53BP1: A key player of DNA damage response with critical functions in cancer

53BP1: A key player of DNA damage response with critical functions in cancer

Accepted Manuscript Title: 53BP1: A key player of DNA damage response with critical functions in cancer Authors: Mohammad Mirza-Aghazadeh-Attari, Amir...

906KB Sizes 0 Downloads 15 Views

Accepted Manuscript Title: 53BP1: A key player of DNA damage response with critical functions in cancer Authors: Mohammad Mirza-Aghazadeh-Attari, Amir Mohammadzadeh, Bahman Yousefi, Ainaz Mihanfar, Ansar Karimian, Maryam Majidinia PII: DOI: Reference:

S1568-7864(18)30260-X https://doi.org/10.1016/j.dnarep.2018.11.008 DNAREP 2562

To appear in:

DNA Repair

Received date: Revised date: Accepted date:

13 October 2018 18 November 2018 19 November 2018

Please cite this article as: Mirza-Aghazadeh-Attari M, Mohammadzadeh A, Yousefi B, Mihanfar A, Karimian A, Majidinia M, 53BP1: A key player of DNA damage response with critical functions in cancer, DNA Repair (2018), https://doi.org/10.1016/j.dnarep.2018.11.008 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

53BP1: A key player of DNA damage response with critical functions in cancer Mohammad Mirza-Aghazadeh-Attaria,b, Amir Mohammadzadeha,b, Bahman Yousefic,d, Ainaz

a

Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran;

b

c

SC RI PT

Mihanfare, Ansar Karimianf, Maryam Majidiniag,*

Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran;

Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran;

U

d

e

N

Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences,

A

Urmia, Iran; f

of Medical Sciences, Babol, Iran; g

M

Cellular and Molecular Biology Research Center, Health Research Institute, Babol University

TE

D

Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.

*Corresponding authors:

A

CC

EP

Maryam Majidinia, Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran. Tel: (+98)9033916051. Email: [email protected]

Abstract

Maintenance of genome integrity and stability is a critical responsibility of the DNA damage response (DDR) within cells, such that any disruption in this kinase-based signaling pathway leads to development of various disorders, particularly cancer. The tumor suppressor P53-binding

SC RI PT

protein 1 (53BP1), as one of the main mediators of DDR, plays a pivotal role in orchestrating the choice of double-strand break (DSB) repair pathway and contains interaction surfaces for numerous DSB-responsive proteins. It has been extensively demonstrated that aberrant expression of 53BP1 contributes to tumor occurrence and development. 53BP1 loss of function in tumor tissues is also related to tumor progression and poor prognosis in human malignancies. Due to

U

undeniable importance of this protein in various aspects of cancer initiation/progression,

N

angiogenesis, metastasis and development of drug resistance, as well as its targeting in the

A

treatment of cancer, this review focused on explaining the structure and function of 53BP1 and its

M

contribution to cancer.

A

CC

EP

TE

D

Keywords: 53BP1; DNA damage response; DNA repair; Double strand breaks; Cancer

1. Introduction The tumor suppressor P53-binding protein 1 (53BP1) is a member of Tudor-containing proteins (including JMJD2A, 53BP1, SGF29, Spindlin1, UHRF1, PHF1, PHF19 and SHH1) in ‘reading’

SC RI PT

unique methylation events on histones in order to facilitate DNA damage repair or regulate transcription (1). This protein is also one of the mediator/adaptor proteins of the DNA damage response (DDR), which includes the mediator of the DNA damage checkpoint protein 1 (MDC1), 53BP1, breast cancer-associated gene 1 (BRCA1), topoisomerase II-binding protein 1 (TOPBP1), Claspin and Pax transactivation domain-interacting protein (PTIP) (2). 53BP1 was first described

U

as a binding partner of the p53, almost 25 years ago (3). 53BP1 is a key component of DDR and

N

plays a pivotal role in orchestrating the choice of double-strand break (DSB) repair pathway; it

A

contains interaction surfaces for numerous DSB-responsive proteins (4, 5). This protein has no

M

characterized enzymatic activity and acts as a recruitment platform for other DDR proteins (2, 6).

D

Access of 53BP1 to DSB is modulated by the Tudor-interacting repair regulator (TIRR) through

TE

masking the dimethylated lysine 20 of the histone H4 (H4K20me2) binding surface on 53BP1 (7). 53BP1 promotes non-homologous end-joining (NHEJ)-mediated DSB repair and prevents

EP

homologous recombination (HR) by counteracting the function of BRCA1 in the HR pathway (8).

CC

Interestingly, promotion of microhomology-mediated end-joining (MMEJ) by 53BP1 in G1-phase cells is observed in the presence of the functional BRCA1 (9). A defect in 53BP1 induces DNA

A

damage checkpoint defects, impaired DNA repair and hypersensitivity to ionizing radiation (IR) (10, 11). Moreover, depletion of 53BP1 results in a cell cycle arrest in the G2/M phase and instability in human cells (12). 53BP1 is also essential for the adaptive immune system because of its role in NHEJ of distal DNA ends generated during long-range V(D)J recombination and class switch recombination (CSR), which are important for a functional adaptive immune response (13).

Various studies have shown the role of this protein in various cellular functions. For example, aberrant expression of 53BP1 was found to contribute to tumor occurrence and development. It was revealed that 53BP1 loss in tumor tissues was related with tumor progression and poor

SC RI PT

prognosis in breast cancer, and expression of 53BP1 was correlated with apoptosis in colorectal cancer (14) (15). Furthermore, it has been recently observed that 53BP1 is involved in regulating mitochondrial clearance from the cell via a type of autophagy termed mitophagy (16). In addition to the role of this molecule in the field of cancer, its function in the aging process can be highlighted by its association with lamin A/C (17). Regarding the structure and function of this protein, various

U

studies have been published so far; however, the role of this molecule in various cancers has less

N

been mentioned. Therefore, the present review focused on various roles of this protein in different

A

aspects of cancer including initiation/progression, angiogenesis, metastasis and development of

M

drug resistance.

The protein structure

TE

2.1.

D

2. The structure, genome, and transcription of 53BP1

EP

53BP1 is a large protein with 1972 amino acids and over 200 kDa mass that is encoded by the TP53BP1 gene (18) (19). Important structural elements in 53BP1 include two repeats of BRCA1

CC

carboxy-terminal (BRCT), the tandem Tudor domains, a glycine/arginine-rich region (GAR) methylation stretch, two dynein 8 kD light chain (LC8) binding sites, and numerous

A

phosphorylation sites including 32 PIK kinases and 41 cyclin-dependent (CDK) phosphorylation sites (called amino-terminal Ser/Thr-Gln (S/TQ) sites and Ser/Thr-Pro (S/TP) sites, respectively (20)), which are phosphorylated, at least in part, by the ataxia-telangiectasia mutated (ATM) kinase (18). 53BP1 was originally identified in a yeast two-hybrid screen as a protein that interacted with

the p53 DNA binding domain through its two C-terminal BRCT motifs (21). BRCT domains are 100–150 residue motifs found in a large number of proteins involved in the cellular responses to DNA damages (22, 23). GAR of 53BP1 is methylated by the protein arginine N-methyltransferase

SC RI PT

1 (PRMT1) (24, 25) and has been implicated in a possible DNA-binding function for 53BP1 (24). Some of S/TQ sites have been identified as phosphatidylinositol 3-kinase-related kinase (PIKK)dependent phosphorylation sites following ionizing radiation (IR) (26-28). Some of these sites have characterized roles in DDR signaling and protein interactions. For example, phosphorylation of 53BP1 at Ser25 is required for the interaction between 53BP1 and mediator PTIP, and abrogation

U

of this interaction results in DNA-damage-sensitivity and reduced checkpoint kinase 2 (CHK2)

N

phosphorylation (29). Additionally, phosphorylation of 53BP1 at Ser1219 has been reported to

A

function in DNA damage signaling and G2/M-phase arrest following IR (28). 53BP1 exhibits

M

strong functional similarity to MDC1 (30), BRCA1 (31) and their yeast orthologues (Rad9 in Saccharomyces cerevisiae and Crb2 in Schizosaccharomyces pombe) (22, 32).

D

The gene structure

TE

2.2.

The TP53BP1 gene is on the chromosome 15 and its location is 15q15.3 with 32 exons (33).

EP

Expression of this gene in the brain is more than that of other tissues (34). Homologs of this gene are conserved in chimpanzees, Rhesus monkeys, dogs, cows, mice, rats, chickens, and frogs; a

CC

number of 239 organisms have orthologs with the human gene TP53BP1 (19, 35, 36).

Regulation and post-translational modification

A

2.3.

53BP1 is continuously expressed in the cell nucleus (37) and accumulated in the nucleoplasm. 53BP1 has two subpopulations after DNA damage: the bulk 53BP1 in the nucleoplasm and 53BP1 bound at the damage site (38, 39). Since the bulk 53BP1 is not bound to chromatin epitopes at the damage site, it would be susceptible to ubiquitination and degradation. Bulk 53BP1 prevents the

RAP1-interacting factor 1 (RIF1) from binding to the DNA in the undamaged state. After IR, bulk 53BP1 is degraded, and RIF1 is recruited to damage sites by the bound 53BP1 to execute inhibition of end resection (40-42). RIF1 is one of a few identified proteins which requires 53BP1 for its

SC RI PT

recruitment to DSBs (43) and is involved in C-NHEJ (42). The protein level of 53BP1 at different stages of the cell division cycle is relatively stable with only minor oscillations (44, 45), suggesting that 53BP1 is being constantly transcribed and translated during the cell cycle. On the other hand, a number of studies have reported the post-translational regulation of 53BP1.

Three pathways regulate the cellular levels of 53BP1: an endosomal/lysosomal protease Cathepsin

U

L (CTSL) that cleaves 53BP1 (46-48), UbcH7 that regulates the proteasome-dependent

N

degradation of 53BP1 (49) and lamin A/C, the depletion of which reduces 53BP1 stability (50).

A

Pozo et al. recently showed that UbcH7-dependent proteasomal degradation is the major pathway

M

that controls the level of 53BP1 in the absence or presence of DNA damage in a wide range of cultured human cell lines (51). The mechanism by which lamin A/C protects 53BP1 is shielding it

D

from the UbcH7-dependent degradation machinery (50, 51).

TE

53BP1 is a rare example of a protein whose post-translational modification-binding function can

EP

be switched on and off. Activity of 53BP1 is directly inhibited by 53BP1-binding protein TIRR (52, 53). X-ray crystal structures of TIRR and a designer protein bound to 53BP1 reveal a unique

CC

regulatory mechanism, in which an intricate binding area centered on an essential TIRR arginine residue blocks the methylated-chromatin-binding surface of 53BP1. This 53BP1 inhibition is

A

relieved by TIRR-interacting RNA molecules, providing the proof-of-principle of the RNAtriggered 53BP1 recruitment to DSBs (54). Furthermore, 53BP1 interacts with the structural protein NuMA, which controls 53BP1 diffusion. This interaction between the two proteins is reduced after DNA damage. NuMA prevents 53BP1 accumulation at DNA breaks. Manipulating

NuMA expression alters the PARP inhibitor sensitivity of BRCA1-null cells, end-joining activity, and immunoglobulin class switching that rely on 53BP1(55). There are several mechanisms that limit the accumulation of 53BP1 at sites of DNA damage by

SC RI PT

preventing self-perpetuating activation of DNA damage checkpoints and excessive spreading of DNA repair factors to undamaged chromatin. Several de-ubiquitylating enzymes (DUBs) have been found to counteract DNA damage-dependent histone ubiquitylation and the assembly of 53BP1 at sites of DNA damage. These include BRCC36, USP3, POH1, and USP44 (56-59). Another DUB, called OTUB1, restricts histone ubiquitylation and 53BP1 loading, but in a non-

U

catalytic manner that involves binding to UBC13 and hindering the ubiquitin ligase activity of

N

RNF8 and RNF168 (60, 61). USP16 and USP28 have also been shown to modulate DDR and

A

repair pathways. USP16 mediates histone deubiquitylation-dependent transcription silencing at

M

DSB sites (62), and USP28 possibly regulates 53BP1 stability (59, 63). In addition, histone acetylation is thought to play a part in inhibiting 53BP1 chromatin binding. The acetyltransferase

D

Tip60/Kat5 can acetylate histone H4 on lysine 16, which interferes with the binding of 53BP1 to

TE

the nearby H4K20Me2 (64, 65). Importantly, the H4K16 acetylation concomitantly increases the

EP

BRCA1 recruitment to DNA damage foci; moreover, the knockdown of Tip60/Kat5 or chemical inhibition of histone deacetylases (HDACs) can rescue the homology-directed repair (HDR) defect

CC

of BRCA1-deficient cells, similar to the absence of 53BP1 (59, 64, 66, 67). After detection of DSBs, 53BP1 rapidly accumulates on the chromatin surrounding the break site

A

or at ionizing radiation-induced foci (IRIF) after IR-induced DNA damage (37, 59), because a cascade of protein modification and relocalization is triggered: this signaling cascade is initiated by the ATM-mediated phosphorylation of the histone 2A (H2A) variant H2AX (γH2AX), followed by the recruitment of MDC1 and activation of RNF8 (ring finger 8)-RNF168-dependent chromatin

ubiquitylation (68-70). Phosphorylation of H2AX (γH2AX) results in the recruitment of downstream factors, such as the E3 ubiquitin ligases RNF8 and RNF168, leading to the formation of K63-linked polyubiquitin chains on histones at DSBs (71, 72). This ubiquitination cascade

SC RI PT

regulated by RNF8 and RNF168 is responsible for the localization of repair mediators (73, 74), including BRCA1 and 53BP1, to DNA damage sites (73, 75). Although the initial recruitment of 53BP1 occurs independently of γH2AX (76). The stable association of 53BP1 with DSBs strongly relies on the RNF8-RNF168-mediated ubiquitylation cascade (73, 77), that is the activated downstream of γH2AX and MDC1 (78, 79). Yiheng et al. found that RNF8 and RNF168 not only

U

mark histones at the break site to create a 53BP1 binding site, but these ubiquitin ligases also

N

regulate the proteasome-mediated degradation of 53BP1 (80). Failure to degrade 53BP1 not bound

A

to DSBs leads to mislocalization of a downstream factor RIF1, thereby impairing DSB repair (80,

M

81). Interestingly, the C-terminal BRCT repeats of 53BP1, which mediate the interactions between p53 and EXPAND1, are dispensable for the focal recruitment of 53BP1 to damaged chromatin

D

(82-84). The chromatin modulator EXPAND1 (also known as MUM1) is one of the two proteins

TE

that rely on 53BP1 for their accumulation at DSB sites, which interacts with the BRCT domains

EP

in 53BP1 in a phosphorylation-independent manner (85). The second protein is RIF1, which interacts (directly or indirectly) with ATM-phosphorylated residues in 53BP1 (86). Instead of the

CC

C-terminal BRCT repeats, the minimal focus-forming region of 53BP1 contains an oligomerization motif and tandem Tudor domains that bind to mono and dimethylated H4K20

A

(H4K20me1 and H4K20me2, respectively) as well as an adjacent ubiquitylation-dependent recruitment (UDR) motif that binds to the RNF168-ubiquitylated H2AK15 (36, 87, 88). The domain structure of this minimal focus-forming region in 53BP1 reflects the need to integrate

multiple, independent histone marks that cooperate to recruit 53BP1, specifically to damaged chromatin (19).

SC RI PT

3. DNA damage response and 53BP1 DDR is a plethora of molecules active in a signaling cascade, where DNA damage is sensed, and molecules are translated to actions such as apoptosis, cell cycle arrest, senescence and most importantly, DNA repair via a specific transducer and mediator (89-91). DNA repair is classified to HR, mismatch repair, nucleotide excision repair, base excision repair and NHEJ (92, 93). It has

U

been shown that alterations in the normal function of DDR can lead to defects in DNA repair or

N

disruption of apoptosis or cellular arrest, which could be the starting point for neoplastic cell

A

formation (91), or other human pathologies, aging and hereditary disorders (94, 95). 53BP1, is a

M

molecule active as a DNA damage sensor and damage mediator, which has important roles in signaling of DDR transducers and mediators in the DSB arm, such as ATM, BRCA1 and p53 (96).

Interactions with molecules involved in DDR

EP

3.1.

TE

the cascade.

D

Figure 1 further elicits the role of 53BP1 in DDR and its interactions with multiple molecules in

CC

3.1.1. Interactions with DDR sensors The Mre11/Rad50/Nbs1 (MRN) is the DNA damage sensor in the DSB arm of DDR, and has

A

critical roles in activating downstream kinases and DDR transducers, namely ATM (97). A study by Lee et al. revealed that the interaction between the tandem breast cancer carboxy-terminal repeats of 53BP1 and the MRN complex increased the rate of ATM activation by phosphorylation and the subsequent downstream activation of molecules such as CHK2. Their study also showed that the interaction was facilitated by RAD50 from the complex, and led to the hyper

phosphorylation of Nbs1, and 53BP1 itself. Furthermore, their study indicated that the combined function of 53BP1, MRN complex and ATM was necessary for optimum DDR and DSB repair (98). The RPA complex is a DNA damage sensor in the single strand damage pathway of DDR. A

SC RI PT

study by Yoo et al. showed that there were important interactions between this complex and 53BP1. The researchers found that 53BP1 was involved in the phosphorylation of RPA2 following DNA damage. This was found by inducing DNA damage in U2OS osteosarcoma cells, in which 53BP1 was knockdown by siRNAs. These cells had an augmented apoptosis with camptothecin and showed increased sensitivity (99).

U

3.1.2. Interactions with DDR transducers

N

ATM is the main transducer of the DSB signal in the DDR cascade, and is activated by the presence

A

of DSB. This activation is done in two manners, one is dependent on the activation of the MRN

M

complex and the other is dependent on the change in the chromatins structure, which is mediated

D

by 53BP1 (100). A study by Wilson et al. showed that 53BP1 mediates the ATM

TE

autophosphorylation in ATM S1981, a role which is unique among proteins with BRCT domains. 53BP1 is also able to induce T68 phosphorylation in CHK2, the downstream molecule in ATM

EP

signaling (31). Further, 53BP1 is phosphorylated by ATM in multiple residues, and by ATR in instances of UV-caused DNA damage (27). A study by Shibata et al. showed another relation

CC

between 53BP1 and the two transducers of DNA damage, the ATM and ATR. It was shown that

A

cells needed the signaling of both ATM and ATR in order to maintain G2/M checkpoint arrest, and in high doses of exposure to radiation which triggered cell arrest, there was a need for the function of 53BP1 and MDC1. The authors concluded that lack of 53BP1 enhanced chromosome breakage in response to radiation (101). ATR is the equivalent of ATM in single strand DNA damage signaling, which exerts its role by affecting CHK1, a downstream transducer. Studies have

shown that 53BP1 mediates the interaction between CHK1 and ATR in instances of replication stress, which leads to the protection of replication forks. An absence of 53BP1 causes a defective ATR-Chk1-p53 signaling, leading to increased rates of apoptosis in ex vivo B cells (102).

SC RI PT

Interestingly, a study by Martinez et al. showed that a double lack of 53BP1 and TRF1, which protects telomeres from damage and fusion, caused an increased ATR function and CHK1 phosphorylation coupled with an increased HR (103). 3.1.3. Interactions with BRCA-1

Studies have shown that the effect of 53BP1 and BRCA-1 on each other has important outcomes

U

regarding DDR signaling, namely DNA repair. A study by Xu et al. revealed that these two

N

molecules governed the entrance into two distinct pathways for stalled replication restart. 53BP1

A

promotes a fork cleavage-free pathway while BRCA-1 coupled with SLX-MUS promotes a break-

M

induced replication (BIR) pathway (104). Further, these two molecules antagonize each other in

D

DSB repair pathway selection. BRCA-1 promotes HR by co-localizing with H2AX to the site of

TE

damage and negatively regulating Mre11, a molecule which is part of the MRN complex and possesses nuclease activity. BRCA-1 also has profound roles in the transcriptional response to

EP

DNA damage, which further contribute to the HR progression (105). Further, BRCA-1 antagonizes BP531 by limiting its interaction to the chromatin in the S phase, thereby inhibiting 53BP1

CC

mediated repair (106). Regarding this, another study found that BRCA-1 was able to stop the

A

translocation of RIF1 to breaks in the S2/G phase. This study also found that BRCA-1 needed its two most important domains, i.e. the RING and BRCT domains, to regulate the function of 53BP1 (107).

3.1.4. Interactions with p53 p53 is the centerpiece in DDR and activated downstream signaling cascades, which determines the final fate of cells undergoing DNA damage (108). Studies by Cuella-Martin et al. showed that p53

SC RI PT

had rigorous transcriptional activities, which were dependent on 53BP1 function. It was revealed that the transcription of molecules such as p21 and MDM2 was dependent on the function of p53, which, itself, was dependent on sufficient 53bp1 activation (109, 110). The same team also uncovered that the interaction between the two molecules was mediated by the BRCT and Nterminal domains of 53BP1, and also by a deubiquitylase called USP28 (109, 111, 112). Al Rashid

U

et al. showed that the non-specific DNA binding domain of p53 in the C terminal of the molecule

N

was responsible for these interactions (113). Interestingly, this function of 53BP1 is shown to be

A

independent of the role of 53BP1 in DNA repair (114). Further evidence regarding the concomitant

M

action of these two molecules is shown in a study by Morales et al. They found that animals with deficiencies in the function of both p53 and 53bp1 showed increased rates of T cell lymphomas

D

and increased genomic instabilities in solid tumors compared to animals defective in only one of

TE

these molecules (115). Another study conducted by Ward et al. revealed similar results. Further,

EP

they suggested a 53-indipendent role for 53BP1 in antagonizing carcinogenic events (116). Another functional significance of 53BP1 in p53 action has been shown regarding cell cycle arrest.

CC

A study by Fong CS et al. showed that 53BP1 and USP28 had essential roles in mediating a p53 dependent cell cycle arrest in G1 in response to prolonged mitosis and loss of centrosome (117).

A

Similar results were also obtained by Meitinger et al. (118) and Lambrus et al. (119). 3.1.5. Interactions with DNA repair As mentioned, DNA repair is one of the end points of DDR. 53BP1 is active in regulating DSBs damage, and its most important function is to promote NHEJ and suppress HR (120). In performing

this function, 53BP1 has close ties with BRCA-1, as mentioned previously. The initial step in initiation of repair is the recruitment of 53BP1 to the damage cite via interactions with MDC1, which is then sustained in the region by interactions with H4K20me2. Studies have also shown

SC RI PT

that RNF8 and RNF168 are necessary for recruitment (58, 87, 121). The main involvement of 53BP1 in DNA repair is the choice between NHEJ and HR. These two processes of DNA repair differ in the fact that HR is an error free process while NHEJ is not; however, NHEJ is shown to be a faster alternative to HR in DNA repair (122). After ATM phosphorylates 53BP1, a series of effector molecules such as RIF1 and PTIP prevent resection of DNA 5′ ends and promote NHEJ

N

3.1.6. Interactions with apoptosis and autophagy

U

(123). Further, 53BP1 itself inhibits end resection dependent on CtIP (59, 66).

A

Apoptosis is defined as the programmed death of cells. DDR cascade has essential roles in the

M

progression of apoptosis. Mutations in DDR can translate into functional variations in the process

D

of apoptosis, which enable cells with compromised DNA content to avoid death and possibly

TE

acquire malignant characteristics (124). Evidence regarding the role of 53BP1 in apoptosis has been shown in C. elegans. A study by Ryu et al. analyzed the function of HSR-9, the homolog of

EP

53BP1, after exposure to ionizing radiation and found that HSR-9 promoted apoptosis and a form of the RAD-54 dependent NHEJ. This molecule has no independent role in cell cycle arrest (125).

CC

Hong et al. found that increased expression of 53BP1 in ovarian cancer cells increased the

A

expression of molecules such as Bax, P21 and caspase 3 but decreased that of BCL-2 and AKT, having a net effect of promoting apoptosis (126). Another set of observations revealed that inhibiting the function of 53BP1 could increase apoptosis in cells undergoing DNA damage inducing agents. Yang et al. found that utilizing a glycogen synthesis kinase 3 (GSK3) β (which

phosphorylates and activates 53BP1) inhibitor called SB216763 would render glioblastoma cells more sensitive to apoptosis (127). Autophagy is defined as the clearance of unneeded cellular particles under conditions of stress.

SC RI PT

This function has multiple cross talks with DDR and its end points (128). A study by Youn et al. showed that 53BP1 had p53 independent roles in promoting mitophagy, a subtype of autophagy in H1299 and HeLa cells. They study found that cells with absent 53BP1 had mitochondria with abnormal shapes and mitochondrial aggregation coupled with an increased mitochondrial

U

membrane potential (129).

N

3.1.7. Interactions with cell cycle

A

As mentioned, 53BP1 interacts with p53 and USP28 to contribute to cell cycle arrest. However,

M

53BP1 also has other roles in controlling cell cycle. It was shown that 53BP1 was a target of cell cycle kinases, such as Polo-like kinase-1, which interacted with 53BP1 and led to a successful

D

inactivation of the G(2)/M checkpoint (44). A study by Stiff et al. suggested that ATR enforced

TE

the G2/M arrest by the mediating effects of 53BP1, MDC1 and Nbs1 (130). Regarding the ATR pathway in DNA damage, TopBP1 is a mediator for the effects of 53BP1 in the G1, as shown by

EP

Cescutti et al. (131). A study by Kwak et al. demonstrated that the role of 53BP1 in upholding the

CC

mitotic checkpoint was dependent on the presence of the Jun activation domain-binding protein 1 (Jab1), which is a 53BP1 binding protein. They found in their study that the knock-down of Jab1

A

resulted in reduced mitotic arrest in resistance to chemotherapy. It was hypostatized that this particular interaction could be a target of therapies aiming to sensitize cells to microtubuleinterfering anticancer drugs (132). In addition, a study by Sengupta et al. showed that 53BP1 had important functional interactions with the BLM helicase in upholding the S phase checkpoint. It

was shown that BLM was essential for 53BP1 and p53 interaction and that ATR/CHK1 was responsible for mediating the initial interaction between the BLM helicase and 53BP1 (133, 134).

SC RI PT

4. 53BP1 interaction with signaling pathways Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling: NF-κB signaling is one of the most important signaling pathways regarding human malignancies (135). It has been shown that NF-κB signaling induces cellular proliferation, angiogenesis, epithelialmesenchymal transition, metastasis and alterations in tumor microenvironment, which further

U

promote cancer formation (136). DNA damage is one of the activating agents of NF-κB, which

N

together with cell cycle proteins, initiates events that insure cell survival. This series of events is

A

one of the reasons why tumors gain the ability to tolerate chemotherapy agents (137). The

M

regulatory role of DDR on NF-κB signaling as well as the role of 53BP1 in this relation was shown in a study by Li et al. where 53BP1 activation inhibited NF-κB signaling, which led to decreased

D

rates of tumor progression and metastasis. This regulatory effect (138) was imposed by miR-146a

TE

(139). Moreover, it has been proposed that NF-κB can have regulatory effects on DDR. A study

EP

showed that this signaling pathway could interact with ATM and BRCA-1 in promoting HR by promoting BRCA-1 stabilization and interaction with CtIP (140). Another study revealed that NF-

CC

κB mediated the expression of Ku70 and Ku80, the proteins involved in NHEJ (141). More evidence regarding the role of NF-κB in DNA repair was proposed by Kraft et al. who found that

A

inhibition of NF-κB negatively affected both NHEJ and HR, and that this effect was cell specific and also different pattern of DSB repair was observed based on the cell type and content of other DDR molecules such as p53 (142).

AKT signaling: Another cellular signaling pathway with meaningful interactions with DDR and 53BP1 is the AKT signaling cascade (143, 144), which has established roles in human pathologies, specifically cancer (145, 146). A study by Liu et al. revealed that AKT mediated the

SC RI PT

phosphorylation of the X-ray repair cross complementing 4 (XRCC4)-like factor (XLF), leading to its disassociation from the XRCC4/DNA ligase IV complex. This resulted in the impairment of the functional goal of 53BP1and NHEJ. Further, it was shown that a mutation in this cascade caused increased rates of NHEJ in cancer cell (147). 53BP1 also has regulatory roles in AKT, as it was shown that increased expression of 53BP1 was able to significantly reduce the levels of

U

AKT in A2780 and HO-8910PM cells while having anti-cancer effects (126).

A

53BP1 expression in cancer and its effects on clinical characteristics

M

5.1.

N

5. Role of 53BP1 in cancer

Because of the aforementioned roles of 53BP1 in DDR cascade, it is unsurprising that this

D

molecule has important roles in cancers (148). A study by Djuzenova et al. revealed that the levels

TE

of 53BP1 were significantly higher in specimens of rectal carcinoma compared to normal controls

EP

(149). Similar results were shown in skin cancer and cervical cancer specimens (150). Djuzenova et al. also found that lower levels of 53BP1 mRNA were associated with lymph node involvement

CC

and metastasis (151). Other studies have shown that BP531 could be one of the unique mutations in some cancers, as it is a tumor suppressor. A study by Bouwman et al. revealed that loss of 53BP1

A

was a factor contributing to the survival of BRCA-1 deficient breast cells, thereby causing the emergence of triple negative breast cancer cells (67). Squatrito et al. found that 53BP1 was a haploinsufficient tumor suppressor regarding glioma formation, as it was shown to be heterozygously lost in almost 20% of glioblastoma multiforme cells (152). Further evidence

regarding the role of 53BP1 in cancer has emerged in studies examining the relation between the expression of 53BP1 and clinical characteristics of cancer. Jianping et al. found that loss of BP531 was associated with increased tumor size, lower survival and higher stages in colorectal cancers.

SC RI PT

In vitro, silencing 53BP1 resulted in inhibited apoptosis and increased proliferation of HCT-116 cells (153). Neboori et al. found that low 53BP1 was associated with increased local recurrence in breast cancer patients being under therapy with radiotherapy and surgery (14).

5.2.

53BP1 in resistance to treatment and prognosis of treatment

One significance of the status of 53BP1 expression in cancer is its effect on the success of

U

therapies(154). Studies have shown that depletion of 53BP1 causes resistance to agents such as

N

PARP inhibitors, chemotherapy agents and radiation (155, 156). It also predicts outcomes after

A

undergoing treatment. A study by Schouten et al. revealed that BRCA-1 like breast cancer with

M

low levels of 53BP1 and high levels of XIST had an inferior response to treatment with high dose

D

alkylating agents compared to that with high levels of 53BP1 and low levels of XIST. The

TE

expression profile of these two proteins contributed significantly to survival and disease free survival after treatment (157). Bonanno et al. in their study investigated the predictive power of

EP

53BP1 levels in predicting survival in non-small lung cancer cell patients undergoing treatment

CC

with platinum based agents. They found that patients with low levels of 53BP1 had an overall survival of 19.3 months compared to those with high 53BP1 that had an overall survival of only

A

8.2 months (158). Hong et al. found that transfecting ovarian cancer cells, SKOV3, with 53BP1 resulted in decreased migration and proliferation. However, they found that the increase in DNA repair contributed to increased resistance to cisplatin, as the half maximal inhibitory concentration increased to 7.58±0.51 µg/ml from 2.98±0.27 µg/ml (155). Yao et al. in their study evaluated the effect of 53BP1 loss on colorectal cancer cells and found that it led to chemo-resistance towards

5-fluorouracil by inhibiting the ATM-CHK2-P53 pathway. Further, this loss was shown to affect the balance between pro- and anti-apoptotic molecules and reduce the levels of caspases (159). As mentioned before, 53BP1 has direct influence on the function of PARP inhibitors (146, 160).

SC RI PT

Jaspers et al. found that loss of 53BP1 increased resistance to PARP inhibitors, by the reemergence of homologous recombination, in BRCA-1 deficient mouse mammary tumors. They also observed that AZD2461 had the most effect on these cells, compared to other agents (161). Another study showed that 53BP1 loss was the main culprit of the PARP inhibitor resistance in the phosphatase and Tensin homolog (PTEN)-deficient cells such as the glioblastoma cell line

U

U251 (162). Hassan et al. suggested that the gene signature of 53BP1 should be used in order to

N

select patients for the PARP inhibitor therapy (163). A study by Barazas et al. focused on the role

A

of the CTC1-STN1-TEN1 (CST) complex in the PARP inhibitor sensitivity and revealed that loss

M

of each of the members of this complex resulted in increased resistance to PARP inhibitors. The

D

study suggested that this complex could be a downstream signaling complex of 53BP1/RIF1

TE

signaling (164). REV7 is another molecule in the downstream signaling of 53BP1. A study by Xu et al. showed that loss of this molecule, which is also known as MAD2L2, led to the restoration of

EP

HR, via establishing end resection of DSBs which was dependent on CTIP. This molecule is recruited to DSB sites via upstream signaling from the H2AX-MDC1-RNF8-RNF168-53BP1

CC

pathway. As predictable, disruption of this signaling pathway in any step caused increased

A

resistance to PARP inhibitors (165). Regarding the PARP inhibitor sensitivity, it is noteworthy to mention that targeting the upstream signaling of 53BP1 is also an effective method in increasing sensitivity to these agents. For example, a study revealed that disruption of the catalytic activity of ubiquitin-specific peptidase 11 mis-regulated recruitment of 53BP1 and RAD51, causing

increased sensitivity to PARP inhibitors (166). A similar relation was also found between the E3 ubiquitin ligase ring finger protein 168, 53BP1 and PARP inhibitor sensitivity (167).

5.3.

Effect of 53BP1 on angiogenesis

SC RI PT

Formation of new vessels is necessary for tumors to sustain their nutrients and also to proliferate and disseminate through the vascular system. Angiogenesis is dependent on multiple signaling cascades, including HIF-alpha, growth factor and PI3K/AKT signaling. In a study performed by Li X et al., it was shown that 53BP1 had an inverse relation with levels of phosphorylated AKT, which is a regulator of two important pro-angiogenic molecules, MMP9 and MMP2. The

U

knockdown of 53BP1 in MCF-7 cells resulted in increased levels of MMP9 and 2, which was

Targeting 53BP1 in cancer

M

5.4.

A

N

accompanied with increased angiogenesis in vivo and in vitro (168).

Apart from the diagnostic and prognostic significance of 53BP1, some studies have targeted this

D

molecule in cancer in hopes of favorable results. KIM YJ et al. used Glionitrin A, a

TE

diketopiperazine disulfide on prostate cancer cells of DU145. This agent showed increased cell

EP

cycle stoppage and increased apoptosis, coupled by the activation of caspase 8, 9 and 3. Further, tumors being treated with this agent had a decreased volume size compared to control tumor

CC

tissues. Molecular investigations revealed that these effects were mediated by the phosphorylation of 53BP1, which activated DDR transducers such as ATM and ATR and their downstream

A

signaling pathway (169). The significant anti-tumor effects of Glionitrin A were previously discovered; however, this study uncovered the molecular basis for its potency for the first time (170). Another study examined the effect of vitamin D and cathepsin L in regulating 53BP1. The study revealed that vitamin D upregulated while cathepsin L downregulated 53BP1, causing increased and decreased DSB repair, respectively (47). The expression of 53BP1 was also targeted

in a study by Siping et al., where long non coding RNAs were used to decrease the expression of the BRAF-activated noncoding RNA (BANCR), which directly affected the expression of CSE1L, a regulator of 53BP1 and other DNA repair proteins (171). Targeting of 53BP1 could also be done

5.5.

SC RI PT

to shift the balance between error prone NHEJ and error free HR.

Utilizing 53BP1 as a marker for DNA damage in cancer

One interesting use of 53BP1 measurement has been in identifying the effect of different therapy regimens on cancer cells with regard to implementing DNA damage in cells. Uehara et al. compared UVB and UVC in pancreatic cancer cell lines MiaPaCa-2, which were implemented in

U

skin flaps in mice. They used histocultures and confocal imaging to observe 53BP1 foci and found

N

that UVB had a greater penetration, by observing 53BP1 foci in the deeper parts of the tumor

A

(172). A similar study was also performed by Miwa S et al., where it was found that UVC induced

M

DNA damage within 15 minutes, using a live cell assay of 53BP1 coupled with the green

D

fluorescent protein (GFP) (173). 53BP1 was also used as a marker of DNA damage in other cancers

TE

such as cholangiocarcinoma (174). Another use of detecting 53BP1 is in monitoring DNA repair initiation, specifically non-homologous end joining. An example is a study by Maes et al., in which

EP

the combined effect of decitabine and JNJ-26481585 was studied on multiple myeloma cells.

CC

Further, it was shown that RAD51 inhibition led to apoptosis (175). Table 1 summarizes the significance of 53BP1 in various types of cancers not mentioned above.

A

6.

Conclusion

We aimed to illustrate the significance of 53BP1 in DDR and DNA repair and also its role in the process of carcinogenesis. It was discussed that 53BP1 was capable of interacting with multiple DNA damage sensors, transducers and effectors, mainly BRCA1. 53BP1 promoted NHEJ by this

means and suppressed the BRCA1 affiliated HR. Further, significant interactions with molecules such as ATM, ATR and CHK were shown. 53BP1 was also able to determine the fate of the cell by regulating functions such as apoptosis, cell cycle arrest and autophagy. It was observed that

SC RI PT

53BP1 impeded cancer by affecting all the aforementioned functions, and that any aberrancy in its function was capable of promoting neoplasia. Additionally, 53BP1 was observed to be a key molecule in determining the prognosis and clinical characteristics of multiple cancers; moreover, some cell lines were observed to gain resistance to treatment by alterations in the gene expression and function of 53BP1. We further discussed that targeting this molecule could be of merit in

U

future studies, and illustrated examples to show how targeting the function of 53BP1 increased

N

sensitivity to medications and radiotherapy, especially with regard to the status of BRCA1.

A

Furthermore, evidence was cited regarding the role of 53BP1 in angiogenesis, a necessary process

M

for tumor growth and metastasis. Currently, our knowledge regarding the importance of 53BP1 in cancer is in its primary steps, and future studies can further show how targeting 53BP1 could

CC

References

EP

TE

D

increase our chances in treating various malignancies.

A

1. Lu R, Wang GG. Tudor: a versatile family of histone methylation 'readers'. Trends Biochem Sci. 2013;38(11):546-55. 2. Li L, Zou L. Sensing, signaling, and responding to DNA damage: organization of the checkpoint pathways in mammalian cells. Journal of cellular biochemistry. 2005;94(2):298-306. 3. Iwabuchi K, Bartel PL, Li B, Marraccino R, Fields S. Two cellular proteins that bind to wild-type but not mutant p53. Proc Natl Acad Sci U S A. 1994;91(13):6098-102. 4. Dai Y, Zhang A, Shan S, Gong Z, Zhou Z. Structural basis for recognition of 53BP1 tandem Tudor domain by TIRR. Nat Commun. 2018;9(1):2123. 5. Chapman JR, Sossick AJ, Boulton SJ, Jackson SP. BRCA1-associated exclusion of 53BP1 from DNA damage sites underlies temporal control of DNA repair. J Cell Sci. 2012;125(Pt 15):3529-34.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

6. Lukas C, Melander F, Stucki M, Falck J, Bekker‐Jensen S, Goldberg M, et al. Mdc1 couples DNA double‐strand break recognition by Nbs1 with its H2AX‐dependent chromatin retention. The EMBO journal. 2004;23(13):2674-83. 7. Drane P, Brault ME, Cui G, Meghani K, Chaubey S, Detappe A, et al. TIRR regulates 53BP1 by masking its histone methyl-lysine binding function. Nature. 2017;543(7644):211-6. 8. Bouwman P, Aly A, Escandell JM, Pieterse M, Bartkova J, van der Gulden H, et al. 53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers. Nat Struct Mol Biol. 2010;17(6):688-95. 9. Xiong X, Du Z, Wang Y, Feng Z, Fan P, Yan C, et al. 53BP1 promotes microhomology-mediated end-joining in G1-phase cells. Nucleic Acids Res. 2015;43(3):1659-70. 10. Ward IM, Minn K, van Deursen J, Chen J. p53 Binding protein 53BP1 is required for DNA damage responses and tumor suppression in mice. Mol Cell Biol. 2003;23(7):2556-63. 11. Fernandez-Vidal A, Vignard J, Mirey G. Around and beyond 53BP1 Nuclear Bodies. Int J Mol Sci. 2017;18(12). 12. Gupta A, Hunt CR, Chakraborty S, Pandita RK, Yordy J, Ramnarain DB, et al. Role of 53BP1 in the regulation of DNA double-strand break repair pathway choice. Radiat Res. 2014;181(1):1-8. 13. Difilippantonio S, Gapud E, Wong N, Huang CY, Mahowald G, Chen HT, et al. 53BP1 facilitates long-range DNA end-joining during V(D)J recombination. Nature. 2008;456(7221):529-33. 14. Neboori HJ, Haffty BG, Wu H, Yang Q, Aly A, Goyal S, et al. Low p53 binding protein 1 (53BP1) expression is associated with increased local recurrence in breast cancer patients treated with breastconserving surgery and radiotherapy. International Journal of Radiation Oncology* Biology* Physics. 2012;83(5):e677-e83. 15. Bi J, Huang A, Liu T, Zhang T, Ma H. Expression of DNA damage checkpoint 53BP1 is correlated with prognosis, cell proliferation and apoptosis in colorectal cancer. Int J Clin Exp Pathol. 2015;8(6):6070-82. 16. Youn CK, Kim HB, Wu TT, Park S, Cho SI, Lee JH. 53BP1 contributes to regulation of autophagic clearance of mitochondria. Sci Rep. 2017;7:45290. 17. Gibbs-Seymour I, Markiewicz E, Bekker-Jensen S, Mailand N, Hutchison CJ. Lamin A/Cdependent interaction with 53BP1 promotes cellular responses to DNA damage. Aging Cell. 2015;14(2):162-9. 18. Adams MM, Carpenter PB. Tying the loose ends together in DNA double strand break repair with 53BP1. Cell Div. 2006;1:19. 19. Panier S, Boulton SJ. Double-strand break repair: 53BP1 comes into focus. Nat Rev Mol Cell Biol. 2014;15(1):7-18. 20. FitzGerald JE, Grenon M, Lowndes NF. 53BP1: function and mechanisms of focal recruitment. Portland Press Limited; 2009. 21. Charier G, Couprie J, Alpha-Bazin B, Meyer V, Quéméneur E, Guérois R, et al. The Tudor tandem of 53BP1: a new structural motif involved in DNA and RG-rich peptide binding. Structure. 2004;12(9):1551-62. 22. Bork P, Hofmann K, Bucher P, Neuwald A, Altschul S, Koonin E. A superfamily of conserved domains in DNA damage-responsive cell cycle checkpoint proteins. The FASEB Journal. 1997;11(1):68-76. 23. Clapperton JA, Manke IA, Lowery DM, Ho T, Haire LF, Yaffe MB, et al. Structure and mechanism of BRCA1 BRCT domain recognition of phosphorylated BACH1 with implications for cancer. Nature Structural and Molecular Biology. 2004;11(6):512. 24. Boisvert F-M, Rhie A, Richard S, Doherty AJ. The GAR motif of 53BP1 is arginine methylated by PRMT1 and is necessary for 53BP1 DNA binding activity. Cell cycle. 2005;4(12):1834-41. 25. Adams MM, Wang B, Xia Z, Morales JC, Lu X, Donehower LA, et al. 53BP1 oligomerization is independent of its methylation by PRMT1. Cell cycle. 2005;4(12):1854-61.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

26. Ward IM, Minn K, Jorda KG, Chen J. Accumulation of checkpoint protein 53BP1 at DNA breaks involves its binding to phosphorylated histone H2AX. Journal of Biological Chemistry. 2003;278(22):19579-82. 27. Jowsey P, Morrice NA, Hastie CJ, McLauchlan H, Toth R, Rouse J. Characterisation of the sites of DNA damage-induced 53BP1 phosphorylation catalysed by ATM and ATR. DNA repair. 2007;6(10):153644. 28. Lee H, Kwak H-J, Cho I-t, Park SH, Lee C-H. S1219 residue of 53BP1 is phosphorylated by ATM kinase upon DNA damage and required for proper execution of DNA damage response. Biochemical and biophysical research communications. 2009;378(1):32-6. 29. Munoz IM, Jowsey PA, Toth R, Rouse J. Phospho-epitope binding by the BRCT domains of hPTIP controls multiple aspects of the cellular response to DNA damage. Nucleic acids research. 2007;35(16):5312-22. 30. Minter-Dykhouse K, Ward I, Huen MS, Chen J, Lou Z. Distinct versus overlapping functions of MDC1 and 53BP1 in DNA damage response and tumorigenesis. The Journal of cell biology. 2008;181(5):727-35. 31. Wilson KA, Stern DF. NFBD1/MDC1, 53BP1 and BRCA1 have both redundant and unique roles in the ATM pathway. Cell Cycle. 2008;7(22):3584-94. 32. Koonin E, Altschul S, Bork P. BRCA1 protein products: functional motifs.(1996). Nature Genetics.13:266-68. 33. Chen Y, Weng C, Zhang H, Sun J, Yuan Y. A Direct Interaction Between P53-Binding Protein 1 and Minichromosome Maintenance Complex in Hepg2 Cells. Cellular Physiology and Biochemistry. 2018;47(6):2350-9. 34. von Morgen P, Lidak T, Horejsi Z, Macurek L. Nuclear localisation of 53BP1 is regulated by phosphorylation of the nuclear localisation signal. Biology of the Cell. 2018. 35. Harding SM, Bristow RG. Discordance between phosphorylation and recruitment of 53BP1 in response to DNA double-strand breaks. Cell Cycle. 2012;11(7):1432-44. 36. Fradet-Turcotte A, Canny MD, Escribano-Díaz C, Orthwein A, Leung CC, Huang H, et al. 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature. 2013;499(7456):50. 37. Schultz LB, Chehab NH, Malikzay A, Halazonetis TD. p53 binding protein 1 (53BP1) is an early participant in the cellular response to DNA double-strand breaks. The Journal of cell biology. 2000;151(7):1381-90. 38. Stewart GS. Solving the RIDDLE of 53BP1 recruitment to sites of damage. Cell cycle. 2009;8(10):1532-8. 39. Lottersberger F, Bothmer A, Robbiani DF, Nussenzweig MC, de Lange T. Role of 53BP1 oligomerization in regulating double-strand break repair. Proceedings of the National Academy of Sciences. 2013;110(6):2146-51. 40. Zimmermann M, Lottersberger F, Buonomo SB, Sfeir A, de Lange T. 53BP1 regulates DSB repair using Rif1 to control 5′ end resection. Science. 2013;339(6120):700-4. 41. Chapman JR, Barral P, Vannier J-B, Borel V, Steger M, Tomas-Loba A, et al. RIF1 is essential for 53BP1-dependent nonhomologous end joining and suppression of DNA double-strand break resection. Molecular cell. 2013;49(5):858-71. 42. Escribano-Díaz C, Orthwein A, Fradet-Turcotte A, Xing M, Young JT, Tkáč J, et al. A cell cycledependent regulatory circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Molecular cell. 2013;49(5):872-83. 43. Silverman J, Takai H, Buonomo SB, Eisenhaber F, de Lange T. Human Rif1, ortholog of a yeast telomeric protein, is regulated by ATM and 53BP1 and functions in the S-phase checkpoint. Genes & development. 2004;18(17):2108-19.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

44. Van Vugt MA, Gardino AK, Linding R, Ostheimer GJ, Reinhardt HC, Ong S-E, et al. A mitotic phosphorylation feedback network connects Cdk1, Plk1, 53BP1, and Chk2 to inactivate the G2/M DNA damage checkpoint. PLoS biology. 2010;8(1):e1000287. 45. Jullien D, Vagnarelli P, Earnshaw WC, Adachi Y. Kinetochore localisation of the DNA damage response component 53BP1 during mitosis. Journal of cell science. 2002;115(1):71-9. 46. Grotsky DA, Gonzalez-Suarez I, Novell A, Neumann MA, Yaddanapudi SC, Croke M, et al. BRCA1 loss activates cathepsin L–mediated degradation of 53BP1 in breast cancer cells. J Cell Biol. 2013;200(2):187-202. 47. Gonzalez‐Suarez I, Redwood AB, Grotsky DA, Neumann MA, Cheng EHY, Stewart CL, et al. A new pathway that regulates 53BP1 stability implicates cathepsin L and vitamin D in DNA repair. The EMBO journal. 2011;30(16):3383-96. 48. Gonzalez‐Suarez I, Redwood AB, Perkins SM, Vermolen B, Lichtensztejin D, Grotsky DA, et al. Novel roles for A‐type lamins in telomere biology and the DNA damage response pathway. The EMBO journal. 2009;28(16):2414-27. 49. Han X, Zhang L, Chung J, Pozo FM, Tran A, Seachrist DD, et al. UbcH7 regulates 53BP1 stability and DSB repair. Proceedings of the National Academy of Sciences. 2014;111(49):17456-61. 50. Gibbs‐Seymour I, Markiewicz E, Bekker‐Jensen S, Mailand N, Hutchison CJ. Lamin A/C‐ dependent interaction with 53 BP 1 promotes cellular responses to DNA damage. Aging cell. 2015;14(2):162-9. 51. Pozo FM, Tang J, Bonk KW, Keri RA, Yao X, Zhang Y. Regulatory crosstalk determines the cellular levels of 53BP1, a critical factor in DNA repair. Journal of Biological Chemistry. 2017:jbc. M116. 760645. 52. Zhang A, Peng B, Huang P, Chen J, Gong Z. The p53-binding protein 1-Tudor interacting repair regulator complex participates in the DNA damage response. Journal of Biological Chemistry. 2017:jbc. M117. 777474. 53. Drané P, Brault M-E, Cui G, Meghani K, Chaubey S, Detappe A, et al. TIRR regulates 53BP1 by masking its histone methyl-lysine binding function. Nature. 2017;543(7644):211. 54. Botuyan MV, Cui G, Drané P, Oliveira C, Detappe A, Brault ME, et al. Mechanism of 53BP1 activity regulation by RNA-binding TIRR and a designer protein. Nature structural & molecular biology. 2018;25(7):591. 55. Salvador-Moreno N, Liu J, Haas K, Parker L, Chakraborty C, Kron S, et al. 2017. 56. Butler LR, Densham RM, Jia J, Garvin AJ, Stone HR, Shah V, et al. The proteasomal de‐ ubiquitinating enzyme POH1 promotes the double‐strand DNA break response. The EMBO journal. 2012;31(19):3918-34. 57. Mosbech A, Lukas C, Bekker-Jensen S, Mailand N. The deubiquitylating enzyme USP44 counteracts the DNA double-strand break response mediated by the RNF8 and RNF168 ubiquitin ligases. Journal of Biological Chemistry. 2013:jbc. M113. 459917. 58. Shao G, Lilli DR, Patterson-Fortin J, Coleman KA, Morrissey DE, Greenberg RA. The Rap80BRCC36 de-ubiquitinating enzyme complex antagonizes RNF8-Ubc13-dependent ubiquitination events at DNA double strand breaks. Proceedings of the National Academy of Sciences. 2009;106(9):3166-71. 59. Zimmermann M, de Lange T. 53BP1: pro choice in DNA repair. Trends in cell biology. 2014;24(2):108-17. 60. Nakada S, Tai I, Panier S, Al-Hakim A, Iemura S-i, Juang Y-C, et al. Non-canonical inhibition of DNA damage-dependent ubiquitination by OTUB1. Nature. 2010;466(7309):941. 61. Sato Y, Yamagata A, Goto-Ito S, Kubota K, Miyamoto R, Nakada S, et al. Molecular basis of K63linked polyubiquitination inhibition by the interaction between human deubiquitinating enzyme OTUB1 and ubiquitin-conjugating enzyme UBC13. Journal of Biological Chemistry. 2012:jbc. M112. 364752. 62. Shanbhag NM, Rafalska-Metcalf IU, Balane-Bolivar C, Janicki SM, Greenberg RA. ATM-dependent chromatin changes silence transcription in cis to DNA double-strand breaks. Cell. 2010;141(6):970-81.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

63. Zhang D, Zaugg K, Mak TW, Elledge SJ. A role for the deubiquitinating enzyme USP28 in control of the DNA-damage response. Cell. 2006;126(3):529-42. 64. Tang J, Cho NW, Cui G, Manion EM, Shanbhag NM, Botuyan MV, et al. Acetylation limits 53BP1 association with damaged chromatin to promote homologous recombination. Nature structural & molecular biology. 2013;20(3):317. 65. Hsiao K-Y, Mizzen CA. Histone H4 deacetylation facilitates 53BP1 DNA damage signaling and double-strand break repair. Journal of molecular cell biology. 2013;5(3):157-65. 66. Bunting SF, Callén E, Wong N, Chen H-T, Polato F, Gunn A, et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 2010;141(2):243-54. 67. Bouwman P, Aly A, Escandell JM, Pieterse M, Bartkova J, van der Gulden H, et al. 53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers. Nature structural & molecular biology. 2010;17(6):688. 68. Anderson L, Henderson C, Adachi Y. Phosphorylation and rapid relocalization of 53BP1 to nuclear foci upon DNA damage. Molecular and cellular biology. 2001;21(5):1719-29. 69. Rappold I, Iwabuchi K, Date T, Chen J. Tumor suppressor p53 binding protein 1 (53BP1) is involved in DNA damage–signaling pathways. The Journal of cell biology. 2001;153(3):613-20. 70. Lukas J, Lukas C, Bartek J. More than just a focus: The chromatin response to DNA damage and its role in genome integrity maintenance. Nature cell biology. 2011;13(10):1161. 71. Al-Hakim A, Escribano-Diaz C, Landry M-C, O’Donnell L, Panier S, Szilard RK, et al. The ubiquitous role of ubiquitin in the DNA damage response. DNA repair. 2010;9(12):1229-40. 72. Bekker-Jensen S, Mailand N. The ubiquitin‐and SUMO‐dependent signaling response to DNA double‐strand breaks. FEBS letters. 2011;585(18):2914-9. 73. Doil C, Mailand N, Bekker-Jensen S, Menard P, Larsen DH, Pepperkok R, et al. RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins. Cell. 2009;136(3):435-46. 74. Mailand N, Bekker-Jensen S, Faustrup H, Melander F, Bartek J, Lukas C, et al. RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins. Cell. 2007;131(5):887900. 75. Kolas NK, Chapman JR, Nakada S, Ylanko J, Chahwan R, Sweeney FD, et al. Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science. 2007;318(5856):1637-40. 76. Celeste A, Fernandez-Capetillo O, Kruhlak MJ, Pilch DR, Staudt DW, Lee A, et al. Histone H2AX phosphorylation is dispensable for the initial recognition of DNA breaks. Nature cell biology. 2003;5(7):675. 77. Stewart GS, Panier S, Townsend K, Al-Hakim AK, Kolas NK, Miller ES, et al. The RIDDLE syndrome protein mediates a ubiquitin-dependent signaling cascade at sites of DNA damage. Cell. 2009;136(3):420-34. 78. Bekker-Jensen S, Lukas C, Melander F, Bartek J, Lukas J. Dynamic assembly and sustained retention of 53BP1 at the sites of DNA damage are controlled by Mdc1/NFBD1. The Journal of cell biology. 2005;170(2):201-11. 79. Stewart GS, Wang B, Bignell CR, Taylor AMR, Elledge SJ. MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature. 2003;421(6926):961. 80. Hu Y, Wang C, Huang K, Xia F, Parvin JD, Mondal N. Regulation of 53BP1 protein stability by RNF8 and RNF168 is important for efficient DNA double-strand break repair. PloS one. 2014;9(10):e110522. 81. Mallette FA, Richard S. K48-linked ubiquitination and protein degradation regulate 53BP1 recruitment at DNA damage sites. Cell research. 2012;22(8):1221.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

82. Morales JC, Xia Z, Lu T, Aldrich MB, Wang B, Rosales C, et al. Role for the BRCA1 C-terminal repeats (BRCT) protein 53BP1 in maintaining genomic stability. Journal of Biological Chemistry. 2003;278(17):14971-7. 83. Joo H-Y, Zhai L, Yang C, Nie S, Erdjument-Bromage H, Tempst P, et al. Regulation of cell cycle progression and gene expression by H2A deubiquitination. Nature. 2007;449(7165):1068. 84. Huen MS, Huang J, Leung JW, Sy SM-H, Leung KM, Ching Y-P, et al. Regulation of chromatin architecture by the PWWP domain-containing DNA damage-responsive factor EXPAND1/MUM1. Molecular cell. 2010;37(6):854-64. 85. Huen MS, Huang J, Leung JW, Sy SM, Leung KM, Ching YP, et al. Regulation of chromatin architecture by the PWWP domain-containing DNA damage-responsive factor EXPAND1/MUM1. Mol Cell. 2010;37(6):854-64. 86. Silverman J, Takai H, Buonomo SB, Eisenhaber F, de Lange T. Human Rif1, ortholog of a yeast telomeric protein, is regulated by ATM and 53BP1 and functions in the S-phase checkpoint. Genes Dev. 2004;18(17):2108-19. 87. Botuyan MV, Lee J, Ward IM, Kim J-E, Thompson JR, Chen J, et al. Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair. Cell. 2006;127(7):1361-73. 88. Zgheib O, Pataky K, Brugger J, Halazonetis TD. An oligomerized 53BP1 tudor domain suffices for recognition of DNA double-strand breaks. Molecular and cellular biology. 2009;29(4):1050-8. 89. Majidinia M, Sadeghpour A, Mehrzadi S, Reiter RJ, Khatami N, Yousefi B. Melatonin: A pleiotropic molecule that modulates DNA damage response and repair pathways. Journal of pineal research. 2017;63(1):e12416. 90. Majidinia M, Yousefi B. DNA damage response regulation by microRNAs as a therapeutic target in cancer. DNA repair. 2016;47:1-11. 91. Mirza-Aghazadeh-Attari M, Darband SG, Kaviani M, Mihanfar A, Attari JA, Yousefi B, et al. DNA damage response and repair in colorectal cancer: Defects, regulation and therapeutic implications. DNA repair. 2018. 92. Karimaian A, Majidinia M, Baghi HB, Yousefi B. The crosstalk between Wnt/β-catenin signaling pathway with DNA damage response and oxidative stress: implications in cancer therapy. DNA repair. 2017;51:14-9. 93. Majidinia M, Yousefi B. DNA repair and damage pathways in breast cancer development and therapy. DNA repair. 2017;54:22-9. 94. Maynard S, Fang EF, Scheibye-Knudsen M, Croteau DL, Bohr VA. DNA damage, DNA repair, aging, and neurodegeneration. Cold Spring Harbor perspectives in medicine. 2015:a025130. 95. McKinnon PJ. ATM and ataxia telangiectasia: second in molecular medicine review series. EMBO reports. 2004;5(8):772-6. 96. Bakr A, Köcher S, Volquardsen J, Reimer R, Borgmann K, Dikomey E, et al. Functional crosstalk between DNA damage response proteins 53BP1 and BRCA1 regulates double strand break repair choice. Radiotherapy and oncology. 2016;119(2):276-81. 97. Tehrani SS, Karimian A, Parsian H, Majidinia M, Yousefi B. Multiple functions of long non‐coding RNAs in oxidative stress, DNA damage response and cancer progression. Journal of cellular biochemistry. 2018;119(1):223-36. 98. Lee JH, Goodarzi AA, Jeggo PA, Paull TT. 53BP1 promotes ATM activity through direct interactions with the MRN complex. The EMBO journal. 2010;29(3):574-85. 99. Yoo E, Kim BU, Lee SY, Cho CH, Chung JH, Lee C-H. 53BP1 is associated with replication protein A and is required for RPA2 hyperphosphorylation following DNA damage. Oncogene. 2005;24(35):5423. 100. Zgheib O, Huyen Y, DiTullio Jr RA, Snyder A, Venere M, Stavridi ES, et al. ATM signaling and 53BP1. Radiotherapy and Oncology. 2005;76(2):119-22.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

101. Shibata A, Barton O, Noon AT, Dahm K, Deckbar D, Goodarzi AA, et al. Role of ATM and the damage response mediator proteins 53BP1 and MDC1 in the maintenance of G2/M checkpoint arrest. Molecular and cellular biology. 2010;30(13):3371-83. 102. Her J, Ray C, Altshuler J, Zhang H, Bunting SF. 53BP1 mediates ATR-Chk1 signaling and protects replication forks under conditions of replication stress. Molecular and cellular biology. 2018:MCB. 00472-17. 103. Martínez P, Flores JM, Blasco MA. 53BP1 deficiency combined with telomere dysfunction activates ATR-dependent DNA damage response. J Cell Biol. 2012;197(2):283-300. 104. Xu Y, Ning S, Wei Z, Xu R, Xu X, Xing M, et al. 53BP1 and BRCA1 control pathway choice for stalled replication restart. Elife. 2017;6:e30523. 105. Yoshida K, Miki Y. Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage. Cancer science. 2004;95(11):866-71. 106. Zhang J. The role of BRCA1 in homologous recombination repair in response to replication stress: significance in tumorigenesis and cancer therapy. Cell & bioscience. 2013;3(1):11. 107. Feng L, Li N, Li Y, Wang J, Gao M, Wang W, et al. Cell cycle-dependent inhibition of 53BP1 signaling by BRCA1. Cell discovery. 2015;1:15019. 108. Ou H-L, Schumacher B. DNA damage responses and p53 in the aging process. Blood. 2017:blood2017-07-746396. 109. Cuella-Martin R, Oliveira C, Lockstone HE, Snellenberg S, Grolmusova N, Chapman JR. 53BP1 integrates DNA repair and p53-dependent cell fate decisions via distinct mechanisms. Molecular cell. 2016;64(1):51-64. 110. Iwabuchi K, Li B, Massa HF, Trask BJ, Date T, Fields S. Stimulation of p53-mediated transcriptional activation by the p53-binding proteins, 53BP1 and 53BP2. Journal of Biological Chemistry. 1998;273(40):26061-8. 111. Durocher D, Pelletier L. 53BP1 Goes Back to Its p53 Roots. Molecular cell. 2016;64(1):3-4. 112. Joo WS, Jeffrey PD, Cantor SB, Finnin MS, Livingston DM, Pavletich NP. Structure of the 53BP1 BRCT region bound to p53 and its comparison to the Brca1 BRCT structure. Genes & development. 2002;16(5):583-93. 113. Al Rashid ST, Harding SM, Law C, Coackley C, Bristow RG. Protein-protein interactions occur between p53 phosphoforms and ATM and 53BP1 at sites of exogenous DNA damage. Radiation research. 2011;175(5):588-98. 114. Wrighton KH. UNDERSTANDING THE ACTIONS OF 53BP1. Nature Publishing Group MACMILLAN BUILDING, 4 CRINAN ST, LONDON N1 9XW, ENGLAND; 2016. 115. Morales JC, Franco S, Murphy MM, Bassing CH, Mills KD, Adams MM, et al. 53BP1 and p53 synergize to suppress genomic instability and lymphomagenesis. Proceedings of the National Academy of Sciences. 2006;103(9):3310-5. 116. Ward IM, Difilippantonio S, Minn K, Mueller MD, Molina JR, Yu X, et al. 53BP1 cooperates with p53 and functions as a haploinsufficient tumor suppressor in mice. Molecular and cellular biology. 2005;25(22):10079-86. 117. Fong CS, Mazo G, Das T, Goodman J, Kim M, O'Rourke BP, et al. 53BP1 and USP28 mediate p53dependent cell cycle arrest in response to centrosome loss and prolonged mitosis. Elife. 2016;5:e16270. 118. Meitinger F, Anzola JV, Kaulich M, Richardson A, Stender JD, Benner C, et al. 53BP1 and USP28 mediate p53 activation and G1 arrest after centrosome loss or extended mitotic duration. J Cell Biol. 2016;214(2):155-66. 119. Lambrus BG, Daggubati V, Uetake Y, Scott PM, Clutario KM, Sluder G, et al. A USP28–53BP1– p53–p21 signaling axis arrests growth after centrosome loss or prolonged mitosis. J Cell Biol. 2016;214(2):143-53.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

120. Gupta A, Hunt CR, Chakraborty S, Pandita RK, Yordy J, Ramnarain DB, et al. Role of 53BP1 in the regulation of DNA double-strand break repair pathway choice. Radiation research. 2013;181(1):1-8. 121. Eliezer Y, Argaman L, Rhie A, Doherty AJ, Goldberg M. The direct interaction between 53BP1 and MDC1 is required for the recruitment of 53BP1 to sites of damage. Journal of Biological Chemistry. 2009;284(1):426-35. 122. Mao Z, Bozzella M, Seluanov A, Gorbunova V. Comparison of nonhomologous end joining and homologous recombination in human cells. DNA repair. 2008;7(10):1765-71. 123. Daley JM, Sung P. 53BP1, BRCA1 and the choice between recombination and end joining at DNA double-strand breaks. Molecular and cellular biology. 2014:MCB. 01639-13. 124. Roos WP, Thomas AD, Kaina B. DNA damage and the balance between survival and death in cancer biology. Nature Reviews Cancer. 2016;16(1):20. 125. Ryu J-S, Kang SJ, Koo H-S. The 53BP1 homolog in C. elegans influences DNA repair and promotes apoptosis in response to ionizing radiation. PloS one. 2013;8(5):e64028. 126. Hong S, Li X, Zhao Y, Yang Q, Kong B. 53BP1 suppresses tumor growth and promotes susceptibility to apoptosis of ovarian cancer cells through modulation of the Akt pathway. Oncology reports. 2012;27(4):1251-7. 127. Yang Y, Lei T, Du S, Tong R, Wang H, Yang J, et al. Nuclear GSK3β induces DNA double-strand break repair by phosphorylating 53BP1 in glioblastoma. International journal of oncology. 2018;52(3):709-20. 128. Eliopoulos AG, Havaki S, Gorgoulis VG. DNA damage response and autophagy: a meaningful partnership. Frontiers in genetics. 2016;7:204. 129. Youn CK, Kim HB, Wu TT, Park S, Cho SI, Lee J-H. 53BP1 contributes to regulation of autophagic clearance of mitochondria. Scientific reports. 2017;7:45290. 130. Stiff T, Cerosaletti K, Concannon P, O'driscoll M, Jeggo PA. Replication independent ATR signalling leads to G2/M arrest requiring Nbs1, 53BP1 and MDC1. Human molecular genetics. 2008;17(20):3247-53. 131. Cescutti R, Negrini S, Kohzaki M, Halazonetis TD. TopBP1 functions with 53BP1 in the G1 DNA damage checkpoint. The EMBO journal. 2010;29(21):3723-32. 132. Kwak H-J, Kim SH, Yoo HG, Park SH, Lee C-H. Jun activation domain-binding protein 1 is required for mitotic checkpoint activation via its involvement in hyperphosphorylation of 53BP1. Journal of cancer research and clinical oncology. 2005;131(12):789-96. 133. Sengupta S, Robles AI, Linke SP, Sinogeeva NI, Zhang R, Pedeux R, et al. Functional interaction between BLM helicase and 53BP1 in a Chk1-mediated pathway during S-phase arrest. J Cell Biol. 2004;166(6):801-13. 134. Davalos AR, Kaminker P, Hansen RK, Campisi J. ATR and ATM-dependent movement of BLM helicase during replication stress ensures optimal ATM activation and 53BP1 focus formation. Cell Cycle. 2004;3(12):1579-86. 135. Majidinia M, Alizadeh E, Yousefi B, Akbarzadeh M, Mihanfar A, Rahmati-Yamchi M, et al. Coinhibition of notch and nf-κb signaling pathway decreases proliferation through downregulating iκb-α and hes-1 expression in human ovarian cancer OVCAR-3 cells. Drug research. 2017;67(01):13-9. 136. Xia Y, Shen S, Verma IM. NF-κB, an active player in human cancers. Cancer immunology research. 2014;2(9):823-30. 137. Janssens S, Tschopp J. Signals from within: the DNA-damage-induced NF-κB response. Cell death and differentiation. 2006;13(5):773. 138. Majidinia M, Sadeghpour A, Yousefi B. The roles of signaling pathways in bone repair and regeneration. Journal of cellular physiology. 2018;233(4):2937-48. 139. Li X, Xu B, Moran MS, Zhao Y, Su P, Haffty BG, et al. 53BP1 functions as a tumor suppressor in breast cancer via the inhibition of NF-κB through miR-146a. Carcinogenesis. 2012;33(12):2593-600.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

140. Volcic M, Karl S, Baumann B, Salles D, Daniel P, Fulda S, et al. NF-κB regulates DNA doublestrand break repair in conjunction with BRCA1–CtIP complexes. Nucleic acids research. 2011;40(1):18195. 141. Lim JW, Kim H, Kim KH. Expressions of Ku70 and Ku80 mediated by NF-kB and cyclooxygenase-2 are related to cell proliferation in human gastric cancer cells. Journal of Biological Chemistry. 2002. 142. Kraft D, Rall M, Volcic M, Metzler E, Groo A, Stahl A, et al. NF-κB-dependent DNA damagesignaling differentially regulates DNA double-strand break repair mechanisms in immature and mature human hematopoietic cells. Leukemia. 2015;29(7):1543. 143. Liu Q, Turner KM, Alfred Yung W, Chen K, Zhang W. Role of AKT signaling in DNA repair and clinical response to cancer therapy. Neuro-oncology. 2014;16(10):1313-23. 144. Yousefi B, Azimi A, Majidinia M, Shafiei-Irannejad V, Badalzadeh R, Baradaran B, et al. Balaglitazone reverses P-glycoprotein-mediated multidrug resistance via upregulation of PTEN in a PPARγ-dependent manner in leukemia cells. Tumor Biology. 2017;39(10):1010428317716501. 145. Fresno JV, Casado E, Cejas P, Belda-Iniesta C, González-Barón M. PI3K/Akt signalling pathway and cancer. Cancer treatment reviews. 2004;30(2):193-204. 146. Yousefi B, Zarghami N, Samadi N, Majidinia M. Peroxisome proliferator-activated receptors and their ligands in cancer drug-resistance: opportunity or challenge. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents). 2016;16(12):1541-8. 147. Liu P, Gan W, Guo C, Xie A, Gao D, Guo J, et al. Akt-mediated phosphorylation of XLF impairs non-homologous end-joining DNA repair. Molecular cell. 2015;57(4):648-61. 148. Bartek J, Lukas J. The DNA damage response in tumorigenesis and cancer treatment. Nature Reviews Cancer. 2010;10. 149. Djuzenova CS, Zimmermann M, Katzer A, Fiedler V, Distel LV, Gasser M, et al. A prospective study on histone γ-H2AX and 53BP1 foci expression in rectal carcinoma patients: correlation with radiation therapy-induced outcome. BMC cancer. 2015;15(1):856. 150. Chen Q, Zhou J, Jiang C, Chen J. Reversal of P-glycoprotein-mediated multidrug resistance in SGC7901/VCR cells by PPARγ activation by troglitazone. Journal of Huazhong University of Science and Technology [Medical Sciences]. 2010;30(3):326-31. 151. Zhu H, Yan H, Jin W, Dai C, Chen Z, Zheng F, et al. The staining patterns of 53BP1 nuclear foci and 53BP1 mRNA level are associated with cervical cancer progression and metastasis. International Journal of Gynecological Pathology. 2014;33(3):241-7. 152. Squatrito M, Vanoli F, Schultz N, Jasin M, Holland EC. 53BP1 is a haploinsufficient tumor suppressor and protects cells from radiation response in glioma. Cancer research. 2012:canres. 0045.2012. 153. Bi J, Huang A, Liu T, Zhang T, Ma H. Expression of DNA damage checkpoint 53BP1 is correlated with prognosis, cell proliferation and apoptosis in colorectal cancer. International journal of clinical and experimental pathology. 2015;8(6):6070. 154. Azimi A, Majidinia M, Shafiei-Irannejad V, Jahanban-Esfahlan R, Ahmadi Y, Karimian A, et al. Suppression of p53R2 gene expression with specific siRNA sensitizes HepG2 cells to doxorubicin. Gene. 2018;642:249-55. 155. Hong S, Li X, Zhao Y, Yang Q, Kong B. 53BP1 inhibits the migration and regulates the chemotherapy resistance of ovarian cancer cells. Oncology letters. 2018;15(6):9917-22. 156. Aly A, Ganesan S. BRCA1, PARP, and 53BP1: conditional synthetic lethality and synthetic viability. Journal of molecular cell biology. 2011;3(1):66-74. 157. Schouten PC, Vollebergh MA, Opdam M, Jonkers M, Loden M, Wesseling J, et al. High XIST and low 53BP1 expression predict poor outcome after high dose alkylating chemotherapy in patients with a BRCA1-like breast cancer. Molecular cancer therapeutics. 2015:molcanther. 0470.2015.

A

CC

EP

TE

D

M

A

N

U

SC RI PT

158. Bonanno L, Costa C, Majem M, Sanchez JJ, Gimenez-Capitan A, Rodriguez I, et al. The predictive value of 53BP1 and BRCA1 mRNA expression in advanced non-small-cell lung cancer patients treated with first-line platinum-based chemotherapy. Oncotarget. 2013;4(10):1572. 159. Yao J, Huang A, Zheng X, Liu T, Lin Z, Zhang S, et al. 53BP1 loss induces chemoresistance of colorectal cancer cells to 5-fluorouracil by inhibiting the ATM–CHK2–P53 pathway. Journal of cancer research and clinical oncology. 2017;143(3):419-31. 160. Yousefi B, Samadi N, Baradaran B, Rameshknia V, Shafiei-Irannejad V, Majidinia M, et al. Differential effects of peroxisome proliferator-activated receptor agonists on doxorubicin-resistant human myelogenous leukemia (K562/DOX) cells. Cellular and molecular biology (Noisy-le-Grand, France). 2015;61(8):118-22. 161. Jaspers JE, Kersbergen A, Boon U, Sol W, van Deemter L, Zander SA, et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer discovery. 2012:CD-120049. 162. Wang YT, Yuan B, Chen HD, Xu L, Tian YN, Zhang A, et al. Acquired resistance of phosphatase and tensin homolog‐deficient cells to poly (ADP‐ribose) polymerase inhibitor and Ara‐C mediated by 53BP1 loss and SAMHD1 overexpression. Cancer science. 2018;109(3):821-31. 163. Hassan S, Esch A, Liby T, Gray JW, Heiser LM. Pathway-enriched gene signature associated with 53BP1 response to PARP inhibition in triple-negative breast cancer. Molecular cancer therapeutics. 2017:molcanther. 0170.2017. 164. Barazas M, Annunziato S, Pettitt SJ, de Krijger I, Ghezraoui H, Roobol SJ, et al. The CST Complex Mediates End Protection at Double-Strand Breaks and Promotes PARP Inhibitor Sensitivity in BRCA1Deficient Cells. Cell reports. 2018;23(7):2107-18. 165. Xu G, Chapman JR, Brandsma I, Yuan J, Mistrik M, Bouwman P, et al. REV7 counteracts DNA double-strand break resection and affects PARP inhibition. Nature. 2015;521(7553):541. 166. Wiltshire TD, Lovejoy CA, Wang T, Xia F, O'Connor MJ, Cortez D. Sensitivity to poly (ADP-ribose) polymerase (PARP) inhibition identifies ubiquitin-specific peptidase 11 (USP11) as a regulator of DNA double-strand break repair. Journal of Biological Chemistry. 2010;285(19):14565-71. 167. Chroma K, Mistrik M, Moudry P, Gursky J, Liptay M, Strauss R, et al. Tumors overexpressing RNF168 show altered DNA repair and responses to genotoxic treatments, genomic instability and resistance to proteotoxic stress. Oncogene. 2017;36(17):2405. 168. Li X, Kong X, Wang Y, Yang Q. 53BP1 is a novel regulator of angiogenesis in breast cancer. Cancer science. 2013;104(11):1420-6. 169. Kim Y-J, Park HB, Yoo J-H, Kwon HC, Kim J, Yang HO. Glionitrin A, a new diketopiperazine disulfide, activates ATM-ATR-Chk1/2 via 53BP1 phosphorylation in DU145 cells and shows antitumor effect in xenograft model. Biological and Pharmaceutical Bulletin. 2014;37(3):378-86. 170. Park HB, Kwon HC, Lee C-H, Yang HO. Glionitrin A, an antibiotic− antitumor metabolite derived from competitive interaction between abandoned mine microbes. Journal of natural products. 2009;72(2):248-52. 171. Ma S, Yang D, Liu Y, Wang Y, Lin T, Li Y, et al. LncRNA BANCR promotes tumorigenesis and enhances adriamycin resistance in colorectal cancer. Aging (Albany NY). 2018;10(8):2062. 172. Uehara F, Miwa S, Tome Y, Hiroshima Y, Yano S, Yamamoto M, et al. Comparison of UVB and UVC effects on the DNA damage‐response protein 53BP1 in human pancreatic cancer. Journal of cellular biochemistry. 2014;115(10):1724-8. 173. Miwa S, Tome Y, Yano S, Hiroshima Y, Uehara F, Mii S, et al. Single cell time-lapse imaging of focus formation by the DNA damage-response protein 53BP1 after UVC irradiation of human pancreatic cancer cells. Anticancer research. 2013;33(4):1373-7.

CC

Figures

EP

TE

D

M

A

N

U

SC RI PT

174. Huang C-Y, Hsieh F-S, Wang C-Y, Chen L-J, Chang S-S, Tsai M-H, et al. Palbociclib enhances radiosensitivity of hepatocellular carcinoma and cholangiocarcinoma via inhibiting ataxia telangiectasia– mutated kinase–mediated DNA damage response. European Journal of Cancer. 2018;102:10-22. 175. Maes K, De Smedt E, Lemaire M, De Raeve H, Menu E, Van Valckenborgh E, et al. The role of DNA damage and repair in decitabine-mediated apoptosis in multiple myeloma. Oncotarget. 2014;5(10):3115. 176. Wakai T, Shirai Y, Sakata J, Korita PV, Matsuda Y, Takamura M, et al. Alteration of p53-binding protein 1 expression as a risk factor for local recurrence in patients undergoing resection for extrahepatic cholangiocarcinoma. International journal of oncology. 2011;38(5):1227-36. 177. Mussazhanova Z, Akazawa Y, Matsuda K, Shichijo K, Miura S, Otsubo R, et al. Association between p53-binding protein 1 expression and genomic instability in oncocytic follicular adenoma of the thyroid. Endocrine journal. 2016;63(5):457-67. 178. De Gregoriis G, Ramos JA, Fernandes PV, Vignal GM, Brianese RC, Carraro DM, et al. DNA repair genes PAXIP1 and TP53BP1 expression is associated with breast cancer prognosis. Cancer biology & therapy. 2017;18(6):439-49. 179. Li X, Kong X, Kong X, Wang Y, Yan S, Yang Q. 53BP1 sensitizes breast cancer cells to 5fluorouracil. PLoS one. 2013;8(9):e74928. 180. Liu X, Chen H, Xu X, Ye M, Cao H, Xu L, et al. Insulin‑ like growth factor‑ 1 receptor knockdown enhances radiosensitivity via the HIF‑ 1α pathway and attenuates ATM/H2AX/53BP1 DNA repair activation in human lung squamous carcinoma cells. Oncology Letters. 2018;16(1):1332-40. 181. Lai T-C, Chow K-C, Lin T-Y, Chiang I, Fang H-Y, Chen C-Y, et al. Expression of 53BP1 as a cisplatinresistant marker in patients with lung adenocarcinomas. Oncology reports. 2010;24(2):321-8. 182. Du R, Zheng L, Huang W, Zhang H, Jiang Z. Correlation of 53BP1 gene mutation with prostatic adenocarcinoma. Zhonghua bing li xue za zhi= Chinese journal of pathology. 2011;40(7):449-53. 183. Gou Q, Xie Y, Wu Y, Wang Q, Wang Z, Li P. Downregulation of 53BP1 by short hairpin RNA enhances radiosensitivity in laryngeal carcinoma cells. Sichuan da xue xue bao Yi xue ban= Journal of Sichuan University Medical science edition. 2014;45(5):754-9. 184. Jankovic M, Feldhahn N, Oliveira TY, Silva IT, Kieffer-Kwon K-R, Yamane A, et al. 53BP1 alters the landscape of DNA rearrangements and suppresses AID-induced B cell lymphoma. Molecular cell. 2013;49(4):623-31. 185. Xiao Y, Zheng X, Huang A, Liu T, Zhang T, Ma H. Deficiency of 53BP1 inhibits the radiosensitivity of colorectal cancer. International journal of oncology. 2016;49(4):1600-8.

A

Figure 1: The role of 53BP1 in DNA damage response and its interactions with multiple molecules in the cascade

SC RI PT U N

A

CC

Lung

Prostate adenocarcinoma Laryngeal carcinoma

Human specimens (drug and radiation resistant group) Human specimens HEP-2

53BP1 alteration Inactivation

M

Sporadic breast cancer specimens MCF-7, MDA-MB231, MDA-MB468, and T47D H520

EP

Breast

Not specified

TE

follicular adenoma of the thyroid

D

Cancer type Cell line cholangiocarcinoma Specimens of patients

A

Table 1: The significance of 53BP1 in various types of cancers

Increased activation in nuclear foci Decreased expression Increased ectopic expression

Significance Increased risk of clinically significant recurrence after excision. Activation of 53BP1 marked an increased genomic instability Increased stage and worse prognosis Sensitivity to 5-FU

Ref (176)

(177)

(178) (179)

Decreased signaling Increased expression

Attenuated DNA damage (180) and better radio sensitivity Increased DNA repair caused (181) increased resistance to cisplatin

Mutations in the gene Downregulation by short heparin RNA

Mutations of 53BP1 were seen in prostatic cancer Increased sensitivity to radiotherapy

(182) (183)

B cell lymphoma

Loss of 53BP1

Increased DNA end resection and alteration in translocatome

(184)

Deficiency of 53BP1

Inhibition of radio sensitivity

(185)

A

CC

EP

TE

D

M

A

N

U

SC RI PT

colorectal cancer

activationinduced cytidine deaminase induced HCT116