Volume 16, Issue 5, Pages (December 2004)

Slides:



Advertisements
Similar presentations
AB Supplementary Figure S1. (A) H1299 cells harboring pGC were transfected with I-SceI-expressing vector. Percentage of GFP + cells was assessed after.
Advertisements

Soy Isoflavones Augment Radiation Effect by Inhibiting APE1/Ref-1 DNA Repair Activity in Non-small Cell Lung Cancer  Vinita Singh-Gupta, PhD, Michael.
Evaluation of Severe Combined Immunodeficiency and Combined Immunodeficiency Pediatric Patients on the Basis of Cellular Radiosensitivity  Pavel Lobachevsky,
Volume 53, Issue 4, Pages (February 2014)
MK-8776, a novel Chk1 inhibitor, exhibits an improved radiosensitizing effect compared to UCN-01 by exacerbating radiation-induced aberrant mitosis  Motofumi.
Volume 35, Issue 3, Pages (August 2009)
Effects of Betulinic Acid Alone and in Combination with Irradiation in Human Melanoma Cells  Edgar Selzer, Emilio Pimentel, Volker Wacheck, Werner Schlegel,
Volume 65, Issue 1, Pages (January 2017)
Volume 13, Issue 3, Pages (October 2015)
SMARCAD1 Phosphorylation and Ubiquitination Are Required for Resection during DNA Double-Strand Break Repair  Sharmistha Chakraborty, Raj K. Pandita,
Volume 114, Issue 3, Pages (August 2003)
Supplemental Figure 1 Li Fraumeni (087) 5C tankyrase1 actin Mock
Volume 2, Issue 4, Pages (October 1998)
The Circadian Gene Period2 Plays an Important Role in Tumor Suppression and DNA Damage Response In Vivo  Loning Fu, Helene Pelicano, Jinsong Liu, Peng.
Human chronic lymphocytic leukemia B cells can escape DNA damage-induced apoptosis through the nonhomologous end-joining DNA repair pathway by Ludovic.
A DNA Damage Response Pathway Controlled by Tel1 and the Mre11 Complex
Volume 35, Issue 2, Pages (July 2009)
Jennifer L. Rizzo, Jessica Dunn, Adam Rees, Thomas M. Rünger 
Volume 31, Issue 2, Pages (July 2008)
Volume 18, Issue 5, Pages (May 2005)
Distinct but Concerted Roles of ATR, DNA-PK, and Chk1 in Countering Replication Stress during S Phase  Rémi Buisson, Jessica L. Boisvert, Cyril H. Benes,
Volume 8, Issue 6, Pages (December 2001)
Oocytes Progress beyond Prophase in the Presence of DNA Damage
Volume 13, Issue 8, Pages (November 2015)
Volume 44, Issue 5, Pages (December 2011)
Volume 23, Issue 2, Pages (July 2006)
Lana Bozulic, Banu Surucu, Debby Hynx, Brian A. Hemmings 
Human RAD50 Deficiency in a Nijmegen Breakage Syndrome-like Disorder
Volume 51, Issue 6, Pages (September 2013)
Robert L.S Perry, Maura H Parker, Michael A Rudnicki  Molecular Cell 
Volume 30, Issue 3, Pages (May 2008)
Modeling the Therapeutic Efficacy of p53 Restoration in Tumors
Volume 54, Issue 3, Pages (May 2014)
Volume 35, Issue 3, Pages (August 2009)
Volume 13, Issue 2, Pages (January 2004)
TNF-Induced Activation of the Nox1 NADPH Oxidase and Its Role in the Induction of Necrotic Cell Death  You-Sun Kim, Michael J. Morgan, Swati Choksi, Zheng-gang.
Volume 66, Issue 4, Pages e5 (May 2017)
Volume 7, Issue 4, Pages (April 2001)
Volume 69, Issue 5, Pages e5 (March 2018)
Raymond L. Warters, Patrick J. Adamson, Christopher D. Pond, Sancy A
Volume 35, Issue 4, Pages (August 2009)
Volume 48, Issue 5, Pages (December 2012)
Volume 114, Issue 3, Pages (August 2003)
Volume 55, Issue 5, Pages (September 2014)
Volume 39, Issue 1, Pages (July 2010)
Volume 24, Issue 10, Pages (May 2014)
Volume 59, Issue 4, Pages (August 2015)
Volume 62, Issue 6, Pages (June 2016)
Single-Stranded DNA Orchestrates an ATM-to-ATR Switch at DNA Breaks
Mst1 Is an Interacting Protein that Mediates PHLPPs' Induced Apoptosis
Volume 13, Issue 10, Pages (December 2015)
Volume 64, Issue 3, Pages (November 2016)
Volume 16, Issue 2, Pages (February 2009)
Two Distinct Modes of ATR Activation Orchestrated by Rad17 and Nbs1
Sirt6 Promotes DNA End Joining in iPSCs Derived from Old Mice
Volume 45, Issue 3, Pages (February 2012)
Volume 13, Issue 10, Pages (December 2015)
Shrestha Ghosh, Baohua Liu, Yi Wang, Quan Hao, Zhongjun Zhou 
Volume 123, Issue 6, Pages (December 2005)
SYCE2 potentiates the steady-state DNA repair activity without affecting the cell cycle checkpoint. SYCE2 potentiates the steady-state DNA repair activity.
Markus Löbrich, Penny Jeggo  Trends in Biochemical Sciences 
SIRT1 Regulates the Function of the Nijmegen Breakage Syndrome Protein
Volume 43, Issue 5, Pages (September 2011)
Volume 13, Issue 1, Pages (October 2015)
Single-Stranded DNA Orchestrates an ATM-to-ATR Switch at DNA Breaks
Ataxia telangiectasia and DNA-dependent protein kinase (DNA-PK) catalytic subunit-deficient cell lines efficiently form H2AX foci after exposure to ionizing.
Jörg Hartkamp, Brian Carpenter, Stefan G.E. Roberts  Molecular Cell 
Volume 13, Issue 1, Pages (October 2015)
Interaction of NHEJ and HR pathways in PCO cultures.
Volume 24, Issue 10, Pages (May 2014)
Presentation transcript:

Volume 16, Issue 5, Pages 715-724 (December 2004) A Pathway of Double-Strand Break Rejoining Dependent upon ATM, Artemis, and Proteins Locating to γ-H2AX Foci  Enriqueta Riballo, Martin Kühne, Nicole Rief, Aidan Doherty, Graeme C.M. Smith, Marı́a-José Recio, Caroline Reis, Kirsten Dahm, Andreas Fricke, Andrea Krempler, Antony R. Parker, Stephen P. Jackson, Andrew Gennery, Penny A. Jeggo, Markus Löbrich  Molecular Cell  Volume 16, Issue 5, Pages 715-724 (December 2004) DOI: 10.1016/j.molcel.2004.10.029

Figure 1 ATM- and Artemis-Deficient Cells Exhibit an Identical DSB Repair Defect (A) PFGE analysis in control fibroblasts (MRC-5) and lines defective in ATM (AT1BR) or Artemis (CJ179, CJ179hTert). Confluent nondividing cells were irradiated with 80 Gy X-rays and incubated for 24 hr (left panel) or 21 days (right panel). Similar results were obtained after 40 Gy γ-rays (data not shown). (B) γ-H2AX analysis after 2 Gy X-irradiation. HSF2 is another control fibroblast line. Without irradiation, all cell lines had less than 0.5 foci/cell. Similar results were obtained following 2 Gy γ-rays. (C) Artemis-defective cell lines (CJ179, DB333, F01-240, and F02-385) show a DSB repair phenotype similar to an A-T line (AT7BI) and distinct from a control (1BR3). γ-H2AX analysis was performed after 1.3 Gy γ-irradiation. The mean and SEM of three experiments is shown. Initial foci were assessed at 15 min post IR. (D) γ-H2AX analysis after 2 Gy γ-irradiation in CJ179 cells following transfection with vector cDNA, c-Myc-Artemis cDNA, or D37N c-Myc-Artemis cDNA. Transfection resulted in a small increase in γ-H2AX foci in unirradiated cells likely due to the introduction of DNA ends. The lack of full correction is most likely due to inefficient expression of c-Myc-Artemis cDNA in the primary cells. Transfection of c-Myc-Artemis cDNA in 1BR3 cells did not affect γ-H2AX numbers compared to cells transfected with c-Myc-vector (data not shown). With the exception of the analysis of the D37N mutant (n = 3), error bars represent the SEM from six experiments. Molecular Cell 2004 16, 715-724DOI: (10.1016/j.molcel.2004.10.029)

Figure 2 Epistasis Analysis Shows that ATM, Artemis, and DNA-PK Function in the Same Pathway (A) DSB repair after 2 Gy X-irradiation in the presence or absence of KU-55933 (ATMi). Initial foci were assessed at 30 min post IR. Error bars represent the SEM from two to three experiments. If not visible, they lie within the symbols. (B) γ-H2AX analysis in wild-type, ATM−/−, DNA-PKcs−/−, and LIG4−/− MEFs after 2 Gy X-irradiation demonstrating a repair defect in ATM−/− MEFs. Initial foci were assessed at 15 min post IR. Without irradiation, the number of foci/cell was below 0.5 for all cell lines. Since MEFs cannot be held in confluency for prolonged periods, foci analysis was undertaken maximally at 3 days post irradiation. (C) DSB repair after 2 Gy X-irradiation in the presence or absence of the ATM inhibitor KU-55933 (ATMi). Wild-type MEFs treated with KU-55933 show a repair defect similar to ATM−/− MEFs (compare with [B]). LIG4−/− MEFs exhibit the same level of unrepaired DSBs with and without KU-55933 treatment. The counting of these foci was computer assisted (see Supplemental Data for further details). (D) DSB repair after 2 Gy X-irradiation in the presence or absence of the DNA-PKcs inhibitor NU7026 (PKi). Wild-type MEFs treated with NU7026 show a repair defect similar to DNA-PKcs−/− MEFs, and NU7026 treatment of DNA-PKcs−/− MEFs has no effect. (E) DSB repair after 2 Gy X-irradiation in primary human fibroblasts in the presence or absence of NU7026. Control cells treated with NU7026 show a repair defect greater than that of Artemis cells. Artemis cells treated with NU7026 exhibit the same level of unrepaired DSBs as NU7026-treated control cells. Initial foci were assessed at 30 min post IR. Error bars represent the SEM from two to three experiments. If not visible, they lie within the symbols. Molecular Cell 2004 16, 715-724DOI: (10.1016/j.molcel.2004.10.029)

Figure 3 Nbs1, Mre11, 53BP1, and H2AX Are Required for ATM-Dependent NHEJ (A) γ-H2AX foci analysis at 1 day post 2 Gy X-rays shows a repair defect in ATLD (ATLD2) and NBS (CZD82CH) cells. Addition of KU-55933 (ATMi) and NU7026 (PKi) to ATLD2 and CZD82CH does not cause additivity in foci numbers. Without irradiation, the numbers of foci per cell were below 0.5 for all lines. (B) γ-H2AX analysis at 1 day post 2 Gy in wild-type, Chk2−/−, and 53BP1−/− MEFs. Addition of KU-55933 (ATMi) and NU7026 (PKi) to 53BP1−/− MEFs does not cause additivity in foci numbers. Chk2−/− MEFs respond like wild-type MEFs. (C) PFGE analysis in wild-type, ATM−/−, H2AX−/−, DNA-PKcs−/−, and LIG4−/− MEFs after 80 Gy X-irradiation. H2AX−/− MEFs show a repair defect similar to that of ATM−/− MEFs. The panel displays the fraction of activity released (FAR) versus repair time (in contrast to Figure 1A, which shows percentage of unrepaired DSBs versus repair time) to allow a comparison with Figure 4C (see Supplemental Data). Error bars represent the SEM from two to four experiments. Molecular Cell 2004 16, 715-724DOI: (10.1016/j.molcel.2004.10.029)

Figure 4 The Fraction of ATM-Dependent DSB Repair Depends on the Nature of the Break (A) γ-H2AX analysis in control fibroblasts (MRC-5, HSF2) and lines defective in ATM (AT1BR) or Artemis (CJ179) after exposure to 2 Gy α particles. The fraction of unrepaired DSBs (DSBs remaining/DSBs induced) in AT1BR and CJ179 is 2-fold higher than after X-irradiation (compare with Figure 1B). (B) γ-H2AX analysis in control fibroblasts (48BR) and lines defective in LIG4 (180BR), ATM (AT7BI), or Artemis (F01-240) after exposure to 20 μM etoposide. The concentration of etoposide used induces the same number of γ-H2AX foci as 1.3 Gy γ-rays. No repair defect is observed in ATM- and Artemis-deficient cells (compare with Figure 1C). (C) PFGE analysis in wild-type, ATM−/−, H2AX−/−, DNA-PKcs−/−, and LIG4−/− MEFs after exposure to 500 μM etoposide. In agreement with (B), no DSB repair defect can be observed in ATM−/− MEFs. H2AX−/− MEFs also fail to show a repair defect. The relative requirements for DNA-PKcs and LIG4 in DSB repair after etoposide treatment are similar to those after IR (compare with Figure 3C). Error bars represent the SEM from two to three experiments. Molecular Cell 2004 16, 715-724DOI: (10.1016/j.molcel.2004.10.029)

Figure 5 ATM-Dependent DSB Rejoining Substantially Contributes to Survival and Affects the Repair of Potentially Lethal Damage (A) A-T (AT1BR, AT5BI) and Artemis (CJ179, DB333, F02-385) lines show similar radiosensitivity. (B) H2AX−/− and 53BP1−/− MEFs show similar sensitivity to ATM−/− MEFs. LIG4−/− MEFs, which show a dramatic defect in DSB repair, are more radiosensitive. (C) Control fibroblasts (1BR3) and lines defective in Artemis (CJ179) were either plated immediately (IP) or 1 day after irradiation (DP). CJ179 does not show significant recovery in delayed plating experiments. Error bars represent the SEM from three experiments. Molecular Cell 2004 16, 715-724DOI: (10.1016/j.molcel.2004.10.029)

Figure 6 ATM Is Required for Radiation-Induced Artemis Hyperphosphorylation, and Artemis Coassociates with 53BP1 (A) KU-55933 (ATMi) inhibits IR-induced Artemis hyperphosphorylation. MRC5Vi cells (transformed MRC-5) were stably transfected with c-Myc-Artemis cDNA. Where indicated, KU-55933 or NU7026 (DNA-PKi) was added 1 hr prior to irradiation. Samples were analyzed 1 hr post irradiation with 2 Gy γ-rays (lower panel). The upper panel shows control cells (no drug treatment) irradiated with 10 Gy and treated with phosphatase. (B) IR-induced Artemis hyperphosphorylation is not observed in A-T cells. AT5BIVa cells (transformed AT5BI) were transiently transfected with c-Myc-Artemis cDNA. 48 hr post transfection, cells were irradiated with 10 Gy and analyzed 1 hr later. (C) Analysis of IR-induced Artemis hyperphosporylation in wild-type, ATM−/−, and DNA-PKcs−/− MEFs. MEFs were transiently transfected with c-Myc-Artemis cDNA. 48 hr post transfection, cells were irradiated with 10 Gy and analyzed 1 hr later. Note that (B) and (C) used a Western blotting system distinct from (A) to maximize the separation between the forms of Artemis. (D) Artemis coassociates with 53BP1. MRC5Vi cells were stably transfected with c-Myc-Artemis (Artemis) or c-Myc (Vector) cDNA. WCE (without irradiation) was immunoprecipitated using α-53BP1 or α-c-Myc and subjected to Western blotting using α-c-Myc or α-53BP1. Interaction was observed only in cells expressing c-Myc-Artemis cDNA. The c-Myc-Artemis band observed migrated at the expected ∼100 kDa. Immunoprecipitation was carried out in the presence of ethidium bromide. Molecular Cell 2004 16, 715-724DOI: (10.1016/j.molcel.2004.10.029)