Deletion of C9orf53 promotes the growth of head and neck squamous cell carcinoma and is associated with poor prognosis of patients with head and neck squamous cell carcinoma

  • Authors:
    • Guo Dong Wang
    • Jian Tao Huang
    • Yuan Liu
    • Yang Wu
    • Xiao Qing Chen
    • Yun Fu Zhao
    • Da Lin Wang
  • View Affiliations

  • Published online on: November 8, 2018     https://doi.org/10.3892/ol.2018.9675
  • Pages: 1223-1228
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Head and neck squamous cell carcinoma (HNSCC) is the fifth most common carcinoma worldwide, and accounts for ~600,000 new cases every year. The information on the molecular carcinogenesis of HNSCC is very limited. In the present study, the role of C9orf53 in HNSCC was investigated. The levels of C9orf53 were assayed by reverse transcription‑quantitative polymerase chain reaction. The levels of C9orf53 in cells were overexpressed by overexpression plasmid and inhibited by small‑interfering RNA. Cell proliferation was assayed by MTT, and cell apoptosis was assessed by FACS analysis. It was demonstrated that C9orf53 deletion was associated with a decreased survival of patients. The level of C9orf53 in HNSCC tissues was lower compared with the matched normal tissues adjacent to tumors. A lower expression of C9orf53 promoted cell proliferation, and the overexpression of C9orf53 induced cell apoptosis. In conclusion, a low level of C9orf53 in HNSCC promoted the growth of HNSCC cells, which might be associated with the low survival rate of patients with HNSCC.

Introduction

Head and neck squamous cell carcinoma (HNSCC) encompasses a heterogeneous group of tumors with aggressive nature and is the fifth most common carcinoma worldwide (1,2). HNSCC accounts for ~4% of all malignancies worldwide and 5% of all cancer mortalities (3).

The choice of cancer treatment depends on the site of the primary tumor, the stage of the disease, treatment toxicities and the expected oncological and functional outcomes. One approach to improve treatment efficacy is to add novel molecular targeted agents to the classical treatment regimens (4).

Monoclonal antibodies targeting the epidermal growth factor receptor have shown clinical benefits in palliative and curative settings (5). However, only a minority of patients presenting with current or metastatic HNSCC exhibited tumor regression with these agents, and the majority of patients develop acquired tumor resistance following several months of treatments (4). Therefore in order to develop agents that target novel proteins, the identification of novel genes in HNSCC is required.

In the present study, the mutated genes in HNSCC were screened by searching the Cancer Genome Atlas (TCGA). The TCGA research network has profiled and analyzed large numbers of human tumors in order to identify molecular aberrations at DNA, RNA, protein and epigenetic levels (613). It was identified that there were many genes with amplification and deletion in HNSCC (Table I), including C9orf53. C9orf53 is a protein-coding gene. C9orf53 (also named CDKN2A-AS1) is associated with Alzheimer's disease (14). The present study focused on the role of C9orf53 in HNSCC, and it was found that the main type of C9orf53 mutation in HNSCC is deletion. Importantly, it was observed that deletions in C9orf53 are associated with lower patient survival rates. In the cell experiments, the role of C9orf53 in HNSCC was investigated in vitro by overexpressing or inhibiting C9orf53 expression. In conclusion, the present study revealed the role of C9orf53 in HNSCC, and the study may provide a novel therapeutic target for future investigation.

Table I.

Expression of genes that are deleted or amplified in HNSCC.

Table I.

Expression of genes that are deleted or amplified in HNSCC.

GenesP-value
Amplification
FADDP<0.001
USP13P<0.001
DCUN1D1P<0.001
EGFRP<0.001
CCND1P<0.001
FGF3P<0.001
MIR548KP<0.001
PPFIA1P<0.001
POU5F1BP<0.001
CASC8P<0.001
CCAT1 4.82×10−22
FGFR1 1.15×10−18
RGP1 1.24×10−13
LINC00393 1.34×10−11
MYRFL 5.81×10−11
MLANA0.147
CDK60.259
SAMD9L0.259
SNAI2 9.67×10−9
NFIB 2.38×10−8
E2F1 9.36×10−6
ANKRD39 5.09×10−5
LRRC14B 7.44×10−5
ERBB2 1.16×10−4
TINAG 2.37×10−4
CD44 5.72×10−4
LINC00452 2.99×10−3
PTP4A10.037
Deletion
C9ORF53P<0.001
ZNF532P<0.001
CDKN2AP<0.001
RN7SL5P 1.42×10−39
RNY3P4 3.13×10−23
STK11 1.77×10−19
FAM72C 4.76×10−19
PDE4D 6.60×10−18
RB1 4.14×10−17
PARD3 1.80×10−14
LINC009710.101
KIAA0825 1.25×10−9
MIR3182 4.80×10−9
CBWD3 2.74×10−7
PTPRG 1.29×10−6
GPN2 1.62×10−6
ZNF750 2.67×10−6
KCNIP4 1.51×10−5
RNA5SP431 1.61×10−5
RAB24 1.09×10−4
C9ORF163 1.24×10−3
LINC00645 7.31×10−3
LRRN10.0239

[i] The expression data were acquired from The Cancer Genome Atlas, where there were 302 HNSCC samples analyzed. The mean expression of these genes in tumor tissues and normal tissues were compared by paired t-test. HNSCC, Head and neck squamous cell carcinoma.

Materials and methods

Frequency of gene alteration and patient survival analysis for cancer

The data on the frequency of C9orf53 alteration, mRNA level analysis and patient survival were obtained from TCGA via cBioportal for Cancer Genomics (http://www.cbioportal.org/public-portal/index.do) (15,16).

Patients

Surgical specimens from 19 patients with HNSCC (age range: 36–75 years; mean age: 54.3±7.4 years; Sex ratio: Male to Female 1.7:1) and matched normal tissues adjacent to tumors were obtained postoperatively in June, 2009 from the Department of Head and Neck Surgery, Changhai Hospital, Second Military Medical University (Shanghai, China). All patients gave signed, informed consent for their tissues to be used for scientific research. Ethics approval for the present study was obtained from Changhai Hospital, Second Military Medical University (Shanghai, China). All diagnoses were based on pathological and/or cytological findings. The histological features of the specimens were evaluated by senior pathologists according to the World Health Organization classification criteria (17). The tissues were obtained prior to chemotherapy and radiotherapy. The samples were immediately frozen and stored at −80°C prior to reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay.

Cell culture

293, SCC-5 and SCC-9 cells were obtained from the Cell Bank of Chinese Academy of Science (Shanghai, China). 293, SCC-5 and SCC-9 cells were cultured in 37°C, 5%CO2, and in Dulbecco's modified Eagle's medium (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 10% fetal bovine serum (Hyclone; GE Healthcare Life Sciences), 2 mM L-glutamine and 100 µg/ml penicillin/streptomycin (Sangon Biotech Co., Ltd., Shanghai, China) as described in previous studies (1820).

RNA extraction and RT-qPCR

RNA was extracted with Trizol (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol. The cDNA synthesis and RT-qPCR were subsequently performed using the Qiagen system as described previously (21). RT-qPCR analysis was performed using standard protocols on Applied Biosystem 7500 HT sequence Detection system. The relative mRNA levels of C9orf53 were normalized to levels of the housekeeping gene GAPDH and calculated using the 2−ΔΔCq method (22). The primers used are as follows: GAPDH forward, 5′-CCATGTTCGTCATGGG-TGTGAACCA-3′ and reverse, 5′-GCCAGTAGAGGCAGGGATGATGTTG-3′ and C9orf53 forward, 5′-AAGAATTCGGCACGAGGGTT-3′ and reverse, 5′-CTCTGCCACAGTGGGATTGT-3′.

MTT assay

For MTT assay, 5×103 cells per well were seeded in triplicate in a 96-well plate with complete growth medium. The cells were counted over 5 days using the MTT assay (Promega Corporation, Madison, WI, USA) as described previously (19,20). The data was measured using the Microtiter plate reader (Promega Corporation) at 570 nm.

Transfection of C9orf53 overexpression plasmid and small-interfering (si)RNA

C9orf53 overexpression plasmid (pcDNA3.1-C9orf53;), C9orf53-siRNA (5′-GTGTGATTTCGTAAACAGATA-3′) and control-siRNA were designed and constructed by Sangon Biotech Company (Shanghai, China). Untransfected cells were used as a blank control. Transfections were performed using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. SCC-5 and SCC-9 cells were seeded into 24-well plates at a density of 5×104 cells/well and were allowed to culture overnight. DNA Plasmid (500 ng) with Lipofectamine® 2000 and siRNA (600 ng) with Lipofectamine® 2000 were mixed for transfection. Then DNA-Lipofectamine® 2000 complexes were added into wells at 37°C for 24 h prior to subsequent experimentation.

Apoptosis assay

SCC-5 and SCC-9 cells were labeled with Annexin V-fluorescein isothiocyanate and propidium iodide (PI) using the apoptosis detecting kit (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. Then samples were analyzed by fluorescence-activated cell sorting (FACS) assay with a flow cytometer (BD FACSVerse™ flow cytometer, with BD FACSuite™ software v1.0.6, BD Biosciences, San Jose, CA, USA) (23).

Statistical analysis

Data are presented as the mean ± standard deviation from at least three independent experiments. The difference between groups was analyzed using two-tailed, paired Student's t-test when only two groups were compared. The Wilcoxon matched-pairs signed rank test was used to determine if there was a statistically significant difference in the expression of C9orf53 between matched pairs. The difference between groups was analyzed using one-way ANOVA with post hoc contrasts by Student-Newman-Keuls test, when three or more than three groups were compared. Correlation analysis was performed using two-tailed Person's correlation coefficient analysis. Patient survival was determined by Kaplan-Meier analysis (with log-rank test). Statistical analyses were performed using SPSS software (version 17.0; IBM Corp., Armonk, NY, USA). P<0.05 was considered significantly different.

Results

Screening of novel genes amplified or deleted in HNSCC

To identify the potential target genes of HNSCC, the genes that are amplified or deleted in HNSCC were screened. It was observed that there were a number of genes amplified or deleted in HNSCC (Table I). To date, to the best of our knowledge, there is no study on the role of C9orf53 in HNSCC. Therefore, in the present study, the role of C9orf53 in HNSCC was investigated. Initially, mutations in C9orf53 were investigated in various types of cancer and mutations were identified in 302 HNSCC tissue samples. There were 27.5% HNSCC tissues with C9orf53 deletions (Fig. 1A). Next, it was observed that a low expression of C9orf53 mRNA was associated with C9orf53 deletions (Fig. 1B). Importantly, these data indicated that C9orf53 deletion was associated with a decreased survival rate (Fig. 1C).

Reduced expression of C9orf53 in HNSCC tissues compared with normal tissues

Next, 19 surgical specimens and matched tumor-adjacent normal tissues were obtained from patients with HNSCC. The levels of C9orf53 mRNA were assayed by RT-qPCR. It was identified that 17 of the 19 HNSCC tissues exhibited lower C9orf53 expression compared with matched normal tissues that were adjacent to tumors (Fig. 2A). Furthermore, the median C9orf53 expression in HNSCC tissues was lower compared with normal tissues that were adjacent to tumors, and the standard deviation of C9orf53 expression in HNSCC was higher compared with normal tissues (Fig. 2B).

Reduced expression of C9orf53 promotes proliferation and overexpression of C9orf53-induced apoptosis

Next, the role of C9orf53 in vitro was investigated. The levels of C9orf53 in HNSCC cell lines (SCC-5 and SCC-9) were analyzed, and 293 cells were used as a control. It was observed that the levels of C9orf53 in SCC-5 and SCC-9 were lower compared with 293 cells (Fig. 3A). As SCC-5 cells exhibited lower C9orf53 levels compared with SCC-9 cells, C9orf53 was overexpressed in SCC-5 cells by transfection with pcDNA3.1-C9orf53. C9orf53 expression was suppressed in SCC-9 cells by transfection with si-C9orf53. The levels of C9orf53 were determined by RT-qPCR, 48 h following transfection. It was observed that the level of C9orf53 was successfully increased following transfection of pcDNA3.1-C9orf53 in SCC-5 cells and that C9orf53 expression was successfully inhibited following transfection with si-C9orf53 in SCC-9 cells (Fig. 3B). As transfection with si-C9orf53-1 resulted in the greatest decrease in C9orf53 expression (Fig. 3B), si-C9orf53-1 was selected for subsequent experiments. Following the overexpression or inhibition of C9orf53, proliferation was examined by MTT assay. It was found that overexpressing C9orf53 inhibited proliferation, and suppressing C9orf53 promoted proliferation (Fig. 3C). Next, the cell apoptosis was examined by FACS assay. The overexpression of C9orf53 increased the rate of cell apoptosis (Fig. 3D).

Discussion

In the present study, the role of C9orf53 in HNSCC was studied. It was identified that C9orf53 was deleted in HNSCC, and the levels of C9orf53 in HNSCC tissues were lower in tumor tissues compared with matched normal tissues that were adjacent to tumor tissues, Notably, the deletion of C9orf53 and a lower expression level of C9orf53 were associated with the rate of patient survival. Subsequently, the role of C9orf53 was confirmed by overexpressing and suppressing C9orf53 expression in vitro. The suppression of C9orf53 was able to promote proliferation and induce apoptosis and apoptosis. To the best of our best knowledge, this might be the first report of the role of C9orf53 in cancer.

The data from the present study indicated that the effects of C9orf53 on proliferation and apoptosis were relative minor. However, the difference in survival between patients with C9orf53 deletion and wild-type C9orf53 was significant. It is surprising that these minor influences were able to cause an effect on patient survival. It was hypothesized that although the effects of C9orf53 on cell proliferation and apoptosis were rather minor, the cells proliferated exponentially in vivo. Therefore, minor differences in C9orf53 were able to cause a long-lasting effect on patient survival (130 months).

Recently, a comprehensive landscape of somatic genomic alteration of HNSCC was provided by TCGA (24). It was indicated that human-papillomavirus-associated tumors are dominated by helical domain mutations of the oncogene phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α. Smoking-related HNSCCs demonstrated near universal loss-of-function tumor protein p53 mutations and CDKN2A inactivation. Whether the role of C9orf53 in HNSCC is associated with human papillomavirus infection or smoking will be investigated in further studies.

In conclusion, the present study investigated the role of C9orf53 in HNSCC. The data indicated that the deletion and decreased level of C9orf53 might promote the growth of HNSCC cells. The present study might provide a novel therapeutic target for further investigations.

Acknowledgements

The authors would like to thank Dr. Chaoxiong Zhang (West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China) for assistance with the discussion.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81271353) and PLA Research Project (grant no. 2011XL015) and ‘The 12th Five-Year Plan’ for Medicsal Science Development of PLA Research Project (grant no. CWS11J300), the Key Project on the Integration of Industry, Education, and Research and Medicine of Science and Technology Commission of Shanghai Municipality (grant nos. 12DZ1940503 and 13DZ1942704).

Availability of data and materials

All data generated or analyzed during the present study are included in this published article.

Authors' contributions

GW, HJ and YL collected patient data and performed cell experiments. YW and XC performed transfection and apoptosis analysis. YZ and DW contributed to study design and manuscript writing.

Ethics approval and consent to participate

The present study was approved by Ethics Committee of Second Military Medical University (Shanghai, China).

Patient consent for publication

All patients gave informed consent for the use of their tissues and publication of the data.

Competing interests

The authors declare that they have no competing interests.

References

1 

Liu CJ, Lin SC, Chen YJ, Chang KM and Chang KW: Array-comparative genomic hybridization to detect genomewide changes in microdissected primary and metastatic oral squamous cell carcinomas. Mol Carcinog. 45:721–731. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Shieh TM, Lin SC, Liu CJ, Chang SS, Ku TH and Chang KW: Association of expression aberrances and genetic polymorphisms of lysyl oxidase with areca-associated oral tumorigenesis. Clin Cancer Res. 13:4378–4385. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Schmitz S, Ang KK, Vermorken J, Haddad R, Suarez C, Wolf GT, Hamoir M and Machiels JP: Targeted therapies for squamous cell carcinoma of the head and neck: Current knowledge and future directions. Cancer Treat Rev. 40:390–404. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Allegra CJ, Jessup JM, Somerfield MR, Hamilton SR, Hammond EH, Hayes DF, McAllister PK, Morton RF and Schilsky RL: American society of clinical oncology provisional clinical opinion: Testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy. J Clin Oncol. 27:2091–2096. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Cancer Genome Atlas Research Network, . Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The cancer genome atlas pan-cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, et al: Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 17:98–110. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Cancer Genome Atlas Research Network: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Cancer Genome Atlas Research Network: Comprehensive genomic characterization of squamous cell lung cancers. Nature. 489:519–525. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al: COSMIC: Mining complete cancer genomes in the catalogue of somatic mutations in cancer. Nucleic Acids Res. 29:D945–D950. 2011. View Article : Google Scholar

12 

Collins FS and Barker AD: Mapping the cancer genome. Scientific American. 296:50–57. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Stratton MR, Campbell PJ and Futreal PA: The cancer genome. Nature. 458:719–724. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Emanuele E, Lista S, Ghidoni R, Binetti G, Cereda C, Benussi L, Maletta R, Bruni AC and Politi P: Chromosome 9p21.3 genotype is associated with vascular dementia and Alzheimer's disease. Neurobiol Aging. 32:1231–1235. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

17 

Barnes L, Eveson JW, Reichart P and Sidransky D: Pathology and genetics of head and neck tumours. IARC. 2005.

18 

Wu N, Liu C, Bai C, Han YP, Cho WC and Li Q: Over-expression of deubiquitinating enzyme USP14 in lung adenocarcinoma promotes proliferation through the accumulation of beta-catenin. Int J Mol Sci. 14:10749–10760. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Wu N, Zhang C, Bai C, Han YP and Li Q: miR-4782-3p Inhibited non-small cell lung cancer growth via USP14. Cell Physiol Biochem. 33:457–467. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Song B, Zhang C, Li G, Jin G and Liu C: MiR-940 inhibited pancreatic ductal adenocarcinoma growth by targeting MyD88. Cell Physiol Biochem. 35:1167–1177. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Hou J, Lin L, Zhou W, Wang Z, Ding G, Dong Q, Qin L, Wu X, Zheng Y, Yang Y, et al: Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for hepatocellular carcinoma. Cancer Cell. 19:232–243. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Lu J, Wen M, Huang Y, He X, Wang Y, Wu Q, Li Z, Castellanos-Martin A, Abad M, Cruz-Hernandez JJ, et al: C2ORF40 suppresses breast cancer cell proliferation and invasion through modulating expression of M phase cell cycle genes. Epigenetics. 8:571–583. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Cancer Genome Atlas Network: Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517:576–582. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 17 Issue 1

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
Wang GD, Huang JT, Liu Y, Wu Y, Chen XQ, Zhao YF and Wang DL: Deletion of C9orf53 promotes the growth of head and neck squamous cell carcinoma and is associated with poor prognosis of patients with head and neck squamous cell carcinoma. Oncol Lett 17: 1223-1228, 2019
APA
Wang, G.D., Huang, J.T., Liu, Y., Wu, Y., Chen, X.Q., Zhao, Y.F., & Wang, D.L. (2019). Deletion of C9orf53 promotes the growth of head and neck squamous cell carcinoma and is associated with poor prognosis of patients with head and neck squamous cell carcinoma. Oncology Letters, 17, 1223-1228. https://doi.org/10.3892/ol.2018.9675
MLA
Wang, G. D., Huang, J. T., Liu, Y., Wu, Y., Chen, X. Q., Zhao, Y. F., Wang, D. L."Deletion of C9orf53 promotes the growth of head and neck squamous cell carcinoma and is associated with poor prognosis of patients with head and neck squamous cell carcinoma". Oncology Letters 17.1 (2019): 1223-1228.
Chicago
Wang, G. D., Huang, J. T., Liu, Y., Wu, Y., Chen, X. Q., Zhao, Y. F., Wang, D. L."Deletion of C9orf53 promotes the growth of head and neck squamous cell carcinoma and is associated with poor prognosis of patients with head and neck squamous cell carcinoma". Oncology Letters 17, no. 1 (2019): 1223-1228. https://doi.org/10.3892/ol.2018.9675