1
|
Obata H, Ogawa M, Zalutsky MR. DNA Repair Inhibitors: Potential Targets and Partners for Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1926. [PMID: 37514113 PMCID: PMC10384049 DOI: 10.3390/pharmaceutics15071926] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The present review aims to explore the potential targets/partners for future targeted radionuclide therapy (TRT) strategies, wherein cancer cells often are not killed effectively, despite receiving a high average tumor radiation dose. Here, we shall discuss the key factors in the cancer genome, especially those related to DNA damage response/repair and maintenance systems for escaping cell death in cancer cells. To overcome the current limitations of TRT effectiveness due to radiation/drug-tolerant cells and tumor heterogeneity, and to make TRT more effective, we propose that a promising strategy would be to target the DNA maintenance factors that are crucial for cancer survival. Considering their cancer-specific DNA damage response/repair ability and dysregulated transcription/epigenetic system, key factors such as PARP, ATM/ATR, amplified/overexpressed transcription factors, and DNA methyltransferases have the potential to be molecular targets for Auger electron therapy; moreover, their inhibition by non-radioactive molecules could be a partnering component for enhancing the therapeutic response of TRT.
Collapse
Affiliation(s)
- Honoka Obata
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Departments of Radiology and Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Mikako Ogawa
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Michael R Zalutsky
- Departments of Radiology and Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
2
|
Structural basis of the (in)activity of the apical DNA damage response kinases ATM, ATR and DNA-PKcs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 163:120-129. [DOI: 10.1016/j.pbiomolbio.2020.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
|
3
|
Xie X, Zhang Y, Wang Z, Wang S, Jiang X, Cui H, Zhou T, He Z, Feng H, Guo Q, Song X, Cao L. ATM at the crossroads of reactive oxygen species and autophagy. Int J Biol Sci 2021; 17:3080-3090. [PMID: 34421351 PMCID: PMC8375236 DOI: 10.7150/ijbs.63963] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/16/2021] [Indexed: 01/08/2023] Open
Abstract
Reactive oxygen species (ROS) are generally small, short-lived and highly reactive molecules, initially thought to be a pathological role in the cell. A growing amount of evidence in recent years argues for ROS functioning as a signaling intermediate to facilitate cellular adaptation in response to pathophysiological stress through the regulation of autophagy. Autophagy is an essential cellular process that plays a crucial role in recycling cellular components and damaged organelles to eliminate sources of ROS in response to various stress conditions. A large number of studies have shown that DNA damage response (DDR) transducer ataxia-telangiectasia mutated (ATM) protein can also be activated by ROS, and its downstream signaling pathway is involved in autophagy regulation. This review aims at providing novel insight into the regulatory mechanism of ATM activated by ROS and its molecular basis for inducing autophagy, and revealing a new function that ATM can not only maintain genome homeostasis in the nucleus, but also as a ROS sensor trigger autophagy to maintain cellular homeostasis in the cytoplasm.
Collapse
Affiliation(s)
- Xiaochen Xie
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Ye Zhang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Zhuo Wang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Shanshan Wang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaoyou Jiang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Hongyan Cui
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Tingting Zhou
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Zheng He
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Hao Feng
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Qiqiang Guo
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaoyu Song
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Liu Cao
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| |
Collapse
|
4
|
Ovejero S, Bueno A, Sacristán MP. Working on Genomic Stability: From the S-Phase to Mitosis. Genes (Basel) 2020; 11:E225. [PMID: 32093406 PMCID: PMC7074175 DOI: 10.3390/genes11020225] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/15/2022] Open
Abstract
Fidelity in chromosome duplication and segregation is indispensable for maintaining genomic stability and the perpetuation of life. Challenges to genome integrity jeopardize cell survival and are at the root of different types of pathologies, such as cancer. The following three main sources of genomic instability exist: DNA damage, replicative stress, and chromosome segregation defects. In response to these challenges, eukaryotic cells have evolved control mechanisms, also known as checkpoint systems, which sense under-replicated or damaged DNA and activate specialized DNA repair machineries. Cells make use of these checkpoints throughout interphase to shield genome integrity before mitosis. Later on, when the cells enter into mitosis, the spindle assembly checkpoint (SAC) is activated and remains active until the chromosomes are properly attached to the spindle apparatus to ensure an equal segregation among daughter cells. All of these processes are tightly interconnected and under strict regulation in the context of the cell division cycle. The chromosomal instability underlying cancer pathogenesis has recently emerged as a major source for understanding the mitotic processes that helps to safeguard genome integrity. Here, we review the special interconnection between the S-phase and mitosis in the presence of under-replicated DNA regions. Furthermore, we discuss what is known about the DNA damage response activated in mitosis that preserves chromosomal integrity.
Collapse
Affiliation(s)
- Sara Ovejero
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca-CSIC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Institute of Human Genetics, CNRS, University of Montpellier, 34000 Montpellier, France
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France
| | - Avelino Bueno
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca-CSIC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Departamento de Microbiología y Genética, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - María P. Sacristán
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca-CSIC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Departamento de Microbiología y Genética, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
5
|
Wei Y, Chen L, Xu H, Xie C, Zhou Y, Zhou F. Mitochondrial Dysfunctions Regulated Radioresistance through Mitochondria-to-Nucleus Retrograde Signaling Pathway of NF-κB/PI3K/AKT2/mTOR. Radiat Res 2018; 190:204-215. [PMID: 29863983 DOI: 10.1667/rr15021.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We investigated the relationship between significantly different genes of the mitochondria-to-nucleus retrograde signaling pathway (RTG) in H1299 ρ0 cells (mtDNA depleted cell) and compared their radiosensitivity to that of parental ρ+ cells, to determine the possible intervention targets of radiosensitization. ρ0 cells were depleted of mitochondrial DNA by chronic culturing in ethidium bromide at low concentration. Radiosensitivity was analyzed using clonogenic assay. Western blot was used to analyze the cell cycle-related proteins, serine/threonine kinase ataxia telangiectasia mutant (ATM), ataxia telangiectasia and Rad3-related protein (ATR) and cyclin B1 (CCNB1). The γ-H2AX foci were detected using confocal fluorescence microscopy. RNA samples were hybridized using the Agilent human genome expression microarray. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used for Gene Ontology (GO) Consortium and pathway annotations of differentially expressed genes, respectively. The H1299 ρ0 cells were found to be more radioresistant than ρ+ cells. The ATP production of H1299 ρ0 cells was lower than that of the ρ+ cells before or after irradiation. Both H1299 ρ0 and ρ+ cells had higher ROS levels after irradiation, however, the radiation-induced ROS production in ρ0 cells was significantly lower than in ρ+ cells. In addition, the percentage of apoptosis in H1299 ρ0 cells was lower than in ρ+ cells after 6 Gy irradiation. As for the cell cycle and DNA damage response-related proteins ATM, ATR and CCNB1, the expression levels in ρ0 cells were significantly higher than in ρ+ cells, and there were less γ-H2AX foci in the ρ0 than ρ+ cells after irradiation. Furthermore, the results of the human genome expression microarray demonstrated that the phosphorylated protein levels of the NF-κB/PI3K/AKT2/mTOR signaling pathway were increased after 6 Gy irradiation and were decreased after treatment with the AKT2-specific inhibitor MK-2206 combined with radiation in H1299 ρ0 cells. MK-2206 treatment also led to an increase in pro-apoptotic proteins. In conclusion, these results demonstrate that mtDNA depletion might activate the mitochondria-to-nucleus retrograde signaling pathway of NF-κB/PI3K/AKT2/mTOR and induce radioresistance in H1299 ρ0 cells by evoking mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Yuehua Wei
- a Department of Radiation and Medical Oncology.,b Hubei Key Laboratory of Tumor Biological Behaviors.,d Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lulu Chen
- a Department of Radiation and Medical Oncology.,b Hubei Key Laboratory of Tumor Biological Behaviors.,c Hubei Clinical Cancer Study Centre, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,d Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hui Xu
- a Department of Radiation and Medical Oncology.,b Hubei Key Laboratory of Tumor Biological Behaviors.,c Hubei Clinical Cancer Study Centre, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Conghua Xie
- a Department of Radiation and Medical Oncology.,b Hubei Key Laboratory of Tumor Biological Behaviors.,c Hubei Clinical Cancer Study Centre, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Yunfeng Zhou
- a Department of Radiation and Medical Oncology.,b Hubei Key Laboratory of Tumor Biological Behaviors.,c Hubei Clinical Cancer Study Centre, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Fuxiang Zhou
- a Department of Radiation and Medical Oncology.,b Hubei Key Laboratory of Tumor Biological Behaviors.,c Hubei Clinical Cancer Study Centre, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
6
|
Durant ST, Zheng L, Wang Y, Chen K, Zhang L, Zhang T, Yang Z, Riches L, Trinidad AG, Fok JHL, Hunt T, Pike KG, Wilson J, Smith A, Colclough N, Reddy VP, Sykes A, Janefeldt A, Johnström P, Varnäs K, Takano A, Ling S, Orme J, Stott J, Roberts C, Barrett I, Jones G, Roudier M, Pierce A, Allen J, Kahn J, Sule A, Karlin J, Cronin A, Chapman M, Valerie K, Illingworth R, Pass M. The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models. SCIENCE ADVANCES 2018; 4:eaat1719. [PMID: 29938225 PMCID: PMC6010333 DOI: 10.1126/sciadv.aat1719] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/15/2018] [Indexed: 06/08/2023]
Abstract
Poor survival rates of patients with tumors arising from or disseminating into the brain are attributed to an inability to excise all tumor tissue (if operable), a lack of blood-brain barrier (BBB) penetration of chemotherapies/targeted agents, and an intrinsic tumor radio-/chemo-resistance. Ataxia-telangiectasia mutated (ATM) protein orchestrates the cellular DNA damage response (DDR) to cytotoxic DNA double-strand breaks induced by ionizing radiation (IR). ATM genetic ablation or pharmacological inhibition results in tumor cell hypersensitivity to IR. We report the primary pharmacology of the clinical-grade, exquisitely potent (cell IC50, 0.78 nM), highly selective [>10,000-fold over kinases within the same phosphatidylinositol 3-kinase-related kinase (PIKK) family], orally bioavailable ATM inhibitor AZD1390 specifically optimized for BBB penetration confirmed in cynomolgus monkey brain positron emission tomography (PET) imaging of microdosed 11C-labeled AZD1390 (Kp,uu, 0.33). AZD1390 blocks ATM-dependent DDR pathway activity and combines with radiation to induce G2 cell cycle phase accumulation, micronuclei, and apoptosis. AZD1390 radiosensitizes glioma and lung cancer cell lines, with p53 mutant glioma cells generally being more radiosensitized than wild type. In in vivo syngeneic and patient-derived glioma as well as orthotopic lung-brain metastatic models, AZD1390 dosed in combination with daily fractions of IR (whole-brain or stereotactic radiotherapy) significantly induced tumor regressions and increased animal survival compared to IR treatment alone. We established a pharmacokinetic-pharmacodynamic-efficacy relationship by correlating free brain concentrations, tumor phospho-ATM/phospho-Rad50 inhibition, apoptotic biomarker (cleaved caspase-3) induction, tumor regression, and survival. On the basis of the data presented here, AZD1390 is now in early clinical development for use as a radiosensitizer in central nervous system malignancies.
Collapse
Affiliation(s)
- Stephen T Durant
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, UK
| | - Li Zheng
- Bioscience, Innovative Cancer Centre, Oncology IMED Biotech Unit, AstraZeneca, Shanghai, China
| | - Yingchun Wang
- Bioscience, Innovative Cancer Centre, Oncology IMED Biotech Unit, AstraZeneca, Shanghai, China
| | - Kan Chen
- Drug Metabolism and Pharmacokinetics, Innovative Cancer Centre, Oncology IMED Biotech Unit, AstraZeneca, Shanghai, China
| | - Lingli Zhang
- Drug Metabolism and Pharmacokinetics, Innovative Cancer Centre, Oncology IMED Biotech Unit, AstraZeneca, Shanghai, China
| | - Tianwei Zhang
- Bioscience, Innovative Cancer Centre, Oncology IMED Biotech Unit, AstraZeneca, Shanghai, China
| | - Zhenfan Yang
- Bioscience, Innovative Cancer Centre, Oncology IMED Biotech Unit, AstraZeneca, Shanghai, China
| | - Lucy Riches
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, UK
| | - Antonio G Trinidad
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, UK
| | - Jacqueline H L Fok
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, UK
| | - Tom Hunt
- Chemistry, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Kurt G Pike
- Chemistry, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Joanne Wilson
- Drug Metabolism and Pharmacokinetics, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Aaron Smith
- Drug Metabolism and Pharmacokinetics, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Nicola Colclough
- Drug Metabolism and Pharmacokinetics, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Venkatesh Pilla Reddy
- Drug Metabolism and Pharmacokinetics, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Andrew Sykes
- Drug Metabolism and Pharmacokinetics, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Annika Janefeldt
- Drug Metabolism and Pharmacokinetics, Cardiovascular and Metabolic Diseases IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Peter Johnström
- Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Varnäs
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Stephanie Ling
- Discovery Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Jonathan Orme
- Discovery Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Jonathan Stott
- Discovery Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Caroline Roberts
- Discovery Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Ian Barrett
- Discovery Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Gemma Jones
- Translational Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Martine Roudier
- Translational Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Andrew Pierce
- Translational Sciences, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Jasmine Allen
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298-0058, USA
| | - Jenna Kahn
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298-0058, USA
| | - Amrita Sule
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298-0058, USA
| | - Jeremy Karlin
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298-0058, USA
| | - Anna Cronin
- Drug Safety and Metabolism, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Melissa Chapman
- Drug Safety and Metabolism, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Kristoffer Valerie
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298-0058, USA
| | | | - Martin Pass
- Projects, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| |
Collapse
|
7
|
Baretić D, Pollard HK, Fisher DI, Johnson CM, Santhanam B, Truman CM, Kouba T, Fersht AR, Phillips C, Williams RL. Structures of closed and open conformations of dimeric human ATM. SCIENCE ADVANCES 2017; 3:e1700933. [PMID: 28508083 PMCID: PMC5425235 DOI: 10.1126/sciadv.1700933] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/19/2017] [Indexed: 05/31/2023]
Abstract
ATM (ataxia-telangiectasia mutated) is a phosphatidylinositol 3-kinase-related protein kinase (PIKK) best known for its role in DNA damage response. ATM also functions in oxidative stress response, insulin signaling, and neurogenesis. Our electron cryomicroscopy (cryo-EM) suggests that human ATM is in a dynamic equilibrium between closed and open dimers. In the closed state, the PIKK regulatory domain blocks the peptide substrate-binding site, suggesting that this conformation may represent an inactive or basally active enzyme. The active site is held in this closed conformation by interaction with a long helical hairpin in the TRD3 (tetratricopeptide repeats domain 3) domain of the symmetry-related molecule. The open dimer has two protomers with only a limited contact interface, and it lacks the intermolecular interactions that block the peptide-binding site in the closed dimer. This suggests that the open conformation may be more active. The ATM structure shows the detailed topology of the regulator-interacting N-terminal helical solenoid. The ATM conformational dynamics shown by the structures represent an important step in understanding the enzyme regulation.
Collapse
Affiliation(s)
- Domagoj Baretić
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Hannah K. Pollard
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Darwin Building, Cambridge CB4 0WG, UK
| | - David I. Fisher
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Darwin Building, Cambridge CB4 0WG, UK
| | | | - Balaji Santhanam
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Caroline M. Truman
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Darwin Building, Cambridge CB4 0WG, UK
| | - Tomas Kouba
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Alan R. Fersht
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Christopher Phillips
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Darwin Building, Cambridge CB4 0WG, UK
| | - Roger L. Williams
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| |
Collapse
|
8
|
Hartlerode AJ, Morgan MJ, Wu Y, Buis J, Ferguson DO. Recruitment and activation of the ATM kinase in the absence of DNA-damage sensors. Nat Struct Mol Biol 2015; 22:736-43. [PMID: 26280532 PMCID: PMC4560612 DOI: 10.1038/nsmb.3072] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/22/2015] [Indexed: 12/26/2022]
Abstract
Two kinases, ATM and DNA-PKcs, control rapid responses to DNA double-strand breaks (DSBs). The paradigm for ATM control is recruitment and activation by the Mre11-Rad50-NBS1 (MRN) sensor complex, whereas DNA-PKcs requires the sensor Ku (Ku70-Ku80). Using mouse cells containing targeted mutant alleles of Mre11 (Mre11a) and/or Ku70 (Xrcc6), together with pharmacologic kinase inhibition, we demonstrate that ATM can be activated by DSBs in the absence of MRN. When MRN is deficient, DNA-PKcs efficiently substitutes for ATM in facilitating local chromatin responses. In the absence of both MRN and Ku, ATM is recruited to chromatin, where it phosphorylates H2AX and triggers the G2-M cell-cycle checkpoint, but the DNA-repair functions of MRN are not restored. These results suggest that, in contrast to straightforward recruitment and activation by MRN, a complex interplay between sensors has a substantial role in ATM control.
Collapse
Affiliation(s)
- Andrea J. Hartlerode
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109. USA
| | - Mary J. Morgan
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109. USA
| | - Yipin Wu
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109. USA
| | - Jeffrey Buis
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109. USA
| | - David O. Ferguson
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109. USA
| |
Collapse
|
9
|
Prognostic Significance of Nuclear Phospho-ATM Expression in Melanoma. PLoS One 2015; 10:e0134678. [PMID: 26275218 PMCID: PMC4537129 DOI: 10.1371/journal.pone.0134678] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/13/2015] [Indexed: 12/21/2022] Open
Abstract
UV radiation induced genomic instability is one of the leading causes for melanoma. Phosphorylation of Ataxia Telangiectasia Mutated (ATM) is one of the initial events that follow DNA damage. Phospho-ATM (p-ATM) plays a key role in the activation of DNA repair and several oncogenic pathways as well as in the maintenance of genomic integrity. The present study was therefore performed to understand the significance of p-ATM in melanoma progression and to correlate it with patient prognosis. Tissue microarray and immunohistochemical analysis were employed to study the expression of p-ATM in melanoma patients. A total of 366 melanoma patients (230 primary melanoma and 136 metastatic melanoma) were used for the study. Chi-square test, Kaplan-Meier, univariate and multivariate Cox regression analysis were used to elucidate the prognostic significance of p-ATM expression. Results revealed that both loss of, and gain in, p-ATM expression were associated with progression of melanoma from normal nevi to metastatic melanoma. Patients whose samples showed negative or strong p-ATM staining had significantly worse 5-year survival compared to patients who had weak to moderate expression. Loss of p-ATM expression was associated with relatively better 5-year survival, but the corresponding 10-year survival curve almost overlapped with that of strong p-ATM expression. p-ATM expression was found to be an independent prognostic factor for 5-year but not for 10-year patient survival. In conclusion our findings show that loss of p-ATM expression and gain-in p-ATM expression are indicators of worse melanoma patient survival.
Collapse
|
10
|
Zhang J, Wei W, Jin HC, Ying RC, Zhu AK, Zhang FJ. The roles of APOBEC3B in gastric cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5089-5096. [PMID: 26191203 PMCID: PMC4503075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/20/2015] [Indexed: 06/04/2023]
Abstract
Gastric cancer was the third cause of death in China. In this study, we found that the APOBEC3 (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3) expression was higher in gastric cancer tissues than that in normal tissues and confirmed APOBEC3B expression was correlated with the unfavorable prognosis of the patients with gastric cancer. Furthermore, APOBEC3B expression was associated with gender, tumor size, histological grade, T stage, and TNM staging of the patients with gastric cancer. Down-regulation of APOBEC3B expression in MNK28 cells could enhance the cytotoxicity of PDCD2. No editing took place in PDCD2 positive MKN28 cells with APOBEC3B shRNA. These results indicated that loss of function of PDCD2 may be partly caused by APOBEC3B-induced extensive mutagenesis.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University Hangzhou 310006, P. R. China
| | - Wei Wei
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University Hangzhou 310006, P. R. China
| | - Hui-Cheng Jin
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University Hangzhou 310006, P. R. China
| | - Rong-Chao Ying
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University Hangzhou 310006, P. R. China
| | - A-Kao Zhu
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University Hangzhou 310006, P. R. China
| | - Fang-Jie Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University Hangzhou 310006, P. R. China
| |
Collapse
|
11
|
Notch is a direct negative regulator of the DNA-damage response. Nat Struct Mol Biol 2015; 22:417-24. [PMID: 25895060 DOI: 10.1038/nsmb.3013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/23/2015] [Indexed: 11/08/2022]
Abstract
The DNA-damage response (DDR) ensures genome stability and proper inheritance of genetic information, both of which are essential to survival. It is presently unclear to what extent other signaling pathways modulate DDR function. Here we show that Notch receptor binds and inactivates ATM kinase and that this mechanism is evolutionarily conserved in Caenorhabditis elegans, Xenopus laevis and humans. In C. elegans, the Notch pathway impairs DDR signaling in gonad germ cells. In mammalian cells, activation of human Notch1 leads to reduced ATM signaling in a manner independent of Notch1 transcriptional activity. Notch1 binds directly to the regulatory FATC domain of ATM and inhibits ATM kinase activity. Notch1 and ATM activation are inversely correlated in human breast cancers, and inactivation of ATM by Notch1 contributes to the survival of Notch1-driven leukemia cells upon DNA damage.
Collapse
|
12
|
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase is a master regulator of the DNA damage response, and it coordinates checkpoint activation, DNA repair, and metabolic changes in eukaryotic cells in response to DNA double-strand breaks and oxidative stress. Loss of ATM activity in humans results in the pleiotropic neurodegeneration disorder ataxia-telangiectasia. ATM exists in an inactive state in resting cells but can be activated by the Mre11-Rad50-Nbs1 (MRN) complex and other factors at sites of DNA breaks. In addition, oxidation of ATM activates the kinase independently of the MRN complex. This review discusses these mechanisms of activation, as well as the posttranslational modifications that affect this process and the cellular factors that affect the efficiency and specificity of ATM activation and substrate phosphorylation. I highlight functional similarities between the activation mechanisms of ATM, phosphatidylinositol 3-kinases (PI3Ks), and the other PI3K-like kinases, as well as recent structural insights into their regulation.
Collapse
Affiliation(s)
- Tanya T Paull
- Howard Hughes Medical Institute, Department of Molecular Biosciences, and Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas 78712;
| |
Collapse
|
13
|
Zhang J, Wei W, Jin HC, Ying RC, Zhu AK, Zhang FJ. Programmed cell death 2 protein induces gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis in a p53-dependent manner. Oncol Rep 2014; 33:103-10. [PMID: 25334010 DOI: 10.3892/or.2014.3551] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/02/2014] [Indexed: 11/06/2022] Open
Abstract
Programmed cell death 2 (PDCD2) is a highly conserved nuclear protein, and aberrant PDCD2 expression alters cell apoptosis. The present study aimed to investigate PDCD2 expression in gastric cancer. Tissue specimens from 34 gastric cancer patients were collected for analysis of PDCD2 expression using immunohistochemistry, western blotting and qRT-PCR. Gastric cancer cell lines (a p53-mutated MKN28 line and a wild-type p53 MKN45 line) were used to assess the effects of PDCD2 overexpression. p53-/- nude mice were used to investigate the effect of PDCD2 on ultraviolet B (UVB)-induced skin carcinogenesis. The data showed that PDCD2 expression was reduced in gastric cancer tissue specimens, and loss of PDCD2 expression was associated with the poor survival of patients. PDCD2 expression induced gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis. The antitumor effects of PDCD2 expression were dependent on p53 expression in gastric cancer cells. Moreover, PDCD2 expression inhibited activity of the ATM/Chk1/2/p53 signaling pathway. In addition, PDCD2 expression suppressed UVB-induced skin carcinogenesis in p53+/+ nude mice, but not in p53-/- mice. The data from the present study demonstrated that loss of PDCD2 expression could contribute to gastric cancer development and progression and that PDCD2-induced gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis are p53-dependent.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Wei Wei
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Hui-Cheng Jin
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Rong-Chao Ying
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - A-Kao Zhu
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Fang-Jie Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| |
Collapse
|
14
|
Zhao H, Yang Y, Janga SC, Kao CC, Zhou Y. Prediction and validation of the unexplored RNA-binding protein atlas of the human proteome. Proteins 2014; 82:640-7. [PMID: 24123256 PMCID: PMC3949140 DOI: 10.1002/prot.24441] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/13/2013] [Accepted: 09/26/2013] [Indexed: 12/13/2022]
Abstract
Detecting protein-RNA interactions is challenging both experimentally and computationally because RNAs are large in number, diverse in cellular location and function, and flexible in structure. As a result, many RNA-binding proteins (RBPs) remain to be identified. Here, a template-based, function-prediction technique SPOT-Seq for RBPs is applied to human proteome and its result is validated by a recent proteomic experimental discovery of 860 mRNA-binding proteins (mRBPs). The coverage (or sensitivity) is 42.6% for 1217 known RBPs annotated in the Gene Ontology and 43.6% for 860 newly discovered human mRBPs. Consistent sensitivity indicates the robust performance of SPOT-Seq for predicting RBPs. More importantly, SPOT-Seq detects 2418 novel RBPs in human proteome, 291 of which were validated by the newly discovered mRBP set. Among 291 validated novel RBPs, 61 are not homologous to any known RBPs. Successful validation of predicted novel RBPs permits us to further analysis of their phenotypic roles in disease pathways. The dataset of 2418 predicted novel RBPs along with confidence levels and complex structures is available at http://sparks-lab.org (in publications) for experimental confirmations and hypothesis generation.
Collapse
Affiliation(s)
- Huiying Zhao
- School of Informatics, Indiana University Purdue University, Indianapolis, Indiana, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 719 Indiana Ave Ste 319, Walker Plaza Building, Indianapolis, Indiana 46202, USA
| | - Yuedong Yang
- School of Informatics, Indiana University Purdue University, Indianapolis, Indiana, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 719 Indiana Ave Ste 319, Walker Plaza Building, Indianapolis, Indiana 46202, USA
- Institute for Glycomics and School of Informatics and Communication Technology, Griffith University, Parklands Dr., Southport, QLD4215, Australia
| | - Sarath Chandra Janga
- School of Informatics, Indiana University Purdue University, Indianapolis, Indiana, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 719 Indiana Ave Ste 319, Walker Plaza Building, Indianapolis, Indiana 46202, USA
| | - C. Cheng Kao
- Department of Molecular & Cellular Biochemistry, Indiana University, Bloomington, Indiana, 47405, USA
| | - Yaoqi Zhou
- School of Informatics, Indiana University Purdue University, Indianapolis, Indiana, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 719 Indiana Ave Ste 319, Walker Plaza Building, Indianapolis, Indiana 46202, USA
- Institute for Glycomics and School of Informatics and Communication Technology, Griffith University, Parklands Dr., Southport, QLD4215, Australia
| |
Collapse
|
15
|
González LN, Arruda-Neto JDT, Cotta MA, Carrer H, Garcia F, Silva RAS, Moreau ALD, Righi H, Genofre GC. DNA fragmentation by gamma radiation and electron beams using atomic force microscopy. J Biol Phys 2012; 38:531-42. [PMID: 23729912 DOI: 10.1007/s10867-012-9270-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 04/24/2012] [Indexed: 10/28/2022] Open
Abstract
Double-stranded pBS plasmid DNA was irradiated with gamma rays at doses ranging from 1 to 12 kGy and electron beams from 1 to 10 kGy. Fragment-size distributions were determined by direct visualization, using atomic force microscopy with nanometer-resolution operating in non-tapping mode, combined with an improved methodology. The fragment distributions from irradiation with gamma rays revealed discrete-like patterns at all doses, suggesting that these patterns are modulated by the base pair composition of the plasmid. Irradiation with electron beams, at very high dose rates, generated continuous distributions of highly shattered DNA fragments, similar to results at much lower dose rates found in the literature. Altogether, these results indicate that AFM could supplement traditional methods for high-resolution measurements of radiation damage to DNA, while providing new and relevant information.
Collapse
Affiliation(s)
- Luis Nieto González
- Departamento de Ciência e Tecnologia, Universidade Estadual de Santa Cruz, Ilhéus, BA Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zhuge C, Chang Y, Li Y, Chen Y, Lei J. PDCD5-regulated cell fate decision after ultraviolet-irradiation-induced DNA damage. Biophys J 2012; 101:2582-91. [PMID: 22261045 DOI: 10.1016/j.bpj.2011.10.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/23/2011] [Accepted: 10/28/2011] [Indexed: 10/14/2022] Open
Abstract
Programmed cell death 5 (PDCD5) is a human apoptosis-related molecule that is involved in both the cytoplasmic caspase-3 activity pathway (by regulating Bax translocation from cytoplasm to mitochondria) and the nuclear pathway (by interacting with Tip60). In this study, we developed a mathematical model of the PDCD5-regulated switching of the cell response from DNA repair to apoptosis after ultraviolet irradiation-induced DNA damage. We established the model by combining several hypotheses with experimental observations. Our simulations indicate that the ultimate cell response to DNA damage is dependent on a signal threshold mechanism, and the PDCD5 promotion of Bax translocation plays an essential role in PDCD5-regulated cell apoptosis. Furthermore, the model simulations revealed that PDCD5 nuclear translocation can attenuate cell apoptosis, and PDCD5 interactions with Tip60 can accelerate DNA damage-induced apoptosis, but the final cell fate decision is insensitive to the PDCD5-Tip60 interaction. These results are consistent with experimental observations. The effect of recombinant human PDCD5 was also investigated and shown to sensitize cells to DNA damage by promoting caspase-3 activity.
Collapse
Affiliation(s)
- Changjing Zhuge
- Zhou Pei-Yuan Center for Applied Mathematics, Tsinghua University, Beijing, China
| | | | | | | | | |
Collapse
|
17
|
Davis AJ, So S, Chen DJ. Dynamics of the PI3K-like protein kinase members ATM and DNA-PKcs at DNA double strand breaks. Cell Cycle 2011; 9:2529-36. [PMID: 20543558 DOI: 10.4161/cc.9.13.12148] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The protein kinases ATM and DNA-PKcs play critical roles in the cellular response to DNA double strand breaks (DSBs). ATM and DNA-PKcs are activated in response to DSBs and play several important roles in propagation of the damage signal and for the repair of DNA damage. Recent work from several groups, including ours, has focused on studying the dynamics of each of these proteins at DSBs and the requirements and factors which play a role(s) in this process. The use of live cell imaging of fluorescently-tagged ATM and DNA-PKcs has allowed us to study the real-time response of these proteins to laser-generated DNA damage in vivo. Here, we will extensively discuss the behavior of the ATM and DNA-PKcs proteins at DSB sites.
Collapse
Affiliation(s)
- Anthony J Davis
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | |
Collapse
|
18
|
Rodriguez-Rocha H, Aracely-Garcia-Garcia, Panayiotidis MI, Franco R. DNA damage and autophagy. Mutat Res 2011; 711:158-66. [PMID: 21419786 PMCID: PMC3105359 DOI: 10.1016/j.mrfmmm.2011.03.007] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 03/04/2011] [Accepted: 03/11/2011] [Indexed: 12/15/2022]
Abstract
Both exogenous and endogenous agents are a threat to DNA integrity. Exogenous environmental agents such as ultraviolet (UV) and ionizing radiation, genotoxic chemicals and endogenous byproducts of metabolism including reactive oxygen species can cause alterations in DNA structure (DNA damage). Unrepaired DNA damage has been linked to a variety of human disorders including cancer and neurodegenerative disease. Thus, efficient mechanisms to detect DNA lesions, signal their presence and promote their repair have been evolved in cells. If DNA is effectively repaired, DNA damage response is inactivated and normal cell functioning resumes. In contrast, when DNA lesions cannot be removed, chronic DNA damage triggers specific cell responses such as cell death and senescence. Recently, DNA damage has been shown to induce autophagy, a cellular catabolic process that maintains a balance between synthesis, degradation, and recycling of cellular components. But the exact mechanisms by which DNA damage triggers autophagy are unclear. More importantly, the role of autophagy in the DNA damage response and cellular fate is unknown. In this review we analyze evidence that supports a role for autophagy as an integral part of the DNA damage response.
Collapse
Affiliation(s)
- Humberto Rodriguez-Rocha
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences. University of Nebraska-Lincoln. Lincoln, NE 68583
| | - Aracely-Garcia-Garcia
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences. University of Nebraska-Lincoln. Lincoln, NE 68583
| | | | - Rodrigo Franco
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences. University of Nebraska-Lincoln. Lincoln, NE 68583
| |
Collapse
|
19
|
Kemp MG, Lindsey-Boltz LA, Sancar A. The DNA damage response kinases DNA-dependent protein kinase (DNA-PK) and ataxia telangiectasia mutated (ATM) Are stimulated by bulky adduct-containing DNA. J Biol Chem 2011; 286:19237-46. [PMID: 21487018 DOI: 10.1074/jbc.m111.235036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A variety of environmental, carcinogenic, and chemotherapeutic agents form bulky lesions on DNA that activate DNA damage checkpoint signaling pathways in human cells. To identify the mechanisms by which bulky DNA adducts induce damage signaling, we developed an in vitro assay using mammalian cell nuclear extract and plasmid DNA containing bulky adducts formed by N-acetoxy-2-acetylaminofluorene or benzo(a)pyrene diol epoxide. Using this cell-free system together with a variety of pharmacological, genetic, and biochemical approaches, we identified the DNA damage response kinases DNA-dependent protein kinase (DNA-PK) and ataxia telangiectasia mutated (ATM) as bulky DNA damage-stimulated kinases that phosphorylate physiologically important residues on the checkpoint proteins p53, Chk1, and RPA. Consistent with these results, purified DNA-PK and ATM were directly stimulated by bulky adduct-containing DNA and preferentially associated with damaged DNA in vitro. Because the DNA damage response kinase ATM and Rad3-related (ATR) is also stimulated by bulky DNA adducts, we conclude that a common biochemical mechanism exists for activation of DNA-PK, ATM, and ATR by bulky adduct-containing DNA.
Collapse
Affiliation(s)
- Michael G Kemp
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | | | | |
Collapse
|
20
|
Modifications of p53 and the DNA damage response in cells expressing mutant form of the protein huntingtin. J Mol Neurosci 2011; 45:256-68. [PMID: 21465263 DOI: 10.1007/s12031-011-9516-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 03/16/2011] [Indexed: 10/18/2022]
Abstract
Huntington's disease (HD) occurs through an expansion of the trinucleotide repeat in the HD gene resulting in the lengthening of the polyglutamine stretch within the N terminus of the protein, huntingtin (Htt). While the function of the protein is still being fully elucidated, we have shown that genomic DNA damage is associated with the expression of mutant Htt (mHtt) in a time-dependent fashion. With the accumulation of mHtt and its development into a micro-aggregated complex, the initiation of genomic damage engages a cellular stress signal that activates the DNA damage and stress response pathway. Here we explore the modifications and activation of p53 and keystone regulators of the cell stress response pathway using expression of a fragment of mHtt in HEK293T cells. We find an increase in phosphorylated p53 at serine 15 (S15), diminished acetylation at lysine 382 (K382), altered ubiquitination pattern, and oligomerization activity as a function of mHtt expression. As one might predict, upstream regulators of p53, such as CREB-binding protein/p300 and MDM2, are also seen to be affected by the expression of mHtt, albeit in different ways. These data suggest a possible relationship between p53 and the slow accumulation of DNA damage resulting from the expression of mHtt. The lack of a proper p53-mediated signaling cascade or its alteration in the presence of DNA damage may contribute to the slow progression of cellular dysfunction which is a hallmark of HD pathology.
Collapse
|
21
|
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase is activated by DNA double-strand breaks (DSBs) through the Mre11-Rad50-Nbs1 (MRN) DNA repair complex and orchestrates signaling cascades that initiate the DNA damage response. Cells lacking ATM are also hypersensitive to insults other than DSBs, particularly oxidative stress. We show that oxidation of ATM directly induces ATM activation in the absence of DNA DSBs and the MRN complex. The oxidized form of ATM is a disulfide-cross-linked dimer, and mutation of a critical cysteine residue involved in disulfide bond formation specifically blocked activation through the oxidation pathway. Identification of this pathway explains observations of ATM activation under conditions of oxidative stress and shows that ATM is an important sensor of reactive oxygen species in human cells.
Collapse
Affiliation(s)
- Zhi Guo
- Howard Hughes Medical Institute, Department of Molecular Genetics and Microbiology, and Institute for Cellular and Molecular Biology (ICMB), University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | | | |
Collapse
|
22
|
Walker M, Black EJ, Oehler V, Gillespie DA, Scott MT. Chk1 C-terminal regulatory phosphorylation mediates checkpoint activation by de-repression of Chk1 catalytic activity. Oncogene 2009; 28:2314-23. [PMID: 19421147 PMCID: PMC2857325 DOI: 10.1038/onc.2009.102] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chk1 is phosphorylated within its C-terminal regulatory domain by the upstream ATM/ ATR kinases during checkpoint activation, however how this modulates Chk1 function is poorly understood. Here, we show that Chk1 kinase activity is rapidly stimulated in a cell cycle phase-specific manner in response to both DNA damage and replication arrest, and that the extent and duration of activation correlates closely with regulatory phosphorylation at serines (S) S317, S345, and S366. Despite their evident co-regulation, substitutions of individual Chk1 regulatory sites with alanine (A) residues have differential effects on checkpoint proficiency and kinase activation. Thus, whereas Chk1 S345 is essential for all functions tested, mutants lacking S317 or S366 retain partial proficiency for G2/ M and S/ M checkpoint arrests triggered by DNA damage or replication arrest. These phenotypes reflect defects in Chk1 kinase induction, since the mutants are either partially (317A, 366A) or completely (345A) resistant to kinase activation. Importantly, S345 phosphorylation is impaired in Chk1 S317A and S366A mutants, suggesting that modification of adjacent SQ sites promotes this key regulatory event. Finally, we provide biochemical evidence that Chk1 catalytic activity is stimulated via a de-repression mechanism.
Collapse
Affiliation(s)
- M Walker
- Beatson Institute for Cancer Research, Garscube Estate, Glasgow, UK
| | | | | | | | | |
Collapse
|
23
|
Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signalling and cancer. Nat Rev Mol Cell Biol 2008; 9:759-69. [PMID: 18813293 DOI: 10.1038/nrm2514] [Citation(s) in RCA: 660] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
First described over 80 years ago, ataxia-telangiectasia (A-T) was defined as a clinical entity 50 years ago. Although not encountered by most clinicians, it is a paradigm for cancer predisposition and neurodegenerative disorders and has a central role in our understanding of the DNA-damage response, signal transduction and cell-cycle control. The discovery of the protein A-T mutated (ATM) that is deficient in A-T paved the way for rapid progress on understanding how ATM functions with a host of other proteins to protect against genome instability and reduce the risk of cancer and other pathologies.
Collapse
|
24
|
Chistiakov DA, Voronova NV, Chistiakov PA. Genetic variations in DNA repair genes, radiosensitivity to cancer and susceptibility to acute tissue reactions in radiotherapy-treated cancer patients. Acta Oncol 2008; 47:809-24. [PMID: 18568480 DOI: 10.1080/02841860801885969] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Ionizing radiation is a well established carcinogen for human cells. At low doses, radiation exposure mainly results in generation of double strand breaks (DSBs). Radiation-related DSBs could be directly linked to the formation of chromosomal rearrangements as has been proven for radiation-induced thyroid tumors. Repair of DSBs presumably involves two main pathways, non-homologous end joining (NHEJ) and homologous recombination (HR). A number of known inherited syndromes, such as ataxia telangiectasia, ataxia-telangiectasia like-disorder, radiosensitive severe combined immunodeficiency, Nijmegen breakage syndrome, and LIG4 deficiency are associated with increased radiosensitivity and/or cancer risk. Many of them are caused by mutations in DNA repair genes. Recent studies also suggest that variations in the DNA repair capacity in the general population may influence cancer susceptibility. In this paper, we summarize the current status of DNA repair proteins as potential targets for radiation-induced cancer risk. We will focus on genetic alterations in genes involved in HR- and NHEJ-mediated repair of DSBs, which could influence predisposition to radiation-related cancer and thereby explain interindividual differences in radiosensitivity or radioresistance in a general population.
Collapse
|
25
|
León-Velarde F, Mejía O. Gene Expression in Chronic High Altitude Diseases. High Alt Med Biol 2008; 9:130-9. [DOI: 10.1089/ham.2007.1077] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fabiola León-Velarde
- Laboratorio de Fisiología Comparada, Departamento de Ciencias Biológicas y Fisiológicas, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Olga Mejía
- Laboratorio de Fisiología Comparada, Departamento de Ciencias Biológicas y Fisiológicas, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
26
|
Supreme EnLIGHTenment: damage recognition and signaling in the mammalian UV response. Mol Cell 2008; 29:279-90. [PMID: 18280234 DOI: 10.1016/j.molcel.2008.01.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Indexed: 12/21/2022]
Abstract
Like their prokaryotic counterparts, mammalian cells can sense light, especially in the ultraviolet (UV) range of the spectrum. After UV exposure, cells mount an elaborate response--called the UV response--that mimics physiological signaling responses except that it targets multiple pathways, thereby lacking the defined specificity of receptor-triggered signal transduction. Despite many years of research, it is still not fully clear how UV radiation is sensed and converted into the "language of cells"--signal reception and transduction. This review focuses on how photonic energy and its primary cellular products are sensed to elicit the UV response.
Collapse
|
27
|
Gehen SC, Staversky RJ, Bambara RA, Keng PC, O'Reilly MA. hSMG-1 and ATM sequentially and independently regulate the G1 checkpoint during oxidative stress. Oncogene 2008; 27:4065-74. [PMID: 18332866 DOI: 10.1038/onc.2008.48] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Genotoxic stress activates the phosphatidylinositol 3-kinase-like kinases (PIKKs) that phosphorylate proteins involved in cell cycle arrest, DNA repair and apoptosis. Previous work showed that the PIKK ataxia telangiectasia mutated (ATM) but not ATM and Rad3 related phosphorylates p53 (Ser15) during hyperoxia, a model of prolonged oxidative stress and DNA damage. Here, we show hSMG-1 is responsible for the rapid and early phosphorylation of p53 (Ser15) and that ATM helps maintain phosphorylation after 24 h. Despite reduced p53 phosphorylation and abundance in cells depleted of hSMG-1 or ATM, levels of the p53 target p21 were still elevated and the G(1) checkpoint remained intact. Conditional overexpression of p21 in p53-deficient cells revealed that hyperoxia also stimulates wortmannin-sensitive degradation of p21. siRNA depletion of hSMG-1 or ATM restored p21 stability and the G(1) checkpoint during hyperoxia. These findings establish hSMG-1 as a proximal regulator of DNA damage signaling and reveal that the G(1) checkpoint is tightly regulated during prolonged oxidative stress by both PIKK-dependent synthesis and proteolysis of p21.
Collapse
Affiliation(s)
- S C Gehen
- Department of Environmental Medicine, The University of Rochester, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
28
|
Lavin MF, Kozlov S. DNA damage-induced signalling in ataxia-telangiectasia and related syndromes. Radiother Oncol 2007; 83:231-7. [PMID: 17512070 DOI: 10.1016/j.radonc.2007.04.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Accepted: 04/22/2007] [Indexed: 01/16/2023]
Abstract
ATM, the protein mutated in the human genetic disorder ataxia-telangiectasia, functions by responding to radiation damage to DNA, primarily DNA double strand breaks (dsb), to reduce the risk of genome instability, cancer and neurodegeneration. ATM is rapidly activated as an existing protein to phosphorylate a number of downstream proteins that are involved in DNA repair and cell cycle checkpoint activation. While the exact mechanism of activation of ATM has not been determined, it is now evident that it relies heavily on the Mre11 complex (Mre11/Rad50/Nbs1) and a series of post-translational events for this activation. The Mre11 complex acts as a sensor for the break, recruits ATM to this site where it is autophosphorylated and then is capable of phosphorylating substrates that participate in DNA repair and cell cycle control. A greater understanding of how ATM is activated and functions through different signalling pathways is paramount to devising therapeutic strategies for the treatment of A-T patients. This knowledge can also be used to advantage in sensitizing cells to radiation and ultimately deriving novel therapeutic approaches for the treatment of cancer.
Collapse
Affiliation(s)
- Martin F Lavin
- Queensland Institute of Medical Research, Brisbane, Australia; School of Medicine, The University of Queensland, Brisbane, Australia.
| | | |
Collapse
|
29
|
Matei IR, Guidos CJ, Danska JS. ATM-dependent DNA damage surveillance in T-cell development and leukemogenesis: the DSB connection. Immunol Rev 2006; 209:142-58. [PMID: 16448540 DOI: 10.1111/j.0105-2896.2006.00361.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The immune system is capable of recognizing and eliminating an enormous array of pathogens due to the extremely diverse antigen receptor repertoire of T and B lymphocytes. However, the development of lymphocytes bearing receptors with unique specificities requires the generation of programmed double strand breaks (DSBs) coupled with bursts of proliferation, rendering lymphocytes susceptible to mutations contributing to oncogenic transformation. Consequently, mechanisms responsible for monitoring global genomic integrity must be activated during lymphocyte development to limit the oncogenic potential of antigen receptor locus recombination. Mutations in ATM (ataxia-telangiectasia mutated), a kinase that coordinates DSB monitoring and the response to DNA damage, result in impaired T-cell development and predispose to T-cell leukemia. Here, we review recent evidence providing insight into the mechanisms by which ATM promotes normal lymphocyte development and protects from neoplastic transformation.
Collapse
Affiliation(s)
- Irina R Matei
- Program in Developmental Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | |
Collapse
|
30
|
Jiang G, Sancar A. Recruitment of DNA damage checkpoint proteins to damage in transcribed and nontranscribed sequences. Mol Cell Biol 2006; 26:39-49. [PMID: 16354678 PMCID: PMC1317637 DOI: 10.1128/mcb.26.1.39-49.2006] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We developed a chromatin immunoprecipitation method for analyzing the binding of repair and checkpoint proteins to DNA base lesions in any region of the human genome. Using this method, we investigated the recruitment of DNA damage checkpoint proteins RPA, Rad9, and ATR to base damage induced by UV and acetoxyacetylaminofluorene in transcribed and nontranscribed regions in wild-type and excision repair-deficient human cells in G1 and S phases of the cell cycle. We find that all 3 damage sensors tested assemble at the site or in the vicinity of damage in the absence of DNA replication or repair and that transcription enhances recruitment of checkpoint proteins to the damage site. Furthermore, we find that UV irradiation of human cells defective in excision repair leads to phosphorylation of Chk1 kinase in both G1 and S phase of the cell cycle, suggesting that primary DNA lesions as well as stalled transcription complexes may act as signals to initiate the DNA damage checkpoint response.
Collapse
Affiliation(s)
- Guochun Jiang
- Department of Biochemistry and Biophysics, Mary Ellen Jones Building CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
31
|
Pang D, Rodgers JE, Berman BL, Chasovskikh S, Dritschilo A. Spatial distribution of radiation-induced double-strand breaks in plasmid DNA as resolved by atomic force microscopy. Radiat Res 2006; 164:755-65. [PMID: 16296881 DOI: 10.1667/rr3425.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Atomic force microscopy (AFM) has been used to directly visualize, size and compare the DNA fragments resulting from exposure to low- and high-LET radiation. Double-stranded pUC-19 plasmid ("naked") DNA samples were irradiated by electron-beam or reactor neutron fluxes with doses ranging from 0.9 to 10 kGy. AFM scanning in the tapping mode was used to image and measure the DNA fragment lengths (ranging from a few bp up to 2864 bp long). Double-strand break (DSB) distributions resulting from high-LET neutron and lower-LET electron irradiation revealed a distinct difference between the effects of these two types of radiation: Low-LET radiation-induced DSBs are distributed more uniformly along the DNA, whereas a much larger proportion of neutron-induced DSBs are distributed locally and densely. Furthermore, comparisons with predictions of a random DSB model of radiation damage show that neutron-induced DSBs deviate more from the model than do electron-induced DSBs. In summary, our high-resolution AFM measurements of radiation-induced DNA fragment-length distributions reveal an increased number of very short fragments and hence clustering of DSBs induced by the high-LET neutron radiation compared with low-LET electron radiation and a random DSB model prediction.
Collapse
Affiliation(s)
- Dalong Pang
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | | | |
Collapse
|
32
|
Pathways of DNA Double-Strand Break Repair in Mammalian Cells after Ionizing Radiation. Genome Integr 2006. [DOI: 10.1007/7050_011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
33
|
Murakami M, Narumi I, Satoh K, Furukawa A, Hayata I. Analysis of interaction between DNA and Deinococcus radiodurans PprA protein by atomic force microscopy. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2005; 1764:20-3. [PMID: 16309981 DOI: 10.1016/j.bbapap.2005.10.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Revised: 10/20/2005] [Accepted: 10/20/2005] [Indexed: 11/21/2022]
Abstract
A DNA repair-promoting protein, PprA, was isolated from a radiation resistant bacterium, Deinococcus radiodurans [I. Narumi, K. Sato, S. Cui, T. Funayama, S. Kitayama, H. Watanabe, PprA: a novel protein from Deinococcus radiodurans that stimulates DNA ligation, Mol. Microbiol. 54 (2004) 278-285]. Despite several studies, however, the function of PprA is not still clear. We used atomic force microscopy (AFM) to elucidate the role of this protein in the DNA repair pathway. In the present study, interaction between the linear DNA and PprA protein was imaged and analyzed by AFM without any fixation or staining. Though both end-bound and internally bound PprA was observed, the affinity of the end-bound protein was greater considering the proportion of features of binding analyzed by AFM. In some conditions, looping forms of the DNA-PprA complex were observed. Gel filtration high performance liquid chromatography (HPLC) was also conducted to estimate the molecular weight of this protein. The result of the HPLC analysis suggested that PprA formed multimers in buffer solution without DNA.
Collapse
Affiliation(s)
- Masahiro Murakami
- Radiation Hazards Research Group, Research Center for Radiation Safety, National Institute of Radiological Sciences, 9-1, Anagawa-4-chome, Chiba-shi 263-8555, Japan.
| | | | | | | | | |
Collapse
|
34
|
Itakura E, Sawada I, Matsuura A. Dimerization of the ATRIP protein through the coiled-coil motif and its implication to the maintenance of stalled replication forks. Mol Biol Cell 2005; 16:5551-62. [PMID: 16176973 PMCID: PMC1289401 DOI: 10.1091/mbc.e05-05-0427] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
ATR (ATM and Rad3-related), a PI kinase-related kinase (PIKK), has been implicated in the DNA structure checkpoint in mammalian cells. ATR associates with its partner protein ATRIP to form a functional complex in the nucleus. In this study, we investigated the role of the ATRIP coiled-coil domain in ATR-mediated processes. The coiled-coil domain of human ATRIP contributes to self-dimerization in vivo, which is important for the stable translocation of the ATR-ATRIP complex to nuclear foci that are formed after exposure to genotoxic stress. The expression of dimerization-defective ATRIP diminishes the maintenance of replication forks during treatment with replication inhibitors. By contrast, it does not compromise the G2/M checkpoint after IR-induced DNA damage. These results show that there are two critical functions of ATR-ATRIP after the exposure to genotoxic stress: maintenance of the integrity of replication machinery and execution of cell cycle arrest, which are separable and are achieved via distinct mechanisms. The former function may involve the concentrated localization of ATR to damaged sites for which the ATRIP coiled-coil motif is critical.
Collapse
Affiliation(s)
- Eisuke Itakura
- Department of Geriatric Medicine, National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka-cho, Obu, Aichi 474-8522, Japan
| | | | | |
Collapse
|
35
|
Falck J, Coates J, Jackson SP. Conserved modes of recruitment of ATM, ATR and DNA-PKcs to sites of DNA damage. Nature 2005; 434:605-11. [PMID: 15758953 DOI: 10.1038/nature03442] [Citation(s) in RCA: 938] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Accepted: 02/14/2005] [Indexed: 01/13/2023]
Abstract
Ataxia-telangiectasia mutated (ATM), ataxia-telangiectasia and Rad3-related (ATR) and DNA-dependent protein kinase catalytic subunit (DNA-PKcs) are members of the phosphoinositide-3-kinase-related protein kinase (PIKK) family, and are rapidly activated in response to DNA damage. ATM and DNA-PKcs respond mainly to DNA double-strand breaks, whereas ATR is activated by single-stranded DNA and stalled DNA replication forks. In all cases, activation involves their recruitment to the sites of damage. Here we identify related, conserved carboxy-terminal motifs in human Nbs1, ATRIP and Ku80 proteins that are required for their interaction with ATM, ATR and DNA-PKcs, respectively. These motifs are essential not only for efficient recruitment of ATM, ATR and DNA-PKcs to sites of damage, but are also critical for ATM-, ATR- and DNA-PKcs-mediated signalling events that trigger cell cycle checkpoints and DNA repair. Our findings reveal that recruitment of these PIKKs to DNA lesions occurs by common mechanisms through an evolutionarily conserved motif, and provide direct evidence that PIKK recruitment is required for PIKK-dependent DNA-damage signalling.
Collapse
Affiliation(s)
- Jacob Falck
- The Wellcome Trust and Cancer Research UK Gurdon Institute, and Department of Zoology, Cambridge University, Tennis Court Road, Cambridge CB2 1QN, UK
| | | | | |
Collapse
|
36
|
Kurz EU, Lees-Miller SP. DNA damage-induced activation of ATM and ATM-dependent signaling pathways. DNA Repair (Amst) 2005; 3:889-900. [PMID: 15279774 DOI: 10.1016/j.dnarep.2004.03.029] [Citation(s) in RCA: 342] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Ataxia-telangiectasia mutated (ATM) plays a key role in regulating the cellular response to ionizing radiation. Activation of ATM results in phosphorylation of many downstream targets that modulate numerous damage response pathways, most notably cell cycle checkpoints. In this review, we describe recent developments in our understanding of the mechanism of activation of ATM and its downstream signaling pathways, and explore whether DNA double-strand breaks are the sole activators of ATM and ATM-dependent signaling pathways.
Collapse
Affiliation(s)
- Ebba U Kurz
- Cancer Biology Research Group, Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, Canada
| | | |
Collapse
|
37
|
Ismail IH, Nyström S, Nygren J, Hammarsten O. Activation of Ataxia Telangiectasia Mutated by DNA Strand Break-inducing Agents Correlates Closely with the Number of DNA Double Strand Breaks. J Biol Chem 2005; 280:4649-55. [PMID: 15546858 DOI: 10.1074/jbc.m411588200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is activated when cells are exposed to ionizing radiation (IR). It has been assumed that ATM is specifically activated by the few induced DNA double strand breaks (DSBs), although little direct evidence for this assumption has been presented. DSBs constitute only a few percent of the IR-induced DNA damage, whereas the more frequent single strand DNA breaks (SSBs) and base damage account for over 98% of the overall DNA damage. It is therefore unclear whether DSBs are the only IR-induced DNA lesions that activate ATM. To test directly whether or not DSBs are responsible for ATM activation, we exposed cells to drugs and radiation that produce different numbers of DSBs and SSBs. We determined the resulting ATM activation by measuring the amount of phosphorylated Chk2 and the numbers of SSBs and DSBs in the same cells after short incubation periods. We found a strong correlation between the number of DSBs and ATM activation but no correlation with the number of SSBs. In fact, hydrogen peroxide, which, similar to IR, induces DNA damage through hydroxyl radicals but fails to induce DSBs, did not activate ATM. In contrast, we found that calicheamicin-induced strand breaks activated ATM more efficiently than IR and that ATM activation correlated with the relative DSB induction by these agents. Our data indicate that ATM is specifically activated by IR-induced DSBs, with little or no contribution from SSBs and other types of DNA damage. These findings have implications for how ATM might recognize DSBs in cells.
Collapse
Affiliation(s)
- Ismail Hassan Ismail
- Department of Clinical Chemistry and Transfusion Medicine, Sahlgrenska University Hospital, Göteborg University, SE-413 45 Göteborg, Sweden
| | | | | | | |
Collapse
|
38
|
Abstract
Chk2 is a critical mediator of diverse cellular responses to DNA damage. Activation of Chk2 by DNA damage requires phosphorylation at sites including Thr68. In earlier work, we found that an activity present in rabbit reticulocyte lysates phosphorylates and activates Chk2. We now find that hypophosphorylated Chk2 can be phosphorylated at Thr68 by various subcellular fractions of HEK293 cells. This activity is sensitive to the phosphatidylinositol 3'-kinase-like kinase inhibitor wortmannin, but not to caffeine. DNA enhances the Chk2 phosphorylation by cellular fractions in vitro. The wortmannin-sensitive Chk2 kinase activity is present in fractions from ATM-deficient cells. In contrast, Chk2 was not efficiently phosphorylated at Thr68 in vitro by fractions from cells with a defective DNA-dependent protein kinase (DNA-PK) catalytic subunit. Chk2 is phosphorylated by purified DNA-PK in vitro. Endogenous Chk2 coimmunoprecipitates Ku70 and Ku80. In a series of matched cell lines having and lacking functional DNA-PK, Chk2 activation by exposure of cells to ionizing radiation, or to camptothecin was consistently diminished in the absence of DNA-PK. Down-regulation of DNA-PK(cs) by either siRNA or a chemical inhibitor attenuated radiation-induced Chk2 phosphorylation. Ionizing radiation-induced Chk2 phosphorylation was wortmannin-sensitive in ATM-defective cells with depleted ATR. These results suggest that DNA-PK augments ATM and ATR in activation of Chk2 by DNA damage.
Collapse
Affiliation(s)
- Jia Li
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | |
Collapse
|
39
|
Coutinho G, Xie J, Du L, Brusco A, Krainer AR, Gatti RA. Functional significance of a deep intronic mutation in the
ATM
gene and evidence for an alternative exon 28a. Hum Mutat 2005; 25:118-24. [PMID: 15643608 DOI: 10.1002/humu.20170] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Screening for ATM mutations is usually performed using genomic DNA as a template for PCR amplification across exonic regions, with the consequence that deep intronic sequences are not analyzed. Here we report a novel pseudoexon-retaining deep intronic mutation (IVS28-159A>G; g.75117A>G based on GenBank U82828.1) in a patient with ataxia-telangiectasia (A-T), as well as the identification of a previously unrecognized alternative exon in the ATM gene (exon 28a) expressed in lymphoblastoid cell lines (LCL) derived from normal individuals. cDNA analysis using the A-T patient's LCL showed the retention of two aberrant intronic segments of 112 and 190 nt between exons 28 and 29. Minigenes were constructed to determine the functional significance of two genomic changes in the region of aberrant splicing: IVS28-193C>T (g.75083C>T) and IVS28-159A>G, revealing that: 1) the first is a polymorphism; 2) IVS28-159A>G weakens the 5' splice site of the alternative exon 28a and activates a cryptic 5' splice site (ss) 83 nt downstream; and 3) wild-type constructs also retain a 29-nt segment (exon 28a) as part of both the 112- and 190-nt segments. Maximum entropy estimates of ss strengths corroborate the cDNA and minigene findings. Such mutations may prove relevant in planning therapy that targets specific splicing aberrations.
Collapse
Affiliation(s)
- Gabriela Coutinho
- Department of Pathology and Laboratory Medicine, The David Geffen School of Medicine, University of California, Los Angeles 90095-1732, USA
| | | | | | | | | | | |
Collapse
|
40
|
Goodarzi AA, Lees-Miller SP. Biochemical characterization of the ataxia-telangiectasia mutated (ATM) protein from human cells. DNA Repair (Amst) 2004; 3:753-67. [PMID: 15177184 DOI: 10.1016/j.dnarep.2004.03.041] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2004] [Indexed: 12/21/2022]
Abstract
Ataxia-telangiectasia mutated (ATM) is a serine/threonine protein kinase that plays a central role in controlling the cellular response to ionizing radiation and other DNA-damaging agents. ATM is a 3056 amino acid polypeptide that is present in low abundance in the nucleus of human cells. Here, we describe the purification and characterization of ATM from the nuclear fraction of HeLa cells. Microgram quantities of highly stable, kinase-active ATM were prepared. Purified ATM was phosphorylated on serine 1981 and was active towards a variety of known ATM substrates, including p53 and the Bloom Syndrome helicase, BLM. The protein kinase activity of ATM was selectively inhibited by wortmannin, caffeine and LY294002 and was stimulated by charged biological polymers, including single-stranded M13 DNA (ssDNA), sheared double-stranded calf thymus DNA, heparin sulfate and poly ADP-ribose (PAR), raising the possibility that charged structures may contribute to regulation of ATM activity. However, chemical inhibition of the formation of poly ADP-ribose in cells had no effect on the activation of ATM-dependent pathways by ionizing radiation. Using gel filtration chromatography, we also show that purified ATM, as well as ATM in crude nuclear extracts from unirradiated and irradiated cells elutes with an estimated native molecular weight of approximately 600 kDa. Moreover, dephosphorylation of serine 1981 did not affect the apparent molecular weight of ATM in irradiated extracts. Our results suggest that phosphorylation of serine 1981 alone may not directly regulate the subunit composition of ATM.
Collapse
Affiliation(s)
- Aaron A Goodarzi
- Departments of Biochemistry and Molecular Biology, and Biological Sciences, Cancer Biology Research Group, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB, Canada
| | | |
Collapse
|
41
|
Helt CE, Cliby WA, Keng PC, Bambara RA, O'Reilly MA. Ataxia telangiectasia mutated (ATM) and ATM and Rad3-related protein exhibit selective target specificities in response to different forms of DNA damage. J Biol Chem 2004; 280:1186-92. [PMID: 15533933 DOI: 10.1074/jbc.m410873200] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ataxia telangiectasia mutated (ATM) and ATR (ATM and Rad3-related) protein kinases exert cell cycle delay, in part, by phosphorylating Checkpoint kinase (Chk) 1, Chk2, and p53. It is well established that ATR is activated following UV light-induced DNA damage such as pyrimidine dimers and the 6-(1,2)-dihydro-2-oxo-4-pyrimidinyl-5-methyl-2,4-(1H,3H)-pyrimidinediones, whereas ATM is activated in response to double strand DNA breaks. Here we clarify the activation of these kinases in cells exposed to IR, UV, and hyperoxia, a condition of chronic oxidative stress resulting in clastogenic DNA damage. Phosphorylation on Chk1(Ser-345), Chk2(Thr-68), and p53(Ser-15) following oxidative damage by IR involved both ATM and ATR. In response to ultraviolet radiation-induced stalled replication forks, phosphorylation on Chk1 and p53 required ATR, whereas Chk2 required ATM. Cells exposed to hyperoxia exhibited growth delay in G1, S, and G2 that was disrupted by wortmannin. Consistent with ATM or ATR activation, hyperoxia induced wortmannin-sensitive phosphorylation of Chk1, Chk2, and p53. By using ATM- and ATR-defective cells, phosphorylation on Chk1, Chk2, and p53 was found to be ATM-dependent, whereas ATR also contributed to Chk1 phosphorylation. These data reveal activated ATM and ATR exhibit selective substrate specificity in response to different genotoxic agents.
Collapse
Affiliation(s)
- Christopher E Helt
- Department of Environmental Medicine, School of Medicine and Dentistry, the University of Rochester, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|
42
|
Chun HH, Cary RB, Lansigan F, Whitelegge J, Rawlings DJ, Gatti RA. ATM protein purified from vaccinia virus expression system: DNA binding requirements for kinase activation. Biochem Biophys Res Commun 2004; 322:74-81. [PMID: 15313175 DOI: 10.1016/j.bbrc.2004.07.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2004] [Indexed: 11/13/2022]
Abstract
The ataxia-telangiectasia mutated (ATM) gene product plays a role in responding to double stand DNA breaks. Some biochemical studies of ATM function have been hampered by lack of an efficient expression system and abundant purified ATM protein. We report the construction of a vaccinia virus expressing ATM, vWR-ATM, which was used to produce large amounts of functional FLAG-tagged ATM protein (FLAG-ATM) in HeLa cells. Kinase activity of the purified FLAG-ATM was dependent on manganese and inhibited with wortmannin. Using the FLAG-ATM recombinant protein, GST-p53 serine 15 phosphorylation increased in the presence of damaged DNA. PHAS-1 phosphorylation was found to be DNA independent. Purified FLAG-ATM was recovered in the autophosphorylated form, as demonstrated by phosphorylation of ATM serine 1981. As shown by atomic force microscopy, FLAG-ATM bound to linear DNA both at broken ends and in mid-strands. Vaccinia virus is the most efficient ATM expression system described to date.
Collapse
Affiliation(s)
- Helen H Chun
- Department of Pathology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
43
|
Lavin MF, Scott S, Gueven N, Kozlov S, Peng C, Chen P. Functional consequences of sequence alterations in the ATM gene. DNA Repair (Amst) 2004; 3:1197-205. [PMID: 15279808 DOI: 10.1016/j.dnarep.2004.03.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The product of the gene (ATM) mutated in the human genetic disorder ataxia-telangiectasia (A-T) is a high molecular weight, protein ( approximately 350kDa) containing a C-terminal protein kinase domain and a number of other putative domains not yet functionally defined. The majority of ATM gene mutations in A-T patients are truncating, resulting in prematurely terminated products that are highly unstable. Missense mutations within the kinase domain and elsewhere in the molecule alter the stability of the protein and lead to loss of protein kinase activity. Only rarely are patients observed with two missense mutations and this gives rise to a milder disease phenotype. Evidence for a dominant interfering effect on normal ATM kinase activity has been reported in cell lines transfected with missense mutant ATM and in cell lines from some A-T heterozygotes. The dominant negative effect of mutant ATM is manifested by an enhancement of cellular radiosensitivity and may be responsible for the cancer predisposition observed in carriers of ATM missense mutations. In this review, we explore the domain structure of the ATM molecule, sites of interaction with other proteins and the consequences of specific amino acid changes on function.
Collapse
Affiliation(s)
- Martin F Lavin
- The Queensland Cancer Fund Research Unit, The Queensland Institute of Medical Research, P.O. Box Royal Brisbane Hospital, Herston, Brisbane 4029, Qld, Australia.
| | | | | | | | | | | |
Collapse
|
44
|
Sancar A, Lindsey-Boltz LA, Unsal-Kaçmaz K, Linn S. Molecular Mechanisms of Mammalian DNA Repair and the DNA Damage Checkpoints. Annu Rev Biochem 2004; 73:39-85. [PMID: 15189136 DOI: 10.1146/annurev.biochem.73.011303.073723] [Citation(s) in RCA: 2348] [Impact Index Per Article: 117.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
DNA damage is a relatively common event in the life of a cell and may lead to mutation, cancer, and cellular or organismic death. Damage to DNA induces several cellular responses that enable the cell either to eliminate or cope with the damage or to activate a programmed cell death process, presumably to eliminate cells with potentially catastrophic mutations. These DNA damage response reactions include: (a) removal of DNA damage and restoration of the continuity of the DNA duplex; (b) activation of a DNA damage checkpoint, which arrests cell cycle progression so as to allow for repair and prevention of the transmission of damaged or incompletely replicated chromosomes; (c) transcriptional response, which causes changes in the transcription profile that may be beneficial to the cell; and (d) apoptosis, which eliminates heavily damaged or seriously deregulated cells. DNA repair mechanisms include direct repair, base excision repair, nucleotide excision repair, double-strand break repair, and cross-link repair. The DNA damage checkpoints employ damage sensor proteins, such as ATM, ATR, the Rad17-RFC complex, and the 9-1-1 complex, to detect DNA damage and to initiate signal transduction cascades that employ Chk1 and Chk2 Ser/Thr kinases and Cdc25 phosphatases. The signal transducers activate p53 and inactivate cyclin-dependent kinases to inhibit cell cycle progression from G1 to S (the G1/S checkpoint), DNA replication (the intra-S checkpoint), or G2 to mitosis (the G2/M checkpoint). In this review the molecular mechanisms of DNA repair and the DNA damage checkpoints in mammalian cells are analyzed.
Collapse
Affiliation(s)
- Aziz Sancar
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599-7260, USA.
| | | | | | | |
Collapse
|
45
|
Yu G, Zhu MH, Zhu Z, Ni CR, Zheng JM, Li FM. Expression of ATM protein and its relationship with p53 in pancreatic carcinoma with tissue array. Pancreas 2004; 28:421-6. [PMID: 15097860 DOI: 10.1097/00006676-200405000-00011] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ATM protein anticipates in the initiation of the DNA repair signal pathway and also mediates cell cycle arrest and repair. ATM deficiency predictably results in radiosensitivity, germ cell degeneration, chromosomal instability, immunodeficiency, and an extreme predisposition to tumors. Moreover, studies found that ATM is the upstream gene of the p53 pathway and would phosphorylate p53 directly after DNA damage, which would suppress tumorigenesis. Expression of ATM and p53 in 167 pancreatic cancer and 101 control specimens, benign lesions, and normal pancreata were detected by high-throughput tissue microarray and immunohistochemistry while seeking the role of ATM in the initiation and development of pancreatic carcinoma as well as its relationship with p53. We found that the positive rates of ATM and p53 expression in pancreatic carcinoma and its relative control specimen were 67.7% (113/167) and 82.2% (83/101) (P < 0.05) and 57.5% (96/167) and 5.0% (5/101) (P < 0.01), respectively. ATM positive staining is significantly relative to age and infiltration (P < 0.05), while the expression of p53 was significantly associated with tumor differentiation, lymph node metastasis, and nerve infiltration (P < 0.05). Expression of ATM and p53 was positively correlated. These findings suggest that expression of ATM deficiency may increase the transformative ability of pancreatic cancer cells. ATM may also cooperate with p53 in the repair of cell damage.
Collapse
Affiliation(s)
- Guanzhen Yu
- Department of Pathology, Chang-hai Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
46
|
Debiak M, Nikolova T, Kaina B. Loss of ATM sensitizes against O6-methylguanine triggered apoptosis, SCEs and chromosomal aberrations. DNA Repair (Amst) 2004; 3:359-68. [PMID: 15010311 DOI: 10.1016/j.dnarep.2003.11.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2003] [Indexed: 01/09/2023]
Abstract
A critical pre-cytotoxic and -apoptotic DNA lesion induced by methylating carcinogens and chemotherapeutic drugs is O6-methylguanine (O6MeG). The mechanism by which O6MeG causes cell death via apoptosis is only partially understood. The current model ascribes a role to DNA replication and mismatch repair, which converts O6MeG into a critical distal lesion (presumably a DNA double-strand break) that is finally responsible for genotoxicity and apoptosis. Here we analysed whether the PI3-like kinase ATM is involved in this process. ATM is a major player in recognizing and signaling DNA breaks, but most reports are limited to ionizing radiation. Comparing mouse ATM knockout fibroblasts (ATM-/-) with the corresponding wild-type (ATM+/+) we show that ATM-/- cells are hypersensitive to the cytotoxic and apoptosis-inducing effect of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Inhibition of O6-methylguanine-DNA methyltransferase (MGMT) activity by O6-benzylguanine enhanced cell killing whereas the increase of MGMT activity by transfection with an expression vector provoked MNNG resistance. This was more pronounced in ATM-/- than in ATM+/+ cells, suggesting that O6MeG is responsible, at least in part, for increased MNNG sensitivity of ATM-/- cells. Cytogenetic studies showed that MNNG-induced sister-chromatid exchange frequencies were the same in ATM-/- and ATM+/+ cells in the first mitoses following treatment, but higher in ATM-/- cells than in the wild-type in the second post-treatment mitoses, when MGMT was depleted. Also, a significant higher frequency of MNNG-induced chromosomal aberrations was observed in ATM-/- than in ATM+/+ cells when analysed at a late recovery time, which is consistent with O6MeG being the inducing lesion. In summary, we conclude that ATM is not only involved in resistance to ionizing radiation but also to methylating agents, playing a role in the repair of secondary DNA damage generated from O6MeG lesions. The data also show that ATM is not required for activating the apoptotic pathway in response to O6MeG since ATM-/- cells are able to undergo apoptosis with high frequency.
Collapse
Affiliation(s)
- Malgorzata Debiak
- Division of Applied Toxicology, Institute of Toxicology, University of Mainz, Obere Zahlbacher Street 67, D-55131 Mainz, Germany
| | | | | |
Collapse
|
47
|
Unsal-Kaçmaz K, Sancar A. Quaternary structure of ATR and effects of ATRIP and replication protein A on its DNA binding and kinase activities. Mol Cell Biol 2004; 24:1292-300. [PMID: 14729973 PMCID: PMC321456 DOI: 10.1128/mcb.24.3.1292-1300.2003] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
ATR is an essential protein that functions as a damage sensor and a proximal kinase in the DNA damage checkpoint response in mammalian cells. It is a member of the phosphoinositide 3-kinase-like kinase (PIKK) family, which includes ATM, ATR, and DNA-dependent protein kinase. Recently, it was found that ATM is an oligomeric protein that is converted to an active monomeric form by phosphorylation in trans upon DNA damage, and this raised the possibility that other members of the PIKK family may be regulated in a similar manner. Here we show that ATR is a monomeric protein associated with a smaller protein called ATRIP with moderate affinity. The ATR protein by itself or in the form of the ATR-ATRIP heterodimer binds to naked or replication protein A (RPA)-covered DNAs with comparable affinities. However, the phosphorylation of RPA by ATR is dependent on single-stranded DNA and is stimulated by ATRIP. These findings suggest that the regulation and mechanism of action of ATR are fundamentally different from those of the other PIKK proteins.
Collapse
Affiliation(s)
- Keziban Unsal-Kaçmaz
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | | |
Collapse
|
48
|
Golding SE, Rosenberg E, Khalil A, McEwen A, Holmes M, Neill S, Povirk LF, Valerie K. Double strand break repair by homologous recombination is regulated by cell cycle-independent signaling via ATM in human glioma cells. J Biol Chem 2004; 279:15402-10. [PMID: 14744854 DOI: 10.1074/jbc.m314191200] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To investigate double strand break (DSB) repair and signaling in human glioma cells, we stably transfected human U87 (ATM(+), p53(+)) glioma cells with a plasmid having a single I-SceI site within an inactive green fluorescent protein (GFP) expression cassette, allowing for the detection of homologous recombination repair (HRR) by GFP expression. HRR and nonhomologous end joining (NHEJ) were also determined by PCR. DSB repair was first detected at 12 h postinfection with an adenovirus expressing I-SceI with repair reaching plateau levels between 24 and 48 h. Within this time frame, NHEJ predominated over HRR in the range of 3-50-fold. To assess the involvement of ATM in DSB repair, we first examined whether ATM was associated with the DSB. Chromatin immunoprecipitation showed that ATM was present at the site of the DSB as early as 18 h postinfection. In cells treated with caffeine, an inhibitor of ATM, HRR was reduced, whereas NHEJ was not. In support of this finding, GFP flow cytometry demonstrated that caffeine reduced HRR by 90% under conditions when ATM kinase activity was inhibited. Dominant-negative ATM expressed from adenovirus inhibited HRR by 45%, also having little to no effect on NHEJ. Furthermore, HRR was inhibited by caffeine in serum-starved cells arrested in G(0)/G(1), suggesting that ATM is also important for HRR outside of the S and G(2) cell cycle phases. Altogether, these results demonstrate that HRR contributes substantially to DSB repair in human glioma cells, and, importantly, ATM plays a critical role in regulating HRR but not NHEJ throughout the cell cycle.
Collapse
Affiliation(s)
- Sarah E Golding
- Department of Radiation Oncology, Pharmacology & Toxicology, Medical College of Virginia, Virginia 23298, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ward IM, Minn K, Chen J. UV-induced ataxia-telangiectasia-mutated and Rad3-related (ATR) activation requires replication stress. J Biol Chem 2004; 279:9677-80. [PMID: 14742437 DOI: 10.1074/jbc.c300554200] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Ataxia-telangiectasia-mutated and Rad3-related (ATR) plays an essential role in the maintenance of genome integrity and cell viability. The kinase is activated in response to DNA damage and initiates a checkpoint signaling cascade by phosphorylating a number of downstream substrates including Chk1. Unlike ataxia-telangiectasia-mutated (ATM), which appears to be mainly activated by DNA double-strand breaks, ATR can be activated by a variety of DNA damaging agents. However, it is still unclear what triggers ATR activation in response to such diverse DNA lesions. One model proposes that ATR can directly recognize DNA lesions, while other recent data suggest that ATR is activated by a common single-stranded DNA (ssDNA) intermediate generated during DNA repair. In this study, we show that UV lesions do not directly activate ATR in vivo. In addition, ssDNA lesions created during the repair of UV damage are also not sufficient to activate the ATR-dependent pathway. ATR activation is only observed in replicating cells indicating that replication stress is required to trigger the ATR-mediated checkpoint cascade in response to UV irradiation. Interestingly, H2AX appears to be required for the accumulation of ATR at stalled replication forks. Together our data suggest that ssDNA at arrested replication forks recruits ATR and initiates ATR-mediated phosphorylation of H2AX and Chk1. Phosphorylated H2AX might further facilitate ATR activation by stabilizing ATR at the sites of arrested replication forks.
Collapse
Affiliation(s)
- Irene M Ward
- Guggenheim 1306, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
50
|
Abstract
Cellular response to genotoxic stress is a very complex process, and it usually starts with the “sensing” or “detection” of the DNA damage, followed by a series of events that include signal transduction and activation of transcription factors. The activated transcription factors induce expressions of many genes which are involved in cellular functions such as DNA repair, cell cycle arrest, and cell death. There have been extensive studies from multiple disciplines exploring the mechanisms of cellular genotoxic responses, which have resulted in the identification of many cellular components involved in this process, including the mitogen-activated protein kinases (MAPKs) cascade. Although the initial activation of protein kinase cascade is not fully understood, human protein kinases ATM (ataxia-telangiectasia, mutated) and ATR (ATM and Rad3-related) are emerging as potential sensors of DNA damage. Current progresses in ATM/ATR research and related signaling pathways are discussed in this review, in an effort to facilitate a better understanding of genotoxic stress response.
Collapse
Affiliation(s)
- Jun Yang
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, 353 Yanan Road, Hangzhou, 310031, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|