Browse > Article
http://dx.doi.org/10.5851/kosfa.2020.e39

Optimization of Culture Conditions for Maintaining Pig Muscle Stem Cells In Vitro  

Choi, Kwang-Hwan (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Yoon, Ji Won (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Kim, Minsu (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Jeong, Jinsol (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Ryu, Minkyung (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Park, Sungkwon (Department of Food Science and Biotechnology, Sejong University)
Jo, Cheorun (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Lee, Chang-Kyu (Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University)
Publication Information
Food Science of Animal Resources / v.40, no.4, 2020 , pp. 659-667 More about this Journal
Abstract
Muscle stem cells isolated from domestic animals, including cows and pigs, were recently spotlighted as candidates for the production of alternative protein resources, so-called cultured meat or lab-grown meat. In the present study, we aimed to optimize the in vitro culture conditions for the long-term expansion of pig muscle stem cells via the screening of various signaling molecules. Pig muscle stem cells were collected from the biceps femoris muscles of 3-d-old crossbred pigs (Landrace×Yorkshire×Duroc, LYD) and cultured in minimum essential medium-based growth media. However, the pig muscle stem cells gradually lost their proliferation ability and featured morphologies during the long-term culture over two weeks. To find suitable in vitro culture conditions for an extended period, skeletal muscle growth medium-2, including epidermal growth factor (EGF), dexamethasone, and a p38 inhibitor (SB203580), was used to support the stemness of the pig muscle stem cells. Interestingly, pig muscle stem cells were stably maintained in a long-term culture without loss of the expression of myogenic marker genes as determined by PCR analysis. Immunostaining analysis showed that the stem cells were capable of myogenic differentiation after multiple passaging. Therefore, we found that basal culture conditions containing EGF, dexamethasone, and a p38 inhibitor were suitable for maintaining pig muscle stem cells during expanded culture in vitro. This culture method may be applied for the production of cultured meat and further basic research on muscle development in the pig.
Keywords
pig; muscle stem cells; EGF; dexamethasone; p38 inhibitor;
Citations & Related Records
Times Cited By KSCI : 2  (Citation Analysis)
연도 인용수 순위
1 Rando TA, Blau HM. 1994. Primary mouse myoblast purification, characterization, and transplantation for cell-mediated gene therapy. J Cell Biol 125:1275-1287.   DOI
2 Roe JA, Haji Baba AS, Harper JMM, Buttery PJ. 1995. Effects of growth factors and gut regulatory peptides on nutrient uptake in ovine muscle cell cultures. Comp Biochem Physiol A Physiol 110:107-114.   DOI
3 Syverud BC, Vandusen KW, Larkin LM. 2016. Effects of dexamethasone on satellite cells and tissue engineered skeletal muscle units. Tissue Eng Part A 22:480-489.   DOI
4 Troy A, Cadwallader AB, Fedorov Y, Tyner K, Tanaka KK, Olwin BB. 2012. Coordination of satellite cell activation and selfrenewal by par-complex-dependent asymmetric activation of p38${\alpha}$/${\beta}$ mapk. Cell Stem Cell 11:541-553.   DOI
5 Yin H, Price F, Rudnicki MA. 2013. Satellite cells and the muscle stem cell niche. Physiol Rev 93:23-67.   DOI
6 Zammit PS, Golding JP, Nagata Y, Hudon V, Partridge TA, Beauchamp JR. 2004. Muscle satellite cells adopt divergent fates:A mechanism for self-renewal? J Cell Biol 166:347-357.   DOI
7 Bentzinger CF, Wang YX, Von Maltzahn J, Soleimani VD, Yin H, Rudnicki MA. 2013. Fibronectin regulates wnt7a signaling and satellite cell expansion. Cell Stem Cell 12:75-87.   DOI
8 Bernet JD, Doles JD, Hall JK, Kelly Tanaka K, Carter TA, Olwin BB. 2014. p38 MAPK signaling underlies a cellautonomous loss of stem cell self-renewal in skeletal muscle of aged mice. Nat Med 20:265-271.   DOI
9 Cao Y, Kumar RM, Penn BH, Berkes CA, Kooperberg C, Boyer LA, Young RA, Tapscott SJ. 2006. Global and gene-specific analyses show distinct roles for myod and myog at a common set of promoters. Embo J 25:502-511.   DOI
10 Campion DR, Richardson RL, Reagan JO, Kraeling RR. 1981. Changes in the satellite cell population during postnatal growth of pig skeletal muscle. J Anim Sci 52:1014-1018.   DOI
11 Chakravarthy MV, Davis BS, Booth FW. 2000. IGF-I restores satellite cell proliferative potential in immobilized old skeletal muscle. J Appl Physiol 89:1365-1379.   DOI
12 Choi KH, Lee CK. 2019. Pig pluripotent stem cells as a candidate for biomedical application. J Anim Reprod Biotechnol 34:139-147.   DOI
13 Ding S, Swennen GNM, Messmer T, Gagliardi M, Molin DGM, Li CB, Zhou GH, Post MJ. 2018. Maintaining bovine satellite cells stemness through p38 pathway. Sci Rep 8:10808.   DOI
14 Dodson MV, Allen RE, Hossner KL. 1985. Ovine somatomedin, multiplication-stimulating activity, and insulin promote skeletal muscle satellite cell proliferation in vitro. Endocrinology 117:2357-2363.   DOI
15 Fu X, Xiao J, Wei Y, Li S, Liu Y, Yin J, Sun K, Sun H, Wang H, Zhang Z, Zhang BT, Sheng C, Wang H, Hu P. 2015. Combination of inflammation-related cytokines promotes long-term muscle stem cell expansion. Cell Res 25:655-673.   DOI
16 Doumit ME, Cook DR, Merkel RA. 1993. Fibroblast growth factor, epidermal growth factor, insulin-like growth factors, and platelet-derived growth factor-BB stimulate proliferation of clonally derived porcine myogenic satellite cells. J Cell Physiol 157:326-332.   DOI
17 Doumit ME, Merkel RA. 1992. Conditions for isolation and culture of porcine myogenic satellite cells. Tissue Cell 24:253-262.   DOI
18 Florini JR, Ewton DZ, Evinger-Hodges MJ, Falen SL, Lau RL, Regan JF, Vertel BM. 1984. Stimulation and inhibition of myoblast differentiation by hormones. In Vitro 20:942-958.   DOI
19 Gospodarowicz D, Weseman J, Moran JS, Lindstrom J. 1976. Effect of fibroblast growth factor on the division and fusion of bovine myoblasts. J Cell Biol 70:395-405.   DOI
20 Gilbert PM, Havenstrite KL, Magnusson KEG, Sacco A, Leonardi NA, Kraft P, Nguyen NK, Thrun S, Lutolf MP, Blau HM. 2010. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 329:1078-1081.   DOI
21 Groux-Muscatelli B, Bassaglia Y, Barritault D, Caruelle JP, Gautron J. 1990. Proliferating satellite cells express acidic fibroblast growth factor during in vitro myogenesis. Dev Biol 142:380-385.   DOI
22 Guerriero V Jr, Florini JR. 1980. Dexamethasone effects on myoblast proliferation and differentiation. Endocrinology 106:1198-1202.   DOI
23 Kuang S, Kuroda K, Le Grand F, Rudnicki MA. 2007. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell 129:999-1010.   DOI
24 Jennische E, Skottner A, Hansson HA. 1987. Satellite cells express the trophic factor IGF-I in regenerating skeletal muscle. Acta Physiol Scand 129:9-15.   DOI
25 Jeong JY, Kim JM, Rajesh RV, Suresh S, Jang GW, Lee KT, Kim TH, Park M, Jeong HJ, Kim KW, Cho YM, Lee HJ. 2013. Comparison of gene expression levels of porcine satellite cells from postnatal muscle tissue during differentiation. Reprod Dev Biol 37:219-224.   DOI
26 Jin W, Peng J, Jiang S. 2016. The epigenetic regulation of embryonic myogenesis and adult muscle regeneration by histone methylation modification. Biochem Biophys Rep 6:209-219.
27 Miersch C, Stange K, Rontgen M. 2018. Separation of functionally divergent muscle precursor cell populations from porcine juvenile muscles by discontinuous percoll density gradient centrifugation. BMC Cell Biol 19:2.   DOI
28 Post MJ. 2012. Cultured meat from stem cells: Challenges and prospects. Meat Sci 92:297-301.   DOI
29 Montarras D, Morgan J, Collins C, Relaix F, Zaffran S, Cumano A, Partridge T, Buckingham M. 2005. Direct isolation of satellite cells for skeletal muscle regeneration. Science 309:2064-2067.   DOI
30 Motohashi N, Alexander MS, Kunkel LM. 2012. Skeletal muscle regeneration and muscle progenitor cells. J Phys Fit Sports Med 1:151-154.   DOI
31 Quinn LS, Ong LD, Roeder RA. 1990. Paracrine control of myoblast proliferation and differentiation by fibroblasts. Dev Biol 140:8-19.   DOI