1
|
Saeed N, Mahjabeen I, Hakim F, Hussain MZ, Mehmood A, Nisar A, Ahmed MW, Kayani MA. Role of Chk1 gene in molecular classification and prognosis of gastric cancer using immunohistochemistry and LORD-Q assay. Future Oncol 2022; 18:2827-2841. [PMID: 35762179 DOI: 10.2217/fon-2021-1546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Purpose: The aim of the current study was to assess the prognostic value of the Chk1 gene in the DNA damage response pathway in gastric cancer (GC). Methods: Expression levels of the Chk1 were measured in 220 GC tumor tissues and adjacent healthy/noncancerous tissues using real-time PCR and immunohistochemical staining. Genomic instability in GC patients was measured using the long-run real-time PCR technique for DNA-damage quantification assay and comet assay. Results: Significantly downregulated expression of Chk1 was observed at the mRNA level (p < 0.0001) and protein level (p < 0.0001). Significantly increased frequency of lesions/10 kb and comets was observed in tumor tissues compared with control tissues. Conclusion: The data suggest that downregulated expression of Chk1 and positive Heliobacter pylori infection status may have prognostic significance in GC.
Collapse
Affiliation(s)
- Nadia Saeed
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Ishrat Mahjabeen
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Farzana Hakim
- Department of Biochemistry, Foundation University Medical College, Islamabad, Pakistan
| | | | - Azhar Mehmood
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Asif Nisar
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Malik Waqar Ahmed
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan.,Pakistan Institute of Rehabilitation Sciences (PIRS), Isra University Islamabad Campus, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| |
Collapse
|
2
|
Abstract
Understanding the mechanisms of embryonic cell cycles is a central goal of developmental biology, as the regulation of the cell cycle must be closely coordinated with other events during early embryogenesis. Quantitative imaging approaches have recently begun to reveal how the cell cycle oscillator is controlled in space and time, and how it is integrated with mechanical signals to drive morphogenesis. Here, we discuss how the Drosophila embryo has served as an excellent model for addressing the molecular and physical mechanisms of embryonic cell cycles, with comparisons to other model systems to highlight conserved and species-specific mechanisms. We describe how the rapid cleavage divisions characteristic of most metazoan embryos require chemical waves and cytoplasmic flows to coordinate morphogenesis across the large expanse of the embryo. We also outline how, in the late cleavage divisions, the cell cycle is inter-regulated with the activation of gene expression to ensure a reliable maternal-to-zygotic transition. Finally, we discuss how precise transcriptional regulation of the timing of mitosis ensures that tissue morphogenesis and cell proliferation are tightly controlled during gastrulation.
Collapse
Affiliation(s)
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27705, USA
| |
Collapse
|
3
|
Ditano JP, Eastman A. Comparative Activity and Off-Target Effects in Cells of the CHK1 Inhibitors MK-8776, SRA737, and LY2606368. ACS Pharmacol Transl Sci 2021; 4:730-743. [PMID: 33860197 DOI: 10.1021/acsptsci.0c00201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 01/22/2023]
Abstract
DNA damage activates the checkpoint protein CHK1 to arrest cell cycle progression, providing time for repair and recovery. Consequently, inhibitors of CHK1 (CHK1i) enhance damage-induced cell death. Additionally, CHK1i elicits single agent cytotoxicity in some cell lines. We compared three CHK1i that have undergone clinical trials and exhibited different toxicities. Each CHK1i inhibits other targets at higher concentrations, and whether these contribute to the toxicity is unknown. We compared their sensitivity in a panel of cell lines, their efficacy at inhibiting CHK1 and CHK2, and their ability to induce DNA damage and abrogate damage-induced S phase arrest. Published in vitro kinase analyses were a poor predictor of selectivity and potency in cells. LY2606368 was far more potent at inhibiting CHK1 and inducing growth arrest, while all three CHK1i inhibited CHK2 at concentrations 10- (MK-8776 and SRA737) to 100- (LY2606368) fold higher. MK-8776 and SRA737 exhibited similar off-target effects: higher concentrations demonstrated transient protection from growth inhibition, circumvented DNA damage, and prevented checkpoint abrogation, possibly due to inhibition of CDK2. Acquired resistance to LY2606368 resulted in limited cross-resistance to other CHK1i. LY2606368-resistant cells still abrogated DNA damage-induced S phase arrest, which requires low CDK2 activity, whereas inappropriately high CDK2 activity is responsible for sensitivity to CHK1i alone. All three CHK1i inhibited protein synthesis in a sensitive cell line correlating with cell death, whereas resistant cells failed to inhibit protein synthesis and underwent transient cytostasis. LY2606368 appears to be the most selective CHK1i, suggesting that further clinical development of this drug is warranted.
Collapse
Affiliation(s)
- Jennifer P Ditano
- Department of Molecular and Systems Biology and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, United States
| | - Alan Eastman
- Department of Molecular and Systems Biology and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, United States
| |
Collapse
|
4
|
Fadaka AO, Bakare OO, Sibuyi NRS, Klein A. Gene Expression Alterations and Molecular Analysis of CHEK1 in Solid Tumors. Cancers (Basel) 2020; 12:cancers12030662. [PMID: 32178478 PMCID: PMC7139733 DOI: 10.3390/cancers12030662] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/16/2022] Open
Abstract
Alterations in the Checkpoint kinase (CHEK1) gene, its regulation, and the possible clinical outcomes in human solid tumors have not been previously examined. Therefore, the present study was carried out to evaluate the expression of CHEK1 in solid tumors as well as the mechanism by which it can be regulated through non-coding RNAs. The expression of CHEK1 was investigated using Oncomine analysis. cBioPortal, Kaplan-Meier Plotter, and PrognoScan were performed to identify the prognostic roles of this gene in solid tumors. The copy number alteration, mutation, interactive analysis, and visualization of the altered networks were performed by cBioPortal. The molecular binding analysis was carried out by Schrodinger suite, PATCHDOCK, and discovery studio visualizer. The study demonstrated that the CHEK1 gene was differentially expressed in four different cancers, and that reduced CHEK1 mRNA expression is an unfavorable prognostic factor for patients with gastric and colorectal cancer. The molecular docking results showed that the CHEK1 gene can be regulated by microRNAs (miR-195-5p) due to the number of stable hydrogen atoms observed within the distance of 2.0 Å and the favorable amino acids (Ala221, Ile353, Ile365, Ile756, Val797, Val70, Val154, Ile159, Val347, Tyr804, Phe811, Tyr815, and Phe156) identified in the binding pocket of the argonaute protein. Due to the possibility of CHEK1's involvement in solid tumors, it may potentially be a target for therapeutic intervention in cancer. Further studies into the interaction between CHEK1 and other co-expressed genes may give further insight into other modes of regulation of this gene in cancer patients.
Collapse
Affiliation(s)
- Adewale Oluwaseun Fadaka
- Bioinformatics research group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
- Correspondence: ; Tel.: +27-630511928 or +234-8039242052
| | - Olalekan Olanrewaju Bakare
- Bioinformatics research group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa
| | - Ashwil Klein
- Plant Omics group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| |
Collapse
|
5
|
Entrenching role of cell cycle checkpoints and autophagy for maintenance of genomic integrity. DNA Repair (Amst) 2019; 86:102748. [PMID: 31790874 DOI: 10.1016/j.dnarep.2019.102748] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/19/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022]
Abstract
Genomic integrity of the cell is crucial for the successful transmission of genetic information to the offspring and its survival. Persistent DNA damage induced by endogenous and exogenous agents leads to various metabolic manifestations. To combat this, eukaryotes have developed complex DNA damage response (DDR) pathway which senses the DNA damage and activates an arsenal of enzymes for the repair of damaged DNA. The active pathways for DNA repair are nucleotide excision repair (NER), base excision repair (BER) and mismatch repair (MMR) for single-strand break repair whereas homologous recombination (HR) and non-homologous end-joining (NHEJ) for double-strand break repair. OGG1 is a DNA glycosylase which initiates BER while Mre11-Rad50-Nbs1 (MRN) protein complex is the primary responder to DSBs which gets localized to damage sites. DNA damage response is meticulously executed by three related kinases: ATM, ATR, and DNA-PK. ATM- and ATR-dependent phosphorylation of p53, Chk1, and Chk2 regulate the G1/S, intra-S, or G2/M checkpoints of the cell cycle, respectively. Autophagy is an evolutionarily conserved process that plays a pivotal role in the regulation of DNA repair and maintains the cellular homeostasis. Genotoxic stress-induced altered autophagy occurs in a P53 dependent manner which is also the master regulator of genotoxic stress. A plethora of proteins involved in autophagy is regulated by p53 which involve DRAM, DAPK, and AMPK. As evident, the mtDNA is more prone to damage than nuclear DNA because of its close proximity to the site of ROS generation. Depending on the extent of damage either the repair mechanism or mitophagy gets triggered. SIRT1 is the master regulator which directs the stress response to mitophagy. Nix, a LC3 adapter also participates in Parkin mediated mitophagy. This review highlights the intricate crosstalks between DNA damage and cell cycle checkpoints activation. The DNA damage mediated regulation of autophagy and mitophagy is also reviewed in detail.
Collapse
|
6
|
van Harten AM, Buijze M, van der Mast R, Rooimans MA, Martens-de Kemp SR, Bachas C, Brink A, Stigter-van Walsum M, Wolthuis RMF, Brakenhoff RH. Targeting the cell cycle in head and neck cancer by Chk1 inhibition: a novel concept of bimodal cell death. Oncogenesis 2019; 8:38. [PMID: 31209198 PMCID: PMC6572811 DOI: 10.1038/s41389-019-0147-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/19/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) coincide with poor survival rates. The lack of driver oncogenes complicates the development of targeted treatments for HNSCC. Here, we follow-up on two previous genome-wide RNA and microRNA interference screens in HNSCC to cross-examine tumor-specific lethality by targeting ATM, ATR, CHEK1, or CHEK2. Our results uncover CHEK1 as the most promising target for HNSCC. CHEK1 expression is essential across a panel of HNSCC cell lines but redundant for growth and survival of untransformed oral keratinocytes and fibroblasts. LY2603618 (Rabusertib), which specifically targets Chk1 kinase, kills HNSCC cells effectively and specifically. Our findings show that HNSCC cells depend on Chk1-mediated signaling to progress through S-phase successfully. Chk1 inhibition coincides with stalled DNA replication, replication fork collapses, and accumulation of DNA damage. We further show that Chk1 inhibition leads to bimodal HNSCC cell killing. In the most sensitive cell lines, apoptosis is induced in S-phase, whereas more resistant cell lines manage to bypass replication-associated apoptosis, but accumulate chromosomal breaks that become lethal in subsequent mitosis. Interestingly, CDK1 expression correlates with treatment outcome. Moreover, sensitivity to Chk1 inhibition requires functional CDK1 and CDK4/6 to drive cell cycle progression, arguing against combining Chk1 inhibitors with CDK inhibitors. In contrast, Wee1 inhibitor Adavosertib progresses the cell cycle and thereby increases lethality to Chk1 inhibition in HNSCC cell lines. We conclude that Chk1 has become a key molecule in HNSCC cell cycle regulation and a very promising therapeutic target. Chk1 inhibition leads to S-phase apoptosis or death in mitosis. We provide a potential efficacy biomarker and combination therapy to follow-up in clinical setting.
Collapse
Affiliation(s)
- Anne M van Harten
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marijke Buijze
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Richard van der Mast
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martin A Rooimans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Clinical Genetics, Section Oncogenetics, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sanne R Martens-de Kemp
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Costa Bachas
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Arjen Brink
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marijke Stigter-van Walsum
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Rob M F Wolthuis
- Amsterdam UMC, Vrije Universiteit Amsterdam, Clinical Genetics, Section Oncogenetics, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Section Tumor Biology, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Wang Z, Førsund MS, Trope CG, Nesland JM, Holm R, Slipicevic A. Evaluation of CHK1 activation in vulvar squamous cell carcinoma and its potential as a therapeutic target in vitro. Cancer Med 2018; 7:3955-3964. [PMID: 29963769 PMCID: PMC6089182 DOI: 10.1002/cam4.1638] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 11/05/2022] Open
Abstract
CHK1 is an important regulator of the cell cycle and DNA damage response, and its altered expression has been identified in various tumors. Chk1 inhibitors are currently being evaluated as monotherapy and as potentiators of chemotherapy in clinical settings. However, to our knowledge, no previous study has investigated either the activation status or the therapeutic potential of CHK1 targeting in vulvar cancer. Therefore, we examined the expression status of activated CHK1 forms pCHK1Ser345, pCHK1Ser317, pCHK1Ser296, and pCHK1Ser280 in 294 vulvar squamous cell carcinomas (VSCC) using immunohistochemistry and analyzed their relationships with various clinicopathological variables and clinical outcome. To aid translation of preclinical studies, we also assessed cell sensitivity to the Chk1 inhibition in two vulvar cancer cell lines. Compared to the levels of pCHK1Ser345, pCHK1Ser317, pCHK1Ser296, and pCHK1Ser280 in normal vulvar squamous epithelium, high nuclear pCHK1Ser345 expression was found in 57% of vulvar carcinomas, whereas low nuclear pCHK1Ser317, pCHK1Ser296, and pCHK1Ser280 expressions were observed in 58%, 64%, and 40% of the cases, respectively. Low levels of pCHK1Ser317 and pCHK1Ser280 in the nucleus correlated significantly with advanced tumor behaviors and aggressive features. None of pCHK1Ser345, pCHK1Ser317, pCHK1Ser296, and pCHK1Ser280 forms were identified as prognostic factors. In vitro inhibition of CHK1 by small molecular inhibitors or siRNA reduced viability by inducing DNA damage and apoptosis of vulvar cancer cell lines. In summary, we conclude that cellular functions regulated by CHK1 are phosphorylation/localization‐dependent and deregulation of CHK1 function occurs in VSCC and might contribute to tumorigenesis. Targeting CHK1 might represent as a useful antitumor strategy for the subgroup of VSCC harboring p53 mutations.
Collapse
Affiliation(s)
- Zhihui Wang
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Mette S Førsund
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Claes G Trope
- Department of Obstetrics and Gynecology, The Norwegian Radium Hospital, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Jahn M Nesland
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ruth Holm
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ana Slipicevic
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
8
|
Göder A, Emmerich C, Nikolova T, Kiweler N, Schreiber M, Kühl T, Imhof D, Christmann M, Heinzel T, Schneider G, Krämer OH. HDAC1 and HDAC2 integrate checkpoint kinase phosphorylation and cell fate through the phosphatase-2A subunit PR130. Nat Commun 2018; 9:764. [PMID: 29472538 PMCID: PMC5823910 DOI: 10.1038/s41467-018-03096-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 01/19/2018] [Indexed: 12/18/2022] Open
Abstract
Checkpoint kinases sense replicative stress to prevent DNA damage. Here we show that the histone deacetylases HDAC1/HDAC2 sustain the phosphorylation of the checkpoint kinases ATM, CHK1 and CHK2, activity of the cell cycle gatekeeper kinases WEE1 and CDK1, and induction of the tumour suppressor p53 in response to stalled DNA replication. Consequently, HDAC inhibition upon replicative stress promotes mitotic catastrophe. Mechanistically, HDAC1 and HDAC2 suppress the expression of PPP2R3A/PR130, a regulatory subunit of the trimeric serine/threonine phosphatase 2 (PP2A). Genetic elimination of PR130 reveals that PR130 promotes dephosphorylation of ATM by PP2A. Moreover, the ablation of PR130 slows G1/S phase transition and increases the levels of phosphorylated CHK1, replication protein A foci and DNA damage upon replicative stress. Accordingly, stressed PR130 null cells are very susceptible to HDAC inhibition, which abrogates the S phase checkpoint, induces apoptosis and reduces the homologous recombination protein RAD51. Thus, PR130 controls cell fate decisions upon replicative stress. Checkpoint kinases control cell cycle progression via the regulation of many key regulators. Here the authors demonstrate how HDAC1 and HDAC2 modulate checkpoint kinase signalling via the suppression of PR130, a regulatory subunit of the trimeric serine/threonine phosphatase 2.
Collapse
Affiliation(s)
- Anja Göder
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Claudia Emmerich
- University of Jena, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Hans-Knöll-Strasse 2, 07745, Jena, Germany
| | - Teodora Nikolova
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Nicole Kiweler
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Maria Schreiber
- University of Jena, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Hans-Knöll-Strasse 2, 07745, Jena, Germany
| | - Toni Kühl
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Markus Christmann
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Thorsten Heinzel
- University of Jena, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Hans-Knöll-Strasse 2, 07745, Jena, Germany
| | - Günter Schneider
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Ismaningerstrasse 22, 81675, Munich, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany.
| |
Collapse
|
9
|
Cdk2 strengthens the intra-S checkpoint and counteracts cell cycle exit induced by DNA damage. Sci Rep 2017; 7:13429. [PMID: 29044141 PMCID: PMC5647392 DOI: 10.1038/s41598-017-12868-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/17/2017] [Indexed: 02/03/2023] Open
Abstract
Although cyclin-dependent kinase 2 (Cdk2) controls the G1/S transition and promotes DNA replication, it is dispensable for cell cycle progression due to redundancy with Cdk1. Yet Cdk2 also has non-redundant functions that can be revealed in certain genetic backgrounds and it was reported to promote the G2/M DNA damage response checkpoint in TP53 (p53)-deficient cancer cells. However, in p53-proficient cells subjected to DNA damage, Cdk2 is inactivated by the CDK inhibitor p21. We therefore investigated whether Cdk2 differentially affects checkpoint responses in p53-proficient and deficient cell lines. We show that, independently of p53 status, Cdk2 stimulates the ATR/Chk1 pathway and is required for an efficient DNA replication checkpoint response. In contrast, Cdk2 is not required for a sustained DNA damage response and G2 arrest. Rather, eliminating Cdk2 delays S/G2 progression after DNA damage and accelerates appearance of early markers of cell cycle exit. Notably, Cdk2 knockdown leads to down-regulation of Cdk6, which we show is a non-redundant pRb kinase whose elimination compromises cell cycle progression. Our data reinforce the notion that Cdk2 is a key p21 target in the DNA damage response whose inactivation promotes exit from the cell cycle in G2.
Collapse
|
10
|
Liberio MS, Sadowski MC, Davis RA, Rockstroh A, Vasireddy R, Lehman ML, Nelson CC. The ascidian natural product eusynstyelamide B is a novel topoisomerase II poison that induces DNA damage and growth arrest in prostate and breast cancer cells. Oncotarget 2016; 6:43944-63. [PMID: 26733491 PMCID: PMC4791278 DOI: 10.18632/oncotarget.6267] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 10/08/2015] [Indexed: 12/25/2022] Open
Abstract
As part of an anti-cancer natural product drug discovery program, we recently identified eusynstyelamide B (EB), which displayed cytotoxicity against MDA-MB-231 breast cancer cells (IC50 = 5 μM) and induced apoptosis. Here, we investigated the mechanism of action of EB in cancer cell lines of the prostate (LNCaP) and breast (MDA-MB-231). EB inhibited cell growth (IC50 = 5 μM) and induced a G2 cell cycle arrest, as shown by a significant increase in the G2/M cell population in the absence of elevated levels of the mitotic marker phospho-histone H3. In contrast to MDA-MB-231 cells, EB did not induce cell death in LNCaP cells when treated for up to 10 days. Transcript profiling and Ingenuity Pathway Analysis suggested that EB activated DNA damage pathways in LNCaP cells. Consistent with this, CHK2 phosphorylation was increased, p21CIP1/WAF1 was up-regulated and CDC2 expression strongly reduced by EB. Importantly, EB caused DNA double-strand breaks, yet did not directly interact with DNA. Analysis of topoisomerase II-mediated decatenation discovered that EB is a novel topoisomerase II poison.
Collapse
Affiliation(s)
- Michelle S Liberio
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia.,Eskitis Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Martin C Sadowski
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Rohan A Davis
- Eskitis Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Raj Vasireddy
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Melanie L Lehman
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
CHK1 expression in Gastric Cancer is modulated by p53 and RB1/E2F1: implications in chemo/radiotherapy response. Sci Rep 2016; 6:21519. [PMID: 26867682 PMCID: PMC4751465 DOI: 10.1038/srep21519] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/26/2016] [Indexed: 12/19/2022] Open
Abstract
Radiation has a limited but relevant role in the adjuvant therapy of gastric cancer (GC) patients. Since Chk1 plays a critical function in cellular response to genotoxic agents, we aimed to analyze the role of Chk1 in GC as a biomarker for radiotherapy resistance. We analyzed Chk1 expression in AGS and MKN45 human GC cell lines by RT-QPCR and WB and in a small cohort of human patient’s samples. We demonstrated that Chk1 overexpression specifically increases resistance to radiation in GC cells. Accordingly, abrogation of Chk1 activity with UCN-01 and its expression with shChk1 increased sensitivity to bleomycin and radiation. Furthermore, when we assessed Chk1 expression in human samples, we found a correlation between nuclear Chk1 accumulation and a decrease in progression free survival. Moreover, using a luciferase assay we found that Chk1’s expression is controlled by p53 and RB/E2F1 at the transcriptional level. Additionally, we present preliminary data suggesting a posttranscriptional regulation mechanism, involving miR-195 and miR-503, which are inversely correlated with expression of Chk1 in radioresistant cells. In conclusion, Chk1/microRNA axis is involved in resistance to radiation in GC, and suggests Chk1 as a potential tool for optimal stratification of patients susceptible to receive adjuvant radiotherapy after surgery.
Collapse
|
12
|
DNA Damage Signalling and Repair Inhibitors: The Long-Sought-After Achilles' Heel of Cancer. Biomolecules 2015; 5:3204-59. [PMID: 26610585 PMCID: PMC4693276 DOI: 10.3390/biom5043204] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
For decades, radiotherapy and chemotherapy were the two only approaches exploiting DNA repair processes to fight against cancer. Nowadays, cancer therapeutics can be a major challenge when it comes to seeking personalized targeted medicine that is both effective and selective to the malignancy. Over the last decade, the discovery of new targeted therapies against DNA damage signalling and repair has offered the possibility of therapeutic improvements in oncology. In this review, we summarize the current knowledge of DNA damage signalling and repair inhibitors, their molecular and cellular effects, and future therapeutic use.
Collapse
|
13
|
Smits VAJ, Gillespie DA. DNA damage control: regulation and functions of checkpoint kinase 1. FEBS J 2015. [DOI: 10.1111/febs.13387] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Veronique A. J. Smits
- Unidad de Investigación; Hospital Universitario de Canarias; Instituto de Tecnologías Biomédicas; Tenerife Spain
| | - David A. Gillespie
- Instituto de Tecnologías Biomédicas; Centro de Investigaciones Biomédicas de Canarias; Facultad de Medicina; Campus Ciencias de la Salud; Universidad de La Laguna; Tenerife Spain
| |
Collapse
|
14
|
Checkpoint kinase1 (CHK1) is an important biomarker in breast cancer having a role in chemotherapy response. Br J Cancer 2015; 112:901-11. [PMID: 25688741 PMCID: PMC4453942 DOI: 10.1038/bjc.2014.576] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 09/18/2014] [Accepted: 09/23/2014] [Indexed: 01/16/2023] Open
Abstract
Background: Checkpoint kinase1 (CHK1), which is a key component of DNA-damage-activated checkpoint signalling response, may have a role in breast cancer (BC) pathogenesis and influence response to chemotherapy. This study investigated the clinicopathological significance of phosphorylated CHK1 (pCHK1) protein in BC. Method: pCHK1 protein expression was assessed using immunohistochemistry in a large, well-characterized annotated series of early-stage primary operable invasive BC prepared as tissue microarray (n=1200). Result: pCHK1 showed nuclear and/or cytoplasmic expression. Tumours with nuclear expression showed positive associations with favourable prognostic features such as lower grade, lower mitotic activity, expression of hormone receptor and lack of expression of KI67 and PI3K (P<0.001). On the other hand, cytoplasmic expression was associated with features of poor prognosis such as higher grade, triple-negative phenotype and expression of KI67, p53, AKT and PI3K. pCHK1 expression showed an association with DNA damage response (ATM, RAD51, BRCA1, KU70/KU80, DNA-PKCα and BARD1) and sumoylation (UBC9 and PIASγ) biomarkers. Subcellular localisation of pCHK1 was associated with the expression of the nuclear transport protein KPNA2. Positive nuclear expression predicted better survival outcome in patients who did not receive chemotherapy in the whole series and in ER-positive tumours. In ER-negative and triple-negative subgroups, nuclear pCHK1 predicted shorter survival in patients who received cyclophosphamide, methotrexate and 5-florouracil chemotherapy. Conclusions: Our data suggest that pCHK1 may have prognostic and predictive significance in BC. Subcellular localisation of pCHK1 protein is related to its function.
Collapse
|
15
|
Shaltiel IA, Krenning L, Bruinsma W, Medema RH. The same, only different - DNA damage checkpoints and their reversal throughout the cell cycle. J Cell Sci 2015; 128:607-20. [PMID: 25609713 DOI: 10.1242/jcs.163766] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell cycle checkpoints activated by DNA double-strand breaks (DSBs) are essential for the maintenance of the genomic integrity of proliferating cells. Following DNA damage, cells must detect the break and either transiently block cell cycle progression, to allow time for repair, or exit the cell cycle. Reversal of a DNA-damage-induced checkpoint not only requires the repair of these lesions, but a cell must also prevent permanent exit from the cell cycle and actively terminate checkpoint signalling to allow cell cycle progression to resume. It is becoming increasingly clear that despite the shared mechanisms of DNA damage detection throughout the cell cycle, the checkpoint and its reversal are precisely tuned to each cell cycle phase. Furthermore, recent findings challenge the dogmatic view that complete repair is a precondition for cell cycle resumption. In this Commentary, we highlight cell-cycle-dependent differences in checkpoint signalling and recovery after a DNA DSB, and summarise the molecular mechanisms that underlie the reversal of DNA damage checkpoints, before discussing when and how cell fate decisions after a DSB are made.
Collapse
Affiliation(s)
- Indra A Shaltiel
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Lenno Krenning
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Wytse Bruinsma
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - René H Medema
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
16
|
Daud AI, Ashworth MT, Strosberg J, Goldman JW, Mendelson D, Springett G, Venook AP, Loechner S, Rosen LS, Shanahan F, Parry D, Shumway S, Grabowsky JA, Freshwater T, Sorge C, Kang SP, Isaacs R, Munster PN. Phase I dose-escalation trial of checkpoint kinase 1 inhibitor MK-8776 as monotherapy and in combination with gemcitabine in patients with advanced solid tumors. J Clin Oncol 2015; 33:1060-6. [PMID: 25605849 DOI: 10.1200/jco.2014.57.5027] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE We determined the safety, pharmacokinetics, pharmacodynamics, and recommended phase II dose of MK-8776 (SCH 900776), a potent, selective checkpoint kinase 1 (Chk1) inhibitor, as monotherapy and in combination with gemcitabine in a first-in-human phase I clinical trial in patients with advanced solid tumor malignancies. PATIENTS AND METHODS Forty-three patients were treated by intravenous infusion with MK-8776 at seven dose levels ranging from 10 to 150 mg/m(2) as monotherapy and then in combination with gemcitabine 800 mg/m(2) (part A, n = 26) or gemcitabine 1,000 mg/m(2) (part B, n = 17). Forty percent of patients had three or more prior treatment regimens, and one third of patients had previously received gemcitabine. RESULTS As monotherapy, MK-8776 was well tolerated, with QTc prolongation (19%), nausea (16%), fatigue (14%), and constipation (14%) as the most frequent adverse effects. Combination therapy demonstrated a higher frequency of adverse effects, predominantly fatigue (63%), nausea (44%), decreased appetite (37%), thrombocytopenia (32%), and neutropenia (24%), as well as dose-related, transient QTc prolongation (17%). The median number of doses of MK-8776 administered was five doses, with relative dose-intensity of 0.96. Bioactivity was assessed by γ-H2AX ex vivo assay. Of 30 patients evaluable for response, two showed partial response, and 13 exhibited stable disease. CONCLUSION MK-8776 was well tolerated as monotherapy and in combination with gemcitabine. Early evidence of clinical efficacy was observed. The recommended phase II dose is MK-8776 200 mg plus gemcitabine 1,000 mg/m(2) on days 1 and 8 of a 21-day cycle.
Collapse
Affiliation(s)
- Adil I Daud
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ.
| | - Michelle T Ashworth
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Jonathan Strosberg
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Jonathan W Goldman
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - David Mendelson
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Gregory Springett
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Alan P Venook
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Sabine Loechner
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Lee S Rosen
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Frances Shanahan
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - David Parry
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Stuart Shumway
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Jennifer A Grabowsky
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Tomoko Freshwater
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Christopher Sorge
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Soonmo Peter Kang
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Randi Isaacs
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| | - Pamela N Munster
- Adil I. Daud, Michelle T. Ashworth, Alan P. Venook, Jennifer A. Grabowsky, and Pamela N. Munster, University of California, San Francisco, San Francisco; Jonathan W. Goldman and Lee S. Rosen, University of California, Los Angeles, Santa Monica, CA; Jonathan Strosberg and Gregory Springett, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; David Mendelson, Pinnacle Oncology Hematology, Scottsdale, AZ; and Sabine Loechner, Frances Shanahan, David Parry, Stuart Shumway, Tomoko Freshwater, Christopher Sorge, Soonmo Peter Kang, and Randi Isaacs, Merck, Whitehouse Station, NJ
| |
Collapse
|
17
|
Goto H, Kasahara K, Inagaki M. Novel insights into Chk1 regulation by phosphorylation. Cell Struct Funct 2014; 40:43-50. [PMID: 25748360 DOI: 10.1247/csf.14017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Checkpoint kinase 1 (Chk1) is a conserved protein kinase central to the cell-cycle checkpoint during DNA damage response (DDR). Until recently, ATR, a protein kinase activated in response to DNA damage or stalled replication, has been considered as the sole regulator of Chk1. Recent progress, however, has led to the identification of additional protein kinases involved in Chk1 phosphorylation, affecting the subcellular localization and binding partners of Chk1. In fact, spatio-temporal regulation of Chk1 is of critical importance not only in the DDR but also in normal cell-cycle progression. In due course, many potent inhibitors targeted to Chk1 have been developed as anticancer agents and some of these inhibitors are currently in clinical trials. In this review, we summarize the current knowledge of Chk1 regulation by phosphorylation.
Collapse
Affiliation(s)
- Hidemasa Goto
- Division of Biochemistry, Aichi Cancer Center Research Institute; Department of Cellular Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | | | | |
Collapse
|
18
|
Yuan LL, Green A, David L, Dozier C, Récher C, Didier C, Tamburini J, Manenti S. Targeting CHK1 inhibits cell proliferation in FLT3-ITD positive acute myeloid leukemia. Leuk Res 2014; 38:1342-9. [PMID: 25281057 DOI: 10.1016/j.leukres.2014.08.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 07/04/2014] [Accepted: 08/30/2014] [Indexed: 01/28/2023]
Abstract
CHK1 Ser/Thr kinase, a well characterized regulator of DNA damage response, is also involved in normal cell cycle progression. In this study, we investigate how CHK1 participates to proliferation of acute myeloid leukemia cells expressing the mutated FLT3-ITD tyrosine kinase receptor. Pharmacological inhibition of CHK1 as well as its shRNA mediated down regulation reduced the proliferation rate of FLT-ITD expressing leukemic cell lines in a cytostatic manner. Flow cytometry analysis revealed no accumulation in a specific phase of the cell cycle upon CHK1 inhibition. Accordingly, lentiviral-mediated CHK1 overexpression increased the proliferation rate of FLT3-ITD expressing cells, as judged by cell viability and [3H] thymidine incorporation experiments. By contrast, expression of a ser280 mutant did not, suggesting that phosphorylation of this residue is an important determinant of CHK1 proliferative function. Clonogenic growth of primary leukemic cells from patients in semi-solid medium was reduced upon CHK1 inhibition, confirming the data obtained with leukemic established cell lines. Surprisingly, 3 out of 4 CHK1 inhibitory compounds tested in this study were also potent inhibitors of the FLT3-ITD tyrosine kinase receptor. Altogether, these data identify CHK1 as a regulator of FLT3-ITD-positive leukemic cells proliferation, and they open interesting perspectives in terms of new therapeutic strategies for these pathologies.
Collapse
Affiliation(s)
- Ling Li Yuan
- Cancer Research Center of Toulouse, Inserm Unité Mixte de Recherche 1037, CNRS Equipe de Recherche labellisée 5294, Université de Toulouse, Centre Hospitalier Universitaire Purpan, Toulouse, France; Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Alexa Green
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - Laure David
- Cancer Research Center of Toulouse, Inserm Unité Mixte de Recherche 1037, CNRS Equipe de Recherche labellisée 5294, Université de Toulouse, Centre Hospitalier Universitaire Purpan, Toulouse, France
| | - Christine Dozier
- Cancer Research Center of Toulouse, Inserm Unité Mixte de Recherche 1037, CNRS Equipe de Recherche labellisée 5294, Université de Toulouse, Centre Hospitalier Universitaire Purpan, Toulouse, France
| | - Christian Récher
- Cancer Research Center of Toulouse, Inserm Unité Mixte de Recherche 1037, CNRS Equipe de Recherche labellisée 5294, Université de Toulouse, Centre Hospitalier Universitaire Purpan, Toulouse, France; Service d'Hématologie, Centre Hospitalier Universitaire Purpan, Toulouse, France
| | - Christine Didier
- Cancer Research Center of Toulouse, Inserm Unité Mixte de Recherche 1037, CNRS Equipe de Recherche labellisée 5294, Université de Toulouse, Centre Hospitalier Universitaire Purpan, Toulouse, France
| | - Jérôme Tamburini
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - Stéphane Manenti
- Cancer Research Center of Toulouse, Inserm Unité Mixte de Recherche 1037, CNRS Equipe de Recherche labellisée 5294, Université de Toulouse, Centre Hospitalier Universitaire Purpan, Toulouse, France.
| |
Collapse
|
19
|
Min W, Bruhn C, Grigaravicius P, Zhou ZW, Li F, Krüger A, Siddeek B, Greulich KO, Popp O, Meisezahl C, Calkhoven CF, Bürkle A, Xu X, Wang ZQ. Poly(ADP-ribose) binding to Chk1 at stalled replication forks is required for S-phase checkpoint activation. Nat Commun 2014; 4:2993. [PMID: 24356582 DOI: 10.1038/ncomms3993] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 11/22/2013] [Indexed: 01/02/2023] Open
Abstract
Damaged replication forks activate poly(ADP-ribose) polymerase 1 (PARP1), which catalyses poly(ADP-ribose) (PAR) formation; however, how PARP1 or poly(ADP-ribosyl)ation is involved in the S-phase checkpoint is unknown. Here we show that PAR, supplied by PARP1, interacts with Chk1 via a novel PAR-binding regulatory (PbR) motif in Chk1, independent of ATR and its activity. iPOND studies reveal that Chk1 associates readily with the unperturbed replication fork and that PAR is required for efficient retention of Chk1 and phosphorylated Chk1 at the fork. A PbR mutation, which disrupts PAR binding, but not the interaction with its partners Claspin or BRCA1, impairs Chk1 and the S-phase checkpoint activation, and mirrors Chk1 knockdown-induced hypersensitivity to fork poisoning. We find that long chains, but not short chains, of PAR stimulate Chk1 kinase activity. Collectively, we disclose a previously unrecognized mechanism of the S-phase checkpoint by PAR metabolism that modulates Chk1 activity at the replication fork.
Collapse
Affiliation(s)
- WooKee Min
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Christopher Bruhn
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Paulius Grigaravicius
- 1] Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany [2]
| | - Zhong-Wei Zhou
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Fu Li
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Anja Krüger
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Bénazir Siddeek
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Karl-Otto Greulich
- 1] Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany [2] Faculty of Biology and Pharmacy, Friedrich-Schiller-University Jena, Beutenberger strasse 11, 07745 Jena, Germany
| | - Oliver Popp
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Chris Meisezahl
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Cornelis F Calkhoven
- Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany
| | - Alexander Bürkle
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Xingzhi Xu
- Beijing Key Laboratory of DNA Damage Response, Capital Normal University, 105 Xi San Huan Road (N), Beijing 100048, China
| | - Zhao-Qi Wang
- 1] Leibniz Institute For Age Research-Fritz Lipmann Institute (FLI), Beutenberger strasse 11, 07745 Jena, Germany [2] Faculty of Biology and Pharmacy, Friedrich-Schiller-University Jena, Beutenberger strasse 11, 07745 Jena, Germany
| |
Collapse
|
20
|
Alexandrou AT, Li JJ. Cell cycle regulators guide mitochondrial activity in radiation-induced adaptive response. Antioxid Redox Signal 2014; 20:1463-80. [PMID: 24180340 PMCID: PMC3936506 DOI: 10.1089/ars.2013.5684] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE There are accruing concerns on potential genotoxic agents present in the environment including low-dose ionizing radiation (LDIR) that naturally exists on earth's surface and atmosphere and is frequently used in medical diagnosis and nuclear industry. Although its long-term health risk is being evaluated and remains controversial, LDIR is shown to induce temporary but significant adaptive responses in mammalian cells and animals. The mechanisms guiding the mitochondrial function in LDIR-induced adaptive response represent a unique communication between DNA damage and cellular metabolism. Elucidation of the LDIR-regulated mitochondrial activity may reveal new mechanisms adjusting cellular function to cope with hazardous environmental stress. RECENT ADVANCES Key cell cycle regulators, including Cyclin D1/CDK4 and Cyclin B1/cyclin-dependent kinase 1 (CDK1) complexes, are actively involved in the regulation of mitochondrial functions via phosphorylation of their mitochondrial targets. Accumulating new evidence supports a concept that the Cyclin B1/CDK1 complex acts as a mediator in the cross talk between radiation-induced DNA damage and mitochondrial functions to coordinate cellular responses to low-level genotoxic stresses. CRITICAL ISSUES The LDIR-mediated mitochondrial activity via Cyclin B1/CDK1 regulation is an irreplaceable network that is able to harmonize vital cellular functions with adjusted mitochondrial metabolism to enhance cellular homeostasis. FUTURE DIRECTIONS Further investigation of the coordinative mechanism that regulates mitochondrial activities in sublethal stress conditions, including LDIR, will reveal new insights of how cells cope with genotoxic injury and will be vital for future targeted therapeutic interventions that reduce environmental injury and cancer risk.
Collapse
Affiliation(s)
- Aris T Alexandrou
- Department of Radiation Oncology, NCI-Designated Comprehensive Cancer Center, University of California at Davis , Sacramento, California
| | | |
Collapse
|
21
|
Gamper AM, Rofougaran R, Watkins SC, Greenberger JS, Beumer JH, Bakkenist CJ. ATR kinase activation in G1 phase facilitates the repair of ionizing radiation-induced DNA damage. Nucleic Acids Res 2013; 41:10334-44. [PMID: 24038466 PMCID: PMC3905881 DOI: 10.1093/nar/gkt833] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The kinase ATR is activated by RPA-coated single-stranded DNA generated at aberrant replicative structures and resected double strand breaks. While many hundred candidate ATR substrates have been identified, the essential role of ATR in the replicative stress response has impeded the study of ATR kinase-dependent signalling. Using recently developed selective drugs, we show that ATR inhibition has a significantly more potent effect than ATM inhibition on ionizing radiation (IR)-mediated cell killing. Transient ATR inhibition for a short interval after IR has long-term consequences that include an accumulation of RPA foci and a total abrogation of Chk1 S345 phosphorylation. We show that ATR kinase activity in G1 phase cells is important for survival after IR and that ATR colocalizes with RPA in the absence of detectable RPA S4/8 phosphorylation. Our data reveal that, unexpectedly, ATR kinase inhibitors may be more potent cellular radiosensitizers than ATM kinase inhibitors, and that this is associated with a novel role for ATR in G1 phase cells.
Collapse
Affiliation(s)
- Armin M Gamper
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA, Department of Cell Biology and Physiology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA and Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Pim kinases phosphorylate Chk1 and regulate its functions in acute myeloid leukemia. Leukemia 2013; 28:293-301. [PMID: 23748345 DOI: 10.1038/leu.2013.168] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 11/08/2022]
Abstract
Phosphorylation by Akt on Ser 280 was reported to induce cytoplasmic retention and inactivation of CHK1 with consequent genetic instability in PTEN-/- cells. In acute myeloid leukemia cells carrying the FLT3-internal tandem duplication (ITD) mutation, we observed high rates of FLT3-ITD-dependent CHK1 Ser 280 phosphorylation. Pharmacological inhibition and RNA interference identified Pim1/2, not Akt, as effectors of this phosphorylation. Pim1 catalyzed Ser 280 phosphorylation in vitro and ectopic expression of Pim1/2-induced CHK1 phosphorylation. Ser 280 phosphorylation did not modify CHK1 localization, but facilitated its cell cycle and resistance functions in leukemic cells. FLT3, PIM or CHK1 inhibitors synergized with DNA-damaging agents to induce apoptosis, allowing cells to bypass the etoposide-induced G2/M arrest. Consistently, etoposide-induced CHK1-dependent phosphorylations of CDC25C on Ser 216 and histone H3 on Thr11 were decreased upon FLT3 inhibition. Accordingly, ectopic expression of CHK1 improved the resistance of FLT3-ITD cells and maintained histone H3 phosphorylation in response to DNA damage, whereas expression of unphosphorylated Ser 280Ala mutant did not. Finally, FLT3- and Pim-dependent phosphorylation of CHK1 on Ser 280 was confirmed in primary blasts from patients. These results identify a new pathway involved in the resistance of FLT3-ITD leukemic cells to genotoxic agents, and they constitute the first report of CHK1 Ser 280 regulation in myeloid malignancies.
Collapse
|
23
|
Peng A. Working hard for recovery: mitotic kinases in the DNA damage checkpoint. Cell Biosci 2013; 3:20. [PMID: 23618492 PMCID: PMC3641994 DOI: 10.1186/2045-3701-3-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/28/2013] [Indexed: 11/10/2022] Open
Abstract
Cell division in mitosis is tightly regulated via a group of protein kinases. Activation of these mitotic kinases is inhibited by the DNA damage checkpoint that arrests the cell cycle in interphase and prevents mitotic entry. Interestingly, it has been shown that the DNA damage checkpoint is feedback regulated by several mitotic kinases. These kinases are reactivated from checkpoint arrest to deactivate the checkpoint and restart cell cycle progression, thereby allowing the cell to recover from the DNA damage checkpoint. The emerging role of mitotic kinases in the DNA damage pathway provides important insights into cancer progression and treatment.
Collapse
Affiliation(s)
- Aimin Peng
- Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, NE 68583, USA.
| |
Collapse
|
24
|
Nek1 kinase associates with ATR-ATRIP and primes ATR for efficient DNA damage signaling. Proc Natl Acad Sci U S A 2013; 110:2175-80. [PMID: 23345434 DOI: 10.1073/pnas.1217781110] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The master checkpoint kinase ATR (ATM and Rad3-related) and its partner ATRIP (ATR-interacting protein) exist as a complex and function together in the DNA damage response. Unexpectedly, we found that the stability of the ATR-ATRIP complex is regulated by an unknown kinase independently of DNA damage. In search for this regulator of ATR-ATRIP, we found that a single member of the NIMA (never in mitosis A)-related kinase family, Nek1, is critical for initiating the ATR response. Upon DNA damage, cells lacking Nek1 failed to efficiently phosphorylate multiple ATR substrates and support ATR autophosphorylation at threnine 1989, one of the earliest events during the ATR response. The ability of Nek1 to promote ATR activation relies on the kinase activity of Nek1 and its interaction with ATR-ATRIP. Importantly, even in undamaged cells, Nek1 is required for maintaining the levels of ATRIP, the association between ATR and ATRIP, and the basal kinase activity of ATR. Thus, as an ATR-associated kinase, Nek1, enhances the stability and activity of ATR-ATRIP before DNA damage, priming ATR-ATRIP for a robust DNA damage response.
Collapse
|
25
|
Wohlbold L, Merrick KA, De S, Amat R, Kim JH, Larochelle S, Allen JJ, Zhang C, Shokat KM, Petrini JHJ, Fisher RP. Chemical genetics reveals a specific requirement for Cdk2 activity in the DNA damage response and identifies Nbs1 as a Cdk2 substrate in human cells. PLoS Genet 2012; 8:e1002935. [PMID: 22927831 PMCID: PMC3426557 DOI: 10.1371/journal.pgen.1002935] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 07/17/2012] [Indexed: 12/20/2022] Open
Abstract
The cyclin-dependent kinases (CDKs) that promote cell-cycle progression are targets for negative regulation by signals from damaged or unreplicated DNA, but also play active roles in response to DNA lesions. The requirement for activity in the face of DNA damage implies that there are mechanisms to insulate certain CDKs from checkpoint inhibition. It remains difficult, however, to assign precise functions to specific CDKs in protecting genomic integrity. In mammals, Cdk2 is active throughout S and G2 phases, but Cdk2 protein is dispensable for survival, owing to compensation by other CDKs. That plasticity obscured a requirement for Cdk2 activity in proliferation of human cells, which we uncovered by replacement of wild-type Cdk2 with a mutant version sensitized to inhibition by bulky adenine analogs. Here we show that transient, selective inhibition of analog-sensitive (AS) Cdk2 after exposure to ionizing radiation (IR) enhances cell-killing. In extracts supplemented with an ATP analog used preferentially by AS kinases, Cdk2(as) phosphorylated the Nijmegen Breakage Syndrome gene product Nbs1-a component of the conserved Mre11-Rad50-Nbs1 complex required for normal DNA damage repair and checkpoint signaling-dependent on a consensus CDK recognition site at Ser432. In vivo, selective inhibition of Cdk2 delayed and diminished Nbs1-Ser432 phosphorylation during S phase, and mutation of Ser432 to Ala or Asp increased IR-sensitivity. Therefore, by chemical genetics, we uncovered both a non-redundant requirement for Cdk2 activity in response to DNA damage and a specific target of Cdk2 within the DNA repair machinery.
Collapse
Affiliation(s)
- Lara Wohlbold
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Karl A. Merrick
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Program in Biochemistry and Program in Cell and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, New York, United States of America
| | - Saurav De
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ramon Amat
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jun Hyun Kim
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Stéphane Larochelle
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jasmina J. Allen
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | - Chao Zhang
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | - Kevan M. Shokat
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | - John H. J. Petrini
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Robert P. Fisher
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, New York, United States of America
| |
Collapse
|
26
|
Wang J, Han X, Feng X, Wang Z, Zhang Y. Coupling cellular localization and function of checkpoint kinase 1 (Chk1) in checkpoints and cell viability. J Biol Chem 2012; 287:25501-9. [PMID: 22692200 DOI: 10.1074/jbc.m112.350397] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chk1 plays a key role in regulating the replication checkpoint and DNA damage response. Recent evidence suggests that mammalian Chk1 regulates both the nuclear and cytoplasmic checkpoint events. However, mechanisms regulating cellular mobilization of Chk1 were not well understood. Here, we report the identification of regions of human Chk1 that regulate its protein cellular localization and checkpoint function. We demonstrate that the two highly conserved motifs (CM1 and CM2) at the C terminus of Chk1 function as a nuclear export signal and nuclear localization signal, respectively. Mutating five highly conserved residues within these two motifs of Chk1 resulted in its accumulation mainly in the cytoplasm. These cytoplasmic Chk1 mutants were less stable and exhibited significantly reduced phosphorylation by DNA damage treatment, yet they retained, at least partially, checkpoint function. Using an adenovirus-mediated gene targeting technique, we attempted to create an HCT116 cell line in which endogenous Chk1 is mutated so that it is expressed exclusively in the cytoplasm. However, we failed to obtain homozygous mutant cell lines. We found that even the heterozygous mutant cell lines showed cell survival defects accompanied by spontaneous cell death. Together, these results reveal novel regulatory mechanisms that couple protein cellular localization with the checkpoint response and cell viability of Chk1.
Collapse
Affiliation(s)
- Jingna Wang
- Department of Pharmacology, Case Comprehensive Cancer Center, Cleveland, Ohio 44106, USA
| | | | | | | | | |
Collapse
|