Skip to main content
Top
Published in: Cancer Cell International 1/2020

Open Access 01-12-2020 | Kidney Cancer | Primary research

Identifying the novel key genes in renal cell carcinoma by bioinformatics analysis and cell experiments

Authors: Yeda Chen, Di Gu, Yaoan Wen, Shuxin Yang, Xiaolu Duan, Yongchang Lai, Jianan Yang, Daozhang Yuan, Aisha Khan, Wenqi Wu, Guohua Zeng

Published in: Cancer Cell International | Issue 1/2020

Login to get access

Abstract

Background

Although major driver gene have been identified, the complex molecular heterogeneity of renal cell cancer (RCC) remains unclear. Therefore, more relevant genes need to be identified to explain the pathogenesis of renal cancer.

Methods

Microarray datasets GSE781, GSE6344, GSE53000 and GSE68417 were downloaded from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified by employing GEO2R tool, and function enrichment analyses were performed by using DAVID. The protein-protein interaction network (PPI) was constructed and the module analysis was performed using STRING and Cytoscape. Survival analysis was performed using GEPIA. Differential expression was verified in Oncomine. Cell experiments (cell viability assays, transwell migration and invasion assays, wound healing assay, flow cytometry) were utilized to verify the roles of the hub genes on the proliferation of kidney cancer cells (A498 and OSRC-2 cell lines).

Results

A total of 215 DEGs were identified from four datasets. Six hub gene (SUCLG1, PCK2, GLDC, SLC12A1, ATP1A1, PDHA1) were identified and the overall survival time of patients with RCC were significantly shorter. The expression levels of these six genes were significantly decreased in six RCC cell lines(A498, OSRC-2, 786- O, Caki-1, ACHN, 769-P) compared to 293t cell line. The expression level of both mRNA and protein of these genes were downregulated in RCC samples compared to those in paracancerous normal tissues. Cell viability assays showed that overexpressions of SUCLG1, PCK2, GLDC significantly decreased proliferation of RCC. Transwell migration, invasion, wound healing assay showed overexpression of three genes(SUCLG1, PCK2, GLDC) significantly inhibited the migration, invasion of RCC. Flow cytometry analysis showed that overexpression of three genes(SUCLG1, PCK2, GLDC) induced G1/S/G2 phase arrest of RCC cells.

Conclusion

Based on our current findings, it is concluded that SUCLG1, PCK2, GLDC may serve as a potential prognostic marker of RCC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Arakaki R, Yamasaki T, Kanno T, et al. CCL2 as a potential therapeutic target for clear cell renal cell carcinoma. Cancer Med. 2016;5:2920–33.CrossRef Arakaki R, Yamasaki T, Kanno T, et al. CCL2 as a potential therapeutic target for clear cell renal cell carcinoma. Cancer Med. 2016;5:2920–33.CrossRef
2.
go back to reference Vasudev NS, Selby PJ, Banks RE. Renal cancer biomarkers: the promise of personalized care. BMC Med. 2012;10:112.CrossRef Vasudev NS, Selby PJ, Banks RE. Renal cancer biomarkers: the promise of personalized care. BMC Med. 2012;10:112.CrossRef
3.
go back to reference Zhu X, Liang J, Shrubsole MJ, et al. Calcium intake and ion transporter genetic polymorphisms interact in human colorectal neoplasia risk in a 2-phase study. J Nutr. 2014;144:1734–41.CrossRef Zhu X, Liang J, Shrubsole MJ, et al. Calcium intake and ion transporter genetic polymorphisms interact in human colorectal neoplasia risk in a 2-phase study. J Nutr. 2014;144:1734–41.CrossRef
4.
go back to reference He Y, Zhang P, Zhang D, et al. Combined assessment of low PGRMC1/positive ATP1A1 levels has enhanced prognostic value for renal cell carcinoma. Oncol Rep. 2018;40:1467–76.PubMed He Y, Zhang P, Zhang D, et al. Combined assessment of low PGRMC1/positive ATP1A1 levels has enhanced prognostic value for renal cell carcinoma. Oncol Rep. 2018;40:1467–76.PubMed
5.
go back to reference Lin CS, Lee HT, Lee MH, et al. Role of mitochondrial DNA copy number alteration in human renal cell carcinoma. Int J Mol Sci. 2016;17(6):814–28.CrossRef Lin CS, Lee HT, Lee MH, et al. Role of mitochondrial DNA copy number alteration in human renal cell carcinoma. Int J Mol Sci. 2016;17(6):814–28.CrossRef
6.
go back to reference Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–10.CrossRef Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–10.CrossRef
7.
go back to reference Lenburg ME, Liou LS, Gerry NP, Frampton GM, Cohen HT, Christman MF. Previously unidentified changes in renal cell carcinoma gene expression identified by parametric analysis of microarray data. BMC Cancer. 2003;3:31.CrossRef Lenburg ME, Liou LS, Gerry NP, Frampton GM, Cohen HT, Christman MF. Previously unidentified changes in renal cell carcinoma gene expression identified by parametric analysis of microarray data. BMC Cancer. 2003;3:31.CrossRef
8.
go back to reference Gumz ML, Zou H, Kreinest PA, et al. Secreted frizzled-related protein 1 loss contributes to tumor phenotype of clear cell renal cell carcinoma. Clin Cancer Res. 2007;13:4740–9.CrossRef Gumz ML, Zou H, Kreinest PA, et al. Secreted frizzled-related protein 1 loss contributes to tumor phenotype of clear cell renal cell carcinoma. Clin Cancer Res. 2007;13:4740–9.CrossRef
9.
go back to reference Gerlinger M, Horswell S, Larkin J, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46:225–33.CrossRef Gerlinger M, Horswell S, Larkin J, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46:225–33.CrossRef
10.
go back to reference Thibodeau BJPD, Fulton MMD, Fortier LEMS, et al. Characterization of clear cell renal cell carcinoma by gene expression profiling. Urol Oncol Semin Original Investigations. 2015;34:161–8. Thibodeau BJPD, Fulton MMD, Fortier LEMS, et al. Characterization of clear cell renal cell carcinoma by gene expression profiling. Urol Oncol Semin Original Investigations. 2015;34:161–8.
11.
go back to reference Huang DW, Sherman BT, Tan Q, et al. The DAVID gene functional classification tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 2007;8:R183.CrossRef Huang DW, Sherman BT, Tan Q, et al. The DAVID gene functional classification tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 2007;8:R183.CrossRef
12.
go back to reference Kanehisa M. The KEGG database. Novartis Found Symp. 2002; 247: 91-101, 101-103, 119-128, 244-252. Kanehisa M. The KEGG database. Novartis Found Symp. 2002; 247: 91-101, 101-103, 119-128, 244-252.
13.
go back to reference Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–9.CrossRef Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–9.CrossRef
14.
go back to reference Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47:D607–13.CrossRef Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47:D607–13.CrossRef
15.
go back to reference Van Parys T, Melckenbeeck I, Houbraken M, et al. A cytoscape app for motif enumeration with ISMAGS. Bioinformatics. 2017;33:461–3.PubMed Van Parys T, Melckenbeeck I, Houbraken M, et al. A cytoscape app for motif enumeration with ISMAGS. Bioinformatics. 2017;33:461–3.PubMed
16.
go back to reference Bader GD, Hogue CW. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 2003;4:2.CrossRef Bader GD, Hogue CW. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 2003;4:2.CrossRef
17.
go back to reference Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:l1.CrossRef Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:l1.CrossRef
18.
go back to reference Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.CrossRef Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.CrossRef
19.
go back to reference Maere S, Heymans K, Kuiper M. BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics (Oxford, England). 2005;21:3448–9.CrossRef Maere S, Heymans K, Kuiper M. BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics (Oxford, England). 2005;21:3448–9.CrossRef
20.
go back to reference Haeussler M, Zweig AS, Tyner C, et al. The UCSC genome browser database: 2019 update. Nucleic Acids Res. 2019;2019(47):D853–8.CrossRef Haeussler M, Zweig AS, Tyner C, et al. The UCSC genome browser database: 2019 update. Nucleic Acids Res. 2019;2019(47):D853–8.CrossRef
21.
go back to reference Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:W98–102.CrossRef Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:W98–102.CrossRef
22.
go back to reference Cardaci S, Zheng L, MacKay G, et al. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat Cell Biol. 2015;17:1317–26.CrossRef Cardaci S, Zheng L, MacKay G, et al. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat Cell Biol. 2015;17:1317–26.CrossRef
23.
go back to reference Liu Y, Li W, Duan Y. Effect of H2O2 induced oxidative stress (OS) on volatile organic compounds (VOCs) and intracellular metabolism in MCF-7 breast cancer cells. J Breath Res. 2019;13:36005.CrossRef Liu Y, Li W, Duan Y. Effect of H2O2 induced oxidative stress (OS) on volatile organic compounds (VOCs) and intracellular metabolism in MCF-7 breast cancer cells. J Breath Res. 2019;13:36005.CrossRef
24.
go back to reference Grange C, Brossa A, Bussolati B. Extracellular vesicles and carried miRNAs in the progression of renal cell carcinoma. Int J Mol Sci. 2019;20:1832.CrossRef Grange C, Brossa A, Bussolati B. Extracellular vesicles and carried miRNAs in the progression of renal cell carcinoma. Int J Mol Sci. 2019;20:1832.CrossRef
25.
go back to reference Grabmaier K, Weijert MC, Verhaegh GW, Schalken JA, Oosterwijk E. Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene. 2004;23:5624–31.CrossRef Grabmaier K, Weijert MC, Verhaegh GW, Schalken JA, Oosterwijk E. Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene. 2004;23:5624–31.CrossRef
26.
go back to reference Woodard J, Sassano A, Hay N, Platanias LC. Statin-dependent suppression of the Akt/mammalian target of rapamycin signaling cascade and programmed cell death 4 up-regulation in renal cell carcinoma. Clin Cancer Res. 2008;14:4640–9.CrossRef Woodard J, Sassano A, Hay N, Platanias LC. Statin-dependent suppression of the Akt/mammalian target of rapamycin signaling cascade and programmed cell death 4 up-regulation in renal cell carcinoma. Clin Cancer Res. 2008;14:4640–9.CrossRef
27.
go back to reference Wilson MR, Hou Z, Wilson LJ, Ye J, Matherly LH. Functional and mechanistic roles of the human proton-coupled folate transporter transmembrane domain 6–7 linker. Biochem J. 2016;473:3545–62.CrossRef Wilson MR, Hou Z, Wilson LJ, Ye J, Matherly LH. Functional and mechanistic roles of the human proton-coupled folate transporter transmembrane domain 6–7 linker. Biochem J. 2016;473:3545–62.CrossRef
28.
go back to reference Zhao J, Li J, Fan TWM, Hou SX. Glycolytic reprogramming through PCK2 regulates tumor initiation of prostate cancer cells. Oncotarget. 2017;8:83602.CrossRef Zhao J, Li J, Fan TWM, Hou SX. Glycolytic reprogramming through PCK2 regulates tumor initiation of prostate cancer cells. Oncotarget. 2017;8:83602.CrossRef
29.
go back to reference Luo S, Li Y, Ma R, et al. Downregulation of PCK2 remodels tricarboxylic acid cycle in tumor-repopulating cells of melanoma. Oncogene. 2017;36:3609–17.CrossRef Luo S, Li Y, Ma R, et al. Downregulation of PCK2 remodels tricarboxylic acid cycle in tumor-repopulating cells of melanoma. Oncogene. 2017;36:3609–17.CrossRef
30.
go back to reference Liu M, Jin L, Sun S, et al. Metabolic reprogramming by PCK1 promotes TCA cataplerosis, oxidative stress and apoptosis in liver cancer cells and suppresses hepatocellular carcinoma. Oncogene. 2018;37:1637–53.CrossRef Liu M, Jin L, Sun S, et al. Metabolic reprogramming by PCK1 promotes TCA cataplerosis, oxidative stress and apoptosis in liver cancer cells and suppresses hepatocellular carcinoma. Oncogene. 2018;37:1637–53.CrossRef
31.
go back to reference Leithner K, Hrzenjak A, Trötzmüller M, et al. PCK2 activation mediates an adaptive response to glucose depletion in lung cancer. Oncogene. 2015;34:1044–50.CrossRef Leithner K, Hrzenjak A, Trötzmüller M, et al. PCK2 activation mediates an adaptive response to glucose depletion in lung cancer. Oncogene. 2015;34:1044–50.CrossRef
32.
go back to reference Vincent EE, Sergushichev A, Griss T, et al. Mitochondrial phosphoenolpyruvate carboxykinase regulates metabolic adaptation and enables glucose-independent tumor growth. Mol Cell. 2015;60:195–207.CrossRef Vincent EE, Sergushichev A, Griss T, et al. Mitochondrial phosphoenolpyruvate carboxykinase regulates metabolic adaptation and enables glucose-independent tumor growth. Mol Cell. 2015;60:195–207.CrossRef
33.
go back to reference Zhang WC, Shyh-Chang N, Yang H, et al. Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis. Cell. 2012;148:259–72.CrossRef Zhang WC, Shyh-Chang N, Yang H, et al. Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis. Cell. 2012;148:259–72.CrossRef
34.
go back to reference Jäger K, Larribère L, Wu H, Weiss C, Gebhardt C, Utikal J. Expression of neural crest markers GLDC and ERRFI1 is correlated with melanoma prognosis. Cancers. 2019;11:76.CrossRef Jäger K, Larribère L, Wu H, Weiss C, Gebhardt C, Utikal J. Expression of neural crest markers GLDC and ERRFI1 is correlated with melanoma prognosis. Cancers. 2019;11:76.CrossRef
35.
go back to reference Min HL, Kim J, Kim WH, Jang BG, Kim MA. Epigenetic silencing of the putative tumor suppressor gene GLDC (Glycine Dehydrogenase) in gastric carcinoma. Anticancer Res. 2016;36:179.PubMed Min HL, Kim J, Kim WH, Jang BG, Kim MA. Epigenetic silencing of the putative tumor suppressor gene GLDC (Glycine Dehydrogenase) in gastric carcinoma. Anticancer Res. 2016;36:179.PubMed
Metadata
Title
Identifying the novel key genes in renal cell carcinoma by bioinformatics analysis and cell experiments
Authors
Yeda Chen
Di Gu
Yaoan Wen
Shuxin Yang
Xiaolu Duan
Yongchang Lai
Jianan Yang
Daozhang Yuan
Aisha Khan
Wenqi Wu
Guohua Zeng
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2020
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-020-01405-6

Other articles of this Issue 1/2020

Cancer Cell International 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine