Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-wxhwt Total loading time: 0 Render date: 2024-07-11T11:27:04.688Z Has data issue: false hasContentIssue false

34 - The ATM-mediated DNA-damage response

from Part 2.4 - Molecular pathways underlying carcinogenesis: DNA repair

Published online by Cambridge University Press:  05 February 2015

Yosef Shiloh
Affiliation:
The David and Inez Myers Laboratory for Genetic Research, Department of Human Molecular Genetics and Biochemistry, Sackler School ofMedicine, Tel Aviv University, Tel Aviv, Israel
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Maintenance of genomic stability and cancer

Cancer is a genetic disease of the somatic cells. Its initiation and development are caused by accumulating genomic alterations, ranging from point mutations to gross chromosomal aberrations. Maintenance of genomic stability and integrity is thus essential for prevention of neoplasia (1,2). DNA damage is arguably the greatest threat to genome stability. DNA-damaging agents induce a plethora of DNA lesions that can be cytotoxic and/or mutagenic, with consequences ranging from malfunction of the cell, to cell death or malignant transformation (3,4). Many DNA-damaging agents are therefore potent carcinogens (5–7).

The cellular defense system against this threat is the DNA damage response (DDR) – an elaborate signaling network activated by DNA damage that swiftly modulates many physiological processes (4,8–12). It is not surprising that various players in the DDR are tumor suppressors; germline mutations in damage response genes lead to inherited predisposition to cancer (13–20) or to complex genomic instability syndromes characterized by a predisposition to develop cancer (21–25). Functional dissection of the DDR is therefore expected to identify additional players in cancer formation. The DDR is also highly relevant to cancer treatment, as radiotherapy and many chemotherapeutic drugs are DNA-damaging agents. Understanding the DDR is thus crucial to design of better treatment regimens, minimization of side effects, identification of new targets for drug therapy, discovery of new methods for radiosensitization and chemosensitization of tumor cells, and resolution of the major problem of radio- and drug-resistance.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 403 - 422
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Hahn, WC, Weinberg, RA. Modelling the molecular circuitry of cancer. Nature Reviews Cancer 2002;2:331–41.CrossRef
Murga, M, Fernandez-Capetillo, O. Genomic instability: on the birth and death of cancer. Clinical and Translational Oncology 2007;9(4):216–20.CrossRef
Norbury, CJ, Hickson, ID. Cellular responses to DNA damage. Annual Review of Pharmacology and Toxicology 2001;41:367–401.CrossRef
Hoeijmakers, JH. Genome maintenance mechanisms for preventing cancer. Nature 2001;411:366–74.CrossRef
Poirier, MC. Chemical-induced DNA damage and human cancer risk. Nature Reviews Cancer 2004;4:630–7.CrossRef
Kawanishi, S, Hiraku, Y. Oxidative and nitrative DNA damage as biomarker for carcinogenesis with special reference to inflammation. Antioxidants and Redox Signaling 2006;8:1047–58.CrossRef
Hwang, ES, Bowen, PE. DNA damage, a biomarker of carcinogenesis: its measurement and modulation by diet and environment. Critical Reviews in Food Science and Nutrition 2007;47:27–50.CrossRef
Bakkenist, CJ, Kastan, MB. Initiating cellular stress responses. Cell 2004;118:9–17.CrossRef
Callegari, AJ, Kelly, TJ. Shedding light on the DNA damage checkpoint. Cell Cycle 2007;6:660–6.CrossRef
Shiloh, Y. ATM and related protein kinases: safeguarding genome integrity. Nature Reviews Cancer 2003;3:155–68.CrossRef
Harrison, JC, Haber, JE. Surviving the breakup: the DNA damage checkpoint. Annual Review of Genetics 2006;40:209–35.CrossRef
Su, TT. Cellular responses to DNA damage: one signal, multiple choices. Annual Review of Genetics 2006;40:187–208.CrossRef
Narod, SA, Foulkes, WD. BRCA1 and BRCA2: 1994 and beyond. Nature Reviews Cancer 2004;4:665–76.CrossRef
Varley, JM. Germline TP53 mutations and Li-Fraumeni syndrome. Human Mutation 2003;21:313–20.CrossRef
Arai, M, Utsunomiya, J, Miki, Y. Familial breast and ovarian cancers. International Journal of Clinical Oncology 2004;9:270–82.CrossRefGoogle ScholarPubMed
Staalesen, V, Falck, J, Geisler, S, et al. Alternative splicing and mutation status of CHEK2 in stage III breast cancer. Oncogene 2004;23:8535–44.CrossRef
Varley, J, Haber, DA. Familial breast cancer and the hCHK2 1100delC mutation: assessing cancer risk. Breast Cancer Research 2003;5:123–5.CrossRef
Tort, F, Hernandez, S, Bea, S, et al. Checkpoint kinase 1 (CHK1) protein and mRNA expression is downregulated in aggressive variants of human lymphoid neoplasms. Leukemia 2004;19:112–17.CrossRef
Celeste, A, Difilippantonio, S, Difilippantonio, MJ, et al. H2AX haploinsufficiency modifies genomic stability and tumor susceptibility. Cell 2003;114:371–83.CrossRef
Bassing, CH, Suh, H, Ferguson, DO, et al. Histone H2AX: a dosage-dependent suppressor of oncogenic translocations and tumors. Cell 2003;114:359–70.CrossRef
Moses, RE. DNA damage processing defects and disease. Annual Review of Genomics and Human Genetics 2001;2:41–68.CrossRef
Vessey, CJ, Norbury, CJ, Hickson, ID. Genetic disorders associated with cancer predisposition and genomic instability. Progress in Nucleic Acid Research and Molecular Biology 1999;63:189–221.CrossRef
O’Driscoll, M, Gennery, AR, Seidel, J, Concannon, P, Jeggo, PA. An overview of three new disorders associated with genetic instability: LIG4 syndrome, RS-SCID and ATR-Seckel syndrome. DNA Repair (Amsterdam) 2004;3:1227–35.CrossRef
O’Driscoll, M, Jeggo, PA. The role of double-strand break repair – insights from human genetics. Nature Reviews Genetics 2006;7(1):45–54.CrossRef
Eyfjord, JE, Bodvarsdottir, SK. Genomic instability and cancer: networks involved in response to DNA damage. Mutation Research 2005;592(1–2):18–28.
Bassing, CH, Alt, FW. The cellular response to general and programmed DNA double strand breaks. DNA Repair (Amsterdam) 2004;3:781–96.CrossRef
Harrison, JC, Haber, JE. Surviving the breakup: the DNA damage checkpoint. Annual Review of Genetics 2006;40:209–35.CrossRef
Lieber, MR, Ma, Y, Pannicke, U, Schwarz, K. The mechanism of vertebrate nonhomologous DNA end joining and its role in V(D)J recombination. DNA Repair (Amsterdam) 2004;3:817–26.CrossRef
van Gent, DC, van der Burg, M. Non-homologous end-joining, a sticky affair. Oncogene 2007;26:7731–40.CrossRef
Shrivastav, M, De Haro, LP, Nickoloff, JA. Regulation of DNA double-strand break repair pathway choice. Cell Research 2008;18:134–47.CrossRef
Wyman, C, Kanaar, R. DNA double-strand break repair: all's well that ends well. Annual Review of Genetics 2006;40:363–83.CrossRef
Reliene, R, Bishop, AJ, Schiestl RH. Involvement of homologous recombination in carcinogenesis. Advances in Genetics 2007;58:67–87.
Helleday, T, Lo, J, van Gent, DC, Engelward, BP. DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair (Amsterdam) 2007;6:923–35.CrossRef
Wyman, C, Kanaar, R. Homologous recombination: down to the wire. Current Biology 2004;14:R629–31.
Shiloh, Y. The ATM-mediated DNA-damage response: taking shape. Trends in Biochemical Sciences 2006;31:402–10.CrossRef
Jeggo, PA, Lobrich, M. Contribution of DNA repair and cell cycle checkpoint arrest to the maintenance of genomic stability. DNA Repair (Amsterdam) 2006;5:1192–8.CrossRef
Muro, I, Hay, BA, Clem, RJ. The Drosophila DIAP1 protein is required to prevent accumulation of a continuously generated, processed form of the apical caspase DRONC. Journal of Biological Chemistry 2002;277:49 644–50.CrossRefGoogle ScholarPubMed
Begley, TJ, Samson, LD. Network responses to DNA damaging agents. DNA Repair (Amsterdam) 2004;3:1123–32.CrossRef
Rashi-Elkeles, S, Elkon, R, Weizman, N, et al. Parallel induction of ATM-dependent pro- and antiapoptotic signals in response to ionizing radiation in murine lymphoid tissue. Oncogene 2006;25:1584–92.CrossRef
Elkon, R, Rashi-Elkeles, S, Lerenthal, Y, et al. Dissection of a DNA-damage-induced transcriptional network using a combination of microarrays, RNA interference and computational promoter analysis. Genome Biology 2005;6:R43.
Kolas, NK, Chapman, JR, Nakada, S, et al. Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science 2007;318:1637–40.CrossRef
Sakasai, R, Tibbetts, RS. RNF8-dependent and independent regulation of 53BP1 in response to DNA damage. Journal of Biological Chemistry 2008;283:13 549–55.CrossRefGoogle ScholarPubMed
Huen, MS, Grant, R, Manke, I, et al. RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly. Cell 2007;131:901–14.CrossRef
Mailand, N, Bekker-Jensen, S, Faustrup, H, et al. RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins. Cell 2007;131:887–900.CrossRef
Wang, B, Elledge, SJ. Ubc13/Rnf8 ubiquitin ligases control foci formation of the Rap80/Abraxas/Brca1/Brcc36 complex in response to DNA damage. Proceedings of the National Academy of Sciences USA 2007;104:20 759–63.
Khosravi, R, Maya, R, Gottlieb, T, et al. Rapid ATM-dependent phosphorylation of MDM2 precedes p53 accumulation in response to DNA damage. Proceedings of the National Academy of Sciences USA 1999;96:14 973–7.
Pereg, Y, Lam, S, Teunisse, A, et al. Differential roles of ATM- and Chk2-mediated phosphorylations of Hdmx in response to DNA damage. Molecular and Cellular Biology 2006;26:6819–31.CrossRef
Pereg, Y, Shkedy, D, de Graaf, P, et al. Phosphorylation of Hdmx mediates its Hdm2- and ATM-dependent degradation in response to DNA damage. Proceedings of the National Academy of Sciences USA 2005;102:5056–61.CrossRef
Braithwaite, AW, Royds, JA, Jackson, P. The p53 story: layers of complexity. Carcinogenesis 2005;26:1161–9.CrossRef
Kaufmann, WK, Heffernan, TP, Beaulieu, LM, et al. Caffeine and human DNA metabolism: the magic and the mystery. Mutation Research 2003;532:85–102.CrossRef
Zamzami, N, Kroemer, G. p53 in apoptosis control: an introduction. Biochemical and Biophysical Research Communications 2005;331:685–7.CrossRef
Helton, ES, Chen, X. p53 modulation of the DNA damage response. Journal of Cell Biochemistry 2006;100:883–96.CrossRefGoogle Scholar
Cusack, JC. Overcoming antiapoptotic responses to promote chemosensitivity in metastatic colorectal cancer to the liver. Annals of Surgical Oncology 2003;10:852–62.CrossRef
Cataldi, A, Rapino, M, Centurione, L, et al. NF-kappaB activation plays an antiapoptotic role in human leukemic K562 cells exposed to ionizing radiation. Journal of Cell Biochemistry 2003;89:956–63.CrossRefGoogle ScholarPubMed
Weaver, KD, Yeyeodu, S, Cusack, JC, Baldwin, AS, Ewend, MG. Potentiation of chemotherapeutic agents following antagonism of nuclear factor kappa B in human gliomas. Journal of Neurooncology 2003;61:187–96.CrossRefGoogle ScholarPubMed
Potapova, O, Gorospe, M, Bost, F, et al. c-Jun N-terminal kinase is essential for growth of human T98G glioblastoma cells. Journal of Biological Chemistry 2000;275:24 767–75.CrossRefGoogle ScholarPubMed
Nehme, A, Baskaran, R, Aebi, S, et al. Differential induction of c-Jun NH2-terminal kinase and c-Abl kinase in DNA mismatch repair-proficient and -deficient cells exposed to cisplatin. Cancer Research 1997;57:3253–7.
van Dam, H, Castellazzi, M. Distinct roles of Jun: Fos and Jun: ATF dimers in oncogenesis. Oncogene 2001;20:2453–64.CrossRef
Hayakawa, J, Depatie, C, Ohmichi, M, Mercola, D. The activation of c-Jun NH2-terminal kinase (JNK) by DNA-damaging agents serves to promote drug resistance via activating transcription factor 2 (ATF2)-dependent enhanced DNA repair. Journal of Biological Chemistry 2003;278:20 582–92.CrossRefGoogle ScholarPubMed
Krones-Herzig, A, Mittal, S, Yule, K, et al. Early growth response 1 acts as a tumor suppressor in vivo and in vitro via regulation of p53. Cancer Research 2005;65(12):5133–43.CrossRef
Bhoumik, A, Takahashi, S, Breitweiser, W, et al. ATM-dependent phosphorylation of ATF2 is required for the DNA damage response. Molecular Cell 2005;18:577–87.CrossRef
Yan, C, Boyd, DD. ATF3 regulates the stability of p53: a link to cancer. Cell Cycle 2006;5:926–9.CrossRef
Lin, WC, Lin, FT, Nevins, JR. Selective induction of E2F1 in response to DNA damage, mediated by ATM-dependent phosphorylation. Genes and Development 2001;15:1833–44.
Prochownik, EV. c-Myc as a therapeutic target in cancer. Expert Review of Anticancer Therapy 2004;4:289–302.CrossRef
Kim, JE, Minter-Dykhouse, K, Chen, J. Signaling networks controlled by the MRN complex and MDC1 during early DNA damage responses. Molecular Carcinogenesis 2006;45:403–8.CrossRef
Zhang, J, Powell, SN. The role of the BRCA1 tumor suppressor in DNA double-strand break repair. Molecular Cancer Research 2005;3:531–9.CrossRef
Wagner, J, Ma, L, Rice, JJ, et al. p53-Mdm2 loop controlled by a balance of its feedback strength and effective dampening using ATM and delayed feedback. Systems Biology (Stevenage) 2005;152:109–18.CrossRef
Zgheib, O, Huyen, Y, DiTullio, RA, et al. ATM signaling and 53BP1. Radiotherapy and Oncology 2005;76:119–22.CrossRef
Stucki, M, Jackson, SP. MDC1/NFBD1: a key regulator of the DNA damage response in higher eukaryotes. DNA Repair (Amsterdam) 2004;3:953–7.CrossRef
Stucki, M, Jackson, SP. gamma-H2AX and MDC1: anchoring the DNA-damage-response machinery to broken chromosomes. DNA Repair (Amsterdam) 2006;5:534–43.CrossRef
Lukas, J, Bartek, J. Watching the DNA repair ensemble dance. Cell 2004;118:666–8.CrossRef
Bekker-Jensen, S, Lukas, C, Kitagawa, R, et al. Spatial organization of the mammalian genome surveillance machinery in response to DNA strand breaks. Journal of Cell Biology 2006;173:195–206.CrossRefGoogle ScholarPubMed
Bekker-Jensen, S, Lukas, C, Melander, F, Bartek, J, Lukas, J. Dynamic assembly and sustained retention of 53BP1 at the sites of DNA damage are controlled by Mdc1/NFBD1. Journal of Cell Biology 2005;170:201–11.CrossRefGoogle ScholarPubMed
Lukas, C, Bartek, J, Lukas, J. Imaging of protein movement induced by chromosomal breakage: tiny “local” lesions pose great “global” challenges. Chromosoma 2005;114:146–54.CrossRef
Lukas, C, Falck, J, Bartkova, J, Bartek, J, Lukas, J. Distinct spatiotemporal dynamics of mammalian checkpoint regulators induced by DNA damage. Nature Cell Biology 2003;5:255–60.CrossRef
Lee, AC, Fernandez-Capetillo, O, Pisupati, V, Jackson, SP, Nussenzweig, A. Specific association of mouse MDC1/NFBD1 with NBS1 at sites of DNA-damage. Cell Cycle 2005;4:177–82.CrossRef
Stracker, TH, Theunissen, JW, Morales, M, Petrini, JH. The Mre11 complex and the metabolism of chromosome breaks: the importance of communicating and holding things together. DNA Repair (Amsterdam) 2004;3:845–54.CrossRef
Moreno-Herrero, F, de Jager, M, Dekker, NH, et al. Mesoscale conformational changes in the DNA-repair complex Rad50/Mre11/Nbs1 upon binding DNA. Nature 2005;437:440–3.CrossRef
Williams, RS, Williams, JS, Tainer, JA. Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochemistry and Cell Biology 2007;85:509–20.CrossRef
Mochan, TA, Venere, M, DiTullio, RA, Halazonetis, TD. 53BP1, an activator of ATM in response to DNA damage. DNA Repair (Amsterdam) 2004;3:945–52.CrossRef
Shiloh, Y, Lehmann, AR. Maintaining integrity. Nature Cell Biology 2004;6:923–8.CrossRef
Shechter, D, Costanzo, V, Gautier, J. Regulation of DNA replication by ATR: signaling in response to DNA intermediates. DNA Repair (Amsterdam) 2004;3:901–8.CrossRef
Hurley, PJ, Bunz, F. ATM and ATR: components of an integrated circuit. Cell Cycle 2007;6(4):414–17.CrossRef
Kurz, EU, Lees-Miller, SP. DNA damage-induced activation of ATM and ATM-dependent signaling pathways. DNA Repair (Amsterdam) 2004;3:889–900.CrossRef
Savitsky, K, Bar-Shira, A, Gilad, S, et al. A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 1995;268:1749–53.CrossRef
Savitsky, K, Sfez, S, Tagle, DA, et al. The complete sequence of the coding region of the ATM gene reveals similarity to cell cycle regulators in different species. Human Molecular Genetics 1995;4:2025–32.CrossRef
Banin, S, Moyal, L, Shieh, S, et al. Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 1998;281:1674–7.CrossRef
Canman, CE, Lim, DS, Cimprich, KA, et al. Activation of the ATM kinase by ionizing radiation and phosphorylation of p53. Science 1998;281:1677–9.CrossRef
Kozlov, SV, Graham, ME, Peng, C, et al. Involvement of novel autophosphoryla-tion sites in ATM activation. EMBO Journal 2006;25:3504–14.CrossRef
Sun, Y, Xu, Y, Roy, K, Price, BD. DNA damage induced acetylation of lysine 3016 of ATM activates ATM kinase activity. Molecular and Cellular Biology 2007;27:8502–9.CrossRef
Matsuoka, S, Ballif, BA, Smogorzewska, A, et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 2007;316:1160–6.CrossRef
Mu, JJ, Wang, Y, Luo, H, et al. A proteomic analysis of ATM/ATR substrates identifies the ubiquitin-proteasome system as a regulator for DNA damage checkpoints. Journal of Biological Chemistry 2007;282:17 330–4.CrossRefGoogle ScholarPubMed
Hurley, PJ, Bunz, F. ATM and ATR: components of an integrated circuit. Cell Cycle 2007;6:414–17.CrossRef
Crawford, TO. Ataxia telangiectasia. Seminars in Pediatric Neurology 1998;5:287–94.CrossRef
Abraham, RT. PI 3-kinase related kinases: ‘big’ players in stress-induced signaling pathways. DNA Repair (Amsterdam) 2004;3:883–7.CrossRef
Burma, S, Chen, DJ. Role of DNA-PK in the cellular response to DNA double-strand breaks. DNA Repair (Amsterdam) 2004;3:909–18.CrossRef
Abraham, RT. The ATM-related kinase, hSMG-1, bridges genome and RNA surveillance pathways. DNA Repair (Amsterdam) 2004;3:919–25.CrossRef
Paulsen, RD, Cimprich, KA. The ATR pathway: fine-tuning the fork. DNA Repair (Amsterdam) 2007;6:953–66.CrossRef
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. Journal of Biological Chemistry 2005;280:1186–92.CrossRefGoogle ScholarPubMed
Jazayeri, A, Falck, J, Lukas, C, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nature Cell Biology 2006;8:37–45.CrossRef
Cortez, D, Guntuku, S, Qin, J, Elledge, SJ. ATR and ATRIP: partners in checkpoint signaling. Science 2001;294:1713–16.CrossRef
Zou, L, Elledge, SJ. Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes. Science 2003;300:1542–8.CrossRef
Itakura, E, Takai, KK, Umeda, K, et al. Amino-terminal domain of ATRIP contributes to intranuclear relocation of the ATR-ATRIP complex following DNA damage. FEBS Letters 2004;577:289–93.CrossRef
Unsal-Kacmaz, K, Sancar, A. Quaternary structure of ATR and effects of ATRIP and replication protein A on its DNA binding and kinase activities. Molecular and Cellular Biology 2004;24:1292–300.CrossRef
Ball, HL, Myers, JS, Cortez, D. ATRIP binding to replication protein A-single-stranded DNA promotes ATR-ATRIP localization but is dispensable for Chk1 phosphorylation. Molecular Biology of the Cell 2005;16:2372–81.CrossRef
Namiki, Y, Zou, L. ATRIP associates with replication protein A-coated ssDNA through multiple interactions. Proceedings of the National Academy of Sciences USA 2006;103:580–5.CrossRef
Uziel, T, Lerenthal, Y, Moyal, L, et al. Requirement of the MRN complex for ATM activation by DNA damage. EMBO Journal 2003;22:5612–21.CrossRef
Carson, CT, Schwartz, RA, Stracker, TH, et al. The Mre11 complex is required for ATM activation and the G2/M checkpoint. EMBO Journal 2003;22:6610–20.CrossRef
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.CrossRef
You, Z, Chahwan, C, Bailis, J, Hunter, T, Russell, P. ATM activation and its recruitment to damaged DNA require binding to the C terminus of Nbs1. Molecular and Cellular Biology 2005;25:5363–79.CrossRef
Costanzo, V, Paull, T, Gottesman, M, Gautier, J. Mre11 assembles linear DNA fragments into DNA damage signaling complexes. PLoS Biology 2004;2:E110.
Lee, JH, Paull, TT. Direct activation of the ATM protein kinase by the Mre11/Rad50/Nbs1 complex. Science 2004;304:93–6.CrossRef
Lee, JH, Paull, TT. ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex. Science 2005;308:551–4.CrossRef
Lee, JH, Paull, TT. Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene 2007;26:7741–8.CrossRef
Block, WD, Merkle, D, Meek, K, Lees-Miller, SP. Selective inhibition of the DNA-dependent protein kinase (DNA-PK) by the radiosensitizing agent caffeine. Nucleic Acids Research 2004;32:1967–72.CrossRef
Lavin, MF. ATM and the Mre11 complex combine to recognize and signal DNA double-strand breaks. Oncogene 2007;26:7749–58.CrossRef
Zhou, J, Lim, CU, Li, JJ, Cai, L, Zhang, Y. The role of NBS1 in the modulation of PIKK family proteins ATM and ATR in the cellular response to DNA damage. Cancer Letters 2006;243:9–15.CrossRef
Adams, KE, Medhurst, AL, Dart, DA, Lakin, ND. Recruitment of ATR to sites of ionising radiation-induced DNA damage requires ATM and components of the MRN protein complex. Oncogene 2006;25:3894–904.CrossRef
Stiff, T, Reis, C, Alderton, GK, et al. Nbs1 is required for ATR-dependent phosphorylation events. EMBO Journal 2005;24:199–208.CrossRef
Myers, JS, Cortez, D. Rapid activation of ATR by ionizing radiation requires ATM and Mre11. Journal of Biological Chemistry 2006;281:9346–50.CrossRefGoogle ScholarPubMed
Nakada, D, Hirano, Y, Sugimoto, K. Requirement of the Mre11 complex and exonuclease 1 for activation of the Mec1 signaling pathway. Molecular and Cellular Biology 2004;24:10 016–25.
Zhong, H, Bryson, A, Eckersdorff, M, Ferguson, DO. Rad50 depletion impacts upon ATR-dependent DNA damage responses. Human Molecular Genetics 2005;14:2685–93.CrossRef
Olson, E, Nievera, CJ, Lee, AY, Chen, L, Wu, X. The Mre11-Rad50-Nbs1 complex acts both upstream and downstream of ataxia telangiectasia mutated and Rad3-related protein (ATR) to regulate the S-phase checkpoint following UV treatment. Journal of Biological Chemistry 2007;282:22 939–52.CrossRefGoogle ScholarPubMed
Lee, AY, Liu, E, Wu, X. The Mre11/Rad50/Nbs1 complex plays an important role in the prevention of DNA rereplication in mammalian cells. Journal of Biological Chemistry 2007;282:32 243–55.CrossRefGoogle ScholarPubMed
Manthey, KC, Opiyo, S, Glanzer, JG, et al. NBS1 mediates ATR-dependent RPA hyperphosphorylation following replication-fork stall and collapse. Journal of Cell Science 2007;120:4221–9.CrossRefGoogle Scholar
Bakkenist, CJ, Kastan, MB. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 2003;421:499–506.CrossRef
Sun, Y, Jiang, X, Chen, S, Fernandes, N, Price, BD. A role for the Tip60 histone acetyltransferase in the acetylation and activation of ATM. Proceedings of the National Academy of Sciences USA 2005;102:13 182–7.
Jiang, X, Sun, Y, Chen, S, Roy, K, Price, BD. The FATC domains of PIKK proteins are functionally equivalent and participate in the Tip60-dependent activation of DNA-PKcs and ATM. Journal of Biological Chemistry 2006;281:15 741–6.CrossRefGoogle ScholarPubMed
Goodarzi, AA, Jonnalagadda, JC, Douglas, P, et al. Autophosphorylation of ataxia-telangiectasia mutated is regulated by protein phosphatase 2A. EMBO Journal 2004;23:4451–61.CrossRef
Ali, A, Zhang, J, Bao, S, et al. Requirement of protein phosphatase 5 in DNA-damage-induced ATM activation. Genes and Development 2004;18:249–54.CrossRef
Andegeko, Y, Moyal, L, Mittelman, L, et al. Nuclear retention of ATM at sites of DNA double strand breaks. Journal of Biological Chemistry 2001;276:38 224–30.Google ScholarPubMed
Lou, Z, Minter-Dykhouse, K, Franco, S, et al. MDC1 maintains genomic stability by participating in the amplification of ATM-dependent DNA damage signals. Molecular Cell 2006;21:187–200.CrossRef
Difilippantonio, S, Nussenzweig, A. The NBS1-ATM connection revisited. Cell Cycle 2007;6:2366–70.CrossRef
Berkovich, E, Monnat, RJ, Kastan, MB. Roles of ATM and NBS1 in chromatin structure modulation and DNA double-strand break repair. Nature Cell Biology 2007;9:683–90.CrossRef
Fernandez-Capetillo, O, Lee, A, Nussenzweig, M, Nussenzweig, A. H2AX: the histone guardian of the genome. DNA Repair (Amsterdam) 2004;3:959–67.CrossRef
Stucki, M, Clapperton, JA, Mohammad, D, et al. MDC1 directly binds phosphorylated histone H2AX to regulate cellular responses to DNA double-strand breaks. Cell 2005;123:1213–26.CrossRef
Huyen, Y, Zgheib, O, Ditullio, RA, et al. Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks. Nature 2004;432:406–11.CrossRef
Lee, MS, Edwards, RA, Thede, GL, Glover, JN. Structure of the BRCT repeat domain of MDC1 and its specificity for the free COOH-terminal end of the gamma-H2AX histone tail. Journal of Biological Chemistry 2005;280:32 053–6.CrossRefGoogle ScholarPubMed
Meek, DW. The p53 response to DNA damage. DNA Repair (Amsterdam) 2004;3:1049–56.CrossRef
Stommel, JM, Wahl, GM. Accelerated MDM2 auto-degradation induced by DNA-damage kinases is required for p53 activation. EMBO Journal 2004;23:1547–56.CrossRef
Dornan, D, Shimizu, H, Mah, A, et al. ATM engages autodegradation of the E3 ubiquitin ligase COP1 after DNA damage. Science 2006;313:1122–6.CrossRef
Chen, L, Gilkes, DM, Pan, Y, Lane, WS, Chen, J. ATM and Chk2-dependent phosphorylation of MDMX contribute to p53 activation after DNA damage. EMBO Journal 2005;24:3411–22.CrossRef
Lukas, J, Lukas, C, Bartek, J. Mammalian cell cycle checkpoints: signalling pathways and their organization in space and time. DNA Repair (Amsterdam) 2004;3:997–1007.CrossRef
Bartek, J, Lukas, J. DNA damage checkpoints: from initiation to recovery or adaptation. Current Opinion in Cell Biology 2007;19:238–45.CrossRef
Burgoyne, PS, Mahadevaiah, SK, Turner, JM. The management of DNA double-strand breaks in mitotic G2, and in mammalian meiosis viewed from a mitotic G2 perspective. Bioessays 2007;29:974–86.CrossRef
Lobrich, M, Jeggo, PA. The impact of a negligent G2/M checkpoint on genomic instability and cancer induction. Nature Reviews Cancer 2007;7:861–9.CrossRef
Ahn, J, Urist, M, Prives, C. The Chk2 protein kinase. DNA Repair (Amsterdam) 2004;3:1039–47.CrossRef
Antoni, L, Sodha, N, Collins, I, Garrett, MD. CHK2 kinase: cancer susceptibility and cancer therapy – two sides of the same coin? Nature Reviews Cancer 2007;7:925–36.
Chen, Y, Sanchez, Y. Chk1 in the DNA damage response: conserved roles from yeasts to mammals. DNA Repair (Amsterdam) 2004;3:1025–32.CrossRef
Mailand, N, Falck, J, Lukas, C, et al. Rapid destruction of human Cdc25A in response to DNA damage. Science 2000;288:1425–9.CrossRef
Karlsson-Rosenthal, C, Millar, JB. Cdc25: mechanisms of checkpoint inhibition and recovery. Trends in Cell Biology 2006;16:285–92.CrossRef
Falck, J, Mailand, N, Syljuasen, RG, Bartek, J, Lukas, J. The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 2001;410:842–7.CrossRef
Costanzo, V, Robertson, K, Ying, CY, et al. Reconstitution of an ATM-dependent checkpoint that inhibits chromosomal DNA replication following DNA damage. Molecular Cell 2000;6:649–59.CrossRef
Yazdi, PT, Wang, Y, Zhao, S, et al. SMC1 is a downstream effector in the ATM/NBS1 branch of the human S-phase checkpoint. Genes and Development 2002;16:571–82.CrossRef
Kim, ST, Xu, B, Kastan, MB. Involvement of the cohesin protein, Smc1, in Atm-dependent and independent responses to DNA damage. Genes and Development 2002;16:560–70.CrossRef
Lim, DS, Kim, ST, Xu, B, et al. ATM phosphorylates p95/nbs1 in an S-phase checkpoint pathway. Nature 2000;404:613–17.CrossRef
Zhao, S, Weng, YC, Yuan, SS, et al. Functional link between ataxia-telangiectasia and Nijmegen breakage syndrome gene products. Nature 2000;405:473–7.CrossRef
Xu, B, Kim, S, Kastan, MB. Involvement of Brca1 in S-phase and G(2)-phase checkpoints after ionizing irradiation. Molecular and Cellular Biology 2001;21:3445–50.CrossRef
Falck, J, Petrini, JH, Williams, BR, Lukas, J, Bartek, J. The DNA damage-dependent intra-S phase checkpoint is regulated by parallel pathways. Nature Genetics 2002;30:290–4.CrossRef
Xu, B, O’Donnell, AH, Kim, ST, Kastan, MB. Phosphorylation of serine 1387 in Brca1 is specifically required for the Atm-mediated S-phase checkpoint after ionizing irradiation. Cancer Research 2002;62:4588–91.
Mailand, N, Podtelejnikov, AV, Groth, A, et al. Regulation of G(2)/M events by Cdc25A through phosphorylation-dependent modulation of its stability. EMBO Journal 2002;21:5911–20.CrossRef
Xu, B, Kim, ST, Lim, DS, Kastan, MB. Two molecularly distinct G(2)/M checkpoints are induced by ionizing irradiation. Molecular and Cellular Biology 2002;22:1049–59.CrossRef
Stankovic, T, Hubank, M, Cronin, D, et al. Microarray analysis reveals that TP53- and ATM-mutant B-CLLs share a defect in activating proapoptotic responses after DNA damage but are distinguished by major differences in activating prosurvival responses. Blood 2004;103:291–300.CrossRef
Wu, ZH, Shi, Y, Tibbetts, RS, Miyamoto, S. Molecular linkage between the kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. Science 2006;311:1141–6.CrossRef
Shi, Y, Venkataraman, SL, Dodson, GE, et al. Direct regulation of CREB transcriptional activity by ATM in response to genotoxic stress. Proceedings of the National Academy of Sciences USA 2004;101:5898–903.CrossRef
Dodson, GE, Tibbetts, RS. DNA replication stress-induced phosphorylation of cyclic AMP response element-binding protein mediated by ATM. Journal of Biological Chemistry 2006;281:1692–7.CrossRefGoogle ScholarPubMed
Kamer, I, Sarig, R, Zaltsman, Y, et al. Proapoptotic BID is an ATM effector in the DNA-damage response. Cell 2005;122:593–603.CrossRef
Zinkel, SS, Hurov, KE, Ong, C, et al. A role for proapoptotic BID in the DNA-damage response. Cell 2005;122:579–91.CrossRef
Ziv, Y, Bielopolski, D, Galanty, Y, et al. Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM- and KAP-1 dependent pathway. Nature Cell Biology 2006;8:870–6.CrossRef
Tsai, WB, Chung, YM, Takahashi, Y, Xu, Z, Hu, MC. Functional interaction between FOXO3a and ATM regulates DNA damage response. Nature Cell Biology 2008;10:460–7.CrossRef
Woodcock, CL. Chromatin architecture. Current Opinion in Structural Biology 2006;16:213–20.CrossRef
Polo, SE, Almouzni, G. Chromatin assembly: a basic recipe with various flavours. Current Opinion in Genetic Development 2006;16:104–11.CrossRef
Horn, PJ, Peterson, CL. Heterochromatin assembly: a new twist on an old model. Chromosome Research 2006;14:83–94.CrossRef
Hediger, F, Gasser, SM. Heterochromatin protein 1: don't judge the book by its cover! Current Opinion in Genetic Development 2006;16:143–50.
McBryant, SJ, Adams, VH, Hansen, JC. Chromatin architectural proteins. Chromosome Research 2006;14:39–51.CrossRef
Mellor, J. Dynamic nucleosomes and gene transcription. Trends in Genetics 2006;22:320–9.CrossRef
Saha, A, Wittmeyer, J, Cairns, BR. Chromatin remodelling: the industrial revolution of DNA around histones. Nature Reviews Molecular and Cellular Biology 2006;7:437–47.CrossRef
Varga-Weisz, PD, Becker, PB. Regulation of higher-order chromatin structures by nucleosome-remodelling factors. Current Opinion in Genetic Development 2006;16:151–6.CrossRef
Peterson, CL, Laniel, MA. Histones and histone modifications. Current Biology 2004;14:R546–51.
Volkel, P, Angrand, PO. The control of histone lysine methylation in epigenetic regulation. Biochimie 2007;89:1–20.CrossRef
Nightingale, KP, O’Neill, LP, Turner, BM. Histone modifications: signalling receptors and potential elements of a heritable epigenetic code. Current Opinion in Genetic Development 2006;16:125–36.CrossRef
Osley, MA, Fleming, AB, Kao, CF. Histone ubiquitylation and the regulation of transcription. Results and Problems in Cell Differentiation 2006;41:47–75.CrossRef
Fischle, W, Tseng, BS, Dormann, HL, et al. Regulation of HP1-chromatin binding by histone H3 methylation and phosphorylation. Nature 2005;438:1116–22.CrossRef
Weake, VM, Workman, JL. Histone ubiquitination: triggering gene activity. Molecular Cell 2008;29:653–63.CrossRef
Gontijo, AM, Green, CM, Almouzni, G. Repairing DNA damage in chromatin. Biochimie 2003;85:1133–47.CrossRef
Verger, A, Crossley, M. Chromatin modifiers in transcription and DNA repair. Cellular and Molecular Life Sciences 2004;61:2154–62.
Karagiannis, TC, El-Osta, A. Chromatin modifications and DNA double-strand breaks: the current state of play. Leukemia 2007;21:195–200.CrossRef
Downs, JA, Nussenzweig, MC, Nussenzweig, A. Chromatin dynamics and the preservation of genetic information. Nature 2007;447:951–8.CrossRef
Tsukuda, T, Fleming, AB, Nickoloff, JA, Osley, MA. Chromatin remodelling at a DNA double-strand break site in Saccharomyces cerevisiae. Nature 2005;438:379–83.CrossRef
Chai, B, Huang, J, Cairns, BR, Laurent, BC. Distinct roles for the RSC and Swi/Snf ATP-dependent chromatin remodelers in DNA double-strand break repair. Genes and Development 2005;19:1656–61.CrossRef
Morrison, AJ, Highland, J, Krogan, NJ, et al. INO80 and gamma-H2AX interaction links ATP-dependent chromatin remodeling to DNA damage repair. Cell 2004;119:767–75.CrossRef
van Attikum, H, Fritsch, O, Hohn, B, Gasser, SM. Recruitment of the INO80 complex by H2A phosphorylation links ATP-dependent chromatin remodeling with DNA double-strand break repair. Cell 2004;119:777–88.CrossRef
van Attikum, H, Gasser, SM. The histone code at DNA breaks: a guide to repair? Nature Reviews Molecular and Cellular Biology 2005;6:757–65.
Sanders, SL, Portoso, M, Mata, J, et al. Methylation of histone H4 lysine 20 controls recruitment of Crb2 to sites of DNA damage. Cell 2004;119:603–14.CrossRef
Kusch, T, Florens, L, Macdonald, WH, et al. Acetylation by Tip60 is required for selective histone variant exchange at DNA lesions. Science 2004;306:2084–7.CrossRef
Fernandez-Capetillo, O, Allis, CD, Nussenzweig, A. Phosphorylation of histone H2B at DNA double-strand breaks. Journal of Experimental Medicine 2004;199:1671–7.CrossRefGoogle ScholarPubMed
Tamburini, BA, Tyler, JK. Localized histone acetylation and deacetylation triggered by the homologous recombination pathway of double-strand DNA repair. Molecular and Cellular Biology 2005;25:4903–13.CrossRef
Foster, ER, Downs, JA. Histone H2A phosphorylation in DNA double-strand break repair. FEBS Journal 2005;272:3231–40.CrossRef
Kruhlak, MJ, Celeste, A, Dellaire, G, et al. Changes in chromatin structure and mobility in living cells at sites of DNA double-strand breaks. Journal of Cell Biology 2006;172:823–34.CrossRefGoogle ScholarPubMed
Bailey, SM, Cornforth, MN. Telomeres and DNA double-strand breaks: ever the twain shall meet? Cellular and Molecular Life Sciences 2007;64:2956–64.
Longhese, MP. DNA damage response at functional and dysfunctional telomeres. Genes and Development 2008;22:125–40.CrossRef
Grandin, N, Charbonneau, M. Protection against chromosome degradation at the telomeres. Biochimie 2008;90:41–59.CrossRef
Verdun, RE, Karlseder, J. The DNA damage machinery and homologous recombination pathway act consecutively to protect human telomeres. Cell 2006;127:709–20.CrossRef
Verdun, RE, Crabbe, L, Haggblom, C, Karlseder, J. Functional human telomeres are recognized as DNA damage in G2 of the cell cycle. Molecular Cell 2005;20:551–61.CrossRef
Denchi, EL, de Lange, T. Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1. Nature 2007;448:1068–71.CrossRef
Dimitrova, N, de Lange, T. MDC1 accelerates nonhomologous end-joining of dysfunctional telomeres. Genes and Development 2006;20:3238–43.CrossRef
Hockemeyer, D, Sfeir, AJ, Shay, JW, Wright, WE, de Lange, T. POT1 protects telomeres from a transient DNA damage response and determines how human chromosomes end. EMBO Journal 2005;24:2667–78.CrossRef
Celli, GB, de Lange, T. DNA processing is not required for ATM-mediated telomere damage response after TRF2 deletion. Nature Cell Biology 2005;7:712–18.CrossRef
Karlseder, J, Hoke, K, Mirzoeva, OK, et al. The telomeric protein TRF2 binds the ATM kinase and can inhibit the ATM-dependent DNA damage response. PLoS Biology 2004;2:E240.
Takai, H, Smogorzewska, A, de Lange, T. DNA damage foci at dysfunctional telomeres. Current Biology 2003;13:1549–56.CrossRef
Cheung, AL, Deng, W. Telomere dysfunction, genome instability and cancer. Frontiers in Bioscience 2008;13:2075–90.CrossRef
Wu, Y, Xiao, S, Zhu, XD. MRE11-RAD50-NBS1 and ATM function as co-mediators of TRF1 in telomere length control. Nature Structural and Molecular Biology 2007;14:832–40.CrossRef
Taylor, AM, Byrd, PJ. Molecular pathology of ataxia telangiectasia. Journal of Clinical Pathology 2005;58:1009–15.CrossRefGoogle ScholarPubMed
Hector, RE, Shtofman, RL, Ray, A, et al. Tel1p preferentially associates with short telomeres to stimulate their elongation. Molecular Cell 2007;27:851–8.CrossRef
Viscardi, V, Clerici, M, Cartagena-Lirola, H, Longhese, MP. Telomeres and DNA damage checkpoints. Biochimie 2005;87:613–24.CrossRef
Viscardi, V, Bonetti, D, Cartagena-Lirola, H, Lucchini, G, Longhese, MP. MRX-dependent DNA damage response to short telomeres. Molecular Biology of the Cell 2007;18:3047–58.CrossRef
Guo, X, Deng, Y, Lin, Y, et al. Dysfunctional telomeres activate an ATM-ATR-dependent DNA damage response to suppress tumorigenesis. EMBO Journal 2007;26:4709–19.CrossRef
de Lange, T. Shelterin: the protein complex that shapes and safeguards human telomeres. Genes and Development 2005;19:2100–10.CrossRef
De la Torre, C, Pincheira, J, Lopez-Saez, JF. Human syndromes with genomic instability and multiprotein machines that repair DNA double-strand breaks. Histology and Histopathology 2003;18:225–43.
Bartkova, J, Bakkenist, CJ, Rajpert-De Meyts, E, et al. ATM activation in normal human tissues and testicular cancer. Cell Cycle 2005;4:838–45.CrossRef
Bartkova, J, Horejsi, Z, Koed, K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 2005;434:864–70.CrossRef
Bartkova, J, Rezaei, N, Liontos, M, et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006;444:633–7.CrossRef
Bartek, J, Bartkova, J, Lukas, J. DNA damage signalling guards against activated oncogenes and tumour progression. Oncogene 2007;26:7773–9.CrossRef
Halazonetis, TD, Gorgoulis, VG, Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 2008;319:1352–5.CrossRef
Crawford, T. Ataxia telangiectaia. Pediatric Neurology 1998;5:287–94.CrossRef
Crawford, TO, Mandir, AS, Lefton-Greif, MA, et al. Quantitative neurologic assessment of ataxia-telangiectasia. Neurology 2000;54:1505–9.CrossRef
Crawford, TO. Neurology problems. In Handbook for Families and Caregivers, A-T Children's Project 2000, Chapter 4.
Lavin, M, Shiloh, Y. Ataxia-Telangiectasia. In: Ochs H, Smith CIE, Puck J, editors. Primary Immunodeficiency Diseases: A Molecular and Genetic Approach, 2nd edn, Volume 1. Oxford: Oxford University Press;2002:306–23.
Chun, HH, Gatti, RA. Ataxia-telangiectasia, an evolving phenotype. DNA Repair (Amsterdam) 2004;3:1187–96.CrossRef
Crawford, T, Shiloh, Y. Ataxia-telangiectasia. In: Brice A, Pulst S-M, editors. Spinocerebellar Degenerations: The Ataxias and Spastic Paraplegias, Volume 1. Amsterdam: Elsevier; 2006:724–39.
Frappart, PO, McKinnon, PJ. Ataxia-telangiectasia and related diseases. Neuromolecular Medicine 2006;8:495–511.CrossRef
Lavin, MF, Gueven, N, Bottle, S, Gatti, RA. Current and potential therapeutic strategies for the treatment of ataxia-telangiectasia. British Medical Bulletin 2007;81–82:129–47.
Lavin, MF, Kozlov, S. ATM activation and DNA damage response. Cell Cycle 2007;6:931–42.CrossRef
Kojis, TL, Gatti, RA, Sparkes, RS. The cytogenetics of ataxia telangiectasia. Cancer Genetics and Cytogenetics 1991;56:143–56.CrossRef
Taylor, AM. Chromosome instability syndromes. Best Practice and Research, Clinical Haematology 2001;14:631–44.CrossRef
Taylor, AM, Metcalfe, JA, Thick, J, Mak, YF. Leukemia and lymphoma in ataxia telangiectasia. Blood 1996;87:423–38.
Hecht, F, Hecht, BK. Ataxia-telangiectasia breakpoints in chromosome rearrangements reflect genes important to T and B lymphocytes. Kroc Foundation Series 1985;19:189–95.
Hecht, F, Hecht, BK. Cancer in ataxia-telangiectasia patients. Cancer Genetics and Cytogenetics 1990;46:9–19.CrossRef
Bredemeyer, AL, Sharma, GG, Huang, CY, et al. ATM stabilizes DNA double-strand-break complexes during V(D)J recombination. Nature 2006;442:466–70.CrossRef
Huang, CY, Sharma, GG, Walker, LM, et al. Defects in coding joint formation in vivo in developing ATM-deficient B and T lymphocytes. Journal of Experimental Medicine 2007;204:1371–81.CrossRefGoogle ScholarPubMed
Callen, E, Jankovic, M, Difilippantonio, S, et al. ATM prevents the persistence and propagation of chromosome breaks in lymphocytes. Cell 2007;130:63–75.CrossRef
Callen, E, Nussenzweig, MC, Nussenzweig, A. Breaking down cell cycle checkpoints and DNA repair during antigen receptor gene assembly. Oncogene 2007;26:7759–64.CrossRef
Morgan, JL, Holcomb, TM, Morrissey, RW. Radiation reaction in ataxia telangiectasia. American Journal of Diseases of Children 1968;116:557–8.Google ScholarPubMed
Becker-Catania, SG, Chen, G, Hwang, MJ, et al. Ataxia-telangiectasia: phenotype/genotype studies of ATM protein expression, mutations, and radiosensitivity. Molecular Genetics and Metabolism 2000;70:122–33.CrossRef
Gilad, S, Khosravi, R, Shkedy, D, et al. Predominance of null mutations in ataxia-telangiectasia. Human Molecular Genetics 1996;5:433–9.CrossRef
Alterman, N, Fattal-Valevski, A, Moyal, L, et al. Ataxia-telangiectasia: mild neurological presentation despite null ATM mutation and severe cellular phenotype. American Journal of Medical Genetics, Part A 2007;143:1827–34.CrossRefGoogle Scholar
Taylor, AM, Flude, E, Laher, B, et al. Variant forms of ataxia telangiectasia. Journal of Medical Genetics 1987;24:669–77.CrossRefGoogle ScholarPubMed
McConville, CM, Stankovic, T, Byrd, PJ, et al. Mutations associated with variant phenotypes in ataxia-telangiectasia. American Journal of Human Genetics 1996;59:320–30.Google ScholarPubMed
Chun, HH, Sun, X, Nahas, SA, et al. Improved diagnostic testing for ataxia-telangiectasia by immunoblotting of nuclear lysates for ATM protein expression. Molecular Genetics and Metabolism 2003;80:437–43.CrossRef
Dork, T, Bendix-Waltes, R, Wegner, RD, Stumm, M. Slow progression of ataxia-telangiectasia with double missense and in frame splice mutations. American Journal of Medical Genetics, Part A 2004;126:272–7.CrossRefGoogle Scholar
Gilad, S, Khosravi, R, Harnik, R, et al. Identification of ATM mutations using extended RT-PCR and restriction endonuclease fingerprinting, and elucidation of the repertoire of A-T mutations in Israel. Human Mutation 1998;11:69–75.3.0.CO;2-X>CrossRef
Saviozzi, S, Saluto, A, Taylor, AM, et al. A late onset variant of ataxia-telangiectasia with a compound heterozygous genotype, A8030G/7481insA. Journal of Medical Genetics 2002;39:57–61.CrossRefGoogle ScholarPubMed
Stankovic, T, Kidd, AM, Sutcliffe, A, et al. ATM mutations and phenotypes in ataxia-telangiectasia families in the British Isles: expression of mutant ATM and the risk of leukemia, lymphoma, and breast cancer. American Journal of Human Genetics 1998;62:334–45.CrossRefGoogle ScholarPubMed
Stewart, GS, Last, JI, Stankovic, T, et al. Residual ataxia telangiectasia mutated protein function in cells from ataxia telangiectasia patients, with 5762ins137 and 7271T–>G mutations, showing a less severe phenotype. Journal of Biological Chemistry 2001;276:30 133–41.CrossRefGoogle ScholarPubMed
Sutton, IJ, Last, JI, Ritchie, SJ, et al. Adult-onset ataxia telangiectasia due to ATM 5762ins137 mutation homozygosity. Annals of Neurology 2004;55:891–5.CrossRef
Gilad, S, Chessa, L, Khosravi, R, et al. Genotype-phenotype relationships in ataxia-telangiectasia and variants. American Journal of Human Genetics 1998;62:551–61.CrossRefGoogle ScholarPubMed
Taylor, AM, Groom, A, Byrd, PJ. Ataxia-telangiectasia-like disorder (ATLD)-its clinical presentation and molecular basis. DNA Repair (Amsterdam) 2004;3:1219–25.CrossRef
Stewart, GS, Maser, RS, Stankovic, T, et al. The DNA double-strand break repair gene hMRE11 is mutated in individuals with an ataxia-telangiectasia-like disorder. Cell 1999;99:577–87.CrossRef
Delia, D, Piane, M, Buscemi, G, et al. MRE11 mutations and impaired ATM-dependent responses in an Italian family with Ataxia-Telangiectasia Like Disorder (ATLD). Human Molecular Genetics 2004;13:2155–63.CrossRef
Fernet, M, Gribaa, M, Salih, MA, et al. Identification and functional consequences of a novel MRE11 mutation affecting 10 Saudi Arabian patients with the ataxia telangiectasia-like disorder. Human Molecular Genetics 2005;14:307–18.CrossRef
Dupre, A, Boyer-Chatenet, L, Sattler, RM, et al. A forward chemical genetic screen reveals an inhibitor of the Mre11-Rad50-Nbs1 complex. Nature Chemical Biology 2008;4:119–25.CrossRef
Petrini, JH, Theunissen, JW. Double strand break metabolism and cancer susceptibility: lessons from the mre11 complex. Cell Cycle 2004;3:541–2.
Paull, TT, Lee, JH. The Mre11/Rad50/Nbs1 complex and its role as a DNA double-strand break sensor for ATM. Cell Cycle 2005;4:737–40.CrossRef
Difilippantonio, S, Celeste, A, Fernandez-Capetillo, O, et al. Role of Nbs1 in the activation of the Atm kinase revealed in humanized mouse models. Nature Cell Biology 2005;7:675–85.CrossRef
Difilippantonio, S, Nussenzweig, A. The NBS1-ATM connection revisited. Cell Cycle 2007;6:2366–70.CrossRef
Tauchi, H, Matsuura, S, Kobayashi, J, Sakamoto, S, Komatsu, K. Nijmegen breakage syndrome gene, NBS1, and molecular links to factors for genome stability. Oncogene 2002;21:8967–80.CrossRef
Matsuura, S, Tauchi, H, Nakamura, A, et al. Positional cloning of the gene for Nijmegen breakage syndrome. Nature Genetics 1998;19:179–81.CrossRef
Varon, R, Vissinga, C, Platzer, M, et al. Nibrin, a novel DNA double-strand break repair protein, is mutated in Nijmegen breakage syndrome. Cell 1998;93:467–76.CrossRef
Carney, JP, Maser, RS, Olivares, H, et al. The hMre11/hRad50 protein complex and Nijmegen breakage syndrome: linkage of double-strand break repair to the cellular DNA damage response. Cell 1998;93:477–86.CrossRef
Cornforth, MN, Bedford, JS. On the nature of a defect in cells from individuals with ataxia-telangiectasia. Science 1985;227:1589–91.CrossRef
Lavin, MF, Shiloh, Y. The genetic defect in ataxia-telangiectasia. Annual Review of Immunology 1997;15:177–202.CrossRef
Riballo, E, Kuhne, M, Rief, N, et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci. Molecular Cell 2004;16:715–24.CrossRef
Pluth, JM, Yamazaki, V, Cooper, BA, et al. DNA double-strand break and chromosomal rejoining defects with misrejoining in Nijmegen breakage syndrome cells. DNA Repair (Amsterdam) 2007;7:108–18.CrossRef
Barlow, C, Hirotsune, S, Paylor, R, et al. Atm-deficient mice: a paradigm of ataxia telangiectasia. Cell 1996;86:159–71.CrossRef
Xu, Y, Ashley, T, Brainerd, EE, et al. Targeted disruption of ATM leads to growth retardation, chromosomal fragmentation during meiosis, immune defects, and thymic lymphoma. Genes and Development 1996;10:2411–22.CrossRef
Elson, A, Wang, Y, Daugherty, CJ, et al. Pleiotropic defects in ataxia-telangiectasia protein-deficient mice. Proceedings of the National Academy of Sciences USA 1996;93:13 084–9.
Borghesani, PR, Alt, FW, Bottaro, A, et al. Abnormal development of Purkinje cells and lymphocytes in Atm mutant mice. Proceedings of the National Academy of Sciences USA 2000;97:3336–41.CrossRef
Frappart, PO, Tong, WM, Demuth, I, et al. An essential function for NBS1 in the prevention of ataxia and cerebellar defects. Nature Medicine 2005;11:538–44.CrossRef
Yang, YG, Frappart, PO, Frappart, L, Wang, ZQ, Tong, WM. A novel function of DNA repair molecule Nbs1 in terminal differentiation of the lens fibre cells and cataractogenesis. DNA Repair (Amsterdam) 2006;5:885–93.CrossRef
Paterson, MC, Anderson, AK, Smith, BP, Smith, PJ. Enhanced radiosensitivity of cultured fibroblasts from ataxia telangiectasia heterozygotes manifested by defective colony-forming ability and reduced DNA repair replication after hypoxic gamma-irradiation. Cancer Research 1979;39:3725–34.
Arlett, CF, Harcourt, SA. Survey of radiosensitivity in a variety of human cell strains. Cancer Research 1980;40:926–32.
Shiloh, Y, Tabor, E, Becker, Y. Cellular hypersensitivity to neocarzinostatin in ataxia-telangiectasia skin fibroblasts. Cancer Research 1982;42:2247–9.
Parshad, R, Sanford, KK, Jones, GM, Tarone, RE. G2 chromosomal radiosensitivity of ataxia-telangiectasia heterozygotes. Cancer Genetics and Cytogenetics 1985;14:163–8.CrossRef
Shiloh, Y, Parshad, R, Sanford, KK, Jones, GM. Carrier detection in ataxia-telangiectasia. Lancet 1986;1:689–90.CrossRef
Shiloh, Y, Parshad, R, Frydman, M, et al. G2 chromosomal radiosensitivity in families with ataxia-telangiectasia. Human Genetics 1989;84:15–18.CrossRef
Sanford, KK, Parshad, R, Price, FM, et al. Enhanced chromatid damage in blood lymphocytes after G2 phase x irradiation, a marker of the ataxia-telangiectasia gene. Journal of the National Cancer Institute 1990;82:1050–4.CrossRefGoogle ScholarPubMed
Paterson, MC, MacFarlane, SJ, Gentner, NE, Smith, BP. Cellular hypersensitivity to chronic gamma-radiation in cultured fibroblasts from ataxia-telangiectasia heterozygotes. Kroc Foundation Series 1985;19:73–87.
Arlett, CF, Priestley, A. An assessment of the radiosensitivity of ataxia-telangiectasia heterozygotes. Kroc Foundation Series 1985;19:101–9.
Rosin, MP, Ochs, HD. In vivo chromosomal instability in ataxia-telangiectasia homozygotes and heterozygotes. Human Genetics 1986;74:335–40.CrossRef
Weissberg, JB, Huang, DD, Swift, M. Radiosensitivity of normal tissues in ataxia-telangiectasia heterozygotes. International Journal of Radiation Oncology, Biology, Physics 1998;42:1133–6.CrossRefGoogle ScholarPubMed
Broeks, A, Braaf, LM, Huseinovic, A, et al. Identification of women with an increased risk of developing radiation-induced breast cancer: a case only study. Breast Cancer Research 2007;9:R26.
Ho, AY, Fan, G, Atencio, DP, et al. Possession of ATM sequence variants as predictor for late normal tissue responses in breast cancer patients treated with radiotherapy. International Journal of Radiation Oncology, Biology, Physics 2007;69:677–84.CrossRefGoogle ScholarPubMed
Cesaretti, JA, Stock, RG, Lehrer, S, et al. ATM sequence variants are predictive of adverse radiotherapy response among patients treated for prostate cancer. International Journal of Radiation Oncology, Biology, Physics 2005;61:196–202.CrossRefGoogle ScholarPubMed
Andreassen, CN, Overgaard, J, Alsner, J, et al. ATM sequence variants and risk of radiation-induced subcutaneous fibrosis after postmastectomy radiotherapy. International Journal of Radiation Oncology, Biology, Physics 2006;64:776–83.CrossRefGoogle ScholarPubMed
Swift, M, Sholman, L, Perry, M, Chase, C. Malignant neoplasms in the families of patients with ataxia-telangiectasia. Cancer Research 1976;36:209–15.
Swift, M, Chase, C. Cancer and cardiac deaths in obligatory ataxia-telangiectasia heterozygotes. Lancet 1983;1:1049–50.CrossRef
Swift, M, Reitnauer, PJ, Morrell, D, Chase, CL. Breast and other cancers in families with ataxia-telangiectasia. New England Journal of Medicine 1987;316:1289–94.CrossRefGoogle ScholarPubMed
Morrell, D, Chase, CL, Swift, M. Cancers in 44 families with ataxia-telangiectasia. Cancer Genetics and Cytogenetics 1990;50:119–23.CrossRef
Swift, M, Chase, CL, Morrell, D. Cancer predisposition of ataxia-telangiectasia heterozygotes. Cancer Genetics and Cytogenetics 1990;46:21–7.CrossRef
Swift, M, Morrell, D, Massey, RB, Chase, CL. Incidence of cancer in 161 families affected by ataxia-telangiectasia. New England Journal of Medicine 1991;325:1831–6.CrossRefGoogle ScholarPubMed
Swift, M. Ataxia telangiectasia and risk of breast cancer. Lancet 1997;350:740.CrossRef
Swift, M. Diabetes-predisposing genes. Lancet 1973;2:497.CrossRef
Swift, M, Su, Y. Link between breast cancer and ATM gene is strong. British Medical Journal 1999;318:400.CrossRef
Su, Y, Swift, M. Mortality rates among carriers of ataxia-telangiectasia mutant alleles. Annals of Internal Medicine 2000;133:770–8.CrossRef
Swift, M. Public health burden of cancer in ataxia-telangiectasia heterozygotes. Journal of the National Cancer Institute 2001;93:84–5.CrossRefGoogle ScholarPubMed
Pippard, EC, Hall, AJ, Barker, DJ, Bridges, BA. Cancer in homozygotes and heterozygotes of ataxia-telangiectasia and xeroderma pigmentosum in Britain. Cancer Research 1988;48:2929–32.
Janin, N, Andrieu, N, Ossian, K, et al. Breast cancer risk in ataxia telangiectasia (AT) heterozygotes: haplotype study in French AT families. British Journal of Cancer 1999;80:1042–5.CrossRefGoogle Scholar
Geoffroy-Perez, B, Janin, N, Ossian, K, et al. Cancer risk in heterozygotes for ataxia-telangiectasia. International Journal of Cancer 2001;93:288–93.CrossRefGoogle ScholarPubMed
Geoffroy-Perez, B, Janin, N, Ossian, K, et al. Variation in breast cancer risk of heterozygotes for ataxia-telangiectasia according to environmental factors. International Journal of Cancer 2002;99:619–23.CrossRefGoogle ScholarPubMed
Andrieu, N, Cavaciuti, E, Lauge, A, et al. Ataxia-telangiectasia genes and breast cancer risk in a French family study. Journal of Dairy Research 2005;72 Spec No:73–80.CrossRefGoogle Scholar
Olsen, JH, Hahnemann, JM, Borresen-Dale, AL, et al. Cancer in patients with ataxia-telangiectasia and in their relatives in the nordic countries. Journal of the National Cancer Institute 2001;93:121–7.CrossRefGoogle ScholarPubMed
Easton, DF. Cancer risks in A-T heterozygotes. International Journal of Radiation Biology 1994;66:S177–82.CrossRefGoogle ScholarPubMed
Wooster, R, Ford, D, Mangion, J, et al. Absence of linkage to the ataxia telangiectasia locus in familial breast cancer. Human Genetics 1993;92:91–4.CrossRef
Khanna, KK, Chenevix-Trench, G. ATM and genome maintenance: defining its role in breast cancer susceptibility. Journal of Mammary Gland Biology and Neoplasia 2004;9:247–62.CrossRefGoogle ScholarPubMed
Khanna, KK. Cancer risk and the ATM gene: a continuing debate. Journal of the National Cancer Institute 2000;92:795–802.CrossRefGoogle ScholarPubMed
Athma, P, Rappaport, R, Swift, M. Molecular genotyping shows that ataxia-telangiectasia heterozygotes are predisposed to breast cancer. Cancer Genetics and Cytogenetics 1996;92:130–4.CrossRef
FitzGerald, MG, Bean, JM, Hegde, SR, et al. Heterozygous ATM mutations do not contribute to early onset of breast cancer. Nature Genetics 1997;15:307–10.CrossRef
Chen, J, Birkholtz, GG, Lindblom, P, Rubio, C, Lindblom, A. The role of ataxia-telangiectasia heterozygotes in familial breast cancer. Cancer Research 1998;58:1376–9.
Bay, JO, Grancho, M, Pernin, D, et al. No evidence for constitutional ATM mutation in breast/gastric cancer families. International Journal of Oncology 1998;12:1385–90.Google Scholar
Bebb, DG, Yu, Z, Chen, J, et al. Absence of mutations in the ATM gene in forty-seven cases of sporadic breast cancer. British Journal of Cancer 1999;80:1979–81.CrossRefGoogle ScholarPubMed
Broeks, A, Urbanus, JH, Floore, AN, et al. ATM-heterozygous germline mutations contribute to breast cancer-susceptibility. American Journal of Human Genetics 2000;66:494–500.CrossRefGoogle ScholarPubMed
Gatti, RA, Tward, A, Concannon, P. Cancer risk in ATM heterozygotes: a model of phenotypic and mechanistic differences between missense and truncating mutations. Molecular Genetics and Metabolism 1999;68:419–23.CrossRef
Waddell, N, Jonnalagadda, J, Marsh, A, et al. Characterization of the breast cancer associated ATM 7271T>G (V2424G) mutation by gene expression profiling. Genes Chromosomes Cancer 2006;45:1169–81.CrossRef
Stredrick, DL, Garcia-Closas, M, Pineda, MA, et al. The ATM missense mutation p.Ser49Cys (c.146C>G) and the risk of breast cancer. Human Mutation 2006;27:538–44.CrossRef
Thompson, D, Duedal, S, Kirner, J, et al. Cancer risks and mortality in heterozygous ATM mutation carriers. Journal of the National Cancer Institute 2005;97:813–22.CrossRefGoogle ScholarPubMed
Renwick, A, Thompson, D, Seal, S, et al. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nature Genetics 2006;38:873–5.CrossRef
Ahmed, M, Rahman, N. ATM and breast cancer susceptibility. Oncogene 2006;25:5906–11.CrossRef
Prokopcova, J, Kleibl, Z, Banwell, CM, Pohlreich, P. The role of ATM in breast cancer development. Breast Cancer Research and Treatment 2007;104:121–8.CrossRef
Stankovic, T, Stewart, GS, Byrd, P, et al. ATM mutations in sporadic lymphoid tumours. Leukemia and Lymphoma 2002;43:1563–71.CrossRef
Stilgenbauer, S, Schaffner, C, Winkler, D, et al. The ATM gene in the pathogenesis of mantle-cell lymphoma. Annals of Oncology 2000;11:127–30.CrossRef
Schaffner, C, Idler, I, Stilgenbauer, S, Dohner, H, Lichter, P. Mantle cell lymphoma is characterized by inactivation of the ATM gene. Proceedings of the National Academy of Sciences USA 2000;97:2773–8.CrossRef
Fang, NY, Greiner, TC, Weisenburger, DD, et al. Oligonucleotide microarrays demonstrate the highest frequency of ATM mutations in the mantle cell subtype of lymphoma. Proceedings of the National Academy of Sciences USA 2003;100:5372–7.CrossRef
Jares, P, Campo, E. Advances in the understanding of mantle cell lymphoma. British Journal of Haematology 2008;142:149–65.CrossRefGoogle ScholarPubMed
Stilgenbauer, S, Schaffner, C, Litterst, A, et al. Biallelic mutations in the ATM gene in T-prolymphocytic leukemia. Nature Medicine 1997;3:1155–9.CrossRef
Schaffner, C, Stilgenbauer, S, Rappold, GA, Dohner, H, Lichter, P. Somatic ATM mutations indicate a pathogenic role of ATM in B-cell chronic lymphocytic leukemia. Blood 1999;94:748–53.
Stankovic, T, Stewart, GS, Fegan, C, et al. Ataxia telangiectasia mutated-deficient B-cell chronic lymphocytic leukemia occurs in pregerminal center cells and results in defective damage response and unrepaired chromosome damage. Blood 2002;99:300–9.CrossRef
Gronbaek, K, Worm, J, Ralfkiaer, E, et al. ATM mutations are associated with inactivation of the ARF-TP53 tumor suppressor pathway in diffuse large B-cell lymphoma. Blood 2002;100:1430–7.CrossRef
Fernandez, V, Hartmann, E, Ott, G, Campo, E, Rosenwald, A. Pathogenesis of mantle-cell lymphoma: all oncogenic roads lead to dysregulation of cell cycle and DNA damage response pathways. Journal of Clinical Oncology 2005;23:6364–9.CrossRefGoogle ScholarPubMed
Greiner, TC, Dasgupta, C, Ho, VV, et al. Mutation and genomic deletion status of ataxia telangiectasia mutated (ATM) and p53 confer specific gene expression profiles in mantle cell lymphoma. Proceedings of the National Academy of Sciences USA 2006;103:2352–7.CrossRef
Boultwood, J. Ataxia telangiectasia gene mutations in leukaemia and lymphoma. Journal of Clinical Pathology 2001;54:512–16.CrossRefGoogle ScholarPubMed
Camacho, E, Hernandez, L, Hernandez, S, et al. ATM gene inactivation in mantle cell lymphoma mainly occurs by truncating mutations and missense mutations involving the phosphatidylinositol-3 kinase domain and is associated with increasing numbers of chromosomal imbalances. Blood 2002;99:238–44.CrossRef
Elkon, R, Vesterman, R, Amit, N, et al. SPIKE–a database, visualization and analysis tool of cellular signaling pathways. BMC Bioinformatics 2008;9:110.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×