Identification of AUNIP as a candidate diagnostic and prognostic biomarker for oral squamous cell carcinoma

Zongcheng Yang, Xiuming Liang, Yue Fu, Yingjiao Liu, Lixin Zheng, Fen Liu, Tongyu Li, Xiaolin Yin, Xu Qiao, Xin Xu, Zongcheng Yang, Xiuming Liang, Yue Fu, Yingjiao Liu, Lixin Zheng, Fen Liu, Tongyu Li, Xiaolin Yin, Xu Qiao, Xin Xu

Abstract

Background: Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors worldwide. Patients with poorly differentiated OSCC often exhibit a poor prognosis. AUNIP (Aurora Kinase A and Ninein Interacting Protein), also known as AIBp, plays a key role in cell cycle and DNA damage repair. However, the function of AUNIP in OSCC remains elusive.

Methods: The differentially expressed genes (DEGs) were obtained using R language. Receiver operating characteristic curve analysis was performed to identify diagnostic markers for OSCC. The effectiveness of AUNIP in diagnosing OSCC was evaluated by machine learning. AUNIP expression was analyzed in publicly available databases and clinical specimens. Bioinformatics analysis and in vitro experiments were conducted to explore biological functions and prognostic value of AUNIP in OSCC.

Findings: The gene integration analysis revealed 90 upregulated DEGs. One candidate biomarker, AUNIP, for the diagnosis of OSCC was detected, and its expression gradually increased along with malignant differentiation of OSCC. Bioinformatics analysis demonstrated that AUNIP could be associated with tumor microenvironment, human papillomavirus infection, and cell cycle in OSCC. The suppression of AUNIP inhibited OSCC cell proliferation and resulted in G0/G1 phase arrest in OSCC cells. The survival analysis showed that AUNIP overexpression predicted poor prognosis of OSCC patients.

Interpretation: AUNIP could serve as a candidate diagnostic and prognostic biomarker for OSCC and suppression of AUNIP may be a potential approach to preventing and treating OSCC. FUND: Taishan Scholars Project in Shandong Province (ts201511106) and the National Natural Science Foundation of China (Nos. 61603218).

Keywords: AUNIP; Biomarker; Oral squamous cell carcinoma; Receiver operating characteristic curve; Survival analysis; Weighted gene co-expression network analysis.

Conflict of interest statement

The authors declare no conflicts of interest.

Copyright © 2019. Published by Elsevier B.V.

Figures

Fig. 1
Fig. 1
Identification of differentially upregulated expressed genes. a–c. Volcano plots of gene expression profiles in GSE3524, GSE30784, and GSE78060. Red/blue symbols classify the upregulated/downregulated genes according to the criteria: log2FC > 1 and adjusted P-value <0·01. d. Common upregulated DEGs among GSE3524, GSE30784, and GSE78060. e. The expression matrix of 90 common upregulated DEGs in 29 pairs of OSCC and adjacent normal tissues followed by unsupervised hierarchical clustering in TCGA database. Blue, red and white respectively represents a lower expression level, a higher expression level and no expression difference among the genes. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 2
Fig. 2
Identification of key genes for diagnosis of OSCC. a. ROC curve analysis of the top ten DEGs in TCGA database. Genes with an AUC value are shown. b. ROC curve analysis of the selected ten genes in GSE30784. Genes with an AUC value are shown. c, d. ROC curve to assess sensitivity and specificity of AUNIP expression as a diagnostic biomarker for OSCC in TCGA database and GSE30784 dataset. e. ROC curve of AUNIP in the five-fold cross-validation. f. Construction of confusion matrix. g. Evaluation metrics of each fold. All data are represented by mean ± SD.
Fig. 3
Fig. 3
AUNIP expression is elevated in OSCC samples from TCGA database. a. TCGA database analysis of the relatively differential expression level (log2) of AUNIP in OSCC (n = 306) and adjacent normal oral (n = 29) tissues. b. Quantification of AUNIP mRNA levels (log2) in HPV-negative (n = 274) and HPV-positive (n = 32) OSCC tissues from TCGA database. c, d. Analysis of AUNIP mRNA levels (log2) in different stromal score and immune score (low, high, n = 153) of OSCC tissues from TCGA database. e, f. Analysis of AUNIP mRNA levels (log2) in different histologic grades (well, n = 48; moderate, n = 191; poor, n = 63) and tumor T stages (T1 + T2, n = 122; T3 + T4, n = 166) of OSCC tissues from TCGA database. g. The negative correlation between AUNIP mRNA expression level (log2) and ESTIMATE score in TCGA database. P-values were obtained by Student's t-test, One-way ANOVA test, and Pearson correlation analysis. All data are represented by mean ± SD.
Fig. 4
Fig. 4
AUNIP expression is upregulated in clinical OSCC samples. a. The protein levels of AUNIP in four pairs of OSCC tissues (T) and adjacent non-tumor tissues (N) measured by western blot. b. Quantification of AUNIP IHC staining in OSCC (n = 16) and normal oral (n = 89) tissues. c. Representative images of IHC staining for AUNIP in normal oral tissues and different histologic grades of OSCC tissues. Scale bars: 100 μm (insets 50 μm). d. Quantification of AUNIP IHC staining in OSCC and paired adjacent normal tissues (n = 9). e. Quantification of AUNIP IHC staining in different histologic grades of OSCC tissues (well, n = 48; moderate, n = 35; poor, n = 6). f ROC curve from IHC staining shows AUNIP is a marker to distinguish OSCC tissues from normal oral tissues. P-values were obtained by Student's t-test and One-way ANOVA test. All data are represented by mean ± SD.
Fig. 5
Fig. 5
Weighted co-expression network construction and identification of the key module containing AUNIP. a. Hierarchical clustering dendogram of samples from GSE41613. The clinical traits of survival status and survival time are displayed at the bottom. b, c. Analysis of scale-free fit index and the mean connectivity for various soft-thresholding powers. Testing the scale free topology when β = 8. d. Hierarchical clustering dendogram of genes with dissimilarity based on topological overlap. Modules are the branches of the clustering tree. e. The heat map describes the TOM among selected 1000 genes in WGCNA, and darker colour represents higher overlap and lighter colour corresponds to lower overlap. The gene dendrogram and module assignment are shown along the left side and the top. f. Correlation between module eigengenes and clinical traits. Each row corresponds to a module eigengene and columns represent clinical traits. Each cell contains the correlation and P-value, and yellow module containing AUNIP is selected. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 6
Fig. 6
Genes positively associated with AUNIP are enriched in cell cycle. a. Scatter plot of genes in yellow module. The vertical line represents cutoff of module membership = 0·8, and the horizontal line represents cutoff of gene significances for survival time = 0·2. Genes on upper right contains AUNIP. b. Network of genes positively associated with AUNIP expression according to the topological overlap. c. Heat map describes expression profiles of 134 genes positively associated with AUNIP in 306 OSCC samples in TCGA database. d. Gene ontology analysis of genes positively associated with AUNIP. e. KEGG pathway enrichment analysis of genes positively associated with AUNIP. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 7
Fig. 7
AUNIP is associated with cell cycle and HPV status in OSCC. a–f. GSEA of Hallmarks, GO, KEGG pathway gene sets in AUNIP high versus low samples from TCGA database. Normalized enrichment score (NES), nominal P-value and FDR are shown in each plot. g. Correlation between AUNIP and cell cycle, HPV infection related molecules in OSCC samples from TCGA database. *P ≤ 0·05, **P ≤ 0·01, and ***P ≤ 0·001 were obtained by Pearson correlation analysis.
Fig. 8
Fig. 8
AUNIP controls cell cycle progression in vitro and its expression is associated with poor survival in OSCC patients. a. AUNIP mRNA levels in indicated cells transfected with AUNIP siRNA. b. AUNIP protein levels in indicated cells transfected with AUNIP siRNA. c. Representative images (left) and quantification (right) of Blank, Negative Control (NC) siRNA-, and AUNIP siRNA- transfected SCC-9 and SCC-15 cells were analyzed in a colony formation assay. d. Representative images (left) and quantification (right) of Blank, Negative Control (NC) siRNA-, and AUNIP siRNA- transfected SCC-9 and SCC-15 cells were analyzed in cell cycle assay. e, f. Kaplan-Meier analysis of overall survival was performed to indicate higher expression of AUNIP was correlated with poor survival of OSCC patients in GSE41613 and TCGA datasets. P-values were obtained from the log-rank test. n.s, not significant, *P ≤ 0·05, **P ≤ 0·01, ***P ≤ 0·001, and ****P ≤ 0·0001 were obtained by Student's t-test. All data are represented by mean ± SD.

References

    1. Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.
    1. Siegel R.L., Miller K.D., Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7–30.
    1. Shi J., Bao X., Liu Z., Zhang Z., Chen W., Xu Q. Serum miR-626 and miR-5100 are promising prognosis predictors for Oral squamous cell carcinoma. Theranostics. 2019;9(4):920–931.
    1. Huang T.H., Li K.Y., Choi W.S. Lymph node ratio as prognostic variable in oral squamous cell carcinomas: systematic review and meta-analysis. Oral Oncol. 2019;89:133–143.
    1. Gharat S.A., Momin M., Bhavsar C. Oral squamous cell carcinoma: current treatment strategies and nanotechnology-based approaches for prevention and therapy. Crit Rev Ther Drug Carrier Syst. 2016;33(4):363–400.
    1. Zhao X., Sun S., Zeng X., Cui L. Expression profiles analysis identifies a novel three-mRNA signature to predict overall survival in oral squamous cell carcinoma. Am J Cancer Res. 2018;8(3):450–461.
    1. Yen C.J., Tsou H.H., Hsieh C.Y. Sequential therapy of neoadjuvant biochemotherapy with cetuximab, paclitaxel, and cisplatin followed by cetuximab-based concurrent bioradiotherapy in high-risk locally advanced oral squamous cell carcinoma: final analysis of a phase 2 clinical trial. Head Neck. 2019;41(6):1703–1712.
    1. Gan C.P., Sam K.K., Yee P.S. IFITM3 knockdown reduces the expression of CCND1 and CDK4 and suppresses the growth of oral squamous cell carcinoma cells. Cell Oncol (Dordr) 2019;42(4):477–490.
    1. Li J.M., Tseng C.W., Lin C.C. Upregulation of LGALS1 is associated with oral cancer metastasis. Ther Adv Med Oncol. 2018;10 1758835918794622.
    1. Hussein A.A., Forouzanfar T., Bloemena E. A review of the most promising biomarkers for early diagnosis and prognosis prediction of tongue squamous cell carcinoma. Br J Cancer. 2018;119(6):724–736.
    1. Rivera C., Oliveira A.K., Costa R.A.P., De Rossi T., Paes Leme A.F. Prognostic biomarkers in oral squamous cell carcinoma: a systematic review. Oral Oncol. 2017;72:38–47.
    1. Liu X., Wu J., Zhang D. Identification of potential key genes associated with the pathogenesis and prognosis of gastric cancer based on integrated bioinformatics analysis. Front Genet. 2018;9:265.
    1. Ni M., Liu X., Wu J. Identification of candidate biomarkers correlated with the pathogenesis and prognosis of non-small cell lung cancer via integrated bioinformatics analysis. Front Genet. 2018;9:469.
    1. Lieu A.S., Cheng T.S., Chou C.H. Functional characterization of AIBp, a novel Aurora-a binding protein in centrosome structure and spindle formation. Int J Oncol. 2010;37(2):429–436.
    1. Chou C.H., Loh J.K., Yang M.C. AIBp regulates mitotic entry and mitotic spindle assembly by controlling activation of both Aurora-A and Plk1. Cell Cycle. 2015;14(17):2764–2776.
    1. Lou J., Chen H., Han J. AUNIP/C1orf135 directs DNA double-strand breaks towards the homologous recombination repair pathway. Nat Commun. 2017;8(1):985.
    1. Foy J.P., Bertolus C., Ortiz-Cuaran S. Immunological and classical subtypes of oral premalignant lesions. Oncoimmunology. 2018;7(12)
    1. Wang Y., Fan H., Zheng L. Biological information analysis of differentially expressed genes in oral squamous cell carcinoma tissues in GEO database. J BUON. 2018;23(6):1662–1670.
    1. Wang R., Zhou X., Wang H. Integrative analysis of gene expression profiles reveals distinct molecular characteristics in oral tongue squamous cell carcinoma. Oncol Lett. 2019;17(2):2377–2387.
    1. Bai S., Zhang P., Zhang J.C. A gene signature associated with prognosis and immune processes in head and neck squamous cell carcinoma. Head Neck. 2019;41(8):2581–2590.
    1. Chabanais J., Labrousse F., Chaunavel A., Germot A., Maftah A. POFUT1 as a promising novel biomarker of colorectal cancer. Cancers (Basel) 2018;10(11)
    1. Yoshihara K., Shahmoradgoli M., Martinez E. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.
    1. Hosmer D.W., Lemeshow S., Sturdivant R.X. 3rd ed. Wiley; Hoboken, New Jersey: 2013. Applied logistic regression.
    1. Müller A.C., Guido S. 1st ed. O'Reilly Media, Inc; Sebastopol, CA: 2016. Introduction to machine learning with Python: A guide for data scientists.
    1. Azim H.A., Jr., Peccatori F.A., Brohee S. RANK-ligand (RANKL) expression in young breast cancer patients and during pregnancy. Breast Cancer Res. 2015;17:24.
    1. Beckerman P., Qiu C., Park J. Human kidney tubule-specific gene expression based dissection of chronic kidney disease traits. EBioMedicine. 2017;24:267–276.
    1. Tang J., Kong D., Cui Q. Prognostic genes of breast cancer identified by gene co-expression network analysis. Front Oncol. 2018;8:374.
    1. Rosen E.Y., Wexler E.M., Versano R. Functional genomic analyses identify pathways dysregulated by progranulin deficiency, implicating Wnt signaling. Neuron. 2011;71(6):1030–1042.
    1. Carnielli C.M., Macedo C.C.S., De Rossi T. Combining discovery and targeted proteomics reveals a prognostic signature in oral cancer. Nat Commun. 2018;9(1):3598.
    1. Wang S., Yin S., Zhang Z.L., Su X., Xu Z.F. Quality of life after oral cancer resection and free flap reconstruction. J Oral Maxillofac Surg. 2019;77(8):1724–1732.
    1. Kansy K., Hoffmann J., Alhalabi O. Subjective and objective appearance of head and neck cancer patients following microsurgical reconstruction and associated quality of life horizontal line a cross-sectional study. J Craniomaxillofac Surg. 2018;46(8):1275–1284.
    1. Sequeira I., Neves J.F., Carrero D. Immunomodulatory role of keratin 76 in oral and gastric cancer. Nat Commun. 2018;9(1):3437.
    1. Wang Y., Guo W., Li Z. Role of the EZH2/miR-200 axis in STAT3-mediated OSCC invasion. Int J Oncol. 2018;52(4):1149–1164.
    1. Keshavarzi M., Darijani M., Momeni F. Molecular imaging and oral cancer diagnosis and therapy. J Cell Biochem. 2017;118(10):3055–3060.
    1. Baykul T., Yilmaz H.H., Aydin U., Aydin M.A., Aksoy M., Yildirim D. Early diagnosis of oral cancer. J Int Med Res. 2010;38(3):737–749.
    1. Murphy J., Isaiah A., Wolf J.S., Lubek J.E. Quality of life factors and survival after total or extended maxillectomy for sinonasal malignancies. J Oral Maxillofac Surg. 2015;73(4):759–763.
    1. Yang B., Chen Z., Huang Y., Han G., Li W. Identification of potential biomarkers and analysis of prognostic values in head and neck squamous cell carcinoma by bioinformatics analysis. Onco Targets Ther. 2017;10:2315–2321.
    1. Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674.
    1. Otto T., Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer. 2017;17(2):93–115.
    1. Zhang P., Kawakami H., Liu W. Targeting CDK1 and MEK/ERK overcomes apoptotic resistance in BRAF-mutant human colorectal cancer. Mol Cancer Res. 2018;16(3):378–389.
    1. Peltanova B., Raudenska M., Masarik M. Effect of tumor microenvironment on pathogenesis of the head and neck squamous cell carcinoma: a systematic review. Mol Cancer. 2019;18(1):63.
    1. Huertas P., Cortes-Ledesma F., Sartori A.A., Aguilera A., Jackson S.P. CDK targets Sae2 to control DNA-end resection and homologous recombination. Nature. 2008;455(7213):689–692.
    1. Chapman J.R., Taylor M.R., Boulton S.J. Playing the end game: DNA double-strand break repair pathway choice. Mol Cell. 2012;47(4):497–510.
    1. Kanao R., Masutani C. Regulation of DNA damage tolerance in mammalian cells by post-translational modifications of PCNA. Mutat Res. 2017;803–805:82–88.
    1. Fakhry C., Blackford A.L., Neuner G. Association of oral human papillomavirus DNA persistence with cancer progression after primary treatment for oral cavity and oropharyngeal squamous cell carcinoma. JAMA Oncol. 2019;5(7):985–992.
    1. Slebos R.J., Yi Y., Ely K. Gene expression differences associated with human papillomavirus status in head and neck squamous cell carcinoma. Clin Cancer Res. 2006;12(3):701–709. Pt 1.

Source: PubMed

3
Abonnere