated ATR-chromatin interaction. Thus, CDC6 might regulate replication-checkpoint activation through the interaction with ATR in higher eukaryotic cells.
Collapse
Affiliation(s)
- Kazumasa Yoshida
- Virology Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuohku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | |
Collapse
|
79
|
Petermann E, Orta ML, Issaeva N, Schultz N, Helleday T. Hydroxyurea-stalled replication forks become progressively inactivated and require two different RAD51-mediated pathways for restart and repair. Mol Cell 2010; 37:492-502. [PMID: 20188668 PMCID: PMC2958316 DOI: 10.1016/j.molcel.2010.01.021] [Citation(s) in RCA: 655] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 10/21/2009] [Accepted: 12/10/2009] [Indexed: 11/30/2022]
Abstract
Faithful DNA replication is essential to all life. Hydroxyurea (HU) depletes the cells of dNTPs, which initially results in stalled replication forks that, after prolonged treatment, collapse into DSBs. Here, we report that stalled replication forks are efficiently restarted in a RAD51-dependent process that does not trigger homologous recombination (HR). The XRCC3 protein, which is required for RAD51 foci formation, is also required for replication restart of HU-stalled forks, suggesting that RAD51-mediated strand invasion supports fork restart. In contrast, replication forks collapsed by prolonged replication blocks do not restart, and global replication is rescued by new origin firing. We find that RAD51-dependent HR is triggered for repair of collapsed replication forks, without apparent restart. In conclusion, our data suggest that restart of stalled replication forks and HR repair of collapsed replication forks require two distinct RAD51-mediated pathways.
Collapse
Affiliation(s)
- Eva Petermann
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Oxford OX3 7DQ, UK
| | - Manuel Luís Orta
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Oxford OX3 7DQ, UK
- Department of Cell Biology, University of Seville, E-41012 Seville, Spain
| | - Natalia Issaeva
- Department of Genetics Microbiology and Toxicology, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Niklas Schultz
- Department of Genetics Microbiology and Toxicology, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Thomas Helleday
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Oxford OX3 7DQ, UK
- Department of Genetics Microbiology and Toxicology, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
80
|
Schwab RA, Blackford AN, Niedzwiedz W. ATR activation and replication fork restart are defective in FANCM-deficient cells. EMBO J 2010; 29:806-18. [PMID: 20057355 PMCID: PMC2829160 DOI: 10.1038/emboj.2009.385] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 11/24/2009] [Indexed: 01/20/2023] Open
Abstract
Fanconi anaemia is a chromosomal instability disorder associated with cancer predisposition and bone marrow failure. Among the 13 identified FA gene products only one, the DNA translocase FANCM, has homologues in lower organisms, suggesting a conserved function in DNA metabolism. However, a precise role for FANCM in DNA repair remains elusive. Here, we show a novel function for FANCM that is distinct from its role in the FA pathway: promoting replication fork restart and simultaneously limiting the accumulation of RPA-ssDNA. We show that in DT40 cells this process is controlled by ATR and PLK1, and that in the absence of FANCM, stalled replication forks are unable to resume DNA synthesis and genome duplication is ensured by excess origin firing. Unexpectedly, we also uncover an early role for FANCM in ATR-mediated checkpoint signalling by promoting chromatin retention of TopBP1. Failure to retain TopBP1 on chromatin impacts on the ability of ATR to phosphorylate downstream molecular targets, including Chk1 and SMC1. Our data therefore indicate a fundamental role for FANCM in the maintenance of genome integrity during S phase.
Collapse
Affiliation(s)
- Rebekka A Schwab
- Department of Molecular Oncology, Weatherall Institute of Molecular Medicine, Oxford University, Oxford, UK
| | - Andrew N Blackford
- Department of Molecular Oncology, Weatherall Institute of Molecular Medicine, Oxford University, Oxford, UK
| | - Wojciech Niedzwiedz
- Department of Molecular Oncology, Weatherall Institute of Molecular Medicine, Oxford University, Oxford, UK
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawinskiego 5a, Warsaw, Poland
| |
Collapse
|
81
|
Carrassa L, Sanchez Y, Erba E, Damia G. U2OS cells lacking Chk1 undergo aberrant mitosis and fail to activate the spindle checkpoint. J Cell Mol Med 2010; 13:1565-76. [PMID: 19778378 DOI: 10.1111/j.1582-4934.2009.00362.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Chk1 is a conserved protein kinase originally identified in fission yeast, required to delay entry of cells with damaged or unreplicated DNA into mitosis. The requirement of Chk1 for both S and G2/M checkpoints has been elucidated while only few studies have connected Chk1 to the mitotic spindle checkpoint. We used a small interference RNA strategy to investigate the role of Chk1 in unstressed conditions. Chk1 depletion in U2OS human osteosarcoma cells inhibited cell proliferation and raised the percentage of cells with a 4N DNA content, which correlated with accumulation of giant polynucleated cells morphologically distinct from apoptotic cells, while no increased number of cells in G2 or mitosis could be detected. Down-regulation of Chk1 also caused accumulation of cells in the last step of cytokinesis, and of tetraploid cells in G1 phase, which coincided with activation of p53 and increased levels of p21. In addition, Chk1-depleted U2OS cells failed to arrest in mitosis after spindle disruption by nocodazole and showed decreased protein levels of Mad2 and BubR1. These studies show that U2OS cells lacking Chk1 undergo abnormal mitosis and fail to activate the spindle checkpoint, suggesting a role of Chk1 in this checkpoint.
Collapse
Affiliation(s)
- Laura Carrassa
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| | | | | | | |
Collapse
|
82
|
Carrassa L, Sanchez Y, Erba E, Damia G. U2OS cells lacking Chk1 undergo aberrant mitosis and fail to activate the spindle checkpoint. J Cell Mol Med 2010. [PMID: 19778378 PMCID: PMC3828867 DOI: 10.1111/j.1582-4934.2008.00362.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Chk1 is a conserved protein kinase originally identified in fission yeast, required to delay entry of cells with damaged or unreplicated DNA into mitosis. The requirement of Chk1 for both S and G2/M checkpoints has been elucidated while only few studies have connected Chk1 to the mitotic spindle checkpoint. We used a small interference RNA strategy to investigate the role of Chk1 in unstressed conditions. Chk1 depletion in U2OS human osteosarcoma cells inhibited cell proliferation and raised the percentage of cells with a 4N DNA content, which correlated with accumulation of giant polynucleated cells morphologically distinct from apoptotic cells, while no increased number of cells in G2 or mitosis could be detected. Down-regulation of Chk1 also caused accumulation of cells in the last step of cytokinesis, and of tetraploid cells in G1 phase, which coincided with activation of p53 and increased levels of p21. In addition, Chk1-depleted U2OS cells failed to arrest in mitosis after spindle disruption by nocodazole and showed decreased protein levels of Mad2 and BubR1. These studies show that U2OS cells lacking Chk1 undergo abnormal mitosis and fail to activate the spindle checkpoint, suggesting a role of Chk1 in this checkpoint.
Collapse
Affiliation(s)
- Laura Carrassa
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| | | | | | | |
Collapse
|
83
|
Buonomo SBC, Wu Y, Ferguson D, de Lange T. Mammalian Rif1 contributes to replication stress survival and homology-directed repair. ACTA ACUST UNITED AC 2010; 187:385-98. [PMID: 19948482 PMCID: PMC2779251 DOI: 10.1083/jcb.200902039] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Rif1, originally recognized for its role at telomeres in budding yeast, has been implicated in a wide variety of cellular processes in mammals, including pluripotency of stem cells, response to double-strand breaks, and breast cancer development. As the molecular function of Rif1 is not known, we examined the consequences of Rif1 deficiency in mouse cells. Rif1 deficiency leads to failure in embryonic development, and conditional deletion of Rif1 from mouse embryo fibroblasts affects S-phase progression, rendering cells hypersensitive to replication poisons. Rif1 deficiency does not alter the activation of the DNA replication checkpoint but rather affects the execution of repair. RNA interference to human Rif1 decreases the efficiency of homology-directed repair (HDR), and Rif1 deficiency results in aberrant aggregates of the HDR factor Rad51. Consistent with a role in S-phase progression, Rif1 accumulates at stalled replication forks, preferentially around pericentromeric heterochromatin. Collectively, these findings reveal a function for Rif1 in the repair of stalled forks by facilitating HDR.
Collapse
Affiliation(s)
- Sara B C Buonomo
- Laboratory of Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | |
Collapse
|
84
|
Hubbard J, Erlichman C, Toft DO, Qin R, Stensgard BA, Felten S, Ten Eyck C, Batzel G, Ivy SP, Haluska P. Phase I study of 17-allylamino-17 demethoxygeldanamycin, gemcitabine and/or cisplatin in patients with refractory solid tumors. Invest New Drugs 2010; 29:473-80. [PMID: 20082116 DOI: 10.1007/s10637-009-9381-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 12/20/2009] [Indexed: 11/27/2022]
Abstract
PURPOSE To determine the maximum tolerated dose (MTD) and characterize the dose-limiting toxicities (DLT) of 17-AAG, gemcitabine and/or cisplatin. Levels of the proteins Hsp90, Hsp70 and ILK were measured in peripheral blood mononuclear cell (PMBC) lysates to assess the effects of 17-AAG. EXPERIMENTAL DESIGN Phase I dose-escalating trial using a "3 + 3" design performed in patients with advanced solid tumors. Once the MTD of gemcitabine + 17-AAG + cisplatin was determined, dose escalation of 17-AAG with constant doses of gemcitabine and cisplatin was attempted. After significant hematologic toxicity occurred, the protocol was amended to evaluate three cohorts: gemcitabine and 17-AAG; 17-AAG and cisplatin; and gemcitabine, 17-AAG and cisplatin with modified dosing. RESULTS The 39 patients enrolled were evaluable for toxicity and response. The MTD for cohort A was 154 mg/m(2) of 17-AAG, 750 mg/m(2) of gemcitabine, and 40 mg/m(2) of cisplatin. In cohort A, DLTs were observed at the higher dose level and included neutropenia, hyperbilirubinemia, dehydration, GGT elevation, hyponatremia, nausea, vomiting, and thrombocytopenia. The MTD for cohort C was 154 mg/m(2) of 17-AAG and 750 mg/m(2) of gemcitabine, with one DLT observed (alkaline phosphatase elevation) observed. In cohort C, DLTs of thrombocytopenia, fever and dyspnea were seen at the higher dose level. The remaining cohorts were closed to accrual due to toxicity. Six patients experienced partial responses. Mean Hsp90 levels were decreased and levels of Hsp70 were increased compared to baseline. CONCLUSIONS 17-AAG in combination with gemcitabine and cisplatin demonstrated antitumor activity, but significant hematologic toxicities were encountered. 17-AAG combined with gemcitabine is tolerable and has demonstrated evidence of activity at the MTD. The recommended phase II dose is defined as 154 mg/m(2) of 17-AAG and 750 mg/m(2) of gemcitabine, and is currently being investigated in phase II studies in ovarian and pancreatic cancers. There is no recommended phase II dose for the cisplatin-containing combinations.
Collapse
Affiliation(s)
- Joleen Hubbard
- Division of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Gagou ME, Zuazua-Villar P, Meuth M. Enhanced H2AX phosphorylation, DNA replication fork arrest, and cell death in the absence of Chk1. Mol Biol Cell 2010; 21:739-52. [PMID: 20053681 PMCID: PMC2828961 DOI: 10.1091/mbc.e09-07-0618] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
H2AX phosphorylation at serine 139 (gammaH2AX) is a sensitive indicator of both DNA damage and DNA replication stress. Here we show that gammaH2AX formation is greatly enhanced in response to replication inhibitors but not ionizing radiation in HCT116 or SW480 cells depleted of Chk1. Although H2AX phosphorylation precedes the induction of apoptosis in such cells, our results suggest that cells containing gammaH2AX are not committed to death. gammaH2AX foci in these cells largely colocalize with RPA foci and their formation is dependent upon the essential replication helicase cofactor Cdc45, suggesting that H2AX phosphorylation occurs at sites of stalled forks. However Chk1-depleted cells released from replication inhibitors retain gammaH2AX foci and do not appear to resume replicative DNA synthesis. BrdU incorporation only occurs in a minority of Chk1-depleted cells containing gammaH2AX foci after release from thymidine arrest and, in cells incorporating BrdU, DNA synthesis does not occur at sites of gammaH2AX foci. Furthermore activated ATM and Chk2 persist in these cells. We propose that the gammaH2AX foci in Chk1-depleted cells may represent sites of persistent replication fork damage or abandonment that are unable to resume DNA synthesis but do not play a direct role in the Chk1 suppressed death pathway.
Collapse
Affiliation(s)
- Mary E Gagou
- Institute for Cancer Studies, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | | | | |
Collapse
|
86
|
Smith KD, Fu MA, Brown EJ. Tim-Tipin dysfunction creates an indispensible reliance on the ATR-Chk1 pathway for continued DNA synthesis. ACTA ACUST UNITED AC 2009; 187:15-23. [PMID: 19805627 PMCID: PMC2762102 DOI: 10.1083/jcb.200905006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased amounts of single-stranded DNA accumulate at replication forks when Tim–Tipin isn't around, activating an ATR-mediated DNA damage response necessary for DNA replication to proceed. The Tim (Timeless)–Tipin complex has been proposed to maintain genome stability by facilitating ATR-mediated Chk1 activation. However, as a replisome component, Tim–Tipin has also been suggested to couple DNA unwinding to synthesis, an activity expected to suppress single-stranded DNA (ssDNA) accumulation and limit ATR–Chk1 pathway engagement. We now demonstrate that Tim–Tipin depletion is sufficient to increase ssDNA accumulation at replication forks and stimulate ATR activity during otherwise unperturbed DNA replication. Notably, suppression of the ATR–Chk1 pathway in Tim–Tipin-deficient cells completely abrogates nucleotide incorporation in S phase, indicating that the ATR-dependent response to Tim–Tipin depletion is indispensible for continued DNA synthesis. Replication failure in ATR/Tim-deficient cells is strongly associated with synergistic increases in H2AX phosphorylation and DNA double-strand breaks, suggesting that ATR pathway activation preserves fork stability in instances of Tim–Tipin dysfunction. Together, these experiments indicate that the Tim–Tipin complex stabilizes replication forks both by preventing the accumulation of ssDNA upstream of ATR–Chk1 function and by facilitating phosphorylation of Chk1 by ATR.
Collapse
Affiliation(s)
- Kevin D Smith
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
87
|
DNA damage induces Chk1-dependent threonine-160 phosphorylation and activation of Cdk2. Oncogene 2009; 29:616-24. [PMID: 19838212 DOI: 10.1038/onc.2009.340] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Abnormal centrosome numbers arise in tumours and can cause multipolar mitoses and genome instability. Cdk2 controls normal centrosome duplication, but Chk1-dependent centrosome amplification also occurs after DNA damage. We investigated the involvement of cyclin-dependent kinases (Cdks) in DNA damage-induced centrosome amplification using cells lacking either Cdk2, or both Cdk1 and Cdk2 activity. Cdk2(-/-) DT40 cells showed robust centrosome amplification after ionizing radiation (IR), whereas Cdk1-deficient Cdk2(-/-) cells showed no centrosome amplification, demonstrating that Cdk1 can substitute for Cdk2 in this pathway. Surprisingly, we found that Cdk2 activity was upregulated by IR in wild-type but not in Chk1(-/-) DT40 cells. Cdk2 upregulation also occurred in HeLa cells after IR treatment. Chk1-dependent Cdk2 induction was not accompanied by increased levels of Cdk1, Cdk2, cyclin A or cyclin E, but activating T160 phosphorylation of Cdk2 increased after IR. Moreover, Cdk2 overexpression restored IR-induced centrosome amplification in Cdk1-deficient Cdk2(-/-) cells, but T160A mutation blocked this rescue. Our data suggest that Chk1 signalling causes centrosome amplification after IR by upregulating Cdk2 activity through activating phosphorylation.
Collapse
|
88
|
Saladino C, Bourke E, Conroy PC, Morrison CG. Centriole separation in DNA damage-induced centrosome amplification. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2009; 50:725-732. [PMID: 19274769 DOI: 10.1002/em.20477] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Altered centrosome numbers are seen in tumor cells in response to DNA damaging treatments and are hypothesised to contribute to cancer development. The mechanism by which the centrosome and chromosome cycles become disconnected after DNA damage is not yet clear. Here, we show that centrosome amplification occurs after ionising radiation (IR) in chicken DT40 cells that lack DNA-PK, Ku70, H2AX, Xpa, and Scc1, demonstrating that these activities are not required for centrosome amplification. We show that inhibition of topoisomerase II induces Chk1-dependent centrosome amplification, a similar response to that seen after IR. In the immortalised, nontransformed hTERT-RPE1 line, we observed centriole splitting, followed by dose-dependent centrosome amplification, after IR. We found that IR results in the formation of single, not multiple, daughter centrioles during centrosome amplification in U2OS osteosarcoma cells. Analysis of BRCA1 and BRCA2 mutant tumor cells showed high levels of centriole splitting in the absence of any treatment. IR caused pronounced levels of centrosome amplification in BRCA1 mutant breast cancer cells. These data show that centrosome amplification occurs after different forms of DNA damage in chicken cells, in nontransformed human cells and in human tumor cell lines, indicating that this is a general response to DNA damaging treatments. Together, our data suggest that centriole splitting is a key step in potentiation of the centrosome amplification that is a general response to DNA damage.
Collapse
Affiliation(s)
- Chiara Saladino
- Centre for Chromosome Biology, Department of Biochemistry and NCBES, National University of Ireland-Galway, University Road, Galway, Ireland
| | | | | | | |
Collapse
|
89
|
Chen S, Maya-Mendoza A, Zeng K, Tang CW, Sims PFG, Loric J, Jackson DA. Interaction with checkpoint kinase 1 modulates the recruitment of nucleophosmin to chromatin. J Proteome Res 2009; 8:4693-704. [PMID: 19694479 DOI: 10.1021/pr900396d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Checkpoint kinase 1 (Chk1) plays a central role in the cellular response to DNA damage and also contributes to the efficacy of DNA replication in the absence of genomic stress. However, we have only limited knowledge regarding the molecular mechanisms that regulate differential Chk1 function in the absence and presence of DNA damage. To address this, we used vertebrate cells with compromised Chk1 function to analyze how altered Chk1 activity influences protein interactions in chromatin. Avian and mammalian cells with compromised Chk1 activity were used in combination with genomic stress, induced by UV, and DNA-associated proteomes were analyzed using 2-DE/MS proteomics and Western-blot analysis. Only one protein, the histone chaperone nucelophosmin, was altered consistently in line with changes in chromatin-associated Chk1 and increased in response to DNA damage. Purified Chk1 and NPM were shown to interact in vitro and strong in vivo interactions were implied from immunoprecipitation analysis of chromatin extracts. During chromatin immunoprecipitation, coassociation of the major cell cycle regulator proteins p53 and CDC25A with both Chk1 and NPM suggests that these proteins are components of complex interaction networks that operate to regulate cell proliferation and apoptosis in vertebrate cells.
Collapse
Affiliation(s)
- Songbi Chen
- Manchester Interdisciplinary Biocentre, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
90
|
Affiliation(s)
- Mats Ljungman
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA.
| |
Collapse
|
91
|
Protein phosphatase 2A-dependent dephosphorylation of replication protein A is required for the repair of DNA breaks induced by replication stress. Mol Cell Biol 2009; 29:5696-709. [PMID: 19704001 DOI: 10.1128/mcb.00191-09] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Eukaryotic genomic integrity is safeguarded by cell cycle checkpoints and DNA repair pathways, collectively known as the DNA damage response, wherein replication protein A (RPA) is a key regulator playing multiple critical roles. The genotoxic insult-induced phosphorylation of the 32-kDa subunit of human RPA (RPA32), most notably the ATM/ATR-dependent phosphorylation at T21 and S33, acts to suppress DNA replication and recruit other checkpoint/repair proteins to the DNA lesions. It is not clear, however, how the DNA damage-responsive function of phosphorylated RPA is attenuated and how the replication-associated activity of the unphosphorylated form of RPA is restored when cells start to resume the normal cell cycle. We report here that in cells recovering from hydroxyurea (HU)-induced genotoxic stress, RPA32 is dephosphorylated by the serine/threonine protein phosphatase 2A (PP2A). Interference with PP2A catalytic activity causes persistent RPA32 phosphorylation and increased HU sensitivity. The PP2A catalytic subunit binds to RPA following DNA damage and can dephosphorylate RPA32 in vitro. Cells expressing a RPA32 persistent phosphorylation mimetic exhibit normal checkpoint activation and reenter the cell cycle normally after recovery but display a pronounced defect in the repair of DNA breaks. These data indicate that PP2A-mediated RPA32 dephosphorylation is required for the efficient DNA damage repair.
Collapse
|
92
|
Jin K, Ewton DZ, Park S, Hu J, Friedman E. Mirk regulates the exit of colon cancer cells from quiescence. J Biol Chem 2009; 284:22916-25. [PMID: 19542220 PMCID: PMC2755699 DOI: 10.1074/jbc.m109.035519] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 06/18/2009] [Indexed: 01/12/2023] Open
Abstract
Mirk/Dyrk1B is a serine/threonine kinase widely expressed in colon cancers. Serum starvation induced HD6 colon carcinoma cells to enter a quiescent G0 state, characterized by a 2N DNA content and a lower RNA content than G1 cells. Compared with cycling cells, quiescent cells exhibited 16-fold higher levels of the retinoblastoma protein p130/Rb2, which sequesters E2F4 to block entry into G1, 10-fold elevated levels of the CDK inhibitor p27kip1, and 10-fold higher levels of Mirk. However, depletion of Mirk did not prevent entry into G0, but enabled quiescent HD6, SW480, and colo320 colon carcinoma cells to acquire some biochemical characteristics of G1 cells, including increased levels of cyclin D1 and cyclin D3 because of slower turnover, increased activity of their CDK4/cyclin D complexes, and increased phosphorylation and decreased E2F4 sequestering ability of the CDK4 target, p130/Rb2. As a result, depletion of Mirk allowed some cells to escape quiescence and enabled cells released from quiescence to traverse G1 more quickly. The kinase activity of Mirk was increased by the chemotherapeutic drug 5-fluorouracil (5-FU). Treatment of p53 mutant colon cancer cells with 5-FU led to an elongated G1 in a Mirk-dependent manner, as G1 was shortened by ectopic overexpression of cyclin D1 mutated at the Mirk phosphorylation site (T288A), but not by wild-type cyclin D1. Mirk, through regulating cyclin D turnover, and the CDK inhibitor p27, as shown by depletion studies, functioned independently and additively to regulate the exit of tumor cells from quiescence.
Collapse
Affiliation(s)
- Kideok Jin
- From the Pathology Department, Upstate Medical University of the State University of New York, Syracuse, New York 13210
| | - Daina Z. Ewton
- From the Pathology Department, Upstate Medical University of the State University of New York, Syracuse, New York 13210
| | - Sunju Park
- From the Pathology Department, Upstate Medical University of the State University of New York, Syracuse, New York 13210
| | - Jing Hu
- From the Pathology Department, Upstate Medical University of the State University of New York, Syracuse, New York 13210
| | - Eileen Friedman
- From the Pathology Department, Upstate Medical University of the State University of New York, Syracuse, New York 13210
| |
Collapse
|
93
|
Tibelius A, Marhold J, Zentgraf H, Heilig CE, Neitzel H, Ducommun B, Rauch A, Ho AD, Bartek J, Krämer A. Microcephalin and pericentrin regulate mitotic entry via centrosome-associated Chk1. ACTA ACUST UNITED AC 2009; 185:1149-57. [PMID: 19546241 PMCID: PMC2712957 DOI: 10.1083/jcb.200810159] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary microcephaly, Seckel syndrome, and microcephalic osteodysplastic primordial dwarfism type II (MOPD II) are disorders exhibiting marked microcephaly, with small brain sizes reflecting reduced neuron production during fetal life. Although primary microcephaly can be caused by mutations in microcephalin (MCPH1), cells from patients with Seckel syndrome and MOPD II harbor mutations in ataxia telangiectasia and Rad3 related (ATR) or pericentrin (PCNT), leading to disturbed ATR signaling. In this study, we show that a lack of MCPH1 or PCNT results in a loss of Chk1 from centrosomes with subsequently deregulated activation of centrosomal cyclin B–Cdk1.
Collapse
Affiliation(s)
- Alexandra Tibelius
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Walker M, Black EJ, Oehler V, Gillespie DA, Scott MT. Chk1 C-terminal regulatory phosphorylation mediates checkpoint activation by de-repression of Chk1 catalytic activity. Oncogene 2009; 28:2314-23. [PMID: 19421147 PMCID: PMC2857325 DOI: 10.1038/onc.2009.102] [Citation(s) in RCA: 63] [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: 02/27/2009] [Revised: 03/23/2009] [Accepted: 03/24/2009] [Indexed: 01/19/2023]
Abstract
Chk1 is phosphorylated within its C-terminal regulatory domain by the upstream ATM/ATR kinases during checkpoint activation; however, how this modulates Chk1 function is poorly understood. Here, we show that Chk1 kinase activity is rapidly stimulated in a cell-cycle phase-specific manner in response to both DNA damage and replication arrest, and that the extent and duration of activation correlates closely with regulatory phosphorylation at serines (S) S317, S345 and S366. Despite their evident co-regulation, substitutions of individual Chk1 regulatory sites with alanine (A) residues have differential effects on checkpoint proficiency and kinase activation. Thus, whereas Chk1 S345 is essential for all functions tested, mutants lacking S317 or S366 retain partial proficiency for G2/M and S/M checkpoint arrests triggered by DNA damage or replication arrest. These phenotypes reflect defects in Chk1 kinase induction, as the mutants are either partially (317A and 366A) or completely (345A) resistant to kinase activation. Importantly, S345 phosphorylation is impaired in Chk1 S317A and S366A mutants, suggesting that modification of adjacent SQ sites promotes this key regulatory event. Finally, we provide biochemical evidence that Chk1 catalytic activity is stimulated by a de-repression mechanism.
Collapse
Affiliation(s)
- M Walker
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - EJ Black
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - V Oehler
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - DA Gillespie
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
- Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - MT Scott
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| |
Collapse
|
95
|
Peddibhotla S, Lam MH, Gonzalez-Rimbau M, Rosen JM. The DNA-damage effector checkpoint kinase 1 is essential for chromosome segregation and cytokinesis. Proc Natl Acad Sci U S A 2009; 106:5159-64. [PMID: 19289837 PMCID: PMC2663996 DOI: 10.1073/pnas.0806671106] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Indexed: 11/18/2022] Open
Abstract
Defective genome maintenance mechanisms, involving DNA repair and cell-cycle checkpoint pathways, initiate genetic instability in many sporadic and hereditary cancers. The DNA damage effector Checkpoint kinase 1 (Chk1) is a critical component of DNA replication, intra-S phase, and G(2)/M phase checkpoints and a recently reported mitotic spindle-assembly checkpoint. Here, we report for the first time that haploinsufficiency of Chk1 in mice resulted in multiple mitotic defects and enhanced binucleation. We observed that Aurora B, a critical cytokinetic regulator and a recently identified Chk1 substrate, was mislocalized in mitotic Chk1(+/-) mammary epithelia. Chk1 also exhibited distinct mitotic localization patterns and was active during unperturbed mitosis and cytokinesis in mammalian cells. Active Chk1 expression was not dependent on treatment with spindle poisons such as colcemid during mitosis and cytokinesis. Furthermore, two different complementary approaches demonstrated that abrogation of Chk1 in mitotic mammalian cells resulted in cytokinetic regression and binucleation, increased chromosome lagging and/or nondisjunction, and abnormal localization of Aurora B at late mitotic structures. Thus, Chk1 is a multifunctional kinase that serves as a nexus between the DNA damage response and the mitotic exit pathways during cell-cycle progression to prevent genomic instability and cancer.
Collapse
Affiliation(s)
- Sirisha Peddibhotla
- Interdepartmental Program in Cell and Molecular Biology and
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Michael H. Lam
- Interdepartmental Program in Cell and Molecular Biology and
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Maria Gonzalez-Rimbau
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Jeffrey M. Rosen
- Interdepartmental Program in Cell and Molecular Biology and
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
96
|
Greenow KR, Clarke AR, Jones RH. Chk1 deficiency in the mouse small intestine results in p53-independent crypt death and subsequent intestinal compensation. Oncogene 2009; 28:1443-53. [PMID: 19169280 PMCID: PMC2659326 DOI: 10.1038/onc.2008.482] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 11/19/2008] [Accepted: 12/11/2008] [Indexed: 01/21/2023]
Abstract
Chk1 is a serine/threonine protein kinase that is activated by a wide range of DNA-damaging agents to slow the cell cycle during S phase and G2/M. Abrogation of these cell-cycle checkpoints using Chk1 inhibitors results in hypersensitivity to DNA-damaging agents in vitro and may provide a potential therapeutic tool to sensitize tumour cells in vivo. We have generated a Cre-Lox-based mouse model in which Chkl can be inducibly deleted from somatic epithelial cells in the adult mouse small intestine and liver. Loss of Chk1 in the liver is tolerated with no apparent phenotype. In contrast, the loss of Chk1 within the small intestine results in immediate DNA damage and high levels of p53-independent apoptosis leading to crypt death. However, the intestine is able to compensate for this death by undergoing complete re-population with Chk1-proficient cells. These data therefore show that Chk1 deficiency is cell lethal, but the intestine can tolerate such lethality at the organ level.
Collapse
Affiliation(s)
- Kirsty R. Greenow
- Cardiff School of Biosciences, Cardiff University, Cardiff, CF10 3US
| | - Alan R. Clarke
- Cardiff School of Biosciences, Cardiff University, Cardiff, CF10 3US
| | - Robert H. Jones
- Cardiff School of Biosciences, Cardiff University, Cardiff, CF10 3US
- Dept Cellular and Molecular Medicine, Bristol University, Bristol, BS8 1TD
| |
Collapse
|
97
|
Chanoux RA, Yin B, Urtishak KA, Asare A, Bassing CH, Brown EJ. ATR and H2AX cooperate in maintaining genome stability under replication stress. J Biol Chem 2009; 284:5994-6003. [PMID: 19049966 PMCID: PMC2645842 DOI: 10.1074/jbc.m806739200] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 12/02/2008] [Indexed: 11/06/2022] Open
Abstract
Chromosomal abnormalities are frequently caused by problems encountered during DNA replication. Although the ATR-Chk1 pathway has previously been implicated in preventing the collapse of stalled replication forks into double-strand breaks (DSB), the importance of the response to fork collapse in ATR-deficient cells has not been well characterized. Herein, we demonstrate that, upon stalled replication, ATR deficiency leads to the phosphorylation of H2AX by ATM and DNA-PKcs and to the focal accumulation of Rad51, a marker of homologous recombination and fork restart. Because H2AX has been shown to play a facilitative role in homologous recombination, we hypothesized that H2AX participates in Rad51-mediated suppression of DSBs generated in the absence of ATR. Consistent with this model, increased Rad51 focal accumulation in ATR-deficient cells is largely dependent on H2AX, and dual deficiencies in ATR and H2AX lead to synergistic increases in chromatid breaks and translocations. Importantly, the ATM and DNA-PK phosphorylation site on H2AX (Ser(139)) is required for genome stabilization in the absence of ATR; therefore, phosphorylation of H2AX by ATM and DNA-PKcs plays a pivotal role in suppressing DSBs during DNA synthesis in instances of ATR pathway failure. These results imply that ATR-dependent fork stabilization and H2AX/ATM/DNA-PKcs-dependent restart pathways cooperatively suppress double-strand breaks as a layered response network when replication stalls.
Collapse
MESH Headings
- Animals
- Ataxia Telangiectasia Mutated Proteins
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/metabolism
- Cell Cycle Proteins/physiology
- Cells, Cultured
- DNA Breaks, Double-Stranded
- DNA Replication
- DNA-Activated Protein Kinase/metabolism
- DNA-Binding Proteins/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Embryo, Mammalian/radiation effects
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Fibroblasts/radiation effects
- Genomic Instability
- Histones/physiology
- Metaphase
- Mice
- Mice, Knockout
- Mitosis
- Nuclear Proteins/metabolism
- Phosphorylation
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Rad51 Recombinase/metabolism
- Radiation, Ionizing
- Reverse Transcriptase Polymerase Chain Reaction
- S Phase/physiology
- Spectral Karyotyping
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Rebecca A Chanoux
- Abramson Family Cancer Research Institute and Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
98
|
Smith E, Dejsuphong D, Balestrini A, Hampel M, Lenz C, Takeda S, Vindigni A, Costanzo V. An ATM- and ATR-dependent checkpoint inactivates spindle assembly by targeting CEP63. Nat Cell Biol 2009; 11:278-85. [DOI: 10.1038/ncb1835] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Accepted: 11/21/2008] [Indexed: 12/22/2022]
|
99
|
Cell-cycle control in the face of damage--a matter of life or death. Trends Cell Biol 2009; 19:89-98. [PMID: 19168356 DOI: 10.1016/j.tcb.2008.12.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 12/09/2008] [Accepted: 12/09/2008] [Indexed: 12/31/2022]
Abstract
Cells respond to DNA damage or defects in the mitotic spindle by activating checkpoints that arrest the cell cycle. Alternatively, damaged cells can undergo cell death by the process of apoptosis. The correct balance between these pathways is important for the maintenance of genomic integrity while preventing unnecessary cell death. Although the molecular mechanisms of the cell cycle and apoptosis have been elucidated, the links between them have not been clear. Recent work, however, indicates that common components directly link the regulation of apoptosis with cell-cycle checkpoints operating during interphase, whereas in mitosis, the control of apoptosis is directly coupled to the cell-cycle machinery. These findings shed new light on how the balance between cell-cycle progression and cell death is controlled.
Collapse
|
100
|
Myers K, Gagou ME, Zuazua-Villar P, Rodriguez R, Meuth M. ATR and Chk1 suppress a caspase-3-dependent apoptotic response following DNA replication stress. PLoS Genet 2009; 5:e1000324. [PMID: 19119425 PMCID: PMC2607051 DOI: 10.1371/journal.pgen.1000324] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 12/02/2008] [Indexed: 01/30/2023] Open
Abstract
The related PIK-like kinases Ataxia-Telangiectasia Mutated (ATM) and ATM- and Rad3-related (ATR) play major roles in the regulation of cellular responses to DNA damage or replication stress. The pro-apoptotic role of ATM and p53 in response to ionizing radiation (IR) has been widely investigated. Much less is known about the control of apoptosis following DNA replication stress. Recent work indicates that Chk1, the downstream phosphorylation target of ATR, protects cells from apoptosis induced by DNA replication inhibitors as well as IR. The aim of the work reported here was to determine the roles of ATM- and ATR-protein kinase cascades in the control of apoptosis following replication stress and the relationship between Chk1-suppressed apoptotic pathways responding to replication stress or IR. ATM and ATR/Chk1 signalling pathways were manipulated using siRNA-mediated depletions or specific inhibitors in two tumour cell lines or fibroblasts derived from patients with inherited mutations. We show that depletion of ATM or its downstream phosphorylation targets, NBS1 and BID, has relatively little effect on apoptosis induced by DNA replication inhibitors, while ATR or Chk1 depletion strongly enhances cell death induced by such agents in all cells tested. Furthermore, early events occurring after the disruption of DNA replication (accumulation of RPA foci and RPA34 hyperphosphorylation) in ATR- or Chk1-depleted cells committed to apoptosis are not detected in ATM-depleted cells. Unlike the Chk1-suppressed pathway responding to IR, the replication stress-triggered apoptotic pathway did not require ATM and is characterized by activation of caspase 3 in both p53-proficient and -deficient cells. Taken together, our results show that the ATR-Chk1 signalling pathway plays a major role in the regulation of death in response to DNA replication stress and that the Chk1-suppressed pathway protecting cells from replication stress is clearly distinguishable from that protecting cells from IR. The integrity of the genetic information in cells is protected by elaborate mechanisms that ensure that an accurate DNA copy is passed from generation to generation. These mechanisms repair errors in DNA sequence or stop growth if DNA structure is compromised. However, if the level of DNA damage is too severe, cells may also respond by inducing death rather than attempt repair. Relatively little is known about how cells decide whether to repair damage or commit to death. The purpose of our work was to identify genes that control this decision-making process while cells are duplicating DNA. We show that two genes play a major role in this process; however, our work also suggests considerable complexity in this death response as different death pathways are triggered in response to different forms of DNA damage. Since DNA replication inhibitors are used widely in the treatment of cancer, our work may enable us to more effectively kill cancer cells in treatment protocols employing these agents.
Collapse
Affiliation(s)
- Katie Myers
- Institute for Cancer Studies, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Mary E. Gagou
- Institute for Cancer Studies, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Pedro Zuazua-Villar
- Institute for Cancer Studies, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Rene Rodriguez
- Institute for Cancer Studies, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Mark Meuth
- Institute for Cancer Studies, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|