Browse > Article
http://dx.doi.org/10.5713/ab.20.0585

Growth factors improve the proliferation of Jeju black pig muscle cells by regulating myogenic differentiation 1 and growth-related genes  

Park, Jinryong (Department of Animal Biotechnology, Jeonbuk National University)
Lee, Jeongeun (Department of Animal Biotechnology, Jeonbuk National University)
Song, Ki-Duk (The Animal Molecular Genetics and Breeding Center, Jeonbuk National University)
Kim, Sung-Jo (Division of Cosmetics and Biotechnology, Hoseo University)
Kim, Dae Cheol (Livestock Promotion Institute, Jeju Special Self-Governing Province)
Lee, Sang Cheol (Cronex Inc)
Son, Young June (Cronex Inc)
Choi, Hyun Woo (Department of Agricultural Convergence Technology, Jeonbuk National University)
Shim, Kwanseob (Department of Animal Biotechnology, Jeonbuk National University)
Publication Information
Animal Bioscience / v.34, no.8, 2021 , pp. 1392-1402 More about this Journal
Abstract
Objective: The growth rate of pigs is related to differentiation and proliferation of muscle cells, which are regulated by growth factors and expression of growth-related genes. Thus, the objective of this study was to establish optimal culture conditions for Jeju black pig (JBP) muscle cells and determine the relationship of various factors involved in muscle growth with the proliferation of JBP muscle cells. Methods: Muscles were taken from the femur skeletal muscle of JBP embryos. After isolation of the muscle cells, cells were cultured in a 6-well plate under four different culture conditions to optimize culture conditions for JBP muscle cells. To analyze proliferation rate of JBP muscle cells, these muscle cells were seeded into 6-well plates at a density of 1.5×105 cells per well and cultured for 3 days. Western blot and quantitative real-time polymerase chain reaction were applied to verify the myogenic differentiation 1 (MyoD) expression and growth-related gene expression in JBP muscle cells, respectively. Results: We established a muscle cell line from JBP embryos and optimized its culture conditions. These muscle cells were positive for MyoD, but not for paired box 7. The proliferation rate of these muscle cells was significantly higher in a culture medium containing bFGF and epidermal growth factor + basic fibroblast growth factor (EGF+bFGF) than that without a growth factor or containing EGF alone. Treatment with EGF and bFGF significantly induced the expression of MyoD protein, an important transcription factor in muscle cells. Moreover, we checked the changes of expression of growth-related genes in JBP muscle cells by presence or absence of growth factors. Expression level of collagen type XXI alpha 1 gene was changed only when EGF and bFGF were added together to culture media for JBP muscle cells. Conclusion: Concurrent use of EGF and bFGF increased the expression of MyoD protein, thus regulating the proliferation of JBP muscle cells and the expression of growth-related genes.
Keywords
Pig; Muscle Cell; Growth Factor; MyoD; Growth-related Gene;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Ben-Arye T, Levenberg S. Tissue engineering for clean meat production. Front Sustain Food Syst 2019;3:46. https://doi.org/10.3389/fsufs.2019.00046   DOI
2 Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Physiol Regul Integr Comp Physiol 2010;298:R1173-87. https://doi.org/10.1152/ajpregu.00735.2009   DOI
3 Hernandez-Hernandez JM, Garcia-Gonzalez EG, Brun CE, Rudnicki MA. The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration. Semin Cell Dev Biol 2017;72:10-8. https://doi.org/10.1016/j.semcdb.2017.11.010   DOI
4 Brocco M, Pollevick GD, Frasch ACC. Differential regulation of polysialyltransferase expression during hippocampus development: implications for neuronal survival. J Neurosci Res 2003;74:744-53. https://doi.org/10.1002/jnr.10781   DOI
5 Yucel N, Blau HM. Skeletal muscle stem cells. In: Atala A, Lanza R, Mikos T, Nerem R, editors. Principles of Regenerative Medicine, 3rd ed. London UK: Academic Press; 2019. pp. 273-93.
6 Ghosh M, Sodhi SS, Sharma N, et al. An integrated in silico approach for functional and structural impact of non- synonymous SNPs in the MYH1 gene in Jeju Native Pigs. BMC Genet 2016;17:35. https://doi.org/10.1186/s12863-016-0341-1   DOI
7 Andres-Mateos E, Mejias R, Soleimani A, et al. Impaired skeletal muscle regeneration in the absence of fibrosis during hibernation in 13-lined ground squirrels. PLoS One 2012;7: e48884. https://doi.org/10.1371/journal.pone.0048884   DOI
8 Tehrani HJ, Parivar K, Ai J, et al. Effect of dexamethasone, insulin and EGF on the myogenic potential on human endometrial stem cell. Iran J Pharm Sci 2014;13:659-64.
9 Blais A, Tsikitis M, Acosta-Alvear D, Sharan R, Kluger Y, Dynlacht BD. An initial blueprint for myogenic differentiation. Gene Dev 2005;19:553-69. https://doi.org/10.1101/gad.1281105   DOI
10 Cao Y, Yao Z, Sarkar D, et al. Genome-wide MyoD binding in skeletal muscle cells: a potential for broad cellular reprogramming. Dev Cell 2010;18:662-74. https://doi.org/10.1016/j.devcel.2010.02. 014   DOI
11 Soleimani VD, Yin H, Jahani-Asl A, et al. Snail regulates MyoD binding-site occupancy to direct enhancer switching and differentiation-specific transcription in myogenesis. Mol Cell 2012;47:457-68. https://doi.org/10.1016/j.molcel.2012.05.046   DOI
12 Yang J, Liu H, Wang K, et al. Isolation, culture and biological characteristics of multipotent porcine skeletal muscle satellite cells. Cell Tissue Bank 2017;18:513-25. https://doi.org/10.1007/s10561-017-9614-9   DOI
13 Trippel SB, Coutts RD, Einhorn TA, Mundy GR, Rosenfeld RG. Growth factors as therapeutic agents. Instr Course Lect 1997;46:473-6. https://doi.org/10.1.1.818.6360   DOI
14 Grounds M. Towards understanding skeletal muscle regeneration. Pathol Res Pract 1991;187:1-22. https://doi.org/10.1016/S0344-0338(11)81039-3   DOI
15 Berkes CA, Tapscott SJ. MyoD and the transcriptional control of myogenesis. Semin Cell Dev Biol 2005;16:585-95. https://doi.org/10.1016/j.semcdb.2005.07.006   DOI
16 Menetrey J, Kasemkijwattana C, Day C, et al. Growth factors improve muscle healing in vivo. J Bone Joint Surg Br 2000;82: 131-7. https://doi.org/10.1302/0301-620X.82B1.0820131   DOI
17 Xu C, Tabebordbar M, Iovino S, et al. A zebrafish embryo culture system defines factors that promote vertebrate myogenesis across species. Cell 2013;155:909-21. https://doi.org/10.1016/j.cell.2013.10.023   DOI
18 Aghila Rani KG, Kartha CC. Effects of epidermal growth factor on proliferation and migration of cardiosphere-derived cells expanded from adult human heart. Growth Factors 2010; 28:157-65. https://doi.org/10.3109/08977190903512628   DOI
19 Braun T, Gautel M. Transcriptional mechanisms regulating skeletal muscle differentiation, growth and homeostasis. Nat Rev Mol Cell Biol 2011;12:349-61. https://doi.org/10.1038/nrm3118   DOI
20 Pirskanen A, Kiefer JC, Hauschka SD. IGFs, insulin, Shh, bFGF, and TGF-β1 interact synergistically to promote somite myogenesis in vitro. Dev Biol 2000;224:189-203. https://doi.org/10.1006/dbio.2000.9784   DOI
21 Shi H, Xie H, Zhao Y, et al. Myoprotective effects of bFGF on skeletal muscle injury in pressure-related deep tissue injury in rats. Burns Trauma 2016;4:s41038-016-0051-y. https://doi.org/10.1186/s41038-016-0051-y   DOI
22 Liu X, Zhu Y, Cao D. Proliferative effect of basic fibroblast growth factor and epidermal growth factor on muscle derived stem cells. Chinese J Repar Reconstr Surg 2006;20:936-9.
23 Palade J, Djordjevic D, Hutchins ED, et al. Identification of satellite cells from anole lizard skeletal muscle and demonstration of expanded musculoskeletal potential. Dev Biol 2018;433:344-56. https://doi.org/10.1016/j.ydbio.2017.08.037   DOI
24 Sato C, Hane M, Kitajima K. Relationship between ST8SIA2, polysialic acid and its binding molecules, and psychiatric disorders. Biochim Biophys Acta Gen Subj 2016;1860:1739-52. https://doi.org/10.1016/j.bbagen.2016.04.015   DOI
25 Liang P, Xu Y, Zhang X, et al. CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes. Protein Cell 2015;6: 363-72. https://doi.org/10.1007/s13238-015-0153-5   DOI
26 Sun N, Zhao H. Seamless correction of the sickle cell disease mutation of the HBB gene in human induced pluripotent stem cells using TALENs. Biotechnol Bioeng 2014;111:1048-53. https://doi.org/10.1002/bit.25018|   DOI
27 Maman S, Sagi-Assif O, Yuan W, et al. The beta subunit of hemoglobin (HBB2/HBB) suppresses neuroblastoma growth and metastasis. Cancer Res 2017;77:14-26. https://doi.org/10.1158/0008-5472.CAN-15-2929   DOI
28 Shaw AD, Tiwari Y, Kaplan W, et al. Characterisation of genetic variation in ST8SIA2 and its interaction region in NCAM1 in patients with bipolar disorder. PLoS one 2014;9:e92556. https://doi.org/10.1371/journal.pone.0092556   DOI
29 Goetsch SC, Hawke TJ, Gallardo TD, Richardson JA, Garry DJ. Transcriptional profiling and regulation of the extracellular matrix during muscle regeneration. Physiol Genomics 2003; 14:261-71. https://doi.org/10.1152/physiolgenomics.00056.2003   DOI
30 Hane M, Kitajima K, Sato C. Effects of intronic single nucleotide polymorphisms (iSNPs) of a polysialyltransferase, ST8SIA2 gene found in psychiatric disorders on its gene products. Biochem Biophys Res Commun 2016;478:1123-9. https://doi.org/10.1016/j.bbrc.2016.08.079   DOI
31 Fitzgerald J, Bateman JF. A new FACIT of the collagen family: COL21A1. FEBS Lett 2001;505:275-80. https://doi.org/10.1016/S0014-5793(01)02754-5   DOI
32 Chou M-Y, Li H-C. Genomic organization and characterization of the human type XXI collagen (COL21A1) gene. Genomics 2002;79:395-401. https://doi.org/10.1006/geno.2002.6712   DOI
33 Sodhi SS, Song KD, Ghosh M, et al. Comparative transcriptomic analysis by RNA-seq to discern differential expression of genes in liver and muscle tissues of adult Berkshire and Jeju Native Pig. Gene 2014;546:233-42. https://doi.org/10.1016/j.gene.2014.06.005   DOI
34 Ghosh M, Sharma N, Gera M, et al. The first comprehensive description of the expression profile of genes involved in differential body growth and the immune system of the Jeju Native Pig and miniature pig. Amino Acids 2019;51:495-511. https://doi.org/10.1007/s00726-018-2685-5   DOI
35 Lunney JK. Advances in swine biomedical model genomics. Int J Biol Sci 2007;3:179-84. https://doi.org/10.7150/ijbs.3.179   DOI
36 Cho IC, Han SH, Fang M, et al. The robust phylogeny of Korean wild boar (Sus scrofa coreanus) using partial D-loop sequence of mtDNA. Mol Cells 2009;28:423. https://doi.org/10.1007/s10059-009-0139-3   DOI
37 Nain Z, Islam MA, Chowdhury SH, Afroza S, Hussain I. Current understanding on tail regeneration in green anoles (Anolis carolinensis). Cell Biol 2016;4:9-17. https://doi.org/10.11648/j.cb.2016040 2.11   DOI
38 Bonnet N, Garnero P, Ferrari S. Periostin action in bone. Mol Cell Endocrinol 2016;432:75-82. https://doi.org/10.1016/j.mce.2015.12.014   DOI
39 Kim GW, Yoo JY, Kim KJ, et al. Analysis of carcass characteristics by gender and carcass grades of Jeju native pigs. J Anim Sci 2010;52:313-8. https://doi.org/10.5187/JAST.2010.52.4.313   DOI
40 Post MJ. Cultured meat from stem cells: challenges and prospects. Meat Sci 2012;92:297-301. https://doi.org/10.1016/j.meatsci.2012.04.008   DOI
41 Musaro A, Barberi L. Isolation and culture of mouse satellite cells. In: Ward A, Tosh D, editors. Mouse cell culture. Methods in Molecular Biology (Methods and Protocols), vol 633. Clifton, NJ, USA: Humana Press; 2010. pp. 101-11. https://doi.org/10.1007/978-1-59745-019-5_8
42 Nihashi Y, Umezawa K, Shinji S, et al. Distinct cell proliferation, myogenic differentiation, and gene expression in skeletal muscle myoblasts of layer and broiler chickens. Sci Rep 2019; 9:16527. https://doi.org/10.1038/s41598-019-52946-4   DOI