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

Open Access 01-12-2018 | Research article

The DNA methylation profile of liver tumors in C3H mice and identification of differentially methylated regions involved in the regulation of tumorigenic genes

Authors: Junya Matsushita, Kazuyuki Okamura, Kazuhiko Nakabayashi, Takehiro Suzuki, Yu Horibe, Tomoko Kawai, Toshihiro Sakurai, Satoshi Yamashita, Yoshikazu Higami, Gaku Ichihara, Kenichiro Hata, Keiko Nohara

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

C3H mice have been frequently used in cancer studies as animal models of spontaneous liver tumors and chemically induced hepatocellular carcinoma (HCC). Epigenetic modifications, including DNA methylation, are among pivotal control mechanisms of gene expression leading to carcinogenesis. Although information on somatic mutations in liver tumors of C3H mice is available, epigenetic aspects are yet to be clarified.

Methods

We performed next generation sequencing-based analysis of DNA methylation and microarray analysis of gene expression to explore genes regulated by DNA methylation in spontaneous liver tumors of C3H mice. Overlaying these data, we selected cancer-related genes whose expressions are inversely correlated with DNA methylation levels in the associated differentially methylated regions (DMRs) located around transcription start sites (TSSs) (promoter DMRs). We further assessed mutuality of the selected genes for expression and DNA methylation in human HCC using the Cancer Genome Atlas (TCGA) database.

Results

We obtained data on genome-wide DNA methylation profiles in the normal and tumor livers of C3H mice. We identified promoter DMRs of genes which are reported to be related to cancer and whose expressions are inversely correlated with the DNA methylation, including Mst1r, Slpi and Extl1. The association between DNA methylation and gene expression was confirmed using a DNA methylation inhibitor 5-aza-2′-deoxycytidine (5-aza-dC) in Hepa1c1c7 cells and Hepa1-6 cells. Overexpression of Mst1r in Hepa1c1c7 cells illuminated a novel downstream pathway via IL-33 upregulation. Database search indicated that gene expressions of Mst1r and Slpi are upregulated and the TSS upstream regions are hypomethylated also in human HCC. These results suggest that DMRs, including those of Mst1r and Slpi, are involved in liver tumorigenesis in C3H mice, and also possibly in human HCC.

Conclusions

Our study clarified genome wide DNA methylation landscape of C3H mice. The data provide useful information for further epigenetic studies of mice models of HCC. The present study particularly proposed novel DNA methylation-regulated pathways for Mst1r and Slpi, which may be applied not only to mouse HCC but also to human HCC.
Appendix
Available only for authorised users
Literature
2.
go back to reference Maronpot RR, Fox T, Malarkey DE, Goldsworthy TL. Mutations in the ras proto-oncogene: clues to etiology and molecular pathogenesis of mouse liver tumors. Toxicology. 1995;101(3):125–56.CrossRefPubMed Maronpot RR, Fox T, Malarkey DE, Goldsworthy TL. Mutations in the ras proto-oncogene: clues to etiology and molecular pathogenesis of mouse liver tumors. Toxicology. 1995;101(3):125–56.CrossRefPubMed
3.
go back to reference Delgado E, Okabe H, Preziosi M, Russell JO, Alvarado TF, Oertel M, Nejak-Bowen KN, Zhang Y, Monga SP. Complete response of Ctnnb1-mutated tumours to beta-catenin suppression by locked nucleic acid antisense in a mouse hepatocarcinogenesis model. J Hepatol. 2015;62(2):380–7.CrossRefPubMed Delgado E, Okabe H, Preziosi M, Russell JO, Alvarado TF, Oertel M, Nejak-Bowen KN, Zhang Y, Monga SP. Complete response of Ctnnb1-mutated tumours to beta-catenin suppression by locked nucleic acid antisense in a mouse hepatocarcinogenesis model. J Hepatol. 2015;62(2):380–7.CrossRefPubMed
4.
go back to reference Chen X, Yamamoto M, Fujii K, Nagahama Y, Ooshio T, Xin B, Okada Y, Furukawa H, Nishikawa Y. Differential reactivation of fetal/neonatal genes in mouse liver tumors induced in cirrhotic and non-cirrhotic conditions. Cancer Sci. 2015;106(8):972–81.CrossRefPubMedPubMedCentral Chen X, Yamamoto M, Fujii K, Nagahama Y, Ooshio T, Xin B, Okada Y, Furukawa H, Nishikawa Y. Differential reactivation of fetal/neonatal genes in mouse liver tumors induced in cirrhotic and non-cirrhotic conditions. Cancer Sci. 2015;106(8):972–81.CrossRefPubMedPubMedCentral
5.
go back to reference Buchmann A, Karcier Z, Schmid B, Strathmann J, Schwarz M. Differential selection for B-raf and ha-ras mutated liver tumors in mice with high and low susceptibility to hepatocarcinogenesis. Mutat Res. 2008;638(1–2):66–74.CrossRefPubMed Buchmann A, Karcier Z, Schmid B, Strathmann J, Schwarz M. Differential selection for B-raf and ha-ras mutated liver tumors in mice with high and low susceptibility to hepatocarcinogenesis. Mutat Res. 2008;638(1–2):66–74.CrossRefPubMed
6.
go back to reference Reiberger T, Chen Y, Ramjiawan RR, Hato T, Fan C, Samuel R, Roberge S, Huang P, Lauwers GY, Zhu AX, et al. An orthotopic mouse model of hepatocellular carcinoma with underlying liver cirrhosis. Nat Protoc. 2015;10(8):1264–74.CrossRefPubMedPubMedCentral Reiberger T, Chen Y, Ramjiawan RR, Hato T, Fan C, Samuel R, Roberge S, Huang P, Lauwers GY, Zhu AX, et al. An orthotopic mouse model of hepatocellular carcinoma with underlying liver cirrhosis. Nat Protoc. 2015;10(8):1264–74.CrossRefPubMedPubMedCentral
8.
go back to reference Jaworski M, Buchmann A, Bauer P, Riess O, Schwarz M. B-raf and ha-ras mutations in chemically induced mouse liver tumors. Oncogene. 2005;24(7):1290–5.CrossRefPubMed Jaworski M, Buchmann A, Bauer P, Riess O, Schwarz M. B-raf and ha-ras mutations in chemically induced mouse liver tumors. Oncogene. 2005;24(7):1290–5.CrossRefPubMed
9.
go back to reference Nohara K, Okamura K, Suzuki T, Murai H, Ito T, Shinjo K, Takumi S, Michikawa T, Kondo Y, Hata K. Augmenting effects of gestational arsenite exposure of C3H mice on the hepatic tumors of the F(2) male offspring via the F(1) male offspring. J Appl Toxicol. 2016;36(1):105–12.CrossRefPubMed Nohara K, Okamura K, Suzuki T, Murai H, Ito T, Shinjo K, Takumi S, Michikawa T, Kondo Y, Hata K. Augmenting effects of gestational arsenite exposure of C3H mice on the hepatic tumors of the F(2) male offspring via the F(1) male offspring. J Appl Toxicol. 2016;36(1):105–12.CrossRefPubMed
11.
go back to reference Malouf GG, Tahara T, Paradis V, Fabre M, Guettier C, Yamazaki J, Long H, Lu Y, Raynal NJ, Jelinek J, et al. Methylome sequencing for fibrolamellar hepatocellular carcinoma depicts distinctive features. Epigenetics. 2015;10(9):872–81.CrossRefPubMedPubMedCentral Malouf GG, Tahara T, Paradis V, Fabre M, Guettier C, Yamazaki J, Long H, Lu Y, Raynal NJ, Jelinek J, et al. Methylome sequencing for fibrolamellar hepatocellular carcinoma depicts distinctive features. Epigenetics. 2015;10(9):872–81.CrossRefPubMedPubMedCentral
12.
go back to reference Suzuki T, Yamashita S, Ushijima T, Takumi S, Sano T, Michikawa T, Nohara K. Genome-wide analysis of DNA methylation changes induced by gestational arsenic exposure in liver tumors. Cancer Sci. 2013;104(12):1575–85.CrossRefPubMed Suzuki T, Yamashita S, Ushijima T, Takumi S, Sano T, Michikawa T, Nohara K. Genome-wide analysis of DNA methylation changes induced by gestational arsenic exposure in liver tumors. Cancer Sci. 2013;104(12):1575–85.CrossRefPubMed
13.
go back to reference Tirado-Magallanes R, Rebbani K, Lim R, Pradhan S, Benoukraf T. Whole genome DNA methylation: beyond genes silencing. Oncotarget. 2017;8(3):5629–37.CrossRefPubMed Tirado-Magallanes R, Rebbani K, Lim R, Pradhan S, Benoukraf T. Whole genome DNA methylation: beyond genes silencing. Oncotarget. 2017;8(3):5629–37.CrossRefPubMed
14.
go back to reference Meissner A, Gnirke A, Bell GW, Ramsahoye B, Lander ES, Jaenisch R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 2005;33(18):5868–77.CrossRefPubMedPubMedCentral Meissner A, Gnirke A, Bell GW, Ramsahoye B, Lander ES, Jaenisch R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 2005;33(18):5868–77.CrossRefPubMedPubMedCentral
15.
go back to reference Boyle P, Clement K, Gu H, Smith ZD, Ziller M, Fostel JL, Holmes L, Meldrim J, Kelley F, Gnirke A, et al. Gel-free multiplexed reduced representation bisulfite sequencing for large-scale DNA methylation profiling. Genome Biol. 2012;13(10):R92.CrossRefPubMedPubMedCentral Boyle P, Clement K, Gu H, Smith ZD, Ziller M, Fostel JL, Holmes L, Meldrim J, Kelley F, Gnirke A, et al. Gel-free multiplexed reduced representation bisulfite sequencing for large-scale DNA methylation profiling. Genome Biol. 2012;13(10):R92.CrossRefPubMedPubMedCentral
16.
go back to reference De Souza AP, Planello AC, Marques MR, De Carvalho DD, Line SR. High-throughput DNA analysis shows the importance of methylation in the control of immune inflammatory gene transcription in chronic periodontitis. Clin Epigenetics. 2014;6(1):15.CrossRefPubMedPubMedCentral De Souza AP, Planello AC, Marques MR, De Carvalho DD, Line SR. High-throughput DNA analysis shows the importance of methylation in the control of immune inflammatory gene transcription in chronic periodontitis. Clin Epigenetics. 2014;6(1):15.CrossRefPubMedPubMedCentral
17.
go back to reference Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41(2):178–86.CrossRefPubMedPubMedCentral Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41(2):178–86.CrossRefPubMedPubMedCentral
18.
go back to reference Nohara K, Tateishi Y, Suzuki T, Okamura K, Murai H, Takumi S, Maekawa F, Nishimura N, Kobori M, Ito T. Late-onset increases in oxidative stress and other tumorigenic activities and tumors with a ha-ras mutation in the liver of adult male C3H mice gestationally exposed to arsenic. Toxicol Sci. 2012;129(2):293–304.CrossRefPubMed Nohara K, Tateishi Y, Suzuki T, Okamura K, Murai H, Takumi S, Maekawa F, Nishimura N, Kobori M, Ito T. Late-onset increases in oxidative stress and other tumorigenic activities and tumors with a ha-ras mutation in the liver of adult male C3H mice gestationally exposed to arsenic. Toxicol Sci. 2012;129(2):293–304.CrossRefPubMed
19.
go back to reference Suzuki T, Nohara K. Regulatory factors involved in species-specific modulation of arylhydrocarbon receptor (AhR)-dependent gene expression in humans and mice. J Biochem. 2007;142(4):443–52.CrossRefPubMed Suzuki T, Nohara K. Regulatory factors involved in species-specific modulation of arylhydrocarbon receptor (AhR)-dependent gene expression in humans and mice. J Biochem. 2007;142(4):443–52.CrossRefPubMed
20.
21.
go back to reference Akalin A, Kormaksson M, Li S, Garrett-Bakelman FE, Figueroa ME, Melnick A. Mason CE: methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles. Genome Biol. 2012;13(10):R87.CrossRefPubMedPubMedCentral Akalin A, Kormaksson M, Li S, Garrett-Bakelman FE, Figueroa ME, Melnick A. Mason CE: methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles. Genome Biol. 2012;13(10):R87.CrossRefPubMedPubMedCentral
22.
go back to reference Li S, Garrett-Bakelman FE, Akalin A, Zumbo P, Levine R, To BL, Lewis ID, Brown AL, D'Andrea RJ, Melnick A, et al. An optimized algorithm for detecting and annotating regional differential methylation. BMC Bioinformatics. 2013;14(Suppl 5):S10.CrossRefPubMedPubMedCentral Li S, Garrett-Bakelman FE, Akalin A, Zumbo P, Levine R, To BL, Lewis ID, Brown AL, D'Andrea RJ, Melnick A, et al. An optimized algorithm for detecting and annotating regional differential methylation. BMC Bioinformatics. 2013;14(Suppl 5):S10.CrossRefPubMedPubMedCentral
23.
go back to reference Takumi S, Aoki Y, Sano T, Suzuki T, Nohmi T, Nohara K. In vivo mutagenicity of arsenite in the livers of gpt delta transgenic mice. Mutat Res Genet Toxicol Environ Mutagen. 2014;760:42–7.CrossRefPubMed Takumi S, Aoki Y, Sano T, Suzuki T, Nohmi T, Nohara K. In vivo mutagenicity of arsenite in the livers of gpt delta transgenic mice. Mutat Res Genet Toxicol Environ Mutagen. 2014;760:42–7.CrossRefPubMed
24.
go back to reference Nohara K, Ao K, Miyamoto Y, Ito T, Suzuki T, Toyoshiba H, Tohyama C. Comparison of the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1A1 gene expression profile in lymphocytes from mice, rats, and humans: most potent induction in humans. Toxicology. 2006;225(2–3):204–13.CrossRefPubMed Nohara K, Ao K, Miyamoto Y, Ito T, Suzuki T, Toyoshiba H, Tohyama C. Comparison of the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1A1 gene expression profile in lymphocytes from mice, rats, and humans: most potent induction in humans. Toxicology. 2006;225(2–3):204–13.CrossRefPubMed
25.
go back to reference Sandelin A, Alkema W, Engstrom P, Wasserman WW, Lenhard B. JASPAR: an open-access database for eukaryotic transcription factor binding profiles. Nucleic Acids Res. 2004;32(Database issue):D91–4.CrossRefPubMedPubMedCentral Sandelin A, Alkema W, Engstrom P, Wasserman WW, Lenhard B. JASPAR: an open-access database for eukaryotic transcription factor binding profiles. Nucleic Acids Res. 2004;32(Database issue):D91–4.CrossRefPubMedPubMedCentral
26.
go back to reference Camp ER, Liu W, Fan F, Yang A, Somcio R, Ellis LM. RON, a tyrosine kinase receptor involved in tumor progression and metastasis. Ann Surg Oncol. 2005;12(4):273–81.CrossRefPubMed Camp ER, Liu W, Fan F, Yang A, Somcio R, Ellis LM. RON, a tyrosine kinase receptor involved in tumor progression and metastasis. Ann Surg Oncol. 2005;12(4):273–81.CrossRefPubMed
27.
go back to reference Yao HP, Zhou YQ, Zhang R, Wang MH. MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13(7):466–81.CrossRefPubMed Yao HP, Zhou YQ, Zhang R, Wang MH. MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13(7):466–81.CrossRefPubMed
28.
go back to reference Nukiwa T, Suzuki T, Fukuhara T, Kikuchi T. Secretory leukocyte peptidase inhibitor and lung cancer. Cancer Sci. 2008;99(5):849–55.CrossRefPubMed Nukiwa T, Suzuki T, Fukuhara T, Kikuchi T. Secretory leukocyte peptidase inhibitor and lung cancer. Cancer Sci. 2008;99(5):849–55.CrossRefPubMed
29.
go back to reference Mathysen D, Van Roy N, Van Hul W, Laureys G, Ambros P, Speleman F, Wuyts W. Molecular analysis of the putative tumour-suppressor gene EXTL1 in neuroblastoma patients and cell lines. Eur J Cancer. 2004;40(8):1255–61.CrossRefPubMed Mathysen D, Van Roy N, Van Hul W, Laureys G, Ambros P, Speleman F, Wuyts W. Molecular analysis of the putative tumour-suppressor gene EXTL1 in neuroblastoma patients and cell lines. Eur J Cancer. 2004;40(8):1255–61.CrossRefPubMed
31.
go back to reference Ettou S, Humbrecht C, Benet B, Billot K, d'Allard D, Mariot V, Goodhardt M, Kosmider O, Mayeux P, Solary E, et al. Epigenetic control of NF-kappaB-dependent FAS gene transcription during progression of myelodysplastic syndromes. Mol Cancer Res. 2013;11(7):724–35.CrossRefPubMed Ettou S, Humbrecht C, Benet B, Billot K, d'Allard D, Mariot V, Goodhardt M, Kosmider O, Mayeux P, Solary E, et al. Epigenetic control of NF-kappaB-dependent FAS gene transcription during progression of myelodysplastic syndromes. Mol Cancer Res. 2013;11(7):724–35.CrossRefPubMed
32.
go back to reference Shen ES, Whitlock JP Jr. The potential role of DNA methylation in the response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Biol Chem. 1989;264(30):17754–8.PubMed Shen ES, Whitlock JP Jr. The potential role of DNA methylation in the response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Biol Chem. 1989;264(30):17754–8.PubMed
33.
go back to reference Takahashi Y, Suzuki C, Kamataki T. Silencing of CYP1A1 expression in rabbits by DNA methylation. Biochem Biophys Res Commun. 1998;247(2):383–6.CrossRefPubMed Takahashi Y, Suzuki C, Kamataki T. Silencing of CYP1A1 expression in rabbits by DNA methylation. Biochem Biophys Res Commun. 1998;247(2):383–6.CrossRefPubMed
35.
go back to reference Zheng D, Gui B, Gray KP, Tinay I, Rafiei S, Huang Q, Sweeney CJ, Kibel AS, Jia L. Secretory leukocyte protease inhibitor is a survival and proliferation factor for castration-resistant prostate cancer. Oncogene. 2016;35(36):4807–15.CrossRefPubMed Zheng D, Gui B, Gray KP, Tinay I, Rafiei S, Huang Q, Sweeney CJ, Kibel AS, Jia L. Secretory leukocyte protease inhibitor is a survival and proliferation factor for castration-resistant prostate cancer. Oncogene. 2016;35(36):4807–15.CrossRefPubMed
36.
go back to reference Honda S, Minato M, Suzuki H, Fujiyoshi M, Miyagi H, Haruta M, Kaneko Y, Hatanaka KC, Hiyama E, Kamijo T, et al. Clinical prognostic value of DNA methylation in hepatoblastoma: four novel tumor suppressor candidates. Cancer Sci. 2016;107(6):812–9.CrossRefPubMedPubMedCentral Honda S, Minato M, Suzuki H, Fujiyoshi M, Miyagi H, Haruta M, Kaneko Y, Hatanaka KC, Hiyama E, Kamijo T, et al. Clinical prognostic value of DNA methylation in hepatoblastoma: four novel tumor suppressor candidates. Cancer Sci. 2016;107(6):812–9.CrossRefPubMedPubMedCentral
37.
go back to reference Yang Y, Wang JB, Li YM, Zhao YU, Wang R, Wu Q, Zheng RS, Ou YR. Role of IL-33 expression in oncogenesis and development of human hepatocellular carcinoma. Oncol Lett. 2016;12(1):429–36.CrossRefPubMedPubMedCentral Yang Y, Wang JB, Li YM, Zhao YU, Wang R, Wu Q, Zheng RS, Ou YR. Role of IL-33 expression in oncogenesis and development of human hepatocellular carcinoma. Oncol Lett. 2016;12(1):429–36.CrossRefPubMedPubMedCentral
38.
go back to reference Luedde T, Schwabe RF. NF-kappaB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2011;8(2):108–18.CrossRefPubMedPubMedCentral Luedde T, Schwabe RF. NF-kappaB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2011;8(2):108–18.CrossRefPubMedPubMedCentral
39.
go back to reference Hartwell HJ, Petrosky KY, Fox JG, Horseman ND, Rogers AB. Prolactin prevents hepatocellular carcinoma by restricting innate immune activation of c-Myc in mice. Proc Natl Acad Sci U S A. 2014;111(31):11455–60.CrossRefPubMedPubMedCentral Hartwell HJ, Petrosky KY, Fox JG, Horseman ND, Rogers AB. Prolactin prevents hepatocellular carcinoma by restricting innate immune activation of c-Myc in mice. Proc Natl Acad Sci U S A. 2014;111(31):11455–60.CrossRefPubMedPubMedCentral
40.
go back to reference Chen GG, Leung J, Liang NC, Li L, Wu K, Chan UP, Leung BC, Li M, Du J, Deng YF, et al. Ent-11alpha-hydroxy-15-oxo-kaur-16-en-19-oic-acid inhibits hepatocellular carcinoma in vitro and in vivo via stabilizing IkBalpha. Investig New Drugs. 2012;30(6):2210–8.CrossRef Chen GG, Leung J, Liang NC, Li L, Wu K, Chan UP, Leung BC, Li M, Du J, Deng YF, et al. Ent-11alpha-hydroxy-15-oxo-kaur-16-en-19-oic-acid inhibits hepatocellular carcinoma in vitro and in vivo via stabilizing IkBalpha. Investig New Drugs. 2012;30(6):2210–8.CrossRef
Metadata
Title
The DNA methylation profile of liver tumors in C3H mice and identification of differentially methylated regions involved in the regulation of tumorigenic genes
Authors
Junya Matsushita
Kazuyuki Okamura
Kazuhiko Nakabayashi
Takehiro Suzuki
Yu Horibe
Tomoko Kawai
Toshihiro Sakurai
Satoshi Yamashita
Yoshikazu Higami
Gaku Ichihara
Kenichiro Hata
Keiko Nohara
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4221-0

Other articles of this Issue 1/2018

BMC Cancer 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