BMB Reports 2023; 56(5): 265-274  https://doi.org/10.5483/BMBRep.2023-0037
Ubiquitin E3 ligases in cancer: somatic mutation and amplification
Eun-Hye Jo , Mi-Yeon Kim , Hyung-Ju Lee & Hee-Sae Park*
Department of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Korea
Correspondence to: Tel: +82-62-530-2197; Fax: +82-62-530-2199; E-mail: proteome@jnu.ac.kr
Received: February 24, 2023; Revised: April 14, 2023; Accepted: April 20, 2023; Published online: May 2, 2023.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Defects in DNA double-strand break (DSB) repair signaling permit cancer cells to accumulate genomic alterations that confer their aggressive phenotype. Nevertheless, tumors depend on residual DNA repair abilities to survive the DNA damage induced by genotoxic stress. This is why only isolated DNA repair signaling is inactivated in cancer cells. DNA DSB repair signaling contributes to general mechanism for various types of lesions in diverse cell cycle phases. DNA DSB repair genes are frequently mutated and amplified in cancer; however, limited data exist regarding the overall genomic prospect and functional result of these modifications. We list the DNA repair genes and related E3 ligases. Mutation and expression frequencies of these genes were analyzed in COSMIC and TCGA. The 11 genes with a high frequency of mutation differed between cancers, and mutations in many DNA DSB repair E3 ligase genes were related to a higher total mutation burden. DNA DSB repair E3 ligase genes are involved in tumor suppressive or oncogenic functions, such as RNF168 and FBXW7, by assisting the functionality of these genomic alterations. DNA damage response-related E3 ligases, such as RNF168, FBXW7, and HERC2, were generated with more than 10% mutation in several cancer cells. This study provides a broad list of candidate genes as potential biomarkers for genomic instability and novel therapeutic targets in cancer. As a DSB related proteins considerably appear the possibilities for targeting DNA repair defective tumors or hyperactive DNA repair tumors. Based on recent research, we describe the relationship between unstable DSB repairs and DSB-related E3 ligases.
Keywords: Cancer, E3 ligase, Genomic instability, Somatic mutation, Ubiquitination
E3 LIGASES REGULATE DYSFUNCTION IN CANCER

Post-translational modifications change function of most proteins. The ubiquitin system gives rise to the regulation of almost every cellular activity through proteolytic and non-proteolytic events, including protein degradation by the lysosomal pathway or through the 26S proteasome and autophagy, protein in every cellular activity the activation, and changed localization (1).

This review focuses on an up-to-date understanding of the functions of E3 ligases in cancers and debates the perspectives of cancer cells that rely on inhibition or activation of ubiquitylation of target proteins. The covalent attachment of ubiquitin to a target protein is catalyzed by the sequential action of a ubiquitin-activating enzyme (E1), a ubiquitin-conjugating enzyme (E2), and a ubiquitin ligase (E3). The E3 ubiquitination step determines the overall specificity for ubiquitination of the target protein, and the activated ubiquitin is usually transferred to a lysine residue in the substrate. Ubiquitination of proteins by a small 76 amino acid protein plays an essential role in various aspects of the DNA damage response (DDR) (2). In the case of ubiquitination signaling, the mechanism of cancer differentiation was insufficient due to the complication of ubiquitin signaling and because many aspects of the essential biology of this signaling are not yet completely understood (1). E3 ligases consist of a large family of more than 600 proteins (3), and a few E3 ligases have been classified based on their conserved structural domains (e.g., RING, RING-SCF, or HECT domains) (4, 5).

Ubiquitination of double-strand break (DSB) repair genes and proteins plays an essential role in cancer because their translocation, interaction, alteration, and regulation can affect the DNA repair pathway, making cancer cells more resistant or more prone to genomic instability. The DDR-associated E3 ligases modify histones and DDR-related proteins (Fig. 1). The failure of DNA repair results in genetic instability, and thus the detailed E3 ligase mechanism plays an important role in cancer cell instability. Therefore, it is necessary to approach effective treatment methods through mechanism research.

E3 LIGASE-DEPENDENT RECRUITMENT TO DSB SITES

The scheme of DSBs following IR (Ionizing Radiation) or NCS (Neocarzinostatin) treatment activates the Mre11/Rad50/Nbs1-Ataxia telangiectasia-mutated (MRN/ATM) complex and several E3 ligases at DSBs. In addition to induction of the H2A/γH2AX-dependent signaling, which results in the recruitment of MDC1, RNF8, RNF168, BRCA1, and 53BP1 to DSBs. DDR-related E3 ligase genetic syndromes can also lead to premature aging or cancer. In this scheme, we will describe cancer associated with DDR-related E3 ligases.

E3 ligase has an essential role in recruiting key DNA DSB repair factors to DNA damage sites. E3 ligases add one or multiple ubiquitin chains at one or several lysine residues, such as K6, K11, K27, K29, K33, K48, and K63. In most cases, K48-linked and K11-linked poly-ubiquitin chains are signals for degradation by the proteasome. Conversely, K63-linked poly-ubiquitin chains cause a change in the substrate activity, substrate signaling, and localization of the substrate (6). Activation of the MRN complex promotes initiation of the DDR and ATM recruitment to DSBs. SKP2 affects MRN complex-mediated ATM activation by triggering K63-linked NBS1 at DSB sites. Phosphorylated FBXW7 by ATM promotes K63-linked poly-ubiquitination of XRCC4 to facilitate recruitment of Ku70/80 for nonhomologous end joining (NHEJ) repair (7-9). WWP2 induces DNA-PKcs (Protein Kinase, Catalytic Subunit)-dependent proteasomal degradation of RNAPII. RNAPII inhibits DNA-PK, XRCC4, DNA ligase IV (LigIV), and XLF (10). RNAPII degradation protects NHEJ machinery in DSB repair sites.

Ubiquitination of histones H2A, H2B, H3, and H4 is important for DNA repair signaling. Subsequent to DSBs, γH2A, K13, and K15 are ubiquitinated by RNF168, and K127 and K129 of BRCA1/BRCA1-associated RING domain protein 1 (BARD1) are ubiquitinated (11). The DNA DSB of MDC1 is polyubiquitinated at K63 by RNF8 and subsequently recognized by RNF168, leading to the recruitment of repair proteins, such as BRCA1 and 53BP1 (12). HERC2 recruited by MDC1 promotes RNF8 oligomerization and expedites the formation of the HERC2/MDC1/RNF8 complex, following RNF8-mediated recruitment of RNF168 (13). The dimethylation of H4K20 is recruitment of 53BP1, where it commends repair pathway selection decisions by limiting DNA end resection at DSB sites. Accumulation of 53BP1 disrupt DNA end resection and homologous recombination (HR) whereas implement NHEJ (14). The K127/129 of H2A binds to BRCA1/BARD1 and promotes end resection and HR (15). The PALB2/BRCA1 complex recruits RAD51 and BRCA2 and promotes RAD51 replacement of RPA (16). Although HR is usually valuable for preserve genome integrity, DSB repair by HR is highly inhibited in G1 cells in non-dividing cells. KEAP1 inhibits interaction of PALB2/BRCA1 and dissociation of BRCA1 inhibits HR in G1 phase (17). The ubiquitination of RAP80 binds BRCA1 complex to DSB sites. The RAP80/BRCA1 complex is required for efficient BRCA1 recruitment, that suppresses excessive DSB end processing, HR repair, and chromosomal instability. BRCA1 induces ubiquitination of CtIP and promotes its chromatin-loading at the G2/M checkpoint (18). The interaction of BRCA1/CtIP was initially implicated in the G2/M checkpoint and apoptosis regulation. The relation of BRCA1/CtIP facilitates HR through ubiquitination. However, separation of the BRCA1/CtIP complex in the G2/M checkpoint accelerates apoptosis in damaged cells. RNF138 recruits MRE11 but not CtIP at ssDNA overhangs resembling initial processing of DSBs RNF138 mediates Ku80 ubiquitylation following DSB repair pathway utilization, Ku70/80, and MRN and promotes RPA loading, CtIP, and exonuclease 1 (EXO1), and hence, facilitates HR (19). RNF168, competing with RNF169, is recruited to DSBs, eliminates RAP80 by histone, and facilitates HR. RNF20/RNF40 mono-ubiquitinates histone H2BK120 at DSBs, which leads to chromatin relaxation. RNF126 upregulates HR through BRCA1 interacting with E2F1 (20). RNF126 promotes K48-linked ubiquitination of Ku70/Ku80, loosening from DNA DSB sites, which recruits proteins that mediate repair by NHEJ (21). Finally, RNF126 directly ubiquitinates RNF168, thereby inhibiting RNF168-mediated ubiquitination of γH2AX (22).

Several cancers are caused by endogenous and exogenous DNA damage. Defects in the DNA DSB mechanisms are triggered by chromosomal abnormalities that cause tumorigenesis and genomic instability. DNA DSB factors and ubiquitination of histones play crucial functions in recruiting proteins to damaged DSB sites and the repair operation (23). In this review, we study the DDR-related E3 ligase genes, such as RNF8, RNF20/40, RNF53 (BRCA1), RNF126, RNF138, RNF168, RNF169, SKP2, FBXW7, HERC2, and WWP2 (Fig. 2).

RNF8

Once DSB occurs, auto-activated ATM rapidly phosphorylates γH2AX at S139, recruiting MDC1 to the DSB sites (24-26). γH2AX recruits the E3 ligases RNF8 and RNF168 (27). RNF8 and RNF168 promote the K63-linked polyubiquitination of γH2AX on K13 and K15. As a result, 53BP1 and BRCA1 are competitively combined polyubiquitination of γH2AX to determine repair pathway (28). Furthermore, RNF8-mediated ubiquitination of NBS1 K435 is essential for HR. The genetic mutations and expression of RNF8 were investigated using the cBioPortal for Cancer Genomics (cBioPortal) databases. The results found that across 584 ovarian cancers in the cBioPortal, RNF8 exhibited high gene alteration frequencies (4.97%, respectively), and RNF8 gene amplification was very high. The rates of point mutations in RNF8 were 3.97% (21 cases) in 529 uterine corpus endometrial carcinomas. These datasets showed significantly increased expression and mutation of RNF8 in cancers, such as ovarian serous cystadenocarcinoma, uterine corpus endometrial carcinoma, and skin cutaneous melanoma. MiR-214 inhibits RNF8, thereby disrupting DNA repair to induce chromosomal instability in ovarian cancers (29). According to a recent study, RNF8 causes epithelial-mesenchymal transition (EMT) and CSC self-renewal in triple-negative breast cancer cells. Furthermore, RNF8 triggers cancer cell invasion and migration and cancer metastasis, with the result that cancer cells induce chemoresistance and tumor aggressiveness (30).

RNF20/40

Numerous histone-modifying E3 ligases have been implicated in DSBs. RNF20/RNF40 mono-ubiquitinylates histone H2B on K120 (H2Bub). It is required for DSB repair leading to HR and class switch recombination, and also induces the recruitment of BRCA1 and RAD51 (31, 32). RNF20/40-dependent H2Bub is essential for transcription of H3K4me3 and H3K79me2. The RNF20/RNF40 association with the RNA polymerase II-associated factor 1 (PAF1) complex regulates H2Bub at the coding regions of transcriptionally active downstream genes (33-35). The mutation of RNF20 (>5% of samples) has been detected in three tumor types: uterine corpus endometrial, skin cutaneous melanoma carcinoma, and colorectal adenocarcinoma. Despite that, the prognostic significance of RNF20 mutations in uterine carcinosarcoma remains uncertain. P53 regulates the transcription of p21 and PUMA through the RNF20/RNF40 complex-dependent recruitment of H2Bub. A lack of the RNF20/RNF40 complex inhibited not only H2Bub but also the mature mRNA of p21 and PUMA. PUMA regulates mRNA splicing for tumor suppressor genes in colon cancer (36).

RNF53 (BRCA1)

The breast and ovarian cancer gene, BRCA1, encodes a RING domain flanked by long α-helices. The BRCA1/BARD1 heterodimer has an essential role in E3 ligase activity and participates in chromosome stability and cell proliferation (37). BRCA1/BARD1 ubiquitylates K125/127/129 of H2A (38). SMARCAD1 interacts ubiquitinated H2A. SMARCAD1 is required for 53BP1 repositioning (15). The ubiquitination signaling of γH2AX results in the ultimate recruitment of DSB repair proteins 53BP1 and BRCA1. 53BP1 is associated with cNHEJ repair of the DSB, while BRCA1 regulates HR-mediated repair. The overall incidence of DDR pathway alterations of BRCA1 in the provisional datasets of The Cancer Genome Atlas (TCGA) database was 8.88% of the 529 samples. There were 47 mutations (8.88%) among the altered genes; the most frequently involved gene was RNF168, followed by FBXW7 and HERC2 (10.78%, 38.6%, and 17.58%, respectively; Table 1). BRCA1 somatic mutations are newly recognized as clinically significant factors of genomic instability (39). The highest rates of BRCA1 mutations tend to cause somatic loss-of-function mutations in ovarian cancer due to massive genomic instability and tumor formation (40, 41). BRCA1-mutated tumors regulate EMT in breast cancer cells and cancer metastasis, the cause of death from all cancers (42).

RNF126

RNF126 positively regulates HR through BRCA1 transcription by interacting with E2F1 (20). RNF126/E2F1 facilitates the transcription of CHK1, which is required in repairing replication stress-induced DSB (43). RNF126 promotes NHEJ by K48-linked ubiquitination of Ku70/Ku80 and loosening of the Ku70/80 heterodimer from DNA DSB sites. Interestingly, the release of the Ku70/80 heterodimer from DNA DSB sites facilitates the accomplishment of DSB repair (21). Finally, RNF126 recruitment at DSB sites occurs in an ATM- and RNF8-dependent manner and RNF126 directly ubiquitinates RNF168, which leads to the inhibition of RNF168-mediated ubiquitination of γH2AX (22). By contrast, RNF126 negatively regulates the foci formation of 53BP1, γH2AXub, RNF168, BRCA1, and RAP80 IR-induced DSB (43). The cBioPortal database showed significantly increased somatic mutation of RNF126 in sarcoma. Numerous examples of high RNF126 expression in sarcoma and brain lower grade glioma have been documented. Recently, studies have revealed an oncogenic role of RNF126 in various cancers, such as breast, gastric, prostate, and ovarian cancers (43-46). RNF126 was overexpressed in human colorectal cancer specimens, which was closely related to tumor size, lymph node metastasis, and the poor survival of colorectal cancer (45). Furthermore, RNF126, as an oncogene, causes ubiquitination and the proteasome pathway degradation of PTEN in bladder cancer and, consequently, induces pulmonary metastasis in bladder cancer (43).

RNF138

RNF138 regulates HR, which is initiated by DNA end resection, and this mechanism is associated with the ubiquitylation of Ku80 (19). RNF138 ubiquitylates Ku80 following HR repair pathway utilization. Once Ku is removed, RNF138 recruits RPA loading and CtIP and EXO1; consequently, the DNA ends produce ssDNA overhangs (47). Highly expressed RNF138 increased cell viability and postponed cell cycle progression and cisplatin resistance by inhibiting Chk1 signaling. RNF138 modulates cisplatin resistance in gastric cancer cells. Furthermore, cisplatin resistance leads to a promising drug target to explore chemotherapy failure (47). The cBioPortal database showed significantly increased somatic mutation of RNF138 in uterine corpus endometrial carcinoma and its high amplification in ovarian serous cystadenocarcinoma. According to a recent study, RNF138-mediated rpS3 ubiquitination induces nuclear translocation of rpS3, thereby led to radioresistance in glioblastoma cells (48).

RNF168

Activation of ATM induced by DNA DSBs recruits the ubiquitin ligases, RNF8, and RNF168, which induce chromatin-ubiquitinating histone γH2AX near DSB sites. The two E3 ubiquitin ligases RNF8/RNF168 ubiquitylate γH2AX K13/K15 upon genotoxic stress (11). This complicated ubiquitin network regulates the foci formation to promote the recruitment of DSB factors, such as BRCA1 and 53BP1, required for downstream signaling (49, 50). Recently, the RNF168/UbcH5c complex was shown to promote K27 ubiquitination of nucleosomal H2A/γH2AX. H2A/γH2AX K27 ubiquitination is required to activate DSB factors, such as 53BP1, RAP80, RNF168, and RNF169 (51). RNF168 is aberrantly overexpressed in many cancers, and one reason for this is the presence of mutations in DSBs. RNF168 was overexpressed in various cancers, such as lung, esophageal, and ovarian cancers. RNF168 was the amplified gene in nearly all cancers, with 31.62% of lung cancer showing a few types of RNF168 mutation and were consistent with mutations that influenced DNA repair. Overexpression of RNF168 and activated 53BP1 recruitment induce mutagenic NHEJ at the HR repair, resulting in reinforced genomic instability and radio-resistance in breast cancer cell lines (52). Of 27 cancer types with DSB-related RNF168 has been confirmed to be a mutation in 20 cases (74.07%) in various cancer cells (Table 1). Moreover, we examined the commonness of NHEJ pathway alterations by cancer type and identified the most frequently mutated gene within the signaling pathway for each cancer type. For example, RNF168 exhaustion inhibits the growth of BRCA1/BRCA2-deficient breast and ovarian cancer cells, resulting in DSBs, senescence, and subsequent cell death (53).

RNF169

RNF169 has a similar domain architecture to RNF168. Despite this, the affinity of RNF169 for γH2A K13/K15 is higher than that of RNF168 (54). The UDM2 domain of RNF169 prevents IR-induced 53BP1 and regulates RNF168-dependent signaling by interrupting DNA DSB-induced ubiquitylation of γH2AK13/K15ub (55). Remarkably, RNF169 is necessary for the recruitment of various DDR proteins, including BRCA1, RAP80, and RAD18, near DSB sites (56). Gain-of-function mutations were amplified in RNF169 in esophageal cancer. The cBioPortal database showed that RNF169 amplification was significantly associated with various cancers, such as esophageal, ovarian, and breast cancer. USP7 deubiquitylates and stabilizes RNF169 through nuclear import in breast cancer (57).

SKP2

SKP2, also known as F-Box/LRR-repeat protein 1 (FBXL1) or p45, is one of the members of the F-box protein family and is involved in ubiquitination, cell cycle regulation, and signal transduction regulated by the SKP2-SCF complex (58, 59). SKP2 affects MRN (Mre11-Rad50-Nbs1) complex-mediated ATM activation by triggering K63-linked NBS1 at DSB sites (60). The cBioPortal database showed significantly increased SKP2 amplification in cancers, such as lung squamous cell carcinoma, lung adenocarcinoma, and bladder urothelial carcinoma (60). Numerous examples of high SKP2 somatic mutation in uterine corpus endometrial carcinoma have been documented. Overexpression of SKP2 plays a multifaceted role in cancer by enhancing the interaction between ATM and the MRN complex. SKP2 remarkably promotes PDCD4 phosphorylation, ubiquitination, and degradation. In other cancer cells, SKP2 induces cell proliferation, cell apoptosis, and inhibition of enhanced PDCD4 during DSB repair in breast cancer (61).

FBXW7

FBXW7 is established as the substrate recognition subunit for the SCF E3 ubiquitin ligase (62, 63). Activated ATM phosphorylates S26 of FBXW7 to promote its recruitment to DSB sites. In the DSB pathway, Ku70/80 quickly binds to DNA ends and serves as the scaffold to recruit the core NHEJ machinery. Subsequently, The DNA-PKcs, XRCC4, Artemis, XLF, and LigIV are independently recruited to the Ku-DNA complex (7-9). FBXW7-mediated polyubiquitination of K63-linked XRCC4 on K296 promotes NHEJ, expedites NHEJ complex formation, and promotes NHEJ repair (64). Highly mutations have been found in FBXW7 (>10% of samples) in at least three tumor types: uterine carcinosarcoma, uterine corpus endometrial carcinoma, and colorectal adenocarcinoma. Despite numerous studies, the prognostic importance of FBXW7 mutations in uterine carcinosarcoma remains uncertain. FBXW7 regulates hematopoietic stem cell (HSC) inactivity and self-renewal (65, 66). FBXW7 regulates the degradation of cyclin E, upregulates Myc overexpression in HSC, and causes defective HSC self-renewal after hematologic stress (67, 68). Decreased cyclin E causes extraordinary cell cycle control, epithelial cell proliferation, impaired genome instability, and erythroid cell differentiation (69, 70).

HERC2

The HERC2 E3 ubiquitin ligase interacts with NEURL4 and MDM2 to regulate p53-dependent gene stability in ATM- and ATR-induced DNA DSB repair (71, 72). The DSB repair pathway begins with the recognition of the sites of DNA damage by the MRN complex, then NBS1 recruitment, which promotes the binding of ATM to the DSB site, in turn, leading to phosphorylation of γH2AX S139 and bind MDC1 (73). The γH2AX/MDC1 complex promotes the recruitment of HERC2 and RNF8. HERC2 promotes RNF8 oligomerization and expedites the formation of the HERC2-MDC1-RNF8 complex, following RNF8-mediated recruitment of RNF168 (13). In addition to regulating the MDM2-p53 transcriptional pathway, HERC2 can inhibit potentially oncogenic mutations from being passed to daughter cells by regulating DNA DSBs during the S and G2-M phases of mitosis (74, 75). The cBioPortal database showed significantly increased overexpression and mutation of HERC2 in cancers, such as skin cutaneous melanoma, uterine corpus endometrial carcinoma, and lung adenocarcinoma. Furthermore, amplification of HERC2 has been discovered in several human cancers, such as cutaneous cancer, uveal melanoma, and non-small cell lung cancer (76). Additionally, the types of cancer associated with HERC2 include pheochromocytoma, paraganglioma, and T-cell prolymphocytic leukemia (77, 78).

WWP2

Human WWP2 contains a HECT domain E3 ligase for NHEJ repairs. RNA polymerase II (RNAPII) is recruited to DNA double-strand breaks for inhibiting of transcription. As WWP2 promotes the degradation of RPB1 of RNAPII subunit through K48-linked ubiquitination regulates DNA-PK-dependent transcription arrest and repair at DNA breaks. The cBioPortal database showed significantly increased somatic mutation of WWP2 in cancers, such as uterine corpus endometrial carcinoma. Numerous examples of high expression of WWP2 in breast cancer and liver cancer have been documented. WWP2 has key roles in the evasion of apoptosis and the over-proliferation of liver cancer; WWP2 overexpression was upregulated in cancer tissues. WWP2 knockdown remarkably decreased the proliferation, caused apoptosis, and induced cell-cycle arrest in Huh7 and BEL-7404 cells (79-82). Moreover, WWP2 is highly expressed in hepatocellular carcinoma and activation of AKT signaling to increase tumor metastasis (83). DNA damage enhances the differentiation of glioma stem cells by WWP2. SOX2 regulates to maintain the self-renewal potential of glioma stem cells in glioblastoma. WWP2 ubiquitylates SOX2 through its HECT domain to enhance Sox2 ubiquitination, thus stabilizing glioma stem cell maintenance (84). 

DISCUSSION

Over the last decades, DSB-associated E3 ligases have been continuously studied for their importance of somatic mutations in cancer cells, but they have not yet solved all the mechanisms. In this study, we revealed that genomic instability caused by DSB-related E3 ligases is manifested as distinct cancer somatic mutations in RNF20/40, BRCA1, RNF168, FBXW7, HERC2, and WWP2 and the expression levels of these genes. Based on the mutation expression of DDR-related E3 ligase through the TCGA, associated with gene instability and cancer. Most of the DSB-related E3 ligases have known functions, but the physical functions in cancer somatic mutations and over-amplification have not been elucidated. It is necessary to study genomic instability through mutations found in patients with cancer. Although many studies have been conducted on the association between gene amplification and cancer, research on the association with mutations is still lacking. The realization that DNA DSB repair pathways, which protect the genome against DSB-related gene changes, can serve as essential targets for cancers is highly encouraging. DSB-related genes are highly expressed in particular cancers, induce radioresistance or chemoresistance. Consequently, strategies utilizing the DNA DSB repair pathways as targets for anticancer treatment can result in the development of potent candidates for therapeutics. Research on gene amplification and mutation-induced gene instability in cancer cells is continuously needed based on the TCGA database and samples from patients with cancer. 

ACKNOWLEDGEMENTS

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (Ministry of Science and ICT) (No. NRF-2022R1H1A2091883, NRF-2019R1A2C1007197).

CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. Several E3 ligases in cancer. Ubiquitination in tumorigenesis and senescence.
Fig. 2. DSB proteins and their regulator in various cancers. We have shown DSB related E3 ligases mentioned in several cancers and how they function in regulating cancer cell. For example, the DSB related E3 ligases RNF20/40, RNF8, RNF126, RNF138, RNF168, RNF169, BRCA1, FBXW7, HERC2, WWP2 are shown to regulate various cancer cell DSB repair by targeting substrates for ubiquitination.
TABLE

Analysis of data in The Cancer Genome Atlas (TCGA) cBioPortal indicates genetic alterations of E3 ligase in several types of cancer

E3 ligase Related E3 ligase in cancer Mutation (%) Mutation sites Amplification (%) Reference
RNF8
  • Ovarian serous cystadenocarcinoma (40)

  • Uterine corpus endometrial carcinoma (UCEC)

  • Skin cutaneous melanoma

UCEC: genes altered in 4.91% of 529 cases 3.97% (21 cases)
  • R356H

  • S465R

  • K279Q

  • M460I

  • K315T

OSC: genes altered in 4.97% of 584 cases 4.97% (29 cases) (85, 86)
RNF20/40
  • UCEC

  • Sarcoma

  • Lung adenocarcinoma

UCEC: genes altered in 7.18% of 529 cases 6.81% (36 cases)
  • R971H

  • K612T

  • K689N

  • R105H

  • K668N

Adrenocortical carcinoma: genes altered in 3.3% of 91 cases 3.3% (3 cases) (31)
RNF53 (BRCA1)
  • UCEC

  • Diffuse large B-cell lymphoma

  • Skin cutaneous melanoma

UCEC: genes altered in 9.26% of 529 cases 8.88% (47 cases)
  • T1561A

  • S1360I

  • L1084F

  • K793T

  • P1315L

Pancreatic adenocarcinoma: genes altered in 3.26% of 184 cases 2.17% (4 cases) (49, 87)
RNF126
  • Sarcoma

  • Cervical squamous cell carcinoma

  • Brain lower grade glioma

Skin cutaneous melanoma: genes altered in 2.7% of 444 cases 2.25% (10 cases)
  • P217L

  • Q77H

  • E92D

  • E38K

  • P76Q

Sarcoma: genes altered in 8.24% of 255 cases 5.49% (14 cases) (20, 22, 88)
RNF138
  • OSC

  • Pancreatic adenocarcinoma

  • Esophageal adenocarcinoma

UCEC: genes altered in 3.02% of 529 cases 2.65% (14 cases)
  • R97C

  • R171C

  • D74N

  • K91Q

  • A236V

OSC: genes altered in 5.14% of 584 cases 4.11% (24 cases) (19, 89)
RNF168
  • Lung squamous cell carcinoma

  • Esophageal adenocarcinoma

  • OSC

UCEC: genes altered in 10.78% of 529 cases 4.35% (23 cases)
  • V486L

  • E307K

  • K528N

  • D516G

  • P209S

Lung squamous cell carcinoma: genes altered in 31.62% of 487 cases 30.18% (147 cases) (54, 90)
RNF169
  • Esophageal adenocarcinoma

  • OSC

  • Breast invasive carcinoma

UCEC: genes altered in 4.35% of 529 cases 3.02% (16 cases)
  • R641*

  • R620Q

  • R690Q

  • E377K

  • R245H

Esophageal adenocarcinoma: genes altered in 8.79% of 182 cases 6.59% (12 cases) (91, 92)
Skp2
  • Lung squamous cell carcinoma

  • Lung adenocarcinoma

  • Bladder urothelial carcinoma

UCEC: genes altered in 6.24% of 529 cases mutation 3.21% (17 cases)
  • E45A

  • K28N

  • R83C

  • Y128H

  • T225I

Lung squamous cell carcinoma: genes altered in 10.47% of 487 cases 10.06% (49 cases) (60)
FBW7
  • Uterine carcinosarcoma

  • UCEC

  • Carcinoma colorectal adenocarcinoma

Uterine carcinosarcoma: genes altered in 38.6% of 57 cases 38.6% (22 cases)
  • R441Q

  • R473Kfs*4

  • L290F

  • R505C

  • T653A

Sarcoma: genes altered in 3.53% of 255 cases 2.75% (7 cases) (64)
HERC2
  • Skin cutaneous melanoma

  • UCEC

  • Lung adenocarcinoma

UCEC: genes altered in 18.53% of 529 cases 17.58% (93 cases)
  • D2901N

  • A335T

  • V3245M

  • R2031Q

  • R2022W

Sarcoma: genes altered in 5.1% of 255 cases 1.96% (5 cases) (13)
WWP2
  • UCEC

  • Cholangiocarcinoma

  • Stomach adenocarcinoma

UCEC: genes altered in 7.37% of 529 cases 5.29% (28 cases)
  • G817R

  • T830I

  • E703*

  • I139N

  • X561_splice

Cholangiocarcinoma: genes altered in 5.56% of 36 cases 2.78% (1 case) (10)

REFERENCES
  1. Swatek KN and Komander D (2016) Ubiquitin modifications. Cell Res 26, 399-422
    Pubmed KoreaMed CrossRef
  2. Lukas J, Lukas C and Bartek J (2011) More than just a focus: the chromatin response to DNA damage and its role in genome integrity maintenance. Nat Cell Biol 13, 1161-1169
    Pubmed CrossRef
  3. Li W, Bengtson MH, Ulbrich A et al (2008) Genome-wide and functional annotation of human E3 ubiquitin ligases identifies MULAN, a mitochondrial E3 that regulates the organelle's dynamics and signaling. PLoS One 3, e1487
    Pubmed KoreaMed CrossRef
  4. Buetow L and Huang DT (2016) Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nat Rev Mol Cell Biol 17, 626-642
    Pubmed KoreaMed CrossRef
  5. Metzger MB, Hristova VA and Weissman AM (2012) HECT and RING finger families of E3 ubiquitin ligases at a glance. J Cell Sci 125, 531-537
    Pubmed KoreaMed CrossRef
  6. Sharma N, Zhu Q, Wani G, He J, Wang QE and Wani AA (2014) USP3 counteracts RNF168 via deubiquitinating H2A and γH2AX at lysine 13 and 15. Cell Cycle 13, 106-114
    Pubmed KoreaMed CrossRef
  7. Critchlow SE, Bowater RP and Jackson SP (1997) Mammalian DNA double-strand break repair protein XRCC4 interacts with DNA ligase IV. Curr Biol 7, 588-598
    Pubmed CrossRef
  8. Davis AJ and Chen DJ (2013) DNA double strand break repair via non-homologous end-joining. Transl Cancer Res 2, 130-143
    Pubmed KoreaMed CrossRef
  9. Lieber MR (2010) The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem 79, 181-211
    Pubmed KoreaMed CrossRef
  10. Caron P, Pankotai T, Wiegant WW et al (2019) WWP2 ubiquitylates RNA polymerase II for DNA-PK-dependent transcription arrest and repair at DNA breaks. Genes Dev 33, 684-704
    Pubmed KoreaMed CrossRef
  11. Gatti M, Pinato S, Maspero E, Soffientini P, Polo S and Penengo L (2012) A novel ubiquitin mark at the N-terminal tail of histone H2As targeted by RNF168 ubiquitin ligase. Cell Cycle 11, 2538-2544
    Pubmed KoreaMed CrossRef
  12. Nowsheen S, Aziz K, Aziz A et al (2018) L3MBTL2 orchestrates ubiquitin signalling by dictating the sequential recruitment of RNF8 and RNF168 after DNA damage. Nat Cell Biol 20, 455-464
    Pubmed KoreaMed CrossRef
  13. Oestergaard VH, Pentzold C, Pedersen RT et al (2012) RNF8 and RNF168 but not HERC2 are required for DNA damage-induced ubiquitylation in chicken DT40 cells. DNA Repair (Amst) 11, 892-905
    Pubmed CrossRef
  14. Pei H, Zhang L, Luo K et al (2011) MMSET regulates histone H4K20 methylation and 53BP1 accumulation at DNA damage sites. Nature 470, 124-128
    Pubmed KoreaMed CrossRef
  15. Densham RM, Garvin AJ, Stone HR et al (2016) Human BRCA1-BARD1 ubiquitin ligase activity counteracts chromatin barriers to DNA resection. Nat Struct Mol Biol 23, 647-655
    Pubmed KoreaMed CrossRef
  16. Zhang F, Ma J, Wu J et al (2009) PALB2 links BRCA1 and BRCA2 in the DNA-damage response. Curr Biol 19, 524-529
    Pubmed KoreaMed CrossRef
  17. Orthwein A, Noordermeer SM, Wilson MD et al (2015) A mechanism for the suppression of homologous recombination in G1 cells. Nature 528, 422-426
    Pubmed KoreaMed CrossRef
  18. Yu X, Fu S, Lai M, Baer R and Chen J (2006) BRCA1 ubiquitinates its phosphorylation-dependent binding partner CtIP. Genes Dev 20, 1721-1726
    Pubmed KoreaMed CrossRef
  19. Ismail IH, Gagné JP, Genois MM et al (2015) The RNF138 E3 ligase displaces Ku to promote DNA end resection and regulate DNA repair pathway choice. Nat Cell Biol 17, 1446-1457
    Pubmed CrossRef
  20. Wang Y, Deng O, Feng Z et al (2016) RNF126 promotes homologous recombination via regulation of E2F1-mediated BRCA1 expression. Oncogene 35, 1363-1372
    Pubmed KoreaMed CrossRef
  21. Ishida N, Nakagawa T, Iemura SI et al (2017) Ubiquitylation of Ku80 by RNF126 promotes completion of nonhomologous end joining-mediated DNA repair. Mol Cell Biol 37, e00347-00316
    Pubmed KoreaMed CrossRef
  22. Zhang L, Wang Z, Shi R et al (2018) RNF126 quenches RNF168 function in the DNA damage response. Genomics Proteomics Bioinformatics 16, 428-438
    Pubmed KoreaMed CrossRef
  23. Jackson SP and Bartek J (2009) The DNA-damage response in human biology and disease. Nature 461, 1071-1078
    Pubmed KoreaMed CrossRef
  24. Coster G and Goldberg M (2010) The cellular response to DNA damage: a focus on MDC1 and its interacting proteins. Nucleus 1, 166-178
    Pubmed CrossRef
  25. Iacovoni JS, Caron P, Lassadi I et al (2010) High-resolution profiling of gammaH2AX around DNA double strand breaks in the mammalian genome. Embo J 29, 1446-1457
    Pubmed KoreaMed CrossRef
  26. Kozlov SV, Graham ME, Jakob B et al (2011) Autophosphorylation and ATM activation: additional sites add to the complexity. J Biol Chem 286, 9107-9119
    Pubmed KoreaMed CrossRef
  27. Bartocci C and Denchi EL (2013) Put a RING on it: regulation and inhibition of RNF8 and RNF168 RING finger E3 ligases at DNA damage sites. Front Genet 4, 128
    Pubmed KoreaMed CrossRef
  28. Nakada S (2016) Opposing roles of RNF8/RNF168 and deubiquitinating enzymes in ubiquitination-dependent DNA double-strand break response signaling and DNA-repair pathway choice. J Radiat Res 57 Suppl 1, i33-i40
    Pubmed KoreaMed CrossRef
  29. Wang Z, Yin H, Zhang Y et al (2014) miR-214-mediated downregulation of RNF8 induces chromosomal instability in ovarian cancer cells. Cell Cycle 13, 3519-3528
    Pubmed KoreaMed CrossRef
  30. Lee HJ, Li CF, Ruan D et al (2016) The DNA damage transducer RNF8 facilitates cancer chemoresistance and progression through twist activation. Mol Cell 63, 1021-1033
    Pubmed KoreaMed CrossRef
  31. Moyal L, Lerenthal Y, Gana-Weisz M et al (2011) Requirement of ATM-dependent monoubiquitylation of histone H2B for timely repair of DNA double-strand breaks. Mol Cell 41, 529-542
    Pubmed KoreaMed CrossRef
  32. Nakamura K, Kato A, Kobayashi J et al (2011) Regulation of homologous recombination by RNF20-dependent H2B ubiquitination. Mol Cell 41, 515-528
    Pubmed CrossRef
  33. Kim J, Guermah M, McGinty RK et al (2009) RAD6-Mediated transcription-coupled H2B ubiquitylation directly stimulates H3K4 methylation in human cells. Cell 137, 459-471
    Pubmed KoreaMed CrossRef
  34. Pavri R, Zhu B, Li G et al (2006) Histone H2B monoubiquitination functions cooperatively with FACT to regulate elongation by RNA polymerase II. Cell 125, 703-717
    Pubmed CrossRef
  35. Shiloh Y, Shema E, Moyal L and Oren M (2011) RNF20-RNF40: a ubiquitin-driven link between gene expression and the DNA damage response. FEBS Lett 585, 2795-2802
    Pubmed CrossRef
  36. Wu C, Cui Y, Liu X, Zhang F, Lu L-Y and Yu X (2020) The RNF20/40 complex regulates p53-dependent gene transcription and mRNA splicing. J Mol Cell Biol 12, 113-124
    Pubmed KoreaMed CrossRef
  37. Joukov V, Groen AC, Prokhorova T et al (2006) The BRCA1/BARD1 heterodimer modulates ran-dependent mitotic spindle assembly. Cell 127, 539-552
    Pubmed CrossRef
  38. Kalb R, Mallery Donna L, Larkin C, Huang Jeffrey TJ and Hiom K (2014) BRCA1 Is a histone-H2A-specific ubiquitin ligase. Cell Rep 8, 999-1005
    Pubmed KoreaMed CrossRef
  39. Piek JM, Torrenga B, Hermsen B et al (2003) Histopathological characteristics of BRCA1- and BRCA2-associated intraperitoneal cancer: a clinic-based study. Fam Cancer 2, 73-78
    Pubmed CrossRef
  40. Moschetta M, George A, Kaye SB and Banerjee S (2016) BRCA somatic mutations and epigenetic BRCA modifications in serous ovarian cancer. Ann Oncol 27, 1449-1455
    Pubmed CrossRef
  41. Pal T, Permuth-Wey J, Betts JA et al (2005) BRCA1 and BRCA2 mutations account for a large proportion of ovarian carcinoma cases. Cancer 104, 2807-2816
    Pubmed CrossRef
  42. Zavitsanos PJ, Wazer DE, Hepel JT, Wang Y, Singh K and Leonard KL (2018) BRCA1 mutations associated with increased risk of brain metastases in breast cancer: a 1: 2 matched-pair analysis. Am J Clin Oncol 41, 1252-1256
    Pubmed CrossRef
  43. Zhi X, Zhao D, Wang Z et al (2013) E3 ubiquitin ligase RNF126 promotes cancer cell proliferation by targeting the tumor suppressor p21 for ubiquitin-mediated degradation. Cancer Res 73, 385-394
    Pubmed CrossRef
  44. Migita K, Matsumoto S, Wakatsuki K et al (2020) RNF126 as a marker of prognosis and proliferation of gastric cancer. Anticancer Res 40, 1367-1374
    Pubmed CrossRef
  45. Wang S, Wang T, Wang L, Zhong L and Li K (2020) Overexpression of RNF126 promotes the development of colorectal cancer via enhancing p53 ubiquitination and degradation. Onco Targets Ther 13, 10917-10929
    Pubmed KoreaMed CrossRef
  46. Yang X, Pan Y, Qiu Z et al (2018) RNF126 as a biomarker of a poor prognosis in invasive breast cancer and CHEK1 inhibitor efficacy in breast cancer cells. Clin Cancer Res 24, 1629-1643
    Pubmed KoreaMed CrossRef
  47. Lu Y, Han D, Liu W et al (2018) RNF138 confers cisplatin resistance in gastric cancer cells via activating Chk1 signaling pathway. Cancer Biol Ther 19, 1128-1138
    Pubmed KoreaMed CrossRef
  48. Kim W, Youn H, Lee S et al (2018) RNF138-mediated ubiquitination of rpS3 is required for resistance of glioblastoma cells to radiation-induced apoptosis. Exp Mol Med 50, e434
    Pubmed KoreaMed CrossRef
  49. Morris JR and Solomon E (2004) BRCA1 : BARD1 induces the formation of conjugated ubiquitin structures, dependent on K6 of ubiquitin, in cells during DNA replication and repair. Hum Mol Genet 13, 807-817
    Pubmed CrossRef
  50. Pinato S, Scandiuzzi C, Arnaudo N, Citterio E, Gaudino G and Penengo L (2009) RNF168, a new RING finger, MIU-containing protein that modifies chromatin by ubiquitination of histones H2A and H2AX. BMC Mol Biol 10, 55
    Pubmed KoreaMed CrossRef
  51. Gatti M, Pinato S, Maiolica A et al (2015) RNF168 promotes noncanonical K27 ubiquitination to signal DNA damage. Cell Rep 10, 226-238
    Pubmed CrossRef
  52. Chroma K, Mistrik M, Moudry P et al (2017) Tumors overexpressing RNF168 show altered DNA repair and responses to genotoxic treatments, genomic instability and resistance to proteotoxic stress. Oncogene 36, 2405-2422
    Pubmed CrossRef
  53. Patel PS, Abraham KJ, Guturi KKN et al (2021) RNF168 regulates R-loop resolution and genomic stability in BRCA1/2-deficient tumors. J Clin Invest 131, e140105
    Pubmed KoreaMed CrossRef
  54. Panier S, Ichijima Y, Fradet-Turcotte A et al (2012) Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks. Mol Cell 47, 383-395
    Pubmed CrossRef
  55. Kitevski-LeBlanc J, Fradet-Turcotte A, Kukic P 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
    Pubmed KoreaMed CrossRef
  56. Stewart GS, Stankovic T, Byrd PJ et al (2007) RIDDLE immunodeficiency syndrome is linked to defects in 53BP1-mediated DNA damage signaling. Proc Natl Acad Sci U S A 104, 16910-16915
    Pubmed KoreaMed CrossRef
  57. An L, Jiang Y, Ng HHW et al (2017) Dual-utility NLS drives RNF169-dependent DNA damage responses. Proc Natl Acad Sci U S A 114, E2872-E2881
    Pubmed KoreaMed CrossRef
  58. Bai C, Sen P, Hofmann K et al (1996) SKP1 connects cell cycle regulators to the ubiquitin proteolysis machinery through a novel motif, the F-box. Cell 86, 263-274
    Pubmed CrossRef
  59. Craig KL and Tyers M (1999) The F-box: a new motif for ubiquitin dependent proteolysis in cell cycle regulation and signal transduction. Prog Biophys Mol Biol 72, 299-328
    Pubmed CrossRef
  60. Wu J, Zhang X, Zhang L et al (2012) Skp2 E3 ligase integrates ATM activation and homologous recombination repair by ubiquitinating NBS1. Mol Cell 46, 351-361
    Pubmed KoreaMed CrossRef
  61. Li C, Du L, Ren Y et al (2019) SKP2 promotes breast cancer tumorigenesis and radiation tolerance through PDCD4 ubiquitination. J Exp Clin Cancer Res 38, 76
    Pubmed KoreaMed CrossRef
  62. Nakayama KI and Nakayama K (2006) Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Cancer 6, 369-381
    Pubmed CrossRef
  63. Welcker M and Clurman BE (2008) FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation. Nat Rev Cancer 8, 83-93
    Pubmed CrossRef
  64. Zhang Q, Karnak D, Tan M, Lawrence TS, Morgan MA and Sun Y (2016) FBXW7 facilitates nonhomologous end-joining via K63-linked polyubiquitylation of XRCC4. Mol Cell 61, 419-433
    Pubmed KoreaMed CrossRef
  65. Matsuoka S, Oike Y, Onoyama I et al (2008) Fbxw7 acts as a critical fail-safe against premature loss of hematopoietic stem cells and development of T-ALL. Genes Dev 22, 986-991
    Pubmed KoreaMed CrossRef
  66. Thompson BJ, Jankovic V, Gao J et al (2008) Control of hematopoietic stem cell quiescence by the E3 ubiquitin ligase Fbw7. J Exp Med 205, 1395-1408
    Pubmed KoreaMed CrossRef
  67. Reavie L, Della Gatta G, Crusio K et al (2010) Regulation of hematopoietic stem cell differentiation by a single ubiquitin ligase-substrate complex. Nat Immunol 11, 207-215
    Pubmed KoreaMed CrossRef
  68. Siu KT, Xu Y, Swartz KL et al (2014) Chromosome instability underlies hematopoietic stem cell dysfunction and lymphoid neoplasia associated with impaired Fbw7-mediated cyclin E regulation. Mol Cell Biol 34, 3244-3258
    Pubmed KoreaMed CrossRef
  69. Loeb KR, Kostner H, Firpo E et al (2005) A mouse model for cyclin E-dependent genetic instability and tumorigenesis. Cancer Cell 8, 35-47
    Pubmed CrossRef
  70. Minella AC, Loeb KR, Knecht A et al (2008) Cyclin E phosphorylation regulates cell proliferation in hematopoietic and epithelial lineages in vivo. Genes Dev 22, 1677-1689
    Pubmed KoreaMed CrossRef
  71. Cubillos-Rojas M, Amair-Pinedo F, Peiró-Jordán R, Bartrons R, Ventura F and Rosa JL (2014) The E3 ubiquitin protein ligase HERC2 modulates the activity of tumor protein p53 by regulating its oligomerization. J Biol Chem 289, 14782-14795
    Pubmed KoreaMed CrossRef
  72. García-Cano J, Sánchez-Tena S, Sala-Gaston J et al (2020) Regulation of the MDM2-p53 pathway by the ubiquitin ligase HERC2. Mol Oncol 14, 69-86
    Pubmed KoreaMed CrossRef
  73. Bekker-Jensen S and Mailand N (2010) Assembly and function of DNA double-strand break repair foci in mammalian cells. DNA Repair (Amst) 9, 1219-1228
    Pubmed CrossRef
  74. Bonanno L, Costa C, Majem M et al (2016) Combinatory effect of BRCA1 and HERC2 expression on outcome in advanced non-small-cell lung cancer. BMC Cancer 16, 312
    Pubmed KoreaMed CrossRef
  75. Yuan J, Luo K, Deng M et al (2014) HERC2-USP20 axis regulates DNA damage checkpoint through Claspin. Nucleic Acids Res 42, 13110-13121
    Pubmed KoreaMed CrossRef
  76. Laino AM, Berry EG, Jagirdar K et al (2018) Iris pigmented lesions as a marker of cutaneous melanoma risk: an Australian case-control study. Br J Dermatol 178, 1119-1127
    Pubmed CrossRef
  77. Johansson P, Klein-Hitpass L, Choidas A et al (2018) SAMHD1 is recurrently mutated in T-cell prolymphocytic leukemia. Blood Cancer J 8, 11
    Pubmed KoreaMed CrossRef
  78. Suh YJ, Choe JY and Park HJ (2017) Malignancy in pheochromocytoma or paraganglioma: integrative analysis of 176 cases in TCGA. Endocr Pathol 28, 159-164
    Pubmed CrossRef
  79. Hopkins BD, Hodakoski C, Barrows D, Mense SM and Parsons RE (2014) PTEN function: the long and the short of it. Trends Biochem Sci 39, 183-190
    Pubmed KoreaMed CrossRef
  80. Ma S and Kosorok MR (2010) Detection of gene pathways with predictive power for breast cancer prognosis. BMC Bioinformatics 11, 1
    Pubmed KoreaMed CrossRef
  81. Subik K, Shu L, Wu C et al (2012) The ubiquitin E3 ligase WWP1 decreases CXCL12-mediated MDA231 breast cancer cell migration and bone metastasis. Bone 50, 813-823
    Pubmed KoreaMed CrossRef
  82. Xu SQ, Qin Y, Pan DB et al (2016) Inhibition of WWP2 suppresses proliferation, and induces G1 cell cycle arrest and apoptosis in liver cancer cells. Mol Med Rep 13, 2261-2266
    Pubmed CrossRef
  83. Qin Y, Wang CJ, Ye HL et al (2022) WWP2 overexpression inhibits the antitumor effects of doxorubicin in hepatocellular carcinoma. Cell Biol Int 46, 1682-1692
    Pubmed CrossRef
  84. Fang X, Huang Z, Zhai K et al (2021) Inhibiting DNA-PK induces glioma stem cell differentiation and sensitizes glioblastoma to radiation in mice. Sci Transl Med 13, eabc7275
    Pubmed CrossRef
  85. Kolas NK, Chapman JR, Nakada S et al (2007) Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science 318, 1637-1640
    Pubmed KoreaMed CrossRef
  86. Thorslund T, Ripplinger A, Hoffmann S et al (2015) Histone H1 couples initiation and amplification of ubiquitin signalling after DNA damage. Nature 527, 389-393
    Pubmed CrossRef
  87. Polanowska J, Martin JS, Garcia-Muse T, Petalcorin MI and Boulton SJ (2006) A conserved pathway to activate BRCA1-dependent ubiquitylation at DNA damage sites. Embo J 25, 2178-2188
    Pubmed KoreaMed CrossRef
  88. Lee NS, Chang HR, Kim S et al (2018) Ring finger protein 126 (RNF126) suppresses ionizing radiation-induced p53-binding protein 1 (53BP1) focus formation. J Biol Chem 293, 588-598
    Pubmed KoreaMed CrossRef
  89. Schmidt CK, Galanty Y, Sczaniecka-Clift M et al (2015) Systematic E2 screening reveals a UBE2D-RNF138-CtIP axis promoting DNA repair. Nat Cell Biol 17, 1458-1470
    Pubmed KoreaMed CrossRef
  90. Mattiroli F, Vissers JH, van Dijk WJ et al (2012) RNF168 ubiquitinates K13-15 on H2A/H2AX to drive DNA damage signaling. Cell 150, 1182-1195
    Pubmed CrossRef
  91. Chen J, Feng W, Jiang J, Deng Y and Huen MSY (2012) Ring finger protein RNF169 antagonizes the ubiquitin-dependent signaling cascade at sites of DNA damage. J Biol Chem 287, 27715-27722
    Pubmed KoreaMed CrossRef
  92. 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
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Author ORCID Information

Funding Information
  • National Research Foundation of Korea
      10.13039/501100003725
      NRF-2022R1H1A2091883, NRF-2019R1A2C1007197
  • Ministry of Science and ICT, South Korea
      10.13039/501100014188
      NRF-2022R1H1A2091883, NRF-2019R1A2C1007197

Collections

Services
Social Network Service

e-submission

Archives