ATM and Artemis promote homologous recombination of radiation-induced DNA double-strand breaks in G2

Andrea Beucher, Julie Birraux, Leopoldine Tchouandong, Olivia Barton, Atsushi Shibata, Sandro Conrad, Aaron A Goodarzi, Andrea Krempler, Penny A Jeggo, Markus Löbrich, Andrea Beucher, Julie Birraux, Leopoldine Tchouandong, Olivia Barton, Atsushi Shibata, Sandro Conrad, Aaron A Goodarzi, Andrea Krempler, Penny A Jeggo, Markus Löbrich

Abstract

Homologous recombination (HR) and non-homologous end joining (NHEJ) represent distinct pathways for repairing DNA double-strand breaks (DSBs). Previous work implicated Artemis and ATM in an NHEJ-dependent process, which repairs a defined subset of radiation-induced DSBs in G1-phase. Here, we show that in G2, as in G1, NHEJ represents the major DSB-repair pathway whereas HR is only essential for repair of approximately 15% of X- or gamma-ray-induced DSBs. In addition to requiring the known HR proteins, Brca2, Rad51 and Rad54, repair of radiation-induced DSBs by HR in G2 also involves Artemis and ATM suggesting that they promote NHEJ during G1 but HR during G2. The dependency for ATM for repair is relieved by depleting KAP-1, providing evidence that HR in G2 repairs heterochromatin-associated DSBs. Although not core HR proteins, ATM and Artemis are required for efficient formation of single-stranded DNA and Rad51 foci at radiation-induced DSBs in G2 with Artemis function requiring its endonuclease activity. We suggest that Artemis endonuclease removes lesions or secondary structures, which inhibit end resection and preclude the completion of HR or NHEJ.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Figure 1
Figure 1
(A) Identification of cell-cycle phases in human fibroblasts (HSF1). Cells were scanned under the microscope and the γH2AX signal was plotted against the DAPI signal. S-phase cells exhibited an intermediate DAPI signal, a weak CENP-F signal and a high pan-nuclear γH2AX signal due to aphidicolin treatment. G1- and G2-phase cells were distinguished from S-phase cells by their dotted instead of a pan-nuclear γH2AX signal. G1 and G2 cells were distinguished from each other either by DAPI content (low in G1 versus high in G2) or by CENP-F staining (absent in G1 versus strong pan-nuclear in G2). Mitotic cells exhibited CENP-F staining and condensed chromatin (Deckbar et al, 2007), and were excluded from the analysis. (B) DSB repair in G1- and G2-phase HSF1 cells is unaffected by aphidicolin treatment and aphidicolin itself (designated ‘control' in the figure) does not induce γH2AX foci. (C) γH2AX foci formation in HSF1 cells at 15 min post IR is linear with dose in G1 and G2 up to approximately 80 foci per cell. G2 cells exhibit about twice the number of DSBs as G1 cells, consistent with their two-fold higher DNA content (NB: The number of DSBs observed in G2 phase at later times (e.g. 8 h) is routinely higher than twice the number in G1-phase since the slow component of DSB repair in G2 is slower than in G1). Error bars in panels B and C represent the s.e.m. from analysis of at least 40 cells.
Figure 2
Figure 2
(A) γH2AX foci analysis in primary human fibroblasts. Background foci numbers in primary human fibroblasts were about 2 in G2 and 0.2 in G1, and were subtracted from the foci numbers in the irradiated samples. (B) γH2AX foci analysis in MEFs. Background foci numbers in MEFs were about 2–4 in G2 and 0.5–2 in G1, and were subtracted from the foci numbers in the irradiated samples. The insets magnify the 8-h data for WT, Brca2-; Rad54-, Artemis- and ATM-deficient cells. Statistical analysis was performed at the 6- and 8-h time points, and revealed that Artemis- and ATM-deficient cells in G1 and G2, and Brca2- and Rad54-deficient cells in G2 exhibit significantly elevated foci levels compared with WT cells (P<0.05; one-tailed Welch's test). (C) γH2AX foci analysis in siRNA-treated HeLa cells analysed 48 h after transfection. Efficient knockdown to protein levels <20% was confirmed for all tested siRNAs by Western blotting. Background foci numbers in HeLa cells were about 2–4 in G2 and 0.5–2 in G1, and were subtracted from the foci numbers in the irradiated samples. The number of induced foci measured at 15 min post-IR was similar for all siRNA conditions (data not shown). Samples were evaluated in a blinded manner. Statistical analysis was performed and revealed that cells depleted for Artemis or ATM in G1 and G2, and for Brca2 and Rad51 in G2 exhibit significantly elevated foci levels compared with control siRNA-treated cells at the 8- and 10-h time points (P<0.05; one-tailed Welch's test). Although the repair defect is apparent and similar for the different cell systems, the absolute foci numbers in HeLa cells are higher, likely due to their higher DNA content. Error bars in panels A–C represent the s.e.m. from at least three different experiments.
Figure 3
Figure 3
HR repair sites were visualized by incorporation of BrdU during repair synthesis. HeLa cells were irradiated with 4 Gy and BrdU was added for the entire repair time—BrdU foci were observed after denaturing conditions in G2- but not in G1-phase cells. S-phase cells were identified by pan-nuclear γH2AX or BrdU staining and excluded from the analysis. The image on the left shows control cells analysed 8 h following 4-Gy irradiation treatment. Efficient knockdown to protein levels

Figure 4

SCEs are detected in G2-irradiated…

Figure 4

SCEs are detected in G2-irradiated HeLa cells. Cells were grown for two cell…

Figure 4
SCEs are detected in G2-irradiated HeLa cells. Cells were grown for two cell cycles (48 h) in BrdU-containing medium and aphidicolin was added immediately prior to irradiation with 2 Gy. Colcemid was then added at 8 h post-IR and the samples were harvested at 12 h post-IR. Irradiation was performed 48 h after siRNA transfection. Samples designated ‘mock' were treated with transfection reagents, but no siRNA was added. SCE numbers were normalized to 70 chromosomes to account for the variability in chromosome number between metaphases. At least 100 metaphases from at least three different experiments were analysed. Error bars represent the s.e.m. from all analysed metaphases.

Figure 5

ssDNA formation is compromised in…

Figure 5

ssDNA formation is compromised in ATM/Artemis-deficient cells. ( A ) Analysis of RPA…

Figure 5
ssDNA formation is compromised in ATM/Artemis-deficient cells. (A) Analysis of RPA and Rad51 foci formation in primary human fibroblasts. (B) Analysis of Rad51 foci formation in siRNA-treated HeLa cells. (C) Analysis of ssDNA by measuring BrdU foci in siRNA-treated HeLa cells. Cells were labelled with BrdU for 24 h prior to irradiation and foci were observed after non-denaturing conditions. Error bars represent the s.e.m. from the analysis of at least three different experiments.

Figure 6

( A ) γH2AX foci…

Figure 6

( A ) γH2AX foci in primary human fibroblasts in the presence or…

Figure 6
(A) γH2AX foci in primary human fibroblasts in the presence or absence of the ATM inhibitor, KU55933 (designated ATMi in the figure). The insets magnify the 8-h data for WT, Brca2-; Artemis- and ATM-deficient cells treated with KU55933. (B) γH2AX foci analysis in MEFs in the presence or absence of KU55933. Statistical analysis was performed at the 6- and 8-h time points, and revealed that XLF- and Lig4-deficient cells treated with KU55933 exhibit significantly elevated foci levels compared with untreated XLF- and Lig4-deficient cells in G2 but not in G1 (P<0.05; one-tailed Welch's test). (C) γH2AX foci analysis in siRNA-treated HeLa cells. Samples were evaluated in a blinded manner. Statistical analysis was performed and revealed that cells depleted for Artemis, ATM, Control/Artemis, Rad51/Artemis, Rad51/ATM, Brca2/Artemis and Brca2/ATM in G1 and G2, and Rad51-, Brca2- and Rad51/Brca2-depleted cells in G2 exhibit significantly elevated foci levels compared with control siRNA- and mock-treated cells (P<0.05; one-tailed Welch's test). Samples designated ‘mock' were treated with transfection reagents, but no siRNA was added. The experiments for panels A–C were carried out as for Figure 2. Error bars represent the s.e.m. from at least three different experiments. (D) Analysis of G2 PCC chromosomal breaks in calyculin A-treated primary human fibroblasts in the presence of aphidicolin and in the presence or absence of KU55933. Breaks in un-irradiated samples were less than 0.2 and were subtracted from the breaks in the irradiated samples. Error bars represent the s.e.m. from at least 100 cells. Statistical analysis revealed that Brca2- and Artemis-deficient cells with and without KU55933 exhibit significantly more PCC breaks than WT cells without KU55933 at 4 and 6 h (P<0.05; one-tailed Welch's test).

Figure 7

( A ) γH2AX foci…

Figure 7

( A ) γH2AX foci analysis in primary human fibroblasts in the presence…

Figure 7
(A) γH2AX foci analysis in primary human fibroblasts in the presence or absence of the DNA-PK inhibitor, NU7026 (designated DNA-PKi in the figure). The analysis was carried out as for Figure 2A. Statistical analysis was performed at the 6- and 8-h time points, and revealed that Artemis- and Brca2-deficient cells with NU7026 exhibit significantly elevated foci levels compared with WT cells with NU7026 in G2 but not in G1 (P<0.05; one-tailed Welch's test). (B) γH2AX foci analysis at prolonged times after IR. Using an approach, which circumvents aphidicolin treatment, cells were pulse-labelled with BrdU for 1 h and irradiated with 9 Gy 4 h after labelling (when in G2). Due to the higher dose, the majority of irradiated G2 cells remained in G2 for at least 96 h (Supplementary Figure 7A). Foci were analysed in BrdU-positive cells and taken to represent G2 cells. The few BrdU-positive cells that had progressed from G2 into G1 during repair incubation were excluded from the analysis based on their lower DAPI signal compared with that of G2 cells. Data for G0 cells were obtained in parallel experiments from the analysis of confluent cultures. Results similar to G0 were obtained for G1 from the analysis of BrdU-negative cells (representing mainly G1 cells) in the same samples that were analysed for the G2 data (Supplementary Figure 7B). The continuing activity of NU7026 over 96 h was confirmed in control experiments (Supplementary Figure 7C). Error bars represent the s.e.m. from the analysis of at least three different experiments.

Figure 8

Artemis endonuclease activity promotes IR-induced…

Figure 8

Artemis endonuclease activity promotes IR-induced HR in G2. The DSB-repair defect of Artemis-deficient…

Figure 8
Artemis endonuclease activity promotes IR-induced HR in G2. The DSB-repair defect of Artemis-deficient cells (CJ179) is corrected with WT Artemis but not with an endonuclease-deficient Artemis construct (D37N), and overexpression of D37N confers a repair defect to HSF1 control cells. Analysis of γH2AX and Rad51 foci was conducted in cells positive for c-myc, that is, only cells efficiently transfected were included in the analysis. The image on the left shows two G2-phase, Artemis-deficient cells transfected with the WT Artemis construct at 8 h following 2-Gy irradiation. One of the two cells shows efficient Artemis expression (c-myc signal) and lower foci numbers. Error bars represent the s.e.m. from three different experiments. Statistical analysis revealed that Artemis-deficient cells with or without complementation of the Artemis D37N construct exhibit significantly elevated γH2AX foci levels (at 6 and 8 h) and significantly reduced Rad51 foci levels (at 2 h) compared with Artemis-deficient cells complemented with the Artemis WT construct (P<0.05; one-tailed Welch's test). Control cells overexpressing D37N-mutant Artemis exhibit significantly elevated γH2AX foci levels (at 6 and 8 h) and significantly reduced Rad51 foci levels (at 2 h) compared with control cells with or without overexpression of Artemis WT (P<0.05; one-tailed Welch's test). See Supplementary data for further information.

Figure 9

IR-induced HR in G2 repairs…

Figure 9

IR-induced HR in G2 repairs DSBs in KAP-1-associated heterochromatic DNA regions. ( A…

Figure 9
IR-induced HR in G2 repairs DSBs in KAP-1-associated heterochromatic DNA regions. (A) Quantification of % of KAP-1 in densely staining DAPI regions in G1- versus G2-phase and localization of γH2AX foci (assessed at 8 h following 2-Gy irradiation in NIH 3T3 mouse cells treated with the ATM inhibitor KU55933) and RPA foci (assessed at 2 h following 2-Gy irradiation in untreated NIH 3T3 cells). KAP-1 signal distribution was estimated by measuring the signal intensity within (i) densely staining DAPI regions and (ii) the total nuclear volume. By dividing (i) by (ii), the percentage of total KAP-1 localized to densely staining DAPI was calculated. In G2-phase, the values estimated were similar to that for the % DAPI staining, suggesting a random distribution of KAP-1, which was not the case in G1-phase where >70% of total KAP-1 was localized to densely staining DAPI, equivalent to <20% of the nuclear volume. Typical images analysed are shown in Supplementary Figure 8. The colocalization studies as well as the KAP-1 staining were performed according to Goodarzi et al (2008). (B) γH2AX foci analysis in 1BRneo cells downregulated for KAP-1 with or without the ATM inhibitor KU55933 (designated ATMi in the figure). (C) γH2AX foci analysis in siRNA-treated HeLa cells. Background foci numbers were subtracted from the foci numbers in the irradiated samples. Samples were evaluated in a blinded manner. Error bars represent the s.e.m. from the analysis of at least three different experiments. Statistical analysis was performed at the 8- and 10-h time points, and revealed that cells treated with control siRNA and KU55933 exhibit significantly elevated foci levels compared with cells treated with KAP-1 siRNA and KU55933. Further, cells treated with ATM siRNA exhibit significantly elevated foci levels compared with cells treated with ATM/KAP-1 double siRNA (P<0.05; one-tailed Welch's test). Although the repair defect is apparent and similar for the two different cell systems, the absolute foci numbers in HeLa cells are slightly higher, likely due to their higher DNA content.

Figure 10

Pathways of DSB repair during…

Figure 10

Pathways of DSB repair during the mammalian cell cycle (see text for explanation).

Figure 10
Pathways of DSB repair during the mammalian cell cycle (see text for explanation).
All figures (10)
Figure 4
Figure 4
SCEs are detected in G2-irradiated HeLa cells. Cells were grown for two cell cycles (48 h) in BrdU-containing medium and aphidicolin was added immediately prior to irradiation with 2 Gy. Colcemid was then added at 8 h post-IR and the samples were harvested at 12 h post-IR. Irradiation was performed 48 h after siRNA transfection. Samples designated ‘mock' were treated with transfection reagents, but no siRNA was added. SCE numbers were normalized to 70 chromosomes to account for the variability in chromosome number between metaphases. At least 100 metaphases from at least three different experiments were analysed. Error bars represent the s.e.m. from all analysed metaphases.
Figure 5
Figure 5
ssDNA formation is compromised in ATM/Artemis-deficient cells. (A) Analysis of RPA and Rad51 foci formation in primary human fibroblasts. (B) Analysis of Rad51 foci formation in siRNA-treated HeLa cells. (C) Analysis of ssDNA by measuring BrdU foci in siRNA-treated HeLa cells. Cells were labelled with BrdU for 24 h prior to irradiation and foci were observed after non-denaturing conditions. Error bars represent the s.e.m. from the analysis of at least three different experiments.
Figure 6
Figure 6
(A) γH2AX foci in primary human fibroblasts in the presence or absence of the ATM inhibitor, KU55933 (designated ATMi in the figure). The insets magnify the 8-h data for WT, Brca2-; Artemis- and ATM-deficient cells treated with KU55933. (B) γH2AX foci analysis in MEFs in the presence or absence of KU55933. Statistical analysis was performed at the 6- and 8-h time points, and revealed that XLF- and Lig4-deficient cells treated with KU55933 exhibit significantly elevated foci levels compared with untreated XLF- and Lig4-deficient cells in G2 but not in G1 (P<0.05; one-tailed Welch's test). (C) γH2AX foci analysis in siRNA-treated HeLa cells. Samples were evaluated in a blinded manner. Statistical analysis was performed and revealed that cells depleted for Artemis, ATM, Control/Artemis, Rad51/Artemis, Rad51/ATM, Brca2/Artemis and Brca2/ATM in G1 and G2, and Rad51-, Brca2- and Rad51/Brca2-depleted cells in G2 exhibit significantly elevated foci levels compared with control siRNA- and mock-treated cells (P<0.05; one-tailed Welch's test). Samples designated ‘mock' were treated with transfection reagents, but no siRNA was added. The experiments for panels A–C were carried out as for Figure 2. Error bars represent the s.e.m. from at least three different experiments. (D) Analysis of G2 PCC chromosomal breaks in calyculin A-treated primary human fibroblasts in the presence of aphidicolin and in the presence or absence of KU55933. Breaks in un-irradiated samples were less than 0.2 and were subtracted from the breaks in the irradiated samples. Error bars represent the s.e.m. from at least 100 cells. Statistical analysis revealed that Brca2- and Artemis-deficient cells with and without KU55933 exhibit significantly more PCC breaks than WT cells without KU55933 at 4 and 6 h (P<0.05; one-tailed Welch's test).
Figure 7
Figure 7
(A) γH2AX foci analysis in primary human fibroblasts in the presence or absence of the DNA-PK inhibitor, NU7026 (designated DNA-PKi in the figure). The analysis was carried out as for Figure 2A. Statistical analysis was performed at the 6- and 8-h time points, and revealed that Artemis- and Brca2-deficient cells with NU7026 exhibit significantly elevated foci levels compared with WT cells with NU7026 in G2 but not in G1 (P<0.05; one-tailed Welch's test). (B) γH2AX foci analysis at prolonged times after IR. Using an approach, which circumvents aphidicolin treatment, cells were pulse-labelled with BrdU for 1 h and irradiated with 9 Gy 4 h after labelling (when in G2). Due to the higher dose, the majority of irradiated G2 cells remained in G2 for at least 96 h (Supplementary Figure 7A). Foci were analysed in BrdU-positive cells and taken to represent G2 cells. The few BrdU-positive cells that had progressed from G2 into G1 during repair incubation were excluded from the analysis based on their lower DAPI signal compared with that of G2 cells. Data for G0 cells were obtained in parallel experiments from the analysis of confluent cultures. Results similar to G0 were obtained for G1 from the analysis of BrdU-negative cells (representing mainly G1 cells) in the same samples that were analysed for the G2 data (Supplementary Figure 7B). The continuing activity of NU7026 over 96 h was confirmed in control experiments (Supplementary Figure 7C). Error bars represent the s.e.m. from the analysis of at least three different experiments.
Figure 8
Figure 8
Artemis endonuclease activity promotes IR-induced HR in G2. The DSB-repair defect of Artemis-deficient cells (CJ179) is corrected with WT Artemis but not with an endonuclease-deficient Artemis construct (D37N), and overexpression of D37N confers a repair defect to HSF1 control cells. Analysis of γH2AX and Rad51 foci was conducted in cells positive for c-myc, that is, only cells efficiently transfected were included in the analysis. The image on the left shows two G2-phase, Artemis-deficient cells transfected with the WT Artemis construct at 8 h following 2-Gy irradiation. One of the two cells shows efficient Artemis expression (c-myc signal) and lower foci numbers. Error bars represent the s.e.m. from three different experiments. Statistical analysis revealed that Artemis-deficient cells with or without complementation of the Artemis D37N construct exhibit significantly elevated γH2AX foci levels (at 6 and 8 h) and significantly reduced Rad51 foci levels (at 2 h) compared with Artemis-deficient cells complemented with the Artemis WT construct (P<0.05; one-tailed Welch's test). Control cells overexpressing D37N-mutant Artemis exhibit significantly elevated γH2AX foci levels (at 6 and 8 h) and significantly reduced Rad51 foci levels (at 2 h) compared with control cells with or without overexpression of Artemis WT (P<0.05; one-tailed Welch's test). See Supplementary data for further information.
Figure 9
Figure 9
IR-induced HR in G2 repairs DSBs in KAP-1-associated heterochromatic DNA regions. (A) Quantification of % of KAP-1 in densely staining DAPI regions in G1- versus G2-phase and localization of γH2AX foci (assessed at 8 h following 2-Gy irradiation in NIH 3T3 mouse cells treated with the ATM inhibitor KU55933) and RPA foci (assessed at 2 h following 2-Gy irradiation in untreated NIH 3T3 cells). KAP-1 signal distribution was estimated by measuring the signal intensity within (i) densely staining DAPI regions and (ii) the total nuclear volume. By dividing (i) by (ii), the percentage of total KAP-1 localized to densely staining DAPI was calculated. In G2-phase, the values estimated were similar to that for the % DAPI staining, suggesting a random distribution of KAP-1, which was not the case in G1-phase where >70% of total KAP-1 was localized to densely staining DAPI, equivalent to <20% of the nuclear volume. Typical images analysed are shown in Supplementary Figure 8. The colocalization studies as well as the KAP-1 staining were performed according to Goodarzi et al (2008). (B) γH2AX foci analysis in 1BRneo cells downregulated for KAP-1 with or without the ATM inhibitor KU55933 (designated ATMi in the figure). (C) γH2AX foci analysis in siRNA-treated HeLa cells. Background foci numbers were subtracted from the foci numbers in the irradiated samples. Samples were evaluated in a blinded manner. Error bars represent the s.e.m. from the analysis of at least three different experiments. Statistical analysis was performed at the 8- and 10-h time points, and revealed that cells treated with control siRNA and KU55933 exhibit significantly elevated foci levels compared with cells treated with KAP-1 siRNA and KU55933. Further, cells treated with ATM siRNA exhibit significantly elevated foci levels compared with cells treated with ATM/KAP-1 double siRNA (P<0.05; one-tailed Welch's test). Although the repair defect is apparent and similar for the two different cell systems, the absolute foci numbers in HeLa cells are slightly higher, likely due to their higher DNA content.
Figure 10
Figure 10
Pathways of DSB repair during the mammalian cell cycle (see text for explanation).

References

    1. Ahnesorg P, Smith P, Jackson SP (2006) XLF interacts with the XRCC4–DNA ligase IV complex to promote DNA nonhomologous end-joining. Cell 124: 301–313
    1. Allen C, Kurimasa A, Brenneman MA, Chen DJ, Nickoloff JA (2002) DNA-dependent protein kinase suppresses double-strand break-induced and spontaneous homologous recombination. Proc Natl Acad Sci USA 99: 3758–3763
    1. Asakawa Y, Gotoh E (1997) A method for detecting sister chromatid exchanges using prematurely condensed chromosomes and immunogold–silver staining. Mutagenesis 12: 175–177
    1. Barlow JH, Lisby M, Rothstein R (2008) Differential regulation of the cellular response to DNA double-strand breaks in G1. Mol Cell 30: 73–85
    1. Bekker-Jensen S, Lukas C, Kitagawa R, Melander F, Kastan MB, Bartek J, Lukas J (2006) Spatial organization of the mammalian genome surveillance machinery in response to DNA strand breaks. J Cell Biol 173: 195–206
    1. Buck D, Malivert L, de Chasseval R, Barraud A, Fondanèche MC, Sanal O, Plebani A, Stéphan JL, Hufnagel M, le Deist F, Fischer A, Durandy A, de Villartay JP, Revy P (2006) Cernunnos, a novel nonhomologous end-joining factor, is mutated in human immunodeficiency with microcephaly. Cell 124: 287–299
    1. Darroudi F, Wiegant W, Meijers M, Friedl AA, van der Burg M, Fomina J, van Dongen JJ, van Gent DC, Zdzienicka MZ (2007) Role of Artemis in DSB repair and guarding chromosomal stability following exposure to ionizing radiation at different stages of cell cycle. Mutat Res 615: 111–124
    1. Deckbar D, Birraux J, Krempler A, Tchouandong L, Beucher A, Walker S, Stiff T, Jeggo PA, Löbrich M (2007) Chromosome breakage after G2 checkpoint release. J Cell Biol 176: 749–755
    1. Esashi F, Galkin VE, Yu X, Egelman EH, West SC (2007) Stabilization of RAD51 nucleoprotein filaments by the C-terminal region of BRCA2. Nat Struct Mol Biol 14: 468–474
    1. Fischle W, Tseng BS, Dormann HL, Ueberheide BM, Garcia BA, Shabanowitz J, Hunt DF, Funabiki H, Allis CD (2005) Regulation of HP1–chromatin binding by histone H3 methylation and phosphorylation. Nature 438: 1116–1122
    1. Goodarzi AA, Noon AT, Deckbar D, Ziv Y, Shiloh Y, Löbrich M, Jeggo PA (2008) ATM signaling facilitates repair of DNA double-strand breaks associated with heterochromatin. Mol Cell 31: 167–177
    1. Goodarzi AA, Noon AT, Jeggo PA (2009) The impact of heterochromatin on DSB repair. Biochem Soc Trans 37: 569–576
    1. Goodarzi AA, Yu Y, Riballo E, Douglas P, Walker SA, Ye R, Härer C, Marchetti C, Morrice N, Jeggo PA, Lees-Miller SP (2006) DNA-PK autophosphorylation facilitates Artemis endonuclease activity. EMBO J 25: 3880–3889
    1. Hanada K, Budzowska M, Davies SL, van Drunen E, Onizawa H, Beverloo HB, Maas A, Essers J, Hickson ID, Kanaar R (2007) The structure-specific endonuclease Mus81 contributes to replication restart by generating double-strand DNA breaks. Nat Struct Mol Biol 14: 1096–1104
    1. Hinz JM, Yamada NA, Salazar EP, Tebbs RS, Thompson LH (2005) Influence of double-strand-break repair pathways on radiosensitivity throughout the cell cycle in CHO cells. DNA Rep (Amst) 4: 782–792
    1. Howlett NG, Taniguchi T, Olson S, Cox B, Waisfisz Q, De Die-Smulders C, Persky N, Grompe M, Joenje H, Pals G, Ikeda H, Fox EA, D'Andrea AD (2002) Biallelic inactivation of BRCA2 in Fanconi anemia. Science 297: 606–609
    1. Huertas P, Cortés-Ledesma F, Sartori AA, Aguilera A, Jackson SP (2008) CDK targets Sae2 to control DNA-end resection and homologous recombination. Nature 455: 689–692
    1. Jazayeri A, Falck J, Lukas C, Bartek J, Smith GC, Lukas J, Jackson SP (2006) ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nat Cell Biol 8: 37–45
    1. Johnson RD, Jasin M (2000) Sister chromatid gene conversion is a prominent double-strand break repair pathway in mammalian cells. EMBO J 19: 3398–3407
    1. Kao GD, McKenna WG, Yen TJ (2001) Detection of repair activity during the DNA damage-induced G2 delay in human cancer cells. Oncogene 20: 3486–3496
    1. Kanaar R, Wyman C, Rothstein R (2008) Quality control of DNA break metabolism: in the ‘end', it's a good thing. EMBO J 27: 581–588
    1. Kegel P, Riballo E, Kühne M, Jeggo PA, Löbrich M (2007) X-irradiation of cells on glass slides has a dose doubling impact. DNA Repair (Amst) 6: 1692–1697
    1. Krüger I, Rothkamm K, Löbrich M (2004) Enhanced fidelity for rejoining radiation-induced DNA double-strand breaks in the G2 phase of Chinese Hamster Ovary cells. Nucleic Acids Res 32: 2677–2684
    1. Kühne M, Riballo E, Rief N, Rothkamm K, Jeggo PA, Löbrich M (2004) A double-strand break repair defect in ATM-deficient cells contributes to radiosensitivity. Cancer Res 64: 500–508
    1. Li Y, Chirgadze DY, Bolanos-Garcia VM, Sibanda BL, Davies OR, Ahnesorg P, Jackson SP, Blundell TL (2008) Crystal structure of human XLF/Cernunnos reveals unexpected differences from XRCC4 with implications for NHEJ. EMBO J 27: 290–300
    1. Liang F, Romanienko PJ, Weaver DT, Jeggo PA, Jasin M (1996) Chromosomal double-strand break repair in Ku80-deficient cells. Proc Natl Acad Sci USA 93: 8929–8933
    1. Liao H, Winkfein RJ, Mack G, Rattner JB, Yen TJ (1995) CENP-F is a protein of the nuclear matrix that assembles onto kinetochores at late G2 and is rapidly degraded after mitosis. J Cell Biol 130: 507–518
    1. Lieber MR (2008) The mechanism of human nonhomologous DNA end joining. J Biol Chem 283: 1–5
    1. Löbrich M, Rydberg B, Cooper PK (1995) Repair of X-ray-induced DNA double-strand breaks in specific NotI restriction fragments in human fibroblasts: joining of correct and incorrect ends. Proc Natl Acad Sci USA 92: 12050–12054
    1. Löbrich M, Jeggo PA (2005) Harmonising the response to DSBs: a new string in the ATM bow. DNA Rep (Amst) 4: 749–759
    1. Löbrich M, Jeggo PA (2007) The impact of a negligent G2/M checkpoint on genomic instability and cancer induction. Nat Rev Cancer 7: 861–869
    1. Miyazaki T, Bressan DA, Shinohara M, Haber JE, Shinohara A (2004) In vivo assembly and disassembly of Rad51 and Rad52 complexes during double-strand break repair. EMBO J 25: 939–949
    1. Morrison C, Sonoda E, Takao N, Shinohara A, Yamamoto K, Takeda S (2000) The controlling role of ATM in homologous recombinational repair of DNA damage. EMBO J 19: 463–471
    1. Moynahan ME, Chiu JW, Koller BH, Jasin M (1999) Brca1 controls homology-directed DNA repair. Mol Cell 4: 511–518
    1. Moynahan ME, Pierce AJ, Jasin M (2001) BRCA2 is required for homology-directed repair of chromosomal breaks. Mol Cell 7: 263–272
    1. Nussenzweig A, Nussenzweig MC (2007) A backup DNA repair pathway moves to the forefront. Cell 131: 223–225
    1. Pannicke U, Ma Y, Hopfner KP, Niewolik D, Lieber MR, Schwarz K (2004) Functional and biochemical dissection of the structure-specific nuclease ARTEMIS. EMBO J 23: 1987–1997
    1. Pierce AJ, Johnson RD, Thompson LH, Jasin M (1999) XRCC3 promotes homology-directed repair of DNA damage in mammalian cells. Genes Dev 13: 2633–2638
    1. Riballo E, Kühne M, Rief N, Doherty A, Smith GC, Recio MJ, Reis C, Dahm K, Fricke A, Krempler A, Parker AR, Jackson SP, Gennery A, Jeggo PA, Löbrich M (2004) A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to γH2AX foci. Mol Cell 16: 715–724
    1. Roseaulin L, Yamada Y, Tsutsui Y, Russell P, Iwasaki H, Arcangioli B (2008) Mus81 is essential for sister chromatid recombination at broken replication forks. EMBO J 27: 1378–1387
    1. Rothkamm K, Krüger I, Thompson LH, Löbrich M (2003) Pathways of DNA double-strand break repair during the mammalian cell cycle. Mol Cell Biol 23: 5706–5715
    1. Sartori AA, Lukas C, Coates J, Mistrik M, Fu S, Bartek J, Baer R, Lukas J, Jackson SP (2007) Human CtIP promotes DNA end resection. Nature 22: 509–514
    1. Shrivastav M, Miller CA, De Haro LP, Durant ST, Chen BP, Chen DJ, Nickoloff JA (2009) DNA-PKcs and ATM co-regulate DNA double-strand break repair. DNA Repair (Amst) 8: 920–929
    1. Sonoda E, Sasaki MS, Morrison C, Yamaguchi-Iwai Y, Takata M, Takeda S (1999) Sister chromatid exchanges are mediated by homologous recombination in vertebrate cells. Mol Cell Biol 19: 5166–5169
    1. Soulas-Sprauel P, Rivera-Munoz P, Malivert L, Le Guyader G, Abramowski V, Revy P, Villartay JP (2007) V(D)J and immunoglobulin class switch recombinations: a paradigm to study the regulation of DNA end-joining. Oncogene 26: 7780–7791
    1. Taghian DG, Nickoloff JA (1997) Chromosomal double-strand breaks induce gene conversion at high frequency in mammalian cells. Mol Cell Biol 17: 6386–6393
    1. Thompson LH, Schild D (2002) Recombinational DNA repair and human disease. Mutat Res 509: 49–78
    1. Thorslund T, West SC (2007) BRCA2: a universal recombinase regulator. Oncogene 26: 7720–7730
    1. Trenz K, Smith E, Smith S, Costanzo V (2006) ATM and ATR promote Mre11 dependent restart of collapsed replication forks and prevent accumulation of DNA breaks. ATM and ATR promote Mre11 dependent restart of collapsed replication forks and prevent accumulation of DNA breaks. EMBO J 25: 1764–1774
    1. van der Burg M, Verkaik NS, den Dekker AT, Barendregt BH, Pico-Knijnenburg I, Tezcan I, vanDongen JJ, van Gent DC (2007) Defective Artemis nuclease is characterized by coding joints with microhomology in long palindromic-nucleotide stretches. Eur J Immunol 37: 3522–3528
    1. van Gent DC, van der Burg M (2007) Non-homologous end-joining, a sticky affair. Oncogene 26: 7731–7740
    1. Wang H, Wang H, Powell SN, Iliakis G, Wang Y (2004) ATR affecting cell radiosensitivity is dependent on homologous recombination repair but independent of nonhomologous end joining. Cancer Res 64: 7139–7143
    1. Wang J, Pluth JM, Cooper PK, Cowan MJ, Chen DJ, Yannone SM (2005) Artemis deficiency confers a DNA double-strand break repair defect and Artemis phosphorylation status is altered by DNA damage and cell cycle progression. DNA Repair (Amst) 4: 556–570
    1. West SC (2003) Molecular views of recombination proteins and their control. Nat Rev Mol Cell Biol 4: 435–445
    1. Weterings E, Chen DJ (2008) The endless tale of non-homologous end-joining. Cell Res 18: 114–124
    1. Wu W, Wang M, Wu W, Singh SK, Mussfeldt T, Iliakis G (2008) Repair of radiation induced DNA double strand breaks by backup NHEJ is enhanced in G2. DNA Repair (Amst) 7: 329–338
    1. Wyman C, Kanaar R (2006) DNA double-strand break repair: all's well that ends well. Annu Rev Genet 40: 363–383
    1. Wyman C, Warmerdam DO, Kanaar R (2008) From DNA end chemistry to cell-cycle response: the importance of structure, even when it's broken. Mol Cell 30: 5–6
    1. Yuan SS, Chang HL, Lee EY (2003) Ionizing radiation-induced Rad51 nuclear focus formation is cell cycle-regulated and defective in both ATM(−/−) and c-Abl(−/−) cells. Mutat Res 525: 85–92
    1. Yuan SS, Lee SY, Chen G, Song M, Tomlinson GE, Lee EY (1999) BRCA2 is required for ionizing radiation-induced assembly of Rad51 complex in vivo. Cancer Res 59: 3547–3551
    1. Ziv Y, Bielopolski D, Galanty Y, Lukas C, Taya Y, Schultz DC, Lukas J, Bekker-Jensen S, Bartek J, Shiloh Y (2006) Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM- and KAP-1 dependent pathway. Nat Cell Biol 8: 870–876

Source: PubMed

3
購読する