Genome-wide DNA methylation dynamics in carbon tetrachloride-induced mice liver fibrosis

Document Type : Original Article

Authors

1 State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China

2 Department of Physiology, Changzhi Medical College, Shanxi, China 2.Department of Physiology, Changzhi Medical College

Abstract

Objective(s): Many persistent harmful stimuli can result in chronic liver diseases, which lead to about 2 million deaths per year in the whole world. Liver fibrosis was found to exist in all kinds of chronic liver diseases. Many studies suggested that DNA methylation was associated with the pathogenesis of liver fibrosis. This study aimed to quantitatively detect DNA methylation changes in the whole genome in fibrotic liver tissues of mice.
Materials and Methods: Liver fibrosis was induced by intraperitoneal injection of carbon tetrachloride (CCl4) for 4 weeks. A genome-wide methylome analysis was performed using 850K BeadChips assays. The methylation status of 27 CpG dinucleotides located in 3 genes was detected by pyrosequencing to confirm chip data accuracy, and mRNA expressions of these 3 genes were examined by RT-qPCR methods.
Results: A total of 130,068 differentially methylated sites (DMS, 58,474 hypermethylated, and 71,594 hypomethylated) between fibrotic liver tissues and control mice liver tissues were identified by the 850k BeadChips array. Consistency between pyrosequencing data and 850k BeadChips array data was observed (R=0.928; P<0.01). Apoptosis, positive regulation of transcription of Notch receptor target, and negative regulation of p38MAPK signal cascade activities were significantly enriched in the Gene Ontology (GO) analyses. Cholesterol metabolism, bile secretion, and more biosynthesis and metabolism pathways were enriched in KEGG pathway analyses. Ten key genes were identified by the Cytoscape plugin cytoHubba. 
Conclusion: 7850 genes were found to have methylation change in fibrotic liver tissues of mice, which facilitates future research for clinical application.

Keywords


1. Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol 2019; 70:151-171.
2. Trautwein C, Friedman SL, Schuppan D, Pinzani M. Hepatic fibrosis: Concept to treatment. J Hepatol 2015; 62:S15-24.
3. Schuppan D, Ashfaq-Khan M, Yang AT, Kim YO. Liver fibrosis: Direct antifibrotic agents and targeted therapies. Matrix Biol 2018; 68-69:435-451.
4. Schaefer L. Decoding fibrosis: Mechanisms and translational aspects. Matrix Biol 2018; 68-69:1-7.
5. Ghosh AK, Quaggin SE, Vaughan DE. Molecular basis of organ fibrosis: Potential therapeutic approaches. Exp Biol Med (Maywood) 2013; 238:461-481.
6. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021; 18:151-166.
7. George J, Tsuchishima M, Tsutsumi M. Metabolism of N-nitrosodimethylamine, methylation of macromolecules, and development of hepatic fibrosis in rodent models. J Mol Med (Berl) 2020; 98:1203-1213.
8. Muriel P. Fighting liver fibrosis to reduce mortality associated with chronic liver diseases: The importance of new molecular targets and biomarkers. EBio Medicine 2019; 40:35-36.
9. Luangmonkong T, Suriguga S, Mutsaers HAM, Groothuis GMM, Olinga P, Boersema M. Targeting oxidative stress for the treatment of liver fibrosis. Rev Physiol Biochem Pharmacol 2018; 175:71-102.
10. Ying HZ, Chen Q, Zhang WY, Zhang HH, Ma Y, Zhang SZ, et al. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol Med Rep 2017; 16:7879-7889.
11. Xu F, Liu C, Zhou D, Zhang L. TGF-beta/SMAD pathway and its regulation in hepatic fibrosis. J Histochem Cytochem 2016; 64:157-167.
12. Nishikawa K, Osawa Y, Kimura K. Wnt/beta-catenin signaling as a potential target for the treatment of liver cirrhosis using antifibrotic drugs. Int J Mol Sci 2018; 19:3103.
13. Mederacke I, Hsu CC, Troeger JS, Huebener P, Mu X, Dapito DH, et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun 2013; 4:2823.
14. Forbes SJ, Russo FP, Rey V, Burra P, Rugge M, Wright NA, et al. A significant proportion of myofibroblasts are of bone marrow origin in human liver fibrosis. Gastroenterology 2004; 126:955-963.
15. Wells RG, Kruglov E, Dranoff JA. Autocrine release of TGF-beta by portal fibroblasts regulates cell growth. FEBS Lett 2004; 559:107-110.
16. Zeisberg M, Yang C, Martino M, Duncan MB, Rieder F, Tanjore H, et al. Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymal transition. J Biol Chem 2007; 282:23337-23347.
17. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003; 112:1776-1784.
18. Mutsaers SE, Birnie K, Lansley S, Herrick SE, Lim CB, Prele CM. Mesothelial cells in tissue repair and fibrosis. Front Pharmacol 2015; 6:113.
19. Turner BM. Cellular memory and the histone code. Cell 2002; 111:285-291.
20. O’Reilly S. Epigenetics in fibrosis. Mol Aspects Med 2017; 54:89-102.
21. Moran-Salvador E, Mann J. Epigenetics and liver fibrosis. Cell Mol Gastroenterol Hepatol 2017; 4:125-134.
22. Dowson C, O’Reilly S. DNA methylation in fibrosis. Eur J Cell Biol 2016; 95:323-330.
23. Shenker N, Flanagan JM. Intragenic DNA methylation: Implications of this epigenetic mechanism for cancer research. Br J Cancer 2012; 106:248-253.
24. Henderson NC, Rieder F, Wynn TA. Fibrosis: From mechanisms to medicines. Nature 2020; 587:555-566.
25. Castera L, Pinzani M. Biopsy and non-invasive methods for the diagnosis of liver fibrosis: does it take two to tango? Gut 2010; 59:861-866.
26. Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 2018; 34:i884-i890.
27. Krueger F, Andrews SR. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 2011; 27:1571-1572.
28. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9:357-359.
29. Akalin A, Kormaksson M, Li S, Garrett-Bakelman FE, Figueroa ME, Melnick A, et al. methylKit: A comprehensive R package for the analysis of genome-wide DNA methylation profiles. Genome Biol 2012; 13:R87.
30. Li S, Garrett-Bakelman FE, Akalin A, Zumbo P, Levine R, To BL, et al. An optimized algorithm for detecting and annotating regional differential methylation. BMC Bioinformatics 2013;14:S10.
31. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2 (-Delta Delta C(T)) method. Methods 2001; 25:402-408.
32. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, 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-D613.
33. Wilson CL, Mann DA, Borthwick LA. Epigenetic reprogramming in liver fibrosis and cancer. Adv Drug Deliv Rev 2017; 121:124-132.
34. Eden A, Gaudet F, Waghmare A, Jaenisch R. Chromosomal instability and tumors promoted by DNA hypomethylation. Science 2003; 300:455.
35. Komatsu Y, Waku T, Iwasaki N, Ono W, Yamaguchi C, Yanagisawa J. Global analysis of DNA methylation in early-stage liver fibrosis. BMC Med Genomics 2012; 5:5.
36. Bechtel W, McGoohan S, Zeisberg EM, Muller GA, Kalbacher H, Salant DJ, et al. Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat Med 2010; 16:544-550.
37. Mann J, Oakley F, Akiboye F, Elsharkawy A, Thorne AW, Mann DA. Regulation of myofibroblast transdifferentiation by DNA methylation and MeCP2: Implications for wound healing and fibrogenesis. Cell Death Differ 2007; 14:275-285.
38. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology 2008; 134:1655-1669.
39. Bakker W, Dingenouts CKE, Lodder K, Wiesmeijer KC, de Jong A, Kurakula K, et al. BMP receptor inhibition enhances tissue repair in endoglin heterozygous mice. Int J Mol Sci 2021; 22:2010.
40. Li J, Wang Y, Ma M, Jiang S, Zhang X, Zhang Y, et al. Autocrine CTHRC1 activates hepatic stellate cells and promotes liver fibrosis by activating TGF-beta signaling. EBioMedicine 2019; 40:43-55.
41. Bernabeu C, Lopez-Novoa JM, Quintanilla M. The emerging role of TGF-beta superfamily coreceptors in cancer. Biochim Biophys Acta 2009; 1792:954-973.
42. Clancy CE, Mendoza MG, Naismith TV, Kolman MF, Egelhoff TT. Identification of a protein kinase from dictyostelium with homology to the novel catalytic domain of myosin heavy chain kinase A. J Biol Chem 1997; 272:11812-11815.
43. Kuo TM, Yeh KT, Hsu HT, Chiang SL, Chang JG, Huang CM, et al. ALPK1 affects testosterone mediated regulation of proinflammatory cytokines production. J Steroid Biochem Mol Biol 2015; 154:150-158.
44. Williams LB, Javed A, Sabri A, Morgan DJ, Huff CD, Grigg JR, et al. ALPK1 missense pathogenic variant in five families leads to ROSAH syndrome, an ocular multisystem autosomal dominant disorder. Genet Med 2019; 21:2103-2115.
45. Chen M, Xu R. Motor coordination deficits in Alpk1 mutant mice with the inserted piggyBac transposon. BMC Neurosci 2011; 12:1.
46. Pullinger CR, Eng C, Salen G, Shefer S, Batta AK, Erickson SK, et al. Human cholesterol 7alpha-hydroxylase (CYP7A1) deficiency has a hypercholesterolemic phenotype. J Clin Invest 2002; 110:109-117.
47. Miyake JH, Duong-Polk XT, Taylor JM, Du EZ, Castellani LW, Lusis AJ, et al. Transgenic expression of cholesterol-7-alpha-hydroxylase prevents atherosclerosis in C57BL/6J mice. Arterioscler Thromb Vasc Biol 2002; 22:121-126.
48. Li T, Owsley E, Matozel M, Hsu P, Novak CM, Chiang JY. Transgenic expression of cholesterol 7alpha-hydroxylase in the liver prevents high-fat diet-induced obesity and insulin resistance in mice. Hepatology 2010; 52:678-690.
49. Yang JJ, Tao H, Huang C, Shi KH, Ma TT, Bian EB, et al. DNA methylation and MeCP2 regulation of PTCH1 expression during rats hepatic fibrosis. Cell Signal 2013; 25:1202-1211.
50. Yoshida T, Ogata H, Kamio M, Joo A, Shiraishi H, Tokunaga Y, et al. SOCS1 is a suppressor of liver fibrosis and hepatitis-induced carcinogenesis. J Exp Med 2004; 199:1701-1707.
51. Wu P, Huang R, Xiong YL, Wu C. Protective effects of curcumin against liver fibrosis through modulating DNA methylation. Chin J Nat Med 2016; 14:255-264.
52. Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol 2020; 38:675-678.
53. Liu Y, Sun J, Zhao M. ONGene: A literature-based database for human oncogenes. J Genet Genomics 2017; 44:119-121.
54. Song L, Jia J, Peng X, Xiao W, Li Y. The performance of the SEPT9 gene methylation assay and a comparison with other CRC screening tests: A meta-analysis. Sci Rep 2017; 7:3032.
55. Grant DM. Detoxification pathways in the liver. J Inherit Metab Dis 1991; 14:421-430.
56. Johnson A, DiPietro LA. Apoptosis and angiogenesis: An evolving mechanism for fibrosis. FASEB J 2013; 27:3893-3901.
57. Hu B, Phan SH. Notch in fibrosis and as a target of anti-fibrotic therapy. Pharmacol Res 2016; 108:57-64.
58. Gu L, Tao X, Xu Y, Han X, Qi Y, Xu L, et al. Dioscin alleviates BDL- and DMN-induced hepatic fibrosis via Sirt1/Nrf2-mediated inhibition of p38 MAPK pathway. Toxicol Appl Pharmacol 2016; 292:19-29.