Otud5 promotes end-joining of deprotected telomeres by promoting atm-dependent phosphorylation of kap1s824

Nature

Otud5 promotes end-joining of deprotected telomeres by promoting atm-dependent phosphorylation of kap1s824"


Play all audios:

Loading...

ABSTRACT Appropriate repair of damaged DNA and the suppression of DNA damage responses at telomeres are essential to preserve genome stability. DNA damage response (DDR) signaling consists


of cascades of kinase-driven phosphorylation events, fine-tuned by proteolytic and regulatory ubiquitination. It is not fully understood how crosstalk between these two major classes of


post-translational modifications impact DNA repair at deprotected telomeres. Hence, we performed a functional genetic screen to search for ubiquitin system factors that promote KAP1S824


phosphorylation, a robust DDR marker at deprotected telomeres. We identified that the OTU family deubiquitinase (DUB) OTUD5 promotes KAP1S824 phosphorylation by facilitating ATM activation,


through stabilization of the ubiquitin ligase UBR5 that is required for DNA damage-induced ATM activity. Loss of OTUD5 impairs KAP1S824 phosphorylation, which suppresses end-joining mediated


DNA repair at deprotected telomeres and at DNA breaks in heterochromatin. Moreover, we identified an unexpected role for the heterochromatin factor KAP1 in suppressing DNA repair at


telomeres. Altogether our work reveals an important role for OTUD5 and KAP1 in relaying DDR-dependent kinase signaling to the control of DNA repair at telomeres and heterochromatin. SIMILAR


CONTENT BEING VIEWED BY OTHERS UBE2D3 FACILITATES NHEJ BY ORCHESTRATING ATM SIGNALLING THROUGH MULTI-LEVEL CONTROL OF RNF168 Article Open access 12 June 2024 REGULATION OF ALT-ASSOCIATED


HOMOLOGY-DIRECTED REPAIR BY POLYADP-RIBOSYLATION Article 12 October 2020 CHROMOSOME END PROTECTION BY RAP1-MEDIATED INHIBITION OF DNA-PK Article Open access 16 April 2025 INTRODUCTION Under


constant exposure to both environmental and endogenous sources of DNA damage, efficient repair of DNA lesions is essential to the maintenance of genome integrity1. Paradoxically, in


particular genomic contexts, such as telomeres, activation of DNA repair mechanisms instead leads to chromosomal aberrations and genomic instability2. Telomeres are specialized nucleoprotein


structures located at the natural ends of chromosomes. They consist of kilobases of TTAGGG DNA repeats and serve to counteract the gradual erosion of chromosomal ends upon each cycle of DNA


replication3,4. Telomeres are bound and protected by shelterin, a six-unit protein complex that prevents misrecognition of telomeres as DNA double-strand breaks (DSBs) and unwanted


activation of DNA repair pathways5,6,7,8. Shelterin blocks DDR signaling and DNA repair in large part through promoting the formation of a telomere-loop (t-loop) and inhibiting the


accessibility of telomeric DNA to components of the DDR machinery9. In the absence of shelterin function, deprotected telomeric DNA ends trigger DDR signaling that induces both cellular


senescence and DNA repair reactions that result in chromosome end-to-end fusions8,10. Such fusions are a major threat to chromosomal stability, particularly in cells escaping from


senescence. The signaling pathways involved in the DDR at dysfunctional telomeres share significant similarities with that of DNA DSBs. The DDR is initiated by the recognition of DSBs or


deprotected telomeres by the MRE11-RAD50-NBS1 (MRN) complex, followed by the activation of ATM, the primary kinase responsible for DDR signaling in DSB repair11,12. While the precise


mechanisms underlying ATM activation are still not fully understood, activated ATM phosphorylates several downstream targets, including p53, CHK2, H2A.X, and KAP1 (also known as TRIM28 or


TIF1B)13,14,15,16. These phosphorylation events lead to cell cycle arrest, local chromatin remodeling, and recruitment of essential repair factors, ultimately enabling the repair process. In


particular, phosphorylation of KAP1 has been shown to be functionally indispensable for the repair of DSBs, specifically at heterochromatin17,18. KAP1, in its native, SUMOylated state,


serves as a critical heterochromatin scaffolding factor that promotes transcriptional repression and chromatin condensation via its interaction with various chromatin remodelers. In the


presence of DSBs, the normally SUMOylated KAP1 is phosphorylated by ATM in a manner facilitated by 53BP1, RNF8 and MDC1, and exhibiting a pan-nuclear phosphorylation pattern18.


Phosphorylation of KAP1 triggers temporary decompaction of heterochromatin in a CHD3-dependent manner, and facilitates DNA repair in an otherwise restrictive environment19. While the DDR


signaling cascade is primarily dictated by the flow of signals in the form of protein phosphorylation, post-translational modifications by ubiquitination streamlines DDR signaling by


orchestrating protein turnover, chromatin remodeling, and recruitment of repair factors20,21,22,23. Accordingly, key roles in the DDR have been assigned to multiple ubiquitin-system factors.


However, the full complexity of how the phosphorylation and ubiquitination systems together orchestrate the DDR, and especially at telomeres, is still unknown. Here we addressed this by


performing a functional genetic screen targeted at identifying ubiquitin system factors that promote DDR activation at deprotected telomeres, using ATM-dependent phosphorylation of KAP1 at


its Ser-824 residue (KAP1S824) as the readout. Through this screen, we identified the OTU-family deubiquitinase (DUB) OTUD5 as a promoter of DDR activation at deprotected telomeres. OTUD5 is


a phospho-activated DUB that preferentially cleaves K48- and K63-linked poly- and di-ubiquitin chains in vitro24. Together with the ubiquitin ligase UBR5, OTUD5 has been linked to the


process of transcriptional suppression at damaged chromatin but has so far not been directly implicated in the regulation of DDR signaling and DNA repair at telomeres25. We find that OTUD5


promotes ATM-mediated phosphorylation of KAP1S824 in response to telomere deprotection, as well as in response to genome-wide, ionizing radiation (IR)-induced DNA damage. This is dependent


on the ability of OTUD5 to stabilize UBR5, previously implicated in ATM activation by alleviating ATMIN-dependent inhibition on ATM in response to DSBs26. In addition to its role in


promoting the signaling axis of the DDR, we found that OTUD5 promotes the efficiency of DNA repair exclusively at telomeres and heterochromatin, but not in general, non-specific chromatin


contexts. Such facilitation is independent of ATM-driven recruitment of 53BP1 and RIF1, as the localization of these NHEJ factors to deprotected telomeres is either unaffected or only


marginally compromised upon loss of OTUD5. Instead, we reveal that defective KAP1S824 phosphorylation is the crucial underpinning step that hinders DNA repair at telomeres in cells lacking


OTUD5, as ectopic expression of phosphomimic KAP1S824D is sufficient to restore telomeric NHEJ in OTUD5-depleted cells. This shows that the residual ATM activity in OTUD5-deficient cells is


inadequate to support sufficient phosphorylation of KAP1S824, a key step in DNA repair in KAP1-rich genomic environments such as the telomeres and heterochromatin, thereby translating into a


repair defect specifically in these regions. Moreover, by genetic inactivation of KAP1, we identify an unanticipated role for KAP1 in telomere protection by restricting NHEJ at deprotected


telomeres. Altogether, our work identifies OTUD5 as a key contributor to DDR signaling and DNA repair at deprotected telomeres and heterochromatin and points at KAP1 as a critical


determinant of end-joining mediated repair efficiency at dysfunctional telomeres. RESULTS A FUNCTIONAL GENETIC SCREEN IDENTIFIES UBIQUITIN SYSTEM FACTORS THAT PROMOTE DDR ACTIVATION AT


DEPROTECTED TELOMERES In mammalian cells, loss of function of TRF2, a core component of the shelterin complex, causes telomere deprotection, triggering DDR signaling at telomeres. A


well-characterized experimental system for studying DDR activities at telomeres is a _Trf2__−/−_;_p53__−/−_ mouse embryonic fibroblast (MEF) cell line that expresses the


temperature-sensitive TRF2I468A mutant (TRF2ts)27. At the permissive temperature (32 °C), TRF2ts functionally complements the loss of endogenous TRF2 and keeps telomeres in a protected


state. However, at non-permissive temperatures (37–39 °C) TRF2ts is inactivated, causing rapid and synchronous telomere deprotection and telomere-specific DDR activation (Fig. 1a). We


exploited this system to gain understanding on the requirements of ubiquitin system factors for efficient DDR activation at deprotected telomeres. We designed a fluorescence-activated cell


sorting (FACS)-based functional genetic screen in TRF2ts MEFs that allows identification of factors with a critical role in the activation of DDR signaling at deprotected telomers, so-called


activators of the telomere damage response (ATD). To circumvent complications to DDR dynamics from cells being in different cell cycle phases, we used TRF2ts MEFs modified with the


Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI) system28 to isolate G1 cells for detection of DDR activation. DDR signaling at TRF2-deprotected telomeres is by at large an ATM


kinase-dependent process5. Amongst ATM substrates, pKAP1S824 is strongly induced by telomere deprotection in TRF2ts MEFs27,29. This induction is also readily detectable by FACS, where a


significant upshift in pKAP1S824 intensity could be detected after incubating TRF2ts MEFs at 39 °C for 3 h (Fig. 1b). To validate the specificity of pKAP1S824 detection by FACS, we


pretreated TRF2ts MEFs with an ATM inhibitor or short hairpin RNA (shRNA) against ATM, before subjecting the cells to telomere deprotection (Fig. 1c, d). Indeed, ablation of ATM activity


abolishes induction of pKAP1S824 by telomere deprotection. This reinforces that our FACS detection strategy specifically detects ATM-dependent KAP1S824 phosphorylation at deprotected


telomeres. To conduct the screen, we infected TRF2ts-FUCCI MEFs in triplicate with a lentiviral library of 609 shRNAs (pLKO.1-Puro) that target 123 ubiquitin system factors in mouse (Fig. 


1e). By FACS, we selected the top 5% G1 cells with lowest pKAP1S824 signal after telomere deprotection for 3 h at 39 °C (Fig. 1e, Supplementary Fig. 1a). From this population displaying


impaired pKAP1S824 induction we extracted genomic DNA, recovered shRNA sequences by PCR, and determined the enrichment of shRNAs in this population, relative to the untreated control cell


population, by next generation sequencing and MaGECK-RRA analysis30. Significantly enriched shRNA targets point at gene candidates that are required for DDR activation following telomere


deprotection. The ATD screen identified UBE2D3, UBE2M and OTUD5 as the top three ubiquitin system factors that promote KAP1S824 phosphorylation at deprotected telomeres (Fig. 1f). UBE2D3 is


a ubiquitin-conjugating E2 enzyme of the UBE2D enzyme family that we recently found to affect KAP1S824 phosphorylation in an independent study31, and hence represents an internal positive


control. UBE2M is an E2 enzyme of both the ubiquitination and neddylation system, while OTUD5, also known as DUBA, is a DUB of the OTU (ovarian tumor) deubiquitinase family. For validation,


we individually depleted UBE2D3, UBE2M or OTUD5 in TRF2ts MEFs with multiple independent shRNAs that were included in the screen and subjected them to telomere deprotection. Indeed,


depletion of all three targets significantly impaired KAP1S824 phosphorylation upon telomere deprotection (Fig. 1g, h, Supplementary Fig. 1b, c). Due to the heavy involvement of UBE2M in the


neddylation pathway (being one of only two described E2 enzymes of the neddylation system)32, this study’s focus on ubiquitination system factors, and given that E2 enzymes in general have


more cellular targets than DUBs, we chose to further investigate the role of OTUD5. OTUD5 PROMOTES ATM-DEPENDENT DDR SIGNALING For more specific and robust depletion of OTUD5 in mechanistic


studies we continued using CRISPR/Cas9-mediated gene knockout approaches33. Depletion of OTUD5 in TRF2ts MEFs with 2 independent sgRNAs (g1, g1B) recapitulated the pKAP1S824 defect observed


with shRNA-mediated knockdown of OTUD5 (Fig. 1i, j). To address whether contribution of OTUD5 to DDR activation extends beyond a telomeric context, we challenged OTUD5-depleted human U2OS


cells with ionizing radiation (IR) to induce genome-wide DSBs. Similar to its role in the response to deprotected telomeres, we found that OTUD5 is also required for robust KAP1S824


phosphorylation in response to genome-wide DNA damage, as IR-induced KAP1S824 phosphorylation was significantly reduced in OTUD5-depleted cells (Fig. 2a, b). We then investigated the


mechanism underpinning the requirement of OTUD5 for robust KAP1S824 phosphorylation. We observed that in addition to pKAP1S824, phosphorylation of other ATM kinase substrates, including


pCHK2T68 and γH2A.X, were also reduced upon loss of OTUD5 (Fig. 2c), suggesting that ATM activity is compromised in absence of OTUD5. In line with this, autophosphorylation of ATM on


Ser-1981 (pATMS1981), indicative of active ATM kinase, was significantly reduced in OTUD5-depleted U2OS cells and TRF2ts MEFs, while total ATM levels remained unaltered (Fig. 2c, d,


Supplementary Fig. 2a). This suggests that OTUD5 promotes ATM activation, and thereby supports phosphorylation of its downstream targets, including KAP1, in the DDR signaling cascade. The


Ser-177 and Cys-224 residues of OTUD5 are critical to its phospho-activation and catalytic activity, respectively24. To assess the importance of these functional aspects of OTUD5 in


promoting KAP1S824 phosphorylation, we performed complementation experiments with ectopic expression of OTUD5WT, OTUD5S177A or OTUD5C224S. While expression of sgRNA-resistant,


codon-optimized OTUD5WT cDNA restored KAP1S824 phosphorylation in OTUD5-depleted cells, both OTUD5S177A and OTUD5C224S expression failed to do so (Fig. 2e, f, Supplementary Fig. 2b). Thus,


both the DUB activity and the phospho-activation of OTUD5 are required for ATM-dependent DDR signaling. OTUD5 PROMOTES END-JOINING MEDIATED FUSION OF DYSFUNCTIONAL TELOMERES Loss of


shelterin factor TRF2 results in processing of the deprotected telomeres by ligase IV- and Ku-dependent classical non-homologous end-joining (NHEJ), giving rise to chromosome-type telomere


fusions (abbreviated here as chromosome fusions)34,35. These fusions are generated primarily in G1 phase and appear in metaphase spreads as end-to-end fusions of both sister chromatids of


one chromosome to the sister chromatids of another chromosome. As OTUD5 is required for robust DDR activation at deprotected telomeres, we assessed whether telomeric NHEJ also requires


OTUD5. Indeed, depletion of OTUD5 significantly reduced the frequency of NHEJ-mediated chromosome end-to-end fusions upon telomere deprotection in TRF2ts MEFs, albeit to a lesser extent than


direct inhibition of ATM with an ATM inhibitor (KU-55963) (Fig. 3a, b). This indicates that OTUD5 is required for efficient NHEJ at telomeres. To address whether OTUD5 is required for NHEJ


at deprotected telomeres similarly in human cells, we depleted OTUD5 in HeLa cells harboring a doxycycline-inducible shRNA against TRF2, where telomere deprotection can be induced by


doxycycline treatment36. Consistent with our results in TRF2ts MEFs, ablation of OTUD5 in these HeLa cells significantly reduced both telomere fusions and KAP1S824 phosphorylation upon


doxycycline-induced TRF2 loss (Fig. 3c, Supplementary Fig. 3a, b). Furthermore, the defect in telomeric NHEJ observed in OTUD5-depleted cells is not attributable to aberrant cell cycle


progression, as OTUD5 depletion did not significantly perturb cell cycle distribution under unchallenged or telomere deprotection conditions (Supplementary Figs. 3c and 4a). Besides via


classical NHEJ, deprotected telomeres can also be fused by microhomology mediated end-joining (MMEJ). This is apparent in the absence of the core NHEJ factor Ku70 and, in particular, when


not only the function of TRF2 is lost but also that of TRF110. To assess the impact of OTUD5 on telomeric MMEJ, we depleted OTUD5 in


_Trf1__F/F__;Trf2__F/F__;Ku70__−/−__;p53__−/−__;Cre-ER__T2_ MEFs, and induced telomere deprotection by Cre-mediated deletion of TRF1 and TRF2. Although to a lesser extent than NHEJ-mediated


fusions, also MMEJ-mediated chromosome fusions were significantly reduced in OTUD5-deficient cells (Fig. 3d, e, Supplementary Fig. 3d, e). Taken together, we conclude that OTUD5 is required


both for efficient NHEJ and for efficient MMEJ at deprotected telomeres. Having established that OTUD5 promotes DNA repair by NHEJ and MMEJ at telomeres, we assessed if the role of OTUD5 in


DNA repair extends beyond a telomeric context, especially since we found OTUD5 to promote both telomere-specific and genome-wide DDR activation in mouse and human cells. To determine the


requirement of OTUD5 for end-joining mediated DNA repair in a genome-wide context, we assessed its impact on the NHEJ-dependent integration of a linearized plasmid, randomly into the genome


of U2OS cells37. Unlike loss of MAD2L2, a well-established factor that promotes NHEJ both at telomeres and genome-wide29, loss of OTUD5 did not significantly compromise NHEJ in this assay


(Fig. 3f, g). In addition, we assessed the contribution of OTUD5 to NHEJ outside of a telomeric context in HEK293T cells harboring a NHEJ-HR dual reporter system, in which the efficiency of


NHEJ can be measured by FACS38. Again, loss of OTUD5 did not significantly reduce NHEJ efficiency, in agreement with the results of the random plasmid integration assay (Fig. 3h,


Supplementary Figs. 3f and 4b). Altogether, this indicates that OTUD5 is not required for NHEJ in a general, nonspecific chromatin context, but promotes end-joining mediated DNA repair


pathways (both NHEJ and MMEJ), in a context-specific manner, at telomeres. OTUD5 PROMOTES ATM-DEPENDENT DDR ACTIVATION BY REGULATING THE UBR5-ATMIN AXIS After establishing a role for OTUD5


in promoting ATM-dependent DDR and DNA repair at deprotected telomeres, we sought to further understand the underlying regulatory circuit. OTUD5 was previously reported to play a role in


transcriptional repression at damaged chromatin by stabilizing the E3 ubiquitin ligase UBR525. Independently, UBR5 has been reported to promote ATM activation by inhibiting ATMIN, a negative


regulator of DSB-induced ATM activity, through non-degradative ubiquitination26. These previously established links prompted us to hypothesize that OTUD5 promotes ATM-dependent DDR and


telomeric DNA repair in a UBR5 and ATMIN-dependent manner. To test our hypothesis, we first assessed whether there is any detectable interaction between OTUD5, UBR5, ATMIN and ATM. We


ectopically expressed GFP-tagged OTUD5 in HEK293T cells and performed co-immunoprecipitation. Despite our expectation that OTUD5 interacts directly only with UBR5 and interacts transiently


and indirectly with ATMIN and/or ATM, thereby posing a challenge for detection, we readily detected an interaction of OTUD5 with all three endogenous proteins (UBR5, ATMIN and ATM) (Fig. 


4a). Next, we assessed the potential functional involvement of UBR5 in OTUD5-promoted KAP1S824 phosphorylation, by depleting OTUD5, UBR5, or both in U2OS cells. In line with previous


findings in the context of transcriptional repression, depletion of OTUD5 led to reduced levels of endogenous UBR5 protein, both in the absence and presence of IR (Fig. 4b). Upon IR, U2OS


cells lacking either OTUD5 or UBR5 displayed reduced pKAP1S824 (Fig. 4b), revealing that not only OTUD5, but also UBR5 is important for efficient KAP1S824 phosphorylation upon DNA damage.


Furthermore, depletion of OTUD5 in UBR5-deficient cells did not further aggravate the pKAP1S824 defect of these cells, indicating that OTUD5 and UBR5 act in an epistatic manner to promote


KAP1S824 phosphorylation, by OTUD5 acting upstream of UBR5 to stabilize it. In further support of this, depletion of UBR5 with independent sgRNAs in TRF2ts MEFs recapitulated the KAP1S824


phosphorylation defect displayed by OTUD5-depleted TRF2ts MEFs (Fig. 4c). Moreover, loss of UBR5 also significantly reduced telomere fusions upon telomere deprotection (Fig. 4d, e,


Supplementary Fig. 5a). Of note, we observed that protein abundance of OTUD5 is elevated in UBR5-deficient cells (Fig. 4b, c). This could indicate some form of internal homeostasis in


ubiquitination-mediated regulation of protein stability between OTUD5 and UBR5. UBR5 is known to promote DSB-induced DDR by counteracting ATMIN-mediated inhibition on ATM26. If OTUD5 indeed


promotes ATM-dependent DDR and telomeric DNA repair through the UBR5-ATMIN axis, ablation of ATMIN is expected to reverse the DDR defects exhibited by OTUD5-depleted cells. To test this, we


inhibited ATMIN expression in OTUD5-depleted TRF2ts MEFs by using 3 independent shRNAs (#5, #24, #84) (Supplementary Fig. 5b). In line with our hypothesis, depletion of ATMIN strongly and


significantly rescued both the KAP1S824 phosphorylation defect and the telomere fusion defect of OTUD5-depleted cells (Fig. 4f, g, Supplementary Fig. 5c). Furthermore, depletion of OTUD5 did


not further aggravate the telomere fusion defect of ATM inhibitor-treated cells (Fig. 4h, Supplementary Fig. 5d). Taken together, these results indicate that the functional role of OTUD5 in


DDR activation and end-joining at dysfunctional telomeres relies on its control of the UBR5-ATMIN-ATM regulatory axis. OTUD5 PROMOTES TELOMERIC END-JOINING BY PROMOTING KAP1S824


PHOSPHORYLATION To further investigate the mechanism underlying the control of end-joining mediated repair by OTUD5, we first considered the possibility that OTUD5 (in part) facilitates


telomeric NHEJ by promoting the stability of the core NHEJ factor Ku80. This, since it has been reported that OTUD5 counteracts proteasomal degradation of Ku80 in certain cell lines39. To


address this, we assessed the protein levels of Ku80 in OTUD5-depleted TRF2ts MEFs and U2OS cells. However, contrary to the previously reported finding in different cell lines, loss of OTUD5


did not lead to a reduction in Ku80 protein levels in MEFs or U2OS cells, regardless of DNA damage induction by IR or telomere deprotection (Supplementary Fig. 6a, b). Thus, OTUD5 does not


appear as a critical regulator of Ku stability in the cell types of our study, in line with that multiple E3s and DUBs have been implicated in controlling Ku stability39,40. Moreover,


destabilization of Ku80 is also unlikely to explain the telomere end-joining defects in OTUD5-depleted cells, as OTUD5-depleted cells also display reduced telomere MMEJ in cells deficient


for TRF1, TRF2 and Ku70, where Ku80 is both dispensable and absent. Additionally, destabilization of a pivotal NHEJ factor such as Ku80 would impair global NHEJ significantly, regardless of


genomic context, which is not what we observed for OTUD5-deficient cells. Hence, we conclude that the impact of OTUD5 on telomeric end-joining in the cells and conditions of our assays, is


not mediated via changes in the level of Ku70/80. At deprotected telomeres, ATM-dependent recruitment of 53BP1 is a critical determinant of NHEJ efficiency41. As OTUD5-depleted cells display


impaired ATM activation and ATM-dependent DDR, it reasonably follows that 53BP1 recruitment could be compromised in these cells, giving rise to defective telomere NHEJ. To address this, we


measured co-localization of 53BP1 foci with telomeres by IF-FISH upon TRF2 inactivation in TRF2ts cells. While treatment with an ATM inhibitor completely abolished the appearance of 53BP1


telomere dysfunction induced foci (TIFs), OTUD5 depletion did not hinder 53BP1 accumulation at deprotected telomeres (Fig. 5a, Supplementary Fig. 6d). This indicates that the residual ATM


activity in OTUD5-depleted cells is sufficient to support efficient 53BP1 recruitment to dysfunctional telomeres, and that the telomere end-joining defect in OTUD5-depleted cells is not


caused by failed 53BP1 recruitment. We next assessed the recruitment of RIF1 in response to telomere deprotection in OTUD5-depleted cells, by immunofluorescence. RIF1 is a key NHEJ factor


that is recruited to DSBs and dysfunctional telomeres in an ATM and 53BP1-dependent manner and promotes end-joining by restricting DNA end resection42,43,44,45. In line with previous


findings, ATM inhibitor treatment fully abolished RIF1 foci formation upon telomere deprotection. On the other hand, RIF1 foci formation was only mildly impaired in OTUD5-depleted cells,


reaching statistical significance for only one of the two sgRNAs (sgOTUD5 g1B) used (Fig. 5b, Supplementary Fig. 6e), thereby recapitulating only a fraction of the effect of ATM inhibitor


treatment. Similar to RIF1 foci, formation of γH2A.X foci upon telomere deprotection was marginally compromised in cells treated with sgOTUD5 g1B and not significantly changed with the other


sgRNA (Supplementary Fig. 6c, f). Despite that ATM activity was not compromised to an extent that abolishes 53BP1 and RIF1 recruitment, the impairment in ATM-dependent DDR signaling,


especially that of KAP1S824 phosphorylation, is substantial. Hence, we next considered the possibility that KAP1, the central factor of our screening approach, is itself of key importance


for how OTUD5 promotes efficient DNA repair activity at telomeres. KAP1 is known to be a heterochromatin scaffold protein that orchestrates the formation and maintenance of


heterochromatin46,47. At heterochromatic DSBs, KAP1 imposes a strong hindrance on DNA repair by maintenance of a compact, non-permissive chromatin state that deters DNA repair


factors17,18,19,48. ATM-dependent phosphorylation of KAP1 is essential to potentiate local chromatin decompaction to enable DNA repair, and ectopic overexpression of a KAP1S824D phosphomimic


mutant is able to fully restore DNA repair efficiency at heterochromatic DSBs that is crippled by ATM dysfunction. Although telomeres are not entirely heterochromatic by consent49,50, they


are enriched in heterochromatin-binding factors KAP1 and HP1 in both mouse and human cells51,52,53, and telomere deprotection strongly triggers KAP1S824 phosphorylation. To address the


significance of KAP1 in telomere NHEJ, we first individually, or in combination, depleted KAP1 and OTUD5 in TRF2ts MEFs and assessed telomere fusions upon telomere deprotection. Strikingly,


not only did depletion of KAP1 drastically enhance telomere fusions, but it also completely abolished the telomere fusion defect of OTUD5-deficient cells, indicating that OTUD5 facilitates


telomeric NHEJ in a KAP1-dependent manner (Fig. 5c, d, Supplementary Fig. 7a). To address the functional significance of KAP1S824 phosphorylation at deprotected telomeres, we ectopically


expressed the phosphomimic KAP1S824D mutant and assessed if this rescues the telomeric NHEJ defect of OTUD5-depleted cells. Indeed, overexpression of KAP1S824D restored telomere fusion


efficiency in OTUD5-depleted cells, while ectopic expression of wild-type (KAP1WT) failed to do so (Fig. 5e, f, Supplementary Fig. 7b, c). From this, we conclude that KAP1 and KAP1S824


phosphorylation play a prominent role in telomere NHEJ, and that OTUD5 facilitates telomere NHEJ by promoting ATM-dependent KAP1S824 phosphorylation. Moreover, ectopic expression of


phosphomimic KAP1S824D also rescued the telomere MMEJ defect in OTUD5-depleted _Trf1__F/F__;Trf2__F/F__;Ku70__−/−__;p53__−/−__;Cre-ER__T2_ MEFs (Fig. 5g, Supplementary Fig. 7d, e). Together,


these results indicate that the phosphorylation status of KAP1S824 is an important functional determinant for end-joining efficiency (both NHEJ and MMEJ) at dysfunctional telomeres and is


the critical node through which OTUD5 facilitates telomeric DNA repair. It has been reported that at DSBs in heterochromatic regions, where KAP1 is enriched, the phosphorylation of KAP1


promotes DNA repair by facilitating the displacement of the nucleosome remodeler CHD3, thereby triggering transient chromatin decompaction19. To test whether KAP1 phosphorylation promotes


telomeric DNA repair in a similar manner as it does at DSBs in heterochromatin, we quantified telomere fusions at deprotected telomeres in TRF2ts MEFs depleted of CHD3. However, unlike


ablation of KAP1, we did not observe an increase in telomere fusions in the absence of CHD3 (Supplementary Fig. 8a, b). Besides indicating that CHD3 is irrelevant to NHEJ of TRF2-deprotected


telomeres, this suggests either that KAP1 functions via a different, potentially redundant, chromatin remodeler at telomeres, or that the phosphorylation of KAP1 promotes NHEJ at telomeres


through a mechanism independent of CHD3 or chromatin decompaction. OTUD5 IS REQUIRED FOR THE EFFICIENT REPAIR OF IR-INDUCED DNA DAMAGE IN HETEROCHROMATIN Given the role of OTUD5 in DNA


repair at dysfunctional telomeres by promoting ATM-dependent KAP1S824 phosphorylation, and the established importance of KAP1S824 phosphorylation for repair of heterochromatic


DSBs17,18,19,48, we investigated whether OTUD5 also facilitates DSB repair at heterochromatin. Hereto we tracked the rate of resolution of IR-induced γH2A.X foci associated with


heterochromatic chromocenters in NIH3T3 cells17. After induction of DSBs by IR, over 90% of induced γH2A.X foci associated with chromocenters were resolved within 24 h (Fig. 6a–d,


Supplementary Fig. 8c). In contrast, and as demonstrated before17, cells pre-treated with ATM inhibitor were inefficient in resolving heterochromatin-associated γH2A.X foci, retaining 70% of


induced foci after 24 h of recovery. OTUD5-depleted cells also showed reduced resolution of γH2A.X foci, with ~40% of heterochromatin-associated foci remaining unresolved at 24 h after IR,


indicating inefficient repair of DSBs at heterochromatic regions. Thus, in addition to telomeres, OTUD5 also plays a role in facilitating DNA repair at heterochromatic regions, in line with


the here uncovered role of OTUD5 in regulating KAP1. DISCUSSION Ubiquitin system factors play a major role in orchestrating responses to DNA damage20. The DUB OTUD5 was previously shown to


suppress transcription at DNA breaks25. Using a novel screening approach, we now revealed OTUD5 as a prominent promotor of ATM activation and ATM-dependent KAP1 phosphorylation, in response


to both telomere deprotection and genome-wide DSBs. Moreover, we found that OTUD5 promotes DNA repair at deprotected telomeres and at heterochromatin. The role of OTUD5 in transcriptional


suppression has been ascribed to its ability to stabilize UBR5. Indeed, we confirm destabilization of UBR5 upon loss of OTUD5. As independently, UBR5 was found to inhibit the interaction


between ATM and ATMIN, a negative regulator of canonical DSB-induced ATM activation26, we hypothesized that OTUD5 may promote ATM-dependent DDR signaling via stabilization of UBR5. Indeed,


we found OTUD5 to promote ATM-dependent DDR signaling in a manner that is epistatic with UBR5, dependent on its catalytic activity and involving ATMIN, supporting that OTUD5 acts via UBR5.


Early biochemical studies performed on OTUD5 suggested that phosphorylation at residue Ser-177 is vital for its DUB activity in vitro24. Consistent with this, we showed that the


phospho-activation of OTUD5 is indispensable to its function in promoting DDR signaling, as phospho-dead OTUD5S177A failed to restore the pKAP1S824 level in OTUD5-depleted cells. This


demonstrates the biological significance of phospho-regulation to the functions of OTUD5 in vivo. To date, little is known about the kinases that phosphorylate OTUD5, apart from recent in


vivo evidence that mTOR kinase promotes OTUD5 stability and activity via direct phosphorylation of multiple sites on OTUD5. This in turns activates mTORC1/2, thereby constituting positive


feedback signaling54,55. It is conceivable that also in the DDR, an upstream kinase phosphorylates OTUD5 to amplify DDR signaling through a similar positive feedback mechanism. Of note,


OTUD5 contains multiple conserved SQ sites (S425, S497, S549) roughly within an SCD domain (≥3 S/TQ sites within 100 amino acids), which are features of ATM/ATR kinase substrates56,57. This


suggests the possibility that OTUD5 is directly phosphorylated by ATM to function as an amplifier of ATM-dependent DDR signaling in a potential positive-feedback circuit, resembling its role


in mTOR signaling. In the context of DNA repair, OTUD5 has been suggested to promote NHEJ by counteracting the degradation of Ku80, a core NHEJ factor, in certain cell lines39. Contrary to


that work, we find in MEFs and U2OS cells that Ku80 stability is not compromised in the absence of OTUD5. This indicates that in these cells OTUD5 is not a critical regulator of Ku stability


and that other DUBs, such as potentially UCHL3, are dominantly responsible for stabilizing Ku40. In line with this, we did not observe a significant defect in genome-wide NHEJ in


OTUD5-depleted cells, which would be expected from Ku70/80-deficient cells. Instead, we propose that OTUD5 facilitates DNA repair exclusively in genomic regions that are particularly


sensitive to ATM activity, such as telomeres and heterochromatin5,17, by promoting full DDR-induced ATM activity via the UBR5-ATMIN axis. Despite impaired ATM activation, recruitment of


53BP1 (and most of RIF1) to deprotected telomeres, an ATM-dependent process5, is unaffected in OTUD5-depleted cells. We reason that the loss of OTUD5 is insufficient to abolish all aspects


of ATM function, and that the residual ATM activity is sufficient to support 53BP1 (and RIF1) recruitment. This indicates also that the cause of the end-joining defect in OTUD5-deficient


cells lies downstream of the end-joining promoting activity of 53BP1. We demonstrate here that this is at the level of KAP1. We show that pKAP1S824, amongst other ATM targets, is an


especially sensitive and functionally important substrate that dictates NHEJ efficiency at telomeres. The sensitivity of KAP1 phosphorylation to disturbance in ATM activity is in line with


previous observations stating that KAP1 phosphorylation requires localized, concentrated ATM activity18, which may not be achieved in OTUD5-deficient cells. This localized, concentrated ATM


activity has been ascribed to hyper-accumulation of the MRN complex, mediated via the C-terminal BRCT-domain of 53BP118. Although 53BP1 recruitment seemed unperturbed, the reduced RIF1 foci


in one of the OTUD5 KO cell lines suggest incomplete phosphorylation of the 53BP1 N-terminus. It remains possible that incomplete ATM-dependent 53BP1 phosphorylation contributes to the


reduced KAP1 phosphorylation in OTUD5 deficient cells. Furthermore, telomere deprotection via TRF2 loss/inactivation strictly triggers ATM activation as ATR activation remains inhibited via


shelterin factor POT15. This precludes phosphorylation of ATM substrates via the ATR kinase, which has been shown to compensate for loss of ATM in global NHEJ58. This lack of compensatory


KAP1 phosphorylation via ATR adds to the exquisite sensitivity of telomeric NHEJ to disturbances in ATM activation. The need for robust KAP1 phosphorylation at deprotected telomeres is


evident in the impaired end-joining observed in OTUD5-deficient cells, where KAP1 phosphorylation is lacking and end-joining at deprotected telomeres is restored upon ectopic overexpression


of phosphomimic KAP1. The significance of KAP1S824 phosphorylation in telomeric DNA repair shares similarities with its previously reported function in DNA repair at heterochromatin16,17.


Different from euchromatin, a substantial challenge for DNA repair at heterochromatin is its compact, sterically non-permissive conformation. Without chromatin relaxation, it is difficult to


repair DNA lesions at heterochromatin, as access of DNA repair factors is restricted. Among ATM substrates, KAP1 is central to heterochromatic DNA repair as KAP1S824 phosphorylation is


necessary for the eviction of CHD3 and other heterochromatin factors from heterochromatin to achieve a relaxed conformation that is beneficial to DNA repair19. Ablation of KAP1 or CHD3, or


complementation with a phosphomimic KAP1S824D, is sufficient to bypass the requirement of ATM, demonstrating a pivotal function for KAP1S824 phosphorylation in facilitating heterochromatic


DNA repair, by promoting local chromatin decompaction. Our data here reveal that DNA repair at telomeres phenotypically resembles that of heterochromatin _prima facie_, in the sense that


KAP1S824 phosphorylation is required for efficient DNA repair in both genomic contexts. However, intriguingly, it has been reported that deprotected telomeres do not require, nor undergo,


chromatin decompaction for DNA repair to take place59,60, suggesting that KAP1S824 phosphorylation is unlikely to promote telomere end-joining through decompaction of telomeric chromatin. In


line with this notion, contrary to KAP1 depletion, loss of CHD3 did not elevate telomere fusion. This indicates that despite the mutual requirement of ample KAP1 phosphorylation for DNA


repair at heterochromatin and deprotected telomeres, the underlying mechanism is likely different for the two genomic contexts. Thus, the mechanism underlying the requirement of KAP1


phosphorylation at deprotected telomeres, is not immediately clear and requires further exploration in future studies. One interesting possibility to investigate relates to the ability of


both KAP1 and shelterin components TRF1 and TRF2 to interact with nuclear lamina components61,62,63,64. The interaction of KAP1 with nuclear lamina components has been postulated to


contribute to the transcriptional silencing of KAP1-associated heterochromatin, by tethering heterochromatin to the nuclear lamina62,65. In a similar fashion, KAP1 may tether telomeric DNA


to the nuclear lamina, in a way, involving transcriptional control or not, that negatively impacts telomeric DNA repair and is modulated by KAP1S824 phosphorylation. In addition, the role of


KAP1 in transcriptional silencing may also independently from nuclear lamina interactions affect DNA repair, given the increasing implications of RNA in DNA repair66. Alternatively, it is


possible that KAP1 phosphorylation serves to promote mobility of the damaged ends, as both deprotected telomeres and heterochromatic DSBs have been shown to have an increased mobility within


the nucleus. This increased mobility has been proposed to increase the chance of two free DNA ends reconnecting41,67. In addition to the facilitation of telomeric DNA repair by KAP1S824


phosphorylation, our data also indicates an unexpected protective role of KAP1 at dysfunctional telomeres by inhibiting NHEJ, as direct ablation of KAP1 considerably increased the frequency


of telomere fusion in the absence of TRF2 function. KAP1 and HP1 are heterochromatin scaffold proteins that are enriched at heterochromatic regions and serve as a recruitment hub for histone


methyltransferases, deacetylases, and chromatin remodelers that act in orchestration to promote the formation and maintenance of heterochromatic chromatin state. Despite the knowledge that


KAP1 and HP1 are enriched in the telomeric proteome in both human and mouse cells51,53, little is known about their functional roles at telomeres. Intriguingly, human telomeres were reported


to not be enriched in heterochromatic histone signatures like H3K9me3 or H3K27me349. Thus, it is enigmatic why these heterochromatin factors are present despite that there is no


heterochromatin to maintain. Our data on KAP1 implies the involvement of heterochromatin factors in the maintenance and protection of telomeres against unwarranted DNA repair. This provides


a plausible functional explanation for the presence of heterochromatin factors at telomeres. It is worth exploring through what mechanisms KAP1, and potentially HP1, inhibit telomeric DNA


repair, especially since KAP1 is frequently upregulated in cancers, with correlation to enhanced invasiveness and poor survival in patients68,69,70. Further understanding on the link between


KAP1 and telomeres may potentiate advancement of our understanding on telomere biology in cancers. Altogether, our work identifies an important role for OTUD5 in promoting DDR and


particularly KAP1S824 phosphorylation through an OTUD5-UBR5-ATMIN-ATM axis. Loss of OTUD5 confers a DNA repair defect most prevalent in regions enriched in KAP1, both at deprotected


telomeres and heterochromatic DSBs. In addition, our work points at an unanticipated role of KAP1 in suppressing toxic end-joining activity at dysfunctional telomeres, the mechanistic basis


of which is interesting to explore in future studies. METHODS CELL CULTURE HEK293T, U2OS and NIH3T3 cells were obtained from ATCC. _Trf2__−/−__;p53__−/−_;TRF2ts MEFs (TRF2ts MEFs) were


generated from _Trf2__flox/-_;_p53__−/−_ MEFs as previously described71. _Trf2__flox/-__;p53__−/−_ MEFs and _Trf1__F/F__;Trf2__F/F__;Ku70__−/−__;p53__−/−__;Cre-ER__T2_ MEFs were obtained


from T. de Lange10. HEK293T cells harboring the DSB-Spectrum_V1 reporter system were a gift from H. van Attikum38. HeLa cells with doxycycline-inducible shTRF2 were a gift from J. Lingner36.


All cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal calf serum (FCS, Sigma), 100 U ml−1 penicillin, 100 μg ml−1 streptomycin, and 2 mM


l-Glutamine (Gibco, Life Technologies), at 37 °C in a humidified atmosphere with 5% CO2, except for TRF2ts MEFs, which were cultured at 32 °C. All cell lines were routinely tested for


mycoplasma contamination and scored negatively. FACS-BASED DETECTION OF PKAP1S824 TRF2ts MEFs were trypsinized and resuspended in ice cold PBS/10% FCS, centrifuged at 268 × _g_ for 5 min at


4 °C. Cells pellets were resuspended in 0.5 ml ice-cold PBS and fixed while vortexing, by adding dropwise 4.5 ml of ice-cold 100% methanol. Cells were then incubated overnight at −20 °C


before starting the staining. After adding 5 ml of PBS, fixed cells were centrifuged for 5 min at 420 × _g_. Cell pellets were resuspended in 500 µl PBS/0.5% BSA (PBA) and cells were


permeabilized with 0.1% Triton X-100/PBA on ice for 15 min, followed by 3 washes with PBA and incubation with anti-pKAP1S824 antibody (1:250; Bethyl A300-767A) for 3 h at room temperature.


After 3 washes with PBA cells were incubated with anti-rabbit Alexa 647 (1:5000, Invitrogen A21246) for 1 h at room temperature. Cells were washed 3 times in PBA and finally resuspended in


PBA. FACS was performed with the LSRFortessa Cell Analyzer (BD). FACS data were analyzed by FlowJo 10.4.2 software. ATD SCREEN For the ATD screen, a tailored lentiviral shRNA library


targeting 123 ubiquitin system factors was assembled from the Mission shRNA library (Sigma). A full list of shRNAs is included in Supplementary Data 2. Infected cells were selected by


puromycin, split into triplicates that were each cultured at 39 °C for 3 h and stained for pKAP1S824 as described above. The total G1 cell population was FACS-sorted for the 5% cells with


the lowest pKAP1 signal, from which the shRNA enrichment was determined relative to the untreated control cell population. Genomic DNA (gDNA) was extracted from the sorted cells using the


DNeasy Blood & Tissue Kit (Qiagen). A 2-step PCR recovery of shRNA sequences from gDNA was performed using the Phusion polymerase kit (NEB): PCR 1: Thermocycling conditions: (1) 98 °C


for 30 s; (2) 20 cycles of 98 °C for 30 s, 60 °C for 30 s, 72 °C for 1 min; (3) 72 °C for 5 min; (4) hold at 4 °C. Using PCR products from PCR 1 as template for PCR 2: (1) 98 °C for 30 s;


(2) 15 cycles of 98 °C for 30 s, 60 °C for 30 s, 72 °C for 1 min; (3) 72 °C for 5 min; (4) hold at 4 °C. For primers used in PCR 1 and 2, see Supplementary Data 3. PCR products from PCR 2


were purified using the Wizard® SV Gel and PCR Clean-Up System (Promega). Purified PCR products were subsequently sequenced using Illumina Hiseq 2500. shRNA enrichment and gene ranking were


done using MAGeCK-RRA analysis30. LENTIVIRAL EXPRESSION CONSTRUCTS AND LENTIVIRAL PACKAGING Expression constructs of OTUD5 and KAP1 were used for complementation/rescue experiments. For


OTUD5, full-length cDNA of human OTUD5, codon-optimized, was cloned into pCDH-Puro lentiviral expression vector by NheI-EcoRI restriction cloning. Sequence of the codon-optimized cDNA of


OTUD5 is included in Supplementary Fig. 5. For KAP1, the expression construct in pLX304-BLAST vector was obtained from the CCSB-Broad Lentiviral Expression Library72. Lentiviral particles


were packaged as described previously73, by co-transfecting HEK293T cell with the lentiviral transfer constructs and lentivirus packaging vectors pRRE, pRSV-Rev, pMD2.G. Medium was refreshed


once at 16 h post transfection. Lentivirus-containing supernatant was collected at 24–36 h after medium was refreshed. SITE-DIRECTED MUTAGENESIS Mutants of OTUD5 (S177A and C224S) and KAP1


(S824D) were generated by site-directed mutagenesis reactions on pCDH-OTUD5 and pLX304-KAP1 respectively, using the QuikChange II XL directed mutagenesis kit (200521, Agilent) according to


manufacturer’s instructions. Primers containing the desired mutations were designed using the web based QuikChange Primer Design Program (www.agilent.com/genomics/qcpd). PCR’s cycling


parameters were as follows: (1) 95 °C for 1 min; (2) 18 cycles of 95 °C for 50 s, 62 °C for 50 s, 68 °C for 12 min; (3) 68 °C for 7 min; (4) hold at 4 °C. The PCR product was treated with 20


U of DpnI enzyme for 2 h at 37 °C to digest the parental/template DNA. The DpnI-treated PCR mix was transformed into Stbl3 _Escherichia coli_ (C737303, Thermo Fisher Scientific) following


manufacturer’s instructions. Transformed cultures were incubated overnight at 37 °C. Mutations were confirmed by Sanger sequencing. CRISPR/CAS9 GENE EDITING For CRISPR/Cas9-mediated gene


knockout, sgRNA sequences against OTUD5, UBR5, and KAP1 were designed using the Broad Institute’s GPP sgRNA Designer


(https://portals.broadinstitute.org/gpp/public/analysis-tools/sgrna-design). sgRNA sequences were cloned into a LentiCRISPRv2 plasmid according to standard protocols33. Lentiviral particles


are packaged as described above73. The sgRNA sequences used are included in Supplementary Data 4. LentiCRISPRv2 was a gift from Feng Zhang (Addgene plasmid #52961)72. RNA INTERFERENCE For


RNA interference by shRNAs, TRF2ts MEFs were transduced with pLKO.1-puro shRNA lentiviruses from Mission library clones (Sigma). shRNA sequences used for the knockdown of UBE2D3, UBE2M and


OTUD5, and their respective Mission library numbers, are included in Supplementary Data 4. METAPHASE CHROMOSOME ANALYSIS FOR TELOMERE FUSIONS Cell collection, preparation of metaphase


spreads and telomere FISH with a FITC-OO-(CCCTAA)3 peptide nucleic acid custom probe (Biosynthesis) or Alexa488-labeled C-rich Telomere Probe (Eurogentec) for metaphase chromosome analysis


was done as described27. Digital images of metaphases were captured using the Metafer4/MSearch automated metaphase finder system (MetaSystems) equipped with an AxioImager Z2 microscope (Carl


Zeiss). After scanning metaphase preparations at x10 magnification, high-resolution images of metaphases were acquired using a ‘Plan-Apochromat’ ×63/1.40 oil objective. Chromosome fusions


were quantified from >1500 chromosomes per experimental condition. CO-IMMUNOPRECIPITATION HEK293T cells were transfected with 10 µg of pLVU-GFP-OTUD5 using polyethylenimine. At 16 h post


transfection, medium was refreshed. At 48 h post transfection HEK293T cells were lysed in 500 µL of Co-IP Lysis Buffer: 20 mM Tris-HCl pH 7.5, 100 mM NaCl, 1% Nonidet P-40, 5% glycerol, 1 mM


EDTA, protease inhibitors (Roche). To complete lysis, cells were incubated for 30 min at 4 °C in a rotating wheel. Lysates were centrifuged for 10 min at 13,523 × _g_ and supernatants were


collected. Protein concentration was quantified using Pierce BCA assay (Thermo Fisher Scientific) and lysate containing 2 mg of protein was incubated with 15 µL of washed GFP-Trap® magnetic


agarose beads (ChromoTek) overnight at 4 °C in a rotating wheel. Next, beads were washed 3× for 5 min with lysis buffer. For western blot, protein was eluted from beads with 40 µL of 2× SDS


sample buffer and incubating at 95 °C for 5 min. IMMUNOBLOTTING Immunoblotting was done according to standard protocols and as described before29. Briefly, cells were washed with PBS and


collected by scraping in 2× SDS sample buffer followed by boiling at 95 °C. The lysates obtained were sonicated at 30% amplitude for 10 s. Protein concentrations were measured using Pierce


BCA Protein Assay (Thermo Fisher Scientific) and equal amounts of protein were loaded onto precast 4–12% Bis-Tris gels (Invitrogen). After protein transfer at 100 V for 60 min onto 0.45 μm


nitrocellulose membranes (Amersham), membranes were incubated in blocking buffer and subsequently primary antibodies O/N at 4 °C. Primary antibodies and respective working concentrations are


included in Supplementary Data 5. Membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (Invitrogen), for detection using enhanced chemiluminescence


(SuperSignal West Pico PLUS, Thermo Fisher Scientific) on a Syngene G:BOX, or with IRDye800CW- and IRDye680-labeled secondary antibodies (Li-COR) for detection on the Odyssey CLx Infrared


imager (LI-COR). Blots were analyzed using either the GeneSys software (version 1.6.9.0) or the Image Studio Lite LI-COR software (version 5.2.5). IMMUNOFLUORESCENCE For immunofluorescence,


cells were seeded on 15 mm ø coverslips in 12-well plates and treated as indicated in the individual experiments. Cells were washed with PBS, fixed for 10 min with 2% paraformaldehyde


followed by 10 min permeabilization in 0.5% Triton/PBS. After 1 h of blocking (0.02% Triton, 5% NGS, 5% FCS in PBS), cells were incubated with primary antibody diluted with blocking solution


overnight at 4 °C in a humid chamber. Primary antibodies and respective concentrations used are included in Supplementary Data 5. Next, cells were washed three times with 0.02% Triton/PBS


and incubated with Alexa Fluor 488, 568 or 647 goat anti-mouse or anti-rabbit IgG secondary antibodies (Invitrogen) in blocking solution for 1 h at room temperature in the dark. After


washing three times with 0.02% Triton/PBS, slides were mounted using ProLong Gold Antifade Mountant with DAPI (Invitrogen). Slides were imaged using a LSM 980 confocal with Airyscan2 system


with ×63 oil objective and ZEN software. Image analysis for foci co-localization was performed with the publicly available Foci Analyzer 1.3 macro


(https://github.com/BioImaging-NKI/Foci-analyzer) on ImageJ software. Briefly, maximum intensity projections were first produced from Z-stack images. Nuclei were automatically defined using


the “Stardist nuclei segmentation”. The two channels where the foci were captured were selected for co-localization analysis. Specifically, for the γH2AX-chromocenter co-localization


experiment (Fig. 6), the DAPI channel was selected as one of the two channels for co-localization analysis, “Foci size” was set to “large”, to enable the detection of chromocenters as foci.


RANDOM PLASMID INTEGRATION ASSAY For random plasmid integration assays, 500,000 U2OS cells were seeded in 6 cm dishes 24 h before transfection. With the use of Lipofectamine 2000 (Thermo


Fisher Scientific) cells were transfected with 3 μg of EcoRI/BamHI-linearized peGFP-C1 plasmid that confers G418 resistance upon successful integration. Transfected cells were trypsinized


the following day and seeded in 10 cm dishes at 1000 cells per dish for –G418 controls and 20,000 cells per dish for treatment with G418. Simultaneously, 20,000 cells per sample were seeded


separately for assessment of transfection efficiency by the % GFP-positive cells using FACS. Selection was initiated the following day with G418/Geneticin at 500 µg/ml. At 10–14 days after


seeding, cells were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet. Dishes were scanned and colony counting was performed using ImageJ (1.52p). NHEJ scores were


calculated by normalizing the number of colonies in +G418 dishes to the plating efficiency (-G418) and transfection efficiency (% GFP). DSB-SPECTRUM_V1 REPORTER The DSB-Spectrum_V1 reporter


system was used to assess the NHEJ-HR balance as described38. Briefly, DSB-Spectrum_V1 reporter HEK293T cells were seeded at 300,000 cells/well in 12-well plates. The next day, cells were


transfected wit pX459-Cas9-sgRNA-mCherry using Lipofectamine 2000 (Invitrogen), according to the manufacturer’s protocol. At 48 h post transfection, cells were trypsinized and analyzed by


FACS for the % BFP (NHEJ) and % GFP (HR) of transfected (mCherry+) cells using an LSRFortessa Cell Analyzer (BD). STATISTICAL ANALYSIS AND REPRODUCIBILITY Statistical analyses were performed


using GraphPad Prism (9.2.0) and Microsoft Excel 2016 (16.0.5356.1000). Details on data representation, statistical tests and number of replicate experiments are indicated in the respective


figure legends. Exact p-values are indicated in the figures. REPORTING SUMMARY Further information on research design is available in the Nature Portfolio Reporting Summary linked to this


article. DATA AVAILABILITY All data generated or analyzed during this study are included in this article and its supplementary information files and are available from the corresponding


author upon request. Source data are provided with this paper. REFERENCES * Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. _Cell_ 144, 646–674 (2011). Article 


CAS  PubMed  Google Scholar  * Maciejowski, J. & de Lange, T. Telomeres in cancer: tumour suppression and genome instability. _Nat. Rev. Mol. Cell Biol._ 18, 175–186 (2017). Article  CAS


  PubMed Central  PubMed  Google Scholar  * Shay, J. W. & Wright, W. E. Hayflick, his limit, and cellular ageing. _Nat. Rev. Mol. Cell Biol._ 1, 72–76 (2000). Article  CAS  PubMed 


Google Scholar  * Karlseder, J., Smogorzewska, A. & de Lange, T. Senescence induced by altered telomere state, not telomere loss. _Science_ 295, 2446–2449 (2002). Article  ADS  CAS 


PubMed  Google Scholar  * Denchi, E. L. & de Lange, T. Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1. _Nature_ 448, 1068–1071 (2007). Article  ADS 


CAS  PubMed  Google Scholar  * Bae, N. S. & Baumann, P. A RAP1/TRF2 complex inhibits nonhomologous end-joining at human telomeric DNA ends. _Mol. Cell_ 26, 323–334 (2007). Article  CAS 


PubMed  Google Scholar  * Lazzerini-Denchi, E. & Sfeir, A. Stop pulling my strings - what telomeres taught us about the DNA damage response. _Nat. Rev. Mol. Cell Biol._ 17, 364–378


(2016). Article  CAS  PubMed Central  PubMed  Google Scholar  * van Steensel, B., Smogorzewska, A. & de Lange, T. TRF2 protects human telomeres from end-to-end fusions. _Cell_ 92,


401–413 (1998). Article  PubMed  Google Scholar  * Stansel, R. M., de Lange, T. & Griffith, J. D. T-loop assembly in vitro involves binding of TRF2 near the 3’ telomeric overhang. _EMBO


J._ 20, 5532–5540 (2001). Article  CAS  PubMed Central  PubMed  Google Scholar  * Sfeir, A. & de Lange, T. Removal of shelterin reveals the telomere end-protection problem. _Science_


336, 593–597 (2012). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar  * Uziel, T. et al. Requirement of the MRN complex for ATM activation by DNA damage. _EMBO J._ 22, 5612–5621


(2003). Article  CAS  PubMed Central  PubMed  Google Scholar  * Blackford, A. N. & Jackson, S. P. ATM, ATR, and DNA-PK: the trinity at the heart of the DNA damage response. _Mol. Cell_


66, 801–817 (2017). Article  CAS  PubMed  Google Scholar  * Banin, S. et al. Enhanced phosphorylation of p53 by ATM in response to DNA damage. _Science_ 281, 1674–1677 (1998). Article  ADS 


CAS  PubMed  Google Scholar  * Matsuoka, S. et al. Ataxia telangiectasia-mutated phosphorylates Chk2 in vivo and in vitro. _Proc. Natl Acad. Sci. USA_ 97, 10389–10394 (2000). Article  ADS 


CAS  PubMed Central  PubMed  Google Scholar  * Burma, S., Chen, B. P., Murphy, M., Kurimasa, A. & Chen, D. J. ATM phosphorylates histone H2AX in response to DNA double-strand breaks. _J.


Biol. Chem._ 276, 42462–42467 (2001). Article  CAS  PubMed  Google Scholar  * Ziv, Y. et al. 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 (2006). Article  CAS  PubMed  Google Scholar  * Goodarzi, A. A. et al. ATM signaling facilitates repair of DNA double-strand breaks


associated with heterochromatin. _Mol. Cell_ 31, 167–177 (2008). Article  CAS  PubMed  Google Scholar  * Noon, A. T. et al. 53BP1-dependent robust localized KAP-1 phosphorylation is


essential for heterochromatic DNA double-strand break repair. _Nat. Cell Biol._ 12, 177–184 (2010). Article  CAS  PubMed  Google Scholar  * Goodarzi, A. A., Kurka, T. & Jeggo, P. A.


KAP-1 phosphorylation regulates CHD3 nucleosome remodeling during the DNA double-strand break response. _Nat. Struct. Mol. Biol._ 18, 831–839 (2011). Article  CAS  PubMed  Google Scholar  *


Schwertman, P., Bekker-Jensen, S. & Mailand, N. Regulation of DNA double-strand break repair by ubiquitin and ubiquitin-like modifiers. _Nat. Rev. Mol. Cell Biol._ 17, 379–394 (2016).


Article  CAS  PubMed  Google Scholar  * Mailand, N. et al. RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins. _Cell_ 131, 887–900 (2007).


Article  CAS  PubMed  Google Scholar  * Doil, C. et al. RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins. _Cell_ 136, 435–446


(2009). Article  CAS  PubMed  Google Scholar  * Poulsen, M., Lukas, C., Lukas, J., Bekker-Jensen, S. & Mailand, N. Human RNF169 is a negative regulator of the ubiquitin-dependent


response to DNA double-strand breaks. _J. Cell Biol._ 197, 189–199 (2012). Article  CAS  PubMed Central  PubMed  Google Scholar  * Huang, O. W. et al. Phosphorylation-dependent activity of


the deubiquitinase DUBA. _Nat. Struct. Mol. Biol._ 19, 171–175 (2012). Article  CAS  PubMed  Google Scholar  * de Vivo, A. et al. The OTUD5-UBR5 complex regulates FACT-mediated transcription


at damaged chromatin. _Nucleic Acids Res._ 47, 729–746 (2019). Article  PubMed  Google Scholar  * Zhang, T., Cronshaw, J., Kanu, N., Snijders, A. P. & Behrens, A. UBR5-mediated


ubiquitination of ATMIN is required for ionizing radiation-induced ATM signaling and function. _Proc. Natl Acad. Sci. USA_ 111, 12091–12096 (2014). Article  ADS  CAS  PubMed Central  PubMed


  Google Scholar  * Konishi, A. & de Lange, T. Cell cycle control of telomere protection and NHEJ revealed by a ts mutation in the DNA-binding domain of TRF2. _Genes Dev._ 22, 1221–1230


(2008). Article  CAS  PubMed Central  PubMed  Google Scholar  * Sakaue-Sawano, A. & Miyawaki, A. Visualizing spatiotemporal dynamics of multicellular cell-cycle progressions with fucci


technology. _Cold Spring Harb. Protoc._ 2014, https://doi.org/10.1101/pdb.prot080408 (2014). * Boersma, V. et al. MAD2L2 controls DNA repair at telomeres and DNA breaks by inhibiting 5’ end


resection. _Nature_ 521, 537–540 (2015). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar  * Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale


CRISPR/Cas9 knockout screens. _Genome Biol._ 15, 554 (2014). Article  PubMed Central  PubMed  Google Scholar  * Yalcin, Z. et al. UBE2D3 facilitates NHEJ by orchestrating ATM signalling


through multi-level control of RNF168. _Nat. Commun._ 15, 5032 (2024). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar  * Enchev, R. I., Schulman, B. A. & Peter, M. Protein


neddylation: beyond cullin-RING ligases. _Nat. Rev. Mol. Cell Biol._ 16, 30–44 (2015). Article  CAS  PubMed Central  PubMed  Google Scholar  * Sanjana, N. E., Shalem, O. & Zhang, F.


Improved vectors and genome-wide libraries for CRISPR screening. _Nat. Methods_ 11, 783–784 (2014). Article  CAS  PubMed Central  PubMed  Google Scholar  * Smogorzewska, A., Karlseder, J.,


Holtgreve-Grez, H., Jauch, A. & de Lange, T. DNA ligase IV-dependent NHEJ of deprotected mammalian telomeres in G1 and G2. _Curr. Biol._ 12, 1635–1644 (2002). Article  CAS  Google


Scholar  * Celli, G. B., Denchi, E. L. & de Lange, T. Ku70 stimulates fusion of dysfunctional telomeres yet protects chromosome ends from homologous recombination. _Nat. Cell Biol._ 8,


885–890 (2006). Article  Google Scholar  * Grolimund, L. et al. A quantitative telomeric chromatin isolation protocol identifies different telomeric states. _Nat. Commun._ 4, 2848 (2013).


Article  ADS  Google Scholar  * Ahnesorg, P., Smith, P. & Jackson, S. P. XLF interacts with the XRCC4-DNA ligase IV complex to promote DNA nonhomologous end-joining. _Cell_ 124, 301–313


(2006). Article  CAS  Google Scholar  * van de Kooij, B., Kruswick, A., van Attikum, H. & Yaffe, M. B. Multi-pathway DNA-repair reporters reveal competition between end-joining,


single-strand annealing and homologous recombination at Cas9-induced DNA double-strand breaks. _Nat. Commun._ 13, 5295 (2022). Article  ADS  PubMed Central  Google Scholar  * Li, F. et al.


The deubiquitinase OTUD5 regulates Ku80 stability and non-homologous end joining. _Cell Mol. Life Sci._ 76, 3861–3873 (2019). Article  CAS  PubMed Central  Google Scholar  * Nishi, R. et al.


The deubiquitylating enzyme UCHL3 regulates Ku80 retention at sites of DNA damage. _Sci. Rep._ 8, 17891 (2018). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar  * Dimitrova, N.,


Chen, Y. C., Spector, D. L. & de Lange, T. 53BP1 promotes non-homologous end joining of telomeres by increasing chromatin mobility. _Nature_ 456, 524–528 (2008). Article  ADS  CAS 


PubMed Central  PubMed  Google Scholar  * Zimmermann, M., Lottersberger, F., Buonomo, S. B., Sfeir, A. & de Lange, T. 53BP1 regulates DSB repair using Rif1 to control 5’ end resection.


_Science_ 339, 700–704 (2013). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar  * Chapman, J. R. et al. RIF1 is essential for 53BP1-dependent nonhomologous end joining and


suppression of DNA double-strand break resection. _Mol. Cell_ 49, 858–871 (2013). Article  CAS  PubMed Central  PubMed  Google Scholar  * Di Virgilio, M. et al. Rif1 prevents resection of


DNA breaks and promotes immunoglobulin class switching. _Science_ 339, 711–715 (2013). Article  ADS  PubMed  Google Scholar  * Escribano-Diaz, C. et al. A cell cycle-dependent regulatory


circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. _Mol. Cell_ 49, 872–883 (2013). Article  CAS  PubMed  Google Scholar  * Friedman, J. R. et al. KAP-1, a


novel corepressor for the highly conserved KRAB repression domain. _Genes Dev._ 10, 2067–2078 (1996). Article  CAS  PubMed  Google Scholar  * Moosmann, P., Georgiev, O., Le Douarin, B.,


Bourquin, J. P. & Schaffner, W. Transcriptional repression by RING finger protein TIF1 beta that interacts with the KRAB repressor domain of KOX1. _Nucleic Acids Res._ 24, 4859–4867


(1996). Article  CAS  PubMed Central  PubMed  Google Scholar  * White, D. et al. The ATM substrate KAP1 controls DNA repair in heterochromatin: regulation by HP1 proteins and serine 473/824


phosphorylation. _Mol. Cancer Res._ 10, 401–414 (2012). Article  CAS  PubMed  Google Scholar  * Cubiles, M. D. et al. Epigenetic features of human telomeres. _Nucleic Acids Res._ 46,


2347–2355 (2018). Article  CAS  PubMed Central  PubMed  Google Scholar  * Garcia-Cao, M., O’Sullivan, R., Peters, A. H., Jenuwein, T. & Blasco, M. A. Epigenetic regulation of telomere


length in mammalian cells by the Suv39h1 and Suv39h2 histone methyltransferases. _Nat. Genet._ 36, 94–99 (2004). Article  CAS  PubMed  Google Scholar  * Bartocci, C. et al. Isolation of


chromatin from dysfunctional telomeres reveals an important role for Ring1b in NHEJ-mediated chromosome fusions. _Cell Rep._ 7, 1320–1332 (2014). Article  CAS  PubMed Central  PubMed  Google


Scholar  * Dejardin, J. & Kingston, R. E. Purification of proteins associated with specific genomic Loci. _Cell_ 136, 175–186 (2009). Article  CAS  PubMed Central  PubMed  Google


Scholar  * Lin, C. G. et al. The human telomeric proteome during telomere replication. _Nucleic Acids Res._ 49, 12119–12135 (2021). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar


  * Cho, J. H. et al. Deubiquitinase OTUD5 is a positive regulator of mTORC1 and mTORC2 signaling pathways. _Cell Death Differ._ 28, 900–914 (2021). Article  CAS  PubMed  Google Scholar  *


Hou, T. et al. Deubiquitinase OTUD5 modulates mTORC1 signaling to promote bladder cancer progression. _Cell Death Dis._ 13, 778 (2022). Article  CAS  PubMed Central  PubMed  Google Scholar 


* Kim, S. T., Lim, D. S., Canman, C. E. & Kastan, M. B. Substrate specificities and identification of putative substrates of ATM kinase family members. _J. Biol. Chem._ 274, 37538–37543


(1999). Article  CAS  PubMed  Google Scholar  * Traven, A. & Heierhorst, J. SQ/TQ cluster domains: concentrated ATM/ATR kinase phosphorylation site regions in DNA-damage-response


proteins. _Bioessays_ 27, 397–407 (2005). Article  CAS  PubMed  Google Scholar  * Schlam-Babayov, S. et al. Phosphoproteomics reveals novel modes of function and inter-relationships among


PIKKs in response to genotoxic stress. _EMBO J._ 40, e104400 (2021). Article  CAS  PubMed  Google Scholar  * Vancevska, A., Douglass, K. M., Pfeiffer, V., Manley, S. & Lingner, J. The


telomeric DNA damage response occurs in the absence of chromatin decompaction. _Genes Dev._ 31, 567–577 (2017). Article  CAS  PubMed Central  PubMed  Google Scholar  * Timashev, L. A.,


Babcock, H., Zhuang, X. & de Lange, T. The DDR at telomeres lacking intact shelterin does not require substantial chromatin decompaction. _Genes Dev._ 31, 578–589 (2017). Article  PubMed


Central  PubMed  Google Scholar  * Kubben, N. et al. Identification of differential protein interactors of lamin A and progerin. _Nucleus_ 1, 513–525 (2010). Article  PubMed Central  PubMed


  Google Scholar  * Neumann-Staubitz, P., Kitsberg, D., Buxboim, A. & Neumann, H. A method to map the interaction network of the nuclear lamina with genetically encoded


photo-crosslinkers in vivo. _Front. Chem._ 10, 905794 (2022). Article  ADS  CAS  PubMed Central  PubMed  Google Scholar  * Crabbe, L., Cesare, A. J., Kasuboski, J. M., Fitzpatrick, J. A.


& Karlseder, J. Human telomeres are tethered to the nuclear envelope during postmitotic nuclear assembly. _Cell Rep._ 2, 1521–1529 (2012). Article  CAS  PubMed Central  PubMed  Google


Scholar  * Wood, A. M., Laster, K., Rice, E. L. & Kosak, S. T. A beginning of the end: new insights into the functional organization of telomeres. _Nucleus_ 6, 172–178 (2015). Article 


CAS  PubMed Central  PubMed  Google Scholar  * Gonzalez-Sandoval, A. & Gasser, S. M. On TADs and LADs: spatial control over gene expression. _Trends Genet._ 32, 485–495 (2016). Article 


CAS  PubMed  Google Scholar  * Zong, D., Oberdoerffer, P., Batista, P. J. & Nussenzweig, A. RNA: a double-edged sword in genome maintenance. _Nat. Rev. Genet._ 21, 651–670 (2020).


Article  CAS  PubMed  Google Scholar  * Lottersberger, F., Karssemeijer, R. A., Dimitrova, N. & de Lange, T. 53BP1 and the LINC complex promote microtubule-dependent DSB mobility and DNA


repair. _Cell_ 163, 880–893 (2015). Article  CAS  PubMed Central  PubMed  Google Scholar  * Hu, M. et al. Expression of KAP1 in epithelial ovarian cancer and its correlation with


drug-resistance. _Int. J. Clin. Exp. Med._ 8, 17308–17320 (2015). CAS  PubMed Central  PubMed  Google Scholar  * Cui, Y. et al. High levels of KAP1 expression are associated with aggressive


clinical features in ovarian cancer. _Int. J. Mol. Sci._ 16, 363–377 (2014). Article  PubMed Central  PubMed  Google Scholar  * Yokoe, T. et al. KAP1 is associated with peritoneal


carcinomatosis in gastric cancer. _Ann. Surg. Oncol._ 17, 821–828 (2010). Article  PubMed  Google Scholar  * Peuscher, M. H. & Jacobs, J. J. DNA-damage response and repair activities at


uncapped telomeres depend on RNF8. _Nat. Cell Biol._ 13, 1139–1145 (2011). Article  CAS  PubMed  Google Scholar  * Yang, X. et al. A public genome-scale lentiviral expression library of


human ORFs. _Nat. Methods_ 8, 659–661 (2011). Article  CAS  PubMed Central  PubMed  Google Scholar  * de Krijger, I. et al. MAD2L2 dimerization and TRIP13 control shieldin activity in DNA


repair. _Nat. Commun._ 12, 5421 (2021). Article  ADS  PubMed Central  PubMed  Google Scholar  Download references ACKNOWLEDGEMENTS We thank Titia de Lange for TRF2I468A expression vector and


_Trf2_flox/-;_p53_−/− MEFs, used to generate TRF2ts MEFs, and for _Trf1__F/F__;Trf2__F/F__;Ku70__−/−__;p53__−/−__;Cre-ER__T2_ MEFs. We thank Haico van Attikum and Bert van der Kooij for


HEK293T cells with the DSB-Spectrum_V1 reporter, Joachim Lingner for the HeLa shTRF2 cells, Thomas Kuilman for MAGeCK-RRA analysis of the screen results, Daniëlle Koot for assistance with


IF-FISH and Brigitte Wevers for contributing to initial optimization of the pKAP1 screen. This work was supported by the European Union’s Horizon 2020 research and innovation program


(Marie-Skłodowska-Curie grant agreement 812829) to J.J.L.J, Dutch Cancer Society project grant KWF 2019-2/12826 to J.J.L.J., and an institutional grant of the Dutch Cancer Society and the


Dutch Ministry of Health, Welfare and Sport to the Netherlands Cancer Institute. AUTHOR INFORMATION Author notes * These authors contributed equally: Shiu Yeung Lam, Ruben van der Lugt.


AUTHORS AND AFFILIATIONS * Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands Shiu Yeung Lam, Ruben van der Lugt, Aurora Cerutti, Zeliha Yalçin, Alexander


M. Thouin, Marco Simonetta & Jacqueline J. L. Jacobs Authors * Shiu Yeung Lam View author publications You can also search for this author inPubMed Google Scholar * Ruben van der Lugt


View author publications You can also search for this author inPubMed Google Scholar * Aurora Cerutti View author publications You can also search for this author inPubMed Google Scholar *


Zeliha Yalçin View author publications You can also search for this author inPubMed Google Scholar * Alexander M. Thouin View author publications You can also search for this author inPubMed


 Google Scholar * Marco Simonetta View author publications You can also search for this author inPubMed Google Scholar * Jacqueline J. L. Jacobs View author publications You can also search


for this author inPubMed Google Scholar CONTRIBUTIONS J.J.L.J. conceived the original idea for the screen and together with S.Y.L. and R.v.d.L. conceived the ideas for follow-up work. M.S.


constructed the shRNA library and A.C. conducted the screen. S.Y.L. and R.v.d.L. performed most experiments, Z.Y. assessed telomere fusion upon UBR5 knockdown and A.T. conducted


DSB-spectrum_V1 reporter assays. J.J.L.J. and S.Y.L. co-wrote the manuscript, to which R.v.d.L. contributed with comments, composing part of the figures and responding to peer reviewers.


CORRESPONDING AUTHOR Correspondence to Jacqueline J. L. Jacobs. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. PEER REVIEW PEER REVIEW INFORMATION


_Nature Communications_ thanks Younghoon Kee, Tej Pandita, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. A peer review file is available.


ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. SUPPLEMENTARY INFORMATION


SUPPLEMENTARY INFORMATION DESCRIPTION OF ADDITIONAL SUPPLEMENTARY FILES SUPPLEMENTARY DATA 1-5 REPORTING SUMMARY PEER REVIEW FILE SOURCE DATA SOURCE DATA RIGHTS AND PERMISSIONS OPEN ACCESS


This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and


reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you


modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in


this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative


Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a


copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Lam, S.Y., van der Lugt, R., Cerutti, A. _et al._


OTUD5 promotes end-joining of deprotected telomeres by promoting ATM-dependent phosphorylation of KAP1S824. _Nat Commun_ 15, 8960 (2024). https://doi.org/10.1038/s41467-024-53404-0 Download


citation * Received: 15 December 2023 * Accepted: 08 October 2024 * Published: 17 October 2024 * DOI: https://doi.org/10.1038/s41467-024-53404-0 SHARE THIS ARTICLE Anyone you share the


following link with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer


Nature SharedIt content-sharing initiative


Trending News

Bargain hunt star shares his dismay over 'sad act' after troubling break-in

An auction house owned by Bargain Hunt star Charles Hanson was broken into overnight leaving him fuming. Officers were c...

Grange up for grabs as farm business changes - farmers weekly

14 JUNE 2002 ------------------------- GRANGE UP FOR GRABS AS FARM BUSINESS CHANGES By Andrew Shirley ACROSS England a n...

Vic news - 9news - latest updates and breaking headlines victoria

VICTORIAN PRINCIPALS GRANTED SWEEPING NEW EXPULSION POWERS Principals in Victoria are being given extraordinary new powe...

Otud5 promotes end-joining of deprotected telomeres by promoting atm-dependent phosphorylation of kap1s824

ABSTRACT Appropriate repair of damaged DNA and the suppression of DNA damage responses at telomeres are essential to pre...

Jacksonville port tonnage up 5%

Jacksonville port tonnage up 5%    The Jacksonville Port Authority said terminals under its jurisdiction handled 5 perce...

Latests News

404 - Page not found

HomeNG HindiIndiaIndiaAndhra PradeshArunachal PradeshAssamBiharChhattisgarhGoaGujaratHaryanaHimachal PradeshJharkhandKar...

‘healthcare policies must be robust, and protect both staff and patients’

Ken Spearpoint comments on the case of a nurse cautioned by the Nursing and Midwifery Council for not trying to resuscit...

Nina Dobrev shows off injured leg following bike accident

The “Vampire Diaries” star, who was involved in a horrific bike accident last month, took to Instagram over the weekend ...

Improving upper limb function after stroke | Nursing Times

Upper limb weakness following stroke has a negative impact on daily living and quality of life but the evidence for inte...

Should I Worry About My Chronic Cough?

James Yates Facebook Twitter LinkedIn I have been coughing for several months. Why do I cough so much? Should I be worri...

Top