autophosphorylationandatmactivation239341/uq239341_oa.pdfautophosphorylationandatmactivation...

30
Autophosphorylation and ATM Activation ADDITIONAL SITES ADD TO THE COMPLEXITY * S Received for publication, November 16, 2010 Published, JBC Papers in Press, December 13, 2010, DOI 10.1074/jbc.M110.204065 Sergei V. Kozlov ‡1 , Mark E. Graham §2 , Burkhard Jakob , Frank Tobias , Amanda W. Kijas , Marcel Tanuji , Philip Chen , Phillip J. Robinson § , Gisela Taucher-Scholz , Keiji Suzuki , Sairai So**, David Chen**, and Martin F. Lavin ‡ ‡‡3 From Radiation Biology and Oncology, Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia, the § Children’s Medical Research Institute, University of Sydney, Westmead, New South Wales 2145, Australia, GSI Helmholtzzentrum fu ¨r Schwerionenforschung GmbH, Biophysik, Planckstrasse 1, D-64291 Darmstadt, Germany, the Department of Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan, the **University of Texas Southwestern Medical Center, Dallas, Texas 75390, and the ‡‡ University of Queensland Centre for Clinical Research, Brisbane, Queensland 4029, Australia The recognition and signaling of DNA double strand breaks involves the participation of multiple proteins, in- cluding the protein kinase ATM (mutated in ataxia-telangi- ectasia). ATM kinase is activated in the vicinity of the break and is recruited to the break site by the Mre11-Rad50-Nbs1 complex, where it is fully activated. In human cells, the acti- vation process involves autophosphorylation on three sites (Ser 367 , Ser 1893 , and Ser 1981 ) and acetylation on Lys 3016 . We now describe the identification of a new ATM phosphoryla- tion site, Thr(P) 1885 and an additional autophosphorylation site, Ser(P) 2996 , that is highly DNA damage-inducible. We also confirm that human and murine ATM share five identi- cal phosphorylation sites. We targeted the ATM phospho- rylation sites, Ser 367 and Ser 2996 , for further study by gener- ating phosphospecific antibodies against these sites and demonstrated that phosphorylation of both was rapidly in- duced by radiation. These phosphorylations were abolished by a specific inhibitor of ATM and were dependent on ATM and the Mre11-Rad50-Nbs1 complex. As found for Ser(P) 1981 , ATM phosphorylated at Ser 367 and Ser 2996 local- ized to sites of DNA damage induced by radiation, but ATM recruitment was not dependent on phosphorylation at these sites. Phosphorylation at Ser 367 and Ser 2996 was functionally important because mutant forms of ATM were defective in correcting the S phase checkpoint defect and restoring ra- dioresistance in ataxia-telangiectasia cells. These data pro- vide further support for the importance of autophosphor- ylation in the activation and function of ATM in vivo. DNA double strand breaks (DSB) 4 arise during normal physiological cell processes, such as immunoglobulin and T cell receptor gene rearrangements (1), but also arise in re- sponse to exposure to a variety of DNA-damaging agents that, if left unrepaired, lead to cell death or genomic instability, cancer, and other pathologies (2). DNA DSB are repaired in mammalian cells by non-homologous end joining and homol- ogous recombination that predominate at different stages of the cell cycle (3). The Mre11-Rad50-Nbs1 (MRN) complex is a primary sensor of DNA DSB, and there is evidence that it participates in both forms of DSB repair (4 –7). Hypomorphic mutations in members of this complex give rise to disorders characterized by sensitivity to agents that generate DNA DSB, cell cycle checkpoint defects, genome instability, and neuro- logical abnormalities (8 –10). These syndromes overlap in their clinical and cellular phenotypes with ataxia-telangiecta- sia (A-T) defective in ATM kinase (11). ATM is a member of the phosphoinositide 3-kinase-like kinase (PIKK) family of proteins and is primarily activated by DNA DSB to signal to both the DNA repair machinery and the cell cycle checkpoints (12, 13). Considerable progress has been made in understanding the mechanism of ATM activa- tion (14 –17). Although the nature of the initial stimulus for activation remains undefined, it seems likely that relaxation of chromatin structure is sufficient to initiate activation (14). ATM is constitutively present as an inactive dimer that mo- nomerizes as part of the activation process, facilitated by both autophosphorylation and acetylation (14, 18). More recently, Sun et al. (19) have shown that after DNA damage, casein kinase 2 phosphorylates and releases HP1 from chromatin, which enables the recruitment of the ATM-Tip60 complex to MRN at the break site. This facilitates interaction between the chromodomain of Tip60 and the unbound histone H3 K9me3, leading to acetylation and activation of the kinase activity of ATM. The initial activation of ATM appears to be incomplete because it relies on recruitment to the site of the break by the MRN complex for full activation (15). This is supported by a variety of in vitro and in vivo studies that dem- * This work was supported, in whole or in part, by National Institutes of Health Grants CA050519 and CA92584 (to D. C.). This work was also sup- ported by grants from the National Health and Medical Research Council (to M. F. L.) and GSI Helmholtzzentrum für Schwerionenforschung GmbH (to G. T.-S. and B. J.). S The on-line version of this article (available at http://www.jbc.org) con- tains supplemental Table 1 and Figs. 1–9. 1 To whom correspondence may be addressed. Tel.: 61-7-3362-0336; Fax: 61-7-3362-0106; E-mail: [email protected]. 2 Supported by a National Health and Medical Research Council Career De- velopment Award. 3 To whom correspondence may be addressed. Tel.: 61-7-3362-0336; Fax: 61-7-3362-0106; E-mail: [email protected]. 4 The abbreviations used are: DSB, double strand break(s); MRN, Mre11- Rad50-Nbs1; A-T, ataxia-telangiectasia; PIKK, phosphoinositide 3-kinase- like kinase; Gy, gray(s); LET, linear energy transfer. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 11, pp. 9107–9119, March 18, 2011 © 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9107 at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from at UQ Library on September 5, 2016 http://www.jbc.org/ Downloaded from

Upload: others

Post on 09-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Autophosphorylation and ATM ActivationADDITIONAL SITES ADD TO THE COMPLEXITY*□S

Received for publication, November 16, 2010 Published, JBC Papers in Press, December 13, 2010, DOI 10.1074/jbc.M110.204065

Sergei V. Kozlov‡1, Mark E. Graham§2, Burkhard Jakob¶, Frank Tobias¶, Amanda W. Kijas‡, Marcel Tanuji‡,Philip Chen‡, Phillip J. Robinson§, Gisela Taucher-Scholz¶, Keiji Suzuki�, Sairai So**, David Chen**,and Martin F. Lavin‡ ‡‡3

From ‡Radiation Biology and Oncology, Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia, the§Children’s Medical Research Institute, University of Sydney, Westmead, New South Wales 2145, Australia, ¶GSI Helmholtzzentrumfur Schwerionenforschung GmbH, Biophysik, Planckstrasse 1, D-64291 Darmstadt, Germany, the �Department of MolecularMedicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan, the**University of Texas Southwestern Medical Center, Dallas, Texas 75390, and the ‡‡University of Queensland Centre for ClinicalResearch, Brisbane, Queensland 4029, Australia

The recognition and signaling of DNA double strandbreaks involves the participation of multiple proteins, in-cluding the protein kinase ATM (mutated in ataxia-telangi-ectasia). ATM kinase is activated in the vicinity of the breakand is recruited to the break site by the Mre11-Rad50-Nbs1complex, where it is fully activated. In human cells, the acti-vation process involves autophosphorylation on three sites(Ser367, Ser1893, and Ser1981) and acetylation on Lys3016. Wenow describe the identification of a new ATM phosphoryla-tion site, Thr(P)1885 and an additional autophosphorylationsite, Ser(P)2996, that is highly DNA damage-inducible. Wealso confirm that human and murine ATM share five identi-cal phosphorylation sites. We targeted the ATM phospho-rylation sites, Ser367 and Ser2996, for further study by gener-ating phosphospecific antibodies against these sites anddemonstrated that phosphorylation of both was rapidly in-duced by radiation. These phosphorylations were abolishedby a specific inhibitor of ATM and were dependent on ATMand the Mre11-Rad50-Nbs1 complex. As found forSer(P)1981, ATM phosphorylated at Ser367 and Ser2996 local-ized to sites of DNA damage induced by radiation, but ATMrecruitment was not dependent on phosphorylation at thesesites. Phosphorylation at Ser367 and Ser2996 was functionallyimportant because mutant forms of ATM were defective incorrecting the S phase checkpoint defect and restoring ra-dioresistance in ataxia-telangiectasia cells. These data pro-vide further support for the importance of autophosphor-ylation in the activation and function of ATM in vivo.

DNA double strand breaks (DSB)4 arise during normalphysiological cell processes, such as immunoglobulin and Tcell receptor gene rearrangements (1), but also arise in re-sponse to exposure to a variety of DNA-damaging agents that,if left unrepaired, lead to cell death or genomic instability,cancer, and other pathologies (2). DNA DSB are repaired inmammalian cells by non-homologous end joining and homol-ogous recombination that predominate at different stages ofthe cell cycle (3). The Mre11-Rad50-Nbs1 (MRN) complex isa primary sensor of DNA DSB, and there is evidence that itparticipates in both forms of DSB repair (4–7). Hypomorphicmutations in members of this complex give rise to disorderscharacterized by sensitivity to agents that generate DNA DSB,cell cycle checkpoint defects, genome instability, and neuro-logical abnormalities (8–10). These syndromes overlap intheir clinical and cellular phenotypes with ataxia-telangiecta-sia (A-T) defective in ATM kinase (11).ATM is a member of the phosphoinositide 3-kinase-like

kinase (PIKK) family of proteins and is primarily activated byDNA DSB to signal to both the DNA repair machinery andthe cell cycle checkpoints (12, 13). Considerable progress hasbeen made in understanding the mechanism of ATM activa-tion (14–17). Although the nature of the initial stimulus foractivation remains undefined, it seems likely that relaxation ofchromatin structure is sufficient to initiate activation (14).ATM is constitutively present as an inactive dimer that mo-nomerizes as part of the activation process, facilitated by bothautophosphorylation and acetylation (14, 18). More recently,Sun et al. (19) have shown that after DNA damage, caseinkinase 2 phosphorylates and releases HP1� from chromatin,which enables the recruitment of the ATM-Tip60 complex toMRN at the break site. This facilitates interaction between thechromodomain of Tip60 and the unbound histone H3K9me3, leading to acetylation and activation of the kinaseactivity of ATM. The initial activation of ATM appears to beincomplete because it relies on recruitment to the site of thebreak by the MRN complex for full activation (15). This issupported by a variety of in vitro and in vivo studies that dem-

* This work was supported, in whole or in part, by National Institutes ofHealth Grants CA050519 and CA92584 (to D. C.). This work was also sup-ported by grants from the National Health and Medical Research Council(to M. F. L.) and GSI Helmholtzzentrum für Schwerionenforschung GmbH(to G. T.-S. and B. J.).

□S The on-line version of this article (available at http://www.jbc.org) con-tains supplemental Table 1 and Figs. 1–9.

1 To whom correspondence may be addressed. Tel.: 61-7-3362-0336; Fax:61-7-3362-0106; E-mail: [email protected].

2 Supported by a National Health and Medical Research Council Career De-velopment Award.

3 To whom correspondence may be addressed. Tel.: 61-7-3362-0336; Fax:61-7-3362-0106; E-mail: [email protected].

4 The abbreviations used are: DSB, double strand break(s); MRN, Mre11-Rad50-Nbs1; A-T, ataxia-telangiectasia; PIKK, phosphoinositide 3-kinase-like kinase; Gy, gray(s); LET, linear energy transfer.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 11, pp. 9107–9119, March 18, 2011© 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9107

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

at U

Q L

ibrary on September 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

onstrate MRN dependence on the activation of ATM (20–23). In addition, although ATM can be activated in cells defi-cient in Nbs1, it fails to localize to nuclear foci at sites of DNAdamage in these cells (24). Once activated, ATM can thenphosphorylate a multitude of substrates that participate indifferent cellular processes, including DNA repair and cellcycle control (25). A cascade of reactions is initiated, startingwith the phosphorylation of H2AX (�H2AX), leading to chro-matin modification that assists in signaling reactions andDNA repair (26). Recent data suggest that dephosphorylationat Tyr142 is a prerequisite for H2AX phosphorylation and thatthis alteration plays a central role in the assembly of the DNAdamage response complex (27). Assembly of DNA damageresponse proteins at the break site is consolidated by phos-phorylation and ubiquitylation (13). ATM phosphorylates anumber of these proteins, including MDC1, Nbs1, 53BP1, andBRCA1. Phosphorylation of MDC1 by a second enzyme, ca-sein kinase-2, is responsible for the retention of the MRNcomplex on chromatin (28, 29). Phosphorylation of MDC1 atATM consensus sites facilitates interaction with the RINGfinger ubiquitin ligase, RNF8, which in turn ubiquitylatesH2A and H2AX, leading to the accumulation of 53BPI,BRCA1, and other proteins at the site of damage (30–32). It isnow evident that this accumulation is consolidated by theinvolvement of a second ubiquitin ligase, RNF168, that alsoubiquitylates H2A and H2AX (33, 34). In addition to activa-tion by DNA DSB recent data show that ATM is also an im-portant sensor of reactive oxygen species in human cells (35).Oxidation directly activates ATM, there is no dependence onthe MRN complex for activation, and the protein is present asa disulfide-linked dimer under these conditions.As for other protein kinases, autophosphorylation is ob-

served at several sites in ATM (14, 36). These sites have beendescribed in human cells employing mass spectrometry andinclude Ser367, Ser1893, and Ser1981. Mutants in these sites failto correct the extent of radiation-induced cell death and cellcycle defects in A-T cells (36). On the other hand, the de-pendence on autophosphorylation for ATM activation is notobserved in mutant Atm mice generated by BAC recom-bineering (37, 38). In these experiments, BAC constructs withS367A, S1899A, and S1987A were introduced into an Atm�/�

background. ATM signaling and downstream events werenormal in these animals. At this stage, these differences be-tween human and murine ATM activation remain unre-solved. In this study, we compared ATM phosphorylation inhuman and murine cells, describing additional sites present inboth. We also confirmed ATM Ser367 and ATM Ser2996 auto-phosphorylation in human cells using phosphospecific anti-bodies against these sites and showed that they localize tosites of DNA DSB and play a role in regulating the S phasecheckpoint.

EXPERIMENTAL PROCEDURES

Cell Culture and Irradiation—Lymphoblastoid cell lineswere established from normal healthy individuals (C3ABR)and A-T patients (AT25ABR and AT1ABR) by Epstein-Barrvirus immortalization. AT1ABR is homozygous for the7636del9 mutation that produces nearly full-length ATM pro-

tein that is kinase-dead, and AT25ABR fails to express ATMprotein. The NBS patient cell line NBS03LA was obtainedfrom Professor R. Gatti, the ATLD cell line (ATLD 6) was akind gift of Professors D. Delia and L. Chessa, and the Rad50-deficient cell line (F239hTert) was kindly provided by Dr.Thilo Dork (10). A-T cell lines GM05823-hTert (39) andGM02052A-hTert (40) were kind gifts of Dr. Tej Pandita andProf. William K. Kaufmann, respectively. The lymphoblastoidcell lines were cultured in RPMI 1640 medium with 10% fetalcalf serum (15% FCS in the case of ATLD and Rad50-deficientcells), 100 units/ml penicillin, and 100 units/ml streptomycin(Invitrogen). All irradiations were performed at room tem-perature using a Gammacell 40 Exactor research irradiator(1 Gy/min; MDS Nordion, Ottawa, Canada).Antibodies—Affinity-purified ATM-specific sheep poly-

clonal antibodies were prepared as described earlier (41).ATM 2C1 monoclonal antibody (Abcam/GeneTex), phospho-Ser1981 ATM rabbit polyclonal (Rockland Immunochemicals),Mre11–2D7 monoclonal (GeneTex), Rad50–2C6 monoclonal(Upstate/Chemicon), Nbs1 polyclonal (GeneTex), and�H2AX (Ser(P)139) polyclonal (Novus Biologicals) antibodieswere used for immunoblotting.Phosphospecific Antibody Production—All peptides were

synthesized by Mimotopes (Clayton, Australia). Phosphopep-tides were conjugated to keyhole limpet hemocyanin, andphospho-Ser367 and phospho-Ser2996 ATM were prepared byimmunizing rabbits with a phospho-Ser367 peptide conjugateof keyhole limpet hemocyanin-SLEIpSQSYTT (where pS rep-resents phosphoserine) and with a phospho-Ser2996 peptideconjugate of keyhole limpet hemocyanin-CKRNLpSDIDQSF(Institute of Medical and Veterinary Science, Salisbury, Aus-tralia) followed by affinity purification on affinity columnsprepared by coupling of the corresponding phosphorylatedand non-phosphorylated ATM peptides to Sulfolink resin(Pierce).Cell Extracts, Immunoblotting, and ATM Immunoprecipita-

tion—Cell extracts were prepared by lysing cells in ATMkinase lysis buffer containing phosphatase and protease inhi-bitors (41). For immunoblotting, 50 �g of cell lysate was elec-trophoresed and transferred to nitrocellulose membranes(Amersham Biosciences). Membranes probed with antibodieswere visualized with the ECL kit (PerkinElmer Life Sciences).ATM protein was immunoprecipitated using sheep polyclonalATM antibodies and resolved on SDS gels (5% acrylamide) asdescribed (41). After transfer to nitrocellulose membranes,they were probed consecutively with phospho-Ser367 ATM,phospho-Ser1981 ATM, phospho-Ser1893 ATM, phospho-Ser2996 ATM, and ATM2C1 antibodies with stripping be-tween each step. Scaled up ATM immunoprecipitations wereused to obtain ATM protein for mass spectrometry analysis.A portion of the ATM immunoprecipitations was used for invitro autophosphorylation reactions with 20 �Ci of [�-32P]ATP to produce 32P-labeled ATM to facilitate analysis bymass spectrometry.Identification of ATM Phosphorylation Sites—Immunopre-

cipitated ATM from untreated or irradiated cells was sepa-rated by SDS-PAGE. Each ATM band (estimated at 0.5–1 �g)was destained, digested with trypsin, and extracted as de-

Autophosphorylation and ATM Activation

9108 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 11 • MARCH 18, 2011

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 3: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

scribed previously (42). Human or mouse ATM tryptic pep-tides from control or irradiated cells, from three SDS-PAGEbands, were enriched for phosphopeptides using titaniumdioxide as described (42). The phosphopeptide-enriched frac-tion or the non-binding non-phosphorylated peptides wereloaded onto an HPLC system (LC Packings Ultimate HPLCsystem, Dionex) for analysis by a QSTAR XL quadrupole-TOF MS (MDS Sciex) as described (42). One-third of thesample was analyzed in information-dependent acquisitionmode where the three most abundant precursors in a 1-s MSscan were selected for MS/MS sequencing in subsequent 2-sscans. Two-thirds of the sample was used to obtain improvedspectra of each of the detected phosphopeptides by fixing theprecursor at a particularm/z to obtain multiple 2-s scans ofthe same phosphopeptide across the elution peak, which werelater summed together.Quantitative Analysis Using Isotope Labeling—Human

ATM tryptic peptides were labeled with light or heavy stableisotopes for quantitative analyses prior to phosphopeptideenrichment. The dimethylation was done similarly to meth-ods described previously (43, 44). In a 100-�l final reactionvolume, 10 �l of 1 M triethylammonium bicarbonate and 10�l of 1% formaldehyde (0.1% final concentration) were addedto ATM tryptic peptides from untreated cells in 60 �l of wa-ter and vortexed for 5 min. 10 �l of 50 mM sodium cyano-borohydride was added, and the solution was vortexed foranother 10 min. ATM tryptic peptides from irradiated cellswere labeled in the same way, except formaldehyde with the“heavy” atoms, 2H and 13C, was used such that the net in-crease in mass compared with the “light” formaldehyde was atleast 6 Da (plus an additional 6 Da for each Lys present in thesequence). The light and heavy labeled peptides were mixedtogether before titanium dioxide phosphopeptide enrichment.The monoisotopic peak, �0.15 units ofm/z, for each light andheavy phosphopeptide signal was used to extract ion chro-matograms, and the peaks were integrated (44). The intensi-ties obtained were normalized for differences in the amountof protein in each of the pair of bands. This was done by com-paring the intensity between light and heavy non-phospho-peptide signals from the non-binding titanium dioxide chro-matography. Three non-phosphopeptides were used fornormalization of protein: 54YLNWDAVFR63, 452TPYVLR457,and 1647LVVNLLQLSK1656. These peptide identities wereconfirmed in the information-dependent acquisition modeanalysis (data not shown). Peptides were identified by export-ing the MS/MS spectra from Analyst QS 1.1 to Mascot 2.2using the Mascot.dll v16b23 script to search the IPI_human3.57 data base (76,541 entries). All MS/MS sequence datawere manually validated.Expression Vectors and Transfection—Construction of full-

length ATM cDNA expression vector pMAT1 was describedpreviously (45). ATM cDNA was placed under the control ofa heavy metal-inducible mehallothionein promoter in pMEP4vector (Invitrogen). Site-directed mutagenesis of the YFP-tagged ATM in pcDNA3 (provided by David Chen) was per-formed using the QuikChange mutagenesis kit essentially asdescribed by the manufacturer (Stratagene) to generate theS367A and S2996A autophosphorylation site mutants. Trans-

fection of hTERT-immortalized A-T fibroblasts was achievedusing the Amaxa Nucleofector system.Immunofluorescence Microscopy—Lymphoblastoid cells

were collected onto glass slides by cytocentrifugation andstained by indirect immunofluorescence. The sheep phospho-Ser1981 ATM antibody was used at 1:100 dilution, and �H2AXantibody (Cell Signaling Technology) was used at 1:300 dilu-tion. Anti-sheep and anti-rabbit secondary antibodies labeledwith Alexa dyes (Molecular Probes) were used to visualizebinding of primary antibodies. Coverslips were mounted withthe Vectashield antifade solution containing DAPI (MolecularProbes) as DNA counterstain. Images were collected using adigital camera attached to a Zeiss Axioskop (Zeiss) fluores-cence microscope. Initial digital image processing was per-formed using Zeiss software. Digital images were prepared forpublication using Adobe Photoshop CS software.Heavy Ion Irradiation and Microscopy—AG1522D human

fibroblasts were subjected to beams of 12C, 64Ni, 129Xe, 238U,and 197Au ions at the UNILAC facility at GSI, as describedpreviously (46). Beam parameters were as follows: linear en-ergy transfer (LET), 12C 170 keV/�m; energy on target (Et),9.8 MeV/unit; 64Ni LET, 3430 keV/�m; Et, 6.0 MeV/unit;129Xe LET, 8800 keV/�m; Et, 4.4 MeV/unit; 197Au LET,12,815 keV/�m; Et, 6.2 MeV/unit). Cells were fixed at the in-dicated time points after irradiation, and immunofluores-cence microscopy was performed with antibodies to Ser(P)367ATM, Ser(P)1981 ATM, Ser(P)2996 ATM, ATM, and �H2AXproteins. DNA was visualized with TOPRO3 or DAPI stain.GM05823-hTert or GM02052A-hTERT A-T cell lines weretransfected with wild type YFP-ATM or S367A, S1981A, andS2996A mutants using the Amaxa Nucleofector I system(Amaxa GmbH). HeLa cells were transfected with the sameATM plasmids using Lipofectamine 2000 reagent (Invitro-gen). Cells were cultured in a Petri dish on glass coverslips or25-�m PTFE foil (In Vitro Systems & Solutions, Gottingen,Germany). A-T cells expressing YFP-tagged ATM kinase wereirradiated with 238U ions (LET, 14350 keV/�m; Et, 3.8 MeV/unit), 197Au ions (LET, 12,815 keV/�m; Et, 6.2MeV/unit), and56Fe ions (LET, 3050 keV/�m; Et, 6.1 MeV/unit) and fixed,and immunofluorescence microscopy was performed withantibodies to �H2AX to visualize tracks of DNA damage. Re-cruitment of YFP-ATM proteins to the sites of ion traversalswas visualized using a beamline microscope as described (47).Monitoring Intra-S Phase Checkpoint—Stable cell lines

were established in the AT1ABR (A-T) lymphoblastoid cellline expressing wild type ATM (pMAT1), ATM S367A, andATM S2996A in pMEP4 vector (Invitrogen) (36). AT1ABRcell line alone or with the wild type or ATM S367A or ATMS2996A mutant form was induced with 5 �M cadmium chlo-ride for 16 h prior to the fork labeling. Induced replicatingcells were labeled consecutively with 25 �M chlorodeoxyuri-dine for 20 min, including a mock or 5-Gy treatment, andthen for a further 20 min with 100 �M iododeoxyuridine.DNA fibers were made following the approach of Parra andWindle (48). Briefly, 2 � 106 cells were mixed with lysis solu-tion (0.5% SDS, 200 mM Tris-HCl, pH 7.4, and 50 mM EDTA),and then after 10 min, slides were tilted to 20° to spread theDNA fibers. Slides were air-dried both pre- and postfixation

Autophosphorylation and ATM Activation

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9109

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 4: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

in methanol/acetic acid (3:1). Immunolabeling of the dena-tured DNA fibers (2.5 M HCl for 60 min) followed the ap-proach of Aten et al. (49), using first a rat monoclonal anti-BrdU (Abcam) to label chlorodeoxyuridine and a goat anti-ratAlexa 488-conjugated secondary antibody (MolecularProbes), followed by a mouse monoclonal anti-BrdU (BD Bio-sciences) to label iododeoxyuridine and donkey anti-mouseAlexa 594-conjugated secondary antibody (MolecularProbes). Fibers were imaged on a Zeiss AxioSkop microscopeusing Axiovision version 3.0 (Zeiss). DNA forks were scored(400–600) for each cell line for mock (unirradiated) and 5-Gytreatments. The percentage of new initiations was calculatedas ((new initiations/(continuing � new initiations)) � 100,and the S.D. value was calculated from three independentexperiments.Statistical Analysis—Statistical analysis was performed us-

ing Microsoft Excel. In experiments with YFP-ATM proteinrecruitment to sites of DNA damage, the error bars representthe 95% confidence interval of the mean. In S phase check-point assays, values were expressed as mean � S.D. In cellsurvival experiments, error bars represent S.E.

RESULTS

Identification of ATM Autophosphorylation Sites—We havepreviously identified three autophosphorylation sites in ATMin response to DNA DSB using tandemMS/MS (36). We em-ployed a more sensitive approach here to identify additionalautophosphorylation sites. Not surprisingly, we detectedphosphopeptides containing the three original sites (Ser(P)367,Ser(P)1893, and Ser(P)1981). A new phosphorylation site wasidentified on Thr1885 adjacent to the previously identifiedSer1893 site. Supplemental Fig. 1, A and B, shows the sequenc-ing of the phosphopeptide containing the novel Thr1885 site.We also confirmed the presence of Ser(P)2996 (supplementalFig. 1F), close to the kinase domain of ATM, which was re-cently detected by Daub et al. (50). A summary of the fivephosphorylation sites detected in human ATM appears inTable 1.We next quantified the change in phosphorylation at each

of these five detectable phosphorylation sites when humancells were irradiated. ATM immunoprecipitated from un-treated and irradiated cells was labeled by dimethylation withlight and heavy stable isotopes, respectively, as described un-der “Experimental Procedures.” Analysis of the signal inten-sity for the labeled peptides allowed relative quantification ofthe phosphopeptides. Thus, the relative level of phosphoryla-tion at each site was measured. The phosphopeptides con-taining either Thr1885 or Ser1893 were separable using reverse

phase chromatography by an �60-s difference in retentiontime, allowing separate quantification. Four of the five detect-able phosphosites, Ser367, Ser1893, Ser1981, and Ser2996, showedincreased phosphorylation after irradiation (Fig. 1A). It wasnot possible to describe the -fold change for most phospho-sites, because the phosphopeptide signal from the untreated

FIGURE 1. The relative abundance of ATM phosphosites after irradiationusing dimethyl labeling with stable isotopes. ATM tryptic peptides fromuntreated cells and irradiated (IR) cells were labeled with light and heavyisotopes and mixed before quantitative MS analysis. A, the relative changein the abundance of each specific phosphorylation site was determinedfrom the abundance of the phosphopeptides, corrected for the amount ofprotein in each band (see “Experimental Procedures”). The abundance inthe irradiated sample was normalized to 100% for each phosphorylationsite. Note that the monophosphorylated peptide that contained Thr1885

eluted �1 min before the peptide monophosphorylated at Ser1893, with thesame sequence and molecular mass, allowing separate quantification. Thedata are represented as the mean � S.E. (error bars), n � 3. B, the relativechange in the abundance of the non-phosphopeptides that harbor phos-phorylation sites was determined. The abundance in the untreated samplewas normalized to 100% for each peptide (ATM 363–375, 1883–1898,1974 –1992, and 2994 –3004, where amino acid residue 3003 is an Asp, not aGln). The data are represented as the mean � S.E., n � 4.

TABLE 1Identification of phosphorylation sites in human and mouse ATM by tandem MS/MS

Human MouseSequence Sites Phosphorylation sites Sequence Sites Phosphorylation sites

363SLEIpSQSYTTTQR375 1 Ser(P)367 363SVEIpSQSYVTQR374 1 Ser(P)3671883STpTPANLDSEpSEHFFR1898 2 Thr(P)1885 1889SApTPANSDSEpSENFLR1904 2 Thr(P)1891

Ser(P)1893 Ser(P)19991974SLAFEEGpSQSTTISSLSEK1992 1 Ser(P)1981 1980SPTFEEGpSQGTTISSLSEK1998 1 Ser(P)19872994NLpSDIDQSFNaK3004 1 Ser(P)2996 3003QSLpSDTDQSFNK3014 1 Ser(P)3006

a We detected human ATM with the Asp30033 Asn polymorphism (74).

Autophosphorylation and ATM Activation

9110 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 11 • MARCH 18, 2011

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 5: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

cells was below the threshold of detection, indicating a low orzero basal phosphorylation. However, there was a measurable6-fold increase in Ser(P)1981 and a 20-fold increase inSer(P)367. The proline-directed (Ser/Thr with Pro in the �1-position) phosphosite, Thr(P)1885, did not change signifi-cantly, indicating that it does not respond to DNA damage.Further insight was gained by measuring the relative changein the amount of each non-phosphopeptide that can harbor aphosphorylation site (Fig. 1B). An �60% drop in the level of1974SLAFEEGSQSTTISSLSEK1992 and 2994NLS-DIDQSFNK3004 indicated that Ser(P)1981 and Ser(P)2996 had asimilar response to DNA damage, and there was a largechange in the stoichiometry of phosphorylation at these sites.The change in stoichiometry at Ser(P)367 (measured as a de-crease in the amount of 363SLEISQSYTTTQR375) was modestin comparison. This peptide may also be influenced by thereported Ser(P)373 site (51). The decrease in the amount of1883STTPANLDSESEHFFR1898 was very slight. However,the inducible Ser(P)1893 shares this tryptic peptide with thenon-inducible Thr1885 (Fig. 1A). Overall, there are large in-creases in the level of phosphorylation at Ser367, Ser1893,Ser1981, and Ser2996 following irradiation of human cells butnot at the new ATM phosphosite Thr1885.We also determined the phosphorylation sites on murine

ATM in irradiated B16 cells. We confirmed that all of the de-tectable phosphorylation sites corresponding to those in hu-man ATM are also phosphorylated in mice (see Table 1 andsupplemental Fig. 1, C–F). The close correlation with humansites indicates similar usage of sites in both organisms forphosphoregulation. Other autophosphorylation sites havebeen detected on ATM in large scale proteomic studies (51,52) (see the PhosphoSitePlus Web site); however, none ofthese have been validated. All of the sites identified appear insupplemental Table 1.The Radiation-inducible Phosphorylation of Ser367 and

Ser2996—We previously generated phosphospecific antibodiesto investigate the importance of the Ser(P)1893 and Ser(P)1981ATM autophosphorylation sites (36). The same approach wasemployed here in generating antibodies against the ATMSer(P)367 site. The specificity of this antibody was demon-strated by detection of a peptide containing Ser(P)367 and noreactivity against the corresponding unphosphorylated pep-tide (supplemental Fig. 2A). Furthermore, no cross-reactivitywas observed against Ser1893- or Ser1981-phosphorylated pep-tides. The effect of increasing doses of radiation on ATMSer(P)367 was initially checked. As is evident from the resultsin Fig. 2A, phosphorylation is detected after 5- and 10-Gy ra-diation exposure. This was comparable with the level of in-duction of ATM Ser(P)1893. A more intense signal was ob-served with anti-ATM Ser(P)1981, but this could be due to thesensitivity of the antibody. After exposure of cells to 10 Gy,ATM Ser(P)367 was detected as early as 6 min postirradiation,comparable with the kinetics of appearance of ATMSer(P)1893 and Ser(P)1981 (Fig. 2B). In all cases, the level ofphosphorylation remained largely unchanged up to 24 hpostirradiation.We also raised phosphospecific antibodies against ATM

Ser2996 and demonstrated its specificity against phosphory-

lated peptide (supplemental Fig. 2B). As in the case of ATMser367 phosphorylation, we also observed time and dose de-pendence for ATM Ser(P)2996 after exposure to radiation (Fig.

FIGURE 2. ATM is autosphosphorylated on Ser367 in response to radia-tion exposure. A, effect of increasing radiation dose on ATM auto-phosphorylation on Ser367, Ser1893, and Ser1981. Cells were irradiated (IR)with the indicated doses and incubated for 1 h prior to preparation of ex-tracts, immunoprecipitation (IP) with anti-ATM antibodies, and immuno-blotting with phosphospecific and anti-ATM antibodies. AT25ABR, whichexpresses no ATM protein, was used as a control. B, time course of ATM au-tophosphorylation on Ser367, Ser1893, and Ser1981 in response to 10 Gy ofradiation. Conditions were the same as described in A.

FIGURE 3. ATM is autosphosphorylated on Ser2996 in response to radia-tion exposure. A, effect of increasing radiation dose on ATM auto-phosphorylation on Ser2996 and Ser1981. Cells were irradiated (IR) with theindicated doses and incubated for 1 h prior to preparation of extracts, im-munoprecipitation (IP) with anti-ATM antibodies, and immunoblotting withphosphospecific and anti-ATM antibodies. B, time course of ATM autophos-phorylation on Ser2996 and Ser1981 in response to 10 Gy of radiation. Condi-tions were the same as described in A.

Autophosphorylation and ATM Activation

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9111

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 6: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

3, A and B). To establish that Ser(P)367 and Ser(P)2996 wereATM-dependent, experiments were carried out with A-Tcells and with ATM inhibitors. Exposure of cells to the spe-cific ATM inhibitor KU55933 abolished radiation-inducedphosphorylation at Ser367 (Fig. 4A). This was also observedwith the more general PIKK inhibitor, wortmannin (Fig. 4B).Under these conditions, the Ser1893 and Ser1981 phosphoryla-tions were also inhibited. The results in Fig. 4C show thatthere is no induction of ATM Ser(P)367 in AT1ABR cells po-stirradiation. This was also the case for Ser(P)1893 andSer(P)1981. Again, as observed for ATM Ser(P)367, the ATM-specific inhibitor (KU55933) abolished ATM Ser2996 phos-phorylation (Fig. 5A), and phosphorylation at this site was notobserved in AT1ABR cells (Fig. 5B). Previous results reveal adependence on the MRN complex for ATM autophosphory-lation and activation (14, 19, 35). Here we employed NBS cellsto demonstrate a marked decrease in DNA damage-inducedATM Ser(P)367 (Fig. 6A). Again, Ser(P)1981 was also defective.This was also observed with ATLD4 cells hypomorphic forMre11 mutations (Fig. 6B). Similarly, we did not detect anyappreciable Ser367 phosphorylation in a cell line, derived froma patient with hypomorphic Rad50 mutations (10) (Fig. 6C).Overall, these results confirm radiation-induced phosphoryla-tion of ATM on Ser367 and Ser2996. They reveal that they aredependent on the presence of active ATM and also demon-strate a dependence of Ser367 phosphorylation on the MRNcomplex.

ATM Ser(P)367 and ATM Ser(P)2996 Localize to Sites of DNADamage—Exposure of cells to ionizing radiation leads to theaccumulation of DNA damage recognition and repair pro-teins at the sites of DNA damage in the form of nuclear foci(53, 54). It has also been shown that ATM Ser(P)1981 co-local-izes with �H2AX in nuclear foci postirradiation (14, 36). Weemployed two different qualities of radiation (low LET andhigh LET), both of which give rise to DNA DSB, to investigateATM Ser(P)367 localization at short times after radiation ex-posure. ATM Ser(P)367 foci were detected at 5 min after 3 Gyof �-rays, and these coincided with �H2AX foci indicative ofDNA DSB (Fig. 7A,middle). �H2AX foci also colocalized withATM Ser(P)1981 foci at the same time point (Fig. 7A, top), andthere was co-localization between ATM Ser(P)367 and ATMSer(P)1981 (bottom). We also employed co-localization with53BP1 to demonstrate the presence of ATM Ser(P)367 at sitesof DNA damage (supplemental Fig. 3). Quantitation ofSer(P)367 and Ser(P)1981 foci showed similar kinetics of accu-mulation (Fig. 7B).Biological imaging of charged particle tracks represents a

useful approach to investigate the association of DNA damageresponse proteins with chromatin (55–57). The results in Fig.8 show that fibroblasts exposed to xenon ions accumulateATM Ser(P)367 along tracks, and these coincide with �H2AXsignal. This was also the case for total ATM and ATMSer(P)1981 (supplemental Fig. 4, A and B). Similar results forSer(P)367 and Ser(P)1981 were obtained after irradiation withcarbon or nickel ions (supplemental Fig. 5, A and B). We have

FIGURE 4. ATM S367 autophosphorylation is dependent on ATM proteinkinase. A, the ATM-specific inhibitor KU55933 prevents ATM auto-phosphorylation on Ser367, Ser1893, and Ser1981 sites after exposure to 10 Gy.B, the effect of the PIKK inhibitor wortmannin on ATM autophosphorylationat the three sites. C, ATM autophosphorylation on Ser367 is not detected inAT1ABR cells. AT1ABR is an A-T cell line expressing a mutant, less stable,and kinase-dead form of ATM (7636del9). C3ABR is a control cell line. ATMwas immunoprecipitated from extracts prepared 1 h after exposure of cellsto 6 Gy, and immunoblotting was carried out as described in Fig. 2. IR, irra-diation; IP, immunoprecipitation.

FIGURE 5. ATM Ser2996 autophosphorylation is dependent on ATM pro-tein kinase. A, the ATM-specific inhibitor KU55933 prevents ATM autophos-phorylation on Ser2996 and Ser1981 sites after exposure to 10 Gy. B, ATM au-tophosphorylation on Ser29967 is not detected in AT1ABR cells, expressing amutant, less stable, and kinase-dead form of ATM. C, C3ABR is a control cellline. ATM was immunoprecipitated from extracts prepared 1 h after expo-sure of cells to 6 Gy, and immunoblotting was carried out as described inFig. 2. IR, irradiation; IP, immunoprecipitation.

Autophosphorylation and ATM Activation

9112 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 11 • MARCH 18, 2011

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 7: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

also observed weak accumulation of ATM Ser(P)367 and ATMSer(P)1981 in Nbs cells after irradiation with 64Ni ions (supple-mental Fig. 6, A and B), and pretreatment of normal fibro-blasts with ATM inhibitor Ku55933 abolishes ATM Ser(P)367signal along the tracks of DNA damage (data not shown).Similar experiments were carried out for localization of ATMSer(P)2996. Exposure of fibroblasts to gold ions led to accumu-lation of ATM Ser(P)2996 at sites of DNA damage as evi-denced by co-localization with �H2AX (Fig. 9). We next de-termined whether phosphosite mutants in ATM would affectits recruitment to chromatin in transfected A-T cells in re-sponse to DNA DSB, using a unique real-time beamline mi-croscopy system as described previously (56, 57). We initiallyestablished localization of YFP-tagged ATM proteins to thesites of DNA damage produced by uranium ions (Fig. 10).Cells were fixed 30 min after irradiation and immunostainedwith antibodies against �H2AX. We observed co-localizationof YFP-ATM wild type, YFP-S1981A ATM, and YFP-S367AATM with �H2AX along “tracks” of trajectories of the ura-nium particles (Fig. 10, A–C). This correlates well with theresults in Fig. 8 and supplemental Fig. 4 showing accumula-tion of ATM, ATM Ser(P)1981, and ATM Ser(P)367 alongtracks produced by heavy ions. No quantitative differences inrecruitment of YFP-ATM and ATM phosphorylation site mu-tants to the sites of DNA damage were evident.We then analyzed the kinetics of accumulation of different

phosphorylated forms of ATM at DSB sites induced by ura-nium ions. Both ATM and the S367A ATMmutant accumu-lated at sites of DNA damage with approximately the same

kinetics (Fig. 11). Both proteins show time constants of accu-mulation of �120–150 s, comparable with those reportedpreviously for Nbs1 and Mdc1 (58). It was not possible to usethis real-time beam microscopy system to analyze bindingover longer periods of time because the signal fades. To fur-ther analyze the recruitment kinetics of ATM, we transfectedHeLa cells with YFP-ATM wild type, S1981A, and S367A andsubjected them to irradiation to determine accumulation ofATM in the presence of endogenous ATMprotein. Similar tothe situation in A-T cells, we did not observe any significant dif-ferences in recruitment of wild type ATM and the twomutantforms of ATM to the sites of DNA damage (supplemental Fig. 7).Use of the ATM-YFP S2996Amutant also demonstrated thatthis form of ATMhad accumulation kinetics at sites of DNAdamage similar to that of wild type (supplemental Fig. 8).We have previously shown that the ATM S367Amutant was

defective in correcting radiosensitivity in A-T cells (36). Trans-fection of AT1ABR cells with ATM S2996A also failed to correctradiosensitivity (supplemental Fig. 9A). Under these conditions,the level of induction of the mutant ATMprotein was compara-ble with that of wild type (supplemental Fig. 9B). It is notable thatin ATM S2996A transfectants, the induced levels of protein de-creased with time of cells in culture (data not shown).We examined the effect of both the S367A and S2996A

mutations on the S phase checkpoint, a hallmark of the A-Tphenotype: radioresistant DNA synthesis (59). RadioresistantDNA synthesis was determined by consecutively labeling sitesof replication to distinguish new initiations from ongoingforks (49). Both the S367A and S2996A mutants were defec-

FIGURE 6. Mutations in members of the MRN complex reduce the efficiency of ATM autophosphorylation on Ser367. A, C3ABR (control) and NBS (Ni-jmegen breakage syndrome) cells were exposed to 3 Gy of radiation and incubated for 15 or 60 min prior to preparation of extracts. ATM was immunoprecipi-tated, and immunoblotting was carried out with the phosphospecific antibodies (top). Separated extracts were also immunoblotted with Nbs1, Mre11, and Rad50antibodies. B, autophosphorylation in control cells (C3ABR) and ATLD4 cells (deficient in Mre11) in response to 3 Gy of radiation. As in A, ATM was immunoprecipi-tated, followed by blotting with phosphospecific antibodies. Mre11, Nbs1, and Rad50 were detected by immunoblotting. C, NFFhTert (control) and F239hTert fi-broblasts (deficient in Rad50) were exposed to 3 Gy of radiation and incubated for 15 or 60 min prior to preparation of extracts. ATM was immunoprecipitated, andimmunoblotting was carried out with the phosphospecific antibodies (top). Extracts were also immunoblotted with Nbs1, Mre11, and Rad50 antibodies. Note thatRad50 mutation in F239hTert cells also affects levels of Mre11 and Nbs1 (10). *, defective protein in the specific cell line.

Autophosphorylation and ATM Activation

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9113

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 8: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

FIGURE 7. Localization of ATM Ser367 to sites of DNA damage. A, control fibroblasts were exposed to 3 Gy of radiation, fixed, and stained for �H2AX focias indicators of DNA DSB using immunofluorescence microscopy. The same cells were also stained for Ser(P)367 or Ser(P)1981 ATM foci, and co-localizationwas carried out using green and red Alexa dye labeling. Co-localization of Ser(P)367 and Ser(P)1981 ATM was also investigated. B, quantitative analysis ofSer(P)367 and Ser(P)1981 foci formation after 3 Gy of radiation. Control fibroblasts were exposed to 3 Gy of radiation, fixed at the indicated time points, andstained with Ser(P)367 and Ser(P)1981 antibodies. Ser(P)367 and Ser(P)1981 foci were analyzed by immunofluorescence microscopy using the DeltaVision de-convolution microscope system (Applied Biosciences). Semiautomated foci counting was performed using macros written for ImageJ software (NationalInstitutes of Health). A histogram of foci distribution per cell nucleus is shown. Combined foci counts from two independent experiments are shown.Counts were obtained from 50 nuclei, and nuclei with two or less foci were excluded from analysis.

Autophosphorylation and ATM Activation

9114 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 11 • MARCH 18, 2011

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 9: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

tive in correcting radioresistant DNA synthesis in A-T cells(Fig. 12). Under these conditions, the control cells showedmarked inhibition of DNA synthesis.

DISCUSSION

The four ATM autophosphorylation sites (Ser(P)367,Ser(P)1893, Ser(P)1981, and Ser(P)2996) described previously

using different approaches (14, 36, 50, 51) were detected, andthe response to DNA damage was quantified for each site. Inaddition, we have described here the existence of Thr(P)1885

on ATM and shown that it is not involved in the DNA dam-age response. The best described of the four radiation-sensi-tive sites is ATM Ser(P)1981, which has been associated withATMmonomerization and activation in the vicinity of DNA

FIGURE 8. Recruitment of ATM kinase phosphorylated at Ser367 to the sites of DNA damage induced by heavy ions. Normal AG1522D human fibro-blasts were subjected to a beam of xenon ions. Cells were fixed at the indicated time points after irradiation, and immunofluorescence microscopy was per-formed with antibodies to Ser(P)367 ATM and �H2AX proteins. DNA was visualized with TOPRO3 stain. Top panels, Ser(P)367 ATM staining; middle, �H2AXstaining; bottom, merged images with TOPRO3 staining.

FIGURE 9. Recruitment of ATM kinase phosphorylated at Ser2996 to the sites of DNA damage induced by heavy ions. Normal AG1522D human fibro-blasts were subjected to a beam of 197Au ions. Cells were fixed at the indicated time points after irradiation, and immunofluorescence microscopy was per-formed with antibodies to Ser(P)2996 ATM and �H2AX proteins. DNA was visualized with DAPI stain.

Autophosphorylation and ATM Activation

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9115

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 10: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

DSB (14, 15). Phosphorylation at this site is widely used as anindicator of ATM activation. The description of a second site,Ser(P)1893, and the availability of a phosphospecific antibodyagainst that site reveal that this phosphorylation can also beused to monitor activity (36). However, this site is not a con-sensus (SEpSE) ATM site and could point to the involvementof another protein kinase closely associated with ATM andinvolved in its activation. However, ATM has been shown tophosphorylate some substrates at non-consensus sites (60).Using a similar approach, we have shown here that theother two sites (Ser367 and Ser2996), are also phosphory-lated in response to DNA DSB. As outlined in supplemen-tal Table 1, five of the reported phosphorylation sites arelocated close to the N terminus of the molecule betweenamino acids 72 and 373, and several of these are in the sub-strate binding region (61). Phosphorylation at these sitesmay alter substrate binding or the interaction of ATM withother proteins.Three other sites are in the region of Ser1981, and the final

site, Ser2996, is located adjacent to the kinase domain in thePIKK regulatory domain. Mordes and Cortez (62) revealedthat TopBP1 interacts with ATRIP and facilitates its bindingto the PIKK regulatory domain of ATR for activation of thatprotein kinase. The PIKK regulatory domains of ATM, DNA-PKcs, and mTOR have also been shown to regulate the activi-ties of these PIKK family members (63). We have shown forthe first time phosphorylation of ATM on Ser2996 and demon-strated that it is particularly responsive to radiation-inducedphosphorylation, similar to Ser1981. It is also of interest thatthis site has been identified by Shiloh and co-workers (65).Perhaps not by coincidence, the Ser(P)2996 site is located close

to an acetylation site (Lys3016) (18). This site is acetylated inparallel to Ser(P)1981 in response to DNA DSB (64). Further-more, mutation in this site inhibited monomerization of theinactive ATM dimer and prevented induction of ATM activa-tion and phosphorylation of downstream substrates. We havealso provided evidence that ATM Ser2996 phosphorylationplays a functional role in radiation signaling. ATM post-trans-lational modifications may influence the interaction of activa-tors with this region of the protein, such as TopBP1 does forATR (62). Identification of such activators will assist furtherin unraveling the mechanism of ATM activation. WhereasATM activation by DNA DSB involves autophosphorylation

FIGURE 10. Recruitment of YFP-ATM, YFP-ATM S367A, and YFP-ATMS1981A to sites of DNA damage induced by heavy ions. YFP-ATM WT,YFP-S1981A ATM, and YFP-S367A ATM were transfected into the A-TGM05823hTert cell line, and cells were subjected to heavy ion irradiation(238U). Cells were fixed 30 min after irradiation and stained with antibodiesagainst �H2AX. DNA was visualized with TOPRO3 stain.

FIGURE 11. Time lapse imaging of YFP-ATM-expressing A-T cells beforeand after irradiation with 238U ions. A, an example of cells expressingYFP-ATM WT. Shown is live cell imaging prior to irradiation and 7 min po-stirradiation. Recruitment of YFP-ATM to the sites of damage produced byheavy ions is visualized in real time as white dots (foci) appearing in the nu-cleus. Images were recorded at 1-s intervals and binned afterward to 5-sintervals. The real-time animation is available upon request. B, kinetics ofWT ATM versus S367A ATM accumulation at the sites of DNA damage in-duced by uranium irradiation. The error bars represent the 95% confidenceinterval of the mean, n � 10.

Autophosphorylation and ATM Activation

9116 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 11 • MARCH 18, 2011

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 11: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

and formation of an active monomer, activation by oxidativestress occurs by formation of a dimeric form of ATM heldtogether by disulfide linkages (35). A critical site for disulfidebond formation is Cys2991. It will be of interest to determinehow the phosphorylation status of Ser2996 impacts on thisprocess. We did not observe here the recently described Cdk5(cyclin-dependent kinase 5) phosphorylation site on ATM,Ser794 (64). Ser794 is located in a small tryptic peptide andtherefore could be easily missed in our MS/MS analysis. Thissite was identified on ATM in postmitotic neurons by indirectapproaches. It is not clear if activation of Cdk5 kinase couldbe achieved in our cell culture model, lymphoblastoid cells.As observed previously for ATM Ser1981 and Ser1893, both

ATM Ser367 and ATM Ser2996 are rapidly phosphorylated inresponse to DNA DSB. In the case of ATM Ser(P)1981, this iscorrelated with dissociation of ATM into active monomers,which is prevented by mutation at this site (14). Mutation atthis site did not interfere with phosphorylation on ATMSer1893, but it reduced ATM-dependent phosphorylation ofp53, Nbs1, SMC1, and Chk2 in A-T cells complemented withATM S1981A cDNA (36). This mutant also failed to correctDNA repair, cell cycle checkpoint defects, and radiosensitivityin A-T cells. These results are at variance with those of Pel-legrini et al. (37), who demonstrated that Atm-dependentresponses are functional at the organism and cellular levels inmice expressing the corresponding mutant (S1987A) form ofAtm. In the case of the human ATM S367A mutant, results

similar to these with ATM S1981A were obtained. Mutationat this site was defective in supporting ATM-dependent sig-naling and did not correct downstream events, such as radio-sensitivity and cell cycle checkpoint defects (36). Although nomouse mutant expressing the ATM S367A mutation has beendescribed, a triple autophosphorylation site mutant (S367A/S1893A/S1981A) revealed that all of these sites were dispen-sable for ATM activation in vivo (38). They propose that thesedata are consistent with Atm autophosphorylation correlatingwith DNA damage-induced activation of the kinase but notbeing required for ATM function in vivo. The difference be-tween human and mouse studies remains unclear at present.However, ATM from these organisms have the same phos-phorylation sites, which implies similar usage of sites forphosphoregulation, although important differences may berevealed by future detailed studies of these sites.We also demonstrated here that ATM Ser(P)367 is localized

to nuclear foci and to sites of DNA damage induced by bothlow and high LET-ionizing radiation (Figs. 7 and 8). This be-havior is also observed for ATM Ser(P)1981 as reported earlier(14). However, we did not observe ATM Ser(P)2996 localizingto foci in response to �-radiation, whereas ATM Ser(P)2996could be detected at higher DSB density damage sites (Fig. 9).More recently, Berkovich et al. (15) showed that althoughwild type ATM was bound to PpoI-induced DNA DSB, nei-ther kinase-inactive ATM nor ATM S1981A was able to bind,suggesting that autophosphorylation is important for localiza-tion to sites of DNA damage. After this initial activation, theMRN complex is instrumental in recruiting ATM to the breaksite for full activation. In contrast, You et al. (16) showed thatATM activation is dispensable for initial recruitment to DNAfragments added to Xenopus egg extracts. This is substanti-ated by a recent report showing that ATM Ser(P)1981 is onlyrequired for sustained retention of ATM at sites of the breaks(35). This is in agreement with the findings of Lee and Paull(17, 22), who used an in vitro system to demonstrate largeincreases in ATM kinase activity without a concomitant in-crease in ATM autophosphorylation. These data suggest thatmutant ATMmay dissociate following the initial recruitment,whereas wild type is retained on chromatin to amplify signal-ing. An alternative model of ATM kinase activation suggests atwo-step mechanism (23), where the level of DNA damagedefines a threshold for MRN complex requirement in the acti-vation process. It is feasible that some discrepancies in theinterpretation of the behavior of ATM phosphorylation mu-tants can be attributed to different doses and types of DNAdamage employed.We have shown here that both the YFP-ATM S367A and

YFP-ATM S2996A mutants are recruited to sites of DNAdamage induced by heavy ions with kinetics similar to that ofwild type YFP-ATM, at least under the experimental condi-tions studied (Fig. 10 and supplemental Figs. 7 and 8). Simi-larly, the recruitment of YFP-ATM S1981A mutant did notseem to differ much from wild type YFP-ATM as can bejudged from immunofluorescence data. We previously pub-lished data demonstrating that an ATM S367A cDNA wasdefective in correcting radiation-induced signaling, DNA re-pair, radiosensitivity, and cell cycle checkpoint defects in A-T

FIGURE 12. ATM S367A and ATM S2996A are unable to correct radiore-sistant DNA synthesis seen in A-T cells. Wild type ATM, mutant ATMS367A, or ATM S2996A was transfected into A-T lymphoblastoid cells(AT1ABR) to investigate the role of Ser367/Ser2996 autophosphorylation sitesin radioresistant DNA synthesis. DNA forks were consecutively labeled withnucleotide analogs to follow the response of cells to ionizing radiation(5 Gy). The percentage of new initiations after irradiation/unirradiated newinitiations is shown. The percentage of new initiations was calculated as((new initiations/(continuing � new initiations)) � 100. At least 500 DNAforks were scored for each cell line for unirradiated and 5-Gy treatmentsfrom three independent experiments. Results are mean � S.D. (error bars),n � 3.

Autophosphorylation and ATM Activation

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9117

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 12: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

cells (36). Because the initial recruitment of YFP-S367A ATMto the sites of DNA damage induced by uranium ions wassimilar to that of wild type YFP-ATM, it seems that functionaleffects of Ser367 mutation can be explained by defective ATMsignaling to downstream substrates (at least at low doses ofirradiation). Indeed, moderate variation in kinase activity ofATM phosphorylation site mutants at the sites of DNA DSBcould influence the effective concentration of “activated”pools of ATM downstream targets (Nbs1, Chk2, p53, etc.).The transient nature of the interaction of ATM with its sub-strates (Chk2 and Nbs1) at sites of DNA DSB and the exist-ence of multiple pools of chromatin-bound ATM substrateshave been observed previously (58, 66). It is also impossible toexclude the possibility that the differences in expression sys-tems and levels of expressed ATM proteins (constitutive ex-pression via CMV promoter in pcDNA3 vector versus heavymetal-inducible metallothionein promoter in pMEP4 vector)could influence the outcome. However, we have been ableagain to detect the functional effect of S367A ATM andS2996A mutations in the S phase checkpoint assay (Fig. 12)and their inability to correct radioresistant DNA synthesis inA-T cells. One intriguing possibility is that Ser367 and Ser2996

phosphorylations might play a distinct role in a specific stageof the cell cycle. From our analysis of Ser367 and Ser2996 ATMphosphorylation, it is clear that phosphospecific Ser367 andSer2996 ATM antibodies can be used as additional and reliablemarkers of ATM activation.Curiously, tethering Cherry-LacR-ATM protein, with a

truncation of the N terminus (residues 1300–3060) lackingthe Ser367 phosphorylation site, to chromatin seems to be suf-ficient to activate DNA damage signaling (67), even in theabsence of DNA damage. In addition, manipulating the N-ter-minal area of ATM by adding the ATRIP-binding N-terminalregion (residues 1–388) of ATR kinase (68) created ATM pro-tein that was capable of binding ATRIP and localizing it tostalled replication forks. Although this chimeric ATM proteinretained normal kinase activity in vitro and was phosphory-lated on Ser1981, it was nevertheless deficient in checkpointsignaling and did not complement the cellular phenotype ofA-T cells, probably due to deficiency in binding to Nbs1. Fer-nandez et al. (69) showed that ATM kinase with mutations inthe N-terminal interaction domain (amino acids 91–97) hadkinase activity and was autophosphorylated on Ser1981 but didnot correct radiosensitivity in A-T cells. The N-terminal “sub-strate binding region” of ATM described previously (61, 70,71) was also identified as a “chromatin association” domain(amino acids 5–224) (72). Importantly, Nbs1 interaction re-gion is also mapped to the N-terminal HEAT repeats of ATM(73). All of these studies underscore the importance of regula-tory phosphorylation sites in the N terminus of ATM andunresolved issues related to the role of N-terminal domains ofATM in its activation as well as potential critical role of C-ter-minal phosphorylation sites in the initiation of DNA damagesignaling. Both the N-terminal and C-terminal ATM phos-phorylation sites described here will assist in elucidating theprecise mechanism of ATM kinase activation after DNAdamage.

Finally, it is clear from the data presented here (Table 1 andsupplemental Table 1) that ATM kinase is capable of phos-phorylation on multiple sites in both human and mouse cells.It is also evident that complex arrays of post-translationalmodifications are involved in regulation of ATM kinase acti-vation (e.g. see Ref. 13; see the PhosphoSitePlus Web site).The combinations of post-translational modifications mightallow precise cell- and damage signal-specific ATM kinaseactivation. Future studies aimed at detailed mapping of ATMmodification sites in different model organisms, especially intissues of neuronal origin, will undoubtedly lead to a greaterunderstanding of ATM kinase activation.

Acknowledgments—We thank Aine Farell and Gudrun Becker forexcellent technical assistance.

REFERENCES1. Bredemeyer, A. L., Sharma, G. G., Huang, C. Y., Helmink, B. A., Walker,

L. M., Khor, K. C., Nuskey, B., Sullivan, K. E., Pandita, T. K., Bassing,C. H., and Sleckman, B. P. (2006) Nature 442, 466–470

2. McKinnon, P. J., and Caldecott, K. W. (2007) Annu. Rev. GenomicsHum. Genet. 8, 37–55

3. Shrivastav, M., De Haro, L. P., and Nickoloff, J. A. (2008) Cell Res. 18,134–147

4. Stracker, T. H., Theunissen, J. W., Morales, M., and Petrini, J. H. (2004)DNA Repair 3, 845–854

5. Helmink, B. A., Bredemeyer, A. L., Lee, B. S., Huang, C. Y., Sharma,G. G., Walker, L. M., Bednarski, J. J., Lee, W. L., Pandita, T. K., Bassing,C. H., and Sleckman, B. P. (2009) J. Exp. Med. 206, 669–679

6. Rass, E., Grabarz, A., Plo, I., Gautier, J., Bertrand, P., and Lopez, B. S.(2009) Nat. Struct. Mol. Biol. 16, 819–824

7. Xie, A., Kwok, A., and Scully, R. (2009) Nat. Struct. Mol. Biol. 16,814–818

8. Carney, J. P., Maser, R. S., Olivares, H., Davis, E. M., Le Beau, M., Yates,J. R., 3rd, Hays, L., Morgan, W. F., and Petrini, J. H. (1998) Cell 93,477–486

9. Stewart, G. S., Maser, R. S., Stankovic, T., Bressan, D. A., Kaplan, M. I.,Jaspers, N. G., Raams, A., Byrd, P. J., Petrini, J. H., and Taylor, A. M.(1999) Cell 99, 577–587

10. Waltes, R., Kalb, R., Gatei, M., Kijas, A. W., Stumm, M., Sobeck, A.,Wieland, B., Varon, R., Lerenthal, Y., Lavin, M. F., Schindler, D., andDork, T. (2009) Am. J. Hum. Genet. 84, 605–616

11. Savitsky, K., Bar-Shira, A., Gilad, S., Rotman, G., Ziv, Y., Vanagaite, L.,Tagle, D. A., Smith, S., Uziel, T., Sfez, S., Ashkenazi, M., Pecker, I., Fryd-man, M., Harnik, R., Patanjali, S. R., Simmons, A., Clines, G. A., Sartiel,A., Gatti, R. A., Chessa, L., Sanal, O., Lavin, M. F., Jaspers, N. G., Taylor,A. M., Arlett, C. F., Miki, T., Weissman, S. M., Lovett, M., Collins, F. S.,and Shiloh, Y. (1995) Science 268, 1749–1753

12. Lavin, M. F., and Shiloh, Y. (1997) Annu. Rev. Immunol. 15, 177–20213. Lavin, M. F. (2008) Nat. Rev. Mol. Cell Biol. 9, 759–76914. Bakkenist, C. J., and Kastan, M. B. (2003) Nature 421, 499–50615. Berkovich, E., Monnat, R. J., Jr., and Kastan, M. B. (2007) Nat. Cell Biol.

9, 683–69016. You, Z., Bailis, J. M., Johnson, S. A., Dilworth, S. M., and Hunter, T.

(2007) Nat. Cell Biol. 9, 1311–131817. Lee, J. H., and Paull, T. T. (2005) Science 308, 551–55418. Sun, Y., Xu, Y., Roy, K., and Price, B. D. (2007)Mol. Cell. Biol. 27,

8502–850919. Sun, Y., Jiang, X., Xu, Y., Ayrapetov, M. K., Moreau, L. A., Whetstine,

J. R., and Price, B. D. (2009) Nat. Cell Biol. 11, 1376–138220. Uziel, T., Lerenthal, Y., Moyal, L., Andegeko, Y., Mittelman, L., and Shi-

loh, Y. (2003) EMBO J. 22, 5612–562121. Cerosaletti, K., and Concannon, P. (2004) J. Biol. Chem. 279,

38813–38819

Autophosphorylation and ATM Activation

9118 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 11 • MARCH 18, 2011

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 13: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

22. Lee, J. H., and Paull, T. T. (2004) Science 304, 93–9623. Dupre, A., Boyer-Chatenet, L., and Gautier, J. (2006) Nat. Struct. Mol.

Biol. 13, 451–45724. Kitagawa, R., Bakkenist, C. J., McKinnon, P. J., and Kastan, M. B. (2004)

Genes Dev. 18, 1423–143825. Kitagawa, R., and Kastan, M. B. (2005) Cold Spring Harb. Symp. Quant.

Biol. 70, 99–10926. van Attikum, H., and Gasser, S. M. (2009) Trends Cell Biol. 19, 207–21727. Cook, P. J., Ju, B. G., Telese, F., Wang, X., Glass, C. K., and Rosenfeld,

M. G. (2009) Nature 458, 591–59628. Chapman, J. R., and Jackson, S. P. (2008) EMBO Rep. 9, 795–80129. Melander, F., Bekker-Jensen, S., Falck, J., Bartek, J., Mailand, N., and Lu-

kas, J. (2008) J. Cell Biol. 181, 213–22630. Huen, M. S., Grant, R., Manke, I., Minn, K., Yu, X., Yaffe, M. B., and

Chen, J. (2007) Cell 131, 901–91431. Kolas, N. K., Chapman, J. R., Nakada, S., Ylanko, J., Chahwan, R.,

Sweeney, F. D., Panier, S., Mendez, M., Wildenhain, J., Thomson, T. M.,Pelletier, L., Jackson, S. P., and Durocher, D. (2007) Science 318,1637–1640

32. Mailand, N., Bekker-Jensen, S., Faustrup, H., Melander, F., Bartek, J.,Lukas, C., and Lukas, J. (2007) Cell 131, 887–900

33. Doil, C., Mailand, N., Bekker-Jensen, S., Menard, P., Larsen, D. H., Pep-perkok, R., Ellenberg, J., Panier, S., Durocher, D., Bartek, J., Lukas, J., andLukas, C. (2009) Cell 136, 435–446

34. Stewart, G. S. (2009) Cell Cycle 8, 1532–153835. Guo, Z., Kozlov, S., Lavin, M. F., Person, M. D., and Paull, T. T. (2010)

Science 330, 517–52136. Kozlov, S. V., Graham, M. E., Peng, C., Chen, P., Robinson, P. J., and

Lavin, M. F. (2006) EMBO J. 25, 3504–351437. Pellegrini, M., Celeste, A., Difilippantonio, S., Guo, R., Wang, W.,

Feigenbaum, L., and Nussenzweig, A. (2006) Nature 443, 222–22538. Daniel, J. A., Pellegrini, M., Lee, J. H., Paull, T. T., Feigenbaum, L., and

Nussenzweig, A. (2008) J. Cell Biol. 183, 777–78339. Wood, L. D., Halvorsen, T. L., Dhar, S., Baur, J. A., Pandita, R. K.,

Wright, W. E., Hande, M. P., Calaf, G., Hei, T. K., Levine, F., Shay, J. W.,Wang, J. J., and Pandita, T. K. (2001) Oncogene 20, 278–288

40. Heffernan, T. P., Simpson, D. A., Frank, A. R., Heinloth, A. N., Paules,R. S., Cordeiro-Stone, M., and Kaufmann, W. K. (2002)Mol. Cell. Biol.22, 8552–8561

41. Kozlov, S., Gueven, N., Keating, K., Ramsay, J., and Lavin, M. F. (2003)J. Biol. Chem. 278, 9309–9317

42. Chan, L. S., Hansra, G., Robinson, P. J., and Graham, M. E. (2010) J. Pro-teome Res. 9, 4028–4037

43. Hsu, J. L., Huang, S. Y., Chow, N. H., and Chen, S. H. (2003) Anal.Chem. 75, 6843–6852

44. Lemeer, S., Jopling, C., Gouw, J., Mohammed, S., Heck, A. J., Slijper, M.,and den Hertog, J. (2008)Mol. Cell Proteomics 7, 2176–2187

45. Zhang, N., Chen, P., Khanna, K. K., Scott, S., Gatei, M., Kozlov, S., Wat-ters, D., Spring, K., Yen, T., and Lavin, M. F. (1997) Proc. Natl. Acad. Sci.U.S.A. 94, 8021–8026

46. Jakob, B., Scholz, M., and Taucher-Scholz, G. (2000) Radiat. Res. 154,398–405

47. Jakob, B., Rudolph, J. H., Gueven, N., Lavin, M. F., and Taucher-Scholz,G. (2005) Radiat. Res. 163, 681–690

48. Parra, I., and Windle, B. (1993) Nat. Genet. 5, 17–2149. Aten, J. A., Bakker, P. J., Stap, J., Boschman, G. A., and Veenhof, C. H.

(1992) Histochem. J. 24, 251–25950. Daub, H., Olsen, J. V., Bairlein, M., Gnad, F., Oppermann, F. S., Korner,

R., Greff, Z., Keri, G., Stemmann, O., and Mann, M. (2008)Mol. Cell 31,438–448

51. Matsuoka, S., Ballif, B. A., Smogorzewska, A., McDonald, E. R., 3rd, Hu-rov, K. E., Luo, J., Bakalarski, C. E., Zhao, Z., Solimini, N., Lerenthal, Y.,Shiloh, Y., Gygi, S. P., and Elledge, S. J. (2007) Science 316, 1160–1166

52. Beausoleil, S. A., Jedrychowski, M., Schwartz, D., Elias, J. E., Villen, J., Li,J., Cohn, M. A., Cantley, L. C., and Gygi, S. P. (2004) Proc. Natl. Acad.Sci. U.S.A. 101, 12130–12135

53. Maser, R. S., Monsen, K. J., Nelms, B. E., and Petrini, J. H. (1997)Mol.Cell. Biol. 17, 6087–6096

54. Paull, T. T., Rogakou, E. P., Yamazaki, V., Kirchgessner, C. U., Gellert,M., and Bonner, W. M. (2000) Curr. Biol. 10, 886–895

55. Lukas, C., Bartek, J., and Lukas, J. (2005) Chromosoma 114, 146–15456. Jakob, B., Scholz, M., and Taucher-Scholz, G. (2003) Radiat Res. 159,

676–68457. Taucher-Scholz, G., and Jakob, B. (2007) Ion Irradiation as a Tool to

Reveal the Spatiotemporal Dynamics of DNA Damage Response Pro-cesses in Genome Dynamics and Stability, Vol. 1 (Lankenau, D.-H., ed)pp. 453–478, Springer-Verlag, Berlin

58. Lukas, C., Melander, F., Stucki, M., Falck, J., Bekker-Jensen, S., Goldberg,M., Lerenthal, Y., Jackson, S. P., Bartek, J., and Lukas, J. (2004) EMBO J.23, 2674–2683

59. Houldsworth, J., and Lavin, M. F. (1983) Biochem. Int. 6, 349–35660. Cortez, D., Wang, Y., Qin, J., and Elledge, S. J. (1999) Science 286,

1162–116661. Khanna, K. K., Keating, K. E., Kozlov, S., Scott, S., Gatei, M., Hobson, K.,

Taya, Y., Gabrielli, B., Chan, D., Lees-Miller, S. P., and Lavin, M. F.(1998) Nat. Genet. 20, 398–400

62. Mordes, D. A., and Cortez, D. (2008) Cell Cycle 7, 2809–281263. Lovejoy, C. A., and Cortez, D. (2009) DNA Repair 8, 1004–100864. Tian, B., Yang, Q., and Mao, Z. (2009) Nat. Cell Biol. 11, 211–21865. Bensimon, A., Schmidt, A., Ziv, Y., Elkon, R., Wang, S. Y., Chen, D. J.,

Aebersold, R., and Shiloh, Y. (2010) Sci. Signal. 3, rs366. So, S., Davis, A. J., and Chen, D. J. (2009) J. Cell Biol. 187, 977–99067. Lukas, C., Falck, J., Bartkova, J., Bartek, J., and Lukas, J. (2003) Nat. Cell

Biol. 5, 255–26068. Soutoglou, E., and Misteli, T. (2008) Science 320, 1507–151069. Chen, X., Zhao, R., Glick, G. G., and Cortez, D. (2007) Exp. Cell Res.

313, 1667–167470. Fernandes, N., Sun, Y., Chen, S., Paul, P., Shaw, R. J., Cantley, L. C., and

Price, B. D. (2005) J. Biol. Chem. 280, 15158–15186471. Beamish, H., Kedar, P., Kaneko, H., Chen, P., Fukao, T., Peng, C., Ber-

esten, S., Gueven, N., Purdie, D., Lees-Miller, S., Ellis, N., Kondo, N., andLavin, M. F. (2002) J. Biol. Chem. 277, 30515–30523

72. Young, D. B., Jonnalagadda, J., Gatei, M., Jans, D. A., Meyn, S., andKhanna, K. K. (2005) J. Biol. Chem. 280, 27587–27594

73. Falck, J., Coates, J., and Jackson, S. P. (2005) Nature 434, 605–61174. Rasio, D., Negrini, M., and Croce, C. M. (1995) Cancer Res. 55,

6053–6057

Autophosphorylation and ATM Activation

MARCH 18, 2011 • VOLUME 286 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 9119

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from

Page 14: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

SUPPLEMENTARY FIGURE LEGENDS Supplementary Fig. 1. Tandem mass spectra of ATM phosphopeptides reveal new human and mouse phosphorylation sites. Titanium dioxide phosphopeptide enriched tryptic digests of human or mouse ATM were analysed by LC-MS/MS in multiple ion monitoring mode (see Methods). Spectra across the eluting phosphopeptide peaks were combined to improve the signal to noise ratio. The signal intensity for each ion is a measure of the average absolute counts for each ion. A. Mass spectrum of phosphorylated hATM 1883-1898. The triply charged precursor ion was at m/z 639.9. The phospho- T1885 was observed between the b2 and b3 ions and the y13 and y14 ions. B. Mass spectrum of phosphorylated mATM 1889-1904. The doubly charged precursor ion was at m/z 902.9. The spectrum was a mixture of two mono-phosphopeptides. Phospho-T1891 was observed between the b2 and b3 ions and the y13 and y14 ions. Ions specific to this phosphopeptide are shown in red. Phospho- S1899 was observed between the y5 and y6 ions. Ions specific to this phosphopeptide are shown in blue. These latter ions were much more abundant than the ions for the phospho-T1891 sequence. Therefore, it is likely that phosphorylation at S1899 was more abundant than phosphorylation at T1891. C. Mass spectrum of phosphorylated mATM 363-374. The doubly charged precursor ion was at m/z 738.9. Phospho-S367 was observed between the y7 and y8 ions. D. Mass spectrum of phosphorylated mATM 1980-1988. The doubly charged precursor ion was at m/z 1033.0. Phospho- T1987 was observed between the y11 and y12 ions. E. Mass spectrum of phosphorylated mATM 3003-3014. The doubly charged precursor ion was at m/z 725.3. Phospho-T3006 was deduced as the only possible phosphorylation site between the y7 and y9 ions or b2 and b4 ions. F. Mass spectrum of phosphorylated hATM 2994-3004 with the D3003→N polymorphism. The doubly charged precursor ion was at m/z 680.8. Phospho-S2996, found previously by Daub et al (49), was observed between the y8 and y9 ions. Supplementary Fig. 2. Specificity of the pS367-ATM and pS2996-ATM antibodies. A.The phosphopeptides containing pS367, pS1893 and pS1981 and unphosphorylated peptides were blotted onto nitrocellulose membranes (Hybond ECL, 0.2μM, Amersham Biosciences), at 250ng and 500ng. The membranes were incubated with affinity-purified rabbit pS367-ATM antibody, antibody specifically recognised the corresponding phosphopeptide. Antibody did not recognise the unphosphorylated peptides. B. The phosphopeptides containing pS2996 and pS1981 and unphosphorylated peptides were spotted onto nitrocellulose membranes (Hybond ECL, 0.2μM, Amersham Biosciences), at 250ng and 500ng. The membranes were incubated with affinity-purified rabbit pS2996-ATM antibody, antibody specifically recognised the corresponding phosphopeptide. Antibody did not recognise the unphosphorylated peptides. Supplementary Fig. 3. Localization of ATM pS367 to sites of DNA damage. Normal human diploid fibroblasts (HE49) were exposed to 1Gy of X-irradiation, fixed after 1 hr incubation and stained for pS367, pS1981, γH2AX and 53BP1. DNA damage–induced foci were visualized by immunofluorescence microscopy. Co-localization of pS367 and pS1981 ATM was evident, as well as co-localization of pS367 ATM with γH2AX and 53BP1. Supplementary Fig. 4. Recruitment of ATM kinase phosphorylated at S1981 to the sites of DNA damage induced by heavy ions. AG1522D human fibroblasts were subjected to the beam of Xe ions (LET of 8800 keV/μm). Cells were fixed at the indicated time points after irradiation and immunofluorescence microscopy was performed with antibodies to A ATM and γ-H2AX proteins and B ATM pS1981 and γH2AX. DNA was visualized with TOPRO3 stain. Supplementary Fig. 5. Recruitment of ATM kinase phosphorylated at S367 to the sites of DNA damage induced by irradiation with 12C and 64Ni ions. A. AG1522D human fibroblasts were subjected to a beam of 12C ions (LET 170 keV/ μm). Cells were fixed 10 min after irradiation and immunofluorescence microscopy was performed with

Page 15: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

antibodies to pS367 ATM and pS1981 ATM proteins. DNA was visualized with TOPRO3 stain. B. AG1522D human fibroblasts were subjected to a beam of 64Ni ions (LET 3430 keV/ μm), fixed 1 h after irradiation and processed as described in A. Supplementary Fig. 6. Recruitment of ATM kinase phosphorylated at S367 to the sites of DNA damage induced by irradiation with 64Ni ions in Nbs cells. AG1522D and Nbs human fibroblasts were subjected to a beam of 64Ni ions (LET 3430 keV/ μm). Cells were fixed 1 hr after irradiation and immunofluorescence microscopy was peformed with antibodies to A. pS1981 ATM and B. pS367 ATM proteins. DNA was visualized with TOPRO3 stain. Supplementary Figure 7 Time-lapse imaging of YFP-ATM expressing HeLa cells before and after irradiation with 56Fe ions. A. Living cells were imaged before irradiation and first 7 min after irradiation with iron ions. An example of cells expressing YFP-ATMwt is shown. Recruitment of YFP-ATM to the sites of damage produced by heavy ions is visualized in real time as white dots (foci) appearing in the nucleus. Images were recorded at 20 s intervals. B. Kinetics of ATM vs S367A ATM accumulation at the sites of DNA damage induced by iron ions irradiation. n=6 for YFP-ATM wt, n=4 for YFP-S367A and YFP-S1981. Supplementary Figure 8 Time-lapse imaging of YFP-ATM S2996A expressing A-T cells before and after irradiation with 197Au ions. Kinetics of wt ATM vs S2996A ATM accumulation at the sites of DNA damage induced by irradiation with gold ions. The error bars represent the 95% confidence interval of the mean; n= 6 for YFP-ATM wt and n= 12 for YFP-ATM S2996A. Supplementary Figure 9. ATM autophosphorylation site mutants S1981A and S2996A fail to correct radiosensitivity of A-T cells. A. Survival of control (C35ABR), AT1ABR and AT1ABR cells transfected with pMAT1, S2996A and S1981A mutant forms of ATM. Cells were induced with CdCl2 for 16 hrs, exposed to radiation over the range 0-4 Gy and incubated for 3 days, prior to cell counting and determination of cell survival. Survival is expressed as a percentage of radiated/unirradiated. An average of triplicate experiments is shown for each point. Error bars represent SEM. B. Expression levels of ATM wt (pMAT1), S2996A and S1981A ATM proteins after 16 hr induction with 5 µM CdCl2.

Page 16: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Supplementary Table 1: Summary of Phosphorylation sites identified in ATM

Sequence

Number of Sites

Phosphorylation Site

KEpTECLRIAKPNVSApSpTQASR

3

pT72 E1

pS85T1

pT86Q1 SLEIpSQ 2 SYTTpTQR

pS367Q1,2

pT373Q1

KKpSP

NKI 1 pS794P5

pSTpTPANLDSEpSE 3 HFFR pS1883T5 pT1885P6

pS1893E2

SLAFEEGpSQ 2 STpTISSLSEK pS1981Q1,2,3,4 pT1985I1

GHSFWEIpYKMTTDPM 1 pY1753K5

TTDPMLApYLQPFRTS 1 pY1763L5 LESVYSLpYPTLSRLQ 1 PY2170P5

NLpSD 1 IDQSFDK pS2996D5,6,7,8

1 Matsuoka et al (51); 2 Kozlov et al (36); 3 Bakkenist and Kastan 2003 (14); 4 Beausoleil et al (52); 5 http://www.phosphosite.org 6 Reported here 7 Shiloh et al (in press) 8 Daub et al (50) Underlined phosphorylation sites were manually confirmed.

Page 17: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

A1883-STpTPANLDSESEHFFR-1898

b3b2

y14 y3y8y11y12y13 y6y9

13 [M+3H]3+13

639.93

[y14-98]3+

544.58

ensi

ty (

coun

ts)

y132+

774.87

b2

189.09b3

370.11

y143+

577.24

[y14-98]2+

816.39

y122+

726.32

y112+

690.81

y142+

865.36

y6

822.40

y8

1038.48y3

469.25y9

1153.50

Abs

olut

e In

te

B

m/z100 200 300 400 500 600 700 800 900 1000 1100 12000

1889-SApTPANSDSESENFLR-1904b4b2 b3

1889-SATPANSDSEpSENFLR-1904

1889 SApTPANSDSESENFLR 1904

b3

y14 y2

b2

y14 y13

y13 y3y4y5

y10

y1

y2y3y4y5 y1y6y11 y8

12 y132+

773.33

[y13-98]2+

724.34

y1

175 12

b2

159.08

b3-98242.12 b4-98

339.16

y132+

733.37

y (c

ount

s)

175.12y14

2+

823.87

y3

435.28

98

y8-98963.55

y5

678.40

y11-981279.54

y8

1061.49

b3

260.13

y13-981447.69

y4

549.34

y2

288.21

y6

845.41

Abs

olut

e In

tens

ity

100 300 500 700 900 1100 1300 15000

y10-981165.56 y13

1545.60

m/z

Page 18: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

C

363-SVEIpSQSYVTQR-374b4b2 b3

3 y3

404.23

a2

159.11[ 98]2+ 2

y10 y2y8 y3y4y5 y1y7 y6y9

unts

)

b2

187.11

[y9-98]2+

532.31

y4

503.32

[M-98+H]2+

689.86

[y10-98]2+

596.84b3

316.15

y5

666.37

y6

753.40

b4

429.25y8-98

950.50

y2

303.18

y1

175.11

olut

e In

tens

ity (

cou

100 200 300 400 500 600 700 800 900 10000

y7

881.49Abs

o

m/z

D

8 y3

1980-SPTFEEGpSQGTTISSLSEK-1998y13 y2

b8b2

y10 y3y4y8 y1

b3 b4 b5 b6

y6y7y11y14 y12

363.20

y6

650.36

b5

562.28

b4

b3

286 15 b8-98

y2

276.16

[M-98+H]2+

984.03

sity

(co

unts

)

y1

147.11

y4

476.28b2

185.09

4

433.22

b6

691.34

286.15

y10

1022.59

8

817.35

y7

763.41

y11

1150.61

y13-981276.60

y12

1317.65y8

864.49

[M+H]2+

1033.00

y14-981405.67

Abs

olut

e In

ten

100 200 300 400 500 600 700 800 900 1000 1100 1200 1300 14000

m/z

Page 19: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

E

3003-QSLpSDTDQSFNK-3014b2 b4

2.2

[M+H]2+

725.31

y4

y6

738.36

[M-98+H]2+

667.32

y2y9 y3y4 y1y6y7

nts)

y4

495.27

y3

408.23

[y9-98]2+

658.81

y2

261.16y1

147.10

b2

216.09

b4

496.21y7

839.44

olut

e In

tens

ity (

cou

100 200 300 400 500 600 700 800 900 10000.0

Abs

o

m/z

2994-NLpSDIDQSFNK-3004y2

b2

y9 y3y5 y1

b3

y6y8

Fb4

y4y7

ty (

coun

ts)

52 a2

200.14

[M-98+H]+

631.80

y5

623 30

Abs

olut

e In

tens

it

b2

228.13 b3-98297.15

623.30y2

261.16

y6

738.34y4

495.26

y3

408.22

[M+H]+

680.78

y9-981035.51

y8

966 48y9

b4

510.17

y1

147.11y7

b3

297.15

m/z100 200 300 400 500 600 700 800 900 1000 1100 12000

966.481133.48

y7

851.45

Page 20: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Anti-pS367-ATM

Supplementary Figure 2

500ng

250ng

250ng

500ng

500ng

250ng

A

B

250ng

500ng

500ng

250ng

Anti-pS2996-ATM

pS-peptide Non-pS-peptide

S2996-peptide

S1981-peptide

Page 21: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Supplementary Figure 3

S367 P S1981 PMerge S367-P S1981-PMerge

S367-P H2AX-PMerge

S367-P 53BP1Mergeg

Page 22: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Supplementary Figure 4

A

ATM

-H2AX

Merge

(ATM + γH2AX+ TOPRO3)

5min 1h 24h

Page 23: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

B

ATM pS1981

-H2AX

Merge

(ATM pS1981 + γH2AX+ TOPRO3)

5min 1h 24h

Page 24: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

scalebar: 5 µm

scalebar: 5 µm

Supplementary Figure 5

pS367-ATMpS1981-ATM DNAMerge

pS367-ATMpS1981-ATMDNAMerge

A

B

Page 25: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Supplementary Figure 6

pS1981-ATM DNAMerge

A

Control

scalebar 5 µm

pS1981-ATM DNApMerge

NBS

scalebar 5 µm

Page 26: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

B

pS367-ATM DNAMerge

Control

scalebar 5 µmpS367-ATM

pS367-ATM DNApMerge

NBS

scalebar 5 µmpS367-ATM

Page 27: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Supplementary Figure 7

wtA

scalebar: 5µm

pre 60s 120s

scalebar: 5µm

180s 240s 300s

360s 420s 480s

ATM-YFP recruitment in Hela cells

1.2

y

B

0.2

0.4

0.6

0.8

1

orm

aliz

ed

flu

ore

sc

en

ce

inte

ns

ity

ATM-WT-YFP

ATM-S367A-YFP

ATM-S1981A-YFP

0

0 50 100 150 200 250 300 350 400 450

Time (s)

n ATM-S1981A-YFP

Page 28: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

pre

wt

30s 60s

120s 150s 180s 210s

240s 270s 300s 360s

pre 30s 90s

Supplementary Figure 8

0

0.2

0.4

0.6

0.8

1

1.2

1.4

-50 0 50 100 150 200 250 300 350 400 450 500

Rela

tive F

luo

rescen

ce In

ten

sit

y

Time (s)

YFP-ATM-S2996A recruitment (gold ions)

S2996A

wt

Page 29: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

1

10

100

0 1 2 3 4

% S

urv

ival

Dose (Gy)

C35ABR

AT+Vector

AT+pMAT1

AT+S1981A

AT+S2996A

Supplementary Figure 9

A

B

- + - + - + Cd2+ pMAT1 S2996A S1981A

IP: FLAG

ATM

Page 30: AutophosphorylationandATMActivation239341/UQ239341_OA.pdfAutophosphorylationandATMActivation ADDITIONALSITESADDTOTHECOMPLEXITY* S Receivedforpublication,November16,2010 Published,JBCPapersinPress,December13,2010,DOI10.1074

Sairai So, David Chen and Martin F. LavinMarcel Tanuji, Philip Chen, Phillip J. Robinson, Gisela Taucher-Scholz, Keiji Suzuki, Sergei V. Kozlov, Mark E. Graham, Burkhard Jakob, Frank Tobias, Amanda W. Kijas,

COMPLEXITYAutophosphorylation and ATM Activation: ADDITIONAL SITES ADD TO THE

doi: 10.1074/jbc.M110.204065 originally published online December 13, 20102011, 286:9107-9119.J. Biol. Chem. 

  10.1074/jbc.M110.204065Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2010/12/13/M110.204065.DC1.html

  http://www.jbc.org/content/286/11/9107.full.html#ref-list-1

This article cites 74 references, 29 of which can be accessed free at

at UQ

Library on Septem

ber 5, 2016http://w

ww

.jbc.org/D

ownloaded from