Role of RNA‑binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer (Review)

  • Authors:
    • Yanling Xu
    • Zhenzhong Su
    • Junyao Li
    • Qi Wang
    • Guangping Meng
    • Yu Zhang
    • Wen Yang
    • Jie Zhang
    • Peng Gao
  • View Affiliations

  • Published online on: December 10, 2018     https://doi.org/10.3892/ol.2018.9818
  • Pages: 2013-2019
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer remains one of the leading causes of cancer‑associated mortality in the world. Lung carcinogenesis is frequently associated with deletions or the loss of heterozygosity at the critical chromosomal region 3p21.3, where RNA‑binding protein 5 (RBM5) is localized. RBM5 regulates cell growth, cell cycle progression and apoptosis in cell homeostasis. In the lungs, altered RBM5 protein expression leads to alterations in cell growth and apoptosis, with subsequent lung pathogenesis and varied responses to treatment in patients with lung cancer. Detection of RBM5 expression may be a tumor marker for diagnosis, prediction and treatment response in lung cancer, and may be developed as a potential therapeutic target for drug resistant lung cancer. This review discusses the most recent progress on the role of RBM5 in lung cancer.

Introduction

Lung cancer is one of the most common cancer types and one of the leading causes of cancer-associated mortality worldwide. In 2012, lung cancer accounted for ~13% of all cancer cases and 26% of cancer-associated mortality, according to recent data (1). In China alone, there were 733,000 new lung cancer cases diagnosed in 2011 (17% of all new cancer cases), and 600,200 lung cancer-associated mortalities (22% of all cancer-associated mortalities) (2). Histologically, lung cancer is classified as small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), the latter of which accounts for up to 85% of all lung cancer cases (3). NSCLC may be further sub-categorized as adenocarcinoma, squamous cell carcinoma, large cell carcinoma and numerous other less common types of cancer, including pleomorphic, carcinoid tumor or undifferentiated carcinoma; however, lung adenocarcinoma makes up 44% and lung squamous cell carcinoma makes up 29% of all NSCLC cases clinically. At present, ~79% of NSCLC patients are diagnosed at advanced stages of the disease, when surgery is not a viable option (4). Thus, early detection, optimal tumor resection and effective chemotherapy, radiotherapy, immunotherapy and tumor-targeting therapy are important for the effective control of NSCLC. Therefore, a better understanding of NSCLC carcinogenesis and the underlying molecular mechanisms is key to developing novel early diagnosis strategies and improving treatment responses for NSCLC.

Lung carcinogenesis, like most human cancer types, is a complex molecular process involving aberrant cell proliferation (5) and apoptosis (6), which leads to the transformation of normal cells into malignant cells and subsequent cell invasion and metastasis. Cell transformation occurs through genetic mutations, loss of cell growth/critical genes, or epigenetic alterations in genomic DNA that silence tumor suppressor genes or activate oncogenes (7), resulting in abnormal cell-cell communication (8), DNA repair (9), chromosome stability (10) and cell motility (6,11). Recently, NSCLC was reported to exhibit abnormal expression of epidermal growth factor receptor (EGFR) (1214), c-Met (15), thyroid transcription factor 1 (TTF-1) (16), phosphoinositide 3-kinase/Rac-α serine/threonine-protein kinase/serine-threonine-protein kinase mTOR signaling (17), Ras-Raf-Mek-extracellular signal-regulated kinase signaling (18,19) and the echinoderm microtubule associated protein like 4-ALK receptor tyrosine kinase fusion gene (20,21). In addition, NSCLC may present with alterations in tumor suppressor genes, including RB transcriptional corepressor 1 (RB), p16-RB (16) and p14-MDM2 proto-oncogene-cellular tumor antigen p53 (p53) signaling (22), and other regulatory molecules, including microRNAs (23) or angiogenesis factors such as vascular endothelial growth factor (24). However, despite marked progress in understanding the molecular basis of human tumorigenesis, including lung cancer, a number of crucial genes and functions remain undefined. For example, RNA-binding protein 5 (RBM5) is localized at chromosome 3p21.3, a critical region associated with lung carcinogenesis. RBM5 regulates cell growth, cell cycle progression and apoptosis. Aberrant RBM5 protein expression leads to the transformation of normal bronchial cells, lung carcinogenesis, and alters the response of patients with lung cancer to treatment (24). In this review, the role of RBM5 in lung cancer is summarized.

Function of RBM5 in human cells

RBM5, also referred to as g15, LUCA-15 and H37, was initially cloned from a tumor suppressor gene (TSG) mapping area at chromosome 3p21.3 (25). RBM5 cDNA contains a full-length 815-amino acid open reading frame, with a predicted protein weight of ~90 kDa (25). The RBM5 protein has two zinc finger motifs, two RNA binding motifs and a bipartite nuclear signal. RBM5 localizes to the cell nucleus where it processes transcribed RNA, due to its DNA/RNA binding function (26). Earlier studies reported that the N-terminal of human RBM5 contains an RNA binding domain and RBM5 epitope marker (27), and that it had a priority function involving the poly(G) RNA polymer in vitro (28). At the C-terminal, RBM5 contains multiple regions, including a rich glutamine domain and a specific site for RNA and DNA binding proteins (28). RBM5 is widely expressed in various human tissues, particularly during embryonic development and in the adult thymus, although it is expressed at low levels in the fetal thymus (27) and normal lung (28). Another study reported a series of splice variants in RBM5 (29). In a normal lung, expression levels of the short transcript of RBM5 are higher compared with lung cancer cell lines, suggesting that the short RBM5 transcript may contribute to its tumor suppressor function in lung cancer (28).

Biologically, RBM5 facilitates DNA/RNA binding to process transcribed RNA, and regulates cell cycle progression and apoptosis during sperm maturation, bone and cardiac cell differentiation (3032). Specifically, RBM5 may modulate the alternative splicing of apoptosis-associated pre-mRNAs, including caspase 2 (CASP2) and Fas cell surface death receptor (FAS/CD95), to regulate cellular apoptosis (33,34). RBM5 may also upregulate the pro-apoptotic apoptosis regulator BAX protein, reduce mitochondrial cytochrome c release into the cytosol and activate caspase 9 and 3, whilst also downregulating the anti-apoptotic apoptosis regulator BCL-2 (BCL-2) and BCL-2 like 1 proteins (30,35,36) [Fig. 1; modified from (37)]. These data suggest that RBM5 may be able to activate the mitochondrial apoptosis pathway. Indeed, RBM5 is able to manipulate the pre-mRNA splicing of multiple target genes, including p53 (30,35,36,3842). Kobayashi et al (43) reported that RBM5 expression enhanced p53 mRNA expression levels and protein expression, whereas knockdown of RBM5 using RBM5 short hairpin RNA inhibited p53 transcription and protein expression, indicating that RBM5 regulates p53-mediated cell apoptosis. Furthermore, RBM5 is able to regulate apoptosis and cell cycle progression by increasing signal transducer and activator of transcription 5B and bone morphogenetic protein 5 expression levels, and reducing nuclear receptor coactivator 3, Pim-1 proto-oncogene serine/threonine kinase, baculoviral IAP repeat containing 3, BCL-2, EGFR and cyclin dependent kinase 2 expression levels (4447). In addition, RBM5 was demonstrated to inhibit cyclin A expression and RB phosphorylation, and thereby regulate cell cycle progression and induce G1 arrest (30). Although initially cloned from a TSG-mapping area at chromosome 3p21.3, RBM5 was previously dismissed as a TSG due to the lack of RBM5 mutations and its expression in the majority of lung cancers. Furthermore, there are multiple RBM5 protein isoforms, each of which differentially regulates apoptosis, leading to its inconsistent role as a tumor suppressor gene (48); however, a previous study did confirm its tumor suppressor function in lung cancer and other cancer types (49).

In eukaryotic cells, gene expression is almost completely regulated through mRNA splicing, and selective mRNA splicing ensures the diversity of functional proteins in cells (50). Thus, the accuracy and effectiveness of mRNA splicing are essential for maintaining homeostasis in eukaryotic cells. Defects in mRNA splicing are associated with various human diseases (2,5153). In this regard, RBM5 is involved in the selective mRNA splicing of apoptosis and cell cycle-associated genes (see above). For example, Fushimi et al (34) demonstrated that RBM5 regulated CASP2 splicing and expression in order to promote tumor suppression, whereas alternative splicing of CASP2 led to a loss of tumor suppressor activity. Thus, modulation of mRNA splicing regulators, like RBM5, may provide a novel therapeutic strategy to control human cancer. Bonnal et al (33) demonstrated that RBM5 was involved in recognizing the mRNA 3′-splice site in order to regulate the alternative splicing of apoptosis-associated mRNAs and their isoforms (including Fas receptor) in angiogenesis and apoptosis. RBM5 is unable to influence the early events of mRNA splicing for FAS at exon 6; however, RBM5 is able to inhibit the transition from the pre-spliceosome around FAS exon 6 into the mature spliceosome between the flanking FAS introns to induce DNA sequence-specific pairing in the distal mRNA splicing site. Jin et al (54) reported that RBM5 overexpression significantly induced exon 4 skipping of activation-induced cytidine deaminase by suppressing intron 3 splicing. This inhibitory effect required a weak mRNA 3′-splice site. As a result, RBM5 is able interfere with the binding of splicing factor U2AF 65 kDa subunit to polypyrimidines at the mRNA 3′-splice site in vitro (50). Taken together, the selective functions and alterations of RBM5 may alter the cell cycle and apoptosis, resulting in human tumorigenesis.

Tobacco use and deletion of the RBM5 locus

Tobacco use is the primary risk factor for developing human lung cancer (55). A number of previous studies have demonstrated that tobacco use contributes to cancer development, including lung, esophageal, bladder, pancreatic and kidney cancer (55). Tobacco contains at least 2,550 known chemical compounds and >60 of these have been demonstrated to be carcinogenic in humans and experimental animals (56). Polycyclic aromatic hydrocarbons (PAHs), including benzo(a)pyrene diol epoxide (BPDE) and N-nitrosamines, are the most highly carcinogenic compounds in tobacco (56,57). PAHs induce DNA adducts, genetic mutations, methylation and chromosome translocation in target organs (58,59). For example, BPDE reacts with cellular macromolecules to form DNA adducts, which are carcinogen metabolites covalently bound to DNA (5860) that induce apoptosis (61). Alternatively, if permanent mutations occur in a critical region of a DNA coding gene, an oncogene may be activated or a tumor suppressor gene deactivated, leading to aberrant cell growth, migration, adhesion and, ultimately, cancer (60). Previous studies that have reported direct evidence of tobacco carcinogen-targeted genes, including hot spot mutations in the KRAS proto-oncogene GTPase (KRAS) and p53 genes (60,62).

Furthermore, previous studies demonstrated that allele loss in a 370 kb region at chromosome 3p21.3 was the earliest alteration detected in pre-malignant lesions of lung cancer, or even in the histologically normal lung epithelium of tobacco smokers (63). Thus, Timmer et al (25) performed a comparative genomic structure and expression pattern analysis of this chromosomal region and identified RBM5. Specifically, RBM5 was involved in EGFR downregulation to prevent lung cancer cell proliferation, angiogenesis, invasion and metastasis (64,65).

One potential EGFR binding partner, the proto-oncoprotein human epidermal growth factor receptor 2 (HER2)/ErbB2, was reported to be overactive in a small percentage of patients with SCLC and non-smoker-associated NSCLC cases (66,67). These activating mutations of EGFR, KRAS and HER2 are mutually exclusive events in lung cancer (68,69). Notably, HER2 overexpression affects the alternative splicing of RBM5 (48). In light of these advances in lung cancer research, it may be speculated that future studies on RBM5 and its potential tumor suppressor activity should consider histological subtypes as well as tobacco smoking history and the mutation status of RBM5 in lung cancer initiation and/or progression. A previous in vitro study reported that RBM5 downregulation and activation of the Wnt/β-catenin signaling pathway are involved in cigarette smoke extract-induced lung epithelial injury, and that RBM5 functions as an upstream molecule to downregulate Wnt/β-catenin signaling (70). Interestingly, RBM5 knockout mice develop lung cancer at similar rates compared with those in wild type mice following exposure to nicotine-derived nitrosamine ketone. Loss of RBM5 expression leads to more aggressive lung cancer. Thus, reduced RBM5 expression and tobacco use increase the risk of an aggressive lung cancer phenotype (64).

Altered RBM5 expression in human cancer types

Lung cancer pathogenesis is multifactorial and results from the interaction between genetic and environmental factors. At the molecular level, genetic alterations are the most direct causes of lung cancer, and lung cancer development is associated with the deletion of tumor suppressor gene loci, including 3p21.3 (52,26), which may be observed in >90% of SCLC and in 50–80% of NSCLC cases (71). RBM5 expression levels are low in Ras-transformed rat embryonic fibroblasts (72), human vestibular schwannoma cells (73), human prostate cancer (74), ovarian cancer (74,75), human breast cancer tissue (48), human pancreatic cancer tissues (76) and lung cancer (77). Overexpression of RBM5 suppresses the growth of prostate cancer cells in vitro (74,75), and RBM5 expression is associated with lung cancer histological subtypes and tobacco use (78).

Although RBM5 expression is frequently reduced in lung, renal and breast cancer (79), RBM5 is not deleted in the majority of lung cancer cases (80). The reduced levels of RBM5 mRNA and protein in NSCLC compared with levels in normal lung tissues are associated with increased EGFR and KRAS expression levels, which are associated with tobacco use, advanced tumor stage and lymph node metastasis (81). Another study reported that RBM5 expression levels were significantly reduced in lung squamous cell tissues and were further associated with deletions at chromosome 3p21.3 and tobacco use (71,78), whereas three out of nine patients with lung adenocarcinoma did not have significant decreases in RBM5 mRNA expression levels (lung adenocarcinoma development may be associated with tobacco use, among which 50% of cases are associated with chromosome 3p21.3 deficiency) (78). Furthermore, Oh et al (82) reported that RBM5 expression was generally lower in lung cancer compared with normal lung tissues. Thus, detection of RBM5 expression may be a useful tumor marker for lung cancer.

RBM5 expression is able to suppress the growth of mouse fibrosarcoma cells or lung adenocarcinoma cells in nude mouse models (30,82,83). Notably, a number of genes were mapped to the common deletion region of chromosome 3p21.3, including RBM5 (25), FUS RNA binding protein (84), Ras association domain family member 1 (85), semaphorin 3B (86), semaphorin 3F (49), hyaluronidase 1 (49) and calcium voltage-gated channel auxiliary subunit α2δ2 (87), and had the ability to modulate lung cancer cell apoptosis. Reduced expression levels of RBM5, as one of nine downregulated genes in this 17-gene metastatic signature for solid tumors (including lung cancer) in humans and mice, was considered important for the development and/or progression of a wide range of human cancer types (88,89). Indeed, a recent study demonstrated that downregulation of RBM5 expression levels may be the key step in malignant lung cell transformation, and that RBM5 is responsible for inhibiting cell cycle progression and inducing apoptosis, in addition to suppressing tumor cell transformation-associated events, including angiogenesis, in SCLC cells (90). Furthermore, at the gene level, a constitutively activated RAS mutant protein (G12V) was demonstrated to be responsible for RBM5 downregulation in rat embryonic fibroblasts (72). Therefore, RBM5 may be a tumor marker for SCLC, and targeting RBM5 may be a potential novel and effective therapeutic option for controlling SCLC.

Thus far, the published data indicate that RBM5 is a lung cancer regulatory protein; however, the detection of various RBM5 isoforms may also be used to the determine association between lung cancer histological subtype and tobacco use, or even RBM5 mutation status (40). Moreover, further research on RBM5 alterations associated with other genes in the transforming growth factor signaling pathway is warranted (37). For example, RBM5 was able to post-transcriptionally regulate RBM10 expression by directly interacting with specific RBM10 splice variants (91).

Targeting RBM5 as a therapeutic strategy for lung cancer

As discussed above, RBM5 mRNA and protein expression levels are significantly reduced in different human cancer types, including lung cancer; thus, targeting RBM5 may be a novel therapeutic strategy for treating lung cancer. Indeed, a previous study revealed that cisplatin-resistant lung adenocarcinoma A549/DDP cells expressed decreased levels of RBM5 compared with parental A549 cells (41). Furthermore, knockdown of RBM5 expression with small interfering RNA in parental A549 cells reduced cisplatin-induced apoptosis. By contrast, exogenous RBM5 expression using a plasmid carrying RBM5 cDNA enhanced the sensitivity of A549/DDP cells to cisplatin treatment (41). In addition, RBM5-enhanced chemosensitivity to cisplatin was associated with cytochrome c release into the cytosol and subsequent activation of CASP9 and CASP3 (38). RBM5 expression inhibits the growth of human lung cancer cells by inducing cell cycle arrest and apoptosis (30). A previous study demonstrated the importance of RBM5 protein expression in normal lung cells and the consequences of RBM5 deletion in SCLC development and progression (90). RBM5 expression slowed the growth of SCLC cells in vitro and increased the sensitivity of tumor cells to cisplatin. Moreover, RBM5 expression inhibited SCLC cell cycle progression and reduced tumor cell membrane integrity (increase in apoptosis) following treatment with cisplatin. In this regard, reduced RBM5 expression was observed in 95% of SCLC cases, indicating the importance of altered RBM5 expression levels in SCLC development (76). Thus, a therapeutic strategy involving RBM5 and/or its direct target genes or pathways may be a very effective approach. In addition, detection of RBM5 expression might be useful for predicting response to chemotherapy in patients with lung cancer. The mechanism by which RBM5 affects gefitinib resistance in lung adenocarcinoma is currently being investigated (Xu et al, unpublished data).

Conclusions

Environmental factors, including tobacco use, may interact with human genes to cause cancer development and progression. As indicated in 2000 by Hanahan and Weinberg (6), there are six essential hallmarks of cancer cells: i) Self-sufficiency in growth signals; ii) insensitivity to growth-inhibitory signals; iii) evasion of apoptosis; iv) limitless replicative potential; v) sustained angiogenesis; and vi) tissue invasion and metastasis. RBM5 possesses at least two of these hallmarks. Thus, further investigations into RBM5 alterations, including chromosomal deletion, loss of heterozygosity, DNA methylation and/or gene-gene interactions, may lead to a better understanding of how RBM5 functions in lung cancer, leading to the development of a novel therapeutic strategy for the treatment of lung cancer. Further research on RBM5 may provide a novel mechanism to induce RBM5 expression or activate its downstream gene pathways. Moreover, detection of RBM5 expression, or its isoforms, may lead to early diagnosis of lung cancer and improve prognosis of patients with lung cancer.

Acknowledgements

Not applicable.

Funding

This review was supported in part by a grant from Nature Science Foundation of China (grant no. 81472169).

Availability of data and materials

Not applicable.

Authors' contributions

YX and PG participated in writing the original draft and in the design of this review; ZS participated in writing the original draft; JL, QW and GM researched the relevant literature; YZ participated in writing and editing the manuscript; WY managed the references and participated in the design of this review; JZ participated in the design of the review and in acquiring funding. All authors contributed substantially to the writing of this review. All authors 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.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

3 

American Cancer Society, Cancer Facts & Figures, 2016. https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2016.htmlJanuary 20–2018

4 

Carrato A, Vergnenègre A, Thomas M, McBride K, Medina J and Cruciani G: Clinical management patterns and treatment outcomes in patients with non-small cell lung cancer (NSCLC) across Europe: EPICLIN-Lung study. Curr Med Res Opin. 30:447–461. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Hunt T and Nasmyth K: Cell multiplication. Curr Opin Cell Biol. 9:765–767. 1997. View Article : Google Scholar : PubMed/NCBI

6 

Mueller BM, Yu YB and Laug WE: Overexpression of plasminogen activator inhibitor 2 in human melanoma cells inhibits spontaneous metastasis in scid/scid mice. Proc Natl Acad Sci USA. 92:205–209. 1995. View Article : Google Scholar : PubMed/NCBI

7 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Hirschi KK, Xu CE, Tsukamoto T and Sager R: Gap junction genes Cx26 and Cx43 individually suppress the cancer phenotype of human mammary carcinoma cells and restore differentiation potential. Cell Growth Differ. 7:861–870. 1996.PubMed/NCBI

9 

Sancar A: DNA repair in humans. Annu Rev Genet. 29:69–105. 1995. View Article : Google Scholar : PubMed/NCBI

10 

Hanawalt PC: Transcription-coupled repair and human disease. Science. 266:1957–1958. 1994. View Article : Google Scholar : PubMed/NCBI

11 

Stetler-Stevenson WG, Aznavoorian S and Liotta LA: Tumor cell interactions with the extracellular matrix during invasion and metastasis. Annu Rev Cell Biol. 9:541–573. 1993. View Article : Google Scholar : PubMed/NCBI

12 

Ichihara S, Toyooka S, Fujiwara Y, Hotta K, Shigematsu H, Tokumo M, Soh J, Asano H, Ichimura K, Aoe K, et al: The impact of epidermal growth factor receptor gene status on gefitinib-treated Japanese patients with non-small-cell lung cancer. Int J Cancer. 120:1239–1247. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Soh J, Okumura N, Lockwood WW, Yamamoto H, Shigematsu H, Zhang W, Chari R, Shames DS, Tang X, MacAulay C, et al: Oncogene mutations, copy number gains and mutant allele specific imbalance (MASI) frequently occur together in tumor cells. PLoS One. 4:e74642009. View Article : Google Scholar : PubMed/NCBI

14 

Wei Y, Zou Z, Becker N, Anderson M, Sumpter R, Xiao G, Kinch L, Koduru P, Christudass CS, Veltri RW, et al: EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell. 154:1269–1284. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Menges CW, Kadariya Y, Altomare D, Talarchek J, Neumann-Domer E, Wu Y, Xiao GH, Shapiro IM, Kolev VN, Pachter JA, et al: Tumor suppressor alterations cooperate to drive aggressive mesotheliomas with enriched cancer stem cells via a p53-miR-34a-c-Met axis. Cancer Res. 74:1261–1271. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Toyooka S, Mitsudomi T, Soh J, Aokage K, Yamane M, Oto T, Kiura K and Miyoshi S: Molecular oncology of lung cancer. Gen Thorac Cardiovasc Surg. 59:527–537. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Guo Y, Du J and Kwiatkowski DJ: Molecular dissection of AKT activation in lung cancer cell lines. Mol Cancer Res. 11:282–293. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Collisson EA, Trejo CL, Silva JM, Gu S, Korkola JE, Heiser LM, Charles RP, Rabinovich BA, Hann B, Dankort D, et al: A central role for RAF→MEK→ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2:685–693. 2012. View Article : Google Scholar : PubMed/NCBI

19 

De Luca A, Maiello MR, D'Alessio A, Pergameno M and Normanno N: The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: Role in cancer pathogenesis and implications for therapeutic approaches. Expert Opin Ther Targets. 16 Suppl 2:S17–S27. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Suda K, Tomizawa K, Yatabe Y and Mitsudomi T: Lung cancers unrelated to smoking: Characterized by single oncogene addiction? Int J Clin Oncol. 16:294–305. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Wu SG, Kuo YW, Chang YL, Shih JY, Chen YH, Tsai MF, Yu CJ, Yang CH and Yang PC: EML4-ALK translocation predicts better outcome in lung adenocarcinoma patients with wild-type EGFR. J Thorac Oncol. 7:98–104. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Paci M, Rapicetta C and Maramotti S: New biomarkers for lung cancer. Expert Opin Med Diagn. 4:201–224. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Shen J, Liu Z, Todd NW, Zhang H, Liao J, Yu L, Guarnera MA, Li R, Cai L, Zhan M and Jiang F: Diagnosis of lung cancer in individuals with solitary pulmonary nodules by plasma microRNA biomarkers. BMC Cancer. 11:3742011. View Article : Google Scholar : PubMed/NCBI

24 

Claesson-Welsh L: Blood vessels as targets in tumor therapy. Ups J Med Sci. 117:178–186. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Timmer T, Terpstra P, van den Berg A, Veldhuis PM, Ter Elst A, Voutsinas G, Hulsbeek MM, Draaijers TG, Looman MW, Kok K, et al: A comparison of genomic structures and expression patterns of two closely related flanking genes in a critical lung cancer region at 3p21.3. Eur J Hum Genet. 7:478–486. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Daigo Y, Nishiwaki T, Kawasoe T, Tamari M, Tsuchiya E and Nakamura Y: Molecular cloning of a candidate tumor suppressor gene, DLC1, from chromosome 3p21.3. Cancer Res. 59:1966–1972. 1999.PubMed/NCBI

27 

Aravind L and Koonin EV: G-patch: A new conserved domain in eukaryotic RNA-processing proteins and type D retroviral polyproteins. Trends Biochem Sci. 24:342–344. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Drabkin HA, West JD, Hotfilder M, Heng YM, Erickson P, Calvo R, Dalmau J, Gemmill RM and Sablitzky F: DEF-3(g16/NY-LU-12), an RNA binding protein from the 3p21.3 homozygous deletion region in SCLC. Oncogene. 18:2589–2597. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Sutherland LC, Rintala-Maki ND, White RD and Morin CD: RNA binding motif (RBM) proteins: A novel family of apoptosis modulators? J Cell Biochem. 94:5–24. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Oh JJ, Razfar A, Delgado I, Reed RA, Malkina A, Boctor B and Slamon DJ: 3p21.3 tumor suppressor gene H37/Luca15/RBM5 inhibits growth of human lung cancer cells through cell cycle arrest and apoptosis. Cancer Res. 66:3419–3427. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Loiselle JJ and Sutherland LC: Differential downregulation of Rbm5 and Rbm10 during skeletal and cardiac differentiation. In Vitro Cell Dev Biol Anim. 50:331–339. 2014. View Article : Google Scholar : PubMed/NCBI

32 

O'Bryan MK, Clark BJ, McLaughlin EA, D'Sylva RJ, O'Donnell L, Wilce JA, Sutherland J, O'Connor AE, Whittle B, Goodnow CC, et al: RBM5 is a male germ cell splicing factor and is required for spermatid differentiation and male fertility. PLoS Genet. 9:e10036282013. View Article : Google Scholar : PubMed/NCBI

33 

Bonnal S, Martinez C, Förch P, Bachi A, Wilm M and Valcárcel J: RBM5/Luca-15/H37 regulates Fas alternative splice site pairing after exon definition. Mol Cell. 32:81–95. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Fushimi K, Ray P, Kar A, Wang L, Sutherland LC and Wu JY: Up-regulation of the proapoptotic caspase 2 splicing isoform by a candidate tumor suppressor, RBM5. Proc Natl Acad Sci USA. 105:15708–15713. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Mourtada-Maarabouni M, Sutherland LC and Williams GT: Candidate tumour suppressor LUCA-15 can regulate multiple apoptotic pathways. Apoptosis. 7:421–432. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Sutherland LC, Lerman M, Williams GT and Miller BA: LUCA-15 suppresses CD95-mediated apoptosis in Jurkat T cells. Oncogene. 20:2713–2719. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Sutherland LC, Wang K and Robinson AG: RBM5 as a putative tumor suppressor gene for lung cancer. J Thorac Oncol. 5:294–298. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Li P, Wang K, Zhang J, Zhao L, Liang H, Shao C and Sutherland LC: The 3p21.3 tumor suppressor RBM5 resensitizes cisplatin-resistant human non-small cell lung cancer cells to cisplatin. Cancer Epidemiol. 36:481–489. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Mourtada-Maarabouni M, Sutherland LC, Meredith JM and Williams GT: Simultaneous acceleration of the cell cycle and suppression of apoptosis by splice variant delta-6 of the candidate tumour suppressor LUCA-15/RBM5. Genes Cells. 8:109–119. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Rintala-Maki ND and Sutherland LC: LUCA-15/RBM5, a putative tumour suppressor, enhances multiple receptor-initiated death signals. Apoptosis. 9:475–484. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Sutherland KD, Lindeman GJ, Choong DY, Wittlin S, Brentzell L, Phillips W, Campbell IG and Visvader JE: Differential hypermethylation of SOCS genes in ovarian and breast carcinomas. Oncogene. 23:7726–7733. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Sutherland LC, Edwards SE, Cable HC, Poirier GG, Miller BA, Cooper CS and Williams GT: LUCA-15-encoded sequence variants regulate CD95-mediated apoptosis. Oncogene. 19:3774–3781. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Kobayashi T, Ishida J, Musashi M, Ota S, Yoshida T, Shimizu Y, Chuma M, Kawakami H, Asaka M, Tanaka J, et al: p53 transactivation is involved in the antiproliferative activity of the putative tumor suppressor RBM5. Int J Cancer. 128:304–318. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Mourtada-Maarabouni M, Keen J, Clark J, Cooper CS and Williams GT: Candidate tumor suppressor LUCA-15/RBM5/H37 modulates expression of apoptosis and cell cycle genes. Exp Cell Res. 312:1745–1752. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Shao C, Yang B, Zhao L, Wang S, Zhang J and Wang K: Tumor suppressor gene RBM5 delivered by attenuated Salmonella inhibits lung adenocarcinoma through diverse apoptotic signaling pathways. World J Surg Oncol. 11:1232013. View Article : Google Scholar : PubMed/NCBI

46 

Shao C, Zhao L, Wang K, Xu W, Zhang J and Yang B: The tumor suppressor gene RBM5 inhibits lung adenocarcinoma cell growth and induces apoptosis. World J Surg Oncol. 10:1602012. View Article : Google Scholar : PubMed/NCBI

47 

Su Z, Yin J, Zhao L, Li R, Liang H, Zhang J and Wang K: Lentiviral vector-mediated RBM5 overexpression downregulates EGFR expression in human non-small cell lung cancer cells. World J Surg Oncol. 12:3672014. View Article : Google Scholar : PubMed/NCBI

48 

Rintala-Maki ND, Goard CA, Langdon CE, Wall VE, Traulsen KE, Morin CD, Bonin M and Sutherland LC: Expression of RBM5-related factors in primary breast tissue. J Cell Biochem. 100:1440–1458. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Zabarovsky ER, Lerman MI and Minna JD: Tumor suppressor genes on chromosome 3p involved in the pathogenesis of lung and other cancers. Oncogene. 21:6915–6935. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Graveley BR: Alternative splicing: Increasing diversity in the proteomic world. Trends Genet. 17:100–107. 2001. View Article : Google Scholar : PubMed/NCBI

51 

Nissim-Rafinia M and Kerem B: Splicing regulation as a potential genetic modifier. Trends Genet. 18:123–127. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Wang Z, Lo HS, Yang H, Gere S, Hu Y, Buetow KH and Lee MP: Computational analysis and experimental validation of tumor-associated alternative RNA splicing in human cancer. Cancer Res. 63:655–657. 2003.PubMed/NCBI

53 

Zhou Z, Licklider LJ, Gygi SP and Reed R: Comprehensive proteomic analysis of the human spliceosome. Nature. 419:182–185. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Jin W, Niu Z, Xu D and Li X: RBM5 promotes exon 4 skipping of AID pre-mRNA by competing with the binding of U2AF65 to the polypyrimidine tract. FEBS Lett. 586:3852–3857. 2012. View Article : Google Scholar : PubMed/NCBI

55 

U.S. Department of Health and Human Services: Smoking, Tobacco, and Cancer Program (1985–1989 Status Report). Public Health Service, USA. 1990.

56 

Hecht SS: Cigarette smoking: Cancer risks, carcinogens, and mechanisms. Langenbecks Arch Surg. 391:603–613. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Harvey RG: Polycyclic aromatic hydrocarbons. Chemistry and Carcinogenicity. Cambridge University Press; Cambridge: pp. 3961991

58 

Beland FA, Cain LG, Felton JS, et al: Chemical Carcinogenesis and Mutagenesis I. Springer-Verlag. 33–572. 1990.

59 

Bartsch H: DNA adducts in human carcinogenesis: Etiological relevance and structure-activity relationship. Mutat Res. 340:67–79. 1996. View Article : Google Scholar : PubMed/NCBI

60 

Mass MJ, Jeffers AJ, Ross JA, Nelson G, Galati AJ, Stoner GD and Nesnow S: Ki-ras oncogene mutations in tumors and DNA adducts formed by benz[j]aceanthrylene and benzo[a]pyrene in the lungs of strain A/J mice. Mol Carcinog. 8:186–192. 1993. View Article : Google Scholar : PubMed/NCBI

61 

Venkatachalam S, Denissenko MF, Alvi N and Wani AA: Rapid activation of apoptosis in human promyelocytic leukemic cells by (+/-)-anti-benzo[a]pyrene diol epoxide induced DNA damage. Biochem Biophys Res Commun. 197:722–729. 1993. View Article : Google Scholar : PubMed/NCBI

62 

Denissenko MF, Pao A, Tang M and Pfeifer GP: Preferential formation of benzo[a]pyrene adducts at lung cancer mutational hotspots in P53. Science. 274:430–432. 1996. View Article : Google Scholar : PubMed/NCBI

63 

Wistuba II, Lam S, Behrens C, Virmani AK, Fong KM, LeRiche J, Samet JM, Srivastava S, Minna JD and Gazdar AF: Molecular damage in the bronchial epithelium of current and former smokers. J Natl Cancer Inst. 89:1366–1373. 1997. View Article : Google Scholar : PubMed/NCBI

64 

Jamsai D, Watkins DN, O'Connor AE, Merriner DJ, Gursoy S, Bird AD, Kumar B, Miller A, Cole TJ, Jenkins BJ, et al: In vivo evidence that RBM5 is a tumour suppressor in the lung. Sci Rep. 7:163232017. View Article : Google Scholar : PubMed/NCBI

65 

Prabhu VV and Devaraj N: Regulating RNA binding Motif 5 gene expression-a novel therapeutic target for lung cancer. J Environ Pathol Toxicol Oncol. 36:99–105. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Canoz O, Ozkan M, Arsav V, Er O, Coskun HS, Soyuer S and Altinbas M: The role of c-erbB-2 expression on the survival of patients with small-cell lung cancer. Lung. 184:267–272. 2006. View Article : Google Scholar : PubMed/NCBI

67 

Hirsch FR, Franklin WA, Veve R, Varella-Garcia M and Bunn PA Jr: HER2/neu expression in malignant lung tumors. Semin Oncol. 29:51–58. 2002. View Article : Google Scholar : PubMed/NCBI

68 

Bae NC, Chae MH, Lee MH, Kim KM, Lee EB, Kim CH, Park TI, Han SB, Jheon S, Jung TH and Park JY: EGFR, ERBB2, and KRAS mutations in Korean non-small cell lung cancer patients. Cancer Genet Cytogenet. 173:107–113. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Buttitta F, Barassi F, Fresu G, Felicioni L, Chella A, Paolizzi D, Lattanzio G, Salvatore S, Camplese PP, Rosini S, et al: Mutational analysis of the HER2 gene in lung tumors from Caucasian patients: Mutations are mainly present in adenocarcinomas with bronchioloalveolar features. Int J Cancer. 119:2586–2591. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Hao YQ, Su ZZ, Lv XJ, Li P, Gao P, Wang C, Bai Y and Zhang J: RNA-binding motif protein 5 negatively regulates the activity of Wnt/β-catenin signaling in cigarette smoke-induced alveolar epithelial injury. Oncol Rep. 33:2438–2444. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Lerman MI and Minna JD: The 630-kb lung cancer homozygous deletion region on human chromosome 3p21.3: Identification and evaluation of the resident candidate tumor suppressor genes. The International Lung Cancer Chromosome 3p21.3 Tumor Suppressor Gene Consortium. Cancer Res. 60:6116–6133. 2000.PubMed/NCBI

72 

Edamatsu H, Kaziro Y and Itoh H: LUCA15, a putative tumour suppressor gene encoding an RNA-binding nuclear protein, is down-regulated in ras-transformed Rat-1 cells. Genes Cells. 5:849–858. 2000. View Article : Google Scholar : PubMed/NCBI

73 

Welling DB, Lasak JM, Akhmametyeva E, Ghaheri B and Chang LS: cDNA microarray analysis of vestibular schwannomas. Otol Neurotol. 23:736–748. 2002. View Article : Google Scholar : PubMed/NCBI

74 

Zhao L, Li R, Shao C, Li P, Liu J and Wang K: 3p21.3 tumor suppressor gene RBM5 inhibits growth of human prostate cancer PC-3 cells through apoptosis. World J Surg Oncol. 10:2472012. View Article : Google Scholar : PubMed/NCBI

75 

Kim YS, Hwan JD, Bae S, Bae DH and Shick WA: Identification of differentially expressed genes using an annealing control primer system in stage III serous ovarian carcinoma. BMC Cancer. 10:5762010. View Article : Google Scholar : PubMed/NCBI

76 

Peng J, Valeshabad AK, Li Q and Wang Y: Differential expression of RBM5 and KRAS in pancreatic ductal adenocarcinoma and their association with clinicopathological features. Oncol Lett. 5:1000–1004. 2013. View Article : Google Scholar : PubMed/NCBI

77 

Oh JJ, Taschereau EO, Koegel AK, Ginther CL, Rotow JK, Isfahani KZ and Slamon DJ: RBM5/H37 tumor suppressor, located at the lung cancer hot spot 3p21.3, alters expression of genes involved in metastasis. Lung Cancer. 70:253–262. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Toyooka S, Maruyama R, Toyooka KO, McLerran D, Feng Z, Fukuyama Y, Virmani AK, Zochbauer-Muller S, Tsukuda K, Sugio K, et al: Smoke exposure, histologic type and geography-related differences in the methylation profiles of non-small cell lung cancer. Int J Cancer. 103:153–160. 2003. View Article : Google Scholar : PubMed/NCBI

79 

Bechara EG, Sebestyén E, Bernardis I, Eyras E and Valcárcel J: RBM5, 6, and 10 differentially regulate NUMB alternative splicing to control cancer cell proliferation. Mol Cell. 52:720–733. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Wistuba II, Behrens C, Virmani AK, Mele G, Milchgrub S, Girard L, Fondon JW III, Garner HR, McKay B, Latif F, et al: High resolution chromosome 3p allelotyping of human lung cancer and preneoplastic/preinvasive bronchial epithelium reveals multiple, discontinuous sites of 3p allele loss and three regions of frequent breakpoints. Cancer Res. 60:1949–1960. 2000.PubMed/NCBI

81 

Liang H, Zhang J, Shao C, Zhao L, Xu W, Sutherland LC and Wang K: Differential expression of RBM5, EGFR and KRAS mRNA and protein in non-small cell lung cancer tissues. J Exp Clin Cancer Res. 31:362012. View Article : Google Scholar : PubMed/NCBI

82 

Oh JJ, West AR, Fishbein MC and Slamon DJ: A candidate tumor suppressor gene, H37, from the human lung cancer tumor suppressor locus 3p21.3. Cancer Res. 62:3207–3213. 2002.PubMed/NCBI

83 

ter Elst A, Hiemstra BE, van der Vlies P, Kamminga W, van der Veen AY, Davelaar I, Terpstra P, te Meerman GJ, Gerbens F, Kok K, et al: Functional analysis of lung tumor suppressor activity at 3p21.3. Genes Chromosomes Cancer. 45:1077–1093. 2006. View Article : Google Scholar : PubMed/NCBI

84 

Ji L and Roth JA: Tumor suppressor FUS1 signaling pathway. J Thorac Oncol. 3:327–330. 2008. View Article : Google Scholar : PubMed/NCBI

85 

Agathanggelou A, Bièche I, Ahmed-Choudhury J, Nicke B, Dammann R, Baksh S, Gao B, Minna JD, Downward J, Maher ER and Latif F: Identification of novel gene expression targets for the Ras association domain family 1 (RASSF1A) tumor suppressor gene in non-small cell lung cancer and neuroblastoma. Cancer Res. 63:5344–5351. 2003.PubMed/NCBI

86 

Castro-Rivera E, Ran S, Thorpe P and Minna JD: Semaphorin 3B (SEMA3B) induces apoptosis in lung and breast cancer, whereas VEGF165 antagonizes this effect. Proc Natl Acad Sci USA. 101:11432–11437. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Carboni GL, Gao B, Nishizaki M, Xu K, Minna JD, Roth JA and Ji L: CACNA2D2-mediated apoptosis in NSCLC cells is associated with alterations of the intracellular calcium signaling and disruption of mitochondria membrane integrity. Oncogene. 22:615–626. 2003. View Article : Google Scholar : PubMed/NCBI

88 

Qiu TH, Chandramouli GV, Hunter KW, Alkharouf NW, Green JE and Liu ET: Global expression profiling identifies signatures of tumor virulence in MMTV-PyMT-transgenic mice: Correlation to human disease. Cancer Res. 64:5973–5981. 2004. View Article : Google Scholar : PubMed/NCBI

89 

Ramaswamy S, Ross KN, Lander ES and Golub TR: A molecular signature of metastasis in primary solid tumors. Nat Genet. 33:49–54. 2003. View Article : Google Scholar : PubMed/NCBI

90 

Loiselle JJ, Roy JG and Sutherland LC: RBM5 reduces small cell lung cancer growth, increases cisplatin sensitivity and regulates key transformation-associated pathways. Heliyon. 2:e002042016. View Article : Google Scholar : PubMed/NCBI

91 

Loiselle JJ, Roy JG and Sutherland LC: RBM10 promotes transformation-associated processes in small cell lung cancer and is directly regulated by RBM5. PLoS One. 12:e01802582017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 17 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu Y, Su Z, Li J, Wang Q, Meng G, Zhang Y, Yang W, Zhang J and Gao P: Role of RNA‑binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer (Review). Oncol Lett 17: 2013-2019, 2019
APA
Xu, Y., Su, Z., Li, J., Wang, Q., Meng, G., Zhang, Y. ... Gao, P. (2019). Role of RNA‑binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer (Review). Oncology Letters, 17, 2013-2019. https://doi.org/10.3892/ol.2018.9818
MLA
Xu, Y., Su, Z., Li, J., Wang, Q., Meng, G., Zhang, Y., Yang, W., Zhang, J., Gao, P."Role of RNA‑binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer (Review)". Oncology Letters 17.2 (2019): 2013-2019.
Chicago
Xu, Y., Su, Z., Li, J., Wang, Q., Meng, G., Zhang, Y., Yang, W., Zhang, J., Gao, P."Role of RNA‑binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer (Review)". Oncology Letters 17, no. 2 (2019): 2013-2019. https://doi.org/10.3892/ol.2018.9818