Open Access

Advances in the role of long non‑coding RNAs and RNA‑binding proteins in regulating DNA damage repair in cancer cells

  • Authors:
    • Songzhu Zou
    • Xiaomei Gou
    • Kunming Wen
  • View Affiliations

  • Published online on: August 23, 2023     https://doi.org/10.3892/ijmm.2023.5296
  • Article Number: 93
  • Copyright: © Zou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA damage and repair play a crucial role in the development, progression and treatment of cancer. In response to various types of DNA damage, the organism initiates a series of DNA damage responses that trigger post‑DNA damage repair processes. Among the most severe forms of DNA damage are DNA double‑strand breaks (DSBs), which can be repaired by the body through two pathways: Homologous recombination and non‑homologous end joining. The repair of DNA damage, particularly DNA DSBs, significantly influences the sensitivity and resistance of cancer cells to chemotherapy and radiotherapy. Numerous studies have demonstrated that long non‑coding RNAs (lncRNAs) can exert multiple regulatory effects on cancer cells by binding to RNA binding proteins (RBPs), thereby influencing DNA damage repair. Based on a comprehensive literature search, the existing research on the regulation of DNA damage repair by lncRNAs interacting with RBPs has primarily focused on the repair of DNA DSBs. Therefore, the present review discusses the regulatory effects of the interaction between lncRNAs and RBPs on DNA damage repair in cancer cells, with a specific focus on the repair of DNA DSBs and its implications in cancer. It is hoped that comprehensive analysis may enhance the current understanding of the molecular mechanisms underlying DNA damage repair in cancer and may lead to the identification of novel diagnostic biomarkers and potential therapeutic targets.

1. Introduction

Various types of DNA damage can occur when cells are exposed to endogenous or exogenous factors, including alterations in base pairs, errors during DNA replication, and twisting and breaking of the DNA double helix strand (1,2). Exogenous factors, such as toxic heavy metals and ionizing radiation are known to cause severe DNA damage (3-7). Endogenous factors are often released during the metabolism of exogenous substances in the body or as a result of cell damage and the loss of cell membrane integrity (8). It is estimated that cells experience ~70,000 DNA lesions per day (9). While the majority of these lesions are single-strand breaks, there are also a few instances of DNA double-strand breaks (DSBs). To cope with this continuously occurring damage, eukaryotic cells have developed a complex and efficient DNA damage response (DDR) system that consists of numerous DNA damage repair pathways (10-12). The primary molecular pathways for DSB repair are homologous recombination (HR) and non-homologous end joining (NHEJ). DSBs are particularly harmful and pose a serious threat to cells. If DSBs are not effectively repaired or undergo error-prone repair, they can lead to carcinogenesis or cell death (13). DNA damage also serves as the foundation of cancer therapy. Chemotherapy and radiotherapy, which are based on inducing severe DNA damage to and the apoptosis of cancer cells, are the preferred treatment regimens for the majority of malignancies (14). However, the activation of DNA damage repair pathways can promote resistance to genotoxic drugs, which remains a significant obstacle in the successful treatment of cancer (15,16). Therefore, it is crucial to elucidate the molecular mechanisms underlying DNA damage repair in order to improve the effectiveness of DNA damage-based anticancer therapies.

Long non-coding RNAs (lncRNAs) are a class of RNAs that exceed 200 nucleotides in length and lack protein-coding potential (17). They have garnered significant attention in recent years. There is mounting evidence to suggest that numerous lncRNAs are dysregulated in various types of cancer and play crucial roles in cancer development and progression (18). The involvement of lncRNAs in drug resistance has also been extensively reported (19,20). Through their interactions with RNA, DNA, or proteins, lncRNAs have emerged as potent regulators of numerous cellular processes (21). RNA-binding proteins (RBPs), a group of proteins, are known to directly bind to single- or double-stranded lncRNAs, participating in lncRNA-mediated regulatory activities (22). Furthermore, the function of certain lncRNAs is dependent on their interactions with specific proteins (23). The interplay between lncRNAs and RBPs plays a critical role in regulating cancer development, progression and drug resistance by influencing DNA damage repair. However, the underlying mechanisms involved in this interplay remain poorly understood, thus necessitating further investigations.

The present review provides a comprehensive summary of the mechanisms through which lncRNAs regulate DDR, DNA DSB repair and influence the sensitivity and resistance to chemotherapy and radiotherapy in DNA damage/repair processes in cancer cells by binding to RBPs. The aim of the present review was to elucidate the regulatory mechanisms of lncRNAs and RBPs associated with cancer development, progression and treatment, thereby aiding the development of novel strategies for cancer therapy. A systematic literature search was conducted using PubMed, employing keywords such as 'long non-coding RNA', 'RNA-binding protein', 'DNA damage', 'DNA repair', 'DNA damage repair', 'DNA double-strand break' and 'cancer'. Articles discussing the regulation of DNA damage repair in cancer by lncRNAs through interactions with RBPs were screened and analyzed.

2. lncRNA binding to RBPs regulates the DNA damage response in cancer cells

Genomic DNA in organisms is highly susceptible to both exogenous and endogenous damage. To maintain genomic integrity and prevent genetic instability, cells and organisms rely on mechanisms to preserve the integrity of their genomic DNA (14,15,24). One crucial mechanism is the DDR, a series of rapid cellular processes that are activated upon the detection of DNA damage (25). The DDR pathway comprises sensors, receptors and effectors that sense DNA damage, propagate signals and initiate appropriate responses, including cell cycle arrest, DNA repair or apoptosis (26-28). In addition to its role in precise cell replication and genome maintenance, there is increasing evidence to indicate that he DDR is involved in resistance to DNA damage-based chemotherapy and radiotherapy (29). It has been observed that molecules expressed as proteins in the DDR pathway can modulate the effects of chemotherapy and radiotherapy (30). Following DNA damage, the ataxia telangiectasia mutated (ATM) kinase is activated through autophosphorylation at the site of damage. ATM, in turn, phosphorylates downstream substrates, including the tumor suppressor p53, breast cancer type 1 susceptibility protein (BRCA1) and checkpoint kinase (CHK)2. These effector molecules transmit DNA damage signals and activate cell cycle checkpoints, DNA repair and apoptosis (24,31,32). Research has highlighted the significant regulatory role of lncRNAs in the DDR, with a number of proteins binding to lncRNAs and participating in their regulatory activities (22). Therefore, the present review provides a comprehensive summary of the role of lncRNA binding to RBPs in these DDR processes (Fig. 1 and Table I).

Table I

lncRNAs bind to RBPs to regulate DNA damage response.

Table I

lncRNAs bind to RBPs to regulate DNA damage response.

RBPMechanismRole(Refs.)
PRC1 and PRC2Inhibition of p15, p16 and p14 expressionInhibits cell cycle checkpoint activation(35)
BCRA1Induced upregulation of JADE1 expression and increased histone H4 acetylation levelInhibits cell cycle checkpoint activation(36)
TRF2Interference with the binding of TRF2 to telomeric double-stranded DNAActivation of cell cycle checkpoints(41)
hnRNP IInhibition of p53 translationInhibition of p53-mediated cell cycle arrest and apoptosis(42)
PTBP3Inhibition of p53 activationInhibition of p53-mediated cell cycle arrest and apoptosis(47)
PCBP2Inhibition of nuclear translocation of p53Inhibition of p53-mediated cell cycle arrest and apoptosis(48)
Sam68Transcriptional co-activator of p53 that enhances p53-mediated cellular responsesPromotes p53-mediated cell cycle arrest and apoptosis(49)
hnRNPKRepression of down-regulated genes as part of the typical p53 transcriptional responsePromotes apoptosis(51)
NF-YABlock or reject the binding of NF-YA to chromatin and inhibit the expression of pro-apoptotic genesInhibition of apoptosis(52)

[i] lncRNAs, long non-coding RNAs; RBP, RNA binding protein; BRCA1, breast cancer type 1 susceptibility protein; TRF2, telomeric-repeat binding factor 2; PTBP3, polypyrimidine tract binding protein 3; PCBP2, poly (rC) binding protein 2; NF-YA, nuclear transcription factor Y, alpha.

Cell cycle checkpoints

DDR involves a series of networks linking tumor suppressor genes to DNA repair pathways, damage tolerance processes, cell cycle checkpoints and apoptosis (24,33). The ATM kinase plays a crucial role as a sensor in the DDR pathway, particularly in detecting DNA DSBs. The ATM-mediated phosphorylation of downstream target proteins initiates signaling cascades that activate cell cycle checkpoints and DNA repair mechanisms (34).

lncRNAs have the ability to directly or indirectly regulate the activation or repression of cell cycle checkpoints through their interactions with RBPs, thereby influencing the DDR. Wan et al (35) discovered that lncRNA ANRIL was induced by the E2F1 transcription factor in an ATM-dependent manner following DNA damage. ANRIL interacted with polycomb repressor complex (PRC)1 and PRC2 to suppress the expression of INK4B-ARF-INK4A motifs, specifically p15(INK4b), p16(INK4a) and p14(ARF). This inhibitory effect on gene expression led to the suppression of cell cycle checkpoint activation, promoting cell proliferation and maintaining the DDR (35). In another study, Wan et al (36) found that lncRNA JADE inhibited the DNA damage checkpoint and enhanced cell proliferation. Similarly, lncRNA JADE expression was induced in an ATM-dependent manner following DNA damage. JADE acted in collaboration with BRCA1 to mediate the transcriptional induction of JADE1 following DNA damage, resulting in the upregulation of JADE1 expression and increased histone H4 acetylation. These molecular events disrupted the DNA damage checkpoint regulation, impaired the DDR and promoted cancer progression (36). Telomeric repeat sequence-containing RNA (TERRA) is a large non-coding RNA localized in mammalian cells and is a component of telomeric heterochromatin (37,38). The inhibition of telomeric-repeat binding factor 2 (TRF2), a protein involved in telomere maintenance, triggers an ATM-dependent DDR pathway and activates cell cycle checkpoints (39,40). Zhang et al (41) demonstrated that TERRA can form a complex with the G-tetraspanin quinoline derivative, CK1-14, which binds to the TERRA G-quadruplex. This complex disrupts the binding of TRF2 to telomeric double-stranded DNA, leading to the induction of a DDR in U2OS cells. Consequently, the cell cycle checkpoint is activated, resulting in cell cycle arrest, the inhibition of cell proliferation and apoptosis. CK1-14 exhibits potential as a lead compound for further development as a novel target for cancer therapy (41).

p53

p53 is a crucial transcription factor involved in stress and the DDR. It plays a pivotal role in activating cell cycle arrest, DNA repair and apoptosis (42). In non-stressed cells, p53 levels are maintained at low levels, while p53 levels are significantly increased during stress (43). In response to stress signals such as DNA damage, p53 is stabilized and activated to perform its function as a sequence-specific transcription factor, inducing genes involved in cell cycle arrest, apoptosis and the expression of its negative regulators (44,45). Apart from protein-coding genes, an increasing number of lncRNAs are recognized as targets of p53 and contribute to p53 regulation and its effector functions (46). lncRNAs can also be involved in the regulation of p53 function through their interactions with RBPs, thereby influencing the DDR.

Inhibition and activation of P53

Zhang et al (42) discovered that following DNA damage, lncRNA ROR interacted with phosphorylated hnRNP I in the cytoplasm. This interaction disrupted the binding of hnRNP I to p53 mRNA, leading to the inhibition of p53 translation. Consequently, p53-mediated cell cycle arrest and apoptosis were suppressed (42). Moreover, p53 forms a self-regulatory feedback loop by regulating ROR and inducing its production. This negative feedback regulation allows for better cellular adaptation to intracellular or extracellular stress (42). Shihabudeen Haider Ali et al also observed that lncRNA Meg3 expression was induced in a p53-dependent manner following DNA damage. p53-dependent lncRNA Meg3 was found to interact with the RBP PTBP3, which inhibited the activation of p53 and suppressed the p53 signaling pathway. This interaction played a role in modulating the DDR (47). In cervical cancer cells, Wen et al (48) found that Linc02535 collaborated with PCBP2 in the cytoplasm to inhibit the nuclear translocation of p53. This led to the promotion of cell cycle progression, DNA damage repair, inhibition of apoptosis and the enhancement of cell proliferation. These events were shown to contribute to the development of cervical cancer in vivo (48). By contrast, Li and Richard (49) discovered that PR-lncRNA-1 interacted with Sam68, enhancing the binding of Sam68 to p53. The presence of Sam68 enhanced the DNA damage-induced expression of PR-lncRNA-1, which in turn promoted the loading of Sam68 and p53 onto the target promoter (49). This upregulation of PR-lncRNA-1 forms a positive feedback regulatory mechanism and enhances p53-mediated cell cycle arrest and the apoptotic regulation of the DDR (49).

p53 transcriptional response

The transcriptional response of p53 involves the activation and repression of numerous genes. It has been discovered that lncRNAs play crucial regulatory roles in the p53 transcriptional response (50). Huarte et al (51) reported that lincRNA p21, located upstream of the CDKN1A gene, was activated by p53 following DNA damage. lincRNA P21 interacted with hnRNPK to participate in a p53-dependent transcriptional repression response. It inhibited the expression of downregulated genes that are typically part of the p53 transcriptional response, thereby promoting apoptosis and contributing to the regulation of the DDR (51). By contrast, Hung et al (52) found that an lncRNA termed PANDA, induced in a p53-dependent manner, restricted apoptosis in the DDR. PANDA was found to bind to NF-YA, preventing or repelling NF-YA from binding to chromatin. This suppression of NF-YA binding led to the downregulation of pro-apoptotic genes, cell cycle arrest and the subsequent regulation of the DDR (52).

3. lncRNA binding to RBPs regulates DNA DSB repair in cancer cells

DNA repair is a critical biological process that ensures the integrity of genomic DNA and enables normal physiological functions, such as cell division (10,53). Under normal conditions, cells possess six major DNA repair pathways that precisely repair DNA damage, thus maintaining genomic stability (54). Among the various types of DNA damage, DSBs are particularly harmful and challenging to repair (55). Fortunately, cells have two primary pathways for repairing DSBs: HR and NHEJ (56). These pathways are typically mediated by proteins belonging to the phosphatidylinositol 3-kinase-like protein kinase family, such as ATM, ATM- and Rad3-related (ATR), and DNA-dependent protein kinase catalytic subunits (DNA-PKcs) (10). The selection of the repair pathway is influenced by the cell cycle phase (57). In the G1 phase, DSBs are primarily repaired through error-prone NHEJ, involving the direct rejoining of DNA ends (58). By contrast, during the S/G2 phase, HR becomes the predominant pathway and utilizes homologous DNA template sequences for error-free repair (59). HR is considered more conserved and error-free due to its reliance on sister chromatids (60,61). However, this property restricts the ability of the HR pathway to repair DSBs to the S/G2 phase, while the NHEJ pathway can repair DSBs throughout the cell cycle (62-64).

The regulatory mechanisms of DNA damage repair play a critical role in the identification of tumor markers and the development of more effective targeted therapies. While the functions of lncRNAs have been extensively studied (65), only a limited number of lncRNAs have been implicated in DNA repair processes (66-68). Furthermore, lncRNAs can bind to RBPs to regulate DNA damage repair. Therefore, it is essential to investigate the regulatory mechanisms involving lncRNAs and RBPs in the two repair pathways, HR and NHEJ, specifically in DSB repair (Fig. 2 and Table II).

Table II

lncRNAs bind to RBPs to regulate DSBs repair.

Table II

lncRNAs bind to RBPs to regulate DSBs repair.

lncRNARBPMechanismRole(Refs.)
LINP1Ku80 and DNA-PKcsEnhancement of molecular interaction between Ku80 and DNA-PKcs and stabilization of Ku80-DNA-PKcs complexesPromote NHEJ(72)
Ku heterodimerReplaces the auxiliary NHEJ protein PAXX in the NHEJ complexPromote NHEJ(71)
LRIKKu heterodimerEnhanced formation of repair complexes at DSB sites in chromatinPromote NHEJ(76)
Linc00312DNA-PKcsInhibition of the recruitment of DNA-PKcs to Ku80Inhibition of NHEJ(77)
DDSR1hnRNPUL1 and BRCA1Isolates the formation of BRCA1-RAP80 complex, derepresses DNA end resection, and regulates the recruitment of BRCA1 and RAP80 to DSBPromote HR(55)
BGL3PARP1BRCA1/BARD1 complex retention at the DSB site and enhanced RAD51 recombinase activity regulates DNA end resectionPromote HR(91)
ANRIL-LEZH2 and Ring1BRecruiting BRCA1, BRCA2, RAD50, and RAD51 proteinsPromote HR(92)
H19RBBP8Involvement of MRN complexes in DNA end resectionPromote HR(95)
HITTERSRAD50 and MRE11Promote the formation of MRN complexesPromote HR(96)
PRLH1RNF169Formation of a stable repair complex that replaces 53BP1 at the DSB sitePromote HR(56)
HITTATMMasking the site on ATM that binds to NBS1 prevents NBS1-mediated recruitment of ATM to the DSBInhibition of HR(98)
MALAT1PARP1 and LIG3Promoting PARP1/LIG3 complex recognition of DSB γH2AX sites on DNAPromote A-NHEJ(108)

[i] lncRNA, long non-coding RNA; DNA-PKcs, DNA-dependent protein kinase catalytic subunits; NHEJ, non-homologous end joining; HR, homologous recombination; DSB, double-strand break; BRCA1, breast cancer type 1 susceptibility protein; PARP1, poly(ADP) ribose polymerase 1; MRN complex, MRE11-RAD50-NBS1 complex; ATM, ataxia telangiectasia mutated; NBS1, Nijmegen breakage syndrome 1 protein; LIG3, DNA ligase 3; H2AX, H2A histone family member X.

NHEJ pathway

The NHEJ pathway is an error-prone mechanism initiated by the binding of DNA break ends to DNA-PK complexes (69). Upon encountering DNA DSBs, Ku80-Ku70 heterodimers bind to the broken ends, forming a clamp complex that recruits DNA-PKcs to the injury site. Two DNA-PKcs molecules interact with the DSB site, forming a synaptic complex that immobilizes the DSB end and protects it from nuclease digestion. Following DNA end processing by Artemis, DNA ligase (LIG)4 and XRCC4 mediate DNA ligation to facilitate the repair of the broken ends (70).

Ku is an RBP that stabilizes the initial synaptic complex in classical NHEJ DSB repair (71). lncRNAs can regulate this repair pathway by binding to Ku. Zhang et al (72) discovered that in triple-negative breast cancer, lncRNA LINP1 interacted with Ku80 and DNA-PKcs, acting as a molecular scaffold. This interaction enhanced the molecular interactions between Ku80 and DNA-PKcs, stabilized the Ku80-DNA-PKcs complex and promoted NHEJ-mediated DNA repair (72). Similarly, in patients with cervical cancer, Wang et al (73) observed the elevated expression of lncRNA LINP1, which promoted NHEJ-mediated DNA repair through the same mechanism described above. Thapar et al (71) further revealed that the interaction between LINP1 and Ku effectively substituted the auxiliary NHEJ protein PAXX in the NHEJ complex. LINP1 enhanced NHEJ-mediated DNA repair by increasing the net concentration of NHEJ factors at DSBs and facilitating the joining of two Ku heterodimers via DSBs, thereby effectively replacing PAXX and achieving efficient NHEJ (71). The early and long-term binding of repair factors has been shown to play a crucial role in the initiation and signal transduction of DNA damage and repair (74). Repair factors, including DNA-PKcs, XRCC4, LIG4 and XLF, bind to DSBs following the perception of damage by the Ku70-Ku80 heterodimer (75). In various cancer cells, lncRNA LRIK is upregulated upon the induction of DNA damage. LRIK interacts with Ku70-Ku80 heterodimers, prolonging their binding to DSB sites and promoting the recruitment of XRCC4 and DNA-PKcs, thereby enhancing the formation of repair complexes at DSB sites in chromatin and facilitating effective DNA damage repair through the NHEJ pathway (76). By contrast, the study by Guo et al (77) reported that linc00312 expression was downregulated in nasopharyngeal carcinoma, leading to a significant decrease in patient survival. Further investigations revealed that linc00312 directly bound to DNA-PKcs and inhibited its recruitment to Ku80, thereby impairing NHEJ repair (77). This, in turn, reduced the viability of nasopharyngeal carcinoma cells and promoted apoptosis (77).

HR pathway

Upon DNA damage, the meiotic recombination 11 homolog 1 (MRE11)-RAD50-Nijmegen breakage syndrome 1 protein (NBS1) (MRN) complex acts as a sensor for DNA DSBs and binds to the damaged site. BRCA1 and CTIP are subsequently recruited to the site of damage. The MRN complex facilitates the activation of ATM through autophosphorylation (78,79). Activated ATM phosphorylates various DNA repair factors, including core histone variants H2AX, CTIP, BRCA1, and the exonuclease EXO1 (59). BRCA1 interacts with CTIP, leading to the activation of MRE11 and stimulating the exonuclease and endonuclease activities necessary for excising the 5'-3'DNA strand and generating a 3'single-stranded DNA (ssDNA) overhang. The ssDNA is then coated by replication protein A (RPA), which prevents the formation of DNA secondary structures. The RPA-coated ssDNA activates CHK1 and CHK2 through ATRIP, resulting in cell cycle arrest to allow time for repair (79-82). BRCA2 binds to BRCA1 and promotes the recruitment of RAD51 to the RPA-coated ssDNA, displacing RPA and forming a stable RAD51-ssDNA complex. BRCA2 also inhibits the ATPase activity of RAD51 and stabilizes the RAD51-ssDNA complex (10,79,83,84). Subsequently, a homology search and DNA repair through strand invasion take place (85).

BRCA1 plays a critical role in the HR repair of DNA (86). Its accumulation at the DNA damage site is essential for an appropriate response to DSBs (24). DNA end resection is a crucial step in initiating and facilitating HR, while inhibiting NHEJ (85). However, BRCA1 can interact with the ubiquitin-binding protein, receptor-associated protein 80 (RAP80), and recruit to the DSB (87), and the aberrant activity of this BRCA1-RAP80 complex would limit HR repair by inhibiting DSB end resection (88-90). Sharma et al (55) reported that following induction by DNA damage, lncRNA DDSR1 interacted with hnRNPUL1 and regulated the formation of the BRCA1-RAP80 complex. This interaction derepressed DNA end resection and regulated the recruitment of BRCA1 and RAP80 at the DSB site, thereby promoting HR repair. The deletion of DDSR1, on the other hand, impaired HR repair (55). Similarly, Hu et al (91) induced DNA damage in breast cancer cell lines and observed that lncRNA BGL3 interacted with poly(ADP-ribose) polymerase 1 (PARP1). Upon recruitment of BGL3 to the DNA damage site, it bound to BARD1 and facilitated the interaction of the BRCA1/BARD1 complex with its binding partners (e.g., RAD51). This resulted in the retention of the BRCA1/BARD1 complex at the DSB site and enhanced RAD51 recombinase activity, which regulated DNA end resection and promoted HR repair (91). In pancreatic cancer cell lines, two isoforms of the lncRNA ANRIL exist, one of which is ANRIL-L, which binds to DNA damage sites and forms a complex with EZH2 and Ring1B. This complex recruits BRCA1, BRCA2, RAD50 and RAD51 proteins to facilitate DNA HR repair during DNA damage repair processes (92).

The MRN complex plays a central role in DNA damage repair by sensing damaged DNA, processing broken DNA ends, and activating DNA damage repair pathways (93,94). In osteosarcoma, lncRNA H19 interacts with RBBP8 (also known as CTIP) and participates in the MRN complex in DNA end resection, promoting HR-mediated DSB repair (95). Under endoplasmic reticulum stress, HITTERS interacts with both RAD50 and MRE11, promoting the formation of MRN complexes. This interaction increases the expression of proteins involved in DNA damage repair, facilitates the repair of the HR pathway, protects oral squamous cell carcinoma from endoplasmic reticulum stress-induced apoptosis, and promotes cancer development (96). MRN complexes also contribute to ATM phosphorylation, subsequently triggering the phosphorylation of various ATM effector proteins (97). Zhao et al (98) reported that lncRNA hypoxia inducible factor-1α (HIF-1α) inhibitor at translation level (HITT) was induced and maintained at high levels following DSB in HCT116 cells. HITT bound to the NBS1 binding site in ATM, masking the site on ATM that binds to NBS1 (98). This binding inhibited the association between ATM and NBS1, preventing the NBS1-mediated recruitment of ATM to the DSB and inhibiting HR repair. This highlights the potential role of HITT in sensitizing cancer to genotoxic treatment (98).

During the G1 phase of the cell cycle, the 53BP1 protein blocks the accumulation of BRCA1 at the DSB site and promotes NHEJ (99-101). The E3 ubiquitin ligase RNF169 has been found to replace 53BP1 at the DSB site, facilitating the initiation of HR repair (102,103). Deng et al (56) discovered that lncRNA PRLH1 specifically bound to RNF169 via two GCUUCA boxes in its 5'terminal region, forming a stable repair complex. This complex stabilized RNF169 and controled the recruitment and retention of RNF169 at the DSB site, replacing 53BP1 and facilitating HR repair (56).

Alternative end joining (ALT-EJ) pathway

In addition to the NHEJ and HR pathways, an alternative end-joining repair pathway, known as ALT-EJ or microhomologous gene-mediated end joining, is responsible for the repair of residual DSBs that cannot be resolved by NHEJ or HR. ALT-EJ is associated with frequent chromosomal abnormalities, such as deletions, translocations, inversions and complex rearrangements (104). It is Ku-independent and is dependent on the microhomologous regions on either side of the break site (70). Several proteins have been identified to be involved in the ALT-EJ repair pathway in mammals, including CTIP in complex with MRN, PARP1, LIG3 and DNA polymerase Pol θ (105). Although LIG3 lacks an RNA-binding structural domain, it can interact with PARP1 through the presence of the PARP and DNA-ligase Zn-finger (zf-PARP) region (106). PARP1 and LIG3 are key molecules in the ALT-EJ DNA repair pathway (107). Hu et al (108) reported that in multiple myeloma, lncRNA MALAT1 bound directly to PARP1 and indirectly to LIG3, facilitating the recognition of DSBs γH2AX sites on DNA by the PARP1/LIG3 complex and promoting DNA repair via A-NHEJ (108). It is worth noting that PARP1 has three zinc finger structural domains, with only the Zn3 structural domain capable of binding to RNA (109). Huang et al (110) further demonstrated in NSCLC cells that PARP1 bound to MALAT1 through the Zn3 structural domain, thereby regulating the ALT-EJ repair pathway. Additionally, MALAT1 was found to promote the HR pathway by regulating the expression of BRCA1 for DNA repair (110).

In summary, lncRNAs play a significant role in various DSB repair pathways through their interactions with RBPs, influencing cancer progression. Therefore, further investigations into the regulation of DSB repair in cancer by lncRNAs binding to RBPs are warranted.

4. Impact of DNA damage/repair on chemotherapy and radiation therapy for cancer

Resistance to chemotherapy and radiation therapy remains a significant challenge in clinical cancer treatment. DNA damage serves as a fundamental mechanism of action for these treatments. DSBs represent the most harmful form of DNA damage that can arise from radiotherapy or DNA-based chemotherapy (15). While radiation and chemotherapy are designed to induce substantial DNA damage in cancer cells, the activation of DNA damage repair systems in the body can limit their effectiveness (111). Therefore, it is crucial to investigate the effects of lncRNA binding to RBPs through DNA damage repair on chemotherapy and radiotherapy for cancer (Table III).

Table III

lncRNAs bind to RBPs to influence cancer chemotherapy and radiotherapy.

Table III

lncRNAs bind to RBPs to influence cancer chemotherapy and radiotherapy.

lncRNARBPMechanismRole(Refs.)
PANDARp53Repression of CDKN1A gene transcription, coping with DNA damageInhibits apoptosis and promotes cancer cell proliferation and chemotherapy resistance(117)
SNHG8hnRNPA1Stabilization of TROY expression, promotes DNA damage repairInhibits apoptosis and promotes chemotherapy resistance(124)
LUCAT1PTBP1Regulation of selective splicing of downstream target genes (APP, CD44, CLSTN1, MBNL1 and ZNF207), inhibits DNA damageInhibits apoptosis and promotes chemotherapy resistance(130)
lnc-POP1-1MCM5Inhibition of MCM5 protein ubiquitination to slow down degradation, promotes DNA damage repairPromotes chemotherapy resistance(135)
AL133467.2ZCCHC4 and γH2AXDownregulation of DNA damage intensity in cancer cells and inhibition of DNA damage-induced apoptosisInhibits apoptosis and promotes chemotherapy resistance(140)
HITTATMMasking the site on ATM that binds to NBS1, preventing NBS1-mediated recruitment of ATM to DSBs, inhibition of HR pathway repairLeads to increased cell death and promotes chemotherapy sensitivity(98)
Linc01134IGF2BP2Enhancement of MAPK1 mRNA stability and promotion of MAPK1 expression, regulating DNA damage responseInhibits apoptosis, promotes cancer cell proliferation and radiotherapy resistance(125)
NORADPUM1Promotion of EEPD1 expression, enhances DNA double-strand break repairInhibits apoptosis and promotes resistance to radiotherapy(126)
MALAT1ANKHD1Positive regulation of YAP1 transcriptional activity, promotes DNA double-strand break repairPromotes cell proliferation and resistance to radiotherapy(128)
LINP1Ku80 and DNA-PKcsEnhanced NHEJ-mediated DNA repair activity on DNA double-strand breaksIncreases the survival rate of cancer cells and confers resistance to radiotherapy(72)
Linc00312DNA-PKcsInhibition of DNA-PKcs recruitment to Ku80 and inhibition of NHEJ repair pathwayDecreases cancer cell viability, promotes apoptosis, and enhances radiotherapy sensitivity(77)

[i] lncRNA; long non-coding RNA; PTBP3, polypyrimidine tract binding protein 3; MCM5, minichromosome maintenance deficient 5; H2AX, H2A histone family member X; ATM, ataxia telangiectasia mutated; IGF2BP2, insulin-like growth factor 2 mRNA binding protein 2; PUM1, Pumilio homolog 1; ANKHD1, ankyrin repeat and KH domain containing 1; DSB, double-strand break; DNA-PKcs, DNA-dependent protein kinase catalytic subunits; NHEJ, non-homologous end joining.

Influencing cancer cell chemotherapy and radiotherapy through transcriptional and post-transcriptional regulation

lncRNAs play a significant role in the regulation of various physiological and pathological cellular processes at three distinct levels: Transcriptional, post-transcriptional and epigenetic. Moreover, they are closely associated with the development, progression and prognosis of cancer (112). There is increasing evidence to support the association between lncRNAs and resistance to chemotherapy and radiotherapy in cancer treatment, thereby highlighting the potential of lncRNAs as biomarkers (113-115). One mechanism by which lncRNAs exert their functions is through their interaction with specific binding proteins (77). Taking into account the existing literature, lncRNAs combined with RBPs are mainly discussed herein to regulate DNA damage repair through transcriptional and post-transcriptional levels, which in turn affects cancer cell chemotherapy and radiotherapy. The epigenetic regulation is not further discussed.

Regulation of transcriptional levels

The CDKN1A (p21) gene plays a critical role in cell cycle checkpoint control and facilitates cell cycle arrest (116). Liu et al (117) discovered that in gastric cancer, lncRNA PANDAR was overexpressed and competitively bound to p53 protein, leading to the suppression of CDKN1A gene transcription. This response to DNA damage inhibited apoptosis, promoted gastric cancer cell proliferation and contributes to chemoresistance. The depletion of PANDAR combined with a p53 activator demonstrated notable efficacy in cancer therapy in vivo (117). PANDAR emerged not only as a potent diagnostic biomarker for patients with gastric cancer, but also as a promising target for cancer therapy (117). Additionally, TROY has been identified as a contributor to DNA damage repair (118). lncRNA SNHG8 exhibits an upregulated expression in multiple types of cancer (119-123). Zhu et al (124) revealed that in gastric cancer, lncRNA SNHG8 bound to hnRNPA1, leading to the stabilization of TROY expression. This interaction promoted DNA damage repair, inhibited apoptosis and ultimately promoted chemotherapeutic resistance in gastric cancer (124). The inhibition of SNHG8 impeded DNA damage repair and reduced the resistance of gastric cancer cells to chemotherapy, providing insight into a novel molecular mechanism underlying drug resistance in gastric cancer (124). Furthermore, in hepatocellular carcinoma, linc01134 has been shown to interact with the IGF2BP2 protein, enhancing MAPK1 mRNA stability and promoting MAPK1 expression, regulating DDR (125). This interaction inhibits apoptosis, accelerates cancer cell proliferation, and augments radiotherapy resistance. Consequently, linc01134 may represent a potential therapeutic target for enhancing the effectiveness of radiotherapy in hepatocellular carcinoma (125). Similarly, Sun et al (126) reported that the DNA damage-activated non-coding RNA NORAD competitively bound to PUM1 of pri-miR-199a1, impeding the processing of pri-miR-199a1. Consequently, the expression of miR-199a-5p was suppressed, resulting in the upregulation of EEPD1 expression (126). This process enhanced the HR repair pathway in DNA DSBs and inhibited cell apoptosis, thereby conferring resistance to radiotherapy in ESCC cells (126). Previous research by Yao et al (127) demonstrated that ANKHD1 was highly expressed in colorectal cancer (CRC) and promoted CRC cell proliferation, invasion and migration through the activation of YAP1. Subsequent investigations revealed that ANKHD1 interacted with both lncRNA MALAT1 and YAP1 in CRC. Both ANKHD1 and MALAT1 positively regulated the transcriptional activity of YAP1, which in turn promoted ATM-CHK2 phosphorylation by activating AKT. Consequently, this cascade upregulated MRE11 expression, facilitating DNA DSB repair and ultimately promoting radiotherapy resistance in CRC. This ANKHD1/MALAT1/YAP1 interaction loop, along with the downstream YAP1/AKT axis, may represent a potential therapeutic target for comprehensive CRC treatment (128).

Post-transcriptional regulation

lncRNAs can exert their influence on resistance to chemo- and radiotherapy in cancer cells through post-transcriptional regulation. PTBP1 is an RBP known for its involvement in premature RNA splicing events and its association with cancer progression (129). Huan et al (130) discovered that in CRC, lncRNA LUCAT1 was induced by HIF-1α transcription under hypoxic stress. Elevated levels of LUCAT1 interacted with PTBP1 protein, regulating the selective splicing of downstream target genes (APP, CD44, CLSTN1, MBNL1 and ZNF207) (130). This interaction inhibited DNA damage and apoptosis, leading to chemoresistance and promoting CRC cell survival. These findings suggest that LUCAT1 may serve as a predictive indicator and therapeutic target for patients with CRC undergoing chemotherapy (130).

Influencing cancer cell chemotherapy and radiotherapy by regulating the repair of DNA DSBs

Resistance to chemotherapy and radiotherapy primarily arises from the induction of DNA DSBs. In response, three important DNA damage sensors, ATM, ATR and DNA-PKcs, are immediately activated to assist cancer cells in evading the damage caused by chemo- and radiotherapy. This evasion is accomplished through enhanced DNA repair mechanisms (131-133). Zhang et al (72) reported that LINP1 was highly expressed in triple-negative breast cancer and that the inhibition of LINP1 expression impaired DNA repair activity, thereby sensitizing the cancer cells to radiation therapy. Their study also revealed a positive correlation between LINP1 and epidermal growth factor receptor (EGFR) expression (72). Further investigations demonstrated that EGFR pathway activation, followed by MAPK (RAS-MEK-ERK) pathway activation and AP1 transcription factor induction, led to an increased LINP1 transcription (72). Elevated LINP1 levels stabilized the interaction between Ku80 and DNA-PKcs, enhancing NHEJ-mediated DNA repair activity. This, in turn, increased cancer cell survival and contributed to radiotherapy resistance (72). Similar mechanisms have been observed in cervical cancer, where LINP1 played a role in radiation resistance and served as a prognostic marker and potential therapeutic target (73). Conversely, another study demonstrated that linc00312 expression was downregulated in nasopharyngeal carcinoma and this was associated with a reduced patient survival (77). Subsequent analyses demonstrated that linc00312 directly bound to DNA-PKcs, inhibiting its recruitment to Ku80 and impairing NHEJ repair. This resulted in the decreased viability and increased apoptosis of nasopharyngeal carcinoma cells (77). Moreover, linc00312 inhibited radiation-induced AKT-DNA-PKcs, MRN-ATM-CHK2 and ATR-CHK1 signaling, leading to impaired DNA damage sensing, processing and repair. Consequently, the sensitivity to radiation therapy was increased. These findings provide new insight into the regulation of radiosensitivity by linc00312 in nasopharyngeal carcinoma (77). Additionally, Zhao et al (98) reported that lncRNA HITT was downregulated in multiple types of cancer. However, under DSB induction, HITT transcription was upregulated and maintained at high levels. HITT bound to the NBS1 binding site in ATM, preventing the association between ATM and NBS1 (98). This inhibition hindered the recruitment of ATM to the DSB site, impairing HR pathway repair. In vitro and in vivo analyses demonstrated that the HITT-mediated inhibition of ATM increased the death of cancer cells treated with doxorubicin, suggesting its significant role in enhancing chemosensitivity. Blocking the NBS1/ATM interaction may thus be a potential target for anticancer therapy (98).

Ubiquitination modifications

lncRNAs can also regulate protein levels through ubiquitination modifications (134). Jiang et al (135) discovered that in head and neck squamous cell carcinoma (HNSCC), upregulated lnc-POP1-1 directly bound to the DNA repair protein minichromosome maintenance deficient 5 (MCM5), which attenuated the degradation of MCM5. This interaction promoted DNA damage repair by inhibiting the ubiquitination of MCM5 protein, ultimately leading to cisplatin resistance in HNSCC cells (135). VN1R5 and Lnc-POP1-1 may thus serve as predictive markers for cisplatin resistance and potential therapeutic targets for reversing cisplatin resistance in HNSCC patients (135).

In recent years, the role of RBPs and their partners in cancer progression and treatment has garnered increasing attention (136,137). RBPs were once considered 'non-druggable'; however, the identification of small molecules or chemically modified antisense oligonucleotides targeting RBPs has opened up new possibilities for the treatment of certain diseases (138,139). Zhu et al (140) discovered that a highly expressed RBP, zinc finger CCHC domain-containing protein 4 (ZCCHC4), was associated with a poor prognosis in several types of cancer. ZCCHC4 and the previously unidentified lncRNA AL133467.2 formed nuclear complexes with the DNA damage indicator γH2AX in oxaliplatin-induced DDR. ZCCHC4 attenuated AL133467.2 and γH2AX, resulting in a downregulation of DNA damage intensity in cancer cells (140). This interaction inhibited DNA damage-induced apoptosis in hepatocellular carcinoma cells and promoted chemoresistance. These findings provide a novel understanding of the mechanisms through which RBPs and their interacting molecules regulate cancer progression and chemoresistance. The epigenetic role of RBPs and their partners in solid cancer chemoresistance remains poorly understood and thus requires further investigation (140).

5. Conclusions and future perspectives

DNA damage, DDR, and repair are crucial factors in cancer development, progression and therapy. Despite previous perceptions of lncRNAs as 'junk RNA' due to their lack of protein-coding capacity, it is now evident that they play significant roles in various aspects of cancer biology. lncRNAs interact with RBPs and contribute to numerous cellular processes, including the regulation of DNA damage repair in cancer cells. The present review provides insight into the molecular mechanisms underlying the interaction between lncRNAs and RBPs, specifically in the context of DNA damage repair in cancer cells. This knowledge may open up new avenues for cancer treatment strategies aimed at enhancing the effectiveness of DNA damage-repair-based therapies. Although substantial research has been conducted to elucidate the functions and mechanisms of lncRNAs and their impact on cancer therapy, the precise underlying mechanisms remain largely unknown. Therefore, further investigations are warranted to enhance the current understanding of this intricate interplay.

Availability of data and materials

Not applicable.

Authors' contributions

SZ and KW conceived the study. SZ drafted the manuscript, and prepared the figures and tables. XG participated in the literature search and in the analysis of the data to be included in the review. KW edited and revised the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

DSB

double-strand break

DDR

DNA damage response

HR

homologous recombination

NHEJ

non-homologous end joining

lncRNA

long non-coding RNA

RBP

RNA binding protein

ATM

ataxia telangiectasia mutated

ATR

ATM-and Rad3-related

DNA-PKcs

DNA-dependent protein kinase catalytic subunits

RPA

replication protein A

MRE11

meiotic recombination 11 homolog 1

NBS1

Nijmegen breakage syndrome 1 protein

RAP80

receptor-associated protein 80

ALT-EJ

alternative end joining

HIF-1α

hypoxia inducible factor-1α

EGFR

epidermal growth factor receptor

MCM5

minichromosome maintenance deficient 5

ZCCHC4

zinc finger CCHC domain-containing protein 4

Acknowledgments

Not applicable.

Funding

The present review was supported by the National Natural Science Foundation of China (grant no. 82160575) and the Outstanding Young Technological and Innovative Talent Cultivation Project of Zunyi Municipal Science and Technology Bureau, 2021 (no. 10).

References

1 

Ragunathan K, Upfold NLE and Oksenych V: Interaction between fibroblasts and immune cells following DNA Damage induced by ionizing radiation. Int J Mol Sci. 21:86352020. View Article : Google Scholar : PubMed/NCBI

2 

Marshall CJ and Santangelo TJ: Archaeal DNA repair mechanisms. Biomolecules. 10:14722020. View Article : Google Scholar : PubMed/NCBI

3 

Maremonti E, Brede DA, Olsen AK, Eide DM and Berg ES: Ionizing radiation, genotoxic stress, and mitochondrial DNA copy-number variation in Caenorhabditis elegans: Droplet digital PCR analysis. Mutat Res Genet Toxicol Environ Mutagen. 858-860:5032772020. View Article : Google Scholar : PubMed/NCBI

4 

Pariset E, Malkani S, Cekanaviciute E and Costes SV: Ionizing radiation-induced risks to the central nervous system and countermeasures in cellular and rodent models. Int J Radiat Biol. 97(Suppl): S132–S150. 2021. View Article : Google Scholar

5 

Wu R, Hogberg J, Adner M, Ramos-Ramirez P, Stenius U and Zheng H: Crystalline silica particles cause rapid NLRP3-dependent mitochondrial depolarization and DNA damage in airway epithelial cells. Part Fibre Toxicol. 17:392020. View Article : Google Scholar : PubMed/NCBI

6 

Dussert F, Arthaud PA, Arnal ME, Dalzon B, Torres A, Douki T, Herlin N, Rabilloud T and Carriere M: Toxicity to RAW264.7 macrophages of silica nanoparticles and the E551 food additive, in combination with genotoxic agents. Nanomaterials (Basel). 10:14182020. View Article : Google Scholar : PubMed/NCBI

7 

Huang R, Yu T, Li Y and Hu J: Upregulated has-miR-4516 as a potential biomarker for early diagnosis of dust-induced pulmonary fibrosis in patients with pneumoconiosis. Toxicol Res (Camb). 7:415–422. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Gupta N, Khetan D, Chaudhary R and Shukla JS: Prospective cohort study to assess the effect of storage duration, Leuko-filtration, and gamma irradiation on cell-free DNA in red cell components. Transfus Med Hemother. 47:409–419. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Lindahl T and Barnes DE: Repair of endogenous DNA damage. Cold Spring Harb Symp Quant Biol. 65:127–133. 2000. View Article : Google Scholar

10 

Jackson SP and Bartek J: The DNA-damage response in human biology and disease. Nature. 461:1071–1078. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Aguilera A and Garcia-Muse T: Causes of genome instability. Annu Rev Genet. 47:1–32. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Aguilera A and Gomez-Gonzalez B: Genome instability: A mechanistic view of its causes and consequences. Nat Rev Genet. 9:204–217. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Li J, Sun H, Huang Y, Wang Y, Liu Y and Chen X: Pathways and assays for DNA double-strand break repair by homologous recombination. Acta Biochim Biophys Sin (Shanghai). 51:879–889. 2019. View Article : Google Scholar : PubMed/NCBI

14 

O'Connor MJ: Targeting the DNA damage response in cancer. Mol Cell. 60:547–560. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Lord CJ and Ashworth A: The DNA damage response and cancer therapy. Nature. 481:287–294. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Pilie PG, Tang C, Mills GB and Yap TA: State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 16:81–104. 2019. View Article : Google Scholar

17 

Marchese FP, Raimondi I and Huarte M: The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 18:2062017. View Article : Google Scholar : PubMed/NCBI

18 

Huarte M: The emerging role of lncRNAs in cancer. Nat Med. 21:1253–1261. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Fanale D, Castiglia M, Bazan V and Russo A: Involvement of Non-coding RNAs in Chemo- and Radioresistance of colorectal Cancer. Adv Exp Med Biol. 937:207–228. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Zhou XL, Wang WW, Zhu WG, Yu CH, Tao GZ, Wu QQ, Song YQ, Pan P and Tong YS: High expression of long non-coding RNA AFAP1-AS1 predicts chemoradioresistance and poor prognosis in patients with esophageal squamous cell carcinoma treated with definitive chemoradiotherapy. Mol Carcinog. 55:2095–2105. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Haemmig S, Yang D, Sun X, Das D, Ghaffari S, Molinaro R, Chen L, Deng Y, Freeman D, Moullan N, et al: Long noncoding RNA SNHG12 integrates a DNA-PK-mediated DNA damage response and vascular senescence. Sci Transl Med. 12:eaaw18682020. View Article : Google Scholar : PubMed/NCBI

22 

Zhang Y, Tao Y, Li Y, Zhao J, Zhang L, Zhang X, Dong C, Xie Y, Dai X, Zhang X and Liao Q: The regulatory network analysis of long noncoding RNAs in human colorectal cancer. Funct Integr Genomics. 18:261–275. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Wang Y and Wang Y, Luo W, Song X, Huang L, Xiao J, Jin F, Ren Z and Wang Y: Roles of long non-coding RNAs and emerging RNA-binding proteins in innate antiviral responses. Theranostics. 10:9407–9424. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Ciccia A and Elledge SJ: The DNA damage response: Making it safe to play with knives. Mol Cell. 40:179–204. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Michelini F, Pitchiaya S, Vitelli V, Sharma S, Gioia U, Pessina F, Cabrini M, Wang Y, Capozzo I, Iannelli F, et al: Damage-induced lncRNAs control the DNA damage response through interaction with DDRNAs at individual double-strand breaks. Nat Cell Biol. 19:1400–1411. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Surova O and Zhivotovsky B: Various modes of cell death induced by DNA damage. Oncogene. 32:3789–3797. 2013. View Article : Google Scholar

27 

Roos WP, Thomas AD and Kaina B: DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer. 16:20–33. 2016. View Article : Google Scholar

28 

Sun X, Wang Y, Ji K, Liu Y, Kong Y, Nie S, Li N, Hao J, Xie Y, Xu C, et al: NRF2 preserves genomic integrity by facilitating ATR activation and G2 cell cycle arrest. Nucleic Acids Res. 48:9109–9123. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Yu R, Hu Y, Zhang S, Li X, Tang M, Yang M, Wu X, Li Z, Liao X, Xu Y, et al: LncRNA CTBP1-DT-encoded microprotein DDUP sustains DNA damage response signalling to trigger dual DNA repair mechanisms. Nucleic Acids Res. 50:8060–8079. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Wu CH, Chen CY, Yeh CT and Lin KH: Radiosensitization of hepatocellular carcinoma through targeting radio-associated MicroRNA. Int J Mol Sci. 21:18592020. View Article : Google Scholar : PubMed/NCBI

31 

Kitagawa R and Kastan MB: The ATM-dependent DNA damage signaling pathway. Cold Spring Harb Symp Quant Biol. 70:99–109. 2005. View Article : Google Scholar

32 

Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER III, Hurov KE, Luo J, Bakalarski CE, Zhao Z, Solimini N, Lerenthal Y, et al: ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 316:1160–1166. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Bartek J and Lukas J: DNA damage checkpoints: From initiation to recovery or adaptation. Curr Opin Cell Biol. 19:238–245. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Shiloh Y: ATM and related protein kinases: Safeguarding genome integrity. Nat Rev Cancer. 3:155–168. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Wan G, Mathur R, Hu X, Liu Y, Zhang X, Peng G and Lu X: Long non-coding RNA ANRIL (CDKN2B-AS) is induced by the ATM-E2F1 signaling pathway. Cell Signal. 25:1086–1095. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Wan G, Hu X, Liu Y, Han C, Sood AK, Calin GA, Zhang X and Lu X: A novel non-coding RNA lncRNA-JADE connects DNA damage signalling to histone H4 acetylation. EMBO J. 32:2833–2847. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Schoeftner S and Blasco MA: Developmentally regulated transcription of mammalian telomeres by DNA-dependent RNA polymerase II. Nat Cell Biol. 10:228–236. 2008. View Article : Google Scholar

38 

Xu Y and Komiyama M: Structure, function and targeting of human telomere RNA. Methods. 57:100–105. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Karlseder J, Broccoli D, Dai Y, Hardy S and de Lange T: p53- and ATM-dependent apoptosis induced by telomeres lacking TRF2. Science. 283:1321–1325. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Okamoto K, Bartocci C, Ouzounov I, Diedrich JK, Yates JR III and Denchi EL: A two-step mechanism for TRF2-mediated chromosome-end protection. Nature. 494:502–505. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Zhang Y, Zeng D, Cao J, Wang M, Shu B, Kuang G, Ou TM, Tan JH, Gu LQ, Huang ZS and Li D: Interaction of Quindoline derivative with telomeric repeat-containing RNA induces telomeric DNA-damage response in cancer cells through inhibition of telomeric repeat factor 2. Biochim Biophys Acta Gen Subj. 1861:3246–3256. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Zhang A, Zhou N, Huang J, Liu Q, Fukuda K, Ma D, Lu Z, Bai C, Watabe K and Mo YY: The human long non-coding RNA-RoR is a p53 repressor in response to DNA damage. Cell Res. 23:340–350. 2013. View Article : Google Scholar :

43 

Meek DW and Anderson CW: Posttranslational modification of p53: Cooperative integrators of function. Cold Spring Harb Perspect Biol. 1:a0009502009. View Article : Google Scholar

44 

Zilfou JT and Lowe SW: Tumor suppressive functions of p53. Cold Spring Harb Perspect Biol. 1:a0018832009. View Article : Google Scholar :

45 

Vousden KH and Prives C: Blinded by the light: The growing complexity of p53. Cell. 137:413–431. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Zhang A, Xu M and Mo YY: Role of the lncRNA-p53 regulatory network in cancer. J Mol Cell Biol. 6:181–191. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Shihabudeen Haider Ali MS, Cheng X, Moran M, Haemmig S, Naldrett MJ, Alvarez S, Feinberg MW and Sun X: LncRNA Meg3 protects endothelial function by regulating the DNA damage response. Nucleic Acids Res. 47:1505–1522. 2019. View Article : Google Scholar :

48 

Wen D, Huang Z, Li Z, Tang X, Wen X, Liu J and Li M: LINC02535 co-functions with PCBP2 to regulate DNA damage repair in cervical cancer by stabilizing RRM1 mRNA. J Cell Physiol. 235:7592–7603. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Li N and Richard S: Sam68 functions as a transcriptional coactivator of the p53 tumor suppressor. Nucleic Acids Res. 44:8726–8741. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, et al: Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci USA. 106:11667–11672. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, Khalil AM, Zuk O, Amit I, Rabani M, et al: A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 142:409–419. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, Horlings HM, Shah N, Umbricht C, Wang P, et al: Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet. 43:621–629. 2011. View Article : Google Scholar : PubMed/NCBI

53 

van Gent DC, Hoeijmakers JH and Kanaar R: Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet. 2:196–206. 2001. View Article : Google Scholar : PubMed/NCBI

54 

Sancar A, Lindsey-Boltz LA, Unsal-Kaçmaz K and Linn S: Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem. 73:39–85. 2004. View Article : Google Scholar : PubMed/NCBI

55 

Sharma V, Khurana S, Kubben N, Abdelmohsen K, Oberdoerffer P, Gorospe M and Misteli T: A BRCA1-interacting lncRNA regulates homologous recombination. EMBO Rep. 16:1520–1534. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Deng B, Xu W, Wang Z, Liu C, Lin P, Li B, Huang Q, Yang J, Zhou H and Qu L: An LTR retrotransposon-derived lncRNA interacts with RNF169 to promote homologous recombination. EMBO Rep. 20:e476502019. View Article : Google Scholar : PubMed/NCBI

57 

Branzei D and Foiani M: Regulation of DNA repair throughout the cell cycle. Nat Rev Mol Cell Biol. 9:297–308. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Lieber MR: The mechanism of human nonhomologous DNA end joining. J Biol Chem. 283:1–5. 2008. View Article : Google Scholar

59 

San Filippo J, Sung P and Klein H: Mechanism of eukaryotic homologous recombination. Annu Rev Biochem. 77:229–257. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Kumar A, Purohit S and Sharma NK: Aberrant DNA Double-strand break repair threads in breast carcinoma: Orchestrating genomic insult survival. J Cancer Prev. 21:227–234. 2016. View Article : Google Scholar

61 

Yao Y, Li X, Chen W, Liu H, Mi L, Ren D, Mo A and Lu P: ATM promotes RAD51-mediated meiotic DSB repair by inter-sister-chromatid recombination in Arabidopsis. Front Plant Sci. 11:8392020. View Article : Google Scholar : PubMed/NCBI

62 

Trenner A and Sartori AA: Harnessing DNA Double-strand break repair for cancer treatment. Front Oncol. 9:13882019. View Article : Google Scholar

63 

Gomez-Mejiba SE and Ramirez DC: Trapping of DNA radicals with the nitrone spin trap 5,5-dimethyl-1-pyrroline N-oxide and genotoxic damage: Recent advances using the immuno-spin trapping technology. Mutat Res Rev Mutat Res. 782:1082832019. View Article : Google Scholar : PubMed/NCBI

64 

Dasika GK, Lin SC, Zhao S, Sung P, Tomkinson A and Lee EY: DNA damage-induced cell cycle checkpoints and DNA strand break repair in development and tumorigenesis. Oncogene. 18:7883–7899. 1999. View Article : Google Scholar

65 

Zhao Y, Li H, Fang S, Kang Y, Wu W, Hao Y, Li Z, Bu D, Sun N, Zhang MQ and Chen R: NONCODE 2016: An informative and valuable data source of long non-coding RNAs. Nucleic Acids Res. 44:D203–D208. 2016. View Article : Google Scholar :

66 

Dimitrova N, Zamudio JR, Jong RM, Soukup D, Resnick R, Sarma K, Ward AJ, Raj A, Lee JT, Sharp PA and Jacks T: LincRNA-p21 activates p21 in cis to promote Polycomb target gene expression and to enforce the G1/S checkpoint. Mol Cell. 54:777–790. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Schmitt AM, Garcia JT, Hung T, Flynn RA, Shen Y, Qu K, Payumo AY, Peres-da-Silva A, Broz DK, Baum R, et al: An inducible long noncoding RNA amplifies DNA damage signaling. Nat Genet. 48:1370–1376. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Liu X, Li D, Zhang W, Guo M and Zhan Q: Long non-coding RNA gadd7 interacts with TDP-43 and regulates Cdk6 mRNA decay. EMBO J. 31:4415–4427. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Shen L, Wang Q, Liu R, Chen Z, Zhang X, Zhou P and Wang Z: LncRNA lnc-RI regulates homologous recombination repair of DNA double-strand breaks by stabilizing RAD51 mRNA as a competitive endogenous RNA. Nucleic Acids Res. 46:717–729. 2018. View Article : Google Scholar :

70 

Huang R and Zhou PK: DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct Target Ther. 6:2542021. View Article : Google Scholar : PubMed/NCBI

71 

Thapar R, Wang JL, Hammel M, Ye R, Liang K, Sun C, Hnizda A, Liang S, Maw SS, Lee L, et al: Mechanism of efficient double-strand break repair by a long non-coding RNA. Nucleic Acids Res. 48:10953–10972. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Zhang Y, He Q, Hu Z, Feng Y, Fan L, Tang Z, Yuan J, Shan W, Li C, Hu X, et al: Long noncoding RNA LINP1 regulates repair of DNA double-strand breaks in triple-negative breast cancer. Nat Struct Mol Biol. 23:522–530. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Wang X, Liu H, Shi L, Yu X, Gu Y and Sun X: LINP1 facilitates DNA damage repair through non-homologous end joining (NHEJ) pathway and subsequently decreases the sensitivity of cervical cancer cells to ionizing radiation. Cell Cycle. 17:439–447. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Soutoglou E and Misteli T: Activation of the cellular DNA damage response in the absence of DNA lesions. Science. 320:1507–1510. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Downs JA and Jackson SP: A means to a DNA end: The many roles of Ku. Nat Rev Mol Cell Biol. 5:367–378. 2004. View Article : Google Scholar : PubMed/NCBI

76 

Wang D, Zhou Z, Wu E, Ouyang C, Wei G, Wang Y, He D, Cui Y, Zhang D, Chen X, et al: LRIK interacts with the Ku70-Ku80 heterodimer enhancing the efficiency of NHEJ repair. Cell Death Differ. 27:3337–3353. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Guo Z, Wang YH, Xu H, Yuan CS, Zhou HH, Huang WH, Wang H and Zhang W: LncRNA linc00312 suppresses radiotherapy resistance by targeting DNA-PKcs and impairing DNA damage repair in nasopharyngeal carcinoma. Cell Death Dis. 12:692021. View Article : Google Scholar : PubMed/NCBI

78 

Uziel T, Lerenthal Y, Moyal L, Andegeko Y, Mittelman L and Shiloh Y: Requirement of the MRN complex for ATM activation by DNA damage. EMBO J. 22:5612–5621. 2003. View Article : Google Scholar : PubMed/NCBI

79 

Prakash R, Zhang Y, Feng W and Jasin M: Homologous recombination and human health: The roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harb Perspect Biol. 7:a0166002015. View Article : Google Scholar : PubMed/NCBI

80 

Gorgoulis VG, Pefani DE, Pateras IS and Trougakos IP: Integrating the DNA damage and protein stress responses during cancer development and treatment. J Pathol. 246:12–40. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Heyer WD, Ehmsen KT and Liu J: Regulation of homologous recombination in eukaryotes. Annu Rev Genet. 44:113–139. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Maréchal A and Zou L: DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol. 5:a0127162013. View Article : Google Scholar : PubMed/NCBI

83 

Renkawitz J, Lademann CA and Jentsch S: Mechanisms and principles of homology search during recombination. Nat Rev Mol Cell Biol. 15:369–383. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Ranjha L, Howard SM and Cejka P: Main steps in DNA double-strand break repair: An introduction to homologous recombination and related processes. Chromosoma. 127:187–214. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Yu N, Qin H, Zhang F, Liu T, Cao K, Yang Y, Chen Y and Cai J: The role and mechanism of long non-coding RNAs in homologous recombination repair of radiation-induced DNA damage. J Gene Med. 25:e34702023. View Article : Google Scholar

86 

Ohta T, Sato K and Wu W: The BRCA1 ubiquitin ligase and homologous recombination repair. FEBS Lett. 585:2836–2844. 2011. View Article : Google Scholar : PubMed/NCBI

87 

Kim H, Chen J and Yu X: Ubiquitin-binding protein RAP80 mediates BRCA1-dependent DNA damage response. Science. 316:1202–1205. 2007. View Article : Google Scholar : PubMed/NCBI

88 

Hu Y, Scully R, Sobhian B, Xie A, Shestakova E and Livingston DM: RAP80-directed tuning of BRCA1 homologous recombination function at ionizing radiation-induced nuclear foci. Genes Dev. 25:685–700. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Coleman KA and Greenberg RA: The BRCA1-RAP80 complex regulates DNA repair mechanism utilization by restricting end resection. J Biol Chem. 286:13669–13680. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Hu Y, Petit SA, Ficarro SB, Toomire KJ, Xie A, Lim E, Cao SA, Park E, Eck MJ, Scully R, et al: PARP1-driven poly-ADP-ribosylation regulates BRCA1 function in homologous recombination-mediated DNA repair. Cancer Discov. 4:1430–1447. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Hu Z, Mi S, Zhao T, Peng C, Peng Y, Chen L, Zhu W, Yao Y, Song Q, Li X, et al: BGL3 lncRNA mediates retention of the BRCA1/BARD1 complex at DNA damage sites. EMBO J. 39:e1041332020. View Article : Google Scholar : PubMed/NCBI

92 

Wang ZW, Pan JJ, Hu JF, Zhang JQ, Huang L, Huang Y, Liao CY, Yang C, Chen ZW, Wang YD, et al: SRSF3-mediated regulation of N6-methyladenosine modification-related lncRNA ANRIL splicing promotes resistance of pancreatic cancer to gemcitabine. Cell Rep. 39:1108132022. View Article : Google Scholar : PubMed/NCBI

93 

Syed A and Tainer JA: The MRE11-RAD50-NBS1 complex conducts the orchestration of damage signaling and outcomes to stress in DNA replication and repair. Annu Rev Biochem. 87:263–294. 2018. View Article : Google Scholar : PubMed/NCBI

94 

Stracker TH and Petrini JH: The MRE11 complex: Starting from the ends. Nat Rev Mol Cell Biol. 12:90–103. 2011. View Article : Google Scholar : PubMed/NCBI

95 

Xu A, Huang MF, Zhu D, Gingold JA, Bazer DA, Chang B, Wang D, Lai CC, Lemischka IR, Zhao R and Lee DF: LncRNA H19 suppresses Osteosarcomagenesis by regulating snoRNAs and DNA repair protein complexes. Front Genet. 11:6118232020. View Article : Google Scholar

96 

Wu C, Chen W, Yu F, Yuan Y, Chen Y, Hurst DR, Li Y, Li L and Liu Z: Long noncoding RNA HITTERS protects oral squamous cell carcinoma cells from endoplasmic reticulum stress-induced apoptosis via promoting MRE11-RAD50-NBS1 complex formation. Adv Sci (Weinh). 7:20027472020. View Article : Google Scholar : PubMed/NCBI

97 

Paull TT: Mechanisms of ATM Activation. Annu Rev Biochem. 84:711–738. 2015. View Article : Google Scholar : PubMed/NCBI

98 

Zhao K, Wang X, Xue X, Li L and Hu Y: A long noncoding RNA sensitizes genotoxic treatment by attenuating ATM activation and homologous recombination repair in cancers. PLoS Biol. 18:e30006662020. View Article : Google Scholar : PubMed/NCBI

99 

Bunting SF, Callén E, Wong N, Chen HT, Polato F, Gunn A, Bothmer A, Feldhahn N, Fernandez-Capetillo O, Cao L, et al: 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 141:243–254. 2010. View Article : Google Scholar : PubMed/NCBI

100 

Escribano-Díaz C, Orthwein A, Fradet-Turcotte A, Xing M, Young JT, Tkáč J, Cook MA, Rosebrock AP, Munro M, Canny MD, et al: A cell cycle-dependent regulatory circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Mol Cell. 49:872–883. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Zimmermann M, Lottersberger F, Buonomo SB, Sfeir A and de Lange T: 53BP1 regulates DSB repair using Rif1 to control 5' end resection. Science. 339:700–704. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Poulsen M, Lukas C, Lukas J, Bekker-Jensen S and Mailand N: Human RNF169 is a negative regulator of the ubiquitin-dependent response to DNA double-strand breaks. J Cell Biol. 197:189–199. 2012. View Article : Google Scholar : PubMed/NCBI

103 

Hu Q, Botuyan MV, Cui G, Zhao D and Mer G: Mechanisms of Ubiquitin-nucleosome recognition and regulation of 53BP1 chromatin recruitment by RNF168/169 and RAD18. Mol Cell. 66:473–487.e479. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Muvarak N, Kelley S, Robert C, Baer MR, Perrotti D, Gambacorti-Passerini C, Civin C, Scheibner K and Rassool FV: c-MYC generates repair errors via increased transcription of Alternative-NHEJ Factors, LIG3 and PARP1, in tyrosine kinase-activated leukemias. Mol Cancer Res. 13:699–712. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Ahrabi S, Sarkar S, Pfister SX, Pirovano G, Higgins GS, Porter AC and Humphrey TC: A role for human homologous recombination factors in suppressing microhomology-mediated end joining. Nucleic Acids Res. 44:5743–5757. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Leppard JB, Dong Z, Mackey ZB and Tomkinson AE: Physical and functional interaction between DNA ligase IIIalpha and poly(ADP-Ribose) polymerase 1 in DNA single-strand break repair. Mol Cell Biol. 23:5919–5927. 2003. View Article : Google Scholar : PubMed/NCBI

107 

Chiruvella KK, Liang Z and Wilson TE: Repair of double-strand breaks by end joining. Cold Spring Harb Perspect Biol. 5:a0127572013. View Article : Google Scholar : PubMed/NCBI

108 

Hu Y, Lin J, Fang H, Fang J, Li C, Chen W, Liu S, Ondrejka S, Gong Z, Reu F, et al: Targeting the MALAT1/PARP1/LIG3 complex induces DNA damage and apoptosis in multiple myeloma. Leukemia. 32:2250–2262. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Langelier MF, Ruhl DD, Planck JL, Kraus WL and Pascal JM: The Zn3 domain of human poly(ADP-ribose) polymerase-1 (PARP-1) functions in both DNA-dependent poly(ADP-ribose) synthesis activity and chromatin compaction. J Biol Chem. 285:18877–18887. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Huang J, Lin C, Dong H, Piao Z, Jin C, Han H and Jin D: Targeting MALAT1 induces DNA damage and sensitize non-small cell lung cancer cells to cisplatin by repressing BRCA1. Cancer Chemother Pharmacol. 86:663–672. 2020. View Article : Google Scholar : PubMed/NCBI

111 

Goldstein M and Kastan MB: The DNA damage response: Implications for tumor responses to radiation and chemotherapy. Annu Rev Med. 66:129–143. 2015. View Article : Google Scholar

112 

Yao RW, Wang Y and Chen LL: Cellular functions of long noncoding RNAs. Nat Cell Biol. 21:542–551. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Kang M, Ren M, Li Y, Fu Y, Deng M and Li C: Exosome-mediated transfer of lncRNA PART1 induces gefitinib resistance in esophageal squamous cell carcinoma via functioning as a competing endogenous RNA. J Exp Clin Cancer Res. 37:1712018. View Article : Google Scholar : PubMed/NCBI

114 

Xiong XD, Ren X, Cai MY, Yang JW, Liu X and Yang JM: Long non-coding RNAs: An emerging powerhouse in the battle between life and death of tumor cells. Drug Resist Updat. 26:28–42. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Li Z, Zhou Y, Tu B, Bu Y, Liu A and Kong J: Long noncoding RNA MALAT1 affects the efficacy of radiotherapy for esophageal squamous cell carcinoma by regulating Cks1 expression. J Oral Pathol Med. 46:583–590. 2017. View Article : Google Scholar

116 

Sun M, Jin FY, Xia R, Kong R, Li JH, Xu TP, Liu YW, Zhang EB, Liu XH and De W: Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer. BMC Cancer. 14:3192014. View Article : Google Scholar : PubMed/NCBI

117 

Liu J, Ben Q, Lu E, He X, Yang X, Ma J, Zhang W, Wang Z, Liu T, Zhang J and Wang H: Long noncoding RNA PANDAR blocks CDKN1A gene transcription by competitive interaction with p53 protein in gastric cancer. Cell Death Dis. 9:1682018. View Article : Google Scholar : PubMed/NCBI

118 

Shao L, Zuo X, Yang Y, Zhang Y, Yang N, Shen B, Wang J, Wang X, Li R, Jin G, et al: The inherited variations of a p53-responsive enhancer in 13q12.12 confer lung cancer risk by attenuating TNFRSF19 expression. Genome Biol. 20:1032019. View Article : Google Scholar : PubMed/NCBI

119 

Zhen Y, Ye Y, Wang H, Xia Z, Wang B, Yi W and Deng X: Knockdown of SNHG8 repressed the growth, migration, and invasion of colorectal cancer cells by directly sponging with miR-663. Biomed Pharmacother. 116:1090002019. View Article : Google Scholar : PubMed/NCBI

120 

Liu J, Yang C, Gu Y, Li C, Zhang H, Zhang W, Wang X, Wu N and Zheng C: Knockdown of the lncRNA SNHG8 inhibits cell growth in Epstein-Barr virus-associated gastric carcinoma. Cell Mol Biol Lett. 23:172018. View Article : Google Scholar : PubMed/NCBI

121 

Tian X, Liu Y, Wang Z and Wu S: lncRNA SNHG8 promotes aggressive behaviors of nasopharyngeal carcinoma via regulating miR-656-3p/SATB1 axis. Biomed Pharmacother. 131:1105642020. View Article : Google Scholar : PubMed/NCBI

122 

Miao W, Lu T, Liu X, Yin W and Zhang H: LncRNA SNHG8 induces ovarian carcinoma cells cellular process and stemness through Wnt/β-catenin pathway. Cancer Biomark. 28:459–471. 2020. View Article : Google Scholar

123 

Fan D, Qiu B, Yang XJ, Tang HL, Peng SJ, Yang P, Dong YM, Yang L, Bao GQ and Zhao HD: LncRNA SNHG8 promotes cell migration and invasion in breast cancer cell through miR-634/ZBTB20 axis. Eur Rev Med Pharmacol Sci. 24:11639–11649. 2020.PubMed/NCBI

124 

Zhu W, Tan L, Ma T, Yin Z and Gao J: Long noncoding RNA SNHG8 promotes chemoresistance in gastric cancer via binding with hnRNPA1 and stabilizing TROY expression. Dig Liver Dis. 54:1573–1582. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Wang Z, Wang X, Rong Z, Dai L, Qin C, Wang S and Geng W: LncRNA LINC01134 contributes to radioresistance in hepatocellular carcinoma by regulating DNA damage response via MAPK signaling pathway. Front Pharmacol. 12:7918892021. View Article : Google Scholar

126 

Sun Y, Wang J, Ma Y, Li J, Sun X, Zhao X, Shi X, Hu Y, Qu F and Zhang X: Radiation induces NORAD expression to promote ESCC radiotherapy resistance via EEPD1/ATR/Chk1 signalling and by inhibiting pri-miR-199a1 processing and the exosomal transfer of miR-199a-5p. J Exp Clin Cancer Res. 40:3062021. View Article : Google Scholar : PubMed/NCBI

127 

Yao P, Li Y, Shen W, Xu X, Zhu W, Yang X, Cao J and Xing C: ANKHD1 silencing suppresses the proliferation, migration and invasion of CRC cells by inhibiting YAP1-induced activation of EMT. Am J Cancer Res. 8:2311–2324. 2018.PubMed/NCBI

128 

Yao PA, Wu Y, Zhao K, Li Y, Cao J and Xing C: The feedback loop of ANKHD1/lncRNA MALAT1/YAP1 strengthens the radioresistance of CRC by activating YAP1/AKT signaling. Cell Death Dis. 13:1032022. View Article : Google Scholar : PubMed/NCBI

129 

Takahashi H, Nishimura J, Kagawa Y, Kano Y, Takahashi Y, Wu X, Hiraki M, Hamabe A, Konno M, Haraguchi N, et al: Significance of Polypyrimidine Tract-binding Protein 1 expression in colorectal cancer. Mol Cancer Ther. 14:1705–1716. 2015. View Article : Google Scholar : PubMed/NCBI

130 

Huan L, Guo T, Wu Y, Xu L, Huang S, Xu Y, Liang L and He X: Hypoxia induced LUCAT1/PTBP1 axis modulates cancer cell viability and chemotherapy response. Mol Cancer. 19:112020. View Article : Google Scholar : PubMed/NCBI

131 

Jin MH and Oh DY: ATM in DNA repair in cancer. Pharmacol Ther. 203:1073912019. View Article : Google Scholar : PubMed/NCBI

132 

Cimprich KA and Cortez D: ATR: An essential regulator of genome integrity. Nat Rev Mol Cell Biol. 9:616–627. 2008. View Article : Google Scholar : PubMed/NCBI

133 

Panzarino NJ, Krais JJ, Cong K, Peng M, Mosqueda M, Nayak SU, Bond SM, Calvo JA, Doshi MB, Bere M, et al: Replication gaps underlie BRCA deficiency and therapy response. Cancer Res. 81:1388–1397. 2021. View Article : Google Scholar

134 

Zhang B, Bao W, Zhang S, Chen B, Zhou X, Zhao J, Shi Z, Zhang T, Chen Z, Wang L, et al: LncRNA HEPFAL accelerates ferroptosis in hepatocellular carcinoma by regulating SLC7A11 ubiquitination. Cell Death Dis. 13:7342022. View Article : Google Scholar : PubMed/NCBI

135 

Jiang Y, Guo H, Tong T, Xie F, Qin X, Wang X, Chen W and Zhang J: lncRNA lnc-POP1-1 upregulated by VN1R5 promotes cisplatin resistance in head and neck squamous cell carcinoma through interaction with MCM5. Mol Ther. 30:448–467. 2022. View Article : Google Scholar :

136 

Choi PS and Thomas-Tikhonenko A: RNA-binding proteins of COSMIC importance in cancer. J Clin Invest. 131:e1516272021. View Article : Google Scholar : PubMed/NCBI

137 

Fabbri L, Chakraborty A, Robert C and Vagner S: The plasticity of mRNA translation during cancer progression and therapy resistance. Nat Rev Cancer. 21:558–577. 2021. View Article : Google Scholar : PubMed/NCBI

138 

Duffy AG, Makarova-Rusher OV, Ulahannan SV, Rahma OE, Fioravanti S, Walker M, Abdullah S, Raffeld M, Anderson V, Abi-Jaoudeh N, et al: Modulation of tumor eIF4E by antisense inhibition: A phase I/II translational clinical trial of ISIS 183750-an antisense oligonucleotide against eIF4E-in combination with irinotecan in solid tumors and irinotecan-refractory colorectal cancer. Int J Cancer. 139:1648–1657. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Shen L and Pelletier J: Selective targeting of the DEAD-box RNA helicase eukaryotic initiation factor (eIF) 4A by natural products. Nat Prod Rep. 37:609–616. 2020. View Article : Google Scholar

140 

Zhu H, Chen K, Chen Y, Liu J, Zhang X, Zhou Y, Liu Q, Wang B, Chen T and Cao X: RNA-binding protein ZCCHC4 promotes human cancer chemoresistance by disrupting DNA-damage-induced apoptosis. Signal Transduct Target Ther. 7:2402022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 52 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zou S, Gou X and Wen K: Advances in the role of long non‑coding RNAs and RNA‑binding proteins in regulating DNA damage repair in cancer cells. Int J Mol Med 52: 93, 2023
APA
Zou, S., Gou, X., & Wen, K. (2023). Advances in the role of long non‑coding RNAs and RNA‑binding proteins in regulating DNA damage repair in cancer cells. International Journal of Molecular Medicine, 52, 93. https://doi.org/10.3892/ijmm.2023.5296
MLA
Zou, S., Gou, X., Wen, K."Advances in the role of long non‑coding RNAs and RNA‑binding proteins in regulating DNA damage repair in cancer cells". International Journal of Molecular Medicine 52.4 (2023): 93.
Chicago
Zou, S., Gou, X., Wen, K."Advances in the role of long non‑coding RNAs and RNA‑binding proteins in regulating DNA damage repair in cancer cells". International Journal of Molecular Medicine 52, no. 4 (2023): 93. https://doi.org/10.3892/ijmm.2023.5296