RAD52 motif‑containing protein 1 promotes non‑small cell lung cancer cell proliferation and survival via cell cycle regulation

  • Authors:
    • Guihua Xu
    • Jiya Du
    • Feng Wang
    • Fan Zhang
    • Ruimin Hu
    • Dejun Sun
    • Jie Shen
  • View Affiliations

  • Published online on: May 23, 2018     https://doi.org/10.3892/or.2018.6459
  • Pages: 833-840
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA repair proteins such as RAD52 have been implicated in tumor progression and response to chemotherapy. RAD52 motif‑containing protein 1 (RDM1) has been implicated in the response to chemotherapeutic agent cisplatin; however, its function in lung cancer progression remains unclear. This study aimed to investigate the role of RDM1 in the progression of non‑small cell lung cancer (NSCLC). We found elevated RDM1 mRNA and protein expression in NSCLC tissues and cell lines compared to levels in normal lung cells. RDM1 protein expression in lung cancer tissues was found to correlate with tumor size, histological differentiation, lymph node metastasis and tumor‑node‑metastasis (TNM) stage. Knockdown of the RDM1 gene with siRNA significantly reduced the cellular proliferation rate and increased apoptosis in the human NSCLC cell line, NCI‑H1299. Compared to wild‑type NCI‑H1299 cells, RDM1 knockdown enhanced the activity of caspase‑3 and caspase‑7, and decreased the proportion of cells in the S‑phase of the cell cycle. Taken together, these data imply that RDM1 promotes the survival and proliferation of NSCLC cells. Due to its similarity to RAD52, we hypothesized that RDM1 potentially repairs DNA double‑strand breaks arising through DNA replication, thereby preventing G2/M cell cycle arrest. Accordingly, specific targeting of RDM1 may be a novel therapeutic strategy in the treatment of NSCLC.

Introduction

Lung cancer is a leading cause of tumor-related morbidity and mortality worldwide (1,2). Despite recent breakthroughs in the diagnosis and treatment of this disease, the prognosis of patients remains poor, especially for those with metastatic disease (2). Standard treatment for lung cancer includes traditional chemotherapeutic agents such as cisplatin and docetaxel; however, their efficacy is somewhat hindered by the development of drug resistance (3). In the case of non-small cell lung cancer (NSCLC), newer targeted-molecular therapies have shown great promise (4,5), yet the majority of patients with advanced disease still develop drug resistance within one year (6). Therefore, there is a pressing need to develop new therapeutic options in the fight against lung cancer, and to better understand the mechanisms leading to drug resistance.

Drug resistance has been linked to changes in the DNA repair capacity within tumor cells (7). Chemotherapeutic drugs such as cisplatin induce DNA damage in tumor cells, and, in turn, activate the DNA damage response. Activation of the DNA damage response by chemotherapeutic drugs can have the desired effect of tumor cell apoptosis, or the cells may repair the lesion, leading to chemoresistance.

Components of the DNA damage response pathway have not only been implicated in chemoresistance, but they themselves may play oncogenic roles (8,9). Indeed, the capacity to repair damaged DNA through homologous recombination has previously been implicated in patient susceptibility to lung cancer (10,11). For example, NSCLC patients expressing high levels of RAD51, a protein involved in homologous recombination and DNA repair, had significantly poorer 5-year survival rates compared to those with low RAD51 expression (12). Similarly, the DNA repair protein RAD52 was implicated as an oncogene in lung squamous cell carcinoma, with enhanced tumor cell death observed in mouse cells ablated of RAD52 (9,13). Therefore, it is worth exploring components of the DNA damage response pathway, such as RAD1/2, as antitumor drug targets in lung cancer.

In the present study, we examined the role of RAD52 motif-containing protein 1 (RDM1) in NSCLC. The RDM1 gene was identified through a database search looking for proteins similar to RAD52 (14). Like RAD52, RDM1 is involved in DNA double-strand break repair and homologous recombination (14,15). Previous research has shown that RDM1 binds to cisplatin-damaged DNA in vitro, suggesting its possible role in the development of chemoresistance (14). As further evidence, RDM1 ablation in a chicken B cell line DT40 resulted in a more than 3-fold increase in cisplatin sensitivity (14). However, RDM1-knockout cells were not hypersensitive to DNA damage caused by ionizing radiation, ultraviolet irradiation, or alkylation (14). Therefore, the precise role of RDM1 in cisplatin resistance remains unclear.

Subsequent research has shown that RDM1 expression is modulated by heat shock (16). More recent studies revealed that the DNA-binding capability of RDM1 is enhanced at low pH, indicating that it may function in acidic conditions such as the tumor microenvironment (17). Furthermore, RDM1 has been shown to be upregulated at the mRNA level in lung squamous cell carcinoma (18). Nonetheless, despite the growing evidence, the precise function of RDM1 in NSCLC remains largely unknown. Therefore, this study aimed to clarify the role of RDM1 in NSCLC.

Materials and methods

Patients and tissue specimens

Paraffin-embedded tumors and adjacent non-cancerous tissue specimens were collected from a series of 103 patients with NSCLC who had only undergone surgical resection, but had not received chemotherapy between January 1, 2015 and 31 December 31, 2016 at the Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China. The study group consisted of 77 men (75%) and 26 women (25%), and the mean age was 60.7 years (range, 32–81 years). The specimens were classified by their tumor-node-metastasis (TNM) stage (19). Detailed patient information including gender, age, tumor size, histological differentiation, and lymph node metastasis is documented in Table I. The study was approved by the Medical Ethics Committee of Inner Mongolia Autonomous Region People's Hospital, and all participants provided written, informed consent.

Table I.

Association between RDM1 expression and the clinicopathological characteristics of the NSCLC cases.

Table I.

Association between RDM1 expression and the clinicopathological characteristics of the NSCLC cases.

RDM1 expression

Clinicopathological parametersTotalNegative (n=45)Positive (n=58)P-value
Age (mean ± SD, years)10361.8±10.259.6±11.70.368
Gender, n (%)
  Male7732 (41.5)45 (58.5)0.447
  Female2613 (50)13 (50)
Tumor size (cm), n (%)
  ≤33827 (71.1)11 (28.9)0.007a
  >36518 (27.7)47 (72.3)
Histological differentiation, n (%)
  Well75 (71.4)2 (28.6)0.013a
  Moderate3519 (54.3)16 (45.7)
  Poor6121 (34.4)40 (65.6)
Histological classification, n (%)
  Squamous carcinoma61 (59)24 (39.3)37 (61.7)0.653
  Adenocarcinoma12 (12)7 (58.3)5 (41.7)
  Adenosquamous10 (10)4 (40)6 (60)
  Others16 (3)9 (56.2)7 (43.8)
Lymph node metastasis, n (%)
  Absent4224 (57.1)18 (42.9)0.034a
  Present6121 (34.4)40 (65.6)
TNM stage, n (%)
  I+II5937 (62.7)22 (37.3)0.004a
  III+IV448 (18.2)36 (81.8)
1-year survival rate, n (%)48 (46.6)31 (68.9)16 (27.6)0.045a

a P<0.05, statistical significance. RDM1, RAD52 motif-containing protein 1; NSCLC, non-small cell lung cancer; TNM, tumor-node-metastasis.

Immunohistochemistry

Paraffin-embedded 4-µm tissue sections were stained with the anti-RDM1 antibody. In brief, deparaffinized tissue sections were processed for antigen retrieval using target retrieval solution (pH 9.0; Dako; Agilent Technologies, Inc., Santa Clara, CA, USA) at 120°C for 4 min. Non-specific immunoreactions were blocked at room temperature for 30 min using the Dako Protein-Block kit (Agilent Technologies, Inc.). Sections were incubated at 4°C overnight with the Invitrogen RDM1 polyclonal primary antibody (1:100; cat. no. PA5-50103; Thermo Fisher Scientific, Inc., Waltham, MA, US)A, followed by incubation with 3,3′-diaminobenzidine tetrahydrochloride (Dako; Agilent Technologies, Inc.) for 5 min. Nuclear counter staining was conducted with hematoxylin. Images were captured using a light microscope (Olympus BX50; Olympus Corp., Tokyo, Japan) and processed using Photoshop software (version 7.0; Adobe, San Jose, CA, USA).

Cell culture

Human lung cancer cell lines, including NCI-H1299, A549, H1688 and H1975, were obtained from the Cell Bank of the Chinese Academy of Science (Shanghai, China). Human normal lung epithelial BEAS-2B cells were used as control. Cells were cultured in Gibco™ RPMI-1640 medium (Thermo Fisher Scientific, Inc.), containing 10% HyClone™ fetal bovine serum (FBS; GE Healthcare Bio-Sciences, Pittsburgh, PA, USA) and 100 U/ml penicillin/streptomycin. All cells were incubated at 37°C in a humidified atmosphere containing 5% CO2.

Knockdown of RDM1 by lentivirus

The target sequence for RDM1-specific shRNA (shRDM1: 5′-GGCCCATCCTGGTTTCTAT-3′) and the negative control shRNA (shCtrl: 5′-TTCTCCGAACGTGTCACGT-3′) were obtained from GeneChem Biotech (Shanghai, China). The NCI-H1299 cell line was transfected with shRDM1 or shCtrl using transfection reagents (c1507; Applygen Technologies, Inc., Beijing, China) according to the manufacturer's instructions.

Western blot analysis. Lung tissue samples and human lung cancer cell lines were homogenized in RIPA buffer (50 mmol/l Tris-HCl, 150 mmol/l NaCl, 0.5% sodium deoxycholate, 1% NP-40, 0.1% SDS, 2 mmol/l Na3VO4, 500 mmol/l NaF, 58 µg/ml aprotinin, 10 µg/ml leupeptin, and 2 mmol/l phenylmethylsulfonyl fluoride) using the Multi Beads Shocker system (Yasui-Kikai, Osaka, Japan) at 4°C. Protein samples (15 µg) were separated by SDS-PAGE using a gradient gel (5–20%; ATTO, Tokyo, Japan), followed by electroblotting onto polyvinylidene difluoride membranes (ATTO) and incubation in a blocking buffer of 5% non-fat milk for 1 h at room temperature.

Membranes were incubated with the following primary antibodies: rabbit anti-RDM1 (1:1,000; cat. no. ab201293; Abcam) and mouse anti-glyceraldehyde 3-phosphate dehydrogenase (GAPDH; 1:5,000; Chemicon, Temecula, CA, USA) at 4°C overnight. Membranes were visualized by incubation with appropriate horseradish peroxidase-linked secondary antibodies, followed by enhanced chemiluminescence (ECL) detection using ECL Plus western blotting detection reagents (Amersham Biosciences, Little Chalfont, UK). The immunoreactive bands of targeted proteins were quantified by VH analyzer software (VH-H1A5; Keyence Corp., Osaka, Japan), normalized by GAPDH levels, and determined relative to the sham values. All experiments were repeated three times.

Quantitative polymerase chain reaction (qPCR)

Total RNA was extracted from lung tissue samples and human lung cancer cell lines using the RNeasy Mini Kit (Qiagen Sciences, Inc., Gaithersburg, MD, USA), purified with the RNase-Free DNase Set (Qiagen), and used as a template for cDNA synthesis by the PrimeScript RT Reagent Kit (Takara, Shiga, Japan). PCR reactions were performed in a Thermal Cycler Dice Real Time System Tp800 (Takara) using SYBR Premix ex Taq (Takara) according to the manufacturer's instructions as follows: 30 sec at 95°C, then 40 cycles of 5 sec at 95°C and 30 sec at 60°C; then for the dissociation stage, 15 sec at 95°C, 30 sec at 60°C, and 15 sec at 95°C. The sequences of primers were: RDM1 forward primer, 5′-GCCCATCCTGGTTTCTATGCC-3′ and reverse primer, 5′-AGACGAACCTTGACTGGAGAT-3′; GAPDH forward primer, 5′-TGACTTCAACAGCGACACCCA-3′ and reverse primer, 5′-CACCCTGTTGCTGTAGCCAAA-3′. Relative gene expression data were determined by the standard curve method, and fold-changes were normalized by GAPDH, and relative to control values. Each sample was tested in triplicate.

Detection of cell proliferation

The NCI-H1299 cell line was seeded into 96-well plates at a density of 2000 cells per well, and infected with the control or RDM1-siRNA lentivirus. Cell numbers were then counted via Celigo S Image Cytometer (Nexcelom Bioscience, Ltd., Manchester, UK) every day for 5 days to determine the growth rate. All samples were assayed in triplicate.

Detection of caspase-3/-7 activity

After lentivirus infection, caspase-3/-7 activity was detected using the Caspase-Glo® 3/7 Assay (G8091; Promega, Madison, WI, USA) according to the manufacturer's instructions. In brief, the NCI-H1299 cell line was seeded into 96-well plates at a density of 2,000 cells per well, and harvested when the confluence reached 80%. Caspase-Glo® 3/7 Reagent (100 µl) was added to each well of a white-walled 96-well plate containing 100 µl of blank, negative control cells, or treated cells in culture medium, and incubated in the dark at room temperature for 30 min to 3 h. The reaction product was detected at 405 nm using a plate-reading luminometer (M2009PR, Tecan Infinite, Switzerland). All samples were assayed in triplicate.

Determination of apoptosis by flow cytometry

The Invitrogen™ eBioscience™ Annexin V Apoptosis Detection Kit APC (88–8007; Thermo Fisher Scientific, Inc.) was used for labeling of apoptotic cells according to the manufacturer's protocol. The cells from each group were collected after lentivirus infection, washed twice with ice-cold phosphate-buffered saline (PBS) and resuspended in 200 µl binding buffer containing 10 µl Annexin V-APC. Fluorescence intensity was measured by flow cytometry (BD Biosciences). All samples were assayed in triplicate.

Cell cycle analysis by flow cytometry

After lentivirus infection, the NCI-H1299 cell line was seeded into 6-well plates at a density of 2×105 cells per well, and harvested when the confluence reached 80%. The collected cells were fixed in 75% cold ethanol (10009269; Sinopharm Chemical Reagent, Co., Ltd., Shanghai, China) for 1 h at 4°C. The cells were then washed with cold PBS and stained with 500 µl propidium iodide (PI) buffer (P4170 Sigma; Merck KGaA, Darmstadt, Germany) for 1 h at 37°C in the dark. The cell cycle stages were analyzed by flow cytometry. All samples were assayed in triplicate.

Statistical analysis

All experiments were repeated three times. SPSS software v. 17.0 (SPSS, Inc., Chicago, IL, USA) was used to analyze the results using either Student's t-test or one-way analysis of variance. A P-value <0.05 was considered statistically significant.

Results

RDM1 is highly expressed in lung cancer tissues and cell lines

Using immunohistochemistry, we found that RDM1 protein expression was significantly elevated in the cell nuclei of lung cancer tissues compared to that in adjacent non-cancerous tissue (Fig. 1A and B). We also found that expression of RDM1 mRNA was elevated in lung cancer cell lines (NCI-H1299, A549, H1688 and H1975) when compared with that noted in the human normal lung epithelial BEAS-2B cells (Fig. 1C). Therefore, the expression of RDM1 appears to be upregulated in lung cancer.

RDM1 expression correlates with tumor size, histological differentiation, lymph node metastasis and TNM staging in lung cancer

We then assessed the correlation between RDM1 expression and the clinicopathological parameters of lung cancer patients. Among the 103 tissue samples obtained from NSCLC patients, 58 showed positive RDM1 protein expression and 45 were negative. As shown in Table I, RDM1 expression in NSCLC tissues was significantly correlated with tumor size, histological differentiation, the degree of lymph node metastasis, TNM stage and 1-year survival rate (all P<0.05); however, it was not directly associated with age, gender or histological classification.

RDM1 knockdown significantly reduces the cellular proliferation rate and increases apoptosis in a lung cancer cell line

To investigate the role of RDM1 in vitro, we knocked down RDM1 in a human NSCLC cell line (NCI-H1299) using lentiviral vector-expressed siRNA. NCI-H1299 cells infected with RDM1-siRNA showed significantly reduced RDM1 mRNA and protein levels at day 3 post-infection compared to the controls (P<0.01; Fig. 2).

We then monitored the effect of RDM1 knockdown on lung cancer cell growth and apoptosis. We found that NCI-H1299 cells transfected with RDM1-siRNA showed a significant reduction in the cell growth rate over 5 days compared to the controls (P<0.01; Fig. 3). In addition, the activity of caspase-3/-7 in NCI-H1299 cells infected with RDM1-siRNA was enhanced compared to that noted in the controls (P<0.01; Fig. 4A). Flow cytometric analysis confirmed there was a significant increase in the apoptosis rate in the NCI-H1299 cells in which RDM1 had been knocked down (P<0.01; Fig. 4B). Therefore, RDM1 promoted cell proliferation and inhibited apoptosis in a lung cancer cell line.

RDM1 regulates the cell cycle

Next, we examined the effect of RDM1 knockdown on the cell cycle using flow cytometry. RDM1 knockdown resulted in a significant increase in the proportion of lung cancer cells in the G1 and G2/M phases and a decreased proportion in the S phase (P<0.01; Fig. 5). These results suggest that RDM1 may be involved in DNA repair in lung cancer cells, thereby promoting progression through the DNA damage checkpoint (G2/M) of the cell cycle and avoiding apoptosis.

Discussion

The present study aimed to clarify the role of RAD52 motif-containing protein 1 (RDM1) in non-small cell lung cancer (NSCLC). We found elevated expression of RDM1 at both the mRNA and protein levels in NSCLC tissues and cell lines. To the best of our knowledge, this study first reported that RDM1 protein expression in NSCLC tissues correlates with tumor size, histological differentiation, lymph node metastasis, TNM stage and 1-year survival rate. Meanwhile, RDM1 knockdown in a human NSCLC cell line significantly reduced the cellular proliferation rate and increased apoptosis. In addition, RDM1 knockdown enhanced the activity of caspase-3/-7. Finally, we provide evidence that RDM1 may function as a DNA repair protein that promotes cell cycle progression of lung cancer cells. Taken together, our results indicate that RDM1 could be a molecular marker of NSCLC and may represent a novel drug target.

DNA repair proteins, such as RAD52 and RDM1, have previously been implicated in the development of resistance to cancer therapy, including resistance to platinum-based chemotherapy (14,15,20) and radiotherapy (21). In addition to drug resistance, proteins involved in DNA repair pathways have been implicated in tumor progression (22). For example, RAD52 has been linked to both the progression and risk of small cell lung cancer (SCLC) and NSCLC (8,9,23). In addition, RDM1 was previously shown to undergo stress-induced nucleolar accumulation, indicating it may function in the heat-shock response that is implicated in tumorigenesis (16).

As further evidence of the role of RDM1 in lung cancer progression, here we showed that RDM1 expression was correlated with tumor size and metastasis in NSCLC patients. In addition, we found that increased RDM1 expression promoted cell growth and reduced apoptosis in an NSCLC cell line. This indicates that RDM1 plays a similar role to RAD52 in promoting the progression of lung cancer, through increasing cell survival and proliferation. However, the mechanism by which RDM1 promotes cell proliferation requires further investigation.

A previous study found an abundance of RDM1 transcripts in the testis, which implies that RDM1 may play a possible role in spermatogenesis (14). During spermatogenesis, immature sperm cells undergo rounds of mitotic and meiotic division and the control of genetic stability is paramount. We hypothesize that RDM1 may have a similar function in controlling genomic stability in NSCLC cells, which allows the cells to proliferate. Indeed, we found that knockdown of RDM1 resulted in a decreased proportion of NSCLC cells in the S phase, with an increased proportion in the G1 and G2/M phases. This indicates that the cells were arrested at the G2/M DNA damage checkpoint, which serves to protect genomic integrity and prevent cells with damaged DNA from entering the S phase. In turn, cells with unrepairable DNA lesions undergo permanent arrest or apoptosis, whereas if the damage is repaired, for example by RDM1, checkpoint-arrested cells may resume cell cycle progression (24). Therefore, RDM1 appears to promote cell cycle progression, likely through repairing DNA double-strand breaks.

Our results are similar to those previously observed for the related DNA repair protein, RAD52, in lung cancer (8,9). RAD52 is predominantly recruited for DNA repair during the S phase of the cell cycle and plays a crucial role in the regulation of homologous recombination-related genomic instability in humans (25). Indeed, RAD52 has been shown to promote mitotic DNA synthesis following replication stress, such as that occurring during tumor proliferation (26). In particular, RAD52 appears to repair collapsed DNA replication forks in cancer cells (27). In addition, the recruitment of Rad52 to stalled DNA replication forks in yeast has been shown to be controlled by various checkpoint and cell cycle signals (28). Therefore, the recruitment of RDM1 in proliferating lung cancer cells may be under similar control by cell cycle regulators or signals.

Similar findings have been made for RAD51. Inhibition of RAD51 transcription in a chicken DT40 cell line resulted in a high G2/M phase arrest, with high levels of chromosome-type breaks (29). Conversely, overexpression of RAD51 protein in tumor cells was associated with high DNA repair capacity and elevated recombination rates (30). Therefore, together, these results imply that lung cancer cells may specifically overexpress and recruit DNA repair proteins such as RDM1, RAD51 and RAD52 in order to generate genetically stable clones suitable for sustained proliferation.

While inhibiting RDM1 seems a promising therapeutic option for NSCLC, we must remember that DNA repair proteins are multifunctional. Therefore, any RDM1 inhibitor that is developed may have unwanted side effects or toxicities. Furthermore, multiple isoforms of RDM1 have been identified, each with a different subcellular location, and potentially possessing different functions (16). Thus, unraveling the specific functions of each of these isoforms will be key for reducing any off-target effects. Indeed, specific inhibitors of proteins involved in homologous recombination for the treatment of cancer remain in their infancy (22). Nonetheless, better characterization of DNA repair proteins, such as that performed here for RDM1, will aid in the discovery of not only more effective DNA repair inhibitors, but also more efficient anticancer combination therapies that overcome the issues of drug resistance.

In summary, our data revealed that RDM1 plays a pivotal role in the survival and proliferation of NSCLC cells by regulating the cell cycle, likely through its DNA repair capabilities. Further studies are warranted to clarify the role of RDM1 in various subtypes of NSCLC, as well as to determine its prognostic and therapeutic value.

Acknowledgements

We thank Haiyan Zhao, Zhiqiang Wang for the technical assistance (Inner Mongolia Medical University).

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81560013) and the Hospital Fund from Inner Mongolia Autonomous Region People's Hospital (grant no. 201712), Inner Mongolia Autonomous Region, China.

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

GX, DS and JS conceived and designed the study. GX, JD, EW, FZ and RH performed the experiments. GX, DS and JS wrote the paper. GX, JD, EW, FZ, RH, DS and JS reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Medical Ethics Committee of Inner Mongolia Autonomous Region People's Hospital, and all participants provided written, informed consent.

Consent for publication

Not applicable.

Competing interests

The authors state that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

GAPDH

glyceraldehyde 3-phosphate dehydrogenase

NSCLC

non-small cell lung cancer

qPCR

quantitative polymerase chain reaction

RDM1

RAD52 motif-containing protein 1

NSCLC

non-small cell lung cancer

TNM

tumor-node-metastasis

References

1 

Ridge CA, McErlean AM and Ginsberg MS: Epidemiology of lung cancer. Semin Intervent Radiol. 30:93–98. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Torre LA, Siegel RL and Jemal A: Lung cancer statistics. Adv Exp Med Biol. 893:1–19. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Calikusu Z, Yildirim Y, Akcali Z, Sakalli H, Bal N, Unal I and Ozyilkan O: The effect of HER2 expression on cisplatin-based chemotherapy in advanced non-small cell lung cancer patients. J Exp Clin Cancer Res. 28:972009. View Article : Google Scholar : PubMed/NCBI

4 

Chan BA and Hughes BG: Targeted therapy for non-small cell lung cancer: Current standards and the promise of the future. Transl Lung Cancer Res. 4:36–54. 2015.PubMed/NCBI

5 

Shtivelman E, Hensing T, Simon GR, Dennis PA, Otterson GA, Bueno R and Salgia R: Molecular pathways and therapeutic targets in lung cancer. Oncotarget. 5:1392–1433. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Maione P, Sacco PC, Sgambato A, Casaluce F, Rossi A and Gridelli C: Overcoming resistance to targeted therapies in NSCLC: Current approaches and clinical application. Ther Adv Med Oncol. 7:263–273. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Salehan MR and Morse HR: DNA damage repair and tolerance: A role in chemotherapeutic drug resistance. Br J Biomed Sci. 70:31–40. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Lieberman R and You M: Corrupting the DNA damage response: A critical role for Rad52 in tumor cell survival. Aging. 9:1647–1659. 2017.PubMed/NCBI

9 

Lieberman R, Pan J, Zhang Q and You M: Rad52 deficiency decreases development of lung squamous cell carcinomas by enhancing immuno-surveillance. Oncotarget. 8:34032–34044. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Rajaee-Behbahani N, Schmezer P, Risch A, Rittgen W, Kayser KW, Dienemann H, Schulz V, Drings P, Thiel S and Bartsch H: Altered DNA repair capacity and bleomycin sensitivity as risk markers for non-small cell lung cancer. Int J Cancer. 95:86–91. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Spitz MR, Wei Q, Dong Q, Amos CI and Wu X: Genetic susceptibility to lung cancer: The role of DNA damage and repair. Cancer Epidemiol Biomarkers Prev. 12:689–698. 2003.PubMed/NCBI

12 

Qiao GB, Wu YL, Yang XN, Zhong WZ, Xie D, Guan XY, Fischer D, Kolberg HC, Kruger S and Stuerzbecher HW: High-level expression of Rad51 is an independent prognostic marker of survival in non-small-cell lung cancer patients. Br J Cancer. 93:137–143. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Benson FE, Baumann P and West SC: Synergistic actions of Rad51 and Rad52 in recombination and DNA repair. Nature. 391:401–404. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Hamimes S, Arakawa H, Stasiak AZ, Kierzek AM, Hirano S, Yang YG, Takata M, Stasiak A, Buerstedde JM and Van Dyck E: RDM1, a novel RNA recognition motif (RRM)-containing protein involved in the cell response to cisplatin in vertebrates. J Biol Chem. 280:9225–9235. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Hamimes S, Bourgeon D, Stasiak AZ, Stasiak A and Van Dyck E: Nucleic acid-binding properties of the RRM-containing protein RDM1. Biochem Biophys Res Commun. 344:87–94. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Messaoudi L, Yang YG, Kinomura A, Stavreva DA, Yan G, Bortolin-Cavaillé ML, Arakawa H, Buerstedde JM, Hainaut P, Cavaillé J, et al: Subcellular distribution of human RDM1 protein isoforms and their nucleolar accumulation in response to heat shock and proteotoxic stress. Nucleic Acids Res. 35:6571–6587. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Stubbs M, McSheehy PM, Griffiths JR and Bashford CL: Causes and consequences of tumour acidity and implications for treatment. Mol Med Today. 6:15–19. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Zhang F, Chen X, Wei K, Liu D, Xu X, Zhang X and Shi H: Identification of key transcription factors associated with lung squamous cell carcinoma. Med Sci Monit. 23:172–206. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WE, Nicholson AG, Groome P, Mitchell A, Bolejack V, et al: The IASLC lung cancer staging project: Proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 11:39–51. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Li HM, Yuan P, Yu D, Ma F, Tan WW, Feng T, Yang J, Huang Y, Lin DX, Xu BH and Tan W: Genetic variation in DNA repair gene RAD52 is associated with the response to platinum-based chemotherapy in SCLC patients. Zhonghua Zhong Liu Za Zhi. 38:504–509. 2016.(In Chinese). PubMed/NCBI

21 

Ghosh S and Krishna M: Role of Rad52 in fractionated irradiation induced signaling in A549 lung adenocarcinoma cells. Mutat Res. 729:61–72. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Kelley MR, Logsdon D and Fishel ML: Targeting DNA repair pathways for cancer treatment: What's new? Future Oncol. 10:1215–1237. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Han S, Gao F, Yang W, Ren Y, Liang X, Xiong X, Pan W, Zhou L, Zhou C, Ma F and Yang M: Identification of an SCLC susceptibility rs7963551 genetic polymorphism in a previously GWAS-identified 12p13.33 RAD52 lung cancer risk locus in the Chinese population. Int J Clin Exp Med. 8:16528–16535. 2015.PubMed/NCBI

24 

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

25 

Lok BH and Powell SN: Molecular pathways: Understanding the role of Rad52 in homologous recombination for therapeutic advancement. Clin Cancer Res. 18:6400–6406. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Bhowmick R, Minocherhomji S and Hickson ID: RAD52 facilitates mitotic DNA synthesis following replication stress. Mol Cell. 64:1117–1126. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Sotiriou SK, Kamileri I, Lugli N, Evangelou K, Da-Ré C, Huber F, Padayachy L, Tardy S, Nicati NL, Barriot S, et al: Mammalian RAD52 functions in break-induced replication repair of collapsed DNA replication forks. Mol Cell. 64:1127–1134. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Barlow JH and Rothstein R: Timing is everything: Cell cycle control of Rad52. Cell Div. 5:72010. View Article : Google Scholar : PubMed/NCBI

29 

Sonoda E, Sasaki MS, Buerstedde JM, Bezzubova O, Shinohara A, Ogawa H, Takata M, Yamaguchi-Iwai Y and Takeda S: Rad51-deficient vertebrate cells accumulate chromosomal breaks prior to cell death. EMBO J. 17:598–608. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Henning W and Sturzbecher HW: Homologous recombination and cell cycle checkpoints: Rad51 in tumour progression and therapy resistance. Toxicology. 193:91–109. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 40 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu G, Du J, Wang F, Zhang F, Hu R, Sun D and Shen J: RAD52 motif‑containing protein 1 promotes non‑small cell lung cancer cell proliferation and survival via cell cycle regulation. Oncol Rep 40: 833-840, 2018
APA
Xu, G., Du, J., Wang, F., Zhang, F., Hu, R., Sun, D., & Shen, J. (2018). RAD52 motif‑containing protein 1 promotes non‑small cell lung cancer cell proliferation and survival via cell cycle regulation. Oncology Reports, 40, 833-840. https://doi.org/10.3892/or.2018.6459
MLA
Xu, G., Du, J., Wang, F., Zhang, F., Hu, R., Sun, D., Shen, J."RAD52 motif‑containing protein 1 promotes non‑small cell lung cancer cell proliferation and survival via cell cycle regulation". Oncology Reports 40.2 (2018): 833-840.
Chicago
Xu, G., Du, J., Wang, F., Zhang, F., Hu, R., Sun, D., Shen, J."RAD52 motif‑containing protein 1 promotes non‑small cell lung cancer cell proliferation and survival via cell cycle regulation". Oncology Reports 40, no. 2 (2018): 833-840. https://doi.org/10.3892/or.2018.6459