1
|
Chen Y, Ding K, Zheng S, Gao S, Xu X, Wu H, Zhou F, Wang Y, Xu J, Wang C, Ling C, Xu J, Wang L, Wu Q, Giamas G, Chen G, Zhang J, Yi C, Ji J. Post-translational modifications in DNA damage repair: mechanisms underlying temozolomide resistance in glioblastoma. Oncogene 2025:10.1038/s41388-025-03454-5. [PMID: 40419791 DOI: 10.1038/s41388-025-03454-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/04/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025]
Abstract
Temozolomide (TMZ) resistance is one of the critical factors contributing to the poor prognosis of glioblastoma (GBM). As a first-line chemotherapeutic agent for GBM, TMZ exerts its cytotoxic effects through DNA alkylation. However, its therapeutic efficacy is significantly compromised by enhanced DNA damage repair (DDR) mechanisms in GBM cells. Although several DDR-targeting drugs have been developed, their clinical outcomes remain suboptimal. Post-translational modifications (PTMs) in GBM cells play a pivotal role in maintaining the genomic stability of DDR mechanisms, including methylguanine-DNA methyltransferase-mediated repair, DNA mismatch repair dysfunction, base excision repair, and double-strand break repair. This review focuses on elucidating the regulatory roles of PTMs in the intrinsic mechanisms underlying TMZ resistance in GBM. Furthermore, we explore the feasibility of enhancing TMZ-induced cytotoxicity by targeting PTM-related enzymatic to disrupt key steps in PTM-mediated DDR pathways. By integrating current preclinical insights and clinical challenges, this work highlights the potential of modulating PTM-driven networks as a novel therapeutic strategy to overcome TMZ resistance and improve treatment outcomes for GBM patients.
Collapse
Affiliation(s)
- Yike Chen
- Department of Plastic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kaikai Ding
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuyu Zheng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Songting Gao
- Guali Branch of the First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Xiaohui Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Fengqi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Yongjie Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jinfang Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Chun Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Chenhan Ling
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jing Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, UK
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration Zhejiang University, Hangzhou, Zhejiang, China.
| | - Chenggang Yi
- Department of Plastic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jianxiong Ji
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Schwarzenbach C, Rinke J, Vilar JB, Sallbach J, Tatsch L, Schmidt A, Schöneis A, Rasenberger B, Kaina B, Tomicic MT, Christmann M. Therapy-induced senescence of glioblastoma cells is determined by the p21 CIP1-CDK1/2 axis and does not require activation of DREAM. Cell Death Dis 2025; 16:357. [PMID: 40319068 PMCID: PMC12049523 DOI: 10.1038/s41419-025-07651-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/07/2025]
Abstract
Therapy-induced senescence (TIS) is a major challenge in cancer therapy as senescent cancer cells provoke local and systemic inflammation and might be the cause of recurrences. Elucidation of pathways leading to TIS is of utmost importance for establishing strategies to counteract this. Previously we have shown that temozolomide (TMZ), an alkylating drug used forefront in glioma therapy, causes majorly cellular senescence, which is triggered by the primary damage O6-methylguanine, activating the mismatch repair dependent ATR/ATM-CHK1/CHK2-p53 damage response pathway. The downstream pathways leading to TIS remained to be explored. Here, we show that TMZ-induced TIS in glioma cells does not require activation of the DREAM complex, but is bound on a G2-specific response. We show that the CDK inhibitor p21CIP1 does not interact with CDK4, but with CDK1 and CDK2 causing abrogation of the B-Myb and FOXM1-signaling pathway and subsequently arrest of cells in the G2-phase. The induced G2-arrest is incomplete as DNA synthesis can be resumed leading to endoreduplications. This process, which is inhibited by the CDK4-blocking drug palbociclib, is preceded by reactivation of the G1/S-specific E2F1-signaling pathway due to lack of functional DREAM activation. These findings provide an explanation for the polyploidization and giant cell phenotype of anticancer drug-induced senescent cells. Incomplete DREAM activation may also explain the observation that downregulation of DNA repair is a transient phenomenon, which goes along with the entrance of cells into the senescent state.
Collapse
Affiliation(s)
- Christian Schwarzenbach
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Justus Rinke
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Juliana B Vilar
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jason Sallbach
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Larissa Tatsch
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ariane Schmidt
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Anna Schöneis
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Birgit Rasenberger
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Bernd Kaina
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Maja T Tomicic
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Markus Christmann
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
3
|
Duan J, Chen T, Li Q, Zhang Y, Lu T, Xue J, Sun Y, Gao L, Zhang Y. Protein arginine methyltransferase 6 enhances immune checkpoint blockade efficacy via the STING pathway in MMR-proficient colorectal cancer. J Immunother Cancer 2025; 13:e010639. [PMID: 40086819 PMCID: PMC11907083 DOI: 10.1136/jitc-2024-010639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The emergence of immunotherapy has revolutionized the paradigm of cancer treatment with immune checkpoint blockades (ICB) in solid cancers, including colorectal cancer (CRC). However, only a small subset of CRC patients harboring deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) benefits from ICB therapy. A very limited response to ICB therapy has been achieved in MMR-proficient CRC, representing a significant challenge limiting the clinical application of immunotherapy. MMR is the critical DNA repair pathway that maintains genomic integrity by correcting DNA mismatches, which is mediated by the MutSα or MutSβ complex consisting of MSH2 with MSH6 and MSH3, respectively. Given that MMR status directs effective immune response, we sought to determine whether targeting MMR capacity boosts ICB efficacy. METHODS Azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC and xenograft model were used to evaluate the function of PRMT6 and response to PRMT6 inhibitor EPZ020411 and combination therapy of PD1 and EPZ020411. Biochemical assays were performed to elucidate the underlying mechanism of PRMT6-mediated MSH2 methylation and immune evasion. RESULTS We have identified PRMT6 as a crucial regulator of MMR capacity via MSH2 dimethylation at R171 and R219. Such a modification abrogates its MMR capacity and prevents the recruitment of MSH3 and MSH6. PRMT6 loss or inhibition triggers cytosolic DNA accumulation and cGAS-STING signaling activation, leading to enhanced immune response in PRMT6-deficient colon tumors or xenografts. Pharmacological inhibition of PRMT6 using EPZ020411 promotes mutagenesis and destabilizes MutSα or MutSβ assembly, and prolonged EPZ020411 exposure maintains an MSI-like phenotype in microsatellite stability (MSS) cells. EPZ020411 treatment sensitizes ICB efficacy of MSS cells, but not MSI cells in vivo. Similar effects have been observed in MSS colon tumors induced by AOM/DSS. CONCLUSIONS Our study provides a preclinical proof of concept to overcome resistance to immunotherapy by targeting PRMT6 in CRC with MSS.
Collapse
Affiliation(s)
- Jinlin Duan
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pathology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tao Chen
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Qiwei Li
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai, China
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ting Lu
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Junyan Xue
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yang Sun
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yonglong Zhang
- Laboratory of Targeted Therapy and Precision Medicine, Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
- Department of General Surgery, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Zhang H, Lu W, Qiu L, Li S, Qiu L, He M, Chen X, Wang J, Fang J, Zhong C, Lan M, Xu X, Zhou Y. Circ_0025373 inhibits carbon black nanoparticles-induced malignant transformation of human bronchial epithelial cells by affecting DNA damage through binding to MSH2. ENVIRONMENT INTERNATIONAL 2024; 191:109001. [PMID: 39284259 DOI: 10.1016/j.envint.2024.109001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
Carbon black nanoparticles (CBNPs) have been demonstrated to induce DNA damage in epithelial cells. However, the potential of the damage to initiate carcinogenesis and the underlying mechanism remain poorly understood. Therefore, we constructed an in vitro model of malignant transformation of human bronchial epithelial cells (16HBE-T) by treating 40 μg/mL CBNPs for 120 passages. We observed tumor-like transformation and sustained DNA damage. Using transcriptome sequencing and RIP-seq, we identified the overexpression of the critical DNA mismatch repair genes MutS homolog 2 (MSH2) and its related circular RNA, circ_0025373, in the 16HBE-T cells. Mechanistically, circ_0025373 was found to inhibit DNA damage by binding to MSH2, thereby modifying its expression and influencing its nuclear and cytoplasmic distribution, which lead to inhibition of CBNP-induced malignant transformation of human bronchial epithelial cells. Our findings provide novel evidence on the carcinogenicity of CBNPs, and offer biological insights into the potential epigenetic regulation and potential therapeutic targets for lung carcinogenesis.
Collapse
Affiliation(s)
- Han Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenfeng Lu
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Lan Qiu
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Saifeng Li
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Liqiu Qiu
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Mengnan He
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Xintong Chen
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiajing Wang
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Jingwen Fang
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Chenghui Zhong
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Meiqi Lan
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaole Xu
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Yun Zhou
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
5
|
Ma Y, Mu X, Gao R, Zhang Y, Geng Y, Chen X, Yin X, Li F, He J. Maternal exposure to dibutyl phthalate regulates MSH6 crotonylation to impair homologous recombination in fetal oocytes. JOURNAL OF HAZARDOUS MATERIALS 2023; 455:131540. [PMID: 37167869 DOI: 10.1016/j.jhazmat.2023.131540] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/12/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Homologous recombination (HR) during early oogenesis repairs programmed double-strand breaks (DSBs) to ensure female fertility and offspring health. The exposure of fetal ovaries to endocrine disrupting chemicals (EDCs) can cause reproductive disorders in the adulthood. The EDC dibutyl phthalate (DBP) is widely distributed in flexible plastic products, leading to ubiquitous human exposure. Here, we report that maternal exposure to DBP caused gross aberrations in meiotic prophase I of fetal oocytes, including delayed progression, impaired DNA damage response, uncoupled localization of DMC1 and RAD51, and decreased HR. However, programmed DSBs were efficiently repaired. DBP exposure negatively regulated lysine crotonylation (Kcr) of MSH6. Similar meiotic defects were observed in fetal ovaries with targeted disruption of Msh6, and mutation of K544cr of MSH6 impaired its association with Ku70, thereby promoting non-homologous end joining (NHEJ) and inhibiting HR. Unlike mature F1 females, F2 female mice exhibited premature follicular activation, precocious puberty, and anxiety-like behaviors. Therefore, DBP can influence early meiotic events, and Kcr of MSH6 may regulate preferential induction of HR or NHEJ for DNA repair during meiosis.
Collapse
Affiliation(s)
- Yidan Ma
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Xinyi Mu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Rufei Gao
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan Zhang
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuemei Chen
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Yin
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Fangfang Li
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Junlin He
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
6
|
Stoler-Barak L, Harris E, Peres A, Hezroni H, Kuka M, Di Lucia P, Grenov A, Gurwicz N, Kupervaser M, Yip BH, Iannacone M, Yaari G, Crispino JD, Shulman Z. B cell class switch recombination is regulated by DYRK1A through MSH6 phosphorylation. Nat Commun 2023; 14:1462. [PMID: 36927854 PMCID: PMC10020581 DOI: 10.1038/s41467-023-37205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Protection from viral infections depends on immunoglobulin isotype switching, which endows antibodies with effector functions. Here, we find that the protein kinase DYRK1A is essential for B cell-mediated protection from viral infection and effective vaccination through regulation of class switch recombination (CSR). Dyrk1a-deficient B cells are impaired in CSR activity in vivo and in vitro. Phosphoproteomic screens and kinase-activity assays identify MSH6, a DNA mismatch repair protein, as a direct substrate for DYRK1A, and deletion of a single phosphorylation site impaired CSR. After CSR and germinal center (GC) seeding, DYRK1A is required for attenuation of B cell proliferation. These findings demonstrate DYRK1A-mediated biological mechanisms of B cell immune responses that may be used for therapeutic manipulation in antibody-mediated autoimmunity.
Collapse
Affiliation(s)
- Liat Stoler-Barak
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ethan Harris
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ayelet Peres
- Faculty of Engineering, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Hadas Hezroni
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Mirela Kuka
- Vita-Salute San Raffaele University and Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Di Lucia
- Vita-Salute San Raffaele University and Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Amalie Grenov
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Neta Gurwicz
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Meital Kupervaser
- De Botton Institute for Proteomics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Bon Ham Yip
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Matteo Iannacone
- Vita-Salute San Raffaele University and Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gur Yaari
- Faculty of Engineering, Bar Ilan University, Ramat Gan, 52900, Israel
| | - John D Crispino
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ziv Shulman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
7
|
Amar-Schwartz A, Ben Hur V, Jbara A, Cohen Y, Barnabas GD, Arbib E, Siegfried Z, Mashahreh B, Hassouna F, Shilo A, Abu-Odeh M, Berger M, Wiener R, Aqeilan R, Geiger T, Karni R. S6K1 phosphorylates Cdk1 and MSH6 to regulate DNA repair. eLife 2022; 11:79128. [PMID: 36189922 PMCID: PMC9529248 DOI: 10.7554/elife.79128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/26/2022] [Indexed: 11/22/2022] Open
Abstract
The mTORC1 substrate, S6 Kinase 1 (S6K1), is involved in the regulation of cell growth, ribosome biogenesis, glucose homeostasis, and adipogenesis. Accumulating evidence has suggested a role for mTORC1 signaling in the DNA damage response. This is mostly based on the findings that mTORC1 inhibitors sensitized cells to DNA damage. However, a direct role of the mTORC1-S6K1 signaling pathway in DNA repair and the mechanism by which this signaling pathway regulates DNA repair is unknown. In this study, we discovered a novel role for S6K1 in regulating DNA repair through the coordinated regulation of the cell cycle, homologous recombination (HR) DNA repair (HRR) and mismatch DNA repair (MMR) mechanisms. Here, we show that S6K1 orchestrates DNA repair by phosphorylation of Cdk1 at serine 39, causing G2/M cell cycle arrest enabling homologous recombination and by phosphorylation of MSH6 at serine 309, enhancing MMR. Moreover, breast cancer cells harboring RPS6KB1 gene amplification show increased resistance to several DNA damaging agents and S6K1 expression is associated with poor survival of breast cancer patients treated with chemotherapy. Our findings reveal an unexpected function of S6K1 in the DNA repair pathway, serving as a tumorigenic barrier by safeguarding genomic stability. Damage to the DNA in our cells can cause harmful changes that, if unchecked, can lead to the development of cancer. To help prevent this, cellular mechanisms are in place to repair defects in the DNA. A particular process, known as the mTORC1-S6K1 pathway is suspected to be important for repair because when this pathway is blocked, cells become more sensitive to DNA damage. It is still unknown how the various proteins involved in the mTORC1-S6K1 pathway contribute to repairing DNA. One of these proteins, S6K1, is an enzyme involved in coordinating cell growth and survival. The tumor cells in some forms of breast cancer produce more of this protein than normal, suggesting that S6K1 benefits these cells’ survival. However, it is unclear exactly how the enzyme does this. Amar-Schwartz, Ben-Hur, Jbara et al. studied the role of S6K1 using genetically manipulated mouse cells and human cancer cells. These experiments showed that the protein interacts with two other proteins involved in DNA repair and activates them, regulating two different repair mechanisms and protecting cells against damage. These results might explain why some breast cancer tumors are resistant to radiotherapy and chemotherapy treatments, which aim to kill tumor cells by damaging their DNA. If this is the case, these findings could help clinicians choose more effective treatment options for people with cancers that produce additional S6K1. In the future, drugs that block the activity of the enzyme could make cancer cells more susceptible to chemotherapy.
Collapse
Affiliation(s)
- Adi Amar-Schwartz
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Vered Ben Hur
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Amina Jbara
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yuval Cohen
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Georgina D Barnabas
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eliran Arbib
- Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Zahava Siegfried
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Bayan Mashahreh
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Fouad Hassouna
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Asaf Shilo
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Mohammad Abu-Odeh
- Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Michael Berger
- Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Reuven Wiener
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Rami Aqeilan
- Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
8
|
Firnau MB, Brieger A. CK2 and the Hallmarks of Cancer. Biomedicines 2022; 10:1987. [PMID: 36009534 PMCID: PMC9405757 DOI: 10.3390/biomedicines10081987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Casein kinase 2 (CK2) is commonly dysregulated in cancer, impacting diverse molecular pathways. CK2 is a highly conserved serine/threonine kinase, constitutively active and ubiquitously expressed in eukaryotes. With over 500 known substrates and being estimated to be responsible for up to 10% of the human phosphoproteome, it is of significant importance. A broad spectrum of diverse types of cancer cells has been already shown to rely on disturbed CK2 levels for their survival. The hallmarks of cancer provide a rationale for understanding cancer's common traits. They constitute the maintenance of proliferative signaling, evasion of growth suppressors, resisting cell death, enabling of replicative immortality, induction of angiogenesis, the activation of invasion and metastasis, as well as avoidance of immune destruction and dysregulation of cellular energetics. In this work, we have compiled evidence from the literature suggesting that CK2 modulates all hallmarks of cancer, thereby promoting oncogenesis and operating as a cancer driver by creating a cellular environment favorable to neoplasia.
Collapse
Affiliation(s)
| | - Angela Brieger
- Department of Internal Medicine I, Biomedical Research Laboratory, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
9
|
Suganuma T, Workman JL. MPTAC links alkylation damage signaling to sterol biosynthesis. Redox Biol 2022; 51:102270. [PMID: 35189552 PMCID: PMC8866156 DOI: 10.1016/j.redox.2022.102270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 12/19/2022] Open
Abstract
Overproduction of reactive oxygen species (ROS) drives inflammation and mutagenesis. However, the role of the DNA damage response in immune responses remains largely unknown. Here we found that stabilization of the mismatch repair (MMR) protein MSH6 in response to alkylation damage requires interactions with the molybdopterin synthase associating complex (MPTAC) and Ada2a-containing histone acetyltransferase complex (ATAC). Furthermore, MSH6 promotes sterol biosynthesis via the mevalonate pathway in a MPTAC- and ATAC-dependent manner. MPTAC reduces the source of alkylating agents (ROS). Therefore, the association between MMR proteins, MPTAC, and ATAC promotes anti-inflammation response and reduces alkylating agents. The inflammatory responses measured by xanthine oxidase activity are elevated in Lymphoblastoid Cell Lines (LCLs) from some Fragile X-associated disorders (FXD) patients, suggesting that alkylating agents are increased in these FXD patients. However, MPTAC is disrupted in LCLs from some FXD patients. In LCLs from other FXD patients, interaction between MSH6 and ATAC was lost, destabilizing MSH6. Thus, impairment of MPTAC and ATAC may cause alkylation damage resistance in some FXD patients.
Collapse
Affiliation(s)
- Tamaki Suganuma
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO, 64110, USA.
| | - Jerry L Workman
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO, 64110, USA.
| |
Collapse
|
10
|
Ulreich K, Firnau MB, Tagscherer N, Beyer S, Ackermann A, Plotz G, Brieger A. High Expression of Casein Kinase 2 Alpha Is Responsible for Enhanced Phosphorylation of DNA Mismatch Repair Protein MLH1 and Increased Tumor Mutation Rates in Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14061553. [PMID: 35326704 PMCID: PMC8946085 DOI: 10.3390/cancers14061553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Colorectal cancer (CRC) is associated with DNA mismatch repair (MMR) deficiency. The serine/threonine casein kinase 2 alpha (CK2α) is able to phosphorylate and inhibit MMR protein MLH1 in vitro. This study aimed to analyze the relevance of CK2α for MLH1 phosphorylation in vivo. Around 50% of CRCs were identified to express significantly increased nuclear/cytoplasmic CK2α. High nuclear/cytoplasmic CK2α level could be significantly correlated with reduced 5-year survival outcome of patients, increased MLH1 phosphorylation, and enriched somatic tumor mutation rates. Overall, our study demonstrated, in vivo, that enhanced CK2α leads to an increase of MLH1 phosphorylation, higher tumor mutation rates, and is an unfavorable prognosis for patients. Abstract DNA mismatch repair (MMR) deficiency plays an essential role in the development of colorectal cancer (CRC). We recently demonstrated in vitro that the serine/threonine casein kinase 2 alpha (CK2α) causes phosphorylation of the MMR protein MLH1 at position serine 477, which significantly inhibits the MMR. In the present study, CK2α-dependent MLH1 phosphorylation was analyzed in vivo. Using a cohort of 165 patients, we identified 88 CRCs showing significantly increased nuclear/cytoplasmic CK2α expression, 28 tumors with high nuclear CK2α expression and 49 cases showing a general low CK2α expression. Patients with high nuclear/cytoplasmic CK2α expression demonstrated significantly reduced 5-year survival outcome. By immunoprecipitation and Western blot analysis, we showed that high nuclear/cytoplasmic CK2α expression significantly correlates with increased MLH1 phosphorylation and enriched somatic tumor mutation rates. The CK2α mRNA levels tended to be enhanced in high nuclear/cytoplasmic and high nuclear CK2α-expressing tumors. Furthermore, we identified various SNPs in the promotor region of CK2α, which might cause differential CK2α expression. In summary, we demonstrated that high nuclear/cytoplasmic CK2α expression in CRCs correlates with enhanced MLH1 phosphorylation in vivo and seems to be causative for increased mutation rates, presumably induced by reduced MMR. These observations could provide important new therapeutic targets.
Collapse
|
11
|
Song HR, Kim HK, Kim SG, Lim HJ, Kim HY, Han MK. Changes in the phosphorylation of nucleotide metabolism‑associated proteins by leukemia inhibitory factor in mouse embryonic stem cells. Mol Med Rep 2021; 23:431. [PMID: 33846773 PMCID: PMC8060798 DOI: 10.3892/mmr.2021.12070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/22/2020] [Indexed: 11/05/2022] Open
Abstract
Leukemia inhibitory factor (LIF) is a stem cell growth factor that maintains self‑renewal of mouse embryonic stem cells (mESCs). LIF is a cytokine in the interleukin‑6 family and signals via the common receptor subunit gp130 and ligand‑specific LIF receptor. LIF causes heterodimerization of the LIF receptor and gp130, activating the Janus kinase/STAT and MAPK pathways, resulting in changes in protein phosphorylation. The present study profiled LIF‑mediated protein phosphorylation changes in mESCs via proteomic analysis. mESCs treated in the presence or absence of LIF were analyzed via two‑dimensional differential in‑gel electrophoresis and protein and phosphoprotein staining. Protein identification was performed by matrix‑assisted laser desorption/ionization‑time of flight mass spectrophotometry. Increased phosphorylation of 16 proteins and decreased phosphorylation of 34 proteins in response to LIF treatment was detected. Gene Ontology terms enriched in these proteins included 'organonitrogen compound metabolic process', 'regulation of mRNA splicing via spliceosome' and 'nucleotide metabolic process'. The present results revealed that LIF modulated phosphorylation levels of nucleotide metabolism‑associated proteins, thus providing insight into the mechanism underlying LIF action in mESCs.
Collapse
Affiliation(s)
- Hwa-Ryung Song
- Department of Microbiology, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| | - Han-Kyu Kim
- Department of Microbiology, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| | - Seung-Gook Kim
- Department of Microbiology, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| | - Hyung-Jin Lim
- Department of Microbiology, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| | - Hyun-Yi Kim
- Division of Anatomy and Developmental Biology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Myung-Kwan Han
- Department of Microbiology, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| |
Collapse
|
12
|
Allmann S, Mayer L, Olma J, Kaina B, Hofmann TG, Tomicic MT, Christmann M. Benzo[a]pyrene represses DNA repair through altered E2F1/E2F4 function marking an early event in DNA damage-induced cellular senescence. Nucleic Acids Res 2020; 48:12085-12101. [PMID: 33166399 PMCID: PMC7708059 DOI: 10.1093/nar/gkaa965] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 09/25/2020] [Accepted: 10/16/2020] [Indexed: 01/08/2023] Open
Abstract
Transcriptional regulation of DNA repair is of outmost importance for the restoration of DNA integrity upon genotoxic stress. Here we report that the potent environmental carcinogen benzo[a]pyrene (B[a]P) activates a cellular DNA damage response resulting in transcriptional repression of mismatch repair (MMR) genes (MSH2, MSH6, EXO1) and of RAD51, the central homologous recombination repair (HR) component, ultimately leading to downregulation of MMR and HR. B[a]P-induced gene repression is caused by abrogated E2F1 signalling. This occurs through proteasomal degradation of E2F1 in G2-arrested cells and downregulation of E2F1 mRNA expression in G1-arrested cells. Repression of E2F1-mediated transcription and silencing of repair genes is further mediated by the p21-dependent E2F4/DREAM complex. Notably, repression of DNA repair is also observed following exposure to the active B[a]P metabolite BPDE and upon ionizing radiation and occurs in response to a p53/p21-triggered, irreversible cell cycle arrest marking the onset of cellular senescence. Overall, our results suggest that repression of MMR and HR is an early event during genotoxic-stress induced senescence. We propose that persistent downregulation of DNA repair might play a role in the maintenance of the senescence phenotype, which is associated with an accumulation of unrepairable DNA lesions.
Collapse
Affiliation(s)
- Sebastian Allmann
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | - Laura Mayer
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | - Jessika Olma
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | - Thomas G Hofmann
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | - Maja T Tomicic
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | - Markus Christmann
- Institute of Toxicology, University Medical Center, Johannes Gutenberg University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| |
Collapse
|
13
|
Aasland D, Götzinger L, Hauck L, Berte N, Meyer J, Effenberger M, Schneider S, Reuber EE, Roos WP, Tomicic MT, Kaina B, Christmann M. Temozolomide Induces Senescence and Repression of DNA Repair Pathways in Glioblastoma Cells via Activation of ATR-CHK1, p21, and NF-κB. Cancer Res 2018; 79:99-113. [PMID: 30361254 DOI: 10.1158/0008-5472.can-18-1733] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/07/2018] [Accepted: 10/22/2018] [Indexed: 11/16/2022]
Abstract
The DNA-methylating drug temozolomide, which induces cell death through apoptosis, is used for the treatment of malignant glioma. Here, we investigate the mechanisms underlying the ability of temozolomide to induce senescence in glioblastoma cells. Temozolomide-induced senescence was triggered by the specific DNA lesion O6-methylguanine (O6MeG) and characterized by arrest of cells in the G2-M phase. Inhibitor experiments revealed that temozolomide-induced senescence was initiated by damage recognition through the MRN complex, activation of the ATR/CHK1 axis of the DNA damage response pathway, and mediated by degradation of CDC25c. Temozolomide-induced senescence required functional p53 and was dependent on sustained p21 induction. p53-deficient cells, not expressing p21, failed to induce senescence, but were still able to induce a G2-M arrest. p14 and p16, targets of p53, were silenced in our cell system and did not seem to play a role in temozolomide-induced senescence. In addition to p21, the NF-κB pathway was required for senescence, which was accompanied by induction of the senescence-associated secretory phenotype. Upon temozolomide exposure, we found a strong repression of the mismatch repair proteins MSH2, MSH6, and EXO1 as well as the homologous recombination protein RAD51, which was downregulated by disruption of the E2F1/DP1 complex. Repression of these repair factors was not observed in G2-M arrested p53-deficient cells and, therefore, it seems to represent a specific trait of temozolomide-induced senescence. SIGNIFICANCE: These findings reveal a mechanism by which the anticancer drug temozolomide induces senescence and downregulation of DNA repair pathways in glioma cells.
Collapse
Affiliation(s)
- Dorthe Aasland
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Laura Götzinger
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Laura Hauck
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Nancy Berte
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Jessica Meyer
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | | | - Simon Schneider
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Emelie E Reuber
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Wynand P Roos
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Maja T Tomicic
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany.
| | - Bernd Kaina
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany.
| | - Markus Christmann
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
14
|
Weßbecher IM, Brieger A. Phosphorylation meets DNA mismatch repair. DNA Repair (Amst) 2018; 72:107-114. [PMID: 30249411 DOI: 10.1016/j.dnarep.2018.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/14/2022]
Abstract
DNA mismatch repair (MMR) is a highly conserved process and ensures the removal of mispaired DNA bases and insertion-deletion loops right after replication. For this, a MutSα or MutSβ protein complex recognizes the DNA damage, MutLα nicks the erroneous strand, exonuclease 1 removes the wrong nucleotides, DNA polymerase δ refills the gap and DNA ligase I joins the fragments to seal the nicks and complete the repair process. The failure to accomplish these functions is associated with higher mutation rates and may lead to cancer, which highlights the importance of MMR by the maintenance of genomic stability. The post-replicative MMR implies that involved proteins are regulated at several levels, including posttranslational modifications (PTMs). Phosphorylation is one of the most common and major PTMs. Suitable with its regulatory force phosphorylation was shown to influence MMR factors thereby adjusting eukaryotic MMR activity. In this review, we summarized the current knowledge of the role of phosphorylation of MMR process involved proteins and their functional relevance.
Collapse
Affiliation(s)
| | - Angela Brieger
- Medical Clinic I, Biomedical Research Laboratory, Goethe-University, Frankfurt a.M., Germany.
| |
Collapse
|
15
|
Weßbecher IM, Hinrichsen I, Funke S, Oellerich T, Plotz G, Zeuzem S, Grus FH, Biondi RM, Brieger A. DNA mismatch repair activity of MutLα is regulated by CK2-dependent phosphorylation of MLH1 (S477). Mol Carcinog 2018; 57:1723-1734. [DOI: 10.1002/mc.22892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/22/2018] [Accepted: 08/18/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Isabel M. Weßbecher
- Medical Clinic I; Biomedical Research Laboratory; Goethe-University; Frankfurt Germany
| | - Inga Hinrichsen
- Medical Clinic I; Biomedical Research Laboratory; Goethe-University; Frankfurt Germany
| | - Sebastian Funke
- Department of Ophthalmology; Experimental Ophthalmology; University Medical Center; Gutenberg University; Mainz Germany
| | - Thomas Oellerich
- Department of Medicine II, Hematology/Oncology; Goethe-University; Frankfurt Germany
| | - Guido Plotz
- Medical Clinic I; Biomedical Research Laboratory; Goethe-University; Frankfurt Germany
| | - Stefan Zeuzem
- Medical Clinic I; Biomedical Research Laboratory; Goethe-University; Frankfurt Germany
| | - Franz H. Grus
- Department of Ophthalmology; Experimental Ophthalmology; University Medical Center; Gutenberg University; Mainz Germany
| | - Ricardo M. Biondi
- Medical Clinic I; Biomedical Research Laboratory; Goethe-University; Frankfurt Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society; Buenos Aires Argentina
| | - Angela Brieger
- Medical Clinic I; Biomedical Research Laboratory; Goethe-University; Frankfurt Germany
| |
Collapse
|
16
|
Chatzileontiadou DSM, Samiotaki M, Alexopoulou AN, Cotsiki M, Panayotou G, Stamatiadi M, Balatsos NAA, Leonidas DD, Kontou M. Proteomic Analysis of Human Angiogenin Interactions Reveals Cytoplasmic PCNA as a Putative Binding Partner. J Proteome Res 2017; 16:3606-3622. [PMID: 28777577 DOI: 10.1021/acs.jproteome.7b00335] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human Angiogenin (hAng) is a member of the ribonuclease A superfamily and a potent inducer of neovascularization. Protein interactions of hAng in the nucleus and cytoplasm of the human umbilical vein cell line EA.hy926 have been investigated by mass spectroscopy. Data are available via ProteomeXchange with identifiers PXD006583 and PXD006584. The first gel-free analysis of hAng immunoprecipitates revealed many statistically significant potential hAng-interacting proteins involved in crucial biological pathways. Surprisingly, proliferating cell nuclear antigen (PCNA), was found to be immunoprecipitated with hAng only in the cytoplasm. The hAng-PCNA interaction and colocalization in the specific cellular compartment was validated with immunoprecipitation, immunoblotting, and immunocytochemistry. The results revealed that PCNA is predominantly localized in the cytoplasm, while hAng is distributed both in the nucleus and in the cytoplasm. hAng and PCNA colocalize in the cytoplasm, suggesting that they may interact in this compartment.
Collapse
Affiliation(s)
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | | | - Marina Cotsiki
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | - George Panayotou
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | - Melina Stamatiadi
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Nikolaos A A Balatsos
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Demetres D Leonidas
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Maria Kontou
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| |
Collapse
|
17
|
Hinrichsen I, Weßbecher IM, Huhn M, Passmann S, Zeuzem S, Plotz G, Biondi RM, Brieger A. Phosphorylation-dependent signaling controls degradation of DNA mismatch repair protein PMS2. Mol Carcinog 2017; 56:2663-2668. [DOI: 10.1002/mc.22709] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/28/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Inga Hinrichsen
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Isabel M. Weßbecher
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Meik Huhn
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
- Pharmazentrum Frankfurt; Institute of Pharmacology and Toxicology; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Sandra Passmann
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Stefan Zeuzem
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Guido Plotz
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Ricardo M. Biondi
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| | - Angela Brieger
- Medical Clinic I; Biomedical Research Laboratory; University Clinic Frankfurt; Frankfurt a.M. Germany
| |
Collapse
|
18
|
DNA mismatch repair and its many roles in eukaryotic cells. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:174-187. [PMID: 28927527 DOI: 10.1016/j.mrrev.2017.07.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/01/2017] [Accepted: 07/06/2017] [Indexed: 02/06/2023]
Abstract
DNA mismatch repair (MMR) is an important DNA repair pathway that plays critical roles in DNA replication fidelity, mutation avoidance and genome stability, all of which contribute significantly to the viability of cells and organisms. MMR is widely-used as a diagnostic biomarker for human cancers in the clinic, and as a biomarker of cancer susceptibility in animal model systems. Prokaryotic MMR is well-characterized at the molecular and mechanistic level; however, MMR is considerably more complex in eukaryotic cells than in prokaryotic cells, and in recent years, it has become evident that MMR plays novel roles in eukaryotic cells, several of which are not yet well-defined or understood. Many MMR-deficient human cancer cells lack mutations in known human MMR genes, which strongly suggests that essential eukaryotic MMR components/cofactors remain unidentified and uncharacterized. Furthermore, the mechanism by which the eukaryotic MMR machinery discriminates between the parental (template) and the daughter (nascent) DNA strand is incompletely understood and how cells choose between the EXO1-dependent and the EXO1-independent subpathways of MMR is not known. This review summarizes recent literature on eukaryotic MMR, with emphasis on the diverse cellular roles of eukaryotic MMR proteins, the mechanism of strand discrimination and cross-talk/interactions between and co-regulation of MMR and other DNA repair pathways in eukaryotic cells. The main conclusion of the review is that MMR proteins contribute to genome stability through their ability to recognize and promote an appropriate cellular response to aberrant DNA structures, especially when they arise during DNA replication. Although the molecular mechanism of MMR in the eukaryotic cell is still not completely understood, increased used of single-molecule analyses in the future may yield new insight into these unsolved questions.
Collapse
|
19
|
Abstract
The connection between genetic variation and drug response has long been explored to facilitate the optimization and personalization of cancer therapy. Crucial to the identification of drug response related genetic features is the ability to separate indirect correlations from direct correlations across abundant datasets with large number of variables. Here we analyzed proteomic and pharmacogenomic data in cancer tissues and cell lines using a global statistical model connecting protein pairs, genes and anti-cancer drugs. We estimated this model using direct coupling analysis (DCA), a powerful statistical inference method that has been successfully applied to protein sequence data to extract evolutionary signals that provide insights on protein structure, folding and interactions. We used Direct Information (DI) as a metric of connectivity between proteins as well as gene-drug pairs. We were able to infer important interactions observed in cancer-related pathways from proteomic data and predict potential connectivities in cancer networks. We also identified known and potential connections for anti-cancer drugs and gene mutations using DI in pharmacogenomic data. Our findings suggest that gene-drug connections predicted with direct couplings can be used as a reliable guide to cancer therapy and expand our understanding of the effects of gene alterations on drug efficacies.
Collapse
|
20
|
Grañé-Boladeras N, Pérez-Torras S, Lozano JJ, Romero MR, Mazo A, Marín JJ, Pastor-Anglada M. Pharmacogenomic analyzis of the responsiveness of gastrointestinal tumor cell lines to drug therapy: A transportome approach. Pharmacol Res 2016; 113:364-375. [DOI: 10.1016/j.phrs.2016.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/17/2016] [Accepted: 09/07/2016] [Indexed: 01/20/2023]
|
21
|
Christmann M, Boisseau C, Kitzinger R, Berac C, Allmann S, Sommer T, Aasland D, Kaina B, Tomicic MT. Adaptive upregulation of DNA repair genes following benzo(a)pyrene diol epoxide protects against cell death at the expense of mutations. Nucleic Acids Res 2016; 44:10727-10743. [PMID: 27694624 PMCID: PMC5159553 DOI: 10.1093/nar/gkw873] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 09/07/2016] [Accepted: 09/21/2016] [Indexed: 12/17/2022] Open
Abstract
A coordinated and faithful DNA damage response is of central importance for maintaining genomic integrity and survival. Here, we show that exposure of human cells to benzo(a)pyrene 9,10-diol-7,8-epoxide (BPDE), the active metabolite of benzo(a)pyrene (B(a)P), which represents a most important carcinogen formed during food preparation at high temperature, smoking and by incomplete combustion processes, causes a prompt and sustained upregulation of the DNA repair genes DDB2, XPC, XPF, XPG and POLH. Induction of these repair factors on RNA and protein level enhanced the removal of BPDE adducts from DNA and protected cells against subsequent BPDE exposure. However, through the induction of POLH the mutation frequency in the surviving cells was enhanced. Activation of these adaptive DNA repair genes was also observed upon B(a)P treatment of MCF7 cells and in buccal cells of human volunteers after cigarette smoking. Our data provide a rational basis for an adaptive response to polycyclic aromatic hydrocarbons, which occurs however at the expense of mutations that may drive cancer formation.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Catherine Boisseau
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Rebekka Kitzinger
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Christian Berac
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Sebastian Allmann
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Tina Sommer
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Dorthe Aasland
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Bernd Kaina
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | - Maja T Tomicic
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| |
Collapse
|
22
|
Wagner VP, Webber LP, Salvadori G, Meurer L, Fonseca FP, Castilho RM, Squarize CH, Vargas PA, Martins MD. Overexpression of MutSα Complex Proteins Predicts Poor Prognosis in Oral Squamous Cell Carcinoma. Medicine (Baltimore) 2016; 95:e3725. [PMID: 27258499 PMCID: PMC4900707 DOI: 10.1097/md.0000000000003725] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The DNA mismatch repair (MMR) system is responsible for the detection and correction of errors created during DNA replication, thereby avoiding the incorporation of mutations in dividing cells. The prognostic value of alterations in MMR system has not previously been analyzed in oral squamous cell carcinoma (OSCC).The study comprised 115 cases of OSCC diagnosed between 1996 and 2010. The specimens collected were constructed into tissue microarray blocks. Immunohistochemical staining for MutSα complex proteins hMSH2 and hMSH6 was performed. The slides were subsequently scanned into high-resolution images, and nuclear staining of hMSH2 and hMSH6 was analyzed using the Nuclear V9 algorithm. Univariable and multivariable Cox proportional hazard regression models were performed to evaluate the prognostic value of hMSH2 and hMSH6 in OSCC.All cases in the present cohort were positive for hMSH2 and hMSH6 and a direct correlation was found between the expression of the proteins (P < 0.05). The mean number of positive cells for hMSH2 and hMSH6 was 64.44 ± 15.21 and 31.46 ± 22.38, respectively. These values were used as cutoff points to determine high protein expression. Cases with high expression of both proteins simultaneously were classified as having high MutSα complex expression. In the multivariable analysis, high expression of the MutSα complex was an independent prognostic factor for poor overall survival (hazard ratio: 2.75, P = 0.02).This study provides a first insight of the prognostic value of alterations in MMR system in OSCC. We found that MutSα complex may constitute a molecular marker for the poor prognosis of OSCC.
Collapse
Affiliation(s)
- Vivian Petersen Wagner
- From the Department of Oral Pathology, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil (VPW, LPW, MDM); Experimental Pathology Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil (VPW, LPW, LM, MDM); Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Norway (GS); Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil (FPF, PAV); and Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan, School of Dentistry, Ann Arbor, MI (RMC, CHS)
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Radhakrishnan R, Li Y, Xiang S, Yuan F, Yuan Z, Telles E, Fang J, Coppola D, Shibata D, Lane WS, Zhang Y, Zhang X, Seto E. Histone deacetylase 10 regulates DNA mismatch repair and may involve the deacetylation of MutS homolog 2. J Biol Chem 2015. [PMID: 26221039 DOI: 10.1074/jbc.m114.612945] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MutS homolog 2 (MSH2) is an essential DNA mismatch repair (MMR) protein. It interacts with MSH6 or MSH3 to form the MutSα or MutSβ complex, respectively, which recognize base-base mispairs and insertions/deletions and initiate the repair process. Mutation or dysregulation of MSH2 causes genomic instability that can lead to cancer. MSH2 is acetylated at its C terminus, and histone deacetylase (HDAC6) deacetylates MSH2. However, whether other regions of MSH2 can be acetylated and whether other histone deacetylases (HDACs) and histone acetyltransferases (HATs) are involved in MSH2 deacetylation/acetylation is unknown. Here, we report that MSH2 can be acetylated at Lys-73 near the N terminus. Lys-73 is highly conserved across many species. Although several Class I and II HDACs interact with MSH2, HDAC10 is the major enzyme that deacetylates MSH2 at Lys-73. Histone acetyltransferase HBO1 might acetylate this residue. HDAC10 overexpression in HeLa cells stimulates cellular DNA MMR activity, whereas HDAC10 knockdown decreases DNA MMR activity. Thus, our study identifies an HDAC10-mediated regulatory mechanism controlling the DNA mismatch repair function of MSH2.
Collapse
Affiliation(s)
| | - Yixuan Li
- From the Departments of Molecular Oncology and
| | - Shengyan Xiang
- the Department of Pathology and Cell Biology, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida 33612
| | - Fenghua Yuan
- the Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | | | | | - Jia Fang
- From the Departments of Molecular Oncology and
| | - Domenico Coppola
- Gastroenterology, and H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - David Shibata
- Gastroenterology, and H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - William S Lane
- the Mass Spectrometry and Proteomics Resource Laboratory, Harvard University, Cambridge, Massachusetts 02138
| | - Yanbin Zhang
- the Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Xiaohong Zhang
- From the Departments of Molecular Oncology and the Department of Pathology and Cell Biology, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida 33612,
| | - Edward Seto
- From the Departments of Molecular Oncology and
| |
Collapse
|
24
|
NPM-ALK mediates phosphorylation of MSH2 at tyrosine 238, creating a functional deficiency in MSH2 and the loss of mismatch repair. Blood Cancer J 2015; 5:e311. [PMID: 25978431 PMCID: PMC4476014 DOI: 10.1038/bcj.2015.35] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 03/16/2015] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
The vast majority of anaplastic lymphoma kinase-positive anaplastic large cell lymphoma (ALK+ALCL) tumors express the characteristic oncogenic fusion protein NPM-ALK, which mediates tumorigenesis by exerting its constitutive tyrosine kinase activity on various substrates. We recently identified MSH2, a protein central to DNA mismatch repair (MMR), as a novel binding partner and phosphorylation substrate of NPM-ALK. Here, using liquid chromatography–mass spectrometry, we report for the first time that MSH2 is phosphorylated by NPM-ALK at a specific residue, tyrosine 238. Using GP293 cells transfected with NPM-ALK, we confirmed that the MSH2Y238F mutant is not tyrosine phosphorylated. Furthermore, transfection of MSH2Y238F into these cells substantially decreased the tyrosine phosphorylation of endogenous MSH2. Importantly, gene transfection of MSH2Y238F abrogated the binding of NPM-ALK with endogenous MSH2, re-established the dimerization of MSH2:MSH6 and restored the sensitivity to DNA mismatch-inducing drugs, indicative of MMR return. Parallel findings were observed in two ALK+ALCL cell lines, Karpas 299 and SUP-M2. In addition, we found that enforced expression of MSH2Y238F into ALK+ALCL cells alone was sufficient to induce spontaneous apoptosis. In conclusion, our findings have identified NPM-ALK-induced phosphorylation of MSH2 at Y238 as a crucial event in suppressing MMR. Our studies have provided novel insights into the mechanism by which oncogenic tyrosine kinases disrupt MMR.
Collapse
|
25
|
Christmann M, Kaina B. Transcriptional regulation of human DNA repair genes following genotoxic stress: trigger mechanisms, inducible responses and genotoxic adaptation. Nucleic Acids Res 2013; 41:8403-20. [PMID: 23892398 PMCID: PMC3794595 DOI: 10.1093/nar/gkt635] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA repair is the first barrier in the defense against genotoxic stress. In recent years, mechanisms that recognize DNA damage and activate DNA repair functions through transcriptional upregulation and post-translational modification were the focus of intensive research. Most DNA repair pathways are complex, involving many proteins working in discrete consecutive steps. Therefore, their balanced expression is important for avoiding erroneous repair that might result from excessive base removal and DNA cleavage. Amelioration of DNA repair requires both a fine-tuned system of lesion recognition and transcription factors that regulate repair genes in a balanced way. Transcriptional upregulation of DNA repair genes by genotoxic stress is counteracted by DNA damage that blocks transcription. Therefore, induction of DNA repair resulting in an adaptive response is only visible through a narrow window of dose. Here, we review transcriptional regulation of DNA repair genes in normal and cancer cells and describe mechanisms of promoter activation following genotoxic exposures through environmental carcinogens and anticancer drugs. The data available to date indicate that 25 DNA repair genes are subject to regulation following genotoxic stress in rodent and human cells, but for only a few of them, the data are solid as to the mechanism, homeostatic regulation and involvement in an adaptive response to genotoxic stress.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University Medical Center, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | | |
Collapse
|
26
|
Edelbrock MA, Kaliyaperumal S, Williams KJ. Structural, molecular and cellular functions of MSH2 and MSH6 during DNA mismatch repair, damage signaling and other noncanonical activities. Mutat Res 2013; 743-744:53-66. [PMID: 23391514 DOI: 10.1016/j.mrfmmm.2012.12.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/28/2012] [Accepted: 12/31/2012] [Indexed: 11/18/2022]
Abstract
The field of DNA mismatch repair (MMR) has rapidly expanded after the discovery of the MutHLS repair system in bacteria. By the mid 1990s yeast and human homologues to bacterial MutL and MutS had been identified and their contribution to hereditary non-polyposis colorectal cancer (HNPCC; Lynch syndrome) was under intense investigation. The human MutS homologue 6 protein (hMSH6), was first reported in 1995 as a G:T binding partner (GTBP) of hMSH2, forming the hMutSα mismatch-binding complex. Signal transduction from each DNA-bound hMutSα complex is accomplished by the hMutLα heterodimer (hMLH1 and hPMS2). Molecular mechanisms and cellular regulation of individual MMR proteins are now areas of intensive research. This review will focus on molecular mechanisms associated with mismatch binding, as well as emerging evidence that MutSα, and in particular, MSH6, is a key protein in MMR-dependent DNA damage response and communication with other DNA repair pathways within the cell. MSH6 is unstable in the absence of MSH2, however it is the DNA lesion-binding partner of this heterodimer. MSH6, but not MSH2, has a conserved Phe-X-Glu motif that recognizes and binds several different DNA structural distortions, initiating different cellular responses. hMSH6 also contains the nuclear localization sequences required to shuttle hMutSα into the nucleus. For example, upon binding to O(6)meG:T, MSH6 triggers a DNA damage response that involves altered phosphorylation within the N-terminal disordered domain of this unique protein. While many investigations have focused on MMR as a post-replication DNA repair mechanism, MMR proteins are expressed and active in all phases of the cell cycle. There is much more to be discovered about regulatory cellular roles that require the presence of MutSα and, in particular, MSH6.
Collapse
Affiliation(s)
| | - Saravanan Kaliyaperumal
- Division of Comparative Medicine and Pathology, New England Primate Research Center, One Pine Hill Drive, Southborough, MA 01772, USA.
| | - Kandace J Williams
- University of Toledo College of Medicine and Life Sciences, Department of Biochemistry & Cancer Biology, 3000 Transverse Dr., Toledo, OH 43614, USA.
| |
Collapse
|
27
|
Exonuclease 1 (Exo1) is required for activating response to S(N)1 DNA methylating agents. DNA Repair (Amst) 2012; 11:951-64. [PMID: 23062884 DOI: 10.1016/j.dnarep.2012.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 09/08/2012] [Accepted: 09/11/2012] [Indexed: 11/23/2022]
Abstract
S(N)1 DNA methylating agents are genotoxic agents that methylate numerous nucleophilic centers within DNA including the O(6) position of guanine (O(6)meG). Methylation of this extracyclic oxygen forces mispairing with thymine during DNA replication. The mismatch repair (MMR) system recognizes these O(6)meG:T mispairs and is required to activate DNA damage response (DDR). Exonuclease I (EXO1) is a key component of MMR by resecting the damaged strand; however, whether EXO1 is required to activate MMR-dependent DDR remains unknown. Here we show that knockdown of the mouse ortholog (mExo1) in mouse embryonic fibroblasts (MEFs) results in decreased G2/M checkpoint response, limited effects on cell proliferation, and increased cell viability following exposure to the S(N)1 methylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), establishing a phenotype paralleling MMR deficiency. MNNG treatment induced formation of γ-H2AX foci with which EXO1 co-localized in MEFs, but mExo1-depleted MEFs displayed a significant diminishment of γ-H2AX foci formation. mExo1 depletion also reduced MSH2 association with DNA duplexes containing G:T mismatches in vitro, decreased MSH2 association with alkylated chromatin in vivo, and abrogated MNNG-induced MSH2/CHK1 interaction. To determine if nuclease activity is required to activate DDR we stably overexpressed a nuclease defective form of human EXO1 (hEXO1) in mExo1-depleted MEFs. These experiments indicated that expression of wildtype and catalytically null hEXO1 was able to restore normal response to MNNG. This study indicates that EXO1 is required to activate MMR-dependent DDR in response to S(N)1 methylating agents; however, this function of EXO1 is independent of its nucleolytic activity.
Collapse
|
28
|
Abstract
Single strand nicks and gaps in DNA have been reported to increase the efficiency of nucleosome loading mediated by chromatin assembly factor 1 (CAF-1). However, on mismatch-containing substrates, these strand discontinuities are utilized by the mismatch repair (MMR) system as loading sites for exonuclease 1, at which degradation of the error-containing strand commences. Because packaging of DNA into chromatin might inhibit MMR, we were interested to learn whether chromatin assembly is differentially regulated on heteroduplex and homoduplex substrates. We now show that the presence of a mismatch in a nicked plasmid substrate delays nucleosome loading in human cell extracts. Our data also suggest that, once the mismatch is removed, repair of the single-stranded gap is accompanied by efficient nucleosome loading. We postulated that the balance between MMR and chromatin assembly might be governed by proliferating cell nuclear antigen (PCNA), the processivity factor of replicative DNA polymerases, which is loaded at DNA termini and which interacts with the MSH6 subunit of the mismatch recognition factor MutSα, as well as with CAF-1. We now show that this regulation might be more complex; MutSα and CAF-1 interact not only with PCNA, but also with each other. In vivo this interaction increases during S-phase and may be controlled by the phosphorylation status of the p150 subunit of CAF-1.
Collapse
|
29
|
Young LC, Bone KM, Wang P, Wu F, Adam BA, Hegazy S, Gelebart P, Holovati J, Li L, Andrew SE, Lai R. Fusion tyrosine kinase NPM-ALK Deregulates MSH2 and suppresses DNA mismatch repair function novel insights into a potent oncoprotein. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:411-21. [PMID: 21703420 DOI: 10.1016/j.ajpath.2011.03.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 02/09/2011] [Accepted: 03/10/2011] [Indexed: 11/17/2022]
Abstract
The fusion tyrosine kinase NPM-ALK is central to the pathogenesis of ALK-positive anaplastic large cell lymphoma (ALK(+)ALCL). We recently identified that MSH2, a key DNA mismatch repair (MMR) protein integral to the suppression of tumorigenesis, is an NPM-ALK-interacting protein. In this study, we found in vitro evidence that enforced expression of NPM-ALK in HEK293 cells suppressed MMR function. Correlating with these findings, six of nine ALK(+)ALCL tumors displayed evidence of microsatellite instability, as opposed to none of the eight normal DNA control samples (P = 0.007, Student's t-test). Using co-immunoprecipitation, we found that increasing levels of NPM-ALK expression in HEK293 cells resulted in decreased levels of MSH6 bound to MSH2, whereas MSH2·NPM-ALK binding was increased. The NPM-ALK·MSH2 interaction was dependent on the activation/autophosphorylation of NPM-ALK, and the Y191 residue of NPM-ALK was a crucial site for this interaction and NPM-ALK-mediated MMR suppression. MSH2 was found to be tyrosine phosphorylated in the presence of NPM-ALK. Finally, NPM-ALK impeded the expected DNA damage-induced translocation of MSH2 out of the cytoplasm. To conclude, our data support a model in which the suppression of MMR by NPM-ALK is attributed to its ability to interfere with normal MSH2 biochemistry and function.
Collapse
MESH Headings
- Cytoplasm/metabolism
- DNA Damage
- DNA Mismatch Repair
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Microsatellite Instability
- MutS Homolog 2 Protein/genetics
- MutS Homolog 2 Protein/metabolism
- Phosphorylation
- Protein Multimerization
- Protein Transport
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Tumor Cells, Cultured
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Leah C Young
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gassman NR, Clodfelter JE, McCauley AK, Bonin K, Salsbury FR, Scarpinato KD. Cooperative nuclear localization sequences lend a novel role to the N-terminal region of MSH6. PLoS One 2011; 6:e17907. [PMID: 21437237 PMCID: PMC3060103 DOI: 10.1371/journal.pone.0017907] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 02/14/2011] [Indexed: 01/08/2023] Open
Abstract
Human mismatch repair proteins MSH2-MSH6 play an essential role in maintaining genetic stability and preventing disease. While protein functions have been extensively studied, the substantial amino-terminal region (NTR*) of MSH6 that is unique to eukaryotic proteins, has mostly evaded functional characterization. We demonstrate that a cluster of three nuclear localization signals (NLS) in the NTR direct nuclear import. Individual NLSs are capable of partially directing cytoplasmic protein into the nucleus; however only cooperative effects between all three NLSs efficiently transport MSH6 into the nucleus. In striking contrast to yeast and previous assumptions on required heterodimerization, human MSH6 does not determine localization of its heterodimeric partner, MSH2. A cancer-derived mutation localized between two of the three NLS significantly decreases nuclear localization of MSH6, suggesting altered protein localization can contribute to carcinogenesis. These results clarify the pending speculations on the functional role of the NTR in human MSH6 and identify a novel, cooperative nuclear localization signal.
Collapse
Affiliation(s)
- Natalie R. Gassman
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Jill E. Clodfelter
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Anita K. McCauley
- Department of Biology, Wake Forest University, Winston Salem, North Carolina, United States of America
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston Salem, North Carolina, United States of America
| | - Freddie R. Salsbury
- Department of Physics, Wake Forest University, Winston Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Karin D. Scarpinato
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
31
|
Kaliyaperumal S, Patrick SM, Williams KJ. Phosphorylated hMSH6: DNA mismatch versus DNA damage recognition. Mutat Res 2011; 706:36-45. [PMID: 21035467 PMCID: PMC3010429 DOI: 10.1016/j.mrfmmm.2010.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/09/2010] [Accepted: 10/20/2010] [Indexed: 01/30/2023]
Abstract
DNA mismatch repair (MMR) maintains genomic integrity by correction of mispaired bases and insertion-deletion loops. The MMR pathway can also trigger a DNA damage response upon binding of MutSα to specific DNA lesions such as O(6)methylguanine (O(6)meG). Limited information is available regarding cellular regulation of these two different pathways. Within this report, we demonstrate that phosphorylated hMSH6 increases in concentration in the presence of a G:T mismatch, as compared to an O(6)meG:T lesion. TPA, a kinase activator, enhances the phosphorylation of hMSH6 and binding of hMutSα to a G:T mismatch, though not to O(6)meG:T. UCN-01, a kinase inhibitor, decreases both phosphorylation of hMSH6 and binding of hMutSα to G:T and O(6)meG:T. HeLa MR cells, pretreated with UCN-01 and exposed to MNNG, undergo activation of Cdk1 and mitosis despite phosphorylation of Chk1 and inactivating phosphorylation of Cdc25c. These results indicate that UCN-01 may inhibit an alternative cell cycle arrest pathway associated with the MMR pathway that does not involve Cdc25c. In addition, recombinant hMutSα containing hMSH6 mutated at an N-terminal cluster of four phosphoserines exhibits decreased phosphorylation and decreased binding of hMutSα to G:T and O(6)meG:T. Taken together, these results suggest a model in which the amount of phosphorylated hMSH6 bound to DNA is dependent on the presence of either a DNA mismatch or DNA alkylation damage. We hypothesize that both phosphorylation of hMSH6 and total concentration of bound hMutSα are involved in cellular signaling of either DNA mismatch repair or MMR-dependent damage recognition activities.
Collapse
Affiliation(s)
- Saravanan Kaliyaperumal
- Department of Biochemistry & Cancer Biology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Steve M. Patrick
- Department of Biochemistry & Cancer Biology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Kandace J. Williams
- Department of Biochemistry & Cancer Biology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| |
Collapse
|
32
|
Delayed c-Fos activation in human cells triggers XPF induction and an adaptive response to UVC-induced DNA damage and cytotoxicity. Cell Mol Life Sci 2010; 68:1785-98. [PMID: 20976523 PMCID: PMC3078315 DOI: 10.1007/s00018-010-0546-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 09/15/2010] [Accepted: 09/16/2010] [Indexed: 01/16/2023]
Abstract
The oncoprotein c-Fos has been commonly found differently expressed in cancer cells. Our previous work showed that mouse cells lacking the immediate-early gene c-fos are hypersensitive to ultraviolet (UVC) light. Here, we demonstrate that in human diploid fibroblasts UV-triggered induction of c-Fos protein is a delayed and long-lasting event. Sustained upregulation of c-Fos goes along with transcriptional stimulation of the NER gene xpf, which harbors an AP-1 binding site in the promoter. Data gained on c-Fos knockdown and c-Fos overexpressing human cells provide evidence that c-Fos/AP-1 stimulates upregulation of XPF, thereby increasing the cellular repair capacity protecting from UVC-induced DNA damage. When these cells are pre-exposed to a low non-toxic UVC dose and challenged with a subsequent high dose of UVC irradiation, they show accelerated repair of UVC-induced DNA adducts and reduced cell kill. The data indicate a protective role of c-Fos induction by triggering an adaptive response pathway.
Collapse
|
33
|
Nguyen TA, Slattery SD, Moon SH, Darlington YF, Lu X, Donehower LA. The oncogenic phosphatase WIP1 negatively regulates nucleotide excision repair. DNA Repair (Amst) 2010; 9:813-23. [PMID: 20451471 PMCID: PMC2893264 DOI: 10.1016/j.dnarep.2010.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 03/31/2010] [Accepted: 04/10/2010] [Indexed: 01/07/2023]
Abstract
Nucleotide excision repair (NER) is the only mechanism in humans to repair UV-induced DNA lesions such as pyrimidine (6-4) pyrimidone photoproducts and cyclobutane pyrimidine dimers (CPDs). In response to UV damage, the ataxia telangiectasia mutated and Rad3-related (ATR) kinase phosphorylates and activates several downstream effector proteins, such as p53 and XPA, to arrest cell cycle progression, stimulate DNA repair, or initiate apoptosis. However, following the completion of DNA repair, there must be active mechanisms that restore the cell to a prestressed homeostatic state. An important part of this recovery must include a process to reduce p53 and NER activity as well as to remove repair protein complexes from the DNA damage sites. Since activation of the damage response occurs in part through phosphorylation, phosphatases are obvious candidates as homeostatic regulators of the DNA damage and repair responses. Therefore, we investigated whether the serine/threonine wild-type p53-induced phosphatase 1 (WIP1/PPM1D) might regulate NER. WIP1 overexpression inhibits the kinetics of NER and CPD repair, whereas WIP1 depletion enhances NER kinetics and CPD repair. This NER suppression is dependent on WIP1 phosphatase activity, as phosphatase-dead WIP1 mutants failed to inhibit NER. Moreover, WIP1 suppresses the kinetics of UV-induced damage repair largely through effects on NER, as XPD-deficient cells are not further suppressed in repairing UV damage by overexpressed WIP1. Wip1 null mice quickly repair their CPD and undergo less UV-induced apoptosis than their wild-type counterparts. In vitro phosphatase assays identify XPA and XPC as two potential WIP1 targets in the NER pathway. Thus WIP1 may suppress NER kinetics by dephosphorylating and inactivating XPA and XPC and other NER proteins and regulators after UV-induced DNA damage is repaired.
Collapse
Affiliation(s)
- Thuy-Ai Nguyen
- Interdepartmental Graduate Program in Cell and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Scott D. Slattery
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Sung-Hwan Moon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Yolanda F. Darlington
- Interdepartmental Graduate Program in Cell and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Xiongbin Lu
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Lawrence A. Donehower
- Interdepartmental Graduate Program in Cell and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
34
|
Christmann M, Tomicic MT, Aasland D, Berdelle N, Kaina B. Three prime exonuclease I (TREX1) is Fos/AP-1 regulated by genotoxic stress and protects against ultraviolet light and benzo(a)pyrene-induced DNA damage. Nucleic Acids Res 2010; 38:6418-32. [PMID: 20511593 PMCID: PMC2965218 DOI: 10.1093/nar/gkq455] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Cells respond to genotoxic stress with the induction of DNA damage defence functions. Aimed at identifying novel players in this response, we analysed the genotoxic stress-induced expression of DNA repair genes in mouse fibroblasts proficient and deficient for c-Fos or c-Jun. The experiments revealed a clear up-regulation of the three prime exonuclease I (trex1) mRNA following ultraviolet (UV) light treatment. This occurred in the wild-type but not c-fos and c-jun null cells, indicating the involvement of AP-1 in trex1 induction. Trex1 up-regulation was also observed in human cells and was found on promoter, RNA and protein level. Apart from UV light, TREX1 is induced by other DNA damaging agents such as benzo(a)pyrene and hydrogen peroxide. The mouse and human trex1 promoter harbours an AP-1 binding site that is recognized by c-Fos and c-Jun, and its mutational inactivation abrogated trex1 induction. Upon genotoxic stress, TREX1 is not only up-regulated but also translocated into the nucleus. Cells deficient in TREX1 show reduced recovery from the UV and benzo(a)pyrene-induced replication inhibition and increased sensitivity towards the genotoxins compared to the isogenic control. The data revealed trex1 as a novel DNA damage-inducible repair gene that plays a protective role in the genotoxic stress response.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany.
| | | | | | | | | |
Collapse
|
35
|
Mohan RD, Litchfield DW, Torchia J, Tini M. Opposing regulatory roles of phosphorylation and acetylation in DNA mispair processing by thymine DNA glycosylase. Nucleic Acids Res 2009; 38:1135-48. [PMID: 19966277 PMCID: PMC2831317 DOI: 10.1093/nar/gkp1097] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
CpG dinucleotides are mutational hotspots associated with cancer and genetic diseases. Thymine DNA glycosylase (TDG) plays an integral role in CpG maintenance by excising mispaired thymine and uracil in a CpG context and also participates in transcriptional regulation via gene-specific CpG demethylation and functional interactions with the transcription machinery. Here, we report that protein kinase C α (PKCα) interacts with TDG and phosphorylates amino-terminal serine residues adjacent to lysines acetylated by CREB-binding protein (CBP) and p300 (CBP/p300). We establish that acetylation and phosphorylation are mutually exclusive, and their interplay dramatically alters the DNA mispair-processing functions of TDG. Remarkably, acetylation of the amino-terminal region abrogates high-affinity DNA binding and selectively prevents processing of G:T mispairs. In contrast, phosphorylation does not markedly alter DNA interactions, but may preserve G:T processing in vivo by preventing CBP-mediated acetylation. Mutational analysis suggests that the acetyl-acceptor lysines are not directly involved in contacting DNA, but may constitute a conformationally sensitive interface that modulates DNA interactions. These findings reveal opposing roles of CBP/p300 and PKCα in regulating the DNA repair functions of TDG and suggest that the interplay of these modifications in vivo may be critically important in the maintenance of CpG dinucleotides and epigenetic regulation.
Collapse
Affiliation(s)
- Ryan D Mohan
- Department of Physiology and Pharmacology, Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
36
|
Knudsen NØ, Andersen SD, Lützen A, Nielsen FC, Rasmussen LJ. Nuclear translocation contributes to regulation of DNA excision repair activities. DNA Repair (Amst) 2009; 8:682-9. [PMID: 19376751 DOI: 10.1016/j.dnarep.2009.03.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 03/06/2009] [Accepted: 03/14/2009] [Indexed: 11/26/2022]
Abstract
DNA mutations are circumvented by dedicated specialized excision repair systems, such as the base excision repair (BER), nucleotide excision repair (NER), and mismatch repair (MMR) pathways. Although the individual repair pathways have distinct roles in suppressing changes in the nuclear DNA, it is evident that proteins from the different DNA repair pathways interact [Y. Wang, D. Cortez, P. Yazdi, N. Neff, S.J. Elledge, J. Qin, BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures, Genes Dev. 14 (2000) 927-939; M. Christmann, M.T. Tomicic, W.P. Roos, B. Kaina, Mechanisms of human DNA repair: an update, Toxicology 193 (2003) 3-34; N.B. Larsen, M. Rasmussen, L.J. Rasmussen, Nuclear and mitochondrial DNA repair: similar pathways? Mitochondrion 5 (2005) 89-108]. Protein interactions are not only important for function, but also for regulation of nuclear import that is necessary for proper localization of the repair proteins. This review summarizes the current knowledge on nuclear import mechanisms of DNA excision repair proteins and provides a model that categorizes the import by different mechanisms, including classical nuclear import, co-import of proteins, and alternative transport pathways. Most excision repair proteins appear to contain classical NLS sequences directing their nuclear import, however, additional import mechanisms add alternative regulatory levels to protein import, indirectly affecting protein function. Protein co-import appears to be a mechanism employed by the composite repair systems NER and MMR to enhance and regulate nuclear accumulation of repair proteins thereby ensuring faithful DNA repair.
Collapse
Affiliation(s)
- Nina Østergaard Knudsen
- Department of Science, Systems and Models, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | | | | | | | | |
Collapse
|
37
|
Christmann M, Tomicic MT, Gestrich C, Roos WP, Bohr VA, Kaina B. WRN protects against topo I but not topo II inhibitors by preventing DNA break formation. DNA Repair (Amst) 2008; 7:1999-2009. [PMID: 18805512 PMCID: PMC2606036 DOI: 10.1016/j.dnarep.2008.08.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 07/15/2008] [Accepted: 08/25/2008] [Indexed: 12/24/2022]
Abstract
The Werner syndrome helicase/3'-exonuclease (WRN) is a major component of the DNA repair and replication machinery. To analyze whether WRN is involved in the repair of topoisomerase-induced DNA damage we utilized U2-OS cells, in which WRN is stably down-regulated (wrn-kd), and the corresponding wild-type cells (wrn-wt). We show that cells not expressing WRN are hypersensitive to the toxic effect of the topoisomerase I inhibitor topotecan, but not to the topoisomerase II inhibitor etoposide. This was shown by mass survival assays, colony formation and induction of apoptosis. Upon topotecan treatment WRN deficient cells showed enhanced DNA replication inhibition and S-phase arrest, whereas after treatment with etoposide they showed the same cell cycle response as the wild-type. A considerable difference between WRN and wild-type cells was observed for DNA single- and double-strand break formation in response to topotecan. Topotecan induced DNA single-strand breaks 6h after treatment. In both wrn-wt and wrn-kd cells these breaks were repaired at similar kinetics. However, in wrn-kd but not wrn-wt cells they were converted into DNA double-strand breaks (DSBs) at high frequency, as shown by neutral comet assay and phosphorylation of H2AX. Our data provide evidence that WRN is involved in the repair of topoisomerase I, but not topoisomerase II-induced DNA damage, most likely via preventing the conversion of DNA single-strand breaks into DSBs during the resolution of stalled replication forks at topo I-DNA complexes. We suggest that the WRN status of tumor cells impacts anticancer therapy with topoisomerase I, but not topoisomerase II inhibitors.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University of Mainz, Obere Zahlbacher Straße 67, D-55131 Mainz, Germany
| | - Maja T. Tomicic
- Department of Toxicology, University of Mainz, Obere Zahlbacher Straße 67, D-55131 Mainz, Germany
| | - Christopher Gestrich
- Department of Toxicology, University of Mainz, Obere Zahlbacher Straße 67, D-55131 Mainz, Germany
| | - Wynand P. Roos
- Department of Toxicology, University of Mainz, Obere Zahlbacher Straße 67, D-55131 Mainz, Germany
| | - Vilhelm A. Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Bernd Kaina
- Department of Toxicology, University of Mainz, Obere Zahlbacher Straße 67, D-55131 Mainz, Germany
| |
Collapse
|
38
|
Neri S, Pawelec G, Facchini A, Ferrari C, Mariani E. Altered expression of mismatch repair proteins associated with acquisition of microsatellite instability in a clonal model of human T lymphocyte aging. Rejuvenation Res 2008; 11:565-72. [PMID: 18484899 DOI: 10.1089/rej.2007.0639] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The DNA mismatch repair system, the main postreplicative correction pathway in eukaryotic cells, has been shown to be involved in the acquisition of genetic damage during the aging of normal somatic cells, including those of the immune system. Previously, we showed that some but not all human T cell clones (TCC) in an in vitro culture aging model develop microsatellite instability (MSI), which is associated with altered expression of mismatch repair genes. Here, we analyzed levels of mismatch repair proteins as well as the corresponding mRNAs and related this to the development of microsatellite instability in TCC. Msh2, Msh3, Msh6, Pms1, and Pms2 protein expression was quantified by Western blotting. We found that clones not manifesting microsatellite instability in this in vitro model of T cell replicative aging, induced by persistent antigenic stimulation, maintain normal transcriptional control and coordination among the mismatch repair system genes, while clones which do manifest MSI display a general deregulation of gene expression, which is likely to contribute to its occurrence.
Collapse
Affiliation(s)
- Simona Neri
- Laboratorio di Immunologia e Genetica, Istituto di Ricerca Codivilla-Putti, IOR, Bologna, Italy
| | | | | | | | | |
Collapse
|
39
|
Wang CY, Lin YW, Yang JL. Activation of protein kinase Calpha signaling prevents cytotoxicity and mutagenicity following lead acetate in CL3 human lung cancer cells. Toxicology 2008; 250:55-61. [PMID: 18590793 DOI: 10.1016/j.tox.2008.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/04/2008] [Accepted: 06/04/2008] [Indexed: 10/22/2022]
Abstract
Protein kinase C (PKC) family of serine/threonine protein kinases is sensitive signaling transducers in response to lead acetate (Pb) that could transmit phosphorylation cascade for proliferation and de-differentiation of neural cells. However, little is known as to the impact of PKC on Pb genotoxicity. Here we investigate whether Pb activates the conventional/classical subfamily of PKC (cPKC) signaling to affect cytotoxicity and mutagenicity in CL3 human non-small-cell lung adenocarcinoma cells. Pb specifically promoted membrane localization of the alpha isoform of PKC in CL3 cells. Pb also elicited Raf-1 activation as measured by the induction of phospho-Raf-1S338 and the dissociation from the Raf-1 kinase inhibitor protein. Inhibition of cPKC activity using Gö6976 or depletion of PKCalpha by introducing specific small interfering RNA blocked the induction of phospho-Raf-1S338, phospho-MKK1/2 and phospho-ERK1/2 in cells exposed to Pb. Intriguingly, declining PKCalpha enhanced the Pb cytotoxicity and revealed the Pb mutagenicity at the hprt gene. The results suggest that PKCalpha is obligatory for activation of the Raf-1-MKK1/2-ERK1/2 signaling module and plays a defensive role against cytotoxicity and mutagenicity following Pb exposure. Results obtained in this study also support our previous report showing that ERK1/2 activity is involved in preventing Pb genotoxicity.
Collapse
Affiliation(s)
- Chun-Yu Wang
- Molecular Carcinogenesis Laboratory, Institute of Biotechnology & Department of Life Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | |
Collapse
|
40
|
Tomicic MT, Christmann M, Kaina B. Apoptosis in UV-C light irradiated p53 wild-type, apaf-1 and p53 knockout mouse embryonic fibroblasts: interplay of receptor and mitochondrial pathway. Apoptosis 2008; 10:1295-304. [PMID: 16215690 DOI: 10.1007/s10495-005-1392-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Mouse embryonic fibroblasts (MEFs) deficient for the transcription factor p53 are hypersensitive to UV-C light. They also show a reduced recovery from UV-C induced replication blockage and are unable to repair UV-C photoproducts. In this study, we utilized wild-type (wt), Apaf-1 deficient (apaf-1(-/-)) and p53 deficient (p53(-/-)) MEFs in order to elucidate the role of non-repaired UV-C lesions in apoptotic signalling. Corresponding with the cellular sensitivity determined by the WST assay, p53(-/-) cells displayed the highest level of apoptosis, whereas wt cells showed moderate apoptosis after UV-C irradiation. Apaf1(-/-) cells were most resistant. In wt cells apoptosis was executed both via the mitochondrial and the receptor-mediated pathway, as shown by Bcl-2 decline, induction of fasR and activation of caspases-3,8,9. In apaf-1(-/-) (p53(+/+)) cells, the mitochondrial pathway was blocked downstream of Bcl-2, indicating that in this case apoptosis was mediated via the induction of fasR and caspase-3,8 activation. In p53 deficient cells, non-repaired UV-C induced DNA lesions triggered sustained up-regulation of fas ligand (fasL) mRNA, which was not seen in wt and apaf-1(-/-) cells. Therefore, in p53(-/-) MEFs, the receptor/ligand triggered pathway appeared to be dominant. This was confirmed by significant reduction of apoptosis after DN-FADD transfection. As opposed to wt and apaf-1(-/-) cells, p53 deficient MEFs showed no induction of Fas receptor and no Bcl-2 decline. Nevertheless, the resulting caspase-8 and -3 activation was stronger compared to wt and apaf-1(-/-) cells. The data indicate that UV-C light activates in MEFs both the Fas (CD95, Apo-1) receptor and the mitochondrial damage pathways. In p53(-/-) cells, however, the high level of non-repaired DNA damage forces signalling by fasL upregulation, leading to enhanced UV-C-induced apoptosis.
Collapse
Affiliation(s)
- M T Tomicic
- Department of Toxicology, University of Mainz, Germany
| | | | | |
Collapse
|
41
|
Kaina B, Christmann M, Naumann S, Roos WP. MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair (Amst) 2007; 6:1079-99. [PMID: 17485253 DOI: 10.1016/j.dnarep.2007.03.008] [Citation(s) in RCA: 464] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
O(6)-methylguanine-DNA methyltransferase (MGMT) plays a crucial role in the defense against alkylating agents that generate, among other lesions, O(6)-alkylguanine in DNA (collectively termed O(6)-alkylating agents [O(6)AA]). The defense is highly important, since O(6)AA are common environmental carcinogens, are formed endogenously during normal cellular metabolism and possibly inflammation, and are being used in cancer therapy. O(6)AA induced DNA damage is subject to repair, which is executed by MGMT, AlkB homologous proteins (ABH) and base excision repair (BER). Although this review focuses on MGMT, the mechanism of repair by ABH and BER will also be discussed. Experimental systems, in which MGMT has been modulated, revealed that O(6)-methylguanine (O(6)MeG) and O(6)-chloroethylguanine are major mutagenic, carcinogenic, recombinogenic, clastogenic and killing lesions. O(6)MeG-induced clastogenicity and cell death require MutS alpha-dependent mismatch repair (MMR), whereas O(6)-chloroethylguanine-induced killing occurs independently of MMR. Extensive DNA replication is required for O(6)MeG to provoke cytotoxicity. In MGMT depleted cells, O(6)MeG induces apoptosis almost exclusively, barely any necrosis, which is presumably due to the remarkable ability of secondarily formed DNA double-strand breaks (DSBs) to trigger apoptosis via ATM/ATR, Chk1, Chk2, p53 and p73. Depending on the cellular background, O(6)MeG activates both the death receptor and the mitochondrial apoptotic pathway. The inter-individual expression of MGMT in human lymphocytes is highly variable. Given the key role of MGMT in cellular defense, determination of MGMT activity could be useful for assessing a patient's drug sensitivity. MGMT is expressed at highly variable amounts in human tumors. In gliomas, a correlation was found between MGMT activity, MGMT promoter methylation and response to O(6)AA. Although the human MGMT gene is inducible by glucocorticoids and genotoxins such as radiation and alkylating agents, the role of this induction in the protection against carcinogens and the development of chemotherapeutic alkylating drug resistance are still unclear. Modulation of MGMT expression in tumors and normal tissue is currently being investigated as a possible strategy for improving cancer therapy.
Collapse
Affiliation(s)
- Bernd Kaina
- Department of Toxicology, University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| | | | | | | |
Collapse
|
42
|
Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB, Batchelor TT, Futreal PA, Stratton MR, Curry WT, Iafrate AJ, Louis DN. Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment. Clin Cancer Res 2007; 13:2038-45. [PMID: 17404084 PMCID: PMC2873832 DOI: 10.1158/1078-0432.ccr-06-2149] [Citation(s) in RCA: 328] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastomas are treated by surgical resection followed by radiotherapy [X-ray therapy (XRT)] and the alkylating chemotherapeutic agent temozolomide. Recently, inactivating mutations in the mismatch repair gene MSH6 were identified in two glioblastomas recurrent post-temozolomide. Because mismatch repair pathway inactivation is a known mediator of alkylator resistance in vitro, these findings suggested that MSH6 inactivation was causally linked to these two recurrences. However, the extent of involvement of MSH6 in glioblastoma is unknown. We sought to determine the overall frequency and clinical relevance of MSH6 alterations in glioblastomas. EXPERIMENTAL DESIGN The MSH6 gene was sequenced in 54 glioblastomas. MSH6 and O(6)-methylguanine methyltransferase (MGMT) immunohistochemistry was systematically scored in a panel of 46 clinically well-characterized glioblastomas, and the corresponding patient response to treatment evaluated. RESULTS MSH6 mutation was not observed in any pretreatment glioblastoma (0 of 40), whereas 3 of 14 recurrent cases had somatic mutations (P = 0.015). MSH6 protein expression was detected in all pretreatment (17 of 17) cases examined but, notably, expression was lost in 7 of 17 (41%) recurrences from matched post-XRT + temozolomide cases (P = 0.016). Loss of MSH6 was not associated with O(6)-methylguanine methyltransferase status. Measurements of in vivo tumor growth using three-dimensional reconstructed magnetic resonance imaging showed that MSH6-negative glioblastomas had a markedly increased rate of growth while under temozolomide treatment (3.17 versus 0.04 cc/mo for MSH6-positive tumors; P = 0.020). CONCLUSIONS Loss of MSH6 occurs in a subset of post-XRT + temozolomide glioblastoma recurrences and is associated with tumor progression during temozolomide treatment, mirroring the alkylator resistance conferred by MSH6 inactivation in vitro. MSH6 deficiency may therefore contribute to the emergence of recurrent glioblastomas during temozolomide treatment.
Collapse
Affiliation(s)
- Daniel P Cahill
- Molecular Pathology Unit, Neurosurgical Service, Brain Tumor Center, and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Christmann M, Tomicic MT, Origer J, Aasland D, Kaina B. c-Fos is required for excision repair of UV-light induced DNA lesions by triggering the re-synthesis of XPF. Nucleic Acids Res 2006; 34:6530-9. [PMID: 17130154 PMCID: PMC1702502 DOI: 10.1093/nar/gkl895] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cells deficient in c-Fos are hypersensitive to ultraviolet (UV-C) light. Here we demonstrate that mouse embryonic fibroblasts lacking c-Fos (fos−/−) are defective in the repair of UV-C induced DNA lesions. They show a decreased rate of sealing of repair-mediated DNA strand breaks and are unable to remove cyclobutane pyrimidine dimers from DNA. A search for genes responsible for the DNA repair defect revealed that upon UV-C treatment the level of xpf and xpg mRNA declined but, in contrast to the wild type (wt), did not recover in fos−/− cells. The observed decline in xpf and xpg mRNA is due to impaired re-synthesis, as shown by experiments using actinomycin D. Block of xpf transcription resulted in a lack of XPF protein after irradiation of fos−/− cells, whereas the XPF level normalized quickly in the wt. Although the xpg mRNA level was reduced, the amount of XPG protein was not altered in c-Fos-deficient cells after UV-C, due to higher stability of the XPG protein. The data suggest a new role for c-Fos in cells exposed to genotoxic stress. Being part of the transcription factor AP-1, c-Fos stimulates NER via the upregulation of xpf and thus plays a central role in the recovery of cells from UV light induced DNA damage.
Collapse
Affiliation(s)
| | | | | | | | - Bernd Kaina
- To whom correspondence should be addressed. Tel: +49 6131 393 3246; Fax: +49 6131 230506;
| |
Collapse
|
44
|
Jascur T, Boland CR. Structure and function of the components of the human DNA mismatch repair system. Int J Cancer 2006; 119:2030-5. [PMID: 16804905 DOI: 10.1002/ijc.22023] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
DNA mismatch repair (MMR) is one of the several enzyme systems involved in DNA homeostasis. DNA MMR is involved in the repair of specific types of errors that occur during new DNA synthesis; loss of this system leads to an accelerated accumulation of potential mutations, and predisposes to certain types of cancers. Germline mutations in some of the DNA MMR genes cause the hereditary cancer predisposition, Lynch syndrome. This review addresses advances in the biochemistry of DNA MMR and its relationship to carcinogenesis.
Collapse
Affiliation(s)
- Thomas Jascur
- Department of Internal Medicine, Baylor Research Institute, Baylor University Medical Center, Dallas, TX 75246, USA.
| | | |
Collapse
|
45
|
Schroering AG, Edelbrock MA, Richards TJ, Williams KJ. The cell cycle and DNA mismatch repair. Exp Cell Res 2006; 313:292-304. [PMID: 17157834 DOI: 10.1016/j.yexcr.2006.10.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2006] [Revised: 10/13/2006] [Accepted: 10/16/2006] [Indexed: 11/22/2022]
Abstract
The DNA mismatch repair (MMR) pathway contributes to the fidelity of DNA synthesis and recombination by correcting mispaired nucleotides and insertion/deletion loops (IDLs). We have investigated whether MMR protein expression, activity, and subcellular location are altered during discrete phases of the cell cycle in mammalian cells. Two distinct methods have been used to demonstrate that although physiological MMR protein expression, mismatch binding, and nick-directed MMR activity within the nucleus are at highest levels during S phase, MMR is active throughout the cell cycle. Despite equal MMR nuclear protein concentrations in S and G(2) phases, mismatch binding and repair activities within G(2) are significantly lower, indicating a post-translational decrease in MMR activity specific to G(2). We further demonstrate that typical co-localization of MutSalpha to late S phase replication foci can be disrupted by 2 microM N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). This concentration of MNNG does not decrease ongoing DNA synthesis nor induce cell cycle arrest until the second cell cycle, with long-term colony survival decreased by only 24%. These results suggest that low level alkylation damage can selectively disrupt MMR proofreading activity during DNA synthesis and potentially increase mutation frequency within surviving cells.
Collapse
Affiliation(s)
- Allen G Schroering
- Department of Biochemistry and Cancer Biology, Medical University of Ohio, Toledo, OH 43614-5804, USA
| | | | | | | |
Collapse
|
46
|
Keane OM, Zadissa A, Wilson T, Hyndman DL, Greer GJ, Baird DB, McCulloch AF, Crawford AM, McEwan JC. Gene expression profiling of naïve sheep genetically resistant and susceptible to gastrointestinal nematodes. BMC Genomics 2006; 7:42. [PMID: 16515715 PMCID: PMC1450279 DOI: 10.1186/1471-2164-7-42] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Accepted: 03/06/2006] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Gastrointestinal nematodes constitute a major cause of morbidity and mortality in grazing ruminants. Individual animals or breeds, however, are known to differ in their resistance to infection. Gene expression profiling allows us to examine large numbers of transcripts simultaneously in order to identify those transcripts that contribute to an animal's susceptibility or resistance. RESULTS With the goal of identifying genes with a differential pattern of expression between sheep genetically resistant and susceptible to gastrointestinal nematodes, a 20,000 spot ovine cDNA microarray was constructed. This array was used to interrogate the expression of 9,238 known genes in duodenum tissue of four resistant and four susceptible female lambs. Naïve animals were used in order to look at genes that were differentially expressed in the absence of infection with gastrointestinal nematodes. Forty one unique known genes were identified that were differentially expressed between the resistant and susceptible animals. Northern blotting of a selection of the genes confirmed differential expression. The differentially expressed genes had a variety of functions, although many genes relating to the stress response and response to stimulus were more highly expressed in the susceptible animals. CONCLUSION We have constructed the first reported ovine microarray and used this array to examine gene expression in lambs genetically resistant and susceptible to gastrointestinal nematode infection. This study indicates that susceptible animals appear to be generating a hyper-sensitive immune response to non-nematode challenges. The gastrointestinal tract of susceptible animals is therefore under stress and compromised even in the absence of gastrointestinal nematodes. These factors may contribute to the genetic susceptibility of these animals.
Collapse
Affiliation(s)
- Orla M Keane
- AgResearch Molecular Biology Unit, Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Amonida Zadissa
- AgResearch Molecular Biology Unit, Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Theresa Wilson
- AgResearch Molecular Biology Unit, Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Dianne L Hyndman
- AgResearch Molecular Biology Unit, Department of Biochemistry, University of Otago, Dunedin, New Zealand
- AgResearch Invermay Agricultural Centre, Mosgiel, New Zealand
| | - Gordon J Greer
- AgResearch Invermay Agricultural Centre, Mosgiel, New Zealand
| | | | | | | | - John C McEwan
- AgResearch Invermay Agricultural Centre, Mosgiel, New Zealand
| |
Collapse
|
47
|
Christmann M, Tomicic MT, Origer J, Kaina B. Fen1 is induced p53 dependently and involved in the recovery from UV-light-induced replication inhibition. Oncogene 2006; 24:8304-13. [PMID: 16103874 DOI: 10.1038/sj.onc.1208994] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mouse embryonic fibroblasts (MEFs) that lack p53 are hypersensitive to the cytotoxic and genotoxic effect of ultraviolet (UV-C) light. They also display a defect in the recovery from UV-C-induced DNA replication inhibition. An enzyme involved in processing stalled DNA replication forks is flap endonuclease 1 (Fen1). Gene expression profiling of UV-C-irradiated MEFs revealed fen1 to be upregulated, which was confirmed by RT-PCR and Western blot experiments. Increased Fen1 levels upon UV-C exposure are due to transcriptional activation, as revealed by inhibitor studies. Fen1 induction was dose- and time-dependent; it occurred on protein level already 3 h after irradiation. Induction of Fen1 by UV-C requires p53 since it was observed in p53 wild-type (wt) but not in p53 null (p53-/-) fibroblasts. Fen1 upregulation paralleled the increase in p53 protein level in replicating wt cells, whereas in nonreplicating cells both Fen1 and p53 were not induced by UV-C. The mouse fen1 promoter was cloned and shown to harbor a p53 consensus sequence to which p53 binds. In cotransfection experiments, p53 stimulated the expression of a fen1 promoter-reporter construct. Transgenic expression of Fen1 in p53 null cells attenuated UV-C light-induced DNA replication inhibition, supporting the hypothesis that Fen1 induction is involved in the recovery of cells from DNA damage.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University of Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | | | | | | |
Collapse
|
48
|
Abstract
Loss of DNA mismatch repair (MMR) in mammalian cells, as well as having a causative role in cancer, has been linked to resistance to certain DNA damaging agents including clinically important cytotoxic chemotherapeutics. MMR-deficient cells exhibit defects in G2/M cell cycle arrest and cell killing when treated with these agents. MMR-dependent cell cycle arrest occurs, at least for low doses of alkylating agents, only after the second S-phase following DNA alkylation, suggesting that two rounds of DNA replication are required to generate a checkpoint signal. These results point to an indirect role for MMR proteins in damage signalling where aberrant processing of mismatches leads to the generation of DNA structures (single-strand gaps and/or double-strand breaks) that provoke checkpoint activation and cell killing. Significantly, recent studies have revealed that the role of MMR proteins in mismatch repair can be uncoupled from the MMR-dependent damage responses. Thus, there is a threshold of expression of MSH2 or MLH1 required for proper checkpoint and cell-death signalling, even though sub-threshold levels are sufficient for fully functional MMR repair activity. Segregation is also revealed through the identification of mutations in MLH1 or MSH2 that provide alleles functional in MMR but not in DNA damage responses and mutations in MSH6 that compromise MMR but not in apoptotic responses to DNA damaging agents. These studies suggest a direct role for MMR proteins in recognizing and signalling DNA damage responses that is independent of the MMR catalytic repair process. How MMR-dependent G2 arrest may link to cell death remains elusive and we speculate that it is perhaps the resolution of the MMR-dependent G2 cell cycle arrest following DNA damage that is important in terms of cell survival.
Collapse
Affiliation(s)
- Vincent O'Brien
- Centre for Oncology and Applied Pharmacology, Cancer Research UK Beatson Laboratories, Garscube Estate, Glasgow G61 1BD, UK.
| | | |
Collapse
|
49
|
Brieger A, Plotz G, Raedle J, Weber N, Baum W, Caspary WF, Zeuzem S, Trojan J. Characterization of the nuclear import of human MutLalpha. Mol Carcinog 2005; 43:51-8. [PMID: 15754314 DOI: 10.1002/mc.20081] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
DNA mismatch repair (MMR) is essential for the maintenance of replication fidelity. Its major task is to recognize mismatches as well as insertion/deletion loops of newly synthesized DNA strands. Although different players of human MMR have been identified, the regulation of essential steps of MMR is poorly understood. Because MMR is initiated in the nucleus, nuclear import might be a mechanism to regulate MMR. Nuclear targeting is accomplished by conserved signal sequences called nuclear localization signals (NLS), which represent clusters of positively charged amino acids (aa). hMLH1 contains two clusters of positively charged amino acids, which are candidate NLS sequences (aa 469-472 and 496-499), while hPMS2 contains one (aa 574-580). To study the effect of these clusters on nuclear import, NLS mutants of hMLH1 and hPMS2 were generated and expressed in 293T cells. The subcellular localization of the mutant constructs was monitored by confocal laser microscopy. We demonstrated that missense mutations of two signal sequences, one in hMLH1 and one in hPMS2, lead to impaired nuclear import, which was especially prominent for mutants of the hMLH1 residues K471 and R472; and hPMS2 residues K577 and R578.
Collapse
Affiliation(s)
- A Brieger
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt a.M., Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hernandez-Pigeon H, Quillet-Mary A, Louat T, Schambourg A, Humbert O, Selves J, Salles B, Laurent G, Lautier D. hMutSα is Protected from Ubiquitin-proteasome-dependent Degradation by Atypical Protein Kinase Cζ Phosphorylation. J Mol Biol 2005; 348:63-74. [PMID: 15808853 DOI: 10.1016/j.jmb.2005.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Revised: 01/17/2005] [Accepted: 02/01/2005] [Indexed: 10/25/2022]
Abstract
The hMutS alpha (hMSH2-hMSH6) protein heterodimer plays a critical role in the detection of DNA mispairs in the mismatch repair (MMR) process. We recently reported that hMutS alpha proteins were degraded by the ubiquitin-proteasome pathway in a cell-type-dependent manner, indicating that one or several regulator(s) may interfere with hMutS alpha protein ubiquitination and degradation. On the other hand, we and others have shown that protein kinase C (PKC) is involved as a positive regulator of MMR activity. Here, we provide evidence that the atypical PKC zeta regulates ubiquitination, degradation, and levels of hMutS alpha proteins. Using both PKC zeta-transfected U937 and PKC zeta siRNA-transfected MRC-5 cell lines, we found that PKC zeta protein expression was correlated with that of hMutS alpha as well as with MMR activity, but was inversely correlated with hMutS alpha protein ubiquitination and degradation. Interestingly, PKC zeta interacts with hMSH2 and hMSH6 proteins and phosphorylates both. Moreover, in an in vitro assay PKCzeta mediates phosphorylation events decreasing hMutS alpha protein degradation via the ubiquitin-proteasome pathway. Altogether, our results indicate that PKC zeta modulates hMutS alpha stability and protein levels, and suggest a role for PKC zeta in genome stability by regulating MMR activity.
Collapse
Affiliation(s)
- Hélène Hernandez-Pigeon
- INSERM U563, CPTP, Bat B, Pavillon Lefebvre, Place du Dr Baylac, CHU PURPAN, BP 3028, 31024 Toulouse cedex 3, France
| | | | | | | | | | | | | | | | | |
Collapse
|