Maintaining stability of the genome requires dedicated DNA repair and signalling processes that are essential for the faithful duplication and propagation of chromosomes. These DNA damage response (DDR) mechanisms counteract the potentially mutagenic impact of daily genotoxic stresses from both exogenous and endogenous sources. Inherent to these DNA repair pathways is the activity of protein factors that instigate repair processes in response to DNA lesions. The regulation, coordination, and orchestration of these DDR factors is carried out, in a large part, by post-translational modifications, such as phosphorylation, ubiquitylation, and modification with ubiquitin-like proteins (UBLs). The importance of ubiquitylation and UBLylation with SUMO in DNA repair is well established, with the modified targets and downstream signalling consequences relatively well characterised. However, the role of dedicated erasers for ubiquitin and UBLs, known as deubiquitylases (DUBs) and ubiquitin-like proteases (ULPs) respectively, in genome stability is less well established, particularly for emerging UBLs such as ISG15 and UFM1. In this review, we provide an overview of the known regulatory roles and mechanisms of DUBs and ULPs involved in genome stability pathways. Expanding our understanding of the molecular agents and mechanisms underlying the removal of ubiquitin and UBL modifications will be fundamental for progressing our knowledge of the DDR and likely provide new therapeutic avenues for relevant human diseases, such as cancer.

The genome is constantly undergoing genotoxic stress due to endogenous processes, such as oxidation due to free radicals produced during cellular metabolism, or exogenous factors, such as ultraviolet (UV) light and ionising radiation (IR). In response, eukaryotic cells have evolved specialised mechanisms to detect and repair these DNA lesions to prevent their propagation to the next generation of cells in a process collectively known as the DNA damage response (DDR). Critically, the inability to detect or repair damaged DNA can lead to blocks in DNA replication or transcription, and mutation or chromosomal aberrations can occur leading to genome instability, a major hallmark of cancer [1]. Hereditary defects in the factors involved in these co-ordinated signalling pathways are associated with predisposition to immunodeficiency, neurodegeneration, infertility, premature aging, and most notably tumorigenesis, highlighting the importance of the DDR to human health [2,3].

The DDR enables cells to determine the type of DNA lesion and respond appropriately, such as by halting the cell cycle, altering gene transcription, instigating DNA repair, or to promote senescence or apoptosis if the damage is too severe. Importantly, post-translational modifications (PTMs) such as phosphorylation, ubiquitylation, and ubiquitin-like modifications play a critical role to enable a rapid and co-ordinated response to genomic damage and to switch off the response once genome integrity is reinstated. Phosphorylation brought about by phosphoinositide 3-kinase-like kinase (PIKK) family enzymes, ATM (ataxia-telangiectasia mutated), ATR (ataxia telangiectasia and Rad3-related protein) and DNA-PKcs, is well established to critically regulate DDR signalling pathways by activating specific repair factors and amplifying the cellular response in positive-feedback loops [4]. The factors involved and modified depend on the type of damage and the subsequent repair pathway. Translesion synthesis (TLS) allows replicative DNA polymerases to bypass blocking DNA lesions, while DNA interstrand cross-links (ICLs), caused by reactive mutagenic agents such as mitomycin C (MMC) or cisplatin, are repaired via the Fanconi anaemia (FA) pathway. The majority of IR-induced DNA double-strand breaks (DSBs) are repaired by homologous recombination (HR) or non-homologous end-joining (NHEJ), with HR being a higher fidelity repair pathway restricted to S/G2 phases of the cell cycle using a sister chromatid as a homologous template for repair. NHEJ, however, can often be error-prone but allows repair at any point in the cell cycle outside of mitosis. Other examples of repair pathways include base excision repair (BER) to correct non-helix-distorting base damage due to oxidising agents, nucleotide excision repair (NER) to respond to helix-distorting damage caused by UV light, and mismatch repair to revert base mismatches. Corresponding kinase activities generally relate to repair pathways, with ATM often linked to DSB repair, ATR associated with replication stress-coupled repair, and DNA-PKcs involved in NHEJ.

PTMs such as phosphorylation play numerous regulatory roles in cell signalling pathways in the DDR and beyond, such as inflammation and the cell cycle. Outside of alternative promoter usage and splicing, PTMs provide a key decision point to expand the human cellular proteome. While the role of phosphorylation has been long established in genome stability, ubiquitylation has been extensively studied with respect to proteasome-mediated degradation and in non-degradative roles (e.g. protein-protein interactions and protein localisation). Ubiquitin (Ub) signalling revolves around the addition of the small (8.5 kDa), highly conserved 76 amino acid long protein Ub to target substrates via an isopeptide bond between its C-terminal glycine to, predominantly, substrate lysine residues [5,6]. The covalent attachment occurs via an enzymatic cascade, with an initial ATP-dependent E1-activation step, followed by E2-conjugation, and E3-ligase mediated substrate modification. Ub modifications can exist as single (mono) forms, multi-mono (at multiple different substrate lysine residues) or as polymeric forms, with Ub polymers formed via attachment of Ub to the N-terminal methionine or one of seven internal lysine residues (K6, K11, K27, K29, K33, K48, and K63) within the Ub monomer [7]. The length and linkage type, whether as a homo-polymer, hetero-polymer, or as a branched chain, can dictate the downstream signalling consequences with alternative binding proteins, commonly referred to as readers or receptors, recruited depending on the poly-ubiquitin (polyUb) chain architecture [8–10]. Further complexity is introduced via the addition of PTMs on Ub, such as T12 [11,12] and S20, S57, and S65 phosphorylation [13], inclusion of ubiquitin-like (UBL) proteins, and non-lysine [14] or non-protein ubiquitylation [15,16]. Collectively, the resulting complex signalling repertoire is called the Ub/UBL code [8,17,18].

Other UBLs have been identified that differ in their primary amino acid sequence but share a common 3D structure with the ubiquitin molecule, specifically, a β-grasp fold and often a C-terminal glycine for covalent attachment to target substrates via enzymatic cascades analogous to the Ub system [19–21]. These include several paralogues of a small ubiquitin-like modifier (mainly SUMO1, SUMO2, and SUMO3) [22], neural precursor cell expressed and developmentally down-regulated 8 (NEDD8), interferon-stimulated gene 15 (ISG15), human leukocyte antigen F locus adjacent transcription 10 (FAT10), ubiquitin-fold modifier 1 (UFM1), ubiquitin-related modifier 1 (URM1), autophagy-related protein 12 (ATG12), autophagy-related protein 8 (ATG8), Finkel-Biskis-Reilly murine sarcoma virus (FBR-MuSV) ubiquitously expressed (FUBI), and ubiquitin-like protein 5 (UBL5). The involvement of Ub, SUMO, and NEDD8 in genome stability pathways has been well established [23–29], while the roles of ISG15 and UFM1 have only recently started to emerge as critical regulatory modules for the DDR [19,30].

The identification and characterisation of the substrates, architecture, and readers for some of these UBLs is still in its infancy, but common to Ub and many of the UBLs is the existence of enzymes to cleave them at the terminal glycine residue, known as deubiquitylases (DUBs) for Ub and ubiquitin-like proteases (ULPs) for UBLs (Figure 1). DUBs and ULPs reverse the covalent attachment of Ub or UBLs from substrates or polymeric chains to remove the modification to counter proteolytic degradative pathways, alter the localisation or signalling output of the protein substrate, or in processing Ub/UBLs from their expressed precursor fusions. Therefore, the action of DUBs and ULPs has a key regulatory role within numerous cell signalling pathways, particularly with respect to genome stability [26,31–36]. Herein, we provide an overview of the current knowledge of DUBs and ULPs involved in diverse DDR signalling pathways, highlighting some of the mechanisms involved and future perspectives on identifying DUBs/ULPs in genome stability as potential therapeutic targets or markers for Ub/UBL-associated human diseases, such as cancer.

Circular plot of the human deubiquitylases (DUBs) and ubiquitin-like proteases (ULPs) currently known.

Figure 1.
Circular plot of the human deubiquitylases (DUBs) and ubiquitin-like proteases (ULPs) currently known.

Enzymes are grouped by major DUB families and proteases against UBLs. DUBs with additional ULP activity (e.g. USP18 and USP36) are annotated with the appropriate UBL outside of the circle. DUBs and ULPs referenced in this review to have a role in the DNA damage response (DDR) (Table 1) are highlighted in bold with a filled background. CSN5 and COPS5 (indicated by an *) are the same protein duplicated due to its annotation as a JAMM-family protease while having deNEDDylation activity. DEN1 is also known as NEDP1 or SENP8. ZUP1 is so far the only member of this DUB family and only a single member of USP17 (USP17L1) is shown due to its highly polymorphic copy number variation. ATG4A-D are cysteine proteases related to autophagy signalling pathways.

Figure 1.
Circular plot of the human deubiquitylases (DUBs) and ubiquitin-like proteases (ULPs) currently known.

Enzymes are grouped by major DUB families and proteases against UBLs. DUBs with additional ULP activity (e.g. USP18 and USP36) are annotated with the appropriate UBL outside of the circle. DUBs and ULPs referenced in this review to have a role in the DNA damage response (DDR) (Table 1) are highlighted in bold with a filled background. CSN5 and COPS5 (indicated by an *) are the same protein duplicated due to its annotation as a JAMM-family protease while having deNEDDylation activity. DEN1 is also known as NEDP1 or SENP8. ZUP1 is so far the only member of this DUB family and only a single member of USP17 (USP17L1) is shown due to its highly polymorphic copy number variation. ATG4A-D are cysteine proteases related to autophagy signalling pathways.

Close modal
Table 1.
Summary of key DUBs and ULPs involved in DDR signalling pathways.
DDR pathwayDUB or ULP (with references)Substrates/function
DSB repair BRCC36 [81–83,86,87,91K63 polyUb 
 POH1 [89–91K63 polyUb 
 USP5 [92Unanchored polyUb 
 OTUB2 [93K63 polyUb 
 USP26/USP37/USP44 [95,96RNF168-mediated Ub, H2A 
 USP13 [97RAP80 
 USP3/USP16/USP51 [94,98–101H2A/H2AX-Ub 
 OTUB1 [102,103,105Non-catalytic E2 (Ubc13/UBE2N) inhibitor 
 USP11 [106–111H2AK119/120-Ub; BRCA2/PALB2; RNF4-associated polyUb 
 USP21 [112BRCA2 
 UCHL3 [113RAD51 
 BAP1 [114–116,121H2AK119, INO80 stabilisation 
 USP48 [122H2AK125/127/129-Ub 
 USP34/USP7 [123,124RNF168 
 USP7 [127,128,133TIP60, MDC1, MRN, SUMO 
 UCHL3/OTUD5 [148,149Ku80 
 USP4 [150,151CtIP, MRN 
 UCHL5 [80NFRKB 
 SENP6 [242,252,254,255PolySUMO2/3 
 SENP7 [254KAP1 
 SENP2 [255MDC1 
 ATXN3 [259,260MDC1, RNF8 
 SENP6 [242,261BRCA1-BARD1, 53BP1, BLM, XPF-ERCC1, RPA 
 UfSP2 [274H4, MRN 
 CSN5 (CSN) [290,293,296CRLs, H4, CtIP 
Replication stress USP1 [165,166,174,175PCNA, FANCI-D2 
 USP37 [198Cdt1, BLM 
 VCPIP1 [195,196SPRTN 
 USP7 [132–134SPRTN, SUMO 
 ZUP1 [216–220K63 polyUb 
 SENP6 [246FANCI-D2 
 USP18 [30,280,281PCNA K164/168-ISG15 
NER USP7 [129–131CSB/ERCC6, XPC 
 USP24/USP44 [224,225DDB2 
 USP45 [226XPF-ERCC1 
BER and DNA alkylation USP47 [232Pol β 
 OTUD4-USP9X-USP7 complex [235ALKB2/3 
DDR pathwayDUB or ULP (with references)Substrates/function
DSB repair BRCC36 [81–83,86,87,91K63 polyUb 
 POH1 [89–91K63 polyUb 
 USP5 [92Unanchored polyUb 
 OTUB2 [93K63 polyUb 
 USP26/USP37/USP44 [95,96RNF168-mediated Ub, H2A 
 USP13 [97RAP80 
 USP3/USP16/USP51 [94,98–101H2A/H2AX-Ub 
 OTUB1 [102,103,105Non-catalytic E2 (Ubc13/UBE2N) inhibitor 
 USP11 [106–111H2AK119/120-Ub; BRCA2/PALB2; RNF4-associated polyUb 
 USP21 [112BRCA2 
 UCHL3 [113RAD51 
 BAP1 [114–116,121H2AK119, INO80 stabilisation 
 USP48 [122H2AK125/127/129-Ub 
 USP34/USP7 [123,124RNF168 
 USP7 [127,128,133TIP60, MDC1, MRN, SUMO 
 UCHL3/OTUD5 [148,149Ku80 
 USP4 [150,151CtIP, MRN 
 UCHL5 [80NFRKB 
 SENP6 [242,252,254,255PolySUMO2/3 
 SENP7 [254KAP1 
 SENP2 [255MDC1 
 ATXN3 [259,260MDC1, RNF8 
 SENP6 [242,261BRCA1-BARD1, 53BP1, BLM, XPF-ERCC1, RPA 
 UfSP2 [274H4, MRN 
 CSN5 (CSN) [290,293,296CRLs, H4, CtIP 
Replication stress USP1 [165,166,174,175PCNA, FANCI-D2 
 USP37 [198Cdt1, BLM 
 VCPIP1 [195,196SPRTN 
 USP7 [132–134SPRTN, SUMO 
 ZUP1 [216–220K63 polyUb 
 SENP6 [246FANCI-D2 
 USP18 [30,280,281PCNA K164/168-ISG15 
NER USP7 [129–131CSB/ERCC6, XPC 
 USP24/USP44 [224,225DDB2 
 USP45 [226XPF-ERCC1 
BER and DNA alkylation USP47 [232Pol β 
 OTUD4-USP9X-USP7 complex [235ALKB2/3 

There are ∼100 DUBs encoded in the human genome that can be sub-divided into seven different classes: Ub C-terminal hydrolases (UCHs), Ub-specific proteases (USPs), Machado-Joseph domain-containing proteins (MJDs), otubain domain-containing proteases (OTUs), JAMM (JAB1/MPN/Mov34) metalloproteases, motif interacting with Ub (MIU)-containing novel DUB family (MINDY), and zinc finger-containing Ub-peptidase (ZUP1) [37,38] (Figure 1). Given the complexity of the Ub code, many DUBs display a certain level of selectivity for Ub chain architectures, such as preference for the predominantly proteasome-targeting K11- and K48-linked polyUb chains to protect substrates from degradation. Other DUBs, such as AMSH, BRCC36 and ZUP1, prefer cleavage of non-proteolytic K63-linked chains to alter binding or recruitment platforms in signalling pathways [37–41]. USP-family DUBs are generally promiscuous with respect to Ub chain linkage but are regulated via other means, such as through regulated gene expression, protein-protein interactions or specific recruitment/localisation, and PTMs such as phosphorylation or ubiquitylation [37,40,42–44]. DUB activity is required for the initial maturation of expressed Ub fusions to free Ub monomers from its precursors, predominantly by IsoT/USP5 [45,46], and the recycling of Ub following degradation of cellular proteins, by the DUBs USP14, UCHL5, and POH1 at the proteasome, with USP8 associated with ubiquitin homeostasis in lysosomal degradation pathways [47]. More generally, DUBs act to counteract the action of E3 ligases, which can modulate their signalling outcomes in different ways. For example, by removing proteasome-targeting Ub modifications they can protect proteins from proteolytic pathways. Alternatively, by selectively removing Ub moieties of a certain architecture they can edit the linkage type and topology of the polyUb chains present on the substrate protein. Therefore, DUBs are established to play key roles in several DDR signalling pathways with errors in their activity or function likely contributing to disease progression [31,35,48–50]. As a result, there is increasing appetite and potential for using small molecule inhibitors targeting DUBs in anti-cancer therapeutic strategies, with some already in early development [51–54].

DUBs in DSB repair

An established role of Ub signalling is to recruit repair factors to sites of DNA damage or modulate their activity to co-ordinate the response. This means DUBs and their proper regulation are central to the maintenance of genome stability, particularly in the case of DNA DSBs [55]. DSBs are formed during endogenous processes such as DNA replication, or due to external factors such as IR, and can be repaired by HR during the S/G2 phases of the cell cycle or more error-prone NHEJ throughout interphase, independently of the cell cycle stage [56]. Classical NHEJ (c-NHEJ) relies on the heterodimer Ku70/Ku80 to bind DSB ends, which localises DNA-PKcs, XRCC4-LIG4, XLF, PAXX, plus other NHEJ factors to bring about synapsis of the DNA ends, DSB end-processing, and ligation of the break. HR, however, requires more extensive processing of the DNA that is initiated by the MRN (MRE11/RAD50/Nbs1) endonuclease complex to activate the ATM kinase and help recruit CtIP, BRCA1-BARD1, plus other helicases and nucleases, for long-range resection around the DSB. The resulting single-stranded DNA (ssDNA) is protected by the replication protein A (RPA) complex before BRCA2-mediated RAD51 loading, leading to strand invasion of the complimentary strand on the sister chromatid and subsequent repair. The cell cycle stage and the recruitment of either 53BP1 and RAP80 for NHEJ, or BRCA1-BARD1 for HR, are critical determinants for the method of repair.

Ubiquitylation is central to the choice between HR and NHEJ for recruiting specific factors to the site of damage on chromatin (Figure 2A). Phosphorylation of the histone variant, H2AX, at S139 (forming γH2AX) by ATM kinase is a key initial signalling event [57,58] to recruit MDC1 [59], phosphorylation of which enables recruitment of the Ub E3 ligase RNF8 [60]. K63-linked polyUb chain formation on histone H1 by RNF8 [61–64], utilising the E2 conjugating enzyme Ubc13/UBE2N, leads to subsequent localisation of the Ub E3 ligase RNF168 to the damage site, resulting in further mono-ubiquitylation on histone H2A at K13/K15 (H2AK13/15-Ub) to propagate the signal [65]. RAP80 binding to K63-linked polyUb via a Ub-interacting motif (UIM) is thought to sequester the pro-HR Ub E3 ligase, BRCA1-BARD1, away from sites of DNA damage, while H2AK13/15-Ub in the context of dimethylated K20 on histone H4 enables 53BP1 recruitment [66–70]. 53BP1 accrual at DNA damage sites facilitates NHEJ by suppressing BRCA1 localisation and recruiting RIF1, REV7 and other components of the shieldin complex, which acts to protect DNA ends [71–73] (Figure 2B). The Ub E3 ligase RNF169 can also bind H2AK15-Ub [74–76], therefore competing with 53BP1 recruitment, to limit NHEJ signalling, and BRCA1-BARD1 ubiquitylation at H2AK125/127/129 can displace 53BP1 as an essential step to promote HR [77,78] (Figure 2C). Therefore, regulating target protein ubiquitylation and K63-linked polyUb chain formation is key and numerous DUBs have been identified to counteract aberrant Ub signalling to ensure robust DSB signalling, as detailed for key examples in the following section [31–33,48,79,80].

Overview of DUBs involved in DSB repair signalling pathways.

Figure 2.
Overview of DUBs involved in DSB repair signalling pathways.

(A) Upon DNA damage causing a DSB (red star), a plethora of signalling events involving Ub can occur to regulate the repair pathway choice in the context of other cell signals, such as cell cycle cues. MDC1 (SENP2 and USP7), TIP60 (USP7), RNF168 (USP34 and USP7), and RAP80 (USP13) are all stabilised through the action of DUBs to counteract polyUb-mediated protein degradation. RAP80 can bind both Ub and SUMO signals upon DSB formation to recruit BRCA1-A complex components, enabling sequestration of BRCA1-BARD1 and localisation of K63-linked polyUb DUB activity of BRCC36. Other DUBs, such as OTUB2, POH1, USP26, USP37, and USP44, dampen RNF168-mediated polyUb chain formation ensuring proficient repair of the DSB. USP51 can directly bind H2AK13/15-Ub-containing nucleosomes to remove the Ub signal, with USP3 and USP16 also implicated to remove H2A(X)-Ub modifications. USP11 and BAP1 can remove the polycomb-related H2AK119-Ub mark, likely to regulate transcription during DDR signalling and local chromatin accessibility, further aided by INO80 stabilisation through the DUBs BAP1 and UCHL5. (B) H2AK13/15-Ub combined with H4K20me2 is bound by 53BP1 in a multidentate manner to recruit the shieldin complex to help protect DNA ends and displace the HR repair machinery. Ku80 chromatin retention is regulated by the DUBs OTUD5 and UCHL3. (C) During S/G2 phase, the Ub E3 ligase BRCA1-BARD1 can bind H2AK13/15-Ub combined with unmethylated H4K20 to ubiquitylate H2AK125/127/129 to promote HR-mediated repair signalling. The DUB USP48 can counteract this Ub signal to prevent hyper-resection. The MRN complex and CtIP are stabilised at sites of DSBs through the action of USP4. The BRCA1-PALB2-BRCA2 complex, required for BRCA2-mediated loading of RAD51, is stabilised by cell cycle-regulated USP11, which removes an inhibitory Ub that otherwise disrupts the BRCA1-PALB2 interaction during S/G2 phase. DUB/ULP activity is represented by dashed arrows.

Figure 2.
Overview of DUBs involved in DSB repair signalling pathways.

(A) Upon DNA damage causing a DSB (red star), a plethora of signalling events involving Ub can occur to regulate the repair pathway choice in the context of other cell signals, such as cell cycle cues. MDC1 (SENP2 and USP7), TIP60 (USP7), RNF168 (USP34 and USP7), and RAP80 (USP13) are all stabilised through the action of DUBs to counteract polyUb-mediated protein degradation. RAP80 can bind both Ub and SUMO signals upon DSB formation to recruit BRCA1-A complex components, enabling sequestration of BRCA1-BARD1 and localisation of K63-linked polyUb DUB activity of BRCC36. Other DUBs, such as OTUB2, POH1, USP26, USP37, and USP44, dampen RNF168-mediated polyUb chain formation ensuring proficient repair of the DSB. USP51 can directly bind H2AK13/15-Ub-containing nucleosomes to remove the Ub signal, with USP3 and USP16 also implicated to remove H2A(X)-Ub modifications. USP11 and BAP1 can remove the polycomb-related H2AK119-Ub mark, likely to regulate transcription during DDR signalling and local chromatin accessibility, further aided by INO80 stabilisation through the DUBs BAP1 and UCHL5. (B) H2AK13/15-Ub combined with H4K20me2 is bound by 53BP1 in a multidentate manner to recruit the shieldin complex to help protect DNA ends and displace the HR repair machinery. Ku80 chromatin retention is regulated by the DUBs OTUD5 and UCHL3. (C) During S/G2 phase, the Ub E3 ligase BRCA1-BARD1 can bind H2AK13/15-Ub combined with unmethylated H4K20 to ubiquitylate H2AK125/127/129 to promote HR-mediated repair signalling. The DUB USP48 can counteract this Ub signal to prevent hyper-resection. The MRN complex and CtIP are stabilised at sites of DSBs through the action of USP4. The BRCA1-PALB2-BRCA2 complex, required for BRCA2-mediated loading of RAD51, is stabilised by cell cycle-regulated USP11, which removes an inhibitory Ub that otherwise disrupts the BRCA1-PALB2 interaction during S/G2 phase. DUB/ULP activity is represented by dashed arrows.

Close modal

BRCC36 in the BRCA1-A complex

BRCC36 is a K63-specific metalloprotease of the JAMM-family of DUBs and was initially linked to the DDR through localisation studies indicating its presence at nuclear foci for DSBs [81,82]. This recruitment was dependent on RAP80 as part of the BRCA1-A complex, which also contains BRCA1, BARD1, MERIT40, BRCC45 and Abraxas, and plays an important role to sequester BRCA1 away from DSB sites and preventing end-resection pathways [83–85] (Figure 2A). Recent structural data shows how the BRCA1-A complex, different from the cytoplasmic BRISC complex that also contains BRCC36, can engage and sequester BRCA1 while enabling RAP80 to bind mixed Ub-SUMO linked chains, relevant for its role in regulating DSB repair outcomes [86]. Depletion of BRCC36 results in elevated K63-linked polyUb at DSBs, leading to increased 53BP1 foci formation, but cells exhibit a hyper-HR phenotype with increased resection, RAD51 loading, and IR sensitivity, suggesting loss of BRCC36 leads to unproductive repair [87,88]. Therefore, preventing excessive K63-polyUb chain formation at DSBs seems to balance the signals alongside other histone PTMs and cell cycle stage, enabling conditions conducive to both HR and NHEJ.

POH1 linking DDR signalling to the proteasome

POH1, otherwise known as Rpn11 or PSMD14, is a proteasome-associated DUB that has been linked to the DDR [89,90]. Like BRCC36, POH1 is a JAMM-family metalloprotease able to cleave K63-linked polyUb chains, as well as other chain types [90,91]. Depletion of POH1 results in increased sensitivity to DSBs in response to hydroxyurea (HU) with elevated polyUb foci at damaged sites. POH1 DUB activity against K63-linked polyUb restricts 53BP1 accumulation at DSB foci, antagonising RNF8/RNF168, and therefore modulating NHEJ-mediated DNA repair and promoting HR-mediated resection pathways [90]. POH1 also acts independently of 53BP1 to promote RAD51 loading in HR-mediated repair during G2 phase of the cell cycle, meaning POH1 is a key component of DSB repair pathways to limit anchored polyUb conjugates at sites of damage to ensure proficient repair [89]. The activity of POH1/Rpn11 to bulk-cleave polyUb chains from substrates, such as histone proteins to counteract 53BP1 binding, may be linked to another DUB USP5, which has known DUB activity against unanchored polyUb chains [46], to recycle the Ub protein for signalling elsewhere. Depletion of USP5 also results in DNA repair defects and USP5 is thought to be recruited to sites of DNA damage [80,92].

OTUB2 as an antagonist to RNF8/168 signalling

An RNAi screen identified another K63-specific DUB, a member of the OTU-family of DUBs OTUB2, which antagonises RNF8/RNF168-mediated Ub signalling in the DDR [93]. Depletion leads to increased Ub, 53BP1, RAP80, and RNF168 foci at damage sites, resulting in increased preference for NHEJ-mediated repair pathways, while overexpression does not alter recruitment of other upstream factors such as RNF8 and MDC1. Therefore, OTUB2 likely supresses K63-linked polyUb chain formation to prevent RNF168, and therefore 53BP1, recruitment and favouring HR-mediated repair. Despite both being K63-specific DUBs, depletion of BRCC36 and OTUB2 have opposing outcomes, with BRCC36 depletion leading to a hyper-resection phenotype. Therefore, differing DUB actions, such as substrate choice and site of action, may influence the recruitment and coordination of the HR-suppressive RNF168-RAP80-53BP1 components, or they may have additional functions in repair outside of their intrinsic DUB activity (e.g. BRCC36 as part of the BRCA1-A complex) resulting in alternative outcomes. Additional DUBs that limit or counteract RNF168-mediated ubiquitylation include USP16, which is thought to regulate DSB signalling through H2A(X) deubiquitylation [94], and USP26, USP37 [95], and USP44 [96] that have been identified as DUBs recruited to sites of DSBs to counteract RNF168-mediated ubiquitylation. USP13, on the other hand, reverses an inhibitory ubiquitylation on RAP80 close to its UIM, which disrupts its binding to K63-linked polyUb, enabling BRCA1-A recruitment to DSBs [97].

DUBs linked to histone H2AK13/15-Ub

In addition to the polyUb DUB activity described above, several DUBs have been identified to act directly on the H2AK13/15-Ub mark. USP3 [98,99], and USP51 [100] have been described to have such activity and counteract RNF168-mediated action on H2A and/or H2AX, so suppressing DSB repair signalling. In the case of USP51, it has been shown to be recruited to chromatin and directly bind the site of action on the H2A/H2B dimer upon DNA damage to specifically remove Ub from H2AK13/15 [101], with knockdown resulting in increased spontaneous DNA damage and elevated 53BP1 and BRCA1 foci formation due to increased H2AK13/15-Ub, while not affecting upstream signalling events.

A common theme is the apparent redundancy between DUBs in the DDR, such as for USP3, USP16, and USP51 noted above for H2A ubiquitylation, with likely effects on how the response to targeted drugs impacts on the signalling outcome. Whether some of this redundancy is due to cell-type differences in expression, lack of specificity (common for many DUBs in vitro), or if they have experimental explanations would need to be determined. Another possibility is that the non-catalytic roles of some DUBs may have additional effects on the DDR signal. One such example is OTUB1 [102]. OTUB1 is a negative regulator of RNF168-mediated polyUb chain formation at sites of DNA damage [103]. OTUB1 inhibits the E2 enzyme, Ubc13/UBE2N plus other E2 conjugating enzymes, independently of its DUB activity by binding directly to the charged E2∼Ub via an N-terminal extension that contains a Ub-binding motif [104,105]. This prevents the transfer of Ub from the E2 to the target substrate. OTUB1 depletion results in spontaneous and persistent polyUb and 53BP1 foci, and therefore regulates DSB repair pathway choice.

USP11 in DSB repair pathway choice

USP11 is another DUB, identified in an overexpression screen with ectopic RNF8, to act on ubiquitylated H2A [106,107]. Validation with USP11 depletion showed increased H2AX-Ub, increased 53BP1 and polyUb foci, and increased sensitivity to IR suggesting improper DSB repair signalling. USP11 can physically interact with H2AX and localises to sites of DNA damage, reinforcing its role in DSB repair signalling, while its activity against H2A-Ub is proposed to be at K119/K120 in H2A, rather than the RNF168/53BP1-relevant H2AK13/15 [107] (Figure 2A). USP11 has also, however, been implicated as an interacting partner and DUB for various other factors in the DDR, particularly in DSB repair, making its mode of action much more complex. USP11 is regulated in a cell cycle-dependent manner by KEAP1 [108], with USP11 levels being low during G1. Indeed, USP11 has been shown to bind and remove Ub from PALB2 to facilitate the formation of the BRCA1-PALB2-BRCA2 complex enabling RAD51 loading and HR repair [108,109], with USP11 abundance during S/G2 able to facilitate this (Figure 2C). It has also been identified as an interactor of the SUMO-targeted Ub ligase (STUbL) RNF4 to counteract its ubiquitylation activity, preventing SUMO-Ub hybrid chains from forming, and regulating DSB repair signalling choice [110]. Consistent with these roles in HR-mediated DSB repair, USP11 depletion sensitises cells to olaparib (PARPi) and IR [111]. Additionally, USP21 was identified to stabilise the BRCA2-RAD51 complex by antagonising BRCA2 ubiquitylation [112], while UCHL3 counteracts RAD51 ubiquitylation [113], both ensuring competent RAD51 loading and proficient HR repair.

BAP1 stabilises the INO80 remodelling complex

BRCA1-associated protein 1 (BAP1) is a nuclear-localised UCH-family DUB originally reported as a tumour suppressor, with its DUB activity linked to promoting HR-mediated repair of DSBs [114]. Depletion of BAP1 leads to reduced BRCA1 foci, and reduced RAD51 loading, with reduced HR repair and increased IR sensitivity coinciding with elevated H2A and H2AX ubiquitylation. The target for BAP1 is H2AK119, a PRC1-related histone ubiquitylation, potentiating a role for BAP1 in the transcriptional or chromatin response to DNA damage, rather than a direct role in the repair process [115]. Linked to this is the role of BAP1 in replication-coupled repair, where BAP1 aids to recruit and stabilise the chromatin remodeller INO80 to sites of DNA damage or replication stress, possibly to promote resection through its remodelling activity [116] (Figures 2A and 3A). BAP1 recruitment to sites of DNA damage is rapid and dependent on PARP1 and/or ATM-mediated modification of BAP1, with it also having a role in NER signalling pathways via PARP1 [54,117,118]. Recent structures of BAP1/ASXL1 with H2AK119-Ub-containing nucleosomes highlighted the role of many BAP1 mutations identified in cancer, likely to have roles in transcriptional regulation or DNA repair pathways linked to its role as the polycomb-repressive DUB (PR-DUB) [119–121].

Overview of DUBs in DDR signalling pathways associated with replication stress, NER, and BER.
Figure 3.
Overview of DUBs in DDR signalling pathways associated with replication stress, NER, and BER.

(A) DUBs act on central signalling nodes within the DDR signalling pathways upon replication stress. PCNA is mono-ubiquitylated by RAD6/RAD18 upon stalling or slowing of the replication fork due to DNA damage. Mono-ubiquitylated PCNA forms a platform to recruit more error-prone TLS polymerases to bypass the lesion. RAD18 and the TLS polymerases are themselves protected from polyUb-mediated degradation via the proteasome due to the action of USP7. To restore normal replicative processes, the mono-Ub is removed by USP1-UAF1. Template switching (TS) is thought to be instigated by HLTF- and SHPRH-dependent poly-ubiquitylation of PCNA, recruiting the helicase and endonuclease ZRANB3 as the downstream effector. Replication stalling can lead to excessive ssDNA bound by ATR phosphorylated RPA, which can lead to replication fork reversal or TLS via RFWD3-dependent poly-ubiquitylation of surrounding proteins, including of PCNA and RPA. ZUP1 is hypothesised to be a candidate DUB to counteract polyUb chain formation during this process. Interstrand cross-links (ICLs) are generally repaired via the Fanconi anaemia (FA) repair pathway. FANCI-D2 is ubiquitylated by the FA core complex in an ATR-dependent manner, resulting in the FANCI-D2 heterodimer forming a DNA clamp at the site of damage to facilitate downstream repair. USP1-UAF1 can remove the mono-Ub mark. SENP6 is a deSUMOylase that counteracts polySUMO2/3 chain formation on FANCI-D2, preventing RNF4-mediated poly-ubiquitylation leading to VCP/p97-mediated extraction of FANCI-D2 from chromatin. One mechanism to remove DNA-protein cross-links (DPCs) is through the DNA-dependent metalloprotease SPRTN whose activity requires removal of an inhibitory mono-Ub by USP7 and/or VCPIP1. BAP1 stabilises the chromatin remodeller INO80 through its DUB activity to provide a DNA repair-proficient chromatin environment upon damage. (B) DUBs in nucleotide excision repair (NER) primarily protect repair factors from degradation. CUL4-DDB2 forms an E3 ligase for the process, with USP24 and USP44 thought to stabilise DDB2 by removing polyUb chains. One of the substrates for CUL4-DDB2 is XPC, which is protected from chromatin extraction and degradation by the action of USP7, with USP7 also removing polyUb from CSA via UVSSA-mediated recruitment. The endonuclease, XPF-ERCC1, is retained at sites of damage by USP45. (C) During base excision repair (BER), Pol β abundance and chromatin engagement is regulated through Ub signalling by the E3 ligases Mule and CHIP, with the DUB USP47 able to remove the polyUb signal. Repair of DNA base alkylation by ALKB2 and ALKB3 is enabled by a DUB complex made up of USP9X and USP7, with OTUD4 forming a non-catalytic scaffold subunit. DUB/ULP activity is represented by dashed arrows.

Figure 3.
Overview of DUBs in DDR signalling pathways associated with replication stress, NER, and BER.

(A) DUBs act on central signalling nodes within the DDR signalling pathways upon replication stress. PCNA is mono-ubiquitylated by RAD6/RAD18 upon stalling or slowing of the replication fork due to DNA damage. Mono-ubiquitylated PCNA forms a platform to recruit more error-prone TLS polymerases to bypass the lesion. RAD18 and the TLS polymerases are themselves protected from polyUb-mediated degradation via the proteasome due to the action of USP7. To restore normal replicative processes, the mono-Ub is removed by USP1-UAF1. Template switching (TS) is thought to be instigated by HLTF- and SHPRH-dependent poly-ubiquitylation of PCNA, recruiting the helicase and endonuclease ZRANB3 as the downstream effector. Replication stalling can lead to excessive ssDNA bound by ATR phosphorylated RPA, which can lead to replication fork reversal or TLS via RFWD3-dependent poly-ubiquitylation of surrounding proteins, including of PCNA and RPA. ZUP1 is hypothesised to be a candidate DUB to counteract polyUb chain formation during this process. Interstrand cross-links (ICLs) are generally repaired via the Fanconi anaemia (FA) repair pathway. FANCI-D2 is ubiquitylated by the FA core complex in an ATR-dependent manner, resulting in the FANCI-D2 heterodimer forming a DNA clamp at the site of damage to facilitate downstream repair. USP1-UAF1 can remove the mono-Ub mark. SENP6 is a deSUMOylase that counteracts polySUMO2/3 chain formation on FANCI-D2, preventing RNF4-mediated poly-ubiquitylation leading to VCP/p97-mediated extraction of FANCI-D2 from chromatin. One mechanism to remove DNA-protein cross-links (DPCs) is through the DNA-dependent metalloprotease SPRTN whose activity requires removal of an inhibitory mono-Ub by USP7 and/or VCPIP1. BAP1 stabilises the chromatin remodeller INO80 through its DUB activity to provide a DNA repair-proficient chromatin environment upon damage. (B) DUBs in nucleotide excision repair (NER) primarily protect repair factors from degradation. CUL4-DDB2 forms an E3 ligase for the process, with USP24 and USP44 thought to stabilise DDB2 by removing polyUb chains. One of the substrates for CUL4-DDB2 is XPC, which is protected from chromatin extraction and degradation by the action of USP7, with USP7 also removing polyUb from CSA via UVSSA-mediated recruitment. The endonuclease, XPF-ERCC1, is retained at sites of damage by USP45. (C) During base excision repair (BER), Pol β abundance and chromatin engagement is regulated through Ub signalling by the E3 ligases Mule and CHIP, with the DUB USP47 able to remove the polyUb signal. Repair of DNA base alkylation by ALKB2 and ALKB3 is enabled by a DUB complex made up of USP9X and USP7, with OTUD4 forming a non-catalytic scaffold subunit. DUB/ULP activity is represented by dashed arrows.

Close modal

USP48 counters hyper-resection

BRCA1-BARD1 mediated H2A ubiquitylation at K125/127/129 also needs to be regulated to dampen HR and end-resection repair [77,78]. USP48 has been identified to reverse this modification by specifically removing Ub at this site [122] (Figure 2C). Depletion of USP48 results in hyper-resection leading to single-strand annealing (SSA) as a method of repair, which is highly mutagenic due to the use of homologous repeats for bridging DSBs leading to genomic deletions.

Other DUBs have been identified to regulate the HR/NHEJ pathway choice through directly regulating RNF168, rather than the downstream H2A/H2AX ubiquitylation. USP34 [123] and USP7 [124] directly associate with RNF168 to prevent its poly-ubiquitylation and subsequent degradation via the proteasome (Figure 2A). Therefore, their role is to promote Ub conjugation at DSBs to enable downstream recruitment of 53BP1 and BRCA1, conferring resistance to DNA damaging agents. However, levels need to be regulated to prevent improper spreading to undamaged chromatin and two HECT-family ligases, UBR5 and TRIP12, have been shown to negatively regulate RNF168 abundance and spreading at DSBs [125]. While an alternative mechanism to DUBs described above, a common paradigm in DSB repair signalling is regulated RNF168 recruitment, and subsequent deposition of H2A-Ub marks, to enable proficient repair in the context of other signals such as cell cycle cues.

Multifaceted roles of USP7 in DDR signalling

The role of USP7 in genome stability is reviewed more thoroughly elsewhere [126], given its vast number of substrates identified and roles in a multitude of cell signalling pathways. In the context of DSBs, it stabilises the acetyltransferase TIP60 that acetylates ATM to promote and propagate DNA damage signalling [127], as well as binding to, and stabilising, MDC1 and [128] (Figure 2A). USP7 is involved in NER in complex with UVSSA to regulate CSB/ERCC6 stability [129,130] or as a DUB for XPC to promote its chromatin loading and stability [131] (Figure 3B). USP7 has also been implicated in the protection against replication stress as a DUB for removing Ub from SUMO chains to protect RAD18 and Pol Eta (η) from proteasome-mediated degradation [132,133], and to remove the inhibitory Ub modification on the DNA-dependent metalloprotease involved in removing DNA-protein cross-links (DPCs), SPRTN [134] (Figure 3A). While not explored in detail here, USP7, along with its role in p53 signalling [135], is critical for genome stability and disease progression, and is actively being investigated from a therapeutic perspective [136–142], with structural characterisation of highly selective USP7 inhibition well established [143–147].

Several other DUBs have been identified to play a role in regulating components of DSB repair UCHL3 [148] and OTUD5 [149] have both been postulated to counteract Ku80 ubiquitylation to stabilise chromatin retention of the Ku heterodimer to promote NHEJ (Figure 2B). USP4 has been linked to positively regulate DNA end resection. USP4 interacts with CtIP, in addition to NBS1 as part of the MRN complex, with USP4 depletion reducing the recruitment of CtIP, RPA, and RAD51 loading to DSB sites [150]. The interaction relies on auto-deubiquitylation of USP4, as a catalytic mutant, unable to remove the polyUb chains on itself, could not interact with CtIP or be recruited to sites of DSBs [151]. UCHL5/UCH37 was identified in a siRNA screen to promote end resection and HR-mediated repair of DSBs [80]. UCHL5 is associated with both the proteasome lid and the INO80 chromatin remodelling complex. With respect to genome stability, the role of UCHL5 is to remove polyUb chains, and subsequent degradation, of NFRKB (nuclear factor related to kB-binding protein), a factor known to promote the interaction between UCHL5 and INO80 (Figure 2A). As noted above, INO80 is required to promote end resection and overall DSB repair.

DUBs in DNA replication-coupled repair

DUB involvement in HR and NHEJ pathways enables proper regulation of the DSB repair choice, dependent on other PTMs, proteins, and cell cycle cues that influence proficient DNA repair. During DNA replication, several repair pathways converge (e.g. HR; SSA; TLS; ICL repair; template switching, TS) because of replication stress to prevent fork collapse and replication catastrophe [152–154]. The type of damage is thought to recruit specific DDR factors to ensure competent repair. Central to the response to replication stress, DPCs, and ICLs is Ub modification of proteins at the fork [155–157], and a few key DUBs have been identified to enable tight regulation of these processes (Figure 3A).

USP1 in DNA replication-coupled repair pathways

USP1 is a DUB that is established to play a key role in the protection against ICL damage and replication stress. A central node for Ub signalling in replication stress is PCNA (proliferating cell nuclear antigen), which functions as the sliding clamp for replicative polymerases ensuring processivity during DNA synthesis. PCNA can be modified with Ub at the conserved K164 position in response to DNA damaging agents (e.g. HU, aphidicolin, UV), resulting in the slowing or stalling of replication forks [158–160]. The mono-ubiquitylation is brought about by RAD6 (E2) and RAD18 (E3) and leads to the recruitment of lower fidelity Y-family TLS polymerases such as Rev1, Pol η (eta), κ (kappa), and ι (iota) via their Ub binding domains (UBDs) and weak PCNA interacting peptide (PIP) box interactions, that allow for bypass of DNA lesions during replication [161–164]. Once the damage is passed or repaired, the PCNA-Ub mark needs to be reversed to resume high-fidelity DNA replication to maintain genome integrity and prevent error-prone replication. This activity is performed by USP1, in complex with UAF1 (also known as WDR48), with dysregulation of USP1 activity leading to increased PCNA ubiquitylation and mutational frequency, reduced fork speed, and micronuclei formation [165–167]. The recruitment of USP1-UAF1 is thought to be via a SUMO-like domain on UAF1 that recognises the SUMO-interacting motif (SIM) on ELG1, a PCNA-binding RFC-like adaptor for Ub removal from PCNA [168,169]. USP1 is also regulated by a self-cleavage mechanism occurring at an internal GG motif at residues 670–671, leading to rapid degradation of USP1 in response to UV-induced DNA damage [165]. Interestingly, a recent study has identified USP1 autocleavage-defective mutants, similar to a catalytic mutant, that lead to increased retention at sites of DNA synthesis leading to the accumulation of USP1 trapping lesions on DNA, which can partially be alleviated by SPRTN to ensure genome stability [170,171]. USP1 is a topical target for cancer therapeutics [172], particularly given its synthetic lethality with BRCA1 [167], and it is possible that pharmacological inhibition of USP1 may lead to USP1 trapping in addition to abrogation of its DUB activity, and therefore may explain some of the effects seen. A recent study highlights how ML323 may engage with USP1, providing a platform for subsequent drug development for its inhibition [173]. Another small molecule, KSQ-4279, has been identified in CRISPR screens and has entered Phase I clinical trials as a monotherapy or combination therapy with PARP inhibitors or chemotherapy [53], highlighting the promise of therapeutically targeting DUBs, such as USP1.

USP1-UAF1 also contributes to genome stability as a DUB for FA repair of ICLs caused by MMC or cisplatin [166,174,175]. ICLs can block DNA replication and transcription if left unrepaired, with FA signalling coordinating multiple repair pathways including NER, HR, and TLS depending on the specific type of damage. A key step in FA signalling is the Ub modification of the FANCI and FANCD2 (FANCI-D2) heterodimer at residues K523 and K561, respectively, by the FA core complex [165,176–178]. Mono-ubiquitylation of FANCI-D2 is activated by ATR-mediated phosphorylation and peaks during S phase of the cell cycle or in response to ICL-inducing agents [179–182]. Recent structural data indicates that sequential ubiquitylation results in substantial conformational changes, enabling ubiquitylated FANCI-D2 to form a clamp around the double-stranded DNA (dsDNA). This likely protects the underlying DNA during the DDR [183–190] and facilitates downstream repair. Interestingly, the mono-Ub mark does not, to date, seem to mediate the recruitment of repair factors, rather the Ub marks are bound by the other member of the heterodimer in FANCI-D2 effectively shielding it from UBDs in FA factors. For competent repair, however, the Ub modification needs to be removed from FANCI-D2 to facilitate its removal from chromatin [166,174,175]. USP1 was identified in an RNAi screen, as depletion increases spontaneous FANCD2 ubiquitylation and increases cellular sensitivity to ICL-inducing cross-linking agents. Structural data of USP1-UAF1, with Ub and in an enzyme-substrate complex with ubiquitylated FANCI-D2, reveals critical mechanistic and regulatory details for Ub removal from FANCD2 [191–194]. Amino acid residues at the FANCI-UAF1 interface, including those of known ATR phosphorylation sites, are crucial for USP1-mediated removal of the FANCD2-Ub mark, corroborating genetic and biochemical data.

Other DUBs involved in countering replication stress

Other DUBs have been proposed to have roles in other aspects of replication-coupled repair pathways. USP37 has been suggested to stabilise the DNA replication origin licensing factor, Cdt1, to ensure proficient DNA replication and to protect the RecQ helicase, BLM, from proteolysis [195–197]. DPC repair via the DNA-dependent metalloprotease SPRTN is regulated through an inhibitory Ub modification, removal of which is thought to be via USP7 [134] and VCPIP1 [198]. VCPIP1 is activated and shuttled to the nucleus by ATM/ATR-mediated phosphorylation, where it can remove the Ub mark, enabling SPRTN recruitment to chromatin and downstream repair of the DPC.

Removing polyUb signals at sites of replication stress

Ub signalling in replication-coupled repair and its regulation by DUBs has predominantly focussed on mono-Ub modifications on DDR factors. PolyUb chains have been identified to occur on PCNA [199,200]; RPA [201–205]; and the CMG (Cdc45-MCM-GINS) complex [206–210]. TRAIP-dependent polyUb chain formation on the CMG helicase is proposed to be linked to VCP/p97 extraction upon replication termination or helicase stalling for unloading [209,211,212]. The role of PCNA poly-ubiquitylation by Rad5 in yeast, HLTF is the human homologue, has been linked to TS through association with the helicase and endonuclease, ZRANB3, and another Ub E3 ligase, SHPRH, to promote error-free fork restart [199,213]. Similarly, RPA poly-ubiquitylation is non-degradative to promote DNA damage bypass and fork restart (Figure 3A). DUBs for these processes have not been robustly elucidated, with the polyUb signal likely leading to VCP/p97-mediated extraction [214]. A recently discovered DUB with preference for K63-linked polyUb chains, ZUP1, has been associated with replication-coupled genome stability [215–219]. ZUP1 localises to sites of DNA damage, interacts with the RPA complex, and depletion increases spontaneous DNA damage and sensitises cells to DNA damaging agents such as camptothecin (CPT). This activity relies on the DUB activity of ZUP1, and preliminary data suggested it to be the DUB for ubiquitylated RPA. A recent study further proposed that the direct interaction with the RPA complex enhanced the K63-linked polyUb DUB activity of ZUP1, suggesting that ZUP1 is recruited and activated through binding to the critical ssDNA binding protein complex [220]. The identity of polyUb modified substrates for ZUP1 at sites of DNA damage remain unknown, but one hypothesis is that it can counteract ssDNA-associated K63-linked polyUb chain formation from E3 ligases, such as RFWD3.

DUBs in the repair of DNA adducts

The repair of bulky DNA adducts, such as thymidine dimers induced by UV light, is carried out by NER, reviewed in more detail elsewhere [221–223]. Recognition of the damage can occur via transcription-coupled NER (TC-NER) or by global genome NER (GG-NER), before the pathways converge to correct the damage. XPC and DDB proteins act as global genomic DNA damage sensing factors to recognise the lesion and recruit the repair machinery. At transcriptionally active regions, TC-NER relies on stalling of the RNA polymerase, with associated CSA and CSB proteins enabling damage recognition without the need for global distortion surveillance. Dual incision of the damaged site occurs via XPG and XPF-ERCC1 endonucleases before DNA polymerase loading to fill-in the damaged strand and sealing by a DNA ligase.

Ub signalling in NER, and its regulation by DUBs, revolves around the action of USP7 on XPC and CSA, mentioned above (Figure 3B). Another DUB, USP24, is also required for proficient repair and cell survival [224]. USP24 has been found to bind, deubiquitylate, and stabilise DDB2, a component of the CUL4 E3 ligase. CUL4-DDB2 is known to poly-ubiquitylate XPC, a substrate of USP7, so how these DUB activities act in concert to regulate NER is unclear. USP44 is another DUB that has been implicated to stabilise DDB2 and shown to prevent tumour progression in mice [225]. The DNA repair endonuclease XPF-ERCC1 is regulated through the DUB USP45 in response to UV damage. USP45 associates with ERCC1 and depletion results in increased levels of ubiquitylated ERCC1, leading to hypersensitivity to UV irradiation [226]. In cells, USP45 knockout, rather than leading to increased ERCC1 proteolysis, causes aberrant ERCC1 translocation to DNA damage foci, likely accounting for defective DNA repair.

BER is an essential repair pathway to remove lesions due to deamination, oxidation, and alkylation of DNA caused by IR or reactive oxygen species [227,228]. The removal of the damaged base by specific glycosylases leads to formation of an abasic site (AP site) that is cleaved by an AP endonuclease (APE1) resulting in a single-strand break, which is repaired by a complex including DNA polymerase β (beta), XRCC1, and DNA ligase III that are recruited in a PARP1-dependent manner. This process is known as single nucleotide or short-patch BER, while long-patch BER relies on the activity of RECQ1, RPA, XPF-ERCC1, and FEN1, which can remove the 5'-flap structure in a PCNA-dependent manner before filling in by polymerase and DNA Ligase I [229–231]. Pol β forms a central node to the BER process, coupling AP lyase and polymerase activities, and levels are tightly regulated to prevent increased mutagenesis and cancer susceptibility. Pol β levels are normally in proportion to the level of damage, with excess pol β targeted for proteasome-mediated degradation through sequential mono-ubiquitylation then polyUb chain formation by the E3 ligases Mule and CHIP, respectively. USP47 has been identified as the DUB counteracting this process to maintain proper levels of pol β, with depletion resulting in reduced levels and deficient BER [232] (Figure 3C).

A DUB complex involved in repair of DNA alkylation

Alkylation of DNA is a frequent mutagenic event caused by endogenous agents, such as S-adenosyl methionine, or external chemicals such as methyl methanesulfonate. In humans, ALKB2 and ALKB3 are the prominent oxidative demethylases involved to remove the modification, with ALKB2 preferring methylated dsDNA as a substrate and ALKB3 for ssDNA or RNA [233]. The stability of these enzymes is regulated by Ub signalling and the proteasome, and the OTU-family DUB, OTUD4, has been shown to interact with both ALKB proteins. However, the DUB activity of OTUD4 is not required to stabilise ALKB2/3, suggesting it acts as a scaffold for other enzymes to prevent proteolysis [234,235]. USP7 and USP9X were further identified to be associated with OTUD4 and ALKB3, with overexpression of wildtype, but not mutant, USP7 or USP9X suppressing the ubiquitylation and subsequent degradation of ALKB3 in the presence of OTUD4 (Figure 3C). Levels of these oxidative demethylases are kept at low levels under normal conditions, with the response to DNA alkylation requiring a very rapid response and stabilisation by this DUB complex.

DUB regulatory pathways of Ub signalling in genome stability tend to vary widely, given the complexity of the Ub code and the number of DDR pathways and factors involved. However, there are common themes, with DUB activity able to stabilise proteins preventing proteasome-mediated degradation, altering of binding interfaces to recruit specific repair factors, and altering the localisation or activity of the protein(s) to ensure proficient repair. Other UBLs, such as SUMO, NEDD8, UFM1, and ISG15 [19,30], have been implicated in genome stability pathways and as such, deconjugating enzymes play a regulatory role in these processes. As with Ub, several UBLs are linked to the p53/MDM2 signalling axis, which is not described in detail here but reviewed elsewhere [19,236]. Instead, direct deUBLylation of DDR factors will be explored.

DeSUMOylation

SUMO has been established to have a critical regulatory role in the DDR. There are several SUMO paralogues within the human genome, with SUMO2 and SUMO3 sharing 97% sequence similarity, while SUMO1 shares only 47% similarity to SUMO2. SUMO4 and SUMO5/SUMO1P1 have also been reported [237]. Similar to Ub, SUMO1-3 can be conjugated to target substrates through an enzymatic cascade with an E1 (SAE1/2), E2 (Ubc9, also known as UBE2I) and several E3s (e.g. PIAS-family proteins), while removal is predominantly carried out by sentrin-specific proteases (SENPs) [238,239]. SUMO is highly conserved and similarly to Ub, is expressed as an immature precursor that is processed by SENP1 to enable conjugation to substrate lysine residues, with the possibility of polySUMO2/3 chains being formed, predominantly via K11. SENP1-3 and SENP5 are related to yeast Ulp1 in having preferred C-terminal hydrolase activity, with SENP6/7 related to Ulp2 and having preferred isopeptidase activity to clear longer SUMO2/3 chains. Other classes of SUMO proteases include DeSI-1/2 (DeSUMOylated Isopeptidase 1/2) and USPL1, with the latter localised to Cajal bodies and having isopeptidase activity.

Regarding the role of SUMOylation in genome stability, SUMO1 and SUMO2/3 can be detected in IR-induced foci (IRIF), laser-induced damage, DSBs, and can be precipitated from damaged chromatin, with SUMOylation known to play a role in HR and NHEJ pathways [158,240–243]. Several targets are known, such as MDC1 [244], RPA [242,245], FANCI-D2 [246,247], with the outcome of the target SUMOylation often to produce an altered recruitment platform, such as for SIM-containing proteins or STUbLs like RNF4, the latter highlighting the interplay between Ub and SUMO signalling in genome stability [248]. Indeed, activation of RNF4 through binding to polySUMO chains via its tandem SIMs can result in the formation of Ub-SUMO hybrids that is coupled to VCP/p97-mediated protein extraction from damaged chromatin and often resulting in proteasome-mediated degradation [249–251].

SUMO proteases tend to prevent the formation of extended chains and various SENP-family members have been identified to have key roles in DDR pathways. SUMO availability, through maturation or recycling of free SUMO from conjugates, enables proper HR and NHEJ signalling [242,252,253]. Loss or knockdown of SENP6 dramatically increases the level of long SUMO2/3-conjugates in the cell and enlarged SUMO foci at PML bodies, which can be rescued by supplying exogenous SUMO isoforms [254,255]. The non-redundancy, altered localisation, and preferences for SUMO protease activity mean it is not surprising that knockdown of individual SENP-family enzymes results in specific HR or NHEJ deficiencies [242,254,255].

SENP7 in DSB repair and chromatin relaxation

One example is SENP7, which is required for HR-mediated repair of DSBs, with a role in chromatin relaxation to allow proficient DNA repair. KRAB-associated protein 1 (KAP1) is a heterochromatin protein that is SUMOylated in its C-terminal bromodomain (Figure 4A). Upon SUMOylation, KAP1 interacts with the chromatin remodeller CHD3 and the histone methyltransferase SETDB1 via their SIM domains [254]. CHD3 is a chromodomain-containing helicase that binds and distorts nucleosome DNA while SETDB1 is a H3K9 histone methyltransferase. Chromatin compaction by CHD3 and H3K9 trimethylation by SETDB1 results in heterochromatin formation and propagation via the heterochromatin protein, HP1α. Upon DNA damage, however, activated ATM releases the repressive KAP1-HP1α interaction by phosphorylation at S824 adjacent to the KAP1 bromodomain [256]. This damage-dependent phosphorylation also disrupts the SUMO-SIM interaction between SUMOylated KAP1 and CHD3, thereby promoting chromatin decompaction to allow efficient DNA repair. Depletion of SENP7, and therefore hyper-SUMOylated KAP1, fails to relax chromatin in response to damaging reagents due to increased chromatin retention of CHD3, preventing HR-mediated repair. SUMOylated KAP1 is also counteracted by the STUbL RNF4 via a concomitant interaction with the phosphorylated S824, with the resulting ubiquitylation leading to VCP/p97-mediated extraction and subsequent degradation of KAP1, providing a further mechanism for chromatin decompaction [257,258]. Therefore, while SUMO modifications can promote repressive chromatin sometimes advantageous in DDR-related transcriptional regulation, proficient repair in heterochromatin regions requires the removal of these marks either through STUbL-mediated degradation or via direct deSUMOylation.

Overview of the role of ULPs in DDR signalling pathways.
Figure 4.
Overview of the role of ULPs in DDR signalling pathways.

(A) The role of SENPs (SUMO ULPs) and the CSN (NEDD8 ULP) in DSB repair signalling. SENP6 is required to regulate SUMO2/3 levels on critical factors for DSB repair such as BRCA1-BARD1, 53BP1, BLM, XPF-ERCC1, and RPA to maintain active levels in an RNF4- and VCP/p97-dependent manner. SENP7 can deSUMOylate KAP1 to release its interaction with the chromatin remodeller, CHD3, which, along with phosphorylation by ATM, ensures chromatin relaxation in response to DSBs within heterochromatin regions. SENP2 and ATXN3 regulate the chromatin retention of MDC1 upon DNA damage in an RNF4- and VCP/p97-dependent manner, ensuring DDR signalling to repair the damage. The COP9 signalosome (CSN) is recruited to DSBs in a NEDD8-dependent manner, with CSN3 phosphorylated by ATM. CSN can remove NEDD8 modifications from Cullin-RING ligases (CRLs), histones, and other proteins during the DDR. (B) UFMylation of MRE11 and histone H4 in response to DNA damage occurs via the E3 ligase UFL1, which is recruited to DSBs through an interaction with the MRN complex at DSBs. H4 UFMylation enables downstream recruitment of the acetyltransferase TIP60 (via SUV39H1-mediated H3K9me3 formation), which acetylates ATM thereby promoting its activation and interaction with MRN. ATM kinase phosphorylation of itself and H2AX at S139 amplifies the DDR signalling for repair by HR. ATM-mediated phosphorylation of S462 on UFL1 further enhances UFL1 activity, resulting in a positive feedback loop. Recruitment of one of the two known UFM1 ULPs, UfSP2, to DSBs is prevented by inhibitory ATM-mediated phosphorylations at its S374/S381 residues, which are removed by the WIP1 phosphatase, allowing UfSP2 to bind MRN to deUFMylate H4 and dampen the ATM DDR signal upon successful repair. (C) During S phase, DNA damage blocking or stalling of the replication machinery leads to mono-ubiquitylation of PCNA at K164 by RAD6/RAD18, resulting in recruitment of more error-prone Y-family TLS polymerases and bypass of the lesion. To recycle unmodified PCNA and resume normal replicative processes, the E3 ligase EFP/TRIM25 can attach ISG15 to K168 on PCNA, leading to USP10-mediated removal of the mono-Ub on PCNA. EFP/TRIM25 can ISGylate K164, likely to prevent re-addition of Ub, before both ISG15 modifications are removed by USP18. How this mechanistically works in parallel with the established action of USP1-UAF1 is unknown. DUB/ULP activity is represented by dashed arrows.

Figure 4.
Overview of the role of ULPs in DDR signalling pathways.

(A) The role of SENPs (SUMO ULPs) and the CSN (NEDD8 ULP) in DSB repair signalling. SENP6 is required to regulate SUMO2/3 levels on critical factors for DSB repair such as BRCA1-BARD1, 53BP1, BLM, XPF-ERCC1, and RPA to maintain active levels in an RNF4- and VCP/p97-dependent manner. SENP7 can deSUMOylate KAP1 to release its interaction with the chromatin remodeller, CHD3, which, along with phosphorylation by ATM, ensures chromatin relaxation in response to DSBs within heterochromatin regions. SENP2 and ATXN3 regulate the chromatin retention of MDC1 upon DNA damage in an RNF4- and VCP/p97-dependent manner, ensuring DDR signalling to repair the damage. The COP9 signalosome (CSN) is recruited to DSBs in a NEDD8-dependent manner, with CSN3 phosphorylated by ATM. CSN can remove NEDD8 modifications from Cullin-RING ligases (CRLs), histones, and other proteins during the DDR. (B) UFMylation of MRE11 and histone H4 in response to DNA damage occurs via the E3 ligase UFL1, which is recruited to DSBs through an interaction with the MRN complex at DSBs. H4 UFMylation enables downstream recruitment of the acetyltransferase TIP60 (via SUV39H1-mediated H3K9me3 formation), which acetylates ATM thereby promoting its activation and interaction with MRN. ATM kinase phosphorylation of itself and H2AX at S139 amplifies the DDR signalling for repair by HR. ATM-mediated phosphorylation of S462 on UFL1 further enhances UFL1 activity, resulting in a positive feedback loop. Recruitment of one of the two known UFM1 ULPs, UfSP2, to DSBs is prevented by inhibitory ATM-mediated phosphorylations at its S374/S381 residues, which are removed by the WIP1 phosphatase, allowing UfSP2 to bind MRN to deUFMylate H4 and dampen the ATM DDR signal upon successful repair. (C) During S phase, DNA damage blocking or stalling of the replication machinery leads to mono-ubiquitylation of PCNA at K164 by RAD6/RAD18, resulting in recruitment of more error-prone Y-family TLS polymerases and bypass of the lesion. To recycle unmodified PCNA and resume normal replicative processes, the E3 ligase EFP/TRIM25 can attach ISG15 to K168 on PCNA, leading to USP10-mediated removal of the mono-Ub on PCNA. EFP/TRIM25 can ISGylate K164, likely to prevent re-addition of Ub, before both ISG15 modifications are removed by USP18. How this mechanistically works in parallel with the established action of USP1-UAF1 is unknown. DUB/ULP activity is represented by dashed arrows.

Close modal

SENP2 regulates MDC1 chromatin retention

While Ub signalling is widely known to regulate the decision between HR and NHEJ in repairing DSBs, the deSUMOylase SENP2 has also been identified as a key factor in these processes. As noted above, SENP-family proteases are required to maintain the supply of SUMO proteins available for conjugation to target proteins and thereby promote DDR signalling, such as to promote HR. Knockdown of SENP2 results in reduced RAD51 loading and reduced HR proficiency in cells in response to CPT and olaparib, while overexpression of SENP2 leads to increased NHEJ signalling due to the increased chromatin retention of MDC1, resulting in increased chromosome aberrations [255]. SENP2 is constitutively active and retained on chromatin to prevent excessive polySUMO2/3 chains forming on MDC1, thereby preventing engagement by the STUbL RNF4, which otherwise would extract MDC1 from chromatin and target it for degradation via VCP/p97. Loss of SENP2 leads to reduced chromatin bound MDC1, leading to increased sensitivity to IR or other DSB-inducing agents (Figure 4A). The VCP/p97-associated DUB, ATXN3, is also recruited to SUMO at sites of DSBs and is thought to counter RNF4-mediated poly-ubiquitylation to protect MDC1 [259] and RNF8 [260] (Figure 4A). Therefore, the levels of SENP2 need to be precisely regulated and it is worth noting that SENP2 is one of several genes, along with RNF168 and USP13, on the amplified region of chromosome 3q found in many aero-digestive tract cancers for example.

SENP6 enables controlled extraction of proteins from chromatin in the DDR

As an extension to the activity of SENP2 on MDC1, another deSUMOylase, SENP6, also regulates the chromatin retention of the FANCI-D2 heterodimer (Figure 3A). As described above, ATR-mediated phosphorylation and FA core complex-mediated ubiquitylation enables regulated recruitment of FANCI-D2 to sites of ICL damage. To enable proficient repair, the FANCI-D2 clamp needs to be removed or extracted from chromatin, and this is enabled through PIAS4-mediated polySUMO2/3 chain formation leading to RNF4 recruitment and subsequent extraction via VCP/p97 [246]. SENP6 depletion reduces the chromatin retention of FANCI-D2 and may lead to premature removal of FANCI-D2 via RNF4, reducing efficiency of ICL repair. Once again therefore, proper expression and recruitment of deSUMOylases is key to enable proficient DDR signalling.

More recently, the roles of SENP6 in genome stability, in addition to those described above for SUMO availability and FANCI-D2 chromatin retention, have been expanded [261]. Through screening of proteins modified with polySUMO2/3 chains upon SENP6 depletion, an array of DDR proteins, including BRCA1-BARD1, 53BP1, BLM, and XPF-ERCC1, were shown to be regulated through polySUMOylation and RNF4-mediated chromatin extraction (Figure 4A). SENP6 is proposed to maintain these proteins in a hypo-SUMOylated state in basal conditions and counteracts excessive polySUMO2/3 chain formation in response to HU-induced replication stress. Reduced SENP6 levels led to dysregulated recruitment or aberrant retention of proteins at damaged sites, increased SUMO2/3 levels at UV or IR-induced damage sites, and reduced DNA repair proficiency.

Generally, deSUMOylases seem to act as buffers or thermostats to maintain minimal SUMOylation of DDR-related proteins, enabling a rapid response to DNA damaging agents. Excessive SUMOylation can lead to premature removal of proteins from chromatin via RNF4 and VCP/p97, while excessive deSUMOylation can result in the aberrant retention of proteins at sites of DNA damage (e.g. MDC1) leading to more error-prone repair pathways. The role of SUMOylation in chromatin compaction and possibly in nuclear condensate formation [261] also suggests a more global method for regulating the availability of DDR proteins and access to sites of DNA damage to ensure efficient repair. The SENP-family of proteins seem to be central to regulate this, with their expression and constitutive activity crucial to maintain genome stability. Given the global role of SUMOylation in a multitude of signalling pathways, it would be difficult to design related inhibitors to specific SUMO proteases but their expression levels, either at the level of RNA or protein, or cellular localisation could be used as potential markers for disease-driving signalling pathways to explore. Protein SUMOylation tends to act in parallel to Ub-signalling in the context of genome stability, often via RNF4 and VCP/p97. However, for other UBLs, this is not always the case, meaning ULPs for these other modifications likely regulate the DDR via other mechanisms.

DeUFMylation

The latest UBL to be identified is UFM1, an 85 amino acid long, 9.1 kDa protein [262]. As with other UBLs, UFM1 needs to be processed to a mature form by removing two C-terminal amino acids (S84 and C85) to expose a conserved glycine (G83 in humans) enabling conjugation to target proteins. UFM1 substrate conjugation occurs via a stepwise manner analogous to Ub and SUMO with an E1 (UBA5), E2 (UFC1) and E3 (UFL1 with cofactors UFBP1 and CDK5RAP3) enzymatic cascade, with polyUFM1 chains able to form via an internal lysine at amino acid position 69 [19,263–265]. Two UFM1-specific cysteine proteases have been identified, UfSP1 and UfSP2, to reverse UFM1 conjugation on target substrates, possibly to regulate UFC1 activity, and to process the expressed precursor form [266].

UFMylation has predominantly been linked to roles in endoplasmic reticulum (ER) homeostasis, fatty acid metabolism, autophagy, neurodevelopment, and liver development [263,264,267,268]. A major substrate for UFMylation is the 60S ribosomal subunit RPL26, which occurs on stalled ribosomes at the ER. Structural evidence suggests the role of UFM1 modification in this context is to release 60S ribosomes from the SEC61 translocon to regulate protein homeostasis at the ER [269,270]. UFM1 substrates have additionally been identified that have roles in DDR signalling and telomere maintenance pathways, including MRE11 [271] and histone H4 [272]. The UFM1-specific E3 ligase UFL1 is proposed to be recruited to IR-induced sites of DSBs, where it can interact with, and UFMylate MRE11 at K281 and K282. MRE11 UFMylation is thought to stabilise the MRN complex at sites of damage to activate the ATM kinase, while recruited UFL1 can also UFMylate histone H4 at K31, aiding to localise TIP60 to DSBs to further increase ATM activity. A positive feedback loop to amplify the ATM activation signal is thought to exist, with ATM-mediated S462 phosphorylation on UFL1 elevating its E3 activity [272] (Figure 4B). Furthermore, MRE11 mutants at target UFMylation sites abrogated its interaction with telomere repeat-binding protein, TRF2, resulting in reduced telomere length in HeLa cells and haematopoietic zebrafish cells [273]. While no DSB repair defects linked to ATM activation and DSBs were detected in this case, it is possible differences in cell lines used and variable context requirements exist for UFMylation in DDR pathways, although further investigation would be required.

UfSP2 regulation of H4 UFMylation

As opposed to other UBLs that are conjugated to substrates via a C-terminal di-glycine motif, UFM1 exhibits a C-terminal VG, masking it from other DUBs and ULPs ensuring specificity and fidelity in UFM1 signalling pathways. UfSP1 and UfSP2 share close structural homology within their catalytic domains, but several regulatory loops distinguish their proteolytic activities, suggesting they are regulated via allosteric means. Furthermore, an N-terminal extension in UfSP2 localises it to the ER membrane via ODR4 while UfSP1 is predominantly cytosolic, likely establishing alternative substrates, with UfSP1 important for UFM1 maturation and removing the auto-inhibitory UFC1 UFMylation, and UfSP2 for regulating UFMylated ribosomes [265]. Given the sub-cellular localisation of UfSPs, their roles in genome stability are unclear. A recent study, from the same group that identified H4 UFMylation, proposed a role for UfSP2 in modulating ATM activation upon IR-induced DSBs [274]. Like UFL1, recruitment was via the MRN complex but occurred at later time points following DNA damage due to an ATM-dependent phosphorylation on UfSP2 at S374 and S381 reducing the binding to MRE11. However, this could be reversed via the phosphatase WIP1, which enabled UfSP2 recruitment, H4 deUFMylation, and dampening of the ATM activation loop (Figure 4B). The mechanism of how UfSP2 is shuttled to DSB foci in the nucleus, and the kinetics between UFL1 and UfSP2 localisation and activity, will need further exploration.

DeISGylation

ISG15 is a 15 kDa protein that contains two UBL domains separated by a flexible linker. Like UFM1, ISG15 is expressed as a 165 amino acid, 17 kDa precursor, which is processed to the mature form through removal of the C-terminal 8 amino acids to expose a diglycine motif available for substrate conjugation. Modification of substrate proteins at target lysine residues with ISG15 is through an enzymatic cascade via an E1 (UBA7/UbE1L), E2 (UBE2L6/UBCH8), and one of three E3 ligases (HERC5, HHARI, EFP/TRIM25). USP18 [275,276], along with USP5, USP14, USP21 and recently USP16 and USP36, have been identified to have deISGylase activity to reverse the modification [277]. Critically, expression of ISG15 and the ISGylation machinery is stimulated by type I interferons, cytoplasmic nucleic acid detection pathways, and other cellular stresses, with ISG15 predominantly known for its role in the classical innate immune response during viral infections to limit viral replication [30,278,279]. ISG15 has more recently been identified by several groups as acting in response to replication-coupled DNA damage, with ISG15 expression and substrate ISGylation detected in response to UV light in certain cell types [280]. In addition, replication stress leading to the accumulation of cytoplasmic DNA has been proposed to activate innate immune signalling via the cGAS-STING-Tbk1 pathway, resulting in ISG15 expression and target protein ISGylation, including of DNA replication-associated proteins, preventing replication fork stalling and genome instability [281].

USP18 ensures high-fidelity replication resumes following TLS

In the DDR, ISG15 modifications have been identified to overlap with Ub-related signalling nodes for replication-coupled DDR pathways, possibly linked to observations for induced expression of the ISGylation machinery as a result of replication stress [19,30,281,282]. TLS facilitates replication past DNA lesions induced by UV light, for example, with a key event being PCNA mono-ubiquitylation at K164 to promote displacement of replicating polymerases with lower-fidelity polymerases. The ISG15 E3 ligase, EFP/TRIM25, was proposed to bind mono-ubiquitylated PCNA leading to ISGylation at K168, recruitment of USP10 to remove the Ub mark, and causing the release of TLS polymerases and other factors, such as REV1, from PCNA to terminate error-prone replication [280]. ISGylation at K164 likely prevents subsequent TLS initiation before USP18 removes the ISG15 marks to enable reloading of replicative polymerases and resumption of high-fidelity DNA replication. USP18-mediated removal of the ISG15 mark is a key step following damage and ISGylation is critical for preventing excessive UV-mediated mutagenesis and genome instability [280,283] (Figure 4C). How this ISG15- and USP10-mediated signalling functions in parallel to the more established USP1-UAF1 mediated regulation of PCNA mono-ubiquitylation will need further exploration.

DeISGylases may regulate K63-linked polyUb formation in DDR signalling

ISG15 can form mixed chains with Ub through conjugation onto K29 of Ub, indicating a directly overlapping role in Ub signalling [284], while ISGylation of Ubc13/UBE2N may also regulate the DDR. Ubc13 is an E2 conjugating enzyme important for several DDR signalling pathways by generating K63-linked polyUb chains as recruitment platforms for multiple DSB repair factors [285]. ISGylation at K92 of Ubc13 inhibits its activity by disrupting its ability to form a thioester bond to Ub [286]. No direct link to the DDR has been identified for proper removal of ISG15 from Ubc13 as K63-linked polyUb is required for numerous cell signalling pathways but given the intersection between the immune response and DDR signalling, it would be intriguing to see how deISGylation may regulate these processes.

USP18 is predominantly the deISGylase thought to act in cells particularly as its expression aligns with the interferon response and ISG15. Other USP-family members such as USP5, USP14, USP16, USP21, and USP36 [277,287], have all shown deISGylase activity in vitro on top of their primary protease activity against other UBLs and/or Ub. Whether these moonlighting activities are relevant to ISG15 biology remains to be seen, but the development of specific tools to probe activity in response to DNA damage will likely aid future work.

DeNEDDylation

NEDD8 is a UBL expressed as an 81 amino acid precursor before processing by DEN1 (also known as NEDP1 or SENP8) to remove the last 5 amino acids to produce the mature form. NEDDylation is most commonly known to activate Cullin-RING ligases (CRLs) by modification of the core Cullin subunit to enhance their E3 Ub ligase activities, reviewed in more detail elsewhere [288]. NEDD8 has a single E1 (NAE1) and two E2 conjugating enzymes (UBC12, also known as UBE2M, and UBE2F) to transfer to the target Cullin subunit [289]. With respect to genome stability, NEDD8 conjugates are enriched at sites of DNA damage, with RNF111-mediated NEDDylation of H4, to promote RNF168 recruitment, and CtIP regulating the response to DSBs [23,34,290–294]. The COP9 signalosome (CSN) consists of 8 subunits required for the removal of NEDD8 from CRLs, resulting in reduced E3 ligase and auto-ubiquitylation activity. CSN5 is the active JAMM-type metalloprotease within this complex. CSN3 is a substrate for the ATM kinase and the CSN has roles in NER and protection against DSBs, with CSN recruited to DSBs in a NEDD8-dependent manner, indicating a key role in genome stability signalling pathways [293] (Figure 4A). Furthermore, CSN7B can be recruited to sites of DNA damage caused by MMC or IR to remove NEDD8 from CRLs, and possibly other E3 Ub ligases or histone proteins. Knocking out CSN7B, as well as the use of NEDD8-relevant inhibitors such as MLN4924 (NAE1 inhibitor) and CSN5i-3 [295], reduced ATM-dependent signalling and apoptosis, while shifting repair preference towards alt-NHEJ via PARP1 over HR, so altering genome stability pathway choice in response to DSBs [296]. Alt-NHEJ is an error-prone backup repair pathway that relies on PARP1 recognition of the DSB to recruit the MRN complex and CtIP, resulting in short-range resection of the DNA (2–20 nucleotides), filling in by DNA polymerase Q, and sealing of the nick by XRCC1 and DNA Ligase III [297,298].

Redundancy in DUBs/ULPs

The identification of many DUBs, plus additional ULPs, suggests that proper regulation of Ub and UBL modifications is critical to the DDR. Several DUBs, such as USP7, USP1, BAP1, appear to play distinct roles in multiple repair pathways, while critical Ub/UBL signalling nodes, such as modified PCNA and H2A, are targets for multiple DUBs/ULPs. The latter evidence of functional redundancy likely provides a safety net for genome stability, with backup systems to ensure damage is repaired. Another possibility is that multiple DUBs act in temporally or spatially different contexts for genome stability, depending on other signalling cues or the type of damage. For example, different DUBs may act during G0 or G1 versus S/G2 phase of the cell cycle (e.g. USP11), or as noted for the KAP1-CHD3 interaction and relevant to INO80, the chromatin architecture or local environment is likely important to dictate the Ub/UBL signalling events to ensure efficient repair processes.

The complexity of the Ub/UBL code and the likely presence of mixed or branched Ub/UBL chains [299–302] further lends credence that multiple DUBs/ULPs, with different enzyme kinetics or substrate specificities, are required to ensure regulated signalling for repairing DNA damage and to prevent unnecessary spreading of repair processes (e.g. hyper-resection). POH1 and USP5 possibly acting in concert is one example, while the K63-specific activity of ZUP1 may potentially allow Ub chain editing by neighbouring E3 ligases and/or regulated extraction of target proteins from chromatin via VCP/p97. Relevant for VCP/p97 is the combined activity of DUB and SENP partners to regulate the STUbL, RNF4, required to target proteins for degradation. More recent evidence for ISG15 and UFM1 modifications overlapping with that of Ub signalling, such as on PCNA or the MRN complex, lends further speculation that altered activities of DUBs and deUBLylases need tight regulation to ensure co-ordinated removal of the modifications. Additional enzyme activities for non-protein [15] or non-lysine [16] ubiquitylation may also need to be considered.

Some DUBs, such as OTUD4 and OTUB1, promote efficient DDR signalling through non-catalytic roles, either as allosteric regulators of components or as scaffolds to recruit other DUBs to sites of damage. Indeed, large multi-subunit DUB/ULP complexes, such as BRCA1-A, PR-DUB, and CSN, are likely regulated on multiple levels with respect to cellular localisation, substrate specificity, and allosteric regulation. Recent developments in single particle cryo-EM alongside high-quality sample preparation and biophysical techniques [303] allows specific questions about these molecular machines to be answered to clarify their role in cell signalling, such as for genome stability. Preparing uniform and designer substrates may also enable any questions about DUB/ULP target specificity to be addressed, which has helped disentangle some of the apparent DUB redundancy observed for H2A ubiquitylation in DSB repair for example [122].

Regulating the regulators

Many DUBs and ULPs tested in vitro can show varying degrees of activity or specificity in their ability to remove Ub/UBL marks. How they are localised to sites of DNA damage and how they engage with specific substrates is more difficult to ascertain, as few DUBs/ULPs contain cellular localisation signals or substrate binding domains. As noted above, some DUBs exist as larger multi-subunit complexes to ensure correct recruitment and activity, such as BRCC36 that is recruited to DSBs via RAP80. Other DUBs likely have interacting partners such as ZUP1, with several UBDs to restrict its activity to K63-linked polyUb chains, interacting with the RPA complex to localise it to sites of ssDNA. Some DUBs are directly modified, either with Ub/UBLs or via DDR-mediated phosphorylation by ATM/ATR/DNA-PKcs, to enable recruitment or shuttling to sites of damage, such as for VCPIP1 and BAP1. ATR-mediated phosphorylation of the USP1-UAF1 complex and of its substrate, ubiquitylated FANCI-D2, has also been shown to regulate the formation of the enzyme-substrate complex for efficient Ub removal.

Related to USP1-UAF1, is how some DUBs function in multiple different DDR signalling pathways. USP1 can be regulated through auto-cleavage and via its UAF1 cofactor, which itself is a cofactor for other DUBs USP12 and USP46 not involved in genome stability signalling [166]. USP7 is another DUB implicated in various pathways of DNA damage signalling, including for p53-MDM2, HR, NER, alkylation repair, plus others, with scaffolding or interacting partners helping to recruit its activity in some cases.

Technology development for DUB/ULP investigation

Utilisation of genetic screens, such as siRNA or CRISPR-Cas9, has identified many of the DUBs and ULPs described here, with improvements in technology enabling genetic and chemo-genetic interactions to be discovered [304–308]. CRISPR screens have helped overcome some of the potential off-target effects of RNAi screens, which has identified or confirmed major signalling nodes within DDR signalling, while subtle regulators of the pathways, such as E3 ligases or DUBs/ULPs, either show some redundancy or have cell-type specific effects [303,307]. Expanding on these monogenic screens to identify synthetic lethal genetic interactions, such as for USP1 and BRCA1 [167], may expand the repertoire of potentially druggable targets in particular genetic backgrounds, whether through mono- or combination therapy. Alongside the discovery of the factors involved, increasing knowledge of the mechanisms by which they act in the DDR, and how they remove the modification from substrates, has been enabled by recent advances in technology development [303]. Progress in mass spectrometry techniques and proteomics analysis, alongside diglycine-enrichment and other tools, has allowed detection of the Ub/UBL linkages relevant to DNA damage signalling. Coupling this to semi-synthetic and designer approaches for producing Ub/UBL substrates, such as for UBLs [277] and mixed/branched chains [309,310], has helped to clarify the activity and specificity of DUBs and ULPs. Further enhancement in structure prediction using AlphaFold [311–314] and the rapid progress in structural biology using cryo-EM alongside complementary techniques, has provided hitherto unmatched detail regarding the regulation and activity of DUBs/ULPs.

DUB/ULP targets in drug discovery

Selective inhibition of DUBs/ULPs is an active avenue for investigation regarding anti-cancer therapeutics. The involvement of USP1 and USP7 in multiple pathways allows specific targeting of these DUBs to sensitise cells to chemotherapy due to the dysregulation of multiple parallel DDR signalling pathways. For example, ML323 is a selective USP1-UAF1 inhibitor that has shown promise to inhibit FA and TLS signalling to maximise cancer cell cytotoxicity with few off-target effects [315]. Recent structural characterisation of inhibitor binding will further progress drug development in this case, with additional drugs currently in Phase 1 clinical trials such as KSQ-4279 [53]. Indeed, the increased mechanistic and structural characterisation of DUBs/ULPs with their targets or inhibitors, combined with deep sequencing of patient samples, either at the genomic or expression level, will likely enable more precise patient stratification with reduced likelihood of developing resistance. Current efforts to inhibit DUBs/ULPs are reviewed elsewhere in more detail [53,142,316–318]. Overall, an enhanced understanding of Ub/UBL signalling via DUBs and ULPs in DDR signalling will provide a stable grounding to develop targeted strategies for treating human diseases, such as cancers.

Data availability is not applicable to this article as no new data were created or analysed in this study.

The authors declare that there are no competing interests associated with the manuscript.

B.M.F. and C.K.S. acknowledge funding via an MRC research grant [MR/X008754/1]. Z.W. is funded by a BBSRC DTP studentship. Figures were made using InkScape (https://inkscape.org/).

Open access for this article was enabled by the participation of University of Manchester in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.

Benjamin Foster: Conceptualisation, Writing — original draft, Writing — review and editing. Zijuan Wang: Writing — review and editing. Christine Schmidt: Supervision, Funding acquisition, Writing — review and editing.

AP

abasic site

ATG

autophagy-related protein

ATM

ataxia-telangiectasia mutated

ATR

ataxia telangiectasia and Rad3-related protein

BAP1

BRCA1-associated protein 1

BER

base excision repair

CMG

CDC45-MCM-GINS

CPT

camptothecin

CSN

COP9 signalosome

DDR

DNA damage response

DeSI

deSUMOylating isopeptidase

DPC

DNA-protein cross-link

DSB

double-strand break

dsDNA

double-stranded DNA

DUB

deubiquitylase

FA

Fanconi anaemia

FAT10

human leukocyte antigen F locus adjacent transcription 10

FUBI

Finkel-Biskis-Reilly murine sarcoma virus (FBR-MuSV) ubiquitously expressed

GG-NER

global genome nucleotide excision repair

HR

homologous recombination

HU

hydroxyurea

ICL

interstrand cross-link

IR

ionising radiation

IRIF

ionising radiation induced foci

ISG15

interferon-stimulated gene 15

JAMM

JAB1/MPN/Mov34

KAP1

KRAB-associated protein 1

MINDY

motif interacting with ubiquitin (MIU)-containing novel DUB family

MJD

Machado-Joseph domain-containing protein

MMC

mitomycin C

MRN

MRE11-RAD50-NBS1

NEDD8

neural precursor cell expressed and developmentally down-regulated 8

NER

nucleotide excision repair

NHEJ

non-homologous end-joining

OTU

otubain domain-containing protease

PCNA

proliferating cell nuclear antigen

PIKK

phosphoinositide 3-kinase-like kinase

PIP

PCNA interacting peptide

PR-DUB

polycomb-repressive DUB

PTM

post-translational modification

RPA

replication protein A

SENP

sentrin-specific protease

SIM

SUMO-interacting motif

SSA

single-strand annealing

ssDNA

single-stranded DNA

STUbL

SUMO-targeted ubiquitin ligase

SUMO

small ubiquitin-like modifier

TC-NER

transcription-coupled nucleotide excision repair

TLS

translesion synthesis

TS

template switching

UAF

USP1-associated factor 1

Ub

ubiquitin

UBD

ubiquitin binding domain

UBL

ubiquitin-like protein

UCH

ubiquitin C-terminal hydrolase

UFM1

ubiquitin-fold modifier 1

UIM

ubiquitin-interacting motif

ULP

ubiquitin-like protease

URM1

ubiquitin-related modifier 1

USP

ubiquitin-specific protease

UV

ultraviolet

VCP

valosin-containing protein

VCPIP1

valosin-containing protein p97/p47 complex-interacting protein 1

ZUP1

zinc finger-containing ubiquitin peptidase 1

1
Hanahan
,
D.
and
Weinberg
,
R.A.
(
2011
)
Hallmarks of cancer: the next generation
.
Cell
144
,
646
674
2
Ciccia
,
A.
and
Elledge
,
S.J.
(
2010
)
The DNA damage response: making it safe to play with knives
.
Mol. Cell
40
,
179
204
3
Jackson
,
S.P.
and
Bartek
,
J.
(
2009
)
The DNA-damage response in human biology and disease
.
Nature
461
,
1071
1078
4
Blackford
,
A.N.
and
Jackson
,
S.P.
(
2017
)
ATM, ATR, and DNA-PK: the trinity at the heart of the DNA damage response
.
Mol. Cell
66
,
801
817
5
Pickart
,
C.M.
(
2001
)
Mechanisms underlying ubiquitination
.
Annu. Rev. Biochem.
70
,
503
533
6
Pickart
,
C.M.
and
Eddins
,
M.J.
(
2004
)
Ubiquitin: structures, functions, mechanisms
.
Biochim. Biophys. Acta
1695
,
55
72
7
Swatek
,
K.N.
and
Komander
,
D.
(
2016
)
Ubiquitin modifications
.
Cell Res.
26
,
399
422
8
Komander
,
D.
and
Rape
,
M.
(
2012
)
The ubiquitin code
.
Annu. Rev. Biochem.
81
,
203
229
9
Kulathu
,
Y.
and
Komander
,
D.
(
2012
)
Atypical ubiquitylation - the unexplored world of polyubiquitin beyond Lys48 and Lys63 linkages
.
Nat. Rev. Mol. Cell Biol.
13
,
508
523
10
Rape
,
M.
(
2018
)
Ubiquitylation at the crossroads of development and disease
.
Nat. Rev. Mol. Cell Biol.
19
,
59
70
11
Walser
,
F.
,
Mulder
,
M.P.C.
,
Bragantini
,
B.
,
Burger
,
S.
,
Gubser
,
T.
,
Gatti
,
M.
, et al. (
2020
)
Ubiquitin phosphorylation at Thr12 modulates the DNA damage response
.
Mol. Cell
80
,
423
436.e9
12
Mattiroli
,
F.
and
Penengo
,
L.
(
2021
)
Histone ubiquitination: an integrative signaling platform in genome stability
.
Trends Genet.
37
,
566
581
13
Hepowit
,
N.L.
,
Kolbe
,
C.C.
,
Zelle
,
S.R.
,
Latz
,
E.
and
MacGurn
,
J.A.
(
2022
)
Regulation of ubiquitin and ubiquitin-like modifiers by phosphorylation
.
FEBS J.
289
,
4797
4810
14
De Cesare
,
V.
,
Carbajo Lopez
,
D.
,
Mabbitt
,
P.D.
,
Fletcher
,
A.J.
,
Soetens
,
M.
,
Antico
,
O.
, et al. (
2021
)
Deubiquitinating enzyme amino acid profiling reveals a class of ubiquitin esterases
.
Proc. Natl Acad. Sci. U.S.A.
118
,
e2006947118
15
Sakamaki
,
J.I.
and
Mizushima
,
N.
(
2023
)
Ubiquitination of non-protein substrates
.
Trends Cell Biol.
33
,
991
1003
16
Kelsall
,
I.R.
(
2022
)
Non-lysine ubiquitylation: doing things differently
.
Front. Mol. Biosci.
9
,
1008175
17
Yau
,
R.
and
Rape
,
M.
(
2016
)
The increasing complexity of the ubiquitin code
.
Nat. Cell Biol.
18
,
579
586
18
Dikic
,
I.
and
Schulman
,
B.A.
(
2023
)
An expanded lexicon for the ubiquitin code
.
Nat. Rev. Mol. Cell Biol.
24
,
273
287
19
Da Costa
,
I.C.
and
Schmidt
,
C.K.
(
2020
)
Ubiquitin-like proteins in the DNA damage response: the next generation
.
Essays Biochem.
64
,
737
752
20
Cappadocia
,
L.
and
Lima
,
C.D.
(
2018
)
Ubiquitin-like protein conjugation: structures, chemistry, and mechanism
.
Chem. Rev.
118
,
889
918
21
Hochstrasser
,
M.
(
2009
)
Origin and function of ubiquitin-like proteins
.
Nature
458
,
422
429
22
Vertegaal
,
A.C.O.
(
2022
)
Signalling mechanisms and cellular functions of SUMO
.
Nat. Rev. Mol. Cell Biol.
23
,
715
731
23
Brown
,
J.S.
and
Jackson
,
S.P.
(
2015
)
Ubiquitylation, neddylation and the DNA damage response
.
Open Biol.
5
,
150018
24
Morris
,
J.R.
and
Garvin
,
A.J.
(
2017
)
SUMO in the DNA double-stranded break response: similarities, differences, and cooperation with ubiquitin
.
J. Mol. Biol.
429
,
3376
3387
25
Su
,
S.
,
Zhang
,
Y.
and
Liu
,
P.
(
2020
)
Roles of ubiquitination and SUMOylation in DNA damage response
.
Curr. Issues Mol. Biol.
35
,
59
84
26
Serbyn
,
N.
,
Bagdiul
,
I.
,
Noireterre
,
A.
,
Michel
,
A.H.
,
Suhandynata
,
R.T.
,
Zhou
,
H.
, et al. (
2021
)
SUMO orchestrates multiple alternative DNA-protein crosslink repair pathways
.
Cell Rep.
37
,
110034
27
Uckelmann
,
M.
and
Sixma
,
T.K.
(
2017
)
Histone ubiquitination in the DNA damage response
.
DNA Repair (Amst)
56
,
92
101
28
Aquila
,
L.
and
Atanassov
,
B.S.
(
2020
)
Regulation of histone ubiquitination in response to DNA double strand breaks
.
Cells
9
,
1699
29
Sekiguchi
,
M.
and
Matsushita
,
N.
(
2022
)
DNA damage response regulation by histone ubiquitination
.
Int. J. Mol. Sci.
23
,
8187
30
Sandy
,
Z.
,
da Costa
,
I.C.
and
Schmidt
,
C.K.
(
2020
)
More than meets the ISG15: emerging roles in the DNA damage response and beyond
.
Biomolecules
10
,
1557
31
He
,
M.
,
Zhou
,
Z.
,
Shah
,
A.A.
,
Zou
,
H.
,
Tao
,
J.
,
Chen
,
Q.
, et al. (
2016
)
The emerging role of deubiquitinating enzymes in genomic integrity, diseases, and therapeutics
.
Cell Biosci.
6
,
62
32
Pinto-Fernandez
,
A.
and
Kessler
,
B.M.
(
2016
)
DUBbing cancer: deubiquitylating enzymes involved in epigenetics, DNA damage and the cell cycle as therapeutic targets
.
Front. Genet.
7
,
133
33
Kee
,
Y.
and
Huang
,
T.T.
(
2016
)
Role of deubiquitinating enzymes in DNA repair
.
Mol. Cell Biol.
36
,
524
544
34
Garvin
,
A.J.
(
2019
)
Beyond reversal: ubiquitin and ubiquitin-like proteases and the orchestration of the DNA double strand break repair response
.
Biochem. Soc. Trans.
47
,
1881
1893
35
Li
,
Y.
and
Yuan
,
J.
(
2021
)
Role of deubiquitinating enzymes in DNA double-strand break repair
.
J. Zhejiang Univ. Sci. B
22
,
63
72
36
Tokarz
,
P.
and
Wozniak
,
K.
(
2021
)
SENP proteases as potential targets for cancer therapy
.
Cancers (Basel)
13
,
2059
37
Mevissen
,
T.E.T.
and
Komander
,
D.
(
2017
)
Mechanisms of deubiquitinase specificity and regulation
.
Annu. Rev. Biochem.
86
,
159
192
38
Clague
,
M.J.
,
Urbe
,
S.
and
Komander
,
D.
(
2019
)
Breaking the chains: deubiquitylating enzyme specificity begets function
.
Nat. Rev. Mol. Cell Biol.
20
,
338
352
39
Komander
,
D.
,
Clague
,
M.J.
and
Urbe
,
S.
(
2009
)
Breaking the chains: structure and function of the deubiquitinases
.
Nat. Rev. Mol. Cell Biol.
10
,
550
563
40
Reyes-Turcu
,
F.E.
,
Ventii
,
K.H.
and
Wilkinson
,
K.D.
(
2009
)
Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes
.
Annu. Rev. Biochem.
78
,
363
397
41
Ronau
,
J.A.
,
Beckmann
,
J.F.
and
Hochstrasser
,
M.
(
2016
)
Substrate specificity of the ubiquitin and Ubl proteases
.
Cell Res.
26
,
441
456
42
Sowa
,
M.E.
,
Bennett
,
E.J.
,
Gygi
,
S.P.
and
Harper
,
J.W.
(
2009
)
Defining the human deubiquitinating enzyme interaction landscape
.
Cell
138
,
389
403
43
Haq
,
S.
and
Ramakrishna
,
S.
(
2017
)
Deubiquitylation of deubiquitylases
.
Open Biol.
7
,
170016
44
Leznicki
,
P.
and
Kulathu
,
Y.
(
2017
)
Mechanisms of regulation and diversification of deubiquitylating enzyme function
.
J. Cell Sci.
130
,
1997
2006
45
Dayal
,
S.
,
Sparks
,
A.
,
Jacob
,
J.
,
Allende-Vega
,
N.
,
Lane
,
D.P.
and
Saville
,
M.K.
(
2009
)
Suppression of the deubiquitinating enzyme USP5 causes the accumulation of unanchored polyubiquitin and the activation of p53
.
J. Biol. Chem.
284
,
5030
5041
46
Blount
,
J.R.
,
Johnson
,
S.L.
and
Todi
,
S.V.
(
2020
)
Unanchored ubiquitin chains, revisited
.
Front. Cell Dev. Biol.
8
,
582361
47
MacDonald
,
E.
,
Urbe
,
S.
and
Clague
,
M.J.
(
2014
)
USP8 controls the trafficking and sorting of lysosomal enzymes
.
Traffic
15
,
879
888
48
Heideker
,
J.
and
Wertz
,
I.E.
(
2015
)
DUBs, the regulation of cell identity and disease
.
Biochem. J.
465
,
1
26
49
Basar
,
M.A.
,
Beck
,
D.B.
and
Werner
,
A.
(
2021
)
Deubiquitylases in developmental ubiquitin signaling and congenital diseases
.
Cell Death Differ.
28
,
538
556
50
Snyder
,
N.A.
and
Silva
,
G.M.
(
2021
)
Deubiquitinating enzymes (DUBs): regulation, homeostasis, and oxidative stress response
.
J. Biol. Chem.
297
,
101077
51
Wertz
,
I.E.
and
Wang
,
X.
(
2019
)
From discovery to bedside: targeting the ubiquitin system
.
Cell Chem. Biol.
26
,
156
177
52
Harrigan
,
J.A.
,
Jacq
,
X.
,
Martin
,
N.M.
and
Jackson
,
S.P.
(
2018
)
Deubiquitylating enzymes and drug discovery: emerging opportunities
.
Nat. Rev. Drug Discov.
17
,
57
78
53
Dewson
,
G.
,
Eichhorn
,
P.J.A.
and
Komander
,
D.
(
2023
)
Deubiquitinases in cancer
.
Nat. Rev. Cancer
23
,
842
862
54
Estavoyer
,
B.
,
Messmer
,
C.
,
Echbicheb
,
M.
,
Rudd
,
C.E.
,
Milot
,
E.
and
Affar
,
E.B.
(
2022
)
Mechanisms orchestrating the enzymatic activity and cellular functions of deubiquitinases
.
J. Biol. Chem.
298
,
102198
55
Schwertman
,
P.
,
Bekker-Jensen
,
S.
and
Mailand
,
N.
(
2016
)
Regulation of DNA double-strand break repair by ubiquitin and ubiquitin-like modifiers
.
Nat. Rev. Mol. Cell Biol.
17
,
379
394
56
Scully
,
R.
,
Panday
,
A.
,
Elango
,
R.
and
Willis
,
N.A.
(
2019
)
DNA double-strand break repair-pathway choice in somatic mammalian cells
.
Nat. Rev. Mol. Cell Biol.
20
,
698
714
57
Rogakou
,
E.P.
,
Pilch
,
D.R.
,
Orr
,
A.H.
,
Ivanova
,
V.S.
and
Bonner
,
W.M.
(
1998
)
DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139
.
J. Biol. Chem.
273
,
5858
5868
58
Savic
,
V.
,
Yin
,
B.
,
Maas
,
N.L.
,
Bredemeyer
,
A.L.
,
Carpenter
,
A.C.
,
Helmink
,
B.A.
, et al. (
2009
)
Formation of dynamic gamma-H2AX domains along broken DNA strands is distinctly regulated by ATM and MDC1 and dependent upon H2AX densities in chromatin
.
Mol. Cell
34
,
298
310
59
Stucki
,
M.
,
Clapperton
,
J.A.
,
Mohammad
,
D.
,
Yaffe
,
M.B.
,
Smerdon
,
S.J.
and
Jackson
,
S.P.
(
2005
)
MDC1 directly binds phosphorylated histone H2AX to regulate cellular responses to DNA double-strand breaks
.
Cell
123
,
1213
1226
60
Kolas
,
N.K.
,
Chapman
,
J.R.
,
Nakada
,
S.
,
Ylanko
,
J.
,
Chahwan
,
R.
,
Sweeney
,
F.D.
, et al. (
2007
)
Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase
.
Science
318
,
1637
1640
61
Huen
,
M.S.
,
Grant
,
R.
,
Manke
,
I.
,
Minn
,
K.
,
Yu
,
X.
,
Yaffe
,
M.B.
, et al. (
2007
)
RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly
.
Cell
131
,
901
914
62
Mailand
,
N.
,
Bekker-Jensen
,
S.
,
Faustrup
,
H.
,
Melander
,
F.
,
Bartek
,
J.
,
Lukas
,
C.
, et al. (
2007
)
RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins
.
Cell
131
,
887
900
63
Thorslund
,
T.
,
Ripplinger
,
A.
,
Hoffmann
,
S.
,
Wild
,
T.
,
Uckelmann
,
M.
,
Villumsen
,
B.
, et al. (
2015
)
Histone H1 couples initiation and amplification of ubiquitin signalling after DNA damage
.
Nature
527
,
389
393
64
Mandemaker
,
I.K.
,
van Cuijk
,
L.
,
Janssens
,
R.C.
,
Lans
,
H.
,
Bezstarosti
,
K.
,
Hoeijmakers
,
J.H.
, et al. (
2017
)
DNA damage-induced histone H1 ubiquitylation is mediated by HUWE1 and stimulates the RNF8-RNF168 pathway
.
Sci. Rep.
7
,
15353
65
Mattiroli
,
F.
,
Vissers
,
J.H.
,
van Dijk
,
W.J.
,
Ikpa
,
P.
,
Citterio
,
E.
,
Vermeulen
,
W.
, et al. (
2012
)
RNF168 ubiquitinates K13-15 on H2A/H2AX to drive DNA damage signaling
.
Cell
150
,
1182
1195
66
Nakamura
,
K.
,
Saredi
,
G.
,
Becker
,
J.R.
,
Foster
,
B.M.
,
Nguyen
,
N.V.
,
Beyer
,
T.E.
, et al. (
2019
)
H4K20me0 recognition by BRCA1-BARD1 directs homologous recombination to sister chromatids
.
Nat. Cell Biol.
21
,
311
318
67
Becker
,
J.R.
,
Clifford
,
G.
,
Bonnet
,
C.
,
Groth
,
A.
,
Wilson
,
M.D.
and
Chapman
,
J.R.
(
2021
)
BARD1 reads H2A lysine 15 ubiquitination to direct homologous recombination
.
Nature
596
,
433
437
68
Panier
,
S.
,
Ichijima
,
Y.
,
Fradet-Turcotte
,
A.
,
Leung
,
C.C.
,
Kaustov
,
L.
,
Arrowsmith
,
C.H.
, et al. (
2012
)
Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks
.
Mol. Cell
47
,
383
395
69
Fradet-Turcotte
,
A.
,
Canny
,
M.D.
,
Escribano-Diaz
,
C.
,
Orthwein
,
A.
,
Leung
,
C.C.
,
Huang
,
H.
, et al. (
2013
)
53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark
.
Nature
499
,
50
54
70
Horn
,
V.
,
Uckelmann
,
M.
,
Zhang
,
H.
,
Eerland
,
J.
,
Aarsman
,
I.
,
le Paige
,
U.B.
, et al. (
2019
)
Structural basis of specific H2A K13/K15 ubiquitination by RNF168
.
Nat. Commun.
10
,
1751
71
Dev
,
H.
,
Chiang
,
T.W.
,
Lescale
,
C.
,
de Krijger
,
I.
,
Martin
,
A.G.
,
Pilger
,
D.
, et al. (
2018
)
Shieldin complex promotes DNA end-joining and counters homologous recombination in BRCA1-null cells
.
Nat. Cell Biol.
20
,
954
965
72
Noordermeer
,
S.M.
,
Adam
,
S.
,
Setiaputra
,
D.
,
Barazas
,
M.
,
Pettitt
,
S.J.
,
Ling
,
A.K.
, et al. (
2018
)
The shieldin complex mediates 53BP1-dependent DNA repair
.
Nature
560
,
117
121
73
Setiaputra
,
D.
and
Durocher
,
D.
(
2019
)
Shieldin - the protector of DNA ends
.
EMBO Rep.
20
,
e47560
74
Chen
,
J.
,
Feng
,
W.
,
Jiang
,
J.
,
Deng
,
Y.
and
Huen
,
M.S.
(
2012
)
Ring finger protein RNF169 antagonizes the ubiquitin-dependent signaling cascade at sites of DNA damage
.
J. Biol. Chem.
287
,
27715
27722
75
Poulsen
,
M.
,
Lukas
,
C.
,
Lukas
,
J.
,
Bekker-Jensen
,
S.
and
Mailand
,
N.
(
2012
)
Human RNF169 is a negative regulator of the ubiquitin-dependent response to DNA double-strand breaks
.
J. Cell Biol.
197
,
189
199
76
Kitevski-LeBlanc
,
J.
,
Fradet-Turcotte
,
A.
,
Kukic
,
P.
,
Wilson
,
M.D.
,
Portella
,
G.
,
Yuwen
,
T.
, et al. (
2017
)
The RNF168 paralog RNF169 defines a new class of ubiquitylated histone reader involved in the response to DNA damage
.
eLife
6
,
e23872
77
Kalb
,
R.
,
Mallery
,
D.L.
,
Larkin
,
C.
,
Huang
,
J.T.
and
Hiom
,
K.
(
2014
)
BRCA1 is a histone-H2A-specific ubiquitin ligase
.
Cell Rep.
8
,
999
1005
78
Densham
,
R.M.
,
Garvin
,
A.J.
,
Stone
,
H.R.
,
Strachan
,
J.
,
Baldock
,
R.A.
,
Daza-Martin
,
M.
, et al. (
2016
)
Human BRCA1-BARD1 ubiquitin ligase activity counteracts chromatin barriers to DNA resection
.
Nat. Struct. Mol. Biol.
23
,
647
655
79
Jacq
,
X.
,
Kemp
,
M.
,
Martin
,
N.M.
and
Jackson
,
S.P.
(
2013
)
Deubiquitylating enzymes and DNA damage response pathways
.
Cell Biochem. Biophys.
67
,
25
43
80
Nishi
,
R.
,
Wijnhoven
,
P.
,
le Sage
,
C.
,
Tjeertes
,
J.
,
Galanty
,
Y.
,
Forment
,
J.V.
, et al. (
2014
)
Systematic characterization of deubiquitylating enzymes for roles in maintaining genome integrity
.
Nat. Cell Biol.
16
,
1016
1026
,
1–8
81
Ng
,
H.M.
,
Wei
,
L.
,
Lan
,
L.
and
Huen
,
M.S.
(
2016
)
The Lys63-deubiquitylating enzyme BRCC36 limits DNA break processing and repair
.
J. Biol. Chem.
291
,
16197
16207
82
Dong
,
Y.
,
Hakimi
,
M.A.
,
Chen
,
X.
,
Kumaraswamy
,
E.
,
Cooch
,
N.S.
,
Godwin
,
A.K.
, et al. (
2003
)
Regulation of BRCC, a holoenzyme complex containing BRCA1 and BRCA2, by a signalosome-like subunit and its role in DNA repair
.
Mol. Cell
12
,
1087
1099
83
Wang
,
B.
and
Elledge
,
S.J.
(
2007
)
Ubc13/Rnf8 ubiquitin ligases control foci formation of the Rap80/Abraxas/Brca1/Brcc36 complex in response to DNA damage
.
Proc. Natl Acad. Sci. U.S.A.
104
,
20759
20763
84
Sobhian
,
B.
,
Shao
,
G.
,
Lilli
,
D.R.
,
Culhane
,
A.C.
,
Moreau
,
L.A.
,
Xia
,
B.
, et al. (
2007
)
RAP80 targets BRCA1 to specific ubiquitin structures at DNA damage sites
.
Science
316
,
1198
1202
85
Patterson-Fortin
,
J.
,
Shao
,
G.
,
Bretscher
,
H.
,
Messick
,
T.E.
and
Greenberg
,
R.A.
(
2010
)
Differential regulation of JAMM domain deubiquitinating enzyme activity within the RAP80 complex
.
J. Biol. Chem.
285
,
30971
30981
86
Rabl
,
J.
,
Bunker
,
R.D.
,
Schenk
,
A.D.
,
Cavadini
,
S.
,
Gill
,
M.E.
,
Abdulrahman
,
W.
, et al. (
2019
)
Structural basis of BRCC36 function in DNA repair and immune regulation
.
Mol. Cell
75
,
483
497.e9
87
Shao
,
G.
,
Lilli
,
D.R.
,
Patterson-Fortin
,
J.
,
Coleman
,
K.A.
,
Morrissey
,
D.E.
and
Greenberg
,
R.A.
(
2009
)
The Rap80-BRCC36 de-ubiquitinating enzyme complex antagonizes RNF8-Ubc13-dependent ubiquitination events at DNA double strand breaks
.
Proc. Natl Acad. Sci. U.S.A.
106
,
3166
3171
88
Coleman
,
K.A.
and
Greenberg
,
R.A.
(
2011
)
The BRCA1-RAP80 complex regulates DNA repair mechanism utilization by restricting end resection
.
J. Biol. Chem.
286
,
13669
13680
89
Butler
,
L.R.
,
Densham
,
R.M.
,
Jia
,
J.
,
Garvin
,
A.J.
,
Stone
,
H.R.
,
Shah
,
V.
, et al. (
2012
)
The proteasomal de-ubiquitinating enzyme POH1 promotes the double-strand DNA break response
.
EMBO J.
31
,
3918
3934
90
Kakarougkas
,
A.
,
Ismail
,
A.
,
Katsuki
,
Y.
,
Freire
,
R.
,
Shibata
,
A.
and
Jeggo
,
P.A.
(
2013
)
Co-operation of BRCA1 and POH1 relieves the barriers posed by 53BP1 and RAP80 to resection
.
Nucleic Acids Res.
41
,
10298
10311
91
Cooper
,
E.M.
,
Cutcliffe
,
C.
,
Kristiansen
,
T.Z.
,
Pandey
,
A.
,
Pickart
,
C.M.
and
Cohen
,
R.E.
(
2009
)
K63-specific deubiquitination by two JAMM/MPN+ complexes: BRISC-associated Brcc36 and proteasomal Poh1
.
EMBO J.
28
,
621
631
92
Nakajima
,
S.
,
Lan
,
L.
,
Wei
,
L.
,
Hsieh
,
C.L.
,
Rapic-Otrin
,
V.
,
Yasui
,
A.
, et al. (
2014
)
Ubiquitin-specific protease 5 is required for the efficient repair of DNA double-strand breaks
.
PLoS One
9
,
e84899
93
Kato
,
K.
,
Nakajima
,
K.
,
Ui
,
A.
,
Muto-Terao
,
Y.
,
Ogiwara
,
H.
and
Nakada
,
S.
(
2014
)
Fine-tuning of DNA damage-dependent ubiquitination by OTUB2 supports the DNA repair pathway choice
.
Mol. Cell
53
,
617
630
94
Zhang
,
Z.
,
Yang
,
H.
and
Wang
,
H.
(
2014
)
The histone H2A deubiquitinase USP16 interacts with HERC2 and fine-tunes cellular response to DNA damage
.
J. Biol. Chem.
289
,
32883
32894
95
Typas
,
D.
,
Luijsterburg
,
M.S.
,
Wiegant
,
W.W.
,
Diakatou
,
M.
,
Helfricht
,
A.
,
Thijssen
,
P.E.
, et al. (
2016
)
The de-ubiquitylating enzymes USP26 and USP37 regulate homologous recombination by counteracting RAP80
.
Nucleic Acids Res.
44
,
2976
96
Mosbech
,
A.
,
Lukas
,
C.
,
Bekker-Jensen
,
S.
and
Mailand
,
N.
(
2013
)
The deubiquitylating enzyme USP44 counteracts the DNA double-strand break response mediated by the RNF8 and RNF168 ubiquitin ligases
.
J. Biol. Chem.
288
,
16579
16587
97
Li
,
Y.
,
Luo
,
K.
,
Yin
,
Y.
,
Wu
,
C.
,
Deng
,
M.
,
Li
,
L.
, et al. (
2017
)
USP13 regulates the RAP80-BRCA1 complex dependent DNA damage response
.
Nat. Commun.
8
,
15752
98
Nicassio
,
F.
,
Corrado
,
N.
,
Vissers
,
J.H.
,
Areces
,
L.B.
,
Bergink
,
S.
,
Marteijn
,
J.A.
, et al. (
2007
)
Human USP3 is a chromatin modifier required for S phase progression and genome stability
.
Curr. Biol.
17
,
1972
1977
99
Sharma
,
N.
,
Zhu
,
Q.
,
Wani
,
G.
,
He
,
J.
,
Wang
,
Q.E.
and
Wani
,
A.A.
(
2014
)
USP3 counteracts RNF168 via deubiquitinating H2A and gammaH2AX at lysine 13 and 15
.
Cell Cycle
13
,
106
114
100
Wang
,
Z.
,
Zhang
,
H.
,
Liu
,
J.
,
Cheruiyot
,
A.
,
Lee
,
J.H.
,
Ordog
,
T.
, et al. (
2016
)
USP51 deubiquitylates H2AK13,15ub and regulates DNA damage response
.
Genes Dev.
30
,
946
959
101
Ai
,
H.
,
Guo
,
Y.
,
Sun
,
D.
,
Liu
,
S.
,
Qi
,
Y.
,
Guo
,
J.
, et al. (
2019
)
Examination of the deubiquitylation site selectivity of USP51 by using chemically synthesized ubiquitylated histones
.
ChemBioChem
20
,
221
229
102
Nakada
,
S.
,
Tai
,
I.
,
Panier
,
S.
,
Al-Hakim
,
A.
,
Iemura
,
S.
,
Juang
,
Y.C.
, et al. (
2010
)
Non-canonical inhibition of DNA damage-dependent ubiquitination by OTUB1
.
Nature
466
,
941
946
103
Blackford
,
A.N.
and
Stewart
,
G.S.
(
2011
)
When cleavage is not attractive: non-catalytic inhibition of ubiquitin chains at DNA double-strand breaks by OTUB1
.
DNA Repair (Amst)
10
,
245
249
104
Sato
,
Y.
,
Yamagata
,
A.
,
Goto-Ito
,
S.
,
Kubota
,
K.
,
Miyamoto
,
R.
,
Nakada
,
S.
, et al. (
2012
)
Molecular basis of Lys-63-linked polyubiquitination inhibition by the interaction between human deubiquitinating enzyme OTUB1 and ubiquitin-conjugating enzyme UBC13
.
J. Biol. Chem.
287
,
25860
25868
105
Wiener
,
R.
,
Zhang
,
X.
,
Wang
,
T.
and
Wolberger
,
C.
(
2012
)
The mechanism of OTUB1-mediated inhibition of ubiquitination
.
Nature
483
,
618
622
106
Yu
,
M.
,
Liu
,
K.
,
Mao
,
Z.
,
Luo
,
J.
,
Gu
,
W.
and
Zhao
,
W.
(
2016
)
USP11 is a negative regulator to gammaH2AX ubiquitylation by RNF8/RNF168
.
J. Biol. Chem.
291
,
959
967
107
Ting
,
X.
,
Xia
,
L.
,
Yang
,
J.
,
He
,
L.
,
Si
,
W.
,
Shang
,
Y.
, et al. (
2019
)
USP11 acts as a histone deubiquitinase functioning in chromatin reorganization during DNA repair
.
Nucleic Acids Res.
47
,
9721
9740
108
Orthwein
,
A.
,
Noordermeer
,
S.M.
,
Wilson
,
M.D.
,
Landry
,
S.
,
Enchev
,
R.I.
,
Sherker
,
A.
, et al. (
2015
)
A mechanism for the suppression of homologous recombination in G1 cells
.
Nature
528
,
422
426
109
Schoenfeld
,
A.R.
,
Apgar
,
S.
,
Dolios
,
G.
,
Wang
,
R.
and
Aaronson
,
S.A.
(
2004
)
BRCA2 is ubiquitinated in vivo and interacts with USP11, a deubiquitinating enzyme that exhibits prosurvival function in the cellular response to DNA damage
.
Mol. Cell Biol.
24
,
7444
7455
110
Hendriks
,
I.A.
,
Schimmel
,
J.
,
Eifler
,
K.
,
Olsen
,
J.V.
and
Vertegaal
,
A.C.O.
(
2015
)
Ubiquitin-specific protease 11 (USP11) deubiquitinates hybrid small ubiquitin-like modifier (SUMO)-ubiquitin chains to counteract RING finger protein 4 (RNF4)
.
J. Biol. Chem.
290
,
15526
15537
111
Wiltshire
,
T.D.
,
Lovejoy
,
C.A.
,
Wang
,
T.
,
Xia
,
F.
,
O'Connor
,
M.J.
and
Cortez
,
D.
(
2010
)
Sensitivity to poly(ADP-ribose) polymerase (PARP) inhibition identifies ubiquitin-specific peptidase 11 (USP11) as a regulator of DNA double-strand break repair
.
J. Biol. Chem.
285
,
14565
14571
112
Liu
,
J.
,
Kruswick
,
A.
,
Dang
,
H.
,
Tran
,
A.D.
,
Kwon
,
S.M.
,
Wang
,
X.W.
, et al. (
2017
)
Ubiquitin-specific protease 21 stabilizes BRCA2 to control DNA repair and tumor growth
.
Nat. Commun.
8
,
137
113
Luo
,
K.
,
Li
,
L.
,
Li
,
Y.
,
Wu
,
C.
,
Yin
,
Y.
,
Chen
,
Y.
, et al. (
2016
)
A phosphorylation-deubiquitination cascade regulates the BRCA2-RAD51 axis in homologous recombination
.
Genes Dev.
30
,
2581
2595
114
Ismail
,
I.H.
,
Davidson
,
R.
,
Gagne
,
J.P.
,
Xu
,
Z.Z.
,
Poirier
,
G.G.
and
Hendzel
,
M.J.
(
2014
)
Germline mutations in BAP1 impair its function in DNA double-strand break repair
.
Cancer Res.
74
,
4282
4294
115
Sahtoe
,
D.D.
,
van Dijk
,
W.J.
,
Ekkebus
,
R.
,
Ovaa
,
H.
and
Sixma
,
T.K.
(
2016
)
BAP1/ASXL1 recruitment and activation for H2A deubiquitination
.
Nat. Commun.
7
,
10292
116
Lee
,
H.S.
,
Lee
,
S.A.
,
Hur
,
S.K.
,
Seo
,
J.W.
and
Kwon
,
J.
(
2014
)
Stabilization and targeting of INO80 to replication forks by BAP1 during normal DNA synthesis
.
Nat. Commun.
5
,
5128
117
Lee
,
S.A.
,
Lee
,
D.
,
Kang
,
M.
,
Kim
,
S.
,
Kwon
,
S.J.
,
Lee
,
H.S.
, et al. (
2022
)
BAP1 promotes the repair of UV-induced DNA damage via PARP1-mediated recruitment to damage sites and control of activity and stability
.
Cell Death Differ.
29
,
2381
2398
118
Kwon
,
J.
,
Lee
,
D.
and
Lee
,
S.A.
(
2023
)
BAP1 as a guardian of genome stability: implications in human cancer
.
Exp. Mol. Med.
55
,
745
754
119
Foglizzo
,
M.
,
Middleton
,
A.J.
,
Burgess
,
A.E.
,
Crowther
,
J.M.
,
Dobson
,
R.C.J.
,
Murphy
,
J.M.
, et al. (
2018
)
A bidentate Polycomb Repressive-Deubiquitinase complex is required for efficient activity on nucleosomes
.
Nat. Commun.
9
,
3932
120
De
,
I.
,
Chittock
,
E.C.
,
Grotsch
,
H.
,
Miller
,
T.C.R.
,
McCarthy
,
A.A.
and
Muller
,
C.W.
(
2019
)
Structural basis for the activation of the deubiquitinase calypso by the Polycomb protein ASX
.
Structure
27
,
528
536.e4
121
Thomas
JF
,
Valencia-Sanchez
MI
,
Tamburri
S
,
Gloor
SL
,
Rustichelli
S
,
Godinez-Lopez
V
, et al.
Structural basis of histone H2A lysine 119 deubiquitination by Polycomb Repressive Deubiquitinase BAP1/ASXL1. bioRxiv
122
Uckelmann
,
M.
,
Densham
,
R.M.
,
Baas
,
R.
,
Winterwerp
,
H.H.K.
,
Fish
,
A.
,
Sixma
,
T.K.
, et al. (
2018
)
USP48 restrains resection by site-specific cleavage of the BRCA1 ubiquitin mark from H2A
.
Nat Commun.
9
,
229
123
Sy
,
S.M.
,
Jiang
,
J.
,
Sun
,
O.W.
,
Deng
,
Y.
and
Huen
,
M.S.
(
2013
)
The ubiquitin specific protease USP34 promotes ubiquitin signaling at DNA double-strand breaks
.
Nucleic Acids Res.
41
,
8572
8580
124
Zhu
,
Q.
,
Sharma
,
N.
,
He
,
J.
,
Wani
,
G.
and
Wani
,
A.A.
(
2015
)
USP7 deubiquitinase promotes ubiquitin-dependent DNA damage signaling by stabilizing RNF168
.
Cell Cycle
14
,
1413
1425
125
Gudjonsson
,
T.
,
Altmeyer
,
M.
,
Savic
,
V.
,
Toledo
,
L.
,
Dinant
,
C.
,
Grofte
,
M.
, et al. (
2012
)
TRIP12 and UBR5 suppress spreading of chromatin ubiquitylation at damaged chromosomes
.
Cell
150
,
697
709
126
Valles
,
G.J.
,
Bezsonova
,
I.
,
Woodgate
,
R.
and
Ashton
,
N.W.
(
2020
)
USP7 is a master regulator of genome stability
.
Front. Cell Dev. Biol.
8
,
717
127
Dar
,
A.
,
Shibata
,
E.
and
Dutta
,
A.
(
2013
)
Deubiquitination of Tip60 by USP7 determines the activity of the p53-dependent apoptotic pathway
.
Mol. Cell Biol.
33
,
3309
3320
128
Su
,
D.
,
Ma
,
S.
,
Shan
,
L.
,
Wang
,
Y.
,
Wang
,
Y.
,
Cao
,
C.
, et al. (
2018
)
Ubiquitin-specific protease 7 sustains DNA damage response and promotes cervical carcinogenesis
.
J. Clin. Invest.
128
,
4280
4296
129
Schwertman
,
P.
,
Lagarou
,
A.
,
Dekkers
,
D.H.
,
Raams
,
A.
,
van der Hoek
,
A.C.
,
Laffeber
,
C.
, et al. (
2012
)
UV-sensitive syndrome protein UVSSA recruits USP7 to regulate transcription-coupled repair
.
Nat. Genet.
44
,
598
602
130
Schwertman
,
P.
,
Vermeulen
,
W.
and
Marteijn
,
J.A.
(
2013
)
UVSSA and USP7, a new couple in transcription-coupled DNA repair
.
Chromosoma
122
,
275
284
131
He
,
J.
,
Zhu
,
Q.
,
Wani
,
G.
,
Sharma
,
N.
,
Han
,
C.
,
Qian
,
J.
, et al. (
2014
)
Ubiquitin-specific protease 7 regulates nucleotide excision repair through deubiquitinating XPC protein and preventing XPC protein from undergoing ultraviolet light-induced and VCP/p97 protein-regulated proteolysis
.
J. Biol. Chem.
289
,
27278
27289
132
Lecona
,
E.
,
Rodriguez-Acebes
,
S.
,
Specks
,
J.
,
Lopez-Contreras
,
A.J.
,
Ruppen
,
I.
,
Murga
,
M.
, et al. (
2016
)
USP7 is a SUMO deubiquitinase essential for DNA replication
.
Nat. Struct. Mol. Biol.
23
,
270
277
133
Jiang
,
H.K.
,
Kurkute
,
P.
,
Li
,
C.L.
,
Wang
,
Y.H.
,
Chen
,
P.J.
,
Lin
,
S.Y.
, et al. (
2020
)
Revealing USP7 deubiquitinase substrate specificity by unbiased synthesis of ubiquitin tagged SUMO2
.
Biochemistry
59
,
3796
3801
134
Zhao
,
S.
,
Kieser
,
A.
,
Li
,
H.Y.
,
Reinking
,
H.K.
,
Weickert
,
P.
,
Euteneuer
,
S.
, et al. (
2021
)
A ubiquitin switch controls autocatalytic inactivation of the DNA-protein crosslink repair protease SPRTN
.
Nucleic Acids Res.
49
,
902
915
135
Sheng
,
Y.
,
Saridakis
,
V.
,
Sarkari
,
F.
,
Duan
,
S.
,
Wu
,
T.
,
Arrowsmith
,
C.H.
, et al. (
2006
)
Molecular recognition of p53 and MDM2 by USP7/HAUSP
.
Nat. Struct. Mol. Biol.
13
,
285
291
136
Wang
,
Z.
,
Kang
,
W.
,
You
,
Y.
,
Pang
,
J.
,
Ren
,
H.
,
Suo
,
Z.
, et al. (
2019
)
USP7: novel drug target in cancer therapy
.
Front. Pharmacol.
10
,
427
137
Qi
,
S.M.
,
Cheng
,
G.
,
Cheng
,
X.D.
,
Xu
,
Z.
,
Xu
,
B.
,
Zhang
,
W.D.
, et al. (
2020
)
Targeting USP7-mediated deubiquitination of MDM2/MDMX-p53 pathway for cancer therapy: are we there yet?
Front. Cell Dev. Biol.
8
,
233
138
Schauer
,
N.J.
,
Liu
,
X.
,
Magin
,
R.S.
,
Doherty
,
L.M.
,
Chan
,
W.C.
,
Ficarro
,
S.B.
, et al. (
2020
)
Selective USP7 inhibition elicits cancer cell killing through a p53-dependent mechanism
.
Sci. Rep.
10
,
5324
139
Lu
,
J.
,
Zhao
,
H.
,
Yu
,
C.
,
Kang
,
Y.
and
Yang
,
X.
(
2021
)
Targeting ubiquitin-specific protease 7 (USP7) in cancer: a new insight to overcome drug resistance
.
Front. Pharmacol.
12
,
648491
140
Harakandi
,
C.
,
Nininahazwe
,
L.
,
Xu
,
H.
,
Liu
,
B.
,
He
,
C.
,
Zheng
,
Y.C.
, et al. (
2021
)
Recent advances on the intervention sites targeting USP7-MDM2-p53 in cancer therapy
.
Bioorg. Chem.
116
,
105273
141
Nininahazwe
,
L.
,
Liu
,
B.
,
He
,
C.
,
Zhang
,
H.
and
Chen
,
Z.S.
(
2021
)
The emerging nature of Ubiquitin-specific protease 7 (USP7): a new target in cancer therapy
.
Drug Discov. Today
26
,
490
502
142
Oliveira
,
R.I.
,
Guedes
,
R.A.
and
Salvador
,
J.A.R.
(
2022
)
Highlights in USP7 inhibitors for cancer treatment
.
Front. Chem.
10
,
1005727
143
Lamberto
,
I.
,
Liu
,
X.
,
Seo
,
H.S.
,
Schauer
,
N.J.
,
Iacob
,
R.E.
,
Hu
,
W.
, et al. (
2017
)
Structure-guided development of a potent and selective non-covalent active-site inhibitor of USP7
.
Cell Chem. Biol.
24
,
1490
1500.e11
144
Pozhidaeva
,
A.
,
Valles
,
G.
,
Wang
,
F.
,
Wu
,
J.
,
Sterner
,
D.E.
,
Nguyen
,
P.
, et al. (
2017
)
USP7-specific inhibitors target and modify the enzyme's active site via distinct chemical mechanisms
.
Cell Chem. Biol.
24
,
1501
1512.e5
145
Di Lello
,
P.
,
Pastor
,
R.
,
Murray
,
J.M.
,
Blake
,
R.A.
,
Cohen
,
F.
,
Crawford
,
T.D.
, et al. (
2017
)
Discovery of small-molecule inhibitors of ubiquitin specific protease 7 (USP7) using integrated NMR and in silico techniques
.
J. Med. Chem.
60
,
10056
10070
146
Kategaya
,
L.
,
Di Lello
,
P.
,
Rouge
,
L.
,
Pastor
,
R.
,
Clark
,
K.R.
,
Drummond
,
J.
, et al. (
2017
)
USP7 small-molecule inhibitors interfere with ubiquitin binding
.
Nature
550
,
534
538
147
Turnbull
,
A.P.
,
Ioannidis
,
S.
,
Krajewski
,
W.W.
,
Pinto-Fernandez
,
A.
,
Heride
,
C.
,
Martin
,
A.C.L.
, et al. (
2017
)
Molecular basis of USP7 inhibition by selective small-molecule inhibitors
.
Nature
550
,
481
486
148
Nishi
,
R.
,
Wijnhoven
,
P.W.G.
,
Kimura
,
Y.
,
Matsui
,
M.
,
Konietzny
,
R.
,
Wu
,
Q.
, et al. (
2018
)
The deubiquitylating enzyme UCHL3 regulates Ku80 retention at sites of DNA damage
.
Sci. Rep.
8
,
17891
149
Li
,
F.
,
Sun
,
Q.
,
Liu
,
K.
,
Han
,
H.
,
Lin
,
N.
,
Cheng
,
Z.
, et al. (
2019
)
The deubiquitinase OTUD5 regulates Ku80 stability and non-homologous end joining
.
Cell. Mol. Life Sci.
76
,
3861
3873
150
Liu
,
H.
,
Zhang
,
H.
,
Wang
,
X.
,
Tian
,
Q.
,
Hu
,
Z.
,
Peng
,
C.
, et al. (
2015
)
The deubiquitylating enzyme USP4 cooperates with CtIP in DNA double-strand break end resection
.
Cell Rep.
13
,
93
107
151
Wijnhoven
,
P.
,
Konietzny
,
R.
,
Blackford
,
A.N.
,
Travers
,
J.
,
Kessler
,
B.M.
,
Nishi
,
R.
, et al. (
2015
)
USP4 auto-deubiquitylation promotes homologous recombination
.
Mol. Cell
60
,
362
373
152
Zeman
,
M.K.
and
Cimprich
,
K.A.
(
2014
)
Causes and consequences of replication stress
.
Nat. Cell Biol.
16
,
2
9
153
Berti
,
M.
,
Cortez
,
D.
and
Lopes
,
M.
(
2020
)
The plasticity of DNA replication forks in response to clinically relevant genotoxic stress
.
Nat. Rev. Mol. Cell Biol.
21
,
633
651
154
Toledo
,
L.I.
,
Altmeyer
,
M.
,
Rask
,
M.B.
,
Lukas
,
C.
,
Larsen
,
D.H.
,
Povlsen
,
L.K.
, et al. (
2013
)
ATR prohibits replication catastrophe by preventing global exhaustion of RPA
.
Cell
155
,
1088
1103
155
Garcia-Rodriguez
,
N.
,
Wong
,
R.P.
and
Ulrich
,
H.D.
(
2016
)
Functions of ubiquitin and SUMO in DNA replication and replication stress
.
Front. Genet.
7
,
87
156
Mirsanaye
,
A.S.
,
Typas
,
D.
and
Mailand
,
N.
(
2021
)
Ubiquitylation at stressed replication forks: mechanisms and functions
.
Trends Cell Biol.
31
,
584
597
157
Yates
,
M.
and
Marechal
,
A.
(
2018
)
Ubiquitylation at the fork: making and breaking chains to complete DNA replication
.
Int. J. Mol. Sci.
19
,
2909
158
Hoege
,
C.
,
Pfander
,
B.
,
Moldovan
,
G.L.
,
Pyrowolakis
,
G.
and
Jentsch
,
S.
(
2002
)
RAD6-dependent DNA repair is linked to modification of PCNA by ubiquitin and SUMO
.
Nature
419
,
135
141
159
Hibbert
,
R.G.
,
Huang
,
A.
,
Boelens
,
R.
and
Sixma
,
T.K.
(
2011
)
E3 ligase Rad18 promotes monoubiquitination rather than ubiquitin chain formation by E2 enzyme Rad6
.
Proc. Natl Acad. Sci. U.S.A.
108
,
5590
5595
160
Li
,
M.
,
Sengupta
,
B.
,
Benkovic
,
S.J.
,
Lee
,
T.H.
and
Hedglin
,
M.
(
2020
)
PCNA monoubiquitination is regulated by diffusion of Rad6/Rad18 complexes along RPA filaments
.
Biochemistry
59
,
4694
4702
161
Notenboom
,
V.
,
Hibbert
,
R.G.
,
van Rossum-Fikkert
,
S.E.
,
Olsen
,
J.V.
,
Mann
,
M.
and
Sixma
,
T.K.
(
2007
)
Functional characterization of Rad18 domains for Rad6, ubiquitin, DNA binding and PCNA modification
.
Nucleic Acids Res.
35
,
5819
5830
162
Mailand
,
N.
,
Gibbs-Seymour
,
I.
and
Bekker-Jensen
,
S.
(
2013
)
Regulation of PCNA-protein interactions for genome stability
.
Nat. Rev. Mol. Cell Biol.
14
,
269
282
163
Lau
,
W.C.
,
Li
,
Y.
,
Zhang
,
Q.
and
Huen
,
M.S.
(
2015
)
Molecular architecture of the Ub-PCNA/Pol eta complex bound to DNA
.
Sci. Rep.
5
,
15759
164
Tsutakawa
,
S.E.
,
Yan
,
C.
,
Xu
,
X.
,
Weinacht
,
C.P.
,
Freudenthal
,
B.D.
,
Yang
,
K.
, et al. (
2015
)
Structurally distinct ubiquitin- and sumo-modified PCNA: implications for their distinct roles in the DNA damage response
.
Structure
23
,
724
733
165
Huang
,
T.T.
,
Nijman
,
S.M.
,
Mirchandani
,
K.D.
,
Galardy
,
P.J.
,
Cohn
,
M.A.
,
Haas
,
W.
, et al. (
2006
)
Regulation of monoubiquitinated PCNA by DUB autocleavage
.
Nat. Cell Biol.
8
,
339
347
166
Cohn
,
M.A.
,
Kee
,
Y.
,
Haas
,
W.
,
Gygi
,
S.P.
and
D'Andrea
,
A.D.
(
2009
)
UAF1 is a subunit of multiple deubiquitinating enzyme complexes
.
J. Biol. Chem.
284
,
5343
5351
167
Lim
,
K.S.
,
Li
,
H.
,
Roberts
,
E.A.
,
Gaudiano
,
E.F.
,
Clairmont
,
C.
,
Sambel
,
L.A.
, et al. (
2018
)
USP1 is required for replication fork protection in BRCA1-deficient tumors
.
Mol. Cell
72
,
925
941.e4
168
Lee
,
K.Y.
,
Yang
,
K.
,
Cohn
,
M.A.
,
Sikdar
,
N.
,
D'Andrea
,
A.D.
and
Myung
,
K.
(
2010
)
Human ELG1 regulates the level of ubiquitinated proliferating cell nuclear antigen (PCNA) through Its interactions with PCNA and USP1
.
J. Biol. Chem.
285
,
10362
10369
169
Kang
,
M.S.
,
Ryu
,
E.
,
Lee
,
S.W.
,
Park
,
J.
,
Ha
,
N.Y.
,
Ra
,
J.S.
, et al. (
2019
)
Regulation of PCNA cycling on replicating DNA by RFC and RFC-like complexes
.
Nat. Commun.
10
,
2420
170
Coleman
,
K.E.
,
Yin
,
Y.
,
Lui
,
S.K.L.
,
Keegan
,
S.
,
Fenyo
,
D.
,
Smith
,
D.J.
, et al. (
2022
)
USP1-trapping lesions as a source of DNA replication stress and genomic instability
.
Nat. Commun.
13
,
1740
171
Simoneau
,
A.
,
Engel
,
J.L.
,
Bandi
,
M.
,
Lazarides
,
K.
,
Liu
,
S.
,
Meier
,
S.R.
, et al. (
2023
)
Ubiquitinated PCNA drives USP1 synthetic lethality in cancer
.
Mol. Cancer Ther.
22
,
215
226
172
Huang
,
P.
,
Wang
,
Y.
,
Zhang
,
P.
and
Li
,
Q.
(
2023
)
Ubiquitin-specific peptidase 1: assessing its role in cancer therapy
.
Clin. Exp. Med.
23
,
2953
2966
173
Rennie
,
M.L.
,
Arkinson
,
C.
,
Chaugule
,
V.K.
and
Walden
,
H.
(
2022
)
Cryo-EM reveals a mechanism of USP1 inhibition through a cryptic binding site
.
Sci. Adv.
8
,
eabq6353
174
Cohn
,
M.A.
,
Kowal
,
P.
,
Yang
,
K.
,
Haas
,
W.
,
Huang
,
T.T.
,
Gygi
,
S.P.
, et al. (
2007
)
A UAF1-containing multisubunit protein complex regulates the Fanconi anemia pathway
.
Mol. Cell
28
,
786
797
175
Oestergaard
,
V.H.
,
Langevin
,
F.
,
Kuiken
,
H.J.
,
Pace
,
P.
,
Niedzwiedz
,
W.
,
Simpson
,
L.J.
, et al. (
2007
)
Deubiquitination of FANCD2 is required for DNA crosslink repair
.
Mol. Cell
28
,
798
809
176
Joo
,
W.
,
Xu
,
G.
,
Persky
,
N.S.
,
Smogorzewska
,
A.
,
Rudge
,
D.G.
,
Buzovetsky
,
O.
, et al. (
2011
)
Structure of the FANCI-FANCD2 complex: insights into the Fanconi anemia DNA repair pathway
.
Science
333
,
312
316
177
Lopez-Martinez
,
D.
,
Liang
,
C.C.
and
Cohn
,
M.A.
(
2016
)
Cellular response to DNA interstrand crosslinks: the Fanconi anemia pathway
.
Cell. Mol. Life Sci.
73
,
3097
3114
178
Nalepa
,
G.
and
Clapp
,
D.W.
(
2018
)
Fanconi anaemia and cancer: an intricate relationship
.
Nat. Rev. Cancer
18
,
168
185
179
Tan
,
W.
,
van Twest
,
S.
,
Murphy
,
V.J.
and
Deans
,
A.J.
(
2020
)
ATR-mediated FANCI phosphorylation regulates both ubiquitination and deubiquitination of FANCD2
.
Front. Cell Dev. Biol.
8
,
2
180
Ishiai
,
M.
(
2021
)
Regulation of the Fanconi anemia DNA repair pathway by phosphorylation and monoubiquitination
.
Genes (Basel)
12
,
1763
181
Sijacki
,
T.
,
Alcon
,
P.
,
Chen
,
Z.A.
,
McLaughlin
,
S.H.
,
Shakeel
,
S.
,
Rappsilber
,
J.
, et al. (
2022
)
The DNA-damage kinase ATR activates the FANCD2-FANCI clamp by priming it for ubiquitination
.
Nat. Struct. Mol. Biol.
29
,
881
890
182
Kupculak
,
M.
,
Bai
,
F.
,
Luo
,
Q.
,
Yoshikawa
,
Y.
,
Lopez-Martinez
,
D.
,
Xu
,
H.
, et al. (
2023
)
Phosphorylation by ATR triggers FANCD2 chromatin loading and activates the Fanconi anemia pathway
.
Cell Rep.
42
,
112721
183
Shakeel
,
S.
,
Rajendra
,
E.
,
Alcon
,
P.
,
O'Reilly
,
F.
,
Chorev
,
D.S.
,
Maslen
,
S.
, et al. (
2019
)
Structure of the Fanconi anaemia monoubiquitin ligase complex
.
Nature
575
,
234
237
184
Li
,
L.
,
Tan
,
W.
and
Deans
,
A.J.
(
2020
)
Structural insight into FANCI-FANCD2 monoubiquitination
.
Essays Biochem.
64
,
807
817
185
Rennie
,
M.L.
,
Lemonidis
,
K.
,
Arkinson
,
C.
,
Chaugule
,
V.K.
,
Clarke
,
M.
,
Streetley
,
J.
, et al. (
2020
)
Differential functions of FANCI and FANCD2 ubiquitination stabilize ID2 complex on DNA
.
EMBO Rep.
21
,
e50133
186
Tan
,
W.
,
van Twest
,
S.
,
Leis
,
A.
,
Bythell-Douglas
,
R.
,
Murphy
,
V.J.
,
Sharp
,
M.
, et al. (
2020
)
Monoubiquitination by the human Fanconi anemia core complex clamps FANCI:FANCD2 on DNA in filamentous arrays
.
eLife
9
,
e54128
187
Chaugule
,
V.K.
,
Arkinson
,
C.
,
Rennie
,
M.L.
,
Kamarainen
,
O.
,
Toth
,
R.
and
Walden
,
H.
(
2020
)
Allosteric mechanism for site-specific ubiquitination of FANCD2
.
Nat. Chem. Biol.
16
,
291
301
188
Wang
,
R.
,
Wang
,
S.
,
Dhar
,
A.
,
Peralta
,
C.
and
Pavletich
,
N.P.
(
2020
)
DNA clamp function of the monoubiquitinated Fanconi anaemia ID complex
.
Nature
580
,
278
282
189
Alcon
,
P.
,
Shakeel
,
S.
,
Chen
,
Z.A.
,
Rappsilber
,
J.
,
Patel
,
K.J.
and
Passmore
,
L.A.
(
2020
)
FANCD2-FANCI is a clamp stabilized on DNA by monoubiquitination of FANCD2 during DNA repair
.
Nat. Struct. Mol. Biol.
27
,
240
248
190
Wang
,
S.
,
Wang
,
R.
,
Peralta
,
C.
,
Yaseen
,
A.
and
Pavletich
,
N.P.
(
2021
)
Structure of the FA core ubiquitin ligase closing the ID clamp on DNA
.
Nat. Struct. Mol. Biol.
28
,
300
309
191
Rennie
,
M.L.
,
Arkinson
,
C.
,
Chaugule
,
V.K.
,
Toth
,
R.
and
Walden
,
H.
(
2021
)
Structural basis of FANCD2 deubiquitination by USP1-UAF1
.
Nat. Struct. Mol. Biol.
28
,
356
364
192
Arkinson
,
C.
,
Chaugule
,
V.K.
,
Toth
,
R.
and
Walden
,
H.
(
2018
)
Specificity for deubiquitination of monoubiquitinated FANCD2 is driven by the N-terminus of USP1
.
Life Sci. Alliance
1
,
e201800162
193
Liang
,
F.
,
Miller
,
A.S.
,
Longerich
,
S.
,
Tang
,
C.
,
Maranon
,
D.
,
Williamson
,
E.A.
, et al. (
2019
)
DNA requirement in FANCD2 deubiquitination by USP1-UAF1-RAD51AP1 in the Fanconi anemia DNA damage response
.
Nat. Commun.
10
,
2849
194
Lemonidis
,
K.
,
Rennie
,
M.L.
,
Arkinson
,
C.
,
Chaugule
,
V.K.
,
Clarke
,
M.
,
Streetley
,
J.
, et al. (
2023
)
Structural and biochemical basis of interdependent FANCI-FANCD2 ubiquitination
.
EMBO J.
42
,
e111898
195
Hernandez-Perez
,
S.
,
Cabrera
,
E.
,
Amoedo
,
H.
,
Rodriguez-Acebes
,
S.
,
Koundrioukoff
,
S.
,
Debatisse
,
M.
, et al. (
2016
)
USP37 deubiquitinates Cdt1 and contributes to regulate DNA replication
.
Mol. Oncol.
10
,
1196
1206
196
Wu
,
C.
,
Chang
,
Y.
,
Chen
,
J.
,
Su
,
Y.
,
Li
,
L.
,
Chen
,
Y.
, et al. (
2021
)
USP37 regulates DNA damage response through stabilizing and deubiquitinating BLM
.
Nucleic Acids Res.
49
,
11224
11240
197
Chauhan
,
R.
,
Bhat
,
A.A.
,
Masoodi
,
T.
,
Bagga
,
P.
,
Reddy
,
R.
,
Gupta
,
A.
, et al. (
2021
)
Ubiquitin-specific peptidase 37: an important cog in the oncogenic machinery of cancerous cells
.
J. Exp. Clin. Cancer Res.
40
,
356
198
Huang
,
J.
,
Zhou
,
Q.
,
Gao
,
M.
,
Nowsheen
,
S.
,
Zhao
,
F.
,
Kim
,
W.
, et al. (
2020
)
Tandem deubiquitination and acetylation of SPRTN promotes DNA-protein crosslink repair and protects against aging
.
Mol. Cell
79
,
824
835.e5
199
Moore
,
C.E.
,
Yalcindag
,
S.E.
,
Czeladko
,
H.
,
Ravindranathan
,
R.
,
Wijesekara Hanthi
,
Y.
,
Levy
,
J.C.
, et al. (
2023
)
RFWD3 promotes ZRANB3 recruitment to regulate the remodeling of stalled replication forks
.
J. Cell Biol.
222
,
e202106022
200
Gallina
,
I.
,
Hendriks
,
I.A.
,
Hoffmann
,
S.
,
Larsen
,
N.B.
,
Johansen
,
J.
,
Colding-Christensen
,
C.S.
, et al. (
2021
)
The ubiquitin ligase RFWD3 is required for translesion DNA synthesis
.
Mol. Cell
81
,
442
458.e9
201
Dubois
,
J.C.
,
Yates
,
M.
,
Gaudreau-Lapierre
,
A.
,
Clement
,
G.
,
Cappadocia
,
L.
,
Gaudreau
,
L.
, et al. (
2017
)
A phosphorylation-and-ubiquitylation circuitry driving ATR activation and homologous recombination
.
Nucleic Acids Res.
45
,
8859
8872
202
Liu
,
S.
,
Chu
,
J.
,
Yucer
,
N.
,
Leng
,
M.
,
Wang
,
S.Y.
,
Chen
,
B.P.
, et al. (
2011
)
RING finger and WD repeat domain 3 (RFWD3) associates with replication protein A (RPA) and facilitates RPA-mediated DNA damage response
.
J. Biol. Chem.
286
,
22314
22322
203
Elia
,
A.E.
,
Wang
,
D.C.
,
Willis
,
N.A.
,
Boardman
,
A.P.
,
Hajdu
,
I.
,
Adeyemi
,
R.O.
, et al. (
2015
)
RFWD3-dependent ubiquitination of RPA regulates repair at stalled replication forks
.
Mol. Cell
60
,
280
293
204
Feeney
,
L.
,
Munoz
,
I.M.
,
Lachaud
,
C.
,
Toth
,
R.
,
Appleton
,
P.L.
,
Schindler
,
D.
, et al. (
2017
)
RPA-mediated recruitment of the E3 ligase RFWD3 is vital for interstrand crosslink repair and human health
.
Mol. Cell
66
,
610
621.e4
205
Inano
,
S.
,
Sato
,
K.
,
Katsuki
,
Y.
,
Kobayashi
,
W.
,
Tanaka
,
H.
,
Nakajima
,
K.
, et al. (
2017
)
RFWD3-mediated ubiquitination promotes timely removal of both RPA and RAD51 from DNA damage sites to facilitate homologous recombination
.
Mol. Cell
66
,
622
634.e8
206
Wu
,
R.A.
,
Semlow
,
D.R.
,
Kamimae-Lanning
,
A.N.
,
Kochenova
,
O.V.
,
Chistol
,
G.
,
Hodskinson
,
M.R.
, et al. (
2019
)
TRAIP is a master regulator of DNA interstrand crosslink repair
.
Nature
567
,
267
272
207
Sparks
,
J.L.
,
Chistol
,
G.
,
Gao
,
A.O.
,
Raschle
,
M.
,
Larsen
,
N.B.
,
Mann
,
M.
, et al. (
2019
)
The CMG helicase bypasses DNA-protein cross-links to facilitate their repair
.
Cell
176
,
167
181.e21
208
Baretic
,
D.
,
Jenkyn-Bedford
,
M.
,
Aria
,
V.
,
Cannone
,
G.
,
Skehel
,
M.
and
Yeeles
,
J.T.P.
(
2020
)
Cryo-EM structure of the fork protection complex bound to CMG at a replication fork
.
Mol. Cell
78
,
926
940.e13
209
Villa
,
F.
,
Fujisawa
,
R.
,
Ainsworth
,
J.
,
Nishimura
,
K.
,
Lie
,
A.L.M.
,
Lacaud
,
G.
, et al. (
2021
)
CUL2(LRR1), TRAIP and p97 control CMG helicase disassembly in the mammalian cell cycle
.
EMBO Rep.
22
,
e52164
210
Kochenova
,
O.V.
,
Mukkavalli
,
S.
,
Raman
,
M.
and
Walter
,
J.C.
(
2022
)
Cooperative assembly of p97 complexes involved in replication termination
.
Nat. Commun.
13
,
6591
211
Moreno
,
S.P.
and
Gambus
,
A.
(
2020
)
Mechanisms of eukaryotic replisome disassembly
.
Biochem. Soc. Trans.
48
,
823
836
212
Tarcan
,
Z.
,
Poovathumkadavil
,
D.
,
Skagia
,
A.
and
Gambus
,
A.
(
2022
)
The p97 segregase cofactor Ubxn7 facilitates replisome disassembly during S-phase
.
J. Biol. Chem.
298
,
102234
213
Bruhl
,
J.
,
Trautwein
,
J.
,
Schafer
,
A.
,
Linne
,
U.
and
Bouazoune
,
K.
(
2019
)
The DNA repair protein SHPRH is a nucleosome-stimulated ATPase and a nucleosome-E3 ubiquitin ligase
.
Epigenetics Chromatin
12
,
52
214
Dantuma
,
N.P.
,
Acs
,
K.
and
Luijsterburg
,
M.S.
(
2014
)
Should I stay or should I go: VCP/p97-mediated chromatin extraction in the DNA damage response
.
Exp. Cell Res.
329
,
9
17
215
Coleman
,
K.E.
and
Huang
,
T.T.
(
2018
)
In a class of its own: a new family of deubiquitinases promotes genome stability
.
Mol. Cell
70
,
1
3
216
Haahr
,
P.
,
Borgermann
,
N.
,
Guo
,
X.
,
Typas
,
D.
,
Achuthankutty
,
D.
,
Hoffmann
,
S.
, et al. (
2018
)
ZUFSP deubiquitylates K63-linked polyubiquitin chains to promote genome stability
.
Mol. Cell
70
,
165
174.e6
217
Hermanns
,
T.
,
Pichlo
,
C.
,
Woiwode
,
I.
,
Klopffleisch
,
K.
,
Witting
,
K.F.
,
Ovaa
,
H.
, et al. (
2018
)
A family of unconventional deubiquitinases with modular chain specificity determinants
.
Nat. Commun.
9
,
799
218
Hewings
,
D.S.
,
Heideker
,
J.
,
Ma
,
T.P.
,
AhYoung
,
A.P.
,
El Oualid
,
F.
,
Amore
,
A.
, et al. (
2018
)
Reactive-site-centric chemoproteomics identifies a distinct class of deubiquitinase enzymes
.
Nat. Commun.
9
,
1162
219
Kwasna
,
D.
,
Abdul Rehman
,
S.A.
,
Natarajan
,
J.
,
Matthews
,
S.
,
Madden
,
R.
,
De Cesare
,
V.
, et al. (
2018
)
Discovery and characterization of ZUFSP/ZUP1, a distinct deubiquitinase class important for genome stability
.
Mol. Cell
70
,
150
164.e6
220
Foster
B
,
Attwood
M
,
Chong
K
,
Drake
M
,
Gilroy
R
,
Fournier
M
, et al.
The RPA complex orchestrates K63-linked deubiquitination via ZUP1. bioRxiv
221
Kusakabe
,
M.
,
Onishi
,
Y.
,
Tada
,
H.
,
Kurihara
,
F.
,
Kusao
,
K.
,
Furukawa
,
M.
, et al. (
2019
)
Mechanism and regulation of DNA damage recognition in nucleotide excision repair
.
Genes Environ.
41
,
2
222
Krasikova
,
Y.
,
Rechkunova
,
N.
and
Lavrik
,
O.
(
2021
)
Nucleotide excision repair: from molecular defects to neurological abnormalities
.
Int. J. Mol. Sci.
22
,
6220
223
Marteijn
,
J.A.
,
Lans
,
H.
,
Vermeulen
,
W.
and
Hoeijmakers
,
J.H.
(
2014
)
Understanding nucleotide excision repair and its roles in cancer and ageing
.
Nat. Rev. Mol. Cell Biol.
15
,
465
481
224
Zhang
,
L.
,
Nemzow
,
L.
,
Chen
,
H.
,
Lubin
,
A.
,
Rong
,
X.
,
Sun
,
Z.
, et al. (
2015
)
The deubiquitinating enzyme USP24 is a regulator of the UV damage response
.
Cell Rep.
10
,
140
147
225
Zhang
,
Y.
,
Mandemaker
,
I.K.
,
Matsumoto
,
S.
,
Foreman
,
O.
,
Holland
,
C.P.
,
Lloyd
,
W.R.
, et al. (
2021
)
USP44 stabilizes DDB2 to facilitate nucleotide excision repair and prevent tumors
.
Front. Cell Dev. Biol.
9
,
663411
226
Perez-Oliva
,
A.B.
,
Lachaud
,
C.
,
Szyniarowski
,
P.
,
Munoz
,
I.
,
Macartney
,
T.
,
Hickson
,
I.
, et al. (
2015
)
USP45 deubiquitylase controls ERCC1-XPF endonuclease-mediated DNA damage responses
.
EMBO J.
34
,
326
343
227
Gohil
,
D.
,
Sarker
,
A.H.
and
Roy
,
R.
(
2023
)
Base excision repair: mechanisms and impact in biology, disease, and medicine
.
Int. J. Mol. Sci.
24
,
14186
228
Beard
,
W.A.
,
Horton
,
J.K.
,
Prasad
,
R.
and
Wilson
,
S.H.
(
2019
)
Eukaryotic base excision repair: new approaches shine light on mechanism
.
Annu. Rev. Biochem.
88
,
137
162
229
Woodrick
,
J.
,
Gupta
,
S.
,
Camacho
,
S.
,
Parvathaneni
,
S.
,
Choudhury
,
S.
,
Cheema
,
A.
, et al. (
2017
)
A new sub-pathway of long-patch base excision repair involving 5′ gap formation
.
EMBO J.
36
,
1605
1622
230
Carter
,
R.J.
and
Parsons
,
J.L.
(
2016
)
Base excision repair, a pathway regulated by posttranslational modifications
.
Mol. Cell Biol.
36
,
1426
1437
231
Yang
,
F.
,
Hu
,
Z.
and
Guo
,
Z.
(
2022
)
Small-molecule inhibitors targeting FEN1 for cancer therapy
.
Biomolecules
12
,
1007
232
Parsons
,
J.L.
,
Dianova
,
I.I.
,
Khoronenkova
,
S.V.
,
Edelmann
,
M.J.
,
Kessler
,
B.M.
and
Dianov
,
G.L.
(
2011
)
USP47 is a deubiquitylating enzyme that regulates base excision repair by controlling steady-state levels of DNA polymerase beta
.
Mol. Cell
41
,
609
615
233
Soll
,
J.M.
,
Sobol
,
R.W.
and
Mosammaparast
,
N.
(
2017
)
Regulation of DNA alkylation damage repair: lessons and therapeutic opportunities
.
Trends Biochem. Sci.
42
,
206
218
234
Louis
,
M.
,
Hofmann
,
K.
and
Broemer
,
M.
(
2015
)
Evolutionary loss of activity in de-ubiquitylating enzymes of the OTU family
.
PLoS One
10
,
e0143227
235
Zhao
,
Y.
,
Majid
,
M.C.
,
Soll
,
J.M.
,
Brickner
,
J.R.
,
Dango
,
S.
and
Mosammaparast
,
N.
(
2015
)
Noncanonical regulation of alkylation damage resistance by the OTUD4 deubiquitinase
.
EMBO J.
34
,
1687
1703
236
Watson
,
I.R.
and
Irwin
,
M.S.
(
2006
)
Ubiquitin and ubiquitin-like modifications of the p53 family
.
Neoplasia
8
,
655
666
237
Liang
,
Y.C.
,
Lee
,
C.C.
,
Yao
,
Y.L.
,
Lai
,
C.C.
,
Schmitz
,
M.L.
and
Yang
,
W.M.
(
2016
)
SUMO5, a novel poly-SUMO isoform, regulates PML nuclear bodies
.
Sci. Rep.
6
,
26509
238
Tatham
,
M.H.
,
Jaffray
,
E.
,
Vaughan
,
O.A.
,
Desterro
,
J.M.
,
Botting
,
C.H.
,
Naismith
,
J.H.
, et al. (
2001
)
Polymeric chains of SUMO-2 and SUMO-3 are conjugated to protein substrates by SAE1/SAE2 and Ubc9
.
J. Biol. Chem.
276
,
35368
35374
239
Hickey
,
C.M.
,
Wilson
,
N.R.
and
Hochstrasser
,
M.
(
2012
)
Function and regulation of SUMO proteases
.
Nat. Rev. Mol. Cell Biol.
13
,
755
766
240
Galanty
,
Y.
,
Belotserkovskaya
,
R.
,
Coates
,
J.
,
Polo
,
S.
,
Miller
,
K.M.
and
Jackson
,
S.P.
(
2009
)
Mammalian SUMO E3-ligases PIAS1 and PIAS4 promote responses to DNA double-strand breaks
.
Nature
462
,
935
939
241
Morris
,
J.R.
,
Boutell
,
C.
,
Keppler
,
M.
,
Densham
,
R.
,
Weekes
,
D.
,
Alamshah
,
A.
, et al. (
2009
)
The SUMO modification pathway is involved in the BRCA1 response to genotoxic stress
.
Nature
462
,
886
890
242
Dou
,
H.
,
Huang
,
C.
,
Singh
,
M.
,
Carpenter
,
P.B.
and
Yeh
,
E.T.
(
2010
)
Regulation of DNA repair through deSUMOylation and SUMOylation of replication protein A complex
.
Mol. Cell
39
,
333
345
243
Dou
,
H.
,
Huang
,
C.
,
Van Nguyen
,
T.
,
Lu
,
L.S.
and
Yeh
,
E.T.
(
2011
)
SUMOylation and de-SUMOylation in response to DNA damage
.
FEBS Lett.
585
,
2891
2896
244
Luo
,
K.
,
Zhang
,
H.
,
Wang
,
L.
,
Yuan
,
J.
and
Lou
,
Z.
(
2012
)
Sumoylation of MDC1 is important for proper DNA damage response
.
EMBO J.
31
,
3008
3019
245
Marechal
,
A.
and
Zou
,
L.
(
2015
)
RPA-coated single-stranded DNA as a platform for post-translational modifications in the DNA damage response
.
Cell Res.
25
,
9
23
246
Gibbs-Seymour
,
I.
,
Oka
,
Y.
,
Rajendra
,
E.
,
Weinert
,
B.T.
,
Passmore
,
L.A.
,
Patel
,
K.J.
, et al. (
2015
)
Ubiquitin-SUMO circuitry controls activated fanconi anemia ID complex dosage in response to DNA damage
.
Mol. Cell
57
,
150
164
247
Coleman
,
K.E.
and
Huang
,
T.T.
(
2016
)
How SUMOylation fine-tunes the Fanconi anemia DNA repair pathway
.
Front. Genet.
7
,
61
248
Chang
,
Y.-C.
,
Oram
,
M.K.
and
Bielinsky
,
A.-K.
(
2021
)
SUMO-targeted ubiquitin ligases and their functions in maintaining genome stability
.
Int. J. Mol. Sci.
22
,
5391
249
Sun
,
H.
,
Leverson
,
J.D.
and
Hunter
,
T.
(
2007
)
Conserved function of RNF4 family proteins in eukaryotes: targeting a ubiquitin ligase to SUMOylated proteins
.
EMBO J.
26
,
4102
4112
250
Plechanovova
,
A.
,
Jaffray
,
E.G.
McMahon
,
S.A.
,
Johnson
,
K.A.
,
Navratilova
,
I.
,
Naismith
,
J.H.
, et al. (
2011
)
Mechanism of ubiquitylation by dimeric RING ligase RNF4
.
Nat. Struct. Mol. Biol.
18
,
1052
1059
251
Yin
,
Y.
,
Seifert
,
A.
,
Chua
,
J.S.
,
Maure
,
J.F.
,
Golebiowski
,
F.
and
Hay
,
R.T.
(
2012
)
SUMO-targeted ubiquitin E3 ligase RNF4 is required for the response of human cells to DNA damage
.
Genes Dev.
26
,
1196
1208
252
Shima
,
H.
,
Suzuki
,
H.
,
Sun
,
J.
,
Kono
,
K.
,
Shi
,
L.
,
Kinomura
,
A.
, et al. (
2013
)
Activation of the SUMO modification system is required for the accumulation of RAD51 at sites of DNA damage
.
J. Cell Sci.
126
,
5284
5292
253
Dantuma
,
N.P.
,
Groothuis
,
T.A.
,
Salomons
,
F.A.
and
Neefjes
,
J.
(
2006
)
A dynamic ubiquitin equilibrium couples proteasomal activity to chromatin remodeling
.
J. Cell Biol.
173
,
19
26
254
Garvin
,
A.J.
,
Densham
,
R.M.
,
Blair-Reid
,
S.A.
,
Pratt
,
K.M.
,
Stone
,
H.R.
,
Weekes
,
D.
, et al. (
2013
)
The deSUMOylase SENP7 promotes chromatin relaxation for homologous recombination DNA repair
.
EMBO Rep.
14
,
975
983
255
Garvin
,
A.J.
,
Walker
,
A.K.
,
Densham
,
R.M.
,
Chauhan
,
A.S.
,
Stone
,
H.R.
,
Mackay
,
H.L.
, et al. (
2019
)
The deSUMOylase SENP2 coordinates homologous recombination and nonhomologous end joining by independent mechanisms
.
Genes Dev.
33
,
333
347
256
Goodarzi
,
A.A.
,
Kurka
,
T.
and
Jeggo
,
P.A.
(
2011
)
KAP-1 phosphorylation regulates CHD3 nucleosome remodeling during the DNA double-strand break response
.
Nat. Struct. Mol. Biol.
18
,
831
839
257
Kuo
,
C.Y.
,
Li
,
X.
,
Kong
,
X.Q.
,
Luo
,
C.
,
Chang
,
C.C.
,
Chung
,
Y.
, et al. (
2014
)
An arginine-rich motif of ring finger protein 4 (RNF4) oversees the recruitment and degradation of the phosphorylated and SUMOylated Kruppel-associated box domain-associated protein 1 (KAP1)/TRIM28 protein during genotoxic stress
.
J. Biol. Chem.
289
,
20757
20772
258
Kuo
,
C.Y.
,
Li
,
X.
,
Stark
,
J.M.
,
Shih
,
H.M.
and
Ann
,
D.K.
(
2016
)
RNF4 regulates DNA double-strand break repair in a cell cycle-dependent manner
.
Cell Cycle
15
,
787
798
259
Pfeiffer
,
A.
,
Luijsterburg
,
M.S.
,
Acs
,
K.
,
Wiegant
,
W.W.
,
Helfricht
,
A.
,
Herzog
,
L.K.
, et al. (
2017
)
Ataxin-3 consolidates the MDC1-dependent DNA double-strand break response by counteracting the SUMO-targeted ubiquitin ligase RNF4
.
EMBO J.
36
,
1066
1083
260
Singh
,
A.N.
,
Oehler
,
J.
,
Torrecilla
,
I.
,
Kilgas
,
S.
,
Li
,
S.
,
Vaz
,
B.
, et al. (
2019
)
The p97-Ataxin 3 complex regulates homeostasis of the DNA damage response E3 ubiquitin ligase RNF8
.
EMBO J.
38
,
e102361
261
Claessens
,
L.A.
,
Verlaan-de Vries
,
M.
,
de Graaf
,
I.J.
and
Vertegaal
,
A.C.O.
(
2023
)
SENP6 regulates localization and nuclear condensation of DNA damage response proteins by group deSUMOylation
.
Nat. Commun.
14
,
5893
262
Komatsu
,
M.
,
Chiba
,
T.
,
Tatsumi
,
K.
,
Iemura
,
S.
,
Tanida
,
I.
,
Okazaki
,
N.
, et al. (
2004
)
A novel protein-conjugating system for Ufm1, a ubiquitin-fold modifier
.
EMBO J.
23
,
1977
1986
263
Millrine
,
D.
,
Peter
,
J.J.
and
Kulathu
,
Y.
(
2023
)
A guide to UFMylation, an emerging posttranslational modification
.
FEBS J.
290
,
5040
5056
264
Zhou
,
X.
,
Mahdizadeh
,
S.J.
,
Le Gallo
,
M.
,
Eriksson
,
L.A.
,
Chevet
,
E.
and
Lafont
,
E.
(
2024
)
UFMylation: a ubiquitin-like modification
.
Trends Biochem. Sci.
49
,
52
67
265
Millrine
,
D.
,
Peter
,
J.J.
and
Kulathu
,
Y.
(
2023
)
A guide to UFMylation, an emerging posttranslational modification
.
FEBS J.
290
,
5040
5056
266
Millrine
,
D.
,
Cummings
,
T.
,
Matthews
,
S.P.
,
Peter
,
J.J.
,
Magnussen
,
H.M.
,
Lange
,
S.M.
, et al. (
2022
)
Human UFSP1 is an active protease that regulates UFM1 maturation and UFMylation
.
Cell Rep.
40
,
111168
267
Wang
,
X.
,
Xu
,
X.
and
Wang
,
Z.
(
2023
)
The post-translational role of UFMylation in physiology and disease
.
Cells
12
,
2543
268
Komatsu
,
M.
,
Inada
,
T.
and
Noda
,
N.N.
(
2024
)
The UFM1 system: working principles, cellular functions, and pathophysiology
.
Mol. Cell
84
,
156
169
269
DaRosa
,
P.A.
,
Penchev
,
I.
,
Gumbin
,
S.C.
,
Scavone
,
F.
,
Wachalska
,
M.
,
Paulo
,
J.A.
, et al. (
2024
)
UFM1 E3 ligase promotes recycling of 60S ribosomal subunits from the ER
.
Nature
627
,
445
452
270
Makhlouf
,
L.
,
Peter
,
J.J.
,
Magnussen
,
H.M.
,
Thakur
,
R.
,
Millrine
,
D.
,
Minshull
,
T.C.
, et al. (
2024
)
The UFM1 E3 ligase recognizes and releases 60S ribosomes from ER translocons
.
Nature
627
,
437
444
271
Wang
,
Z.
,
Gong
,
Y.
,
Peng
,
B.
,
Shi
,
R.
,
Fan
,
D.
,
Zhao
,
H.
, et al. (
2019
)
MRE11 UFMylation promotes ATM activation
.
Nucleic Acids Res.
47
,
4124
4135
272
Qin
,
B.
,
Yu
,
J.
,
Nowsheen
,
S.
,
Wang
,
M.
,
Tu
,
X.
,
Liu
,
T.
, et al. (
2019
)
UFL1 promotes histone H4 ufmylation and ATM activation
.
Nat. Commun.
10
,
1242
273
Lee
,
L.
,
Perez Oliva
,
A.B.
,
Martinez-Balsalobre
,
E.
,
Churikov
,
D.
,
Peter
,
J.
,
Rahmouni
,
D.
, et al. (
2021
)
UFMylation of MRE11 is essential for telomere length maintenance and hematopoietic stem cell survival
.
Sci. Adv.
7
,
eabc7371
274
Qin
,
B.
,
Yu
,
J.
,
Zhao
,
F.
,
Huang
,
J.
,
Zhou
,
Q.
and
Lou
,
Z.
(
2022
)
Dynamic recruitment of UFM1-specific peptidase 2 to the DNA double-strand breaks regulated by WIP1
.
Genome Instab. Dis.
3
,
217
226
275
Malakhov
,
M.P.
,
Malakhova
,
O.A.
,
Kim
,
K.I.
,
Ritchie
,
K.J.
and
Zhang
,
D.E.
(
2002
)
UBP43 (USP18) specifically removes ISG15 from conjugated proteins
.
J. Biol. Chem.
277
,
9976
9981
276
Basters
,
A.
,
Geurink
,
P.P.
,
Rocker
,
A.
,
Witting
,
K.F.
,
Tadayon
,
R.
,
Hess
,
S.
, et al. (
2017
)
Structural basis of the specificity of USP18 toward ISG15
.
Nat. Struct. Mol. Biol.
24
,
270
278
277
Zhao
,
Z.
,
O'Dea
,
R.
,
Wendrich
,
K.
,
Kazi
,
N.
and
Gersch
,
M.
(
2023
)
Native semisynthesis of isopeptide-linked substrates for specificity analysis of deubiquitinases and Ubl proteases
.
J. Am. Chem. Soc.
145
,
20801
20812
278
Mirzalieva
,
O.
,
Juncker
,
M.
,
Schwartzenburg
,
J.
and
Desai
,
S.
(
2022
)
ISG15 and ISGylation in human diseases
.
Cells
11
,
538
279
Perng
,
Y.C.
and
Lenschow
,
D.J.
(
2018
)
ISG15 in antiviral immunity and beyond
.
Nat. Rev. Microbiol.
16
,
423
439
280
Park
,
J.M.
,
Yang
,
S.W.
,
Yu
,
K.R.
,
Ka
,
S.H.
,
Lee
,
S.W.
,
Seol
,
J.H.
, et al. (
2014
)
Modification of PCNA by ISG15 plays a crucial role in termination of error-prone translesion DNA synthesis
.
Mol. Cell
54
,
626
638
281
Wardlaw
,
C.P.
and
Petrini
,
J.H.J.
(
2022
)
ISG15 conjugation to proteins on nascent DNA mitigates DNA replication stress
.
Nat. Commun.
13
,
5971
282
Moro
,
R.N.
,
Biswas
,
U.
,
Kharat
,
S.S.
,
Duzanic
,
F.D.
,
Das
,
P.
,
Stavrou
,
M.
, et al. (
2023
)
Interferon restores replication fork stability and cell viability in BRCA-defective cells via ISG15
.
Nat. Commun.
14
,
6140
283
Jeon
,
Y.J.
,
Park
,
J.H.
and
Chung
,
C.H.
(
2017
)
Interferon-stimulated gene 15 in the control of cellular responses to genotoxic stress
.
Mol. Cells
40
,
83
89
284
Fan
,
J.B.
,
Arimoto
,
K.
,
Motamedchaboki
,
K.
,
Yan
,
M.
,
Wolf
,
D.A.
and
Zhang
,
D.E.
(
2015
)
Identification and characterization of a novel ISG15-ubiquitin mixed chain and its role in regulating protein homeostasis
.
Sci. Rep.
5
,
12704
285
Schmidt
,
C.K.
,
Galanty
,
Y.
,
Sczaniecka-Clift
,
M.
,
Coates
,
J.
,
Jhujh
,
S.
,
Demir
,
M.
, et al. (
2015
)
Systematic E2 screening reveals a UBE2D-RNF138-CtIP axis promoting DNA repair
.
Nat. Cell Biol.
17
,
1458
1470
286
Takeuchi
,
T.
and
Yokosawa
,
H.
(
2005
)
ISG15 modification of Ubc13 suppresses its ubiquitin-conjugating activity
.
Biochem. Biophys. Res. Commun.
336
,
9
13
287
O'Dea
,
R.
,
Kazi
,
N.
,
Hoffmann-Benito
,
A.
,
Zhao
,
Z.
,
Recknagel
,
S.
,
Wendrich
,
K.
, et al. (
2023
)
Molecular basis for ubiquitin/Fubi cross-reactivity in USP16 and USP36
.
Nat. Chem. Biol.
19
,
1394
1405
288
Harper
,
J.W.
and
Schulman
,
B.A.
(
2021
)
Cullin-RING ubiquitin ligase regulatory circuits: a quarter century beyond the F-box hypothesis
.
Annu. Rev. Biochem.
90
,
403
429
289
Soucy
,
T.A.
,
Dick
,
L.R.
,
Smith
,
P.G.
,
Milhollen
,
M.A.
and
Brownell
,
J.E.
(
2010
)
The NEDD8 conjugation pathway and its relevance in cancer biology and therapy
.
Genes Cancer
1
,
708
716
290
Hannss
,
R.
and
Dubiel
,
W.
(
2011
)
COP9 signalosome function in the DDR
.
FEBS Lett.
585
,
2845
2852
291
Ma
,
T.
,
Chen
,
Y.
,
Zhang
,
F.
,
Yang
,
C.Y.
,
Wang
,
S.
and
Yu
,
X.
(
2013
)
RNF111-dependent neddylation activates DNA damage-induced ubiquitination
.
Mol. Cell
49
,
897
907
292
Chung
,
D.
and
Dellaire
,
G.
(
2015
)
The role of the COP9 signalosome and neddylation in DNA damage signaling and repair
.
Biomolecules
5
,
2388
2416
293
Meir
,
M.
,
Galanty
,
Y.
,
Kashani
,
L.
,
Blank
,
M.
,
Khosravi
,
R.
,
Fernandez-Avila
,
M.J.
, et al. (
2015
)
The COP9 signalosome is vital for timely repair of DNA double-strand breaks
.
Nucleic Acids Res.
43
,
4517
4530
294
Luo
,
Y.
,
Su
,
Y.
and
Rao
,
F.
(
2021
)
Role of NEDD8 and neddylation dynamics in DNA damage response
.
Genome Instab. Dis.
2
,
139
149
295
Schlierf
,
A.
,
Altmann
,
E.
,
Quancard
,
J.
,
Jefferson
,
A.B.
,
Assenberg
,
R.
,
Renatus
,
M.
, et al. (
2016
)
Targeted inhibition of the COP9 signalosome for treatment of cancer
.
Nat. Commun.
7
,
13166
296
Wang
,
J.
,
Dubiel
,
D.
,
Wu
,
Y.
,
Cheng
,
Y.
,
Wolf
,
D.A.
and
Dubiel
,
W.
(
2021
)
CSN7B defines a variant COP9 signalosome complex with distinct function in DNA damage response
.
Cell Rep.
34
,
108662
297
Caracciolo
,
D.
,
Riillo
,
C.
,
Di Martino
,
M.T.
,
Tagliaferri
,
P.
and
Tassone
,
P.
(
2021
)
Alternative non-homologous end-joining: error-prone DNA repair as cancer's achilles’ heel
.
Cancers
13
,
1392
298
Chang
,
H.H.Y.
,
Pannunzio
,
N.R.
,
Adachi
,
N.
and
Lieber
,
M.R.
(
2017
)
Non-homologous DNA end joining and alternative pathways to double-strand break repair
.
Nat. Rev. Mol. Cell Biol.
18
,
495
506
299
Haakonsen
,
D.L.
and
Rape
,
M.
(
2019
)
Branching out: improved signaling by heterotypic ubiquitin chains
.
Trends Cell Biol.
29
,
704
716
300
French
,
M.E.
,
Koehler
,
C.F.
and
Hunter
,
T.
(
2021
)
Emerging functions of branched ubiquitin chains
.
Cell Discov.
7
,
6
301
Trulsson
,
F.
,
Akimov
,
V.
,
Robu
,
M.
,
van Overbeek
,
N.
,
Berrocal
,
D.A.P.
,
Shah
,
R.G.
, et al. (
2022
)
Deubiquitinating enzymes and the proteasome regulate preferential sets of ubiquitin substrates
.
Nat. Commun.
13
,
2736
302
Perez Berrocal
,
D.A.
,
Witting
,
K.F.
,
Ovaa
,
H.
and
Mulder
,
M.P.C.
(
2019
)
Hybrid chains: a collaboration of ubiquitin and ubiquitin-like modifiers introducing cross-functionality to the ubiquitin code
.
Front. Chem.
7
,
931
303
Foster
,
B.
,
Attwood
,
M.
and
Gibbs-Seymour
,
I.
(
2021
)
Tools for decoding ubiquitin signaling in DNA repair
.
Front. Cell Dev. Biol.
9
,
760226
304
Olivieri
,
M.
,
Cho
,
T.
,
Alvarez-Quilon
,
A.
,
Li
,
K.
,
Schellenberg
,
M.J.
,
Zimmermann
,
M.
, et al. (
2020
)
A genetic map of the response to DNA damage in human cells
.
Cell
182
,
481
496.e21
305
Zhao
,
Y.
,
Tabet
,
D.
,
Rubio Contreras
,
D.
,
Lao
,
L.
,
Kousholt
,
A.N.
,
Weile
,
J.
, et al. (
2023
)
Genome-scale mapping of DNA damage suppressors through phenotypic CRISPR-Cas9 screens
.
Mol. Cell
83
,
2792
2809.e9
306
Hussmann
,
J.A.
,
Ling
,
J.
,
Ravisankar
,
P.
,
Yan
,
J.
,
Cirincione
,
A.
,
Xu
,
A.
, et al. (
2021
)
Mapping the genetic landscape of DNA double-strand break repair
.
Cell
184
,
5653
5669.e25
307
Hundley
,
F.V.
,
Sanvisens Delgado
,
N.
,
Marin
,
H.C.
,
Carr
,
K.L.
,
Tian
,
R.
and
Toczyski
,
D.P.
(
2021
)
A comprehensive phenotypic CRISPR-Cas9 screen of the ubiquitin pathway uncovers roles of ubiquitin ligases in mitosis
.
Mol. Cell
81
,
1319
1336.e9
308
Cho
T
,
Zhao
Y
,
Olivieri
M
,
Hoeg
L
,
Setiaputra
D
,
Durocher
D.
Chemogenetic profiling of ubiquitin-like modifier pathways identifies NFATC2IP as a mediator of SUMO-dependent genome integrity. bioRxiv
309
Gorka
,
M.
,
Magnussen
,
H.M.
and
Kulathu
,
Y.
(
2022
)
Chemical biology tools to study deubiquitinases and Ubl proteases
.
Semin. Cell Dev. Biol.
132
,
86
96
310
Michel
,
M.A.
,
Komander
,
D.
and
Elliott
,
P.R.
(
2018
)
Enzymatic assembly of ubiquitin chains
.
Methods Mol. Biol.
1844
,
73
84
311
Jumper
,
J.
,
Evans
,
R.
,
Pritzel
,
A.
,
Green
,
T.
,
Figurnov
,
M.
,
Ronneberger
,
O.
, et al. (
2021
)
Highly accurate protein structure prediction with AlphaFold
.
Nature
596
,
583
589
312
Varadi
,
M.
,
Anyango
,
S.
,
Deshpande
,
M.
,
Nair
,
S.
,
Natassia
,
C.
,
Yordanova
,
G.
, et al. (
2022
)
AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models
.
Nucleic Acids Res.
50
,
D439
D444
313
Mirdita
,
M.
,
Schutze
,
K.
,
Moriwaki
,
Y.
,
Heo
,
L.
,
Ovchinnikov
,
S.
and
Steinegger
,
M.
(
2022
)
ColabFold: making protein folding accessible to all
.
Nat. Methods
19
,
679
682
314
Burke
,
D.F.
,
Bryant
,
P.
,
Barrio-Hernandez
,
I.
,
Memon
,
D.
,
Pozzati
,
G.
,
Shenoy
,
A.
, et al. (
2023
)
Towards a structurally resolved human protein interaction network
.
Nat. Struct. Mol. Biol.
30
,
216
225
315
Dexheimer
,
T.S.
,
Rosenthal
,
A.S.
,
Liang
,
Q.
,
Chen
,
J.
,
Villamil
,
M.A.
,
Kerns
,
E.H.
, et al. (
2010
)
Discovery of ML323 as a Novel Inhibitor of the USP1/UAF1 Deubiquitinase Complex
.
Probe Reports from the NIH Molecular Libraries Program
.
National Center for Biotechnology Information
,
Bethesda, MD
316
Lange
,
S.M.
,
Armstrong
,
L.A.
and
Kulathu
,
Y.
(
2022
)
Deubiquitinases: from mechanisms to their inhibition by small molecules
.
Mol. Cell
82
,
15
29
317
Cleary
,
J.M.
,
Aguirre
,
A.J.
,
Shapiro
,
G.I.
and
D'Andrea
,
A.D.
(
2020
)
Biomarker-guided development of DNA repair inhibitors
.
Mol. Cell
78
,
1070
1085
318
Chen
,
S.
,
Liu
,
Y.
and
Zhou
,
H.
(
2021
)
Advances in the development ubiquitin-specific peptidase (USP) inhibitors
.
Int. J. Mol. Sci.
22
,
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of University of Manchester in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.