Skip to main content
Top
Published in: Cancer Cell International 1/2018

Open Access 01-12-2018 | Primary Research

DNA methylation affects metastasis of renal cancer and is associated with TGF-β/RUNX3 inhibition

Authors: Jianbo Zheng, Yanhui Mei, Ping Xiang, Guangsheng Zhai, Ning Zhao, Chuanbing Xu, Min Liu, Zhengsheng Pan, Kai Tang, Dongsheng Jia

Published in: Cancer Cell International | Issue 1/2018

Login to get access

Abstract

Background

Renal cell carcinoma accounts for 2–3% of all cancers and metastasis increased the malignancy of renal cancer. However, the role of methylation in metastasis of renal cancer is poorly understood.

Methods

We performed targeted gene array to compare the differential expressions of methylation regulated genes in metastatic and primary renal cancer tissues. Quantitative methylation specific PCR was performed to examine the CpG methylation levels of Runt related transcription factor 3 (RUNX3) and transforming growth factor (TGF)-β. Western blot was performed to detect the expression of target genes. Murine xenograft renal cancer model was established to assay gene expression, methylation level, tumor growth and animal survival in vivo.

Results

RUNX3 and TGF-β levels were decreased in metastatic renal cancer tissues as a result of their CpG methylation. Metastatic xenograft model displayed decreased expression levels of RUNX3 and TGF-β and higher CpG methylation levels, bigger tumor size and shorter survival time, all which were restored by treatment with a methylation inhibitor.

Conclusions

Hypermethylation in CpG islands promotes metastasis of renal cancer and is associated with TGF-β and RUNX3 inhibition.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Al-Marhoon MS. Small incidental renal masses in adults: review of the literature. Sultan Qaboos Univ Med J. 2010;10:196–202.PubMedPubMedCentral Al-Marhoon MS. Small incidental renal masses in adults: review of the literature. Sultan Qaboos Univ Med J. 2010;10:196–202.PubMedPubMedCentral
3.
go back to reference Lasseigne BN, Burwell TC, Patil MA, Absher DM, Brooks JD, Myers RM. DNA methylation profiling reveals novel diagnostic biomarkers in renal cell carcinoma. BMC Med. 2014;12:235.CrossRefPubMedPubMedCentral Lasseigne BN, Burwell TC, Patil MA, Absher DM, Brooks JD, Myers RM. DNA methylation profiling reveals novel diagnostic biomarkers in renal cell carcinoma. BMC Med. 2014;12:235.CrossRefPubMedPubMedCentral
4.
go back to reference Xue J, Wu XL, Huang XT, Qu M, Guo F, Sun GY, Zhang PC, Han L, Pan LM. Correlation of RUNX3 expression with microvessel density in colorectal adenocarcinoma tissues and clinical significance. Asian Pac J Trop Med. 2017;10:98–101.CrossRefPubMed Xue J, Wu XL, Huang XT, Qu M, Guo F, Sun GY, Zhang PC, Han L, Pan LM. Correlation of RUNX3 expression with microvessel density in colorectal adenocarcinoma tissues and clinical significance. Asian Pac J Trop Med. 2017;10:98–101.CrossRefPubMed
5.
go back to reference Park J, Kim HJ, Kim KR, Lee SK, Kim H, Park KK, Chung WY. Loss of RUNX3 expression inhibits bone invasion of oral squamous cell carcinoma. Oncotarget. 2016;8(6):9079.PubMedCentral Park J, Kim HJ, Kim KR, Lee SK, Kim H, Park KK, Chung WY. Loss of RUNX3 expression inhibits bone invasion of oral squamous cell carcinoma. Oncotarget. 2016;8(6):9079.PubMedCentral
6.
go back to reference Sato K, Tomizawa Y, Iijima H, Saito R, Ishizuka T, Nakajima T, Mori M. Epigenetic inactivation of the RUNX3 gene in lung cancer. Oncol Rep. 2006;15:129–35.PubMed Sato K, Tomizawa Y, Iijima H, Saito R, Ishizuka T, Nakajima T, Mori M. Epigenetic inactivation of the RUNX3 gene in lung cancer. Oncol Rep. 2006;15:129–35.PubMed
7.
go back to reference He L, Zhao X, Wang H, Zhang P, Guo C, Huang C, Liu X, Yao F, Chen Y, Lou W, Sun S, Fan D. RUNX3 mediates suppression of tumor growth and metastasis of human CCRCC by regulating cyclin related proteins and TIMP-1. PLoS ONE. 2012;7:e32961.CrossRefPubMedPubMedCentral He L, Zhao X, Wang H, Zhang P, Guo C, Huang C, Liu X, Yao F, Chen Y, Lou W, Sun S, Fan D. RUNX3 mediates suppression of tumor growth and metastasis of human CCRCC by regulating cyclin related proteins and TIMP-1. PLoS ONE. 2012;7:e32961.CrossRefPubMedPubMedCentral
8.
go back to reference Chen F, Liu X, Cheng Q, Zhu S, Bai J, Zheng J. RUNX3 regulates renal cell carcinoma metastasis via targeting miR-6780a-5p/E-cadherin/EMT signaling axis. Oncotarget. 2016;8(60):101042. Chen F, Liu X, Cheng Q, Zhu S, Bai J, Zheng J. RUNX3 regulates renal cell carcinoma metastasis via targeting miR-6780a-5p/E-cadherin/EMT signaling axis. Oncotarget. 2016;8(60):101042.
9.
go back to reference Chen F, Bai J, Li W, Mei P, Liu H, Li L, Pan Z, Wu Y, Zheng J. RUNX3 suppresses migration, invasion and angiogenesis of human renal cell carcinoma. PLoS ONE. 2013;8:e56241.CrossRefPubMedPubMedCentral Chen F, Bai J, Li W, Mei P, Liu H, Li L, Pan Z, Wu Y, Zheng J. RUNX3 suppresses migration, invasion and angiogenesis of human renal cell carcinoma. PLoS ONE. 2013;8:e56241.CrossRefPubMedPubMedCentral
10.
go back to reference Slattery ML, Lundgreen A, Herrick JS, Caan BJ, Potter JD, Wolff RK. Associations between genetic variation in RUNX1, RUNX2, RUNX3, MAPK1 and eIF4E and riskof colon and rectal cancer: additional support for a TGF-beta-signaling pathway. Carcinogenesis. 2011;32:318–26.CrossRefPubMed Slattery ML, Lundgreen A, Herrick JS, Caan BJ, Potter JD, Wolff RK. Associations between genetic variation in RUNX1, RUNX2, RUNX3, MAPK1 and eIF4E and riskof colon and rectal cancer: additional support for a TGF-beta-signaling pathway. Carcinogenesis. 2011;32:318–26.CrossRefPubMed
11.
go back to reference Mabuchi M, Kataoka H, Miura Y, Kim TS, Kawaguchi M, Ebi M, Tanaka M, Mori Y, Kubota E, Mizushima T, Shimura T, Mizoshita T, Tanida S, Kamiya T, Asai K, Joh T. Tumor suppressor, AT motif binding factor 1 (ATBF1), translocates to the nucleus with runt domain transcription factor 3 (RUNX3) in response to TGF-beta signal transduction. Biochem Biophys Res Commun. 2010;398:321–5.CrossRefPubMed Mabuchi M, Kataoka H, Miura Y, Kim TS, Kawaguchi M, Ebi M, Tanaka M, Mori Y, Kubota E, Mizushima T, Shimura T, Mizoshita T, Tanida S, Kamiya T, Asai K, Joh T. Tumor suppressor, AT motif binding factor 1 (ATBF1), translocates to the nucleus with runt domain transcription factor 3 (RUNX3) in response to TGF-beta signal transduction. Biochem Biophys Res Commun. 2010;398:321–5.CrossRefPubMed
12.
go back to reference Yokobori T, Nishiyama M. TGF-beta signaling in gastrointestinal cancers: progress in basic and clinical research. J Clin Med. 2017;6(1):11.CrossRefPubMedCentral Yokobori T, Nishiyama M. TGF-beta signaling in gastrointestinal cancers: progress in basic and clinical research. J Clin Med. 2017;6(1):11.CrossRefPubMedCentral
13.
go back to reference Sanjabi S, Oh SA, Li MO. Regulation of the immune response by TGF-beta: from conception to autoimmunity and infection. Cold Spring Harb Perspect Biol. 2017;9(6):a022236.CrossRefPubMed Sanjabi S, Oh SA, Li MO. Regulation of the immune response by TGF-beta: from conception to autoimmunity and infection. Cold Spring Harb Perspect Biol. 2017;9(6):a022236.CrossRefPubMed
14.
go back to reference Zaffaroni N, Mariani L, Giannatempo P, Nicolai N, Raggi D, Pennati M, Morosi C, Lanocita R, Crippa F, Daidone MG, Gianni AM, De Braud FG, Salvioni R. Interleukin-8 (IL8) and transforming growth-factor beta (TGF-beta) as drugable biomarkers of response, progression-free survival (PFS), and overall survival (OS) with pazopanib (PZP): a phase II study in relapsed urothelial cancer (UC). J Clin Oncol. 2012;30:78. Zaffaroni N, Mariani L, Giannatempo P, Nicolai N, Raggi D, Pennati M, Morosi C, Lanocita R, Crippa F, Daidone MG, Gianni AM, De Braud FG, Salvioni R. Interleukin-8 (IL8) and transforming growth-factor beta (TGF-beta) as drugable biomarkers of response, progression-free survival (PFS), and overall survival (OS) with pazopanib (PZP): a phase II study in relapsed urothelial cancer (UC). J Clin Oncol. 2012;30:78.
15.
go back to reference Melchionna R, Iapicca P, Di Modugno F, Trono P, Sperduti I, Fassan M, Cataldo I, Rusev BC, Lawlor RT, Diodoro MG, Milella M, Grazi GL, Bissell MJ, Scarpa A, Nistico P. The pattern of hMENA isoforms is regulated by TGF-beta1 in pancreatic cancer and may predict patient outcome. Oncoimmunology. 2016;5:e1221556.CrossRefPubMedPubMedCentral Melchionna R, Iapicca P, Di Modugno F, Trono P, Sperduti I, Fassan M, Cataldo I, Rusev BC, Lawlor RT, Diodoro MG, Milella M, Grazi GL, Bissell MJ, Scarpa A, Nistico P. The pattern of hMENA isoforms is regulated by TGF-beta1 in pancreatic cancer and may predict patient outcome. Oncoimmunology. 2016;5:e1221556.CrossRefPubMedPubMedCentral
16.
go back to reference Ruskyte K, Liutkeviciene R, Vilkeviciute A, Vaitkiene P, Valiulyte I, Glebauskiene B, Kriauciuniene L, Zaliuniene D. MMP-14 and TGFbeta-1 methylation in pituitary adenomas. Oncol Lett. 2016;12:3013–7.CrossRefPubMedPubMedCentral Ruskyte K, Liutkeviciene R, Vilkeviciute A, Vaitkiene P, Valiulyte I, Glebauskiene B, Kriauciuniene L, Zaliuniene D. MMP-14 and TGFbeta-1 methylation in pituitary adenomas. Oncol Lett. 2016;12:3013–7.CrossRefPubMedPubMedCentral
17.
go back to reference Moller M, Strand SH, Mundbjerg K, Liang G, Gill I, Haldrup C, Borre M, Hoyer S, Orntoft TF, Sorensen KD. Heterogeneous patterns of DNA methylation-based field effects in histologically normal prostate tissue from cancer patients. Sci Rep. 2017;7:40636.CrossRefPubMedPubMedCentral Moller M, Strand SH, Mundbjerg K, Liang G, Gill I, Haldrup C, Borre M, Hoyer S, Orntoft TF, Sorensen KD. Heterogeneous patterns of DNA methylation-based field effects in histologically normal prostate tissue from cancer patients. Sci Rep. 2017;7:40636.CrossRefPubMedPubMedCentral
18.
go back to reference Wang X, Wang J, Jia Y, Wang Y, Han X, Duan Y, Lv W, Ma M, Liu L. Methylation decreases the Bin1 tumor suppressor in ESCC and restoration by decitabine inhibits the epithelial mesenchymal transition. Oncotarget. 2017;8(12):19661.PubMedPubMedCentral Wang X, Wang J, Jia Y, Wang Y, Han X, Duan Y, Lv W, Ma M, Liu L. Methylation decreases the Bin1 tumor suppressor in ESCC and restoration by decitabine inhibits the epithelial mesenchymal transition. Oncotarget. 2017;8(12):19661.PubMedPubMedCentral
19.
go back to reference Wang H, Duan XL, Qi XL, Meng L, Xu YS, Wu T, Dai PG. Concurrent hypermethylation of SFRP2 and DKK2 activates the Wnt/beta-catenin pathway and is associated with poor prognosis in patients with gastric cancer. Mol Cells. 2017;40(1):45.CrossRefPubMedPubMedCentral Wang H, Duan XL, Qi XL, Meng L, Xu YS, Wu T, Dai PG. Concurrent hypermethylation of SFRP2 and DKK2 activates the Wnt/beta-catenin pathway and is associated with poor prognosis in patients with gastric cancer. Mol Cells. 2017;40(1):45.CrossRefPubMedPubMedCentral
20.
22.
go back to reference Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ, Lee KY, Nomura S, Lee CW, Han SB, Kim HM, Kim WJ, Yamamoto H, Yamashita N, Yano T, Ikeda T, Itohara S, Inazawa J, Abe T, Hagiwara A, Yamagishi H, Ooe A, Kaneda A, Sugimura T, Ushijima T, Bae SC, Ito Y. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell. 2002;109:113–24.CrossRefPubMed Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ, Lee KY, Nomura S, Lee CW, Han SB, Kim HM, Kim WJ, Yamamoto H, Yamashita N, Yano T, Ikeda T, Itohara S, Inazawa J, Abe T, Hagiwara A, Yamagishi H, Ooe A, Kaneda A, Sugimura T, Ushijima T, Bae SC, Ito Y. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell. 2002;109:113–24.CrossRefPubMed
23.
go back to reference Ku JL, Kang SB, Shin YK, Kang HC, Hong SH, Kim IJ, Shin JH, Han IO, Park JG. Promoter hypermethylation downregulates RUNX3 gene expression in colorectal cancer cell lines. Oncogene. 2004;23:6736–42.CrossRefPubMed Ku JL, Kang SB, Shin YK, Kang HC, Hong SH, Kim IJ, Shin JH, Han IO, Park JG. Promoter hypermethylation downregulates RUNX3 gene expression in colorectal cancer cell lines. Oncogene. 2004;23:6736–42.CrossRefPubMed
24.
go back to reference Goel A, Arnold CN, Tassone P, Chang DK, Niedzwiecki D, Dowell JM, Wasserman L, Compton C, Mayer RJ, Bertagnolli MM, Boland CR. Epigenetic inactivation of RUNX3 in microsatellite unstable sporadic colon cancers. Int J Cancer. 2004;112:754–9.CrossRefPubMed Goel A, Arnold CN, Tassone P, Chang DK, Niedzwiecki D, Dowell JM, Wasserman L, Compton C, Mayer RJ, Bertagnolli MM, Boland CR. Epigenetic inactivation of RUNX3 in microsatellite unstable sporadic colon cancers. Int J Cancer. 2004;112:754–9.CrossRefPubMed
25.
go back to reference Wada M, Yazumi S, Takaishi S, Hasegawa K, Sawada M, Tanaka H, Ida H, Sakakura C, Ito K, Ito Y, Chiba T. Frequent loss of RUNX3 gene expression in human bile duct and pancreatic cancer cell lines. Oncogene. 2004;23:2401–7.CrossRefPubMed Wada M, Yazumi S, Takaishi S, Hasegawa K, Sawada M, Tanaka H, Ida H, Sakakura C, Ito K, Ito Y, Chiba T. Frequent loss of RUNX3 gene expression in human bile duct and pancreatic cancer cell lines. Oncogene. 2004;23:2401–7.CrossRefPubMed
26.
go back to reference Ito Y, Miyazono K. RUNX transcription factors as key targets of TGF-beta superfamily signaling. Curr Opin Genet Dev. 2003;13:43–7.CrossRefPubMed Ito Y, Miyazono K. RUNX transcription factors as key targets of TGF-beta superfamily signaling. Curr Opin Genet Dev. 2003;13:43–7.CrossRefPubMed
27.
go back to reference Foroutan M, Cursons J, Hediyeh-Zadeh S, Thompson EW, Davis MJ. A transcriptional program for detecting TGFbeta-induced EMT in cancer. Mol Cancer Res. 2017;15(5):619–31.CrossRefPubMed Foroutan M, Cursons J, Hediyeh-Zadeh S, Thompson EW, Davis MJ. A transcriptional program for detecting TGFbeta-induced EMT in cancer. Mol Cancer Res. 2017;15(5):619–31.CrossRefPubMed
28.
go back to reference Kim YJ, Jeon Y, Kim T, Lim WC, Ham J, Park YN, Kim TJ, Ko H. Combined treatment with zingerone and its novel derivative synergistically inhibits TGF-beta1 induced epithelial-mesenchymal transition, migration and invasion of human hepatocellular carcinoma cells. Bioorg Med Chem Lett. 2016;27(4):1081–8.CrossRefPubMed Kim YJ, Jeon Y, Kim T, Lim WC, Ham J, Park YN, Kim TJ, Ko H. Combined treatment with zingerone and its novel derivative synergistically inhibits TGF-beta1 induced epithelial-mesenchymal transition, migration and invasion of human hepatocellular carcinoma cells. Bioorg Med Chem Lett. 2016;27(4):1081–8.CrossRefPubMed
Metadata
Title
DNA methylation affects metastasis of renal cancer and is associated with TGF-β/RUNX3 inhibition
Authors
Jianbo Zheng
Yanhui Mei
Ping Xiang
Guangsheng Zhai
Ning Zhao
Chuanbing Xu
Min Liu
Zhengsheng Pan
Kai Tang
Dongsheng Jia
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2018
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0554-7

Other articles of this Issue 1/2018

Cancer Cell International 1/2018 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