51
|
Fletcher SC, Grou CP, Legrand AJ, Chen X, Soderstrom K, Poletto M, Dianov GL. Sp1 phosphorylation by ATM downregulates BER and promotes cell elimination in response to persistent DNA damage. Nucleic Acids Res 2018; 46:1834-1846. [PMID: 29294106 PMCID: PMC5829641 DOI: 10.1093/nar/gkx1291] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 11/15/2022] Open
Abstract
ATM (ataxia-telangiectasia mutated) is a central molecule for DNA quality control. Its activation by DNA damage promotes cell-cycle delay, which facilitates DNA repair prior to replication. On the other hand, persistent DNA damage has been implicated in ATM-dependent cell death via apoptosis; however, the mechanisms underlying this process remain elusive. Here we find that, in response to persistent DNA strand breaks, ATM phosphorylates transcription factor Sp1 and initiates its degradation. We show that Sp1 controls expression of the key base excision repair gene XRCC1, essential for DNA strand break repair. Therefore, degradation of Sp1 leads to a vicious cycle that involves suppression of DNA repair and further aggravation of the load of DNA damage. This activates transcription of pro-apoptotic genes and renders cells susceptible to elimination via both apoptosis and natural killer cells. These findings constitute a previously unrecognized 'gatekeeper' function of ATM as a detector of cells with persistent DNA damage.
Collapse
Affiliation(s)
- Sally C Fletcher
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Claudia P Grou
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Arnaud J Legrand
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Xin Chen
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
- Department of Marine Technology, College of Ocean, Nantong University, Nantong, Jiangsu, 226007, China
| | - Kalle Soderstrom
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK
| | - Mattia Poletto
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Grigory L Dianov
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
- Institute of Cytology and Genetics, Russian Academy of Sciences, Lavrentyeva 10 Novosibirsk 630090, Russian Federation
- Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russian Federation
| |
Collapse
|
52
|
Ronson GE, Piberger AL, Higgs MR, Olsen AL, Stewart GS, McHugh PJ, Petermann E, Lakin ND. PARP1 and PARP2 stabilise replication forks at base excision repair intermediates through Fbh1-dependent Rad51 regulation. Nat Commun 2018; 9:746. [PMID: 29467415 PMCID: PMC5821833 DOI: 10.1038/s41467-018-03159-2] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/24/2018] [Indexed: 12/22/2022] Open
Abstract
PARP1 regulates the repair of DNA single-strand breaks generated directly, or during base excision repair (BER). However, the role of PARP2 in these and other repair mechanisms is unknown. Here, we report a requirement for PARP2 in stabilising replication forks that encounter BER intermediates through Fbh1-dependent regulation of Rad51. Whereas PARP2 is dispensable for tolerance of cells to SSBs or homologous recombination dysfunction, it is redundant with PARP1 in BER. Therefore, combined disruption of PARP1 and PARP2 leads to defective BER, resulting in elevated levels of replication-associated DNA damage owing to an inability to stabilise Rad51 at damaged replication forks and prevent uncontrolled DNA resection. Together, our results demonstrate how PARP1 and PARP2 regulate two independent, but intrinsically linked aspects of DNA base damage tolerance by promoting BER directly, and by stabilising replication forks that encounter BER intermediates.
Collapse
Affiliation(s)
- George E Ronson
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Ann Liza Piberger
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Martin R Higgs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Anna L Olsen
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Grant S Stewart
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Peter J McHugh
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Eva Petermann
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Nicholas D Lakin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
53
|
Bao Z, Tian B, Wang X, Feng H, Liang Y, Chen Z, Li W, Shen H, Ying S. Oleandrin induces DNA damage responses in cancer cells by suppressing the expression of Rad51. Oncotarget 2018; 7:59572-59579. [PMID: 27449097 PMCID: PMC5312332 DOI: 10.18632/oncotarget.10726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 07/09/2016] [Indexed: 11/25/2022] Open
Abstract
Oleandrin is a monomeric compound extracted from leaves and seeds of Nerium oleander. It had been reported that oleandrin could effectively inhibit the growth of human cancer cells. However, the specific mechanisms of the oleandrin-induced anti-tumor effects remain largely unclear. Genomic instability is one of the main features of cancer cells, it can be the combined effect of DNA damage and tumour-specific DNA repair defects. DNA damage plays important roles during tumorigenesis. In fact, most of the current chemotherapy agents were designed to kill cancer cells by inducing DNA damage. In this study, we found that oleandrin was effective to induce apoptosis in cancer cells, and cause rapid DNA damage response, represented by nuclear RPA (Replication Protein A, a single strand DNA binding protein) and γH2AX(a marker for DNA double strand breaks) foci formation. Interestingly, expression of RAD51, a key protein involved in homologous recombination (HR), was suppressed while XRCC1 was up-regulated in oleandrin treated cancer cells. These results suggested that XRCC1 may play a predominant role in repairing oleandrin-induced DNA damage. Collectively, oleandrin may be a potential anti-tumor agent by suppressing the expression of Rad51.
Collapse
Affiliation(s)
- Zhengqiang Bao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou 310009, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Baoping Tian
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xiaohui Wang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hanrong Feng
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ye Liang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhihua Chen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou 310009, China.,State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong 510120, China
| | - Songmin Ying
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou 310009, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
54
|
Du L, Yu W, Huang X, Zhao N, Liu F, Tong F, Zhang S, Niu B, Liu X, Xu S, Huang Y, Dai X, Xie C, Chen G, Cong X, Qu B. GSTP1 Ile105Val polymorphism might be associated with the risk of radiation pneumonitis among lung cancer patients in Chinese population: A prospective study. J Cancer 2018; 9:726-735. [PMID: 29556330 PMCID: PMC5858494 DOI: 10.7150/jca.20643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/03/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Growing data suggest that DNA damage repair and detoxification pathways play crucial roles in radiation-induced toxicities. To determine whether common functional single-nucleotide polymorphisms (SNPs) in candidate genes from these pathways can be used as predictors of radiation pneumonitis (RP), we conducted a prospective study to evaluate the associations between functional SNPs and risk of RP. Methods: We recruited a total of 149 lung cancer patients who had received intensity modulated radiation therapy (IMRT). GSTP1 and XRCC1 were genotyped using the SurPlexTM-xTAG method in all patients. RP events were prospectively scored using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), version 4.0. Kaplan-Meier analysis was used to determine the cumulative probability of RP of grade ≥ 2. Cox proportional hazard regression was performed to identify clinical variables and SNPs associated with risk of RP grade ≥ 2, using univariate and multivariate analysis, respectively. Results: With a median follow-up of 9 months, the incidence of RP of grade ≥ 2 was 38.3%. A predicting role in RP was observed for the GSTP1 SNP (adjusted hazard ratio 3.543; 95% CI 1.770-7.092; adjusted P< 0.001 for the Ile/Val and Val/Val genotypes versus Ile/Ile genotype). Whereas, we found that patients with XRCC1 399Arg/Gln and Gln/Gln genotypes had a lower risk of RP compares with those carrying Arg/Arg genotype (adjusted HR 0.653; 95% CI 0.342-1.245), but with no statistical significance observed (adjusted P = 0.195). Conclusions: Our results suggested a novel association between GSTP1 SNP 105Ile/Val and risk of RP development, which suggests the potential use of this genetic polymorphism as a predictor of RP. In addition, genetic polymorphisms of XRCC1 399Arg/Gln may also be associated with RP.
Collapse
Affiliation(s)
- Lehui Du
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Wei Yu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Xiang Huang
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Nana Zhao
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Fang Liu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Fang Tong
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Sujing Zhang
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Baolong Niu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Xiaoliang Liu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Shouping Xu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Yurong Huang
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Xiangkun Dai
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Chuanbin Xie
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Gaoxiang Chen
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Xiaohu Cong
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Baolin Qu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| |
Collapse
|
55
|
Azambuja DB, Leguisamo NM, Gloria HC, Kalil AN, Rhoden E, Saffi J. Prognostic impact of changes in base excision repair machinery in sporadic colorectal cancer. Pathol Res Pract 2018; 214:64-71. [DOI: 10.1016/j.prp.2017.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/03/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023]
|
56
|
Horton JK, Stefanick DF, Zhao ML, Janoshazi AK, Gassman NR, Seddon HJ, Wilson SH. XRCC1-mediated repair of strand breaks independent of PNKP binding. DNA Repair (Amst) 2017; 60:52-63. [PMID: 29100039 PMCID: PMC5696015 DOI: 10.1016/j.dnarep.2017.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/03/2017] [Accepted: 10/12/2017] [Indexed: 10/18/2022]
Abstract
Repair of DNA-protein crosslinks and oxidatively damaged DNA base lesions generates intermediates with nicks or gaps with abnormal and blocked 3'-phosphate and 5'-OH ends that prevent the activity of DNA polymerases and ligases. End cleaning in mammalian cells by Tdp1 and PNKP produces the conventional 3'-OH and 5'-phosphate DNA ends suitable for completion of repair. This repair function of PNKP is facilitated by its binding to the scaffold protein XRCC1, and phosphorylation of XRCC1 by CK2 at several consensus sites enables PNKP binding and recruitment to DNA damage. To evaluate this documented repair process, a phosphorylation mutant of XRCC1, designed to eliminate PNKP binding, was stably expressed in Xrcc1-/- mouse fibroblast cells. Analysis of PNKP-GFP accumulation at micro-irradiation induced damage confirmed that the XRCC1 phosphorylation mutant failed to support efficient PNKP recruitment, whereas there was rapid recruitment in cells expressing wild-type XRCC1. Recruitment of additional fluorescently-tagged repair factors PARP-1-YFP, GFF-XRCC1, PNKP-GFP and Tdp1-GFP to micro-irradiation induced damage was assessed in wild-type XRCC1-expressing cells. PARP-1-YFP recruitment was best fit to two exponentials, whereas kinetics for the other proteins were fit to a single exponential. The similar half-times of recruitment suggest that XRCC1 may be recruited with other proteins possibly as a pre-formed complex. Xrcc1-/- cells are hypersensitive to the DNA-protein cross-link inducing agent camptothecin (CPT) and the DNA oxidative agent H2O2 due in part to compromised PNKP-mediated repair. However, cells expressing the PNKP interaction mutant of XRCC1 demonstrated marked reversal of CPT hypersensitivity. This reversal represents XRCC1-dependent repair in the absence of the phosphorylation-dependent PNKP recruitment and suggests either an XRCC1-independent mechanism of PNKP recruitment or a functional back-up pathway for cleaning of blocked DNA ends.
Collapse
Affiliation(s)
- Julie K Horton
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Donna F Stefanick
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Ming-Lang Zhao
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Agnes K Janoshazi
- Signal Transduction Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Natalie R Gassman
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hannah J Seddon
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Samuel H Wilson
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
57
|
Lv MQ, Wang J, Yu XQ, Hong HH, Ren WJ, Ge P, Zhou DX. Association between X-ray repair cross-complementing group 1 (XRCC1) Arg399Gln polymorphism and endometriosis: A systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2017; 218:12-20. [DOI: 10.1016/j.ejogrb.2017.09.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/09/2017] [Accepted: 09/12/2017] [Indexed: 01/27/2023]
|
58
|
Markkanen E. Not breathing is not an option: How to deal with oxidative DNA damage. DNA Repair (Amst) 2017; 59:82-105. [PMID: 28963982 DOI: 10.1016/j.dnarep.2017.09.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 09/20/2017] [Indexed: 02/07/2023]
Abstract
Oxidative DNA damage constitutes a major threat to genetic integrity, and has thus been implicated in the pathogenesis of a wide variety of diseases, including cancer and neurodegeneration. 7,8-dihydro-8oxo-deoxyGuanine (8-oxo-G) is one of the best characterised oxidative DNA lesions, and it can give rise to point mutations due to its miscoding potential that instructs most DNA polymerases (Pols) to preferentially insert Adenine (A) opposite 8-oxo-G instead of the correct Cytosine (C). If uncorrected, A:8-oxo-G mispairs can give rise to C:G→A:T transversion mutations. Cells have evolved a variety of pathways to mitigate the mutational potential of 8-oxo-G that include i) mechanisms to avoid incorporation of oxidized nucleotides into DNA through nucleotide pool sanitisation enzymes (by MTH1, MTH2, MTH3 and NUDT5), ii) base excision repair (BER) of 8-oxo-G in DNA (involving MUTYH, OGG1, Pol λ, and other components of the BER machinery), and iii) faithful bypass of 8-oxo-G lesions during replication (using a switch between replicative Pols and Pol λ). In the following, the fate of 8-oxo-G in mammalian cells is reviewed in detail. The differential origins of 8-oxo-G in DNA and its consequences for genetic stability will be covered. This will be followed by a thorough discussion of the different mechanisms in place to cope with 8-oxo-G with an emphasis on Pol λ-mediated correct bypass of 8-oxo-G during MUTYH-initiated BER as well as replication across 8-oxo-G. Furthermore, the multitude of mechanisms in place to regulate key proteins involved in 8-oxo-G repair will be reviewed. Novel functions of 8-oxo-G as an epigenetic-like regulator and insights into the repair of 8-oxo-G within the cellular context will be touched upon. Finally, a discussion will outline the relevance of 8-oxo-G and the proteins involved in dealing with 8-oxo-G to human diseases with a special emphasis on cancer.
Collapse
Affiliation(s)
- Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, Winterthurerstr. 260, 8057 Zürich, Switzerland.
| |
Collapse
|
59
|
APE1 modulates cellular responses to organophosphate pesticide-induced oxidative damage in non-small cell lung carcinoma A549 cells. Mol Cell Biochem 2017; 441:201-216. [PMID: 28887667 DOI: 10.1007/s11010-017-3186-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/01/2017] [Indexed: 12/15/2022]
Abstract
Monocrotophos (MCP) and chlorpyrifos (CP) are widely used organophosphate pesticides (OPPs), speculated to be linked with human pathologies including cancer. Owing to the fact that lung cells are most vulnerable to the environmental toxins, the development and progression of lung cancer can be caused by the exposure of OPPs. The present study investigates the oxidative DNA damage response evoked by MCP and CP in human non-small cell lung carcinoma A549 cells. A549 cells were exposed to MCP and CP; cytotoxicity and reactive oxygen species (ROS) generation were measured to select the non-toxic dose. In order to establish whether MCP and CP can initiate the DNA repair and cell survival signalling pathways in A549 cells, qRT-PCR and Western blotting techniques were used to investigate the mRNA and protein expression levels of DNA base excision repair (BER)-pathway enzymes and transcription factors (TFs) involved in cell survival mechanisms. A significant increase in cell viability and ROS generation was observed when exposed to low and moderate doses of MCP and CP at different time points (24, 48 and 72 h) studied. A549 cells displayed a dose-dependent accumulation of apurinic/apyrimidinic (AP) sites after 24 h exposure to MCP advocating for the activation of AP endonuclease-mediated DNA BER-pathway. Cellular responses to MCP- and CP-induced oxidative stress resulted in an imbalance in the mRNA and protein expression of BER-pathway enzymes, viz. PARP1, OGG1, APE1, XRCC1, DNA pol β and DNA ligase III α at different time points. The treatment of OPPs resulted in the upregulation of TFs, viz. Nrf2, c-jun, phospho-c-jun and inducible nitric oxide synthase. Immunofluorescent confocal imaging of A549 cells indicated that MCP and CP induces the translocation of APE1 within the cytoplasm at an early 6 h time point, whereas it promotes nuclear localization after 24 h of treatment, which suggests that APE1 subcellular distribution is dynamically regulated in response to OPP-induced oxidative stress. Furthermore, nuclear colocalization of APE1 and the TF c-jun was observed in response to the treatment of CP and MCP for different time points in A549 cells. Therefore, in this study we demonstrate that MCP- and CP-induced oxidative stress alters APE1-dependent BER-pathway and also mediates cell survival signalling mechanisms via APE1 regulation, thereby promoting lung cancer cell survival and proliferation.
Collapse
|
60
|
Horton JK, Seddon HJ, Zhao ML, Gassman NR, Janoshazi AK, Stefanick DF, Wilson SH. Role of the oxidized form of XRCC1 in protection against extreme oxidative stress. Free Radic Biol Med 2017; 107:292-300. [PMID: 28179111 PMCID: PMC5457714 DOI: 10.1016/j.freeradbiomed.2017.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 12/20/2022]
Abstract
The multi-domain protein XRCC1 is without catalytic activity, but can interact with a number of known repair proteins. The interaction between the N-terminal domain (NTD) of XRCC1 and DNA polymerase β (pol β) is critical for recruitment of pol β to sites of DNA damage and repair. Crystallographic and NMR approaches have identified oxidized and reduced forms of the XRCC1 NTD, and the corresponding forms of XRCC1 have been identified in cultured mouse fibroblast cells. Both forms of NTD interact with pol β, but the interaction is much stronger with the oxidized form. The potential for formation of the C12-C20 oxidized conformation can be removed by alanine substitution at C12 (C12A) leading to stabilized reduced XRCC1 with a lower pol β binding affinity. Here, we compare cells expressing C12A XRCC1 (XRE8) with those expressing wild-type XRCC1 (XC5). Reduced C12A XRCC1 is detected at sites of micro-irradiation DNA damage, but provides slower recruitment of pol β. Expression of reduced XRCC1 does not affect sensitivity to MMS or H2O2. In contrast, further oxidative stress imposed by glutathione depletion results in increased sensitization of reduced XRCC1-expressing cells to H2O2 compared with wild-type XRCC1-expressing cells. There is no indication of enhanced H2O2-generated free radicals or DNA strand breaks in XRE8 cells. However, elevated cellular PAR is found following H2O2 exposure, suggesting BER deficiency of H2O2-induced damage in the C12A expressing cells.
Collapse
Affiliation(s)
- Julie K Horton
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hannah J Seddon
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Ming-Lang Zhao
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Natalie R Gassman
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Agnes K Janoshazi
- Signal Transduction Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Donna F Stefanick
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Samuel H Wilson
- Genomic Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
61
|
Zhang Y, Dong S, Xu R, Yang Y, Zheng Z, Wang X, Ren R, Sun R, Li M, Yang H, Huang Y, Zhou F, Zheng A. Prognostic and predictive role of COX-2, XRCC1 and RASSF1 expression in patients with esophageal squamous cell carcinoma receiving radiotherapy. Oncol Lett 2017; 13:2549-2556. [PMID: 28454432 PMCID: PMC5403488 DOI: 10.3892/ol.2017.5780] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 11/07/2016] [Indexed: 01/13/2023] Open
Abstract
Identification of biomarkers for predicting radiosensitivity would be useful for administering individualized radiotherapy (RT) to patients with esophageal cancer. The aim of the present study was to evaluate the association between cyclooxygenase-2 (COX-2), X-ray repair cross complementing group 1 (XRCC1), ras association domain family 1 (RASSF1) protein expression, clinicopathological characteristics, radiosensitivity and survival rate in 76 patients with esophageal squamous cell carcinoma (ESCC) who were treated with RT. Positive expression of COX-2, XRCC1 and RASSF1 was identified by immunohistochemistry in 81.6, 52.6 and 59.2% of ESCC cases, respectively. Negative COX-2 expression was associated with tumor (T) stage, node (N) stage, clinical stage and complete response (P<0.05), but not with gender, age, tumor location, differentiation degree, lesion length, progression-free survival (PFS) or overall survival (OS; P>0.05). XRCC1 expression was not associated with the clinicopathological features of ESCC, response to RT, PFS or OS. Positive RASSF1 expression was associated with the clinical stage, response to RT, PFS and OS (P<0.05), but not with gender, age, tumor location, T stage, N stage, differentiation degree or the lesion length (P>0.05). In the subgroup analysis, RASSF1 positive/XRCC1 negative expression was correlated with a longer median OS and PFS (P<0.05). Multivariate analyses revealed that the tumor response and RASSF1 expression were significant prognostic factors. Therefore, positive RASSF1 expression is associated with ESCC RT sensitivity, and may be a useful independent prognostic factor for ESCC.
Collapse
Affiliation(s)
- Yaowen Zhang
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Shangwen Dong
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Ruiping Xu
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Yanping Yang
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Zhiyong Zheng
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Xiaojing Wang
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Runchuan Ren
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Ronggang Sun
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Ming Li
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Haijun Yang
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Yuting Huang
- Department of Gynecological Oncology, Tianjin Medical Univerisity Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Fuyou Zhou
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| | - Anping Zheng
- Department of Radiation Oncology, Anyang Cancer Hospital, Anyang, Henan 455000, P.R. China
| |
Collapse
|
62
|
Goodfellow E, Senhaji Mouhri Z, Williams C, Jean-Claude BJ. Design, synthesis and biological activity of novel molecules designed to target PARP and DNA. Bioorg Med Chem Lett 2017; 27:688-694. [DOI: 10.1016/j.bmcl.2016.09.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 10/21/2022]
|
63
|
Leguisamo NM, Gloria HC, Kalil AN, Martins TV, Azambuja DB, Meira LB, Saffi J. Base excision repair imbalance in colorectal cancer has prognostic value and modulates response to chemotherapy. Oncotarget 2017; 8:54199-54214. [PMID: 28903334 PMCID: PMC5589573 DOI: 10.18632/oncotarget.14909] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 11/30/2016] [Indexed: 11/25/2022] Open
Abstract
Colorectal cancer (CRC) is prevalent worldwide, and treatment often involves surgery and genotoxic chemotherapy. DNA repair mechanisms, such as base excision repair (BER) and mismatch repair (MMR), may not only influence tumour characteristics and prognosis but also dictate chemotherapy response. Defective MMR contributes to chemoresistance in colorectal cancer. Moreover, BER affects cellular survival by repairing genotoxic base damage in a process that itself can disrupt metabolism. In this study, we characterized BER and MMR gene expression in colorectal tumours and the association between this repair profile with patients’ clinical and pathological features. In addition, we exploited the possible mechanisms underlying the association between altered DNA repair, metabolism and response to chemotherapy. Seventy pairs of sporadic colorectal tumour samples and adjacent non-tumour mucosal specimens were assessed for BER and MMR gene and protein expression and their association with pathological and clinical features. MMR-deficient colon cancer cells (HCT116) transiently overexpressing MPG or XRCC1 were treated with 5-FU or TMZ and evaluated for viability and metabolic intermediate levels. Increase in BER gene and protein expression is associated with more aggressive tumour features and poor pathological outcomes in CRC. However, tumours with reduced MMR gene expression also displayed low MPG, OGG1 and PARP1 expression. Imbalancing BER by overexpression of MPG, but not XRCC1, sensitises MMR-deficient colon cancer cells to 5-FU and TMZ and leads to ATP depletion and lactate accumulation. MPG overexpression alters DNA repair and metabolism and is a potential strategy to overcome 5-FU chemotherapeutic resistance in MMR-deficient CRC.
Collapse
Affiliation(s)
- Natalia M Leguisamo
- Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Helena C Gloria
- Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Antonio N Kalil
- Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.,Oncology and Colorectal Surgery, Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Talita V Martins
- Oncology and Colorectal Surgery, Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Daniel B Azambuja
- Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.,Oncology and Colorectal Surgery, Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Lisiane B Meira
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jenifer Saffi
- Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
64
|
Halim NHA, Chong ETJ, Goh LPW, Chuah JA, See EUH, Chua KH, Lee PC. Variant Alleles in XRCC1 Arg194Trp and Arg399Gln Polymorphisms Increase Risk of Gastrointestinal Cancer in Sabah, North Borneo. Asian Pac J Cancer Prev 2017; 17:1925-31. [PMID: 27221877 DOI: 10.7314/apjcp.2016.17.4.1925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The XRCC1 protein facilitates various DNA repair pathways; single-nucleotide polymorphisms (SNPs) in this gene are associated with a risk of gastrointestinal cancer (GIC) with inconsistent results, but no data have been previously reported for the Sabah, North Borneo, population. We accordingly investigated the XRCC1 Arg194Trp and Arg399Gln SNPs in terms of GIC risk in Sabah. MATERIALS AND METHODS We performed genotyping for both SNPs for 250 GIC patients and 572 healthy volunteers using a polymerase chain reaction- restriction fragment length polymorphism approach. We validated heterozygosity and homozygosity for both SNPs using direct sequencing. RESULTS The presence of a variant 194Trp allele in the Arg194Trp SNP was significantly associated with a higher risk of GIC, especially with gastric and colorectal cancers. We additionally found that the variant 399Gln allele in Arg399Gln SNP was associated with a greater risk of developing gastric cancer. Our combined analysis revealed that inheritance of variant alleles in both SNPs increased the GIC risk in Sabah population. Based on our etiological analysis, we found that subjects ≥50 years and males who carrying the variant 194Trp allele, and Bajau subjects carrying the 399Gln allele had a significantly increased risk of GIC. CONCLUSIONS Our findings suggest that inheritance of variant alleles in XRCC1 Arg194Trp and Arg399Gln SNPs may act as biomarkers for the early detection of GIC, especially for gastric and colorectal cancers in the Sabah population.
Collapse
Affiliation(s)
- Noor Hanis Abu Halim
- Biotechnology Programme, Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Malaysia E-mail :
| | | | | | | | | | | | | |
Collapse
|
65
|
Ooka M, Kobayashi K, Abe T, Akiyama K, Hada M, Takeda S, Hirota K. Determination of genotoxic potential by comparison of structurally related azo dyes using DNA repair-deficient DT40 mutant panels. CHEMOSPHERE 2016; 164:106-112. [PMID: 27580264 DOI: 10.1016/j.chemosphere.2016.08.092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 07/15/2016] [Accepted: 08/20/2016] [Indexed: 06/06/2023]
Abstract
Azo dyes, including Sudan I, Orange II and Orange G, are industrial dyes that are assumed to have genotoxic potential. However, neither the type of DNA damage induced nor the structural features responsible for toxicity have been determined. We used a panel of DNA-repair-pathway-deficient mutants generated from chicken DT40 cells to evaluate the ability of these azo dyes to induce DNA damage and to identify the type of DNA damage induced. We compared the structurally related azo dyes Sudan I, Orange II and Orange G to identify the structural features responsible for genotoxicity. Compared with wild type cells, the double-strand break repair defective RAD54-/-/KU70-/- cells were significantly more sensitive to Sudan I, but not to Orange II or Orange G. The quantum-chemical calculations revealed that Sudan I, but not Orange II or Orange G, has a complete planar aromatic ring structure. These suggest that the planar feature of Sudan I is critical to the inducing of double-strand breaks. In summary, we used a DNA-repair mutant panel in combination with quantum-chemical calculations to provide a clue to the chemical structure responsible for genotoxicity.
Collapse
Affiliation(s)
- Masato Ooka
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan
| | - Koji Kobayashi
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan
| | - Takuya Abe
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan
| | - Kazuhiko Akiyama
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan
| | - Masahiko Hada
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan.
| |
Collapse
|
66
|
Cinesi C, Aeschbach L, Yang B, Dion V. Contracting CAG/CTG repeats using the CRISPR-Cas9 nickase. Nat Commun 2016; 7:13272. [PMID: 27827362 PMCID: PMC5105158 DOI: 10.1038/ncomms13272] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022] Open
Abstract
CAG/CTG repeat expansions cause over 13 neurological diseases that remain without a cure. Because longer tracts cause more severe phenotypes, contracting them may provide a therapeutic avenue. No currently known agent can specifically generate contractions. Using a GFP-based chromosomal reporter that monitors expansions and contractions in the same cell population, here we find that inducing double-strand breaks within the repeat tract causes instability in both directions. In contrast, the CRISPR-Cas9 D10A nickase induces mainly contractions independently of single-strand break repair. Nickase-induced contractions depend on the DNA damage response kinase ATM, whereas ATR inhibition increases both expansions and contractions in a MSH2- and XPA-dependent manner. We propose that DNA gaps lead to contractions and that the type of DNA damage present within the repeat tract dictates the levels and the direction of CAG repeat instability. Our study paves the way towards deliberate induction of CAG/CTG repeat contractions in vivo.
Collapse
Affiliation(s)
- Cinzia Cinesi
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Lorène Aeschbach
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Bin Yang
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Vincent Dion
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
67
|
Subhash VV, Tan SH, Yeo MS, Yan FL, Peethala PC, Liem N, Krishnan V, Yong WP. ATM Expression Predicts Veliparib and Irinotecan Sensitivity in Gastric Cancer by Mediating P53-Independent Regulation of Cell Cycle and Apoptosis. Mol Cancer Ther 2016; 15:3087-3096. [DOI: 10.1158/1535-7163.mct-15-1002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 08/26/2016] [Accepted: 08/26/2016] [Indexed: 11/16/2022]
|
68
|
ZHANG YAOHONG, LUO ZHAOYUN, YANG LIYE, CHEN SENMING, CHEN CHUZHI, LIN ZHIXIONG. The association between four SNPs of X-ray repair cross complementing protein 1 and the sensitivity to radiotherapy in patients with esophageal squamous cell carcinoma. Oncol Lett 2016; 11:3508-3514. [PMID: 27123143 PMCID: PMC4840843 DOI: 10.3892/ol.2016.4384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 01/11/2016] [Indexed: 02/05/2023] Open
Abstract
Early stage diagnosis and therapeutic outcomes of esophageal squamous cell carcinoma remain poor. In order to evaluate the association between 4 single nucleotide polymorphisms (SNPs) of X-ray repair cross complementing protein 1 (XRCC1) and the sensitivity to radiotherapy in patients with esophageal squamous cell carcinoma (ESCC), the present study identified 4 SNPs of XRCC1 and evaluated the distribution of these genotypes among patients with ESCC. Venous blood samples from 175 patients with ESCC were collected and DNA was extracted. The 4 SNPs of the XRCC1 gene fragment were amplified using three primer pairs, which were sequenced. The mismatches were analyzed and identified using Basic Local Alignment Search Tool software. The sensitivity to radiotherapy was graded as effective and non-effective, according to the treatment results of the patients. The present study successfully amplified and sequenced 4 SNPs of XRCC1 in 112 out of the 175 patients with ESCC. The effective response rate of radiotherapy was 84.8% among the 112 patients. The effective response rate of patients with no mutation in the SNPs was 74.3%, and the rate increased to 89.6% in patients that had ≥1 mutation out of the 4 SNPs (χ2=4.389; P=0.036). For G28152A and G28152A mutations the effective response rate of patients was 91.2% (χ2=4.014; P=0.045) and 91.5% (χ2=4.451; P=0.035), respectively, which was significantly different compared to patients with no mutation (P=0.045 and P=0.035, respectively). The present results suggest that the 4 SNPs of XRCC1 are associated with the effective response rate of radiotherapy in patients with ESCC. The mutation of SNP G28152A was particularly important and may be a potential genomic predictor for radiotherapy sensitivity in patients with ESCC.
Collapse
Affiliation(s)
- YAOHONG ZHANG
- Department of Oncology, Chaozhou People's Hospital, Chaozhou, Guangdong 521000, P.R. China
| | - ZHAOYUN LUO
- Medical Laboratory Center, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong 521000, P.R. China
| | - LIYE YANG
- Medical Laboratory Center, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong 521000, P.R. China
| | - SENMING CHEN
- Department of Oncology, Chaozhou People's Hospital, Chaozhou, Guangdong 521000, P.R. China
| | - CHUZHI CHEN
- Medical Laboratory Center, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong 521000, P.R. China
| | - ZHIXIONG LIN
- Department of Radiation Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515031, P.R. China
- Correspondence to: Dr Zhixiong Lin, Department of Radiation Oncology, Affiliated Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, Guangdong 515031, P.R. China, E-mail:
| |
Collapse
|
69
|
Welcsh P, Kehrli K, Lazarchuk P, Ladiges W, Sidorova J. Application of the microfluidic-assisted replication track analysis to measure DNA repair in human and mouse cells. Methods 2016; 108:99-110. [PMID: 27130890 DOI: 10.1016/j.ymeth.2016.04.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/19/2016] [Accepted: 04/25/2016] [Indexed: 11/25/2022] Open
Abstract
Functional studies of the roles that DNA helicases play in human cells have benefited immensely from DNA fiber (or single molecule) technologies, which enable us to discern minute differences in behaviors of individual replication forks in genomic DNA in vivo. DNA fiber technologies are a group of methods that use different approaches to unravel and stretch genomic DNA to its contour length, and display it on a glass surface in order to immuno-stain nucleoside analog incorporation into DNA to reveal tracks (or tracts) of replication. We have previously adopted a microfluidic approach to DNA stretching and used it to analyze DNA replication. This method was introduced under the moniker maRTA or microfluidic-assisted Replication Track Analysis, and we have since used it to analyze roles of the RECQ helicases WRN and BLM, and other proteins in normal and perturbed replication. Here we describe a novel application of maRTA to detect and measure repair of DNA damage produced by three different agents relevant to etiology or therapy of cancer: methyl-methanesulfonate, UV irradiation, and mitomycin C. Moreover, we demonstrate the utility of this method by analyzing DNA repair in cells with reduced levels of WRN or of the base excision repair protein XRCC1.
Collapse
Affiliation(s)
- Piri Welcsh
- Department of Pathology, Box 357705, University of Washington, 1959 NE Pacific St., Seattle, WA 98195-7705, USA
| | - Keffy Kehrli
- Department of Pathology, Box 357705, University of Washington, 1959 NE Pacific St., Seattle, WA 98195-7705, USA
| | - Pavlo Lazarchuk
- Department of Pathology, Box 357705, University of Washington, 1959 NE Pacific St., Seattle, WA 98195-7705, USA
| | - Warren Ladiges
- Department of Comparative Medicine, Box 357340, University of Washington, 1959 NE Pacific St., Seattle, WA 98195-7340, USA
| | - Julia Sidorova
- Department of Pathology, Box 357705, University of Washington, 1959 NE Pacific St., Seattle, WA 98195-7705, USA.
| |
Collapse
|
70
|
Kim T, Freudenthal BD, Beard WA, Wilson SH, Schlick T. Insertion of oxidized nucleotide triggers rapid DNA polymerase opening. Nucleic Acids Res 2016; 44:4409-24. [PMID: 27034465 PMCID: PMC4872097 DOI: 10.1093/nar/gkw174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/04/2016] [Indexed: 11/21/2022] Open
Abstract
A novel mechanism is unveiled to explain why a pro-mutagenic nucleotide lesion (oxidized guanine, 8-oxoG) causes the mammalian DNA repair polymerase-β (pol-β) to rapidly transition to an inactive open conformation. The mechanism involves unexpected features revealed recently in time-lapse crystallography. Specifically, a delicate water network associated with a lesion-stabilizing auxilliary product ion Mg(p) triggers a cascade of events that leads to poor active site geometry and the rupture of crucial molecular interactions between key residues in both the anti(8-oxoG:C) and syn(8-oxoG:A) systems. Once the base pairs in these lesioned systems are broken, dislocation of both Asp192 (a metal coordinating ligand) and the oxoG phosphate group (PO4) interfere with the hydrogen bonding between Asp192 and Arg258, whose rotation toward Asp192 is crucial to the closed-to-open enzyme transition. Energetically, the lesioned open states are similar in energy to those of the corresponding closed complexes after chemistry, in marked contrast to the unlesioned pol-β anti(G:C) system, whose open state is energetically higher than the closed state. The delicate surveillance system offers a fundamental protective mechanism in the cell that triggers DNA repair events which help deter insertion of oxidized lesions.
Collapse
Affiliation(s)
- Taejin Kim
- Department of Chemistry, New York University, 10th Floor Silver Center, 100 Washington Square East, New York, NY 10003, USA
| | - Bret D Freudenthal
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709, USA
| | - William A Beard
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709, USA
| | - Samuel H Wilson
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709, USA
| | - Tamar Schlick
- Department of Chemistry, New York University, 10th Floor Silver Center, 100 Washington Square East, New York, NY 10003, USA Courant Institute of Mathematical Sciences, New York University, 251 Mercer Street, New York, NY 10012, USA
| |
Collapse
|
71
|
Ooka M, Takazawa H, Takeda S, Hirota K. Cytotoxic and genotoxic profiles of benzo[a]pyrene and N-nitrosodimethylamine demonstrated using DNA repair deficient DT40 cells with metabolic activation. CHEMOSPHERE 2016; 144:1901-1907. [PMID: 26547024 DOI: 10.1016/j.chemosphere.2015.10.085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 10/19/2015] [Accepted: 10/21/2015] [Indexed: 06/05/2023]
Abstract
Benzo[a]pyrene and N-nitrosodimethylamine are major genotoxic compounds present in cigarette smoke, food and oil. To examine the type(s) of DNA damage induced by these compounds, we used a panel of DNA-repair-pathway-deficient mutants generated from chicken DT40 cells and achieved metabolic activation of the test compounds by including rat liver S9 mix. Consistent with expections, benzo[a]pyrene and N-nitrosodimethylamine require metabolicactivation to become genotoxic. The REV3(-/-) mutant cell line exhibited the highest sensitivity, in terms of increased cytotoxicity, to the both compounds after metabolic activation consistent with the known ability of these two compounds to induce DNA adducts. Strikingly, we found that the RAD54(-/-)/KU70(-/-) cell line, a mutant defective in the repair of double-strand breaks, is sensitive to benzo[a]pyrene, suggesting that this compound also induces strand breaks in these cells. In this study we combined a previously employed method, metabolic activation by S9 mix, with the use of a DNA-repair mutant panel, thereby broadening the range of compounds that can be screened for potential genotoxicity.
Collapse
Affiliation(s)
- Masato Ooka
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo 192-0397, Japan
| | - Hironori Takazawa
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo 192-0397, Japan
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo 192-0397, Japan.
| |
Collapse
|
72
|
Abstract
Both proteolytic and nonproteolytic functions of ubiquitination are essential regulatory mechanisms for promoting DNA repair and the DNA damage response in mammalian cells. Deubiquitinating enzymes (DUBs) have emerged as key players in the maintenance of genome stability. In this minireview, we discuss the recent findings on human DUBs that participate in genome maintenance, with a focus on the role of DUBs in the modulation of DNA repair and DNA damage signaling.
Collapse
|
73
|
Seibold P, Behrens S, Schmezer P, Helmbold I, Barnett G, Coles C, Yarnold J, Talbot CJ, Imai T, Azria D, Koch CA, Dunning AM, Burnet N, Bliss JM, Symonds RP, Rattay T, Suga T, Kerns SL, Bourgier C, Vallis KA, Sautter-Bihl ML, Claßen J, Debus J, Schnabel T, Rosenstein BS, Wenz F, West CM, Popanda O, Chang-Claude J. XRCC1 Polymorphism Associated With Late Toxicity After Radiation Therapy in Breast Cancer Patients. Int J Radiat Oncol Biol Phys 2015; 92:1084-1092. [PMID: 26072091 DOI: 10.1016/j.ijrobp.2015.04.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/23/2015] [Accepted: 04/06/2015] [Indexed: 12/27/2022]
Abstract
PURPOSE To identify single-nucleotide polymorphisms (SNPs) in oxidative stress-related genes associated with risk of late toxicities in breast cancer patients receiving radiation therapy. METHODS AND MATERIALS Using a 2-stage design, 305 SNPs in 59 candidate genes were investigated in the discovery phase in 753 breast cancer patients from 2 prospective cohorts from Germany. The 10 most promising SNPs in 4 genes were evaluated in the replication phase in up to 1883 breast cancer patients from 6 cohorts identified through the Radiogenomics Consortium. Outcomes of interest were late skin toxicity and fibrosis of the breast, as well as an overall toxicity score (Standardized Total Average Toxicity). Multivariable logistic and linear regression models were used to assess associations between SNPs and late toxicity. A meta-analysis approach was used to summarize evidence. RESULTS The association of a genetic variant in the base excision repair gene XRCC1, rs2682585, with normal tissue late radiation toxicity was replicated in all tested studies. In the combined analysis of discovery and replication cohorts, carrying the rare allele was associated with a significantly lower risk of skin toxicities (multivariate odds ratio 0.77, 95% confidence interval 0.61-0.96, P=.02) and a decrease in Standardized Total Average Toxicity scores (-0.08, 95% confidence interval -0.15 to -0.02, P=.016). CONCLUSIONS Using a stage design with replication, we identified a variant allele in the base excision repair gene XRCC1 that could be used in combination with additional variants for developing a test to predict late toxicities after radiation therapy in breast cancer patients.
Collapse
Affiliation(s)
- Petra Seibold
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Peter Schmezer
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, Heidelberg, Germany
| | - Irmgard Helmbold
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Gillian Barnett
- Department of Oncology, Oncology Centre, Cambridge University Hospital NHS Foundation Trust, United Kingdom (UK)
| | - Charlotte Coles
- Department of Oncology, Oncology Centre, Cambridge University Hospital NHS Foundation Trust, United Kingdom (UK)
| | - John Yarnold
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | | | - Takashi Imai
- Advanced Radiation Biology Research Program, National Institute of Radiological Sciences, Chiba, Japan
| | - David Azria
- Department of Radiation Oncology and Medical Physics, I.C.M. - Institut regional du Cancer Montpellier, Montpellier, France
| | - C Anne Koch
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Neil Burnet
- Department of Oncology, Oncology Centre, Cambridge University Hospital NHS Foundation Trust, University of Cambridge, Cambridge, UK
| | - Judith M Bliss
- The Institute of Cancer Research, Clinical Trials and Statistics Unit, Sutton, UK
| | - R Paul Symonds
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester, UK
| | - Tim Rattay
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester, UK
| | - Tomo Suga
- Advanced Radiation Biology Research Program, National Institute of Radiological Sciences, Chiba, Japan
| | - Sarah L Kerns
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NH
| | - Celine Bourgier
- Department of Radiation Oncology and Medical Physics, I.C.M. - Institut regional du Cancer Montpellier, Montpellier, France
| | - Katherine A Vallis
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Oxford University, Oxford, UK
| | | | - Johannes Claßen
- Clinic for Radiation Therapy and Radiation Oncology, St. Vincentius-Kliniken gAG, Karlsruhe, Germany
| | - Juergen Debus
- Department of Radiation Oncology, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schnabel
- Clinic for Radiotherapy and Radiation Oncology, Klinikum Ludwigshafen, Ludwigshafen am Rhein, Germany
| | - Barry S Rosenstein
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NH
| | - Frederik Wenz
- Department of Radiation Oncology, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Catharine M West
- Institute of Cancer Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Odilia Popanda
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
74
|
Abstract
Scaffold proteins play a central role in DNA repair by recruiting and organizing sets of enzymes required to perform multi-step repair processes. X-ray cross complementing group 1 protein (XRCC1) forms enzyme complexes optimized for single-strand break repair, but participates in other repair pathways as well. Available structural data for XRCC1 interactions is summarized and evaluated in terms of its proposed roles in DNA repair. Mutational approaches related to the abrogation of specific XRCC1 interactions are also discussed. Although substantial progress has been made in elucidating the structural basis for XRCC1 function, the molecular mechanisms of XRCC1 recruitment related to several proposed roles of the XRCC1 DNA repair complex remain undetermined.
Collapse
Affiliation(s)
- Robert E London
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States.
| |
Collapse
|
75
|
DNA polymerases β and λ and their roles in cell. DNA Repair (Amst) 2015; 29:112-26. [DOI: 10.1016/j.dnarep.2015.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 01/29/2015] [Accepted: 02/02/2015] [Indexed: 10/24/2022]
|
76
|
Scott CL, Swisher EM, Kaufmann SH. Poly (ADP-ribose) polymerase inhibitors: recent advances and future development. J Clin Oncol 2015; 33:1397-406. [PMID: 25779564 PMCID: PMC4517072 DOI: 10.1200/jco.2014.58.8848] [Citation(s) in RCA: 274] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have shown promising activity in epithelial ovarian cancers, especially relapsed platinum-sensitive high-grade serous disease. Consistent with preclinical studies, ovarian cancers and a number of other solid tumor types occurring in patients with deleterious germline mutations in BRCA1 or BRCA2 seem to be particularly sensitive. However, it is also becoming clear that germline BRCA1/2 mutations are neither necessary nor sufficient for patients to derive benefit from PARP inhibitors. We provide an update on PARP inhibitor clinical development, describe recent advances in our understanding of PARP inhibitor mechanism of action, and discuss current issues in the development of these agents.
Collapse
Affiliation(s)
- Clare L Scott
- Clare L. Scott, Walter and Eliza Hall Institute of Medical Research and Royal Melbourne Hospital, Parkville, Victoria, Australia; Elizabeth M. Swisher, University of Washington, Seattle, WA; and Scott H. Kaufmann, Mayo Clinic, Rochester, MN
| | - Elizabeth M Swisher
- Clare L. Scott, Walter and Eliza Hall Institute of Medical Research and Royal Melbourne Hospital, Parkville, Victoria, Australia; Elizabeth M. Swisher, University of Washington, Seattle, WA; and Scott H. Kaufmann, Mayo Clinic, Rochester, MN
| | - Scott H Kaufmann
- Clare L. Scott, Walter and Eliza Hall Institute of Medical Research and Royal Melbourne Hospital, Parkville, Victoria, Australia; Elizabeth M. Swisher, University of Washington, Seattle, WA; and Scott H. Kaufmann, Mayo Clinic, Rochester, MN.
| |
Collapse
|
77
|
Yu X, Xiao H, Zhao B, Zhang X, Wang G. DNA repair gene ERCC1 C118T polymorphism predicts sensitivity of recurrent esophageal cancer to radiochemotherapy in a Chinese population. Thorac Cancer 2015; 6:741-8. [PMID: 26557912 PMCID: PMC4632926 DOI: 10.1111/1759-7714.12251] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/04/2015] [Indexed: 02/06/2023] Open
Abstract
Background DNA repair gene polymorphisms could alter DNA repair capacity and therefore associate with tumor sensitivity to radiochemotherapy. This study assessed excision repair cross-complementing group 1 (ERCC1) C118T and X-ray cross-complementing group 1 (XRCC1) G399A single-nucleotide polymorphisms in esophageal patients for an association with sensitivity to radiation and chemotherapy. Methods Esophageal squamous cell carcinoma patients (n = 118) who relapsed after surgery were enrolled for assessment of ERCC1 C118T and XRCC1 G399A polymorphisms by direct DNA sequencing. Results The response rate of treatments was 48.30%: 14 complete response (CR, 11.86%), 43 partial response (PR, 36.44%), 49 stable disease (SD, 41.53%), and 12 progressive disease (PD, 10.17%). ERCC1 C118T was significantly associated with treatment response (C/T vs. C/C + T/T, odds ratio [OR] = 6.035, 95% confidence interval [CI]: 2.114–17.226, P = 0.001) after adjusting for other clinicopathological factors. Patients carrying the C/T genotype had significantly prolonged overall survival (OS) compared with C/C and T/T (median OS 43.00 vs. 27.00, P = 0.027). Multivariate Cox regression showed that a response was only an independent prognostic factor for OS (CR + PR vs. SD+PD, HR = 0.471 95% CI 0.269–0.826, P = 0.009). Grade III and IV adverse events occurred in 12 of 118 patients (10.17%). Only concurrent radiochemotherapy significantly increased these adverse events (OR = 26.529, 95% CI 2.312–304.389, P = 0.008). Conclusion ERCC1 C118T could be a predictive factor for the response to radiotherapy and chemotherapy, but not a prognostic factor for OS in esophageal cancer patients after surgery.
Collapse
Affiliation(s)
- Xian Yu
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University Chongqing, China
| | - He Xiao
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University Chongqing, China
| | - Baojian Zhao
- Beijing Epigenetics Biotechnology Co., Ltd Beijing, China
| | - Xu Zhang
- Beijing Epigenetics Biotechnology Co., Ltd Beijing, China
| | - Ge Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University Chongqing, China
| |
Collapse
|
78
|
Tan X, Wang H, Luo G, Ren S, Li W, Cui J, Gill HS, Fu SW, Lu Y. Clinical significance of a point mutation in DNA polymerase beta (POLB) gene in gastric cancer. Int J Biol Sci 2015; 11:144-55. [PMID: 25561897 PMCID: PMC4279090 DOI: 10.7150/ijbs.10692] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/19/2014] [Indexed: 01/17/2023] Open
Abstract
Gastric cancer (GC) is a major cause of global cancer mortality. Genetic variations in DNA repair genes can modulate DNA repair capability and, consequently, have been associated with risk of developing cancer. We have previously identified a T to C point mutation at nucleotide 889 (T889C) in DNA polymerase beta (POLB) gene, a key enzyme involved in base excision repair in primary GCs. The purpose of this study was to evaluate the mutation and expression of POLB in a larger cohort and to identify possible prognostic roles of the POLB alterations in GC. Primary GC specimens and their matched normal adjacent tissues were collected at the time of surgery. DNA, RNA and protein samples were isolated from GC specimens and cell lines. Mutations were detected by PCR-RFLP/DHPLC and sequencing analysis. POLB gene expression was examined by RT-PCR, tissue microarray, Western blotting and immunofluorescence assays. The function of the mutation was evaluated by chemosensitivity, MTT, Transwell matrigel invasion and host cell reactivation assays. The T889C mutation was detected in 18 (10.17%) of 177 GC patients. And the T889C mutation was associated with POLB overexpression, lymph nodes metastases and poor tumor differentiation. In addition, patients with- the mutation had significantly shorter survival time than those without-, following postoperative chemotherapy. Furthermore, cell lines with T889C mutation in POLB gene were more resistant to the treatment of 5-fluorouracil, cisplatin and epirubicin than those with wild type POLB. Forced expression of POLB gene with T889C mutation resulted in enhanced cell proliferation, invasion and resistance to anticancer drugs, along with increased DNA repair capability. These results suggest that POLB gene with T889C mutation in surgically resected primary gastric tissues may be clinically useful for predicting responsiveness to chemotherapy in patients with GC. The POLB gene alteration may serve as a prognostic biomarker for GC.
Collapse
Affiliation(s)
- Xiaohui Tan
- 1. Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); ; 4. Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Hongyi Wang
- 2. Department of Sugary, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, P.R. China
| | - Guangbin Luo
- 3. Department of Genetics, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Shuyang Ren
- 1. Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)
| | - Wenmei Li
- 1. Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)
| | - Jiantao Cui
- 1. Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)
| | - Harindarpal S Gill
- 4. Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Sidney W Fu
- 4. Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Youyong Lu
- 1. Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)
| |
Collapse
|
79
|
Abdou I, Poirier GG, Hendzel MJ, Weinfeld M. DNA ligase III acts as a DNA strand break sensor in the cellular orchestration of DNA strand break repair. Nucleic Acids Res 2014; 43:875-92. [PMID: 25539916 PMCID: PMC4333375 DOI: 10.1093/nar/gku1307] [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] [Indexed: 01/18/2023] Open
Abstract
In the current model of DNA SSBR, PARP1 is regarded as the sensor of single-strand breaks (SSBs). However, biochemical studies have implicated LIG3 as another possible SSB sensor. Using a laser micro-irradiation protocol that predominantly generates SSBs, we were able to demonstrate that PARP1 is dispensable for the accumulation of different single-strand break repair (SSBR) proteins at sites of DNA damage in live cells. Furthermore, we show in live cells for the first time that LIG3 plays a role in mediating the accumulation of the SSBR proteins XRCC1 and PNKP at sites of DNA damage. Importantly, the accumulation of LIG3 at sites of DNA damage did not require the BRCT domain-mediated interaction with XRCC1. We were able to show that the N-terminal ZnF domain of LIG3 plays a key role in the enzyme's SSB sensing function. Finally, we provide cellular evidence that LIG3 and not PARP1 acts as the sensor for DNA damage caused by the topoisomerase I inhibitor, irinotecan. Our results support the existence of a second damage-sensing mechanism in SSBR involving the detection of nicks in the genome by LIG3.
Collapse
Affiliation(s)
- Ismail Abdou
- Department of Oncology, University of Alberta, and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Guy G Poirier
- Cancer Axis, CHUQ Research Center and Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| | - Michael J Hendzel
- Department of Oncology, University of Alberta, and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Michael Weinfeld
- Department of Oncology, University of Alberta, and Cross Cancer Institute, Edmonton, Alberta, Canada
| |
Collapse
|
80
|
Horton JK, Gassman NR, Dunigan BD, Stefanick DF, Wilson SH. DNA polymerase β-dependent cell survival independent of XRCC1 expression. DNA Repair (Amst) 2014; 26:23-9. [PMID: 25541391 DOI: 10.1016/j.dnarep.2014.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/21/2014] [Accepted: 11/25/2014] [Indexed: 01/09/2023]
Abstract
Base excision repair (BER) is a primary mechanism for repair of base lesions in DNA such as those formed by exposure to the DNA methylating agent methyl methanesulfonate (MMS). Both DNA polymerase β (pol β)- and XRCC1-deficient mouse fibroblasts are hypersensitive to MMS. This is linked to a repair deficiency as measured by accumulation of strand breaks and poly(ADP-ribose) (PAR). The interaction between pol β and XRCC1 is important for recruitment of pol β to sites of DNA damage. Endogenous DNA damage can substitute for MMS-induced damage such that BER deficiency as a result of either pol β- or XRCC1-deletion is associated with sensitivity to PARP inhibitors. Pol β shRNA was used to knock down pol β in Xrcc1(+/+) and Xrcc1(-/-) mouse fibroblasts. We determined whether pol β-mediated cellular resistance to MMS and PARP inhibitors resulted entirely from coordination with XRCC1 within the same BER sub-pathway. We find evidence for pol β-dependent cell survival independent of XRCC1 expression for both types of agents. The results suggest a role for pol β-dependent, XRCC1-independent repair. PAR immunofluorescence data are consistent with the hypothesis of a decrease in repair in both pol β knock down cell variants.
Collapse
Affiliation(s)
- Julie K Horton
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Natalie R Gassman
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Brittany D Dunigan
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Donna F Stefanick
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Samuel H Wilson
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
81
|
Brenerman BM, Illuzzi JL, Wilson DM. Base excision repair capacity in informing healthspan. Carcinogenesis 2014; 35:2643-52. [PMID: 25355293 PMCID: PMC4247524 DOI: 10.1093/carcin/bgu225] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 12/21/2022] Open
Abstract
Base excision repair (BER) is a frontline defense mechanism for dealing with many common forms of endogenous DNA damage, several of which can drive mutagenic or cell death outcomes. The pathway engages proteins such as glycosylases, abasic endonucleases, polymerases and ligases to remove substrate modifications from DNA and restore the genome back to its original state. Inherited mutations in genes related to BER can give rise to disorders involving cancer, immunodeficiency and neurodegeneration. Studies employing genetically defined heterozygous (haploinsufficient) mouse models indicate that partial reduction in BER capacity can increase vulnerability to both spontaneous and exposure-dependent pathologies. In humans, measurement of BER variation has been imperfect to this point, yet tools to assess BER in epidemiological surveys are steadily evolving. We provide herein an overview of the BER pathway and discuss the current efforts toward defining the relationship of BER defects with disease susceptibility.
Collapse
Affiliation(s)
- Boris M Brenerman
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jennifer L Illuzzi
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - David M Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
82
|
Krokan HE, Sætrom P, Aas PA, Pettersen HS, Kavli B, Slupphaug G. Error-free versus mutagenic processing of genomic uracil—Relevance to cancer. DNA Repair (Amst) 2014; 19:38-47. [DOI: 10.1016/j.dnarep.2014.03.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
83
|
Abstract
TDP1 and TDP2 were discovered and named based on the fact they process 3'- and 5'-DNA ends by excising irreversible protein tyrosyl-DNA complexes involving topoisomerases I and II, respectively. Yet, both enzymes have an extended spectrum of activities. TDP1 not only excises trapped topoisomerases I (Top1 in the nucleus and Top1mt in mitochondria), but also repairs oxidative damage-induced 3'-phosphoglycolates and alkylation damage-induced DNA breaks, and excises chain terminating anticancer and antiviral nucleosides in the nucleus and mitochondria. The repair function of TDP2 is devoted to the excision of topoisomerase II- and potentially topoisomerases III-DNA adducts. TDP2 is also essential for the life cycle of picornaviruses (important human and bovine pathogens) as it unlinks VPg proteins from the 5'-end of the viral RNA genome. Moreover, TDP2 has been involved in signal transduction (under the former names of TTRAP or EAPII). The DNA repair partners of TDP1 include PARP1, XRCC1, ligase III and PNKP from the base excision repair (BER) pathway. By contrast, TDP2 repair functions are coordinated with Ku and ligase IV in the non-homologous end joining pathway (NHEJ). This article summarizes and compares the biochemistry, functions, and post-translational regulation of TDP1 and TDP2, as well as the relevance of TDP1 and TDP2 as determinants of response to anticancer agents. We discuss the rationale for developing TDP inhibitors for combinations with topoisomerase inhibitors (topotecan, irinotecan, doxorubicin, etoposide, mitoxantrone) and DNA damaging agents (temozolomide, bleomycin, cytarabine, and ionizing radiation), and as novel antiviral agents.
Collapse
Affiliation(s)
- Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Building 37, Room 5068, NIH, Bethesda, MD 20892, USA.
| | - Shar-yin N Huang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Building 37, Room 5068, NIH, Bethesda, MD 20892, USA
| | - Rui Gao
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Building 37, Room 5068, NIH, Bethesda, MD 20892, USA
| | - Benu Brata Das
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Building 37, Room 5068, NIH, Bethesda, MD 20892, USA; Laboratory of Molecular Biology, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Junko Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Building 37, Room 5068, NIH, Bethesda, MD 20892, USA; Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo-ku 606-8501, Japan
| | - Christophe Marchand
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Building 37, Room 5068, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
84
|
Ensminger M, Iloff L, Ebel C, Nikolova T, Kaina B, Lӧbrich M. DNA breaks and chromosomal aberrations arise when replication meets base excision repair. ACTA ACUST UNITED AC 2014; 206:29-43. [PMID: 24982429 PMCID: PMC4085701 DOI: 10.1083/jcb.201312078] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
DNA double-strand breaks and chromosomal aberrations after treatment with N-alkylating agents likely arise as a result of replication fork collision with single-strand breaks generated during base excision repair. Exposures that methylate DNA potently induce DNA double-strand breaks (DSBs) and chromosomal aberrations, which are thought to arise when damaged bases block DNA replication. Here, we demonstrate that DNA methylation damage causes DSB formation when replication interferes with base excision repair (BER), the predominant pathway for repairing methylated bases. We show that cells defective in the N-methylpurine DNA glycosylase, which fail to remove N-methylpurines from DNA and do not initiate BER, display strongly reduced levels of methylation-induced DSBs and chromosomal aberrations compared with wild-type cells. Also, cells unable to generate single-strand breaks (SSBs) at apurinic/apyrimidinic sites do not form DSBs immediately after methylation damage. In contrast, cells deficient in x-ray cross-complementing protein 1, DNA polymerase β, or poly (ADP-ribose) polymerase 1 activity, all of which fail to seal SSBs induced at apurinic/apyrimidinic sites, exhibit strongly elevated levels of methylation-induced DSBs and chromosomal aberrations. We propose that DSBs and chromosomal aberrations after treatment with N-alkylators arise when replication forks collide with SSBs generated during BER.
Collapse
Affiliation(s)
- Michael Ensminger
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Lucie Iloff
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Christian Ebel
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Teodora Nikolova
- Institute of Toxicology, Medical Center of the University Mainz, 55131 Mainz, Germany
| | - Bernd Kaina
- Institute of Toxicology, Medical Center of the University Mainz, 55131 Mainz, Germany
| | - Markus Lӧbrich
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| |
Collapse
|
85
|
ERCC1 expression and tumor regression predict survival in esophageal squamous cell carcinoma patients receiving combined trimodality therapy. Pathol Res Pract 2014; 210:656-61. [PMID: 25042384 DOI: 10.1016/j.prp.2014.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/25/2014] [Accepted: 06/19/2014] [Indexed: 11/23/2022]
Abstract
PURPOSE Combined trimodality therapy with neoadjuvant chemoradiation followed by surgery has shown promising results for locally advanced operable esophageal cancer. DNA repair proteins may affect treatment efficacy through repairing DNA damage induced by chemotherapy and radiation therapy. We evaluated the associations of XRCC1, ERCC1 and MGMT expression with histopathologic response and survival in patients with locally advanced operable esophageal squamous cell carcinoma (ESCC) who received neoadjuvant chemoradiation. METHODS Paraffin-embedded pre-treatment tissue samples, collected by endoscopic biopsy from patients treated with cisplatin-based neoadjuvant chemoradiation followed by surgery, were immunohistochemically stained for XRCC1, ERCC1 and MGMT expression. RESULTS Of the 44 patients, major histopathologic response was noted in 26 (59.1%) patients. 68.8% of patients with ERCC1-negative tumors had major histopathologic response, compared to 53.6% of those who expressed positive ERCC1, though the difference was not statistically significant (P=0.361). The patients with ERCC1-negative tumor presented much better overall survival than those positive for ERCC1 expression (P=0.018). Patients with major histopathologic response had a 3-year survival rate of 96.2% versus those with minor response, with a 3-year survival rate of 41.5% (P=0.000). Multivariate analysis showed that ERCC1 expression and histopathologic response were independent predictive factors of overall survival in patients with locally advanced operable ESCC receiving neoadjuvant chemoradiation. CONCLUSION Patients with ERCC1-negative tumors show a benefit from neoadjuvant chemoradiation, ERCC1 expression and tumor regression are useful predictive markers in patients with locally advanced operable ESCC receiving neoadjuvant chemoradiation followed by surgery.
Collapse
|
86
|
Choudhury JH, Choudhury B, Kundu S, Ghosh SK. Combined effect of tobacco and DNA repair genes polymorphisms of XRCC1 and XRCC2 influence high risk of head and neck squamous cell carcinoma in northeast Indian population. Med Oncol 2014; 31:67. [PMID: 24958516 DOI: 10.1007/s12032-014-0067-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/03/2014] [Indexed: 01/12/2023]
Abstract
Tobacco consumption in various forms is one of the major risk factor for the development of head and neck squamous cell carcinoma. Polymorphisms in XRCC1 and XRCC2 genes may alter an individual's susceptibility to tobacco-related cancers. Here, we have investigated the interaction of XRCC1 (Arg399Gln) and XRCC2 (Arg188His) polymorphism and tobacco exposure in the progression of HNSCC in northeast Indian population. The population-based case-control study includes 110 HNSCC patients and 140 controls. The polymorphisms of XRCC1 and XRCC2 were studied by means of PCR-RFLP, and the results were confirmed by DNA sequencing. Smokers and tobacco-betel quid chewers were significantly higher in cases (P = 0.045 and 0.033). The variant homozygote AA genotype of XRCC1 Arg399Gln and heterozygote GA genotype of XRCC2 Arg188His has an increased risk toward HNSCC (OR 2.43; P = 0.031 and OR 3.29; P < 0.01, respectively). The interaction between tobacco-betel quid chewing and variant genotypes of XRCC1 and XRCC2 resulted in several fold increase the risk of HNSCC, when compared to non-chewers. Heavy smokers carrying XRCC1 AA and XRCC2 GA genotypes had a significantly higher risk of HNSCC compared to never smokers (P = 0.017 and 0.003, respectively). Upon gene-gene interaction analysis, individuals carrying both XRCC1 GA (Arg/Gln) and XRCC2 GA (Arg/His) genotypes had the highest risk of HNSCC (P = 0.001).Our finding suggests that interaction of tobacco and polymorphisms of XRCC1 and XRCC2 increases the risk of HNSCC. Furthermore, cross talk between these two DNA repair genes might modulate susceptibility toward HNSCC.
Collapse
Affiliation(s)
- Javed Hussain Choudhury
- Department of Biotechnology, Assam University (A Central University), Silchar, 788011, Assam, India
| | | | | | | |
Collapse
|
87
|
The oligonucleotide/oligosaccharide-binding fold motif is a poly(ADP-ribose)-binding domain that mediates DNA damage response. Proc Natl Acad Sci U S A 2014; 111:7278-83. [PMID: 24799691 DOI: 10.1073/pnas.1318367111] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oligonucleotide/oligosaccharide-binding (OB) fold is a ssDNA or RNA binding motif in prokaryotes and eukaryotes. Unexpectedly, we found that the OB fold of human ssDNA-binding protein 1 (hSSB1) is a poly(ADP ribose) (PAR) binding domain. hSSB1 exhibits high-affinity binding to PAR and recognizes iso-ADP ribose (ADPR), the linkage between two ADPR units. This interaction between PAR and hSSB1 mediates the early recruitment of hSSB1 to the sites of DNA damage. Mutations in the OB fold of hSSB1 that disrupt PAR binding abolish the relocation of hSSB1 to the sites of DNA damage. Moreover, PAR-mediated recruitment of hSSB1 is important for early DNA damage repair. We have screened other OB folds and found that several other OB folds also recognize PAR. Taken together, our study reveals a PAR-binding domain that mediates DNA damage repair.
Collapse
|
88
|
Zaanan A, Dalban C, Emile JF, Blons H, Fléjou JF, Goumard C, Istanbullu M, Calmel C, Alhazmi K, Validire P, Louvet C, de Gramont A, Laurent-Puig P, Taïeb J, Praz F. ERCC1, XRCC1 and GSTP1 Single Nucleotide Polymorphisms and Survival of Patients with Colon Cancer Receiving Oxaliplatin-Based Adjuvant Chemotherapy. J Cancer 2014; 5:425-32. [PMID: 24847383 PMCID: PMC4026996 DOI: 10.7150/jca.8594] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/26/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND While single nucleotide polymorphisms (SNP) in genes involved in DNA repair or drug metabolism have been shown to influence survival of metastatic colon cancer patients treated with FOLFOX, data on adjuvant setting are scarce. METHODS This study evaluated the correlation between disease-free survival (DFS) of 210 unselected stage III colon cancer patients receiving FOLFOX chemotherapy, and ERCC1-118 (rs11615, c.354T>C), XRCC1-399 (rs25487, c.1196G>A) and GSTP1-105 (rs1695, c.313A>G) polymorphisms. SNP were determined on tumor DNA using a PCR-based RFLP technique. RESULTS In univariate analysis, a trend towards longer DFS was observed for ERCC1 (C/T + T/T) versus (C/C) (HR=2.29; p=0.06), and XRCC1 (A/A) versus (G/G + G/A) (HR=1.61; p=0.16), but not for GSTP1 genotypes; a statistically significant p value was obtained when combining ERCC1 and XRCC1 favorable genotypes (0 versus ≥ 1 favorable genotypes, HR=2.42; p=0.02). After adjustment on tumor stage, lymph node ratio and differentiation grade, multivariate analysis showed that combining ERCC1 and XRCC1 genotypes gave a p value slightly above the threshold for statistical significance (HR=2.03; p=0.06), which was lower than for tumor stage, lymph node ratio or differentiation grade. CONCLUSION The association of ERCC1 and XRCC1 polymorphisms may influence the prognosis of stage III colon cancer patients treated with FOLFOX adjuvant chemotherapy. Yet, these findings need to be confirmed in independent prospective studies.
Collapse
Affiliation(s)
- Aziz Zaanan
- 1. INSERM, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 2. UPMC Univ Paris 06, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 3. Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris, France; ; 4. Paris Sorbonne Cité, University of Paris Descartes, Paris, France; ; 5. Cancer Research Personalized Medicine (CARPEM), European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Cécile Dalban
- 6. Department of Biostatistics and Epidemiology, EA4184, Georges François Leclerc Center, Dijon, France
| | - Jean-François Emile
- 7. Department of Pathology, Ambroise Paré Hospital, AP-HP, Boulogne, France; ; 8. EA4340, University of Versailles, Saint-Quentin-en-Yvelines, Versailles, France
| | - Hélène Blons
- 4. Paris Sorbonne Cité, University of Paris Descartes, Paris, France; ; 9. Department of Biology, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Jean-François Fléjou
- 1. INSERM, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 2. UPMC Univ Paris 06, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 10. Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - Claire Goumard
- 1. INSERM, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 2. UPMC Univ Paris 06, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France
| | - Melek Istanbullu
- 1. INSERM, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 2. UPMC Univ Paris 06, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France
| | - Claire Calmel
- 1. INSERM, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 2. UPMC Univ Paris 06, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France
| | - Khalid Alhazmi
- 11. Department of Pathology, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Pierre Validire
- 12. Department of Pathology, Mutualiste Montsouris Institute, Paris, France
| | - Christophe Louvet
- 13. Department of Medical Oncology, Mutualiste Montsouris Institute, Paris, France
| | - Aimery de Gramont
- 14. Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - Pierre Laurent-Puig
- 4. Paris Sorbonne Cité, University of Paris Descartes, Paris, France; ; 5. Cancer Research Personalized Medicine (CARPEM), European Georges Pompidou Hospital, AP-HP, Paris, France; ; 9. Department of Biology, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Julien Taïeb
- 3. Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris, France; ; 4. Paris Sorbonne Cité, University of Paris Descartes, Paris, France; ; 5. Cancer Research Personalized Medicine (CARPEM), European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Françoise Praz
- 1. INSERM, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France; ; 2. UPMC Univ Paris 06, UMR_S 938, Saint-Antoine Research Centre, F-75012, Paris, France
| |
Collapse
|
89
|
Nagel ZD, Chaim IA, Samson LD. Inter-individual variation in DNA repair capacity: a need for multi-pathway functional assays to promote translational DNA repair research. DNA Repair (Amst) 2014; 19:199-213. [PMID: 24780560 DOI: 10.1016/j.dnarep.2014.03.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Why does a constant barrage of DNA damage lead to disease in some individuals, while others remain healthy? This article surveys current work addressing the implications of inter-individual variation in DNA repair capacity for human health, and discusses the status of DNA repair assays as potential clinical tools for personalized prevention or treatment of disease. In particular, we highlight research showing that there are significant inter-individual variations in DNA repair capacity (DRC), and that measuring these differences provides important biological insight regarding disease susceptibility and cancer treatment efficacy. We emphasize work showing that it is important to measure repair capacity in multiple pathways, and that functional assays are required to fill a gap left by genome wide association studies, global gene expression and proteomics. Finally, we discuss research that will be needed to overcome barriers that currently limit the use of DNA repair assays in the clinic.
Collapse
Affiliation(s)
- Zachary D Nagel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Isaac A Chaim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leona D Samson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
90
|
Horton JK, Stefanick DF, Prasad R, Gassman NR, Kedar PS, Wilson SH. Base excision repair defects invoke hypersensitivity to PARP inhibition. Mol Cancer Res 2014; 12:1128-39. [PMID: 24770870 DOI: 10.1158/1541-7786.mcr-13-0502] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED PARP-1 is important for the recognition of both endogenous and exogenous DNA damage, and binds to DNA strand breaks including intermediates of base excision repair (BER). Once DNA-bound, PARP-1 becomes catalytically activated synthesizing PAR polymers onto itself and other repair factors (PARylation). As a result, BER repair proteins such as XRCC1 and DNA polymerase β (pol β) are more efficiently and rapidly recruited to sites of DNA damage. In the presence of an inhibitor of PARP activity (PARPi), PARP-1 binds to sites of DNA damage, but PARylation is prevented. BER enzyme recruitment is hindered, but binding of PARP-1 to DNA is stabilized, impeding DNA repair and leading to double-strand DNA breaks (DSB). Deficiencies in pol β(-/-) and Xrcc1(-/-) cells resulted in hypersensitivity to the PARP inhibitor 4-AN and reexpression of pol β or XRCC1, in these contexts, reversed the 4-AN hypersensitivity phenotype. BER deficiencies also showed evidence of replication defects that lead to DSB-induced apoptosis upon PARPi treatment. Finally, the clinically relevant PARP inhibitors olaparib and veliparib also exhibited hypersensitivity in both pol β(-/-) and Xrcc1(-/-) BER-deficient cells. These results reveal heightened sensitivity to PARPi as a function of BER deficiency. IMPLICATIONS BER deficiency represents a new therapeutic opportunity to enhance PARPi efficacy.
Collapse
Affiliation(s)
- Julie K Horton
- Laboratory of Structural Biology, NIEHS, NIH, Research Triangle Park, North Carolina
| | - Donna F Stefanick
- Laboratory of Structural Biology, NIEHS, NIH, Research Triangle Park, North Carolina
| | - Rajendra Prasad
- Laboratory of Structural Biology, NIEHS, NIH, Research Triangle Park, North Carolina
| | - Natalie R Gassman
- Laboratory of Structural Biology, NIEHS, NIH, Research Triangle Park, North Carolina
| | - Padmini S Kedar
- Laboratory of Structural Biology, NIEHS, NIH, Research Triangle Park, North Carolina
| | - Samuel H Wilson
- Laboratory of Structural Biology, NIEHS, NIH, Research Triangle Park, North Carolina
| |
Collapse
|
91
|
Prasad R, Horton JK, Chastain PD, Gassman NR, Freudenthal BD, Hou EW, Wilson SH. Suicidal cross-linking of PARP-1 to AP site intermediates in cells undergoing base excision repair. Nucleic Acids Res 2014; 42:6337-51. [PMID: 24771347 PMCID: PMC4041460 DOI: 10.1093/nar/gku288] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is an abundant nuclear enzyme in mammalian cells. The enzyme synthesizes polymers of ADP-ribose from the coenzyme NAD+ and plays multifaceted roles in cellular responses to genotoxic stress, including DNA repair. It had been shown that mouse fibroblasts treated with a DNA methylating agent in combination with a PARP inhibitor exhibit higher cytotoxicity than cells treated with methylating agent alone. This lethality of the PARP inhibitor is dependent on apurinic/apyrimidinic (AP) sites in the DNA and the presence of PARP-1. Here, we show that purified PARP-1 is capable of forming a DNA-protein cross-link (DPC) by covalently attaching to the AP site. This DPC formation is specific to the presence of the natural AP site in DNA and is accompanied by a single-strand DNA incision. Cellular studies confirm the formation of PARP-1 DPCs during alkylating agent-induced base excision repair (BER) and formation of DPCs is enhanced by a PARP inhibitor. Using an N-terminal and C-terminal truncated PARP-1 we show that a polypeptide fragment comprising the zinc 3 and BRCT sub-domains is sufficient for DPC formation. The covalent attachment of PARP-1 to AP site-containing DNA appears to be a suicidal event when BER is overwhelmed or disrupted.
Collapse
Affiliation(s)
- Rajendra Prasad
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Julie K Horton
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Paul D Chastain
- William Carey University College of Osteopathic Medicine, 498 Tuscan Avenue, Hattiesburg, MS 39401, USA
| | - Natalie R Gassman
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Bret D Freudenthal
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Esther W Hou
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Samuel H Wilson
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| |
Collapse
|
92
|
Liu C, Yin Q, Jiao G, Zhou X, Ying M, Gao W, Hu J, Wei L, Wang N, Wang Y. Association between the XRCC1 Arg399Gln polymorphism and head and neck cancer susceptibility: a meta-analysis based on case-control studies. DNA Cell Biol 2014; 33:378-87. [PMID: 24694255 DOI: 10.1089/dna.2013.2269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Published data regarding the association between the XRCC1 Arg399Gln polymorphism and head and neck cancer (HNC) susceptibility showed inconsistent results. This meta-analysis of eligible literatures was performed to draw a more precise estimation of the relationship. We systematically searched PubMed, Embase, and Web of Science with a time limit of Oct 28, 2013. Summary odds ratios (ORs) with 95% CIs were used to assess the strength of association between XRCC1 Arg399Gln polymorphism and HNC susceptibility using random-effect model. A total of 27 case-control studies including 5942 cases and 9041 controls were included for analysis. Meta-analysis of total studies showed that the XRCC1 Arg399Gln variant carriers were not susceptible to HNC (AA vs. GG: OR=0.92, 95% CI=0.77-1.11; AG vs. GG: OR=1.05, 95% CI=0.76-1.44; the dominant model AA+AG vs. GG: OR=1.00, 95% CI=0.78-1.29; the recessive model AA vs. AG+GG: OR=0.91, 95% CI=0.71-1.16). Further, subgroup analyses by ethnicity and source of controls did not identify any significant associations of XRCC1 Arg399Gln polymorphism with head and neck susceptibility in any populations. Our meta-analysis suggested that the XRCC1 Arg399Gln polymorphism was not a risk factor for HNC susceptibility.
Collapse
Affiliation(s)
- Chuan Liu
- 1 Department of Oncology, Changhai Hospital, Second Military Medical University , Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Hombach-Klonisch S, Natarajan S, Thanasupawat T, Medapati M, Pathak A, Ghavami S, Klonisch T. Mechanisms of therapeutic resistance in cancer (stem) cells with emphasis on thyroid cancer cells. Front Endocrinol (Lausanne) 2014; 5:37. [PMID: 24723911 PMCID: PMC3971176 DOI: 10.3389/fendo.2014.00037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/11/2014] [Indexed: 12/11/2022] Open
Abstract
The two main reasons for death of cancer patients, tumor recurrence and metastasis, are multi-stage cellular processes that involve increased cell plasticity and coincide with elevated resistance to anti-cancer treatments. Epithelial-to-mesenchymal transition (EMT) is a key contributor to metastasis in many cancer types, including thyroid cancer and is known to confer stem cell-like properties onto cancer cells. This review provides an overview of molecular mechanisms and factors known to contribute to cancer cell plasticity and capable of enhancing cancer cell resistance to radio- and chemotherapy. We elucidate the role of DNA repair mechanisms in contributing to therapeutic resistance, with a special emphasis on thyroid cancer. Next, we explore the emerging roles of autophagy and damage-associated molecular pattern responses in EMT and chemoresistance in tumor cells. Finally, we demonstrate how cancer cells, including thyroid cancer cells, can highjack the oncofetal nucleoprotein high-mobility group A2 to gain increased transformative cell plasticity, prevent apoptosis, and enhance metastasis of chemoresistant tumor cells.
Collapse
Affiliation(s)
- Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Suchitra Natarajan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | | | - Manoj Medapati
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Alok Pathak
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Department of Surgery, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Institute of Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
- Department of Surgery, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Institute of Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
94
|
Arian D, Hedayati M, Zhou H, Bilis Z, Chen K, DeWeese TL, Greenberg MM. Irreversible inhibition of DNA polymerase β by small-molecule mimics of a DNA lesion. J Am Chem Soc 2014; 136:3176-83. [PMID: 24517090 DOI: 10.1021/ja411733s] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Abasic sites are ubiquitous DNA lesions that are mutagenic and cytotoxic but are removed by the base excision repair pathway. DNA polymerase β carries out two of the four steps during base excision repair, including a lyase reaction that removes the abasic site from DNA following incision of its 5'-phosphate. DNA polymerase β is overexpressed in cancer cells and is a potential anticancer target. Recently, DNA oxidized abasic sites that are produced by potent antitumor agents were shown to inactivate DNA polymerase β. A library of small molecules whose structures were inspired by the oxidized abasic sites was synthesized and screened for the ability to irreversibly inhibit DNA polymerase β. One candidate (3a) was examined more thoroughly, and modification of its phosphate backbone led to a molecule that irreversibly inactivates DNA polymerase β in solution (IC50 ≈ 21 μM), and inhibits the enzyme's lyase activity in cell lysates. A bisacetate analogue is converted in cell lysates to 3a. The bisacetate is more effective in cell lysates, more cytotoxic in prostate cancer cells than 3a and potentiates the cytotoxicity of methyl methanesulfonate between 2- and 5-fold. This is the first example of an irreversible inhibitor of the lyase activity of DNA polymerase β that works synergistically with a DNA damaging agent.
Collapse
Affiliation(s)
- Dumitru Arian
- Department of Chemistry, Johns Hopkins University , 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | | | | | | | | | | | | |
Collapse
|
95
|
He F, Li G, Man L, Liu N. Association between X-ray repair cross-complementing group 1 Arg194Trp polymorphism and prostate cancer risk. Tumour Biol 2014; 35:4041-5. [DOI: 10.1007/s13277-013-1528-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/06/2013] [Indexed: 10/25/2022] Open
|
96
|
Vriend LEM, Jasin M, Krawczyk PM. Assaying break and nick-induced homologous recombination in mammalian cells using the DR-GFP reporter and Cas9 nucleases. Methods Enzymol 2014; 546:175-91. [PMID: 25398341 PMCID: PMC4408992 DOI: 10.1016/b978-0-12-801185-0.00009-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Thousands of DNA breaks occur daily in mammalian cells, including potentially tumorigenic double-strand breaks (DSBs) and less dangerous but vastly more abundant single-strand breaks (SSBs). The majority of SSBs are quickly repaired, but some can be converted to DSBs, posing a threat to the integrity of the genome. Although SSBs are usually repaired by dedicated pathways, they can also trigger homologous recombination (HR), an error-free pathway generally associated with DSB repair. While HR-mediated DSB repair has been extensively studied, the mechanisms of HR-mediated SSB repair are less clear. This chapter describes a protocol to investigate SSB-induced HR in mammalian cells employing the DR-GFP reporter, which has been widely used in DSB repair studies, together with an adapted bacterial CRISPR/Cas system.
Collapse
Affiliation(s)
- Lianne E M Vriend
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, USA; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, USA.
| | - Przemek M Krawczyk
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, USA; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
97
|
Zhou Y, Zhou W, Liu Q, Fan Z, Yang Z, Tu Q, Li L, Liu H. XRCC1 R399Q polymorphism and risk of normal tissue injury after radiotherapy in breast cancer patients. Tumour Biol 2013; 35:21-5. [PMID: 24292986 DOI: 10.1007/s13277-013-0990-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 07/01/2013] [Indexed: 01/04/2023] Open
Abstract
Radiotherapy is an important weapon in the treatment of breast cancer, but normal tissue injury after radiotherapy can be a threat for patients. Genetic markers conferring the ability to identify hyper-sensitive patients at risk of normal tissue injury in advance would considerably improve therapy. Association studies on genetic variation and occurrence of normal tissue injury can help us identify such markers, but previous studies on the association between XRCC1 R399Q polymorphism and risk of normal tissue injury after radiotherapy in breast cancer patients report conflicting findings. We performed a meta-analysis to comprehensively evaluate the association between XRCC1 R399Q polymorphism and risk of normal tissue injury after radiotherapy in breast cancer patients. The pooled odds ratios (ORs) with their 95% confidence interval (95% CIs) were calculated to assess the strength of the association. Fourteen case-control studies with a total of 2,448 breast cancer cases were finally included into the meta-analysis. Overall, XRCC1 R399Q polymorphism was significantly associated with increased risk of normal tissue injury after radiotherapy under all three models (for QQ versus RR: fixed-effects OR = 1.06, 95% CI 1.00-1.13, P = 0.050; for RQ versus RR: fixed-effects OR = 1.05, 95% CI 1.00-1.10, P = 0.047; for QQ/RQ versus RR: fixed-effects OR = 1.26, 95% CI 1.01-1.58, P = 0.041). The meta-analysis suggests that XRCC1 R399Q polymorphism was significantly associated with increased risk of normal tissue injury after radiotherapy in breast cancer patients, and XRCC1 R399Q polymorphism is a genetic marker of normal tissue injury after radiotherapy in breast cancer patients.
Collapse
Affiliation(s)
- Yingying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Redrejo-Rodríguez M, Salas ML. Repair of base damage and genome maintenance in the nucleo-cytoplasmic large DNA viruses. Virus Res 2013; 179:12-25. [PMID: 24184318 DOI: 10.1016/j.virusres.2013.10.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 10/21/2013] [Accepted: 10/21/2013] [Indexed: 11/27/2022]
Abstract
Among the DNA viruses, the so-called nucleo-cytoplasmic large DNA viruses (NCLDV) constitute a monophyletic group that currently consists of seven families of viruses infecting a very broad variety of eukaryotes, from unicellular marine protists to humans. Many recent papers have analyzed the sequence and structure of NCLDV genomes and their phylogeny, providing detailed analysis about their genomic structure and evolutionary history and proposing their inclusion in a new viral order named Megavirales that, according to some authors, should be considered as a fourth domain of life, aside from Bacteria, Archaea and Eukarya. The maintenance of genetic information protected from environmental attacks and mutations is essential not only for the survival of cellular organisms but also viruses. In cellular organisms, damaged DNA bases are removed in two major repair pathways: base excision repair (BER) and nucleotide incision repair (NIR) that constitute the major pathways responsible for repairing most endogenous base lesions and abnormal bases in the genome by precise repair procedures. Like cells, many NCLDV encode proteins that might constitute viral DNA repair pathways that would remove damages through BER/NIR pathways. However, the molecular mechanisms and, specially, the biological roles of those viral repair pathways have not been deeply addressed in the literature so far. In this paper, we review viral-encoded BER proteins and the genetic and biochemical data available about them. We propose and discuss probable viral-encoded DNA repair mechanisms and pathways, as compared with the functional and molecular features of known homologs proteins.
Collapse
Affiliation(s)
- Modesto Redrejo-Rodríguez
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | - María L Salas
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
99
|
Jacob KD, Noren Hooten N, Tadokoro T, Lohani A, Barnes J, Evans MK. Alzheimer's disease-associated polymorphisms in human OGG1 alter catalytic activity and sensitize cells to DNA damage. Free Radic Biol Med 2013; 63:115-25. [PMID: 23684897 PMCID: PMC3767440 DOI: 10.1016/j.freeradbiomed.2013.05.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 04/09/2013] [Accepted: 05/06/2013] [Indexed: 12/14/2022]
Abstract
Brain tissues from Alzheimer's disease (AD) patients show increased levels of oxidative DNA damage and 7,8-dihydro-8-oxoguanine (8-oxoG) accumulation. In humans, the base excision repair protein 8-oxoguanine-DNA glycosylase (OGG1) is the major enzyme that recognizes and excises the mutagenic DNA base lesion 8-oxoG. Recently, two polymorphisms of OGG1, A53T and A288V, have been identified in brain tissues of AD patients, but little is known about how these polymorphisms may contribute to AD. We characterized the A53T and A288V polymorphic variants and detected a significant reduction in the catalytic activity for both proteins in vitro and in cells. Additionally, the A53T polymorphism has decreased substrate binding, whereas the A288V polymorphism has reduced AP lyase activity. Both variants have decreased binding to known OGG1 binding partners PARP-1 and XRCC1. We found that OGG1(-/-) cells expressing A53T and A288V OGG1 were significantly more sensitive to DNA damage and had significantly decreased survival. Our results provide both biochemical and cellular evidence that A53T and A288V polymorphic proteins have deficiencies in catalytic and protein-binding activities that could be related to the increase in oxidative damage to DNA found in AD brains.
Collapse
Affiliation(s)
- Kimberly D Jacob
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224-6825, USA
| | | | | | | | | | | |
Collapse
|
100
|
Horton JK, Wilson SH. Strategic Combination of DNA-Damaging Agent and PARP Inhibitor Results in Enhanced Cytotoxicity. Front Oncol 2013; 3:257. [PMID: 24137565 PMCID: PMC3786324 DOI: 10.3389/fonc.2013.00257] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/11/2013] [Indexed: 01/01/2023] Open
Abstract
PARP inhibitors (PARPi) are under clinical trial for combination cancer chemotherapy. In the presence of a PARPi, PARP-1 binds DNA strand breaks but cannot produce poly(ADP-ribose) polymers or undergo auto-poly(ADP-ribosyl)ation. DNA binding is persistent, hindering DNA repair. Methylated bases formed as a result of cellular exposure to DNA-methylating agents are repaired by DNA polymerase β (pol β)-dependent base excision repair (BER) producing a 5′-deoxyribose phosphate (5′-dRP) repair intermediate. PARP-1 binds and is activated by the 5′-dRP, and PARPi-mediated sensitization to methylating agents is considerable, especially in pol β-deficient cells. Cells deficient in the BER factor XRCC1 are less sensitized by PARPi than are wild-type cells. PARPi sensitization is reduced in cells expressing forms of XRCC1 deficient in interaction with either pol β or PARP-1. In contrast, agents producing oxidative DNA damage and 3′- rather than 5′-repair intermediates are modestly PARPi sensitized. We summarize PARPi experiments in mouse fibroblasts and confirm the importance of the 5′-dRP repair intermediate and functional pol β and XRCC1 proteins. Understanding the chemistry of repair is key to enhancing the clinical success of PARPi.
Collapse
Affiliation(s)
- Julie K Horton
- Laboratory of Structural Biology, NIEHS, National Institutes of Health , Research Triangle Park, NC , USA
| | | |
Collapse
|