<<

Adamowicz M. J Immunol Sci. (2018); 2(1): 26-31 Journal of Immunological Sciences

Commentary Open Access

Breaking up with ATM Marek Adamowicz1* 1Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RH, UK

Article Info ABSTRACT

Article Notes ATM is a master regulator of the DNA damage response (DDR). A Received: December 17, 2017 recently published report from the d’Adda di Fagagna laboratory1 sheds a light Accepted: January 18, 2018 onto our understanding of ATM activation. In this short-commentary we will *Correspondence: expand on this and other work to perceive better some of the aspects of ATM Dr. Marek Adamowicz, Genome Damage and Stability regulation. Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RH, UK; , E-mail: [email protected] ATM and DNA damage response activation © 2018 Adamowicz M. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License. ATM belongs to PI3K-like kinase family that consists of ATR, DNA-PKcs, mTOR, TRRAP and SMG1 , which share similar structures and ability to phosphorylate their substrates on Serine and Threonine residues2 20 years ago in the laboratory of Yossi Shiloh3. ATM is recruited to DNA double strand breaks. ATM (DSBs) was firstby MRN discovered complex, and composed cloned over of MRE11, and NBS1 , which senses DNA damage and

and triggers a downstream cascade that leads to the engagement of DDRquickly factors localizes like: MDC1to the andDSBs. 53BP1, This processaround DNAinduces lesions ATM ( Figureactivation 1).

P P P ATM MRN RNF8 MDC1 P P P RNF P P MDC1 168 ATM P Ub Ub H1 MRN Ub 53BP1 Ub Ub Ub Ub Me Ub Ub

Figure 1. DNA damage response cascade. First step in the DDR activation is recruitment of ATM kinase by MRN complex to DSBs through its interaction with NBS14,5. At the DSB ATM is activated (for details see Figure 2) and phosphorylates both itself and other substrates (1). One of the most important substrates of ATM is H2AX6, which provides a scaffold for the further accumulation of DDR factors7. γH2AX is recognized and bound by MDC1 that then enables further accumulation of ATM- MRN complex and spreading of γH2AX around DSBs (2)8,9. ATM-mediated of MDC1 allows also for the recruitment of RNF8 (3), which ubiquitinates H1 and enables the recruitment of another ubiquitin ligase RNF168 (4)10,11, which in turn ubiquitinates histone H2A12. Ubiquitination of H2A together with the deposition of methyl group done by MMSET13 results in the recruitment of 53BP1 (5), which coordinates with other factors DNA repair pathway choice14.

Page 26 of 31 Adamowicz M. J Immunol Sci. (2018); 2(1): 26-31 Journal of Immunological Sciences

A KAT5 P Ac FOXO3a P cAbl ATM ATM P P MRN

B

Figure 2. Close-up on ATM activation. A) Upon MRN-mediated recruitment to DSBs ATM undergoes an activation process. First, cAbl kinase phosphorylates KAT5 on Tyrosine 44 (1)18, which together with KAT5 interaction with H3K9m3 leads to KAT5 activation19. This way stimulated KAT5 will mediate acetylation of ATM in its PRD domain (Lysine 3016)(2)15,20. As a consequence of acetylation, ATM undergoes structural changes in its dimer form that allows for the phosphorylation in trans each of ATM monomers (3)16,20. ATM autophosphorylation leads to its monomerization and full activation16. At the end, activated ATM will generate a positive feedback loop and phosphorylate cAbl kinase leading to its increased activation (4)21. B) Structure of the ATM kinase (1-3056aa; Domains: FAT 1966–2566aa; kinase domain 2614–2960aa and FATC 3025–3056aa) together with its most important interactors and their binding sites1,22-25. Competition between FOXO3a and NOTCH1 for the binding to the FATC domain is depicted with arrows.

Upon recruitment to DSBs, ATM dimer undergoes a of already published reports. At the end, I will speculate lysine acetyltransferase 5 (KAT5/TIP60) at Lysine 301615, and phosphorylation of H2AX), putting them in the context whichcomplicated is followed process by of activation.autophosphorylation, First, it is acetylated in-trans onby possible future areas of ATM research. Serine 1981, leading to its monomerization and conferring about the role of NOTCH-FOXO3a competition in cells and ATM phosphorylation – is it still a thing? access of ATM substrates to the kinase domain16,17(Figure 2A). ATM phosphorylation on Serine 1981 is commonly In recent work, Adamowicz et al., show that ATM 26,27 and since localizes to the DSBs in a complex together with used as a marker for activated ATM. ATM phosphorylation was first observed28,29. It almost has been 20 years established ago that ATM then many of ATM’s autophosphorylation sites have FOXO3a and KAT5/Tip60 (from now on referred to as AAC autophosphorylation on Serine 1981 is necessary for ATM been identified 16, 20. Additionally it ATM(ATM and Activation KAT5 (Complex)).Figure 2A Formationand B of this complex is inhibiteddependent by on a FOXO3awell-known bridging transcription an interaction factor, NOTCH between1, has been shown that ATM mutants carrying the S1981A ). Formation of AAC is monomerization and kinase activation 16 ATM binding ability24 in transfection experiments . In agreement with those substitution do not rescue radio-sensitivity of the A-T cells towhich MRN-mediated is a negative recruitment regulator of of ATM ATM activation to DSBs without through the its (Figure 2B). Interestingly this leads of ATM autophosporylation resulted in the attenuation reports we have shown that NOTCH1-mediated inhibition by the lack of autophosphorylation (pSer1891ATM)1. of phosphorylation of ATM downstream substrates and latter being activated at the site of the damage, as observed checkpoint impairment1,24. On the other hand it has been reported that mouse Atm S1987A mutants (mouse (includingStrikingly, althoughATM-mediated all downstream DDR eventslike: , are CHK2,attenuated SMC1), due phosphorylation to NOTCH1-mediated of the ATM ATM main inactivation substrate, 30 Atmequivalent autophosphorylation of human S1981A) sites (S367 are proficient and S1893) in DDRare 1,24. Below I will try to focus on those and ATM kinase activity . Moreover,31. Interestingly it seems in the that in other vitro histone H2AX (Ser139H2AX, from now on referred as kinase assay ATM S1981A mutant can phosphorylate CHK2 γH2AX) is not affected dispensable for Atm activation two aspects of ATM activation (ATM autophosphorylation Page 27 of 31 Adamowicz M. J Immunol Sci. (2018); 2(1): 26-31 Journal of Immunological Sciences and p53 in response to linear dsDNA to the same extent as the wild-type ATM32 inhibiting ATM-mediated p53 phosphorylation in vitro24, presenceHowever unexpectedly,of either DNA-PKcs when orwe ATR performed inhibitors analysis we did of notthe in the light of the new results. Although demonstrating we observed disruption NOTCH1 of γH2AX foci formation in NOTCH1-expressing cells in the 1 it would be interesting to observe any difference in the H2AX phosphorylation (data KAT5 and FOXO3a from thein AACvitro ATM kinase assay in the unpublished – data available upon request). see if addition of: KAT5, FOXO3a, H3K9m3 and cAbl would by protein kinases other than ATM, ATR and DNA- presence of NOTCH1 and mutated ATM (S1981A). These results suggest that H2AX can be phosphorylated give similar results in an PKcs. Indeed, it has been reported that JNK and p38 can It has been suggested that ATM autophosphorylation is necessary for ATM retention at the DSBs33 because the 40,41. Additionally, it has been ATM S1981A mutant, although recruited normally to DSBs, phosphorylate H2AX in response of UV light irradiation was not stabilized properly at the site of the damage33. responseor starvation to IR in respectively parallel to ATM kinase42 reportedwas shown that to be VRK1 necessary kinase for can the phosphorylate accumulation H2AX of DDR in inhibited ATM autophosphorylation it did not affect ATM’s factors around DSBs, which implies more. Moreover,complex role VRK1 of retentionIn contrast at wethe haveDSBs 1. reported To understand that although these apparently NOTCH1 43.

VRK1Because in the DDRNOTCH1, that needs unlike further small elucidation molecule ATM contradictory observations one should look not24, therefore just at the it we can speculate that NOTCH1 binding to ATM could wouldATM autophosphorylation. be better to compare We our have results previously with the reported reports stronglyinhibitors, impair cannot ATM directly substrate inhibit recognition, ATM kinase resulting activity describingthat NOTCH1 recruitment inhibits kinase of the activity ATM ofkinase ATM dead mutant in an inhibition of phosphorylation of some substrates (ATM KD). Indeed it has been shown that ATM KD mutant (that cannot undergo autophosphorylation) is recruited reported that NOTCH1 can bind and hence modulate the to the DSBs without any impairment of retention34,35. This substratesuch as p53 recognition or CHK2, of but LSD1 not H2AX.demethylase It has 44 been. Therefore, already impact on ATM recruitment and autophosphorylation. ATM, NOTCH1 would strongly impair substrate recognition Additionally,is similar to it our was observation reported that of cells the carrying NOTCH1-mediated mutations ofit canATM. be Interestingly,possible that byit hasbinding been to shown the FATC in yeastdomain that of DSBs (without retention impairment); although ATM does can impair Tel1 phosphorylation of Rad53, which was in the KU70 and MRE11 nuclease actively recruit ATM to the36 not undergo autophosphorylation in those conditions . deletion of last 10 amino acids (aa) of the FATC domain 1,24 complex45 reports, show our still incomplete understanding of ATM connected with the loss in its ability to interact with MRX Overall, our results , together with above mentioned ATM in the substrate recognition by stimulating ATM binding to. its Moreover, substrates MRN like complex p53 or CHK2 was 46 shown. On the to other help DDR, that needs further elucidation. autophosporylation and its role in the ATM activity and hand it has been published that in human cells deletion of Meeting γH2AX at the FATC end impairment of ATM MRN-mediated response, but rather its last 10aa of the FATC domain of ATM does47 not lead to the andWe phosphorylation have repeatedly of observed downstream that NOTCH1 substrates inhibits such ATM as ability to activate upon oxidative stress . We have shown1 activation by blocking its autophosphorylation1,24 (Ser1981) not affect interaction between ATM and MRN complex . KAP1, SMC1, p53, DNA-PKcs or CHK2 (Adamowicz et al – that although NOTCH1 binds to the FATC domain it does NOTCH1 is perturbing ATM substrate recognition this using Xenopus laevis egg extract that NOTCH1 blocked a effectThis suggests is rather that mediated if by binding by inhibition to the ATMof KAT5-mediated FATC domain substantialin press). Additionally, amount of we ATM-mediated have observed phosphorylation in our initial study24. acetylation. Impairment of ATM acetylation will then block structural changes in ATM that would lead to its phosphorylation defects, phosphorylation of ATM’s main However, of all observed NOTCH1-mediated ATM of the ATM kinase domain otherwise hindered inside of its monomerization17 and activation, inhibiting this way release substrate,It has been H2AX, suggested remained that unaffected. there is redundancy between dimer structure . Taking ATM down a NOTCH PI3K-like37,38 kinases in terms of H2AX phosphorylation. DNA- PKcsis phosphorylated or ATR kinases to havethe same been shownextent toin phosphorylatethe ATM WT and marked as an due to its ability to induce H2AXand KO cells. Additionally,37,39. Indeed, it experiments has been reported carried thatout in H2AX our tumourNOTCH1 growth was48 very early connected to the tumorogenesis laboratory showed that NOTCH1 neither blocks ATR nor present in many T-cell acute lymphoblastic leukemias (T-ALL)49 or breast. Activatingcancers50 mutations in NOTCH1 are

DNA-PKcs kinase activity (Adamowicz et al – in press). . Indeed, we have found that

Page 28 of 31 Adamowicz M. J Immunol Sci. (2018); 2(1): 26-31 Journal of Immunological Sciences

24. At the NOTCH1same time, expression other group was reported negatively that ectopic correlated expression with the of ATMNOTCH1 activation in cancer in the cells human lead breast to their cancer increased patients resistance ATM Synapsin-I VAMP2 51 . Those data show that expression Peroxisome to DNA damage in vivo ? ATM ? of high levels of NOTCH1 (due to activating mutations or NEMO ? in cancer cells, resulting probably from the inhibition of ATM KAT5? ectopic expression) stimulate radioresistance and survival NEMO FOXO3a? ATM 52 KAT5 ATM ATMIN FOXO3a NOTCH1 induce faster proliferation leading to replication MRN ? ? stress.p53-mediated Although . we showed Additionally, that NOTCH1 increased inhibits levels ATM of ATM H1 H1 cancersactivation, from this replication is not true stress. for ATR kinase (Adamowicz et Figure 3. Existence of different ATM complexes in the cells. al – in press) resulting in the protection of NOTCH1-driven Schematic representation of the existence of different ATM Neural stem cells (NSC) are known to express moderate complexes both in the nucleus as well as in the cytosol. For details please look into the text. proliferation53. Interestingly it has been reported that levelsinduction of activatedof DDR NOTCH1,in NSC leads which to is necessarytheir spontaneous for their differentiation to astrocytes, which is dependent on ATM Figure 3). 54. It is therefore possible to speculate that AAC is preserved while complexed with ATMIN, and if so howWe it istend involved to think in the about ATM ATM activation through ( its role in DDR re tipactivation the balance allowing for DNA damage repair without in many more cellular processes like: stress response60, inducingNOTCH1-mediated differentiation. downregulation It is important of ATM to activationremember could that neuronalgulation, signal although transmission ATM has been61 or described pexophagy to be62,63 involved(Figure 3 physiological levels of NOTCH1 are low as compared to nucleus. This implies that ATM is not always in complex ), which in every case requires its presence outside the those observed in T-ALL cells or those achieved by ectopic with KAT5 or MRN complex. The presence of ATM in beexpression, an outcome therefore of many observed different effects factors of NOTCH1 impacting activation at the might be very mild. Additionally, observed results might which is responsible for ATM peroxisome localization. In same time on DDR. Indeed, it has been shown that SALL4 peroxisomes is a result of its interaction with PEX5,

55 . peroxisomeperoxisomes, phagocytosis ATM is activated63 by reactive oxygen species transcription factor expressed in stem cells favours ATM andthe stressformation response of an activeis connected dimer, allowing to its interaction ATM to control with activationIn summary, by its binding I would to MRN like complex to propose that the physiological role of NOTCH1 is not to inhibit fully ATM NEMO and with shuttling between. The involvement nucleus and of cytosol ATM 60 in. Additionally, ATM has been described to localize in the 64 modulate a balance between the amount of DNA damage cytosol of neuronal cells and has been implicated in the andactivation, DDR signalling. but rather This to would induce result its mild in the impairment, suppression to and Synapsin-I61. It would be interesting to see if NOTCH1, neuronal signal transmission by its interaction with VAMP21 time for necessary repair. , of DNA damage induced apoptosis or differentiation, giving could be used as a tool for identifying new regulatory There is plenty more ATM in the sea componentswhich has very of ATMstrong complexes affinity to in the the FATC cytosol. domain of ATM at DSBs and DDR1,24. Apart from AAC, ATM relies also on MRNFormation complex, ofwhich AAC allows is necessary proper AAC for localization ATM activation and It is thought-provoking to picture ATM in different substrate recognition. Interestingly, in the nucleus, ATM ATMcomplexes complexes that opens differentially new and regulateexciting areas its activity of research and has been described to exist in two different complexes. In substrate recognition. The identification of different active has been shown that there is competition between MRN complex and ATMIN for binding to ATM (Figure 3)56,57. and raises even more fascinating questions. For example, regulated?how is ATM Hopefully, activation in the in near those future complexes we will stimulated, know the answers.and how is ATM substrate recognition and kinase activity andStudies hypotonic have found stresses that 58,59while. It MRN is possible complex that guides like ATM MRN, in theATMIN response by binding to DSBs, to ATM ATMIN regulates is necessary it substrate during recognition oxidative Acknowledgements stimuli. It would be interesting to see if the structure of Caldecott for their critical reading of the manuscript. I and therefore its kinase activity in response to different I thank Sheetal Sharma, Jelena Vermezovic and Keith

Page 29 of 31 Adamowicz M. J Immunol Sci. (2018); 2(1): 26-31 Journal of Immunological Sciences apologize to all the authors whose work I could not include 18. Kaidi A, Jackson SP. KAT5 tyrosine phosphorylation couples due to the space constrains of this short-commentary/ sensing to ATM signalling. Nature. 2013; 498: 70-74. 19.

Conflict of Interest Sun2009; Y, 11Jiang: 1376-1382. X, Xu Y, et al. Histone H3 methylation links DNA damage review. detection to activation of the tumour suppressor Tip60. Nat Cell Biol. 20. 27: Sun8502-8509. Y, Xu Y, Roy K, et al. DNA damage-induced acetylation of lysine References 3016 of ATM activates ATM kinase activity. Mol Cell Biol. 2007; I declare no conflict of interest. 21. Baskaran R, Wood LD, Whitaker LL, et al. Ataxia telangiectasia 1. radiation. Nature. 1997; 387: 516-519. KAT5/Tip60Adamowicz M, Complex. Vermezovic Cell Rep. J, d’Adda 2016; di 16 Fagagna: 2068-2076. F. NOTCH1 Inhibits mutant protein activates c-Abl tyrosine kinase in response to ionizing Activation of ATM by Impairing the Formation of an ATM-FOXO3a- 22. 2. biochemistry. 2015; 84: 711-738. Pessina F, Lowndes NF. The RSF1 Histone-Remodelling Factor12. Paull TT. Mechanisms of ATM Activation. Annual review of Facilitates DNA Double-Strand Break Repair by Recruiting 3. 23. Centromeric and Fanconi Anaemia Proteins. PLoS Biology. 2014; with a product similar to PI-3 kinase. Science. 1995; 268: 1749- phosphorylates p53: mapping the region of interaction. Nature Sa1753.vitsky K, Bar-Shira A, Gilad S, et al. A single ataxia telangiectasia Genetics. Khanna K, 1998; Keating 20: 398-400. KE, Kozlov S, et al. ATM associates with and 4. 24. ATR and DNA-PKcs to sites of DNA damage. Nature. 2005; 434: 605- 611. Falck,J, Coates J, Jackson SP. Conserved modes of recruitment of ATM, V2015;ermezovic 22: 417-424. J, Adamowicz M, Santarpia L, et al. Notch is a direct negative regulator of the DNA-damage response. Nat Struct Mol Biol. 5. Nakada D, Matsumoto K, Sugimoto K. ATM-related Tel1 associates 25.

17: 1957-1962. Jiang X, Sun Y, Chen S, et al. The FATC Domains of PIKK Proteins Are with double-strand breaks through an Xrs2-dependent mechanism. Functionally Equivalent and Participate in the Tip60-dependent 6. 2006; 281: 15741-15746. Dev. 2003; Activation of DNA-PKcs and ATM. Journal of Biological Chemistry. in response to DNA double-strand breaks. The Journal of biological 26. chemistry. Burma S, Chen 2001; BP, 276 Murphy: 42462-42467. M, et al. ATM phosphorylates histone H2AX Kim ST, Lim DS, Canman CE, et al. Substrate specificities and 7. The Journal of biological chemistry. 1999; 274: 37538-37543. identification of putative substrates of ATM kinase family members. phosphorylation is dispensable for the initial recognition of DNA 27. breaks. Celeste Nature A, Fernandez-Capetillo Cell Biology. 2003; O, 5 Kruhlak: 675-679. MJ, et al. Histone H2AX by ionizing radiation and phosphorylation of p53. Science (New York, Canman CE, Lim DS, Cimprich KA, et al. Activation of the ATM kinase 8. Stucki M, Clapperton JA, Mohammad D, et al. MDC1 directly binds N.Y.). 1998; 281: 1677-1679. 28. Matsuoka S, Ballif BA, Smogorzewska A, et al. ATM and ATR substrate double-strand breaks. Cell. 2005; 123: 1213-1226. phosphorylated histone H2AX to regulate cellular responses to DNA 9. Chapman JR, Jackson, SP. Phospho-dependent interactions between damage. Science (New York, N.Y.). 2007; 316: 1160-1166. analysis reveals extensive protein networks responsive to DNA NBS1 and MDC1 mediate chromatin retention of the MRN complex at 29. sites of DNA damage. EMBO reports. 2008; 9: 795-801. Kozlov SV, Graham ME, Peng C, et al. Involvement of novel 10. Thorslund T, Ripplinger A, Hoffmann S, et al. Histone H1 couples 2006; 25: 3504-3514. autophosphorylation sites in ATM activation. The EMBO journal. 30. Nature. 2015; 527: 389–393. initiation and amplification of ubiquitin signalling after DNA damage. Pellegrini M, Celeste A, Difilippantonio S, et al. Autophosphorylation 11. Nature. 2006; 443: 222-225. at serine 1987 is dispensable for murine Atm activation in vivo. at DNA double-strand breaks and promotes assembly of 31. Daniel JA, Pellegrini M, Lee JH, et al. Multiple autophosphorylation repair Mailand proteins. N, Bekker-Jensen Cell. 2007; 131 S, Faustrup: 887-900. H, et al. RNF8 ubiquitylates 12. of cell biology. 2008; 183: 777-783. sites are dispensable for murine ATM activation in vivo. The Journal 32. accumulation Doil C, Mailand of repair N, Bekker-Jensen proteins. Cell. 2009; S, et 136 al. : RNF168435-446. binds and through the Mre11-Rad50-Nbs1 complex. Science. 2005; (New York, amplifies ubiquitin conjugates on damaged to allow Lee JHH, Paull TT. ATM activation by DNA double-strand breaks 13. Pei H, Zhang L, Luo K, et al. MMSET regulates histone H4K20 N.Y.) 308: 551-554. methylation and 53BP1 accumulation at DNA damage sites. Nature. 33. 2011; 470: 124-128. ATM at DNA damage sites. J Cell Biol. 2009; 187: 977-990. So S, Davis AJ, Chen DJ. Autophosphorylation at serine 1981 stabilizes 14. Panier S, Boulton SJ. Double-strand break repair: 53BP1 comes into 34. 15: 7-18. to embryonic lethality in mice. The Journal of cell biology. 2012; 198: Daniel295-304. JA, Pellegrini M, Lee BS, et al. Loss of ATM kinase activity leads 15. focus. Nature reviews. Molecular cell biology. 2014; 35. Yamamoto K, Wang Y, Jiang W, et al. Kinase-dead ATM protein causes 2005; Sun Y, Jiang102: X,13182-13187. Chen S, et al. A role for the Tip60 histone acetyltransferase genomic instability and early embryonic lethality in mice. J Cell Biol. in the acetylation and activation of ATM. Proc Natl Acad Sci U S A. 16. 2012; 198: 305-313. intermolecular autophosphorylation and dimer dissociation. Nature. 36. 2003; Bakkenist 421 : CJ,499-506. Kastan MB. DNA damage activates ATM through the ATM kinase in the absence of DNA-damage sensors. Nat Struct Mol Hartlerode AJ, Morgan MJ, Wu Y, et al. Recruitment and activation of 17. Biol. 2015; 22: 736-743. 37. Stiff T, O’Driscoll M, Rief N, et al. ATM and DNA-PK function redundantly structural Sawicka M, organisation. Wanrooij PH, The Darbari Journal VC, ofet al.biological The dimeric chemistry. architecture 2016; 291of checkpoint: 13436-13447. kinases Mec1ATR and Tel1ATM reveal a common research. 2004; 64: 2390-2396. to phosphorylate H2AX after exposure to ionizing radiation. Cancer

Page 30 of 31 Adamowicz M. J Immunol Sci. (2018); 2(1): 26-31 Journal of Immunological Sciences

38. 52. Mazz one M Selfors LM, Albeck J, et al. Dose-dependent induction dependent manner in response to replicational stress. The Journal of biological Ward IM, chemistry. Chen J. Histone2001; 276 H2AX: 47759-47762. is phosphorylated in an ATR- mammary epithelial cells. Proceedings of the National Academy of distinctSciences phenotypic of the United responses States toof NotchAmerica. pathway 2010; activation 107: 5012- in 39. Riballo E, Kühne M, Rief N, et al. A pathway of double-strand break 5017.

16: 715-724. 53. rejoining dependent upon ATM, Artemis, and proteins locating to essential for the maintenance, but not the generation, of mammalian 40. gamma-H2AX foci. Molecular cell. 2004; Hit oshi S, Alexson T, Tropepe V, et al. Notch pathway16: 846-858. molecules are

Luletters. C, 2008; Shi Y, 582 Wang: 2703-2708. Z, et al. Serum starvation induces H2AX 54. neural stem cells. Genes & Development. 2002; phosphorylation to regulate apoptosis via p38 MAPK pathway. FEBS Neural Stem Cells Leads to Astrocytic Differentiation Mediated by 41. BMP2 Schneider Signaling L, Pellegatta through S, JAK-STAT.Favaro R, etStem al. DNACell Reports.Damage in2013; Mammalian 1: 123- 138. cell.Lu C, 2006; Zhu F, 23 Cho: 121-132. YY, et al. Cell apoptosis: requirement of H2AX in DNA ladder formation, but not for the activation of caspase-3. Molecular 55. 42. DNA damage response in embryonic stem cells. The Journal of Cell XiongBiology. J, Todorova2015; 208 D,: 513-520.Su NY, et al. Stemness factor Sall4 is required for SalzanoDNA damage. M, Sanz-García . M, Monsalve2015; 10 :DM, 373-383. et al. VRK1 chromatin kinase phosphorylates H2AX and is required for foci formation induced by 56. Zhang T, Cronshaw J, Kanu N, et al. UBR5-mediated ubiquitination 43. of ATMIN is required for ionizing radiation-induced ATM signaling and function. Proceedings of the National Academy of Sciences of the Sanz -García M, Monsalve DM, Sevilla A. et al. Vaccinia-related kinase 1 of 53BP1 foci in response to ionizing radiation-induced DNA damage. United States of America. 2014; 111: 12091-12096. The(VRK1) Journal is an of upstream biological nucleosomal chemistry. 2012; kinase 287 required: 23757-23768. for the assembly 57. Zhang T, Penicud K, Bruhn C, et al. Competition between NBS1 and 44. Yatim A, Benne C, Sobhian B, et al. NOTCH1 nuclear interactome ATMIN controls ATM signaling pathway choice. Cell reports. 2012; 2: 1498-1504. function. Mol Cell. 2012; 48: 445-458. reveals key regulators of its transcriptional activity and oncogenic 58. 45. 26: 2933-2941. protein kinase Tel1 for localization to DNA ends and target protein Kanu N, Behrens A. ATMIN defines an NBS1-independent pathway of Ogirecognition. H, Goto GH,Mol GhoshBiol Cell. A, 2015;et al. Requirement26: 3480-3488. of the FATC domain of 59. ATM signalling. The EMBO journal. 2007;

46. Kanubrain. N, The Penicud Journal K, ofHristova biological M, et chemistry. al. The ATM 2010; cofactor 285: ATMIN38534-38542. protects Mre11/Rad50/Nbs1 complex. Science (New York, N.Y.). 2004; 304: 93-96. against oxidative stress and accumulation of DNA damage in the aging Lee JHH, Paull TT. Direct activation of the ATM protein kinase by the 60. W u ZHH, Shi Y, Tibbetts RS, et al. Molecular linkage between the 47. Science (New York, N.Y.). 2010; 330: 517-521. Science (New York, N.Y.). 2006; 311: 1141-1146. Guo Z, Kozlov S, Lavin MF, et al. ATM activation by oxidative stress. kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. 48. 61. Li J, Han YR, Plummer MR, et al. Cytoplasmic ATM in neurons neoplasms in mice transplanted with bone marrow expressing modulates synaptic function. Current biology : CB. 2009; 19: 2091- Pear WS, Aster JC, Scott ML, et al. Exclusive development of T cell 2096. 183: 2283-2291. activated Notch alleles. The Journal of Experimental Medicine. 1996; 62. W atters D, Kedar P, Spring K, et al. Localization of a portion of 49. extranuclear ATM to peroxisomes. The Journal of biological chemistry. NOTCH1 in Human T Cell Acute Lymphoblastic Leukemia. 1999; 274: 34277-34282. WScience.eng AP, 2004; Ferrando 306: 269-271. AA, Lee W, et al. Activating Mutations of 63. Zhang J, Tripathi DN, Jing J, et al. ATM functions at the peroxisome to 50. induce pexophagy in response to ROS. Nature cell biology. 2015; 17: 1259-1269. PJournalece S, of Serresi Cell Biology. M, Santolini 2004; E,167 et: al.215-221. Loss of negative regulation by Numb over Notch is relevant to human breast . The 64. Bar low C, Ribaut-Barassin C, Zwingman TA, et al. ATM is a cytoplasmic 51. radiation resistance in non small cell lung cancer. Radiotherapy and Proceedings of the National Academy of Sciences of the United States Oncology.Theys J, Yahyanejad 2013; 108 :S, 440-445. Habets R et al. High NOTCH activity induces ofprotein America. in mouse 2000; brain97: 871-876. required to prevent lysosomal accumulation.

Page 31 of 31