Browse > Article
http://dx.doi.org/10.5713/ajas.15.0459

Characterization of the Methylation Status of Pax7 and Myogenic Regulator Factors in Cell Myogenic Differentiation  

Chao, Zhe (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Zheng, Xin-Li (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Sun, Rui-Ping (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Liu, Hai-Long (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Huang, Li-Li (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Cao, Zong-Xi (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Deng, Chang-Yan (College of Animal Science, Huazhong Agricultural University)
Wang, Feng (Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences)
Publication Information
Asian-Australasian Journal of Animal Sciences / v.29, no.7, 2016 , pp. 1037-1043 More about this Journal
Abstract
Epigenetic processes in the development of skeletal muscle have been appreciated for over a decade. DNA methylation is a major epigenetic modification important for regulating gene expression and suppressing spurious transcription. Up to now, the importance of epigenetic marks in the regulation of Pax7 and myogenic regulatory factors (MRFs) expression is far less explored. In the present study, semi-quantitative the real-time polymerase chain reaction (RT-PCR) analyses showed MyoD and Myf5 were expressed in activated and quiescent C2C12 cells. MyoG was expressed in a later stage of myogenesis. Pax7 was weakly expressed in differentiated C2C12 cells. To further understand the regulation of expression of these genes, the DNA methylation status of Pax7, MyoD, and Myf5 was determined by bisulfite sequencing PCR. During the C2C12 myoblasts fusion process, the changes of promoter and exon 1 methylation of Pax7, MyoD, and Myf5 genes were observed. In addition, an inverse relationship of low methylation and high expression was found. These results suggest that DNA methylation may be an important mechanism regulating Pax7 and MRFs transcription in cell myogenic differentiation.
Keywords
Pax7; Myogenic Regulatory Factors; C2C12; DNA Methylation; CpG Islands;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Abe, S., S. Rhee, O. Iwanuma, E. Hiroki, N. Yanagisawa, K. Sakiyama, and Y. Ide. 2009. Effect of mechanical stretching on expressions of muscle specific transcription factors MyoD, Myf5, Myogenin and MRF4 in proliferated myoblasts. Anat. Histol. Embryol. 38:305-310.   DOI
2 Baar, K. 2010. Epigenetic control of skeletal muscle fibre type. Acta. Physiol. 199:477487.
3 Butcher, L. M. and S. Beck. 2008. Future impact of integrated high-throughput methylome analyses on human health and disease. J. Genet. Genomics 35:391-401.   DOI
4 Caliaro, F., L. Maccatrozzo, L. Toniolo, C. Reggiani, F. Mascarello, and M. Patruno. 2005. Myogenic regulatory factors expressed during postnatal hyperplastic growth in porcine muscles. Basic Appl. Myol. 15:55-60.
5 Collins, C. A., V. F. Gnocchi, R. B. White, L. Boldrin, A. Perez-Ruiz, F. Relaix, J. E. Morgan, and P. S. Zammit. 2009. Integrated functions of Pax3 and Pax7 in the regulation of proliferation, cell size and myogenic differentiation. PloS one 4:e4475.   DOI
6 Dedieu, S., G. Mazeres, P. Cottin, and J. J. Brustis. 2002. Involvement of myogenic regulator factors during fusion in the cell line C. Int. J. Dev. Biol. 46:235-241.
7 Eckhardt, F., J. Lewin, R. Cortese, V. K. Rakyan, J. Attwood, M. Burger, J. Burton, T. V. Cox, R. Davies, and T. A. Down et al. 2006. DNA methylation profiling of human chromosomes 6, 20 and 22. Nat. Genet. 38:1378-1385.   DOI
8 Fan, H., M. U. Cinar, C. Phatsara, D. Tesfaye, E. Tholen, C. Looft, and K. Schellander. 2011. Molecular mechanism underlying the differential MYF6 expression in postnatal skeletal muscle of Duroc and Pietrain breeds. Gene 486:8-14.   DOI
9 Fan, X., M. Inda, T. Tunon, and J. S. Castresana. 2002. Improvement of the methylation specific PCR technical conditions for the detection of p16 promoter hypermethylation in small amounts of tumor DNA. Oncol. Rep. 9:181-183.
10 Frommer, M., L. E. McDonald, D. S. Millar, C. M. Collis, F. Watt, G. W. Grigg, P. L. Molloy, and C. L. Paul. 1992. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc. Natil. Acad. Sci. USA. 89:1827-1831.   DOI
11 Ghai, S., R. Monga, T. K. Mohanty, M. S. Chauhan, and D. Singh. 2010. Tissue-specific promoter methylation coincides with Cyp19 gene expression in buffalo (Bubalus bubalis) placenta of different stages of gestation. Gen. Comp. Endocrinol. 169:182-189.   DOI
12 Horst, D., S. Ustanina, C. Sergi, G. Mikuz, H. Jurgens, T. Braun, and E. Vorobyov. 2006. Comparative expression analysis of Pax3 and Pax7 during mouse myogenesis. Int. J. Dev. Biol. 50:47-54.   DOI
13 Kim, S. K., H. R. Jang, J. H. Kim, M. Kim, S. M. Noh, K. S. Song, G. H. Kang, H. J. Kim, S. Y. Kim, and H. S. Yoo, and Y. S. Kim. 2008. CpG methylation in exon 1 of transcription factor 4 increases with age in normal gastric mucosa and is associated with gene silencing in intestinal-type gastric cancers. Carcinogenesis 29:1623-1631.   DOI
14 Liu, G. Q., K. Zong, L. L. Zhang, and S. Q. Cao. 2010. Dietary methionine affect meat qulity and myostatin gene exon 1 region methylation in skeletal muscle tissues of broilers. Agric. Sci. China 9:1338-1346.   DOI
15 Qiao. M., H. Y. Wu, F. E. Li, S. W. Jiang, Y. Z. Xiong, and C. Y. Deng. 2010. Molecular characterization, expression profile and association analysis with carcass traits of porcine LCAT gene. Mol. Biol. Rep. 37:2227-2234.   DOI
16 Nakamura, J., T. Tanaka, Y. Kitajima, H. Noshiro, and K. Miyazaki. 2014. Methylation-mediated gene silencing as biomarkers of gastric cancer: a review. World J. Gastroenterol. 20:11991-12006.   DOI
17 Olguin, H. C., Z. Yang, S. J. Tapscott, and B. B. Olwin. 2007. Reciprocal inhibition between Pax7 and muscle regulatory factors modulates myogenic cell fate determination. J. Cell Biol. 177:769-779.   DOI
18 Palacios, D. and P. L. Puri. 2006. The epigenetic network regulating muscle development and regeneration. J. Cell. Physiol. 207:1-11.   DOI
19 Steffens, A. A., G. M. Hong, and L. J. Bain. 2011. Sodium arsenite delays the differentiation of C2C12 mouse myoblast cells and alters methylation patterns on the transcription factor myogenin. Toxicol. Appl. Pharmacol. 250:154-161.   DOI
20 Takai, D. and P. A. Jones. 2002. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc. Nati. Acad. Sci. USA. 99:3740-3745.   DOI
21 Verner, J., P. Humpolícek, and A. Knoll. 2007. Impact of MYOD family genes on pork traits in Large White and Landrace pigs. J. Anim. Breed. Genet. 124:81-85.   DOI
22 Weintraub, H. 1993. The MyoD family and myogenesis: Redundancy, networks, and thresholds. Cell 75:1241-1244.   DOI
23 Wu, Q., R. A. Lothe, T. Ahlquist, I. Silins, C. G. Tropé, F. Micci, J. M. Nesland, Z. Suo, and G. E. Lind. 2007. DNA methylation profiling of ovarian carcinomas and their in vitro models identifies HOXA9, HOXB5, SCGB3A1, and CRABP1 as novel targets. Mol. Cancer 6:45.   DOI
24 Zelko, I. N., M. R. Mueller, and R. J. Folz. 2010. CpG methylation attenuates Sp1 and Sp3 binding to the human extracellular superoxide dismutase promoter and regulates its cell-specific expression. Free Radic. Biol. Med. 48:895-904.   DOI
25 Xiong, Q., J. Chai, P. P. Zhang, J. Wu, S. W. Jiang, R. Zheng, and C. Y. Deng. 2011. MyoD control of SKIP expression during pig skeletal muscle development. Mol. Biol. Rep. 38:267-274.   DOI
26 Yeo, S., S. Jeong, J. Kim, J. S. Han, Y. M. Han, and Y. K. Kang. 2007. Characterization of DNA methylation change in stem cell marker genes during differentiation of human embryonic stem cells. Biochem. Biophys. Res. Commun. 359:536-542.   DOI