Scientia Agricultura Sinica ›› 2014, Vol. 47 ›› Issue (6): 1200-1207.doi: 10.3864/j.issn.0578-1752.2014.06.016

• ANIMAL SCIENCE·VETERINARY SCIENCERE·SOURCE INSECT • Previous Articles     Next Articles

Progresses in Research of the Mechanisms of Skeletal Muscle Fiber Formation

 LI  Bo-Jiang, LI  Ping-Hua, WU  Wang-Jun, LI  Qi-Fa, HUANG  Rui-Hua, LIU  Hong-Lin   

  1. College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095
  • Received:2013-09-18 Online:2014-03-15 Published:2013-12-20

Abstract: Skeletal muscle, the most important component of animal body, constitutes 40% of the animal body, while muscle fibers as the primary part of muscle, its differences is one of the critical factors affecting meat quality. Therefore, the growth, development of the skeletal muscles and their physiological and biochemical characteristics will directly affect the meat quantity and quality. Generally, the number of the muscle fibers is fixed during the embryonic development, while the increase of the muscle mass is due to the hypertrophy of skeletal muscle fibers after birth. Moreover, the skeletal muscle fibers types are not completely fixed, the change will be occurred according the metabolism and the functional demand during the growth and development of the skeletal muscle. The biological processes of the skeletal muscle growth and development, and the formation of muscle fibers are quite complex, involving in many genes and signal pathway. With the development of molecular biological technologies, many methods are applied to elucidate the regulatory mechanisms of the formation of many complex biological phenomena. To date, a great progress has been made in study of the molecular mechanisms related to the skeletal muscle development, and many key factors involved in the skeletal muscle growth and development have been identified. In the early study, the researches on the skeletal muscle fibers were focused on the identification of muscle fibers types and their physiological and biochemical characteristics, whereas the studies on the molecular mechanisms are relatively rare. Recently, a breakthrough has been made in the molecular regulatory mechanisms involved in the formation of skeletal muscle fibers. So, it is necessary to completely review the types and properties of muscle fiber, molecular regulatory mechanisms and the relationship between the muscle fibers types and meat quality. In this review, the types and properties of muscle fiber are summarized firstly. Then, the research progress in molecular regulatory mechanisms of the slow and fast muscle fibers formation was presented, respectively. Finally, the relationship of muscle fibers and meat quality was discussed. In general, this review will be helpful for further understanding the genetic mechanisms of the formation of skeletal muscle fiber types, and could be a reference for further studying on the formation of muscle fibers in the future. Meanwhile, the review is able to provide useful information for revealing the molecular mechanisms of meat quality, and provide molecular theoretical evidences for cultivating new breeds or lines using biotechnology in the future.

Key words: agricultural animal , muscle fiber , regulatory mechanism , meat quality

 [1]Bassel-Duby R, Olson E N. Signaling pathways in skeletal muscle remodeling. Annual Review of Biochemistry, 2006, 75: 19-37.

[2]Schiaffino S, Sandri M, Murgia M. Activity-dependent signaling pathways controlling muscle diversity and plasticity. Physiology, 2007, 22: 269-278.

[3]Chin E R, Olson E N, Richardson J A, Yang Q, Humphries C, Shelton J M, Wu H, Zhu W, Bassel-Duby R, Williams R S. A calcineurin-dependent transcriptional pathway controls skeletal muscle fiber type. Genes Development, 1998, 12(16): 2499-2509.

[4]Brooke M H, Kaiser K K. Muscle fiber types: how many and what kind. Archives of Neurology, 1970, 23(4): 369-379.

[5]Guth L, Samaha F J. Qualitative differences between actomyosin ATPase of slow and fast mammalian muscle. Experimental Neurology, 1969, 5: 138-152.

[6]Peter J B, Barnard R J, Edgerton V R. Metabolic profiles of three fiber types of skeletal muscle in guinea pigs and rabbits. Biochemistry, 1972, 11(14): 2627-2633.

[7]Ashmore C R, Doerr L. Comparative aspects of muscle fibre types in different species. Experimental Neurology, 1971, 31: 408-418.

[8]Schiaffino S, Gorza L, Sartore S, Saggin L, AusoniS V, Gundersen M K, Lomo T. Three myosin heavy chain isoforms in type 2 skeletal muscle fibres. Journal of Muscle Research and Cell Motility, 1989, 10: 197-205.

[9]LaFramboise W A, Daood M J, Guthrie R D, Moretti P, Schiaffino S, Ontell M. Electrophoretic separation and immunological identification of type 2X myosin heavy chain in rat skeletal muscle. Biochimica et Biophysica Acta, 1990, 1035: 109-112.

[10]Bassel-Duby R, Olson E N. Signaling pathways in skeletal muscle remodeling. Annual Review of Biochemistry, 2006, 75: 19-37.

[11]Ausoni S, Gorza L, Schiaffino S, Gundersen K, Lomo T. Expression of myosin heavy chain isoforms in stimulated fast and slow rat muscles. The Journal of Neuroscience, 1990, 10: 153-160.

[12]Wu H, Kanatous S B, Thurmond F A, Gallardo T, Isotani E, Bassel-Duby R, Williams R S. Regulation of mitochondrial biogenesis in skeletal muscle by CaMK. Science, 2002, 296(5566): 349-352.

[13]Chin E R. Role of Ca2+/calmodulin-dependent kinases in skeletal muscle plasticity. Journal of Applied Physiology, 2005, 99(2): 414-423.

[14]Martins K J, St-Louis M, Murdoch G. K, MacLean I M, McDonald P, Dixon W T, Putman C T, Michel R N. Nitric oxide synthase inhibition prevents activity-induced calcineurin-NFATc1 signalling and fast-to-slow skeletal muscle fibre type conversions. The Journal of Physiology, 2012, 590(6): 1427-1442.

[15]Potthoff M J, Wu H, Arnold M A, Shelton J M, Backs J, McAnally J, Richardson J A, Bassel-Duby R, Olson E N. Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers. Journal of Clinical Investigation, 2007, 117(9): 2459-2467.

[16]Lu J, McKinsey T A, Nicol R L, Olson E N. Signal-dependent activation of the MEF2 transcription factor by dissociation from histone deacetylases. Proceedings of National Academy of Sciences of the United States of America, 2000, 97(8): 4070-4075.

[17]McKinsey T A, Zhang C L, Lu J, Olson E N. Signal-dependent nuclear export of a histone deacetylase regulates muscle differentiation. Nature, 2000, 408(6808): 106-111.

[18]McKinsey T A, Zhang C L, Olson E N. Activation of the myocyte enhancer factor-2 transcription factor by calcium/calmodulin- dependent protein kinase-stimulated binding of 14-3-3 to histone deacetylase 5. Proceedings of National Academy of Sciences of the United States of America, 2000, 97(26): 14400-14405.

[19]Grozinger C M, Schreiber S L. Regulation of histone deacetylase 4 and 5 and transcriptional activity by 14-3-3-dependent cellular localization. Proceedings of National Academy of Sciences of the United States of America, 2000, 97(14): 7835-7840.

[20]Wang A H, Kruhlak M J, Wu J, Bertos N R, M Vezmar, Posner B I, Bazett-Jones D P, Yang X J. Regulation of histone deacetylase 4 by binding of 14-3-3 proteins. Molecular and Cellular Biology, 2000, 20(18): 6904-6912.

[21]Kim M S, Fielitz J, McAnally J, Shelton J M, Lemon D D, McKinsey T A, Richardson J A, Bassel-Duby R, Olson E N. Protein kinase D1 stimulates MEF2 activity in skeletal muscle and enhances muscle performance. Molecular and Cellular Biology, 2008, 28(11): 3600-3609.

[22]Murgia M, Serrano A L, Calabria E, Pallafacchina G, Lømo T, Schiaffino S. Ras is involved in nerve-activity-dependent regulation of muscle genes. Nature Cell Biology, 2000, 2: 142-147.

[23]Lin J, Wu H, Tarr P T, Zhang C Y, Wu Z D, Boss O, Michael L F, Puigserver P, Isotani E, Olson E N, Lowell B B, Bassel-Duby R, Spiegelman B M. Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature, 2002, 418(6899): 797-801.

[24]Schuler M, Ali F, Chambon C, Duteil D, Bornert J M, Tardivel A, Desvergne B, Wahli W, Chambon P, Metzger D. PGC1 alpha expression is controlled in skeletal muscles by PPAR beta, whose ablation results in fiber-type switching, obesity, and type 2 diabetes. Cell Metabolism, 2006, 4(5): 407-414.

[25]Wang Y X, Zhang C L, Yu R T, Cho H K, Nelson M C, Bayuga- Ocampo C R, Ham J, Kang H, Evans R M. Regulation of muscle fiber type and running endurance by PPAR delta. Plos Biology, 2004, 2(10): 1532-1539.

[26]Wright D C, Han D H, Garcia-Roves P M, Geiger P C, Jones T E, Holloszy J O. Exercise-induced mitochondrial biogenesis begins before the increase in muscle PGC-1alpha expression. Journal of Biological Chemistry, 2007, 282(1): 194-199.

[27]Akimoto T, Pohnert S C, Li P, Zhang M, Gumbs C, Rosenberg P B, Williams R S, Ya Z n. Exercise stimulates Pgc-1alpha transcription in skeletal muscle through activation of the p38 MAPK pathway. Journal of Biological Chemistry, 2005, 280(20): 19587-19593.

[28]Foster W H, Tidball J G, Wang Y. p38gamma activity is required for maintenance of slow skeletal muscle size. Muscle Nerve, 2012, 45(2): 266-273.

[29]Takemori H, Katoh Hashimoto Y, Nakae J, Olson E N, Okamoto M. Inactivation of HDAC5 by SIK1 in AICAR-treated C2C12 myoblasts. Endocrine Journal, 2009, 56(1): 121-130.

[30]Tsika R W, Schramm C, Simmer G, Fitzsimons D P, Moss R L, Ji J. Overexpression of TEAD-1 in transgenic mouse striated muscles produces a slower skeletal muscle contractile phenotype. Journal of Biological Chemistry, 2008, 283(52): 36154-36167.

[31]Liew H P, Choksi S P, Wong K N, Roy S. Specification of vertebrate slow-twitch muscle fiber fate by the transcriptional regulator Blimp1. Developmental Biology,  2008, 324(2): 226-235.

[32]Issa L L, Palmer S J, Guven K L, Santucci N, Hodgson V R M, Popovic K, Joya J E, Hardeman E C. MusTRD can regulate postnatal fiber-specific expression. Developmental Biology, 2006, 293(1): 104-115.

[33]von Hofsten, J., S. Elworthy, M.J. Gilchrist, J.C. Smith, F.C. Wardle, and P.W. Ingham. Prdm1- and Sox6-mediated transcriptional repression specifies muscle fibre type in the zebrafish embryo. Embo Reports, 2008, 9(7): 683-689.

[34]Kuroda K, Kuang S, Taketo M M, Rudnicki M A. Canonical Wnt signaling induces BMP-4 to specify slow myofibrogenesis of fetal myoblasts. Skeletal Muscle,  2013, 3(1): 5.

[35]Grifone R, Laclef C, Spitz F, Lopez S, Demignon J, Guidotti J E, Kawakami K, Xu P X, Kelly R, Petrof B J, Daegelen D, Concordet J P, Maire P. Six1 and Eya1 expression can reprogram adult muscle from the slow-twitch phenotype into the fast-twitch phenotype. Molecular and Cellular Biology, 2004, 24(14): 6253-6267.

[36]Hamade A, Deries M, Begemann G, Bally-Cuif L, Genet C, Sabatier F, Bonnieu A, Cousin X. Retinoic acid activates myogenesis in vivo through Fgf8 signalling. Developmental Biology, 2006, 289(1): 127-140.

[37]Groves J A, Hammond C L, Hughes S M. Fgf8 drives myogenic progression of a novel lateral fast muscle fibre population in zebrafish. Development, 2005, 132(19): 4211-4222.

[38]Maves L, A J Waskiewicz, B Paul, Y Cao, A Tyler, Moens C B, Tapscott S J. Pbx homeodomain proteins direct Myod activity to promote fast-muscle differentiation. Development, 2007, 134(18): 3371-3382.

[39]Arany Z, Lebrasseur N, Morris C, Smith E, Yang W L, Ma Y H, Chin S, Spiegelman B M. The transcriptional coactivator PGC-1 beta drives the formation of oxidative type IIX fibers in skeletal muscle. Cell Metabolism, 2007, 5(1): 35-46.

[40]Hagiwara N, Yeh M, Liu A. Sox6 is required for normal fiber type differentiation of fetal skeletal muscle in mice. Developmental Dynamics, 2007, 236(8): 2062-2076.

[41]Shi H, Scheffler J M, Pleitner J M, Zeng C, Park S, Hannon K M, Grant A L, Gerrard D E. Modulation of skeletal muscle fiber type by mitogenactivated protein kinase signaling. The FASEB Journal, 2008, 22(8): 2990-3000.

[42]Bessarab D A, Chong S W, Srinivas B P, Korzh V. Six1a is required for the onset of fast muscle differentiation in zebrafish. Developmental Biology, 2008, 323(2): 216-228.

[43]Niro C, Demignon J, Vincent S, Liu Y B, Giordani J, Sgarioto N, Favier M, Guillet-Deniau I, Blais A, Maire P. Six1 and Six4 gene expression is necessary to activate the fast-type muscle gene program in the mouse primary myotome. Developmental Biology, 2010, 338(2): 168-182.

[44]Richard A F, Demignon J, Sakakibara I, Pujol J, Favier M, Strochlic L, Le Grand F, Sgarioto N, Guernec A, Schmitt A, Cagnard N, Huang R, Legay C, Guillet-Deniau I, Maire P. Genesis of muscle fiber-type diversity during mouse embryogenesis relies on Six1 and Six4 gene expression. Developmental Biology, 2011, 359(2): 303-320.

[45]Wu W, Ren Z, Wang Y, Chao Z, Xu D, Xiong Y. Molecular characterization, expression patterns and polymorphism analysis of porcine Six1 gene. Molecular Biology Report, 2011, 38(4): 2619-2632.

[46]Anders H K, Klont R E, ernandez X F. Skeletal muscle fibres as factors for pork quality. Livestock Production Science, 1999, 60(23): 255 - 269.

[47]Picard B, Lefaucheur L, Berri C, Duclos M J. Muscle fibre ontogenesis in farm animal species. Reproduction Nutrition Development, 2002, 42(5): 415-431.

[48]Offer G. Modelling of the formation of pale, soft and exudative meat: Effects of chilling regime and rate and extent of glycolysis. Meat Science, 1991, 30(2): 157-184.

[49]Bowker B C, Grant A L, Forrest J C, Gerrard D E. Muscle metabolism and PSE pork. Journal of Animal Science, 2000, 79: 1-9.

[50]Immonen K, Puolanne E. Variation of residual glycogen-glucose concentration at ultimate pH values below 5.75. Meat Science, 2000, 55(3): 279-283.

[51]Serra X, Gil F, Perez-Enciso, Oliver M, Vazquez M, Gilpert M. A comparison of carsass,meat quality and histochemieal characteristies of Iberian and landrace Pigs. Livestock Production Science, 1998, 56: 215-223.

[52]Leseigneur-Meynier A, Gandemer G. Lipid composition of pork muscle in relation to the metabolic type of the fibres. Meat Science, 1991, 29(3): 229-241.

[53]Devoto S H, Melancon E, Eisen J S, Westerfield M. Identification of separate slow and fast muscle precursor cells in vivo, prior to somite formation. Development, 1996, 122(11): 3371-3380.

[54]Ingham P W, Kim H R. Hedgehog signalling and the specification of muscle cell identity in the zebrafish embryo. Experimental Cell Research, 2005, 306(2): 336-342.
[1] SONG ShuZhen, GAO LiangShuang, LI Hong, GONG XuYin, LIU LiShan, WEI YuBing. Effects of Feeding Levels on Muscle Tissue Structure and Muscle Fiber Composition Related Genes in Sheep [J]. Scientia Agricultura Sinica, 2022, 55(21): 4304-4314.
[2] LIU WangJing,TANG DeFu,AO ChangJin. Effect of Allium mongolicum Regel and Its Extracts on the Growth Performance, Carcass Characteristics, Meat Quality and Serum Biochemical Indices of Captive Small-Tailed Han Sheep [J]. Scientia Agricultura Sinica, 2022, 55(17): 3461-3472.
[3] ZHANG Lan,WANG LiangZhi,HUANG YanLing,LIAO XiuDong,ZHANG LiYang,LÜ Lin,LUO XuGang. Effects of Dietary Supplemental Pattern of Trace Eloments on the Growth Performance, Carcass Traits and Meat Quality of Broilers [J]. Scientia Agricultura Sinica, 2021, 54(22): 4906-4916.
[4] ZHANG MeiQi,LI Yan,LI ShuJing,GAO YanXia,LI JianGuo,CAO YuFeng,LI QiuFeng. Effects of Dietary Energy Levels on Production Performance, Blood Index, Slaughter Performance and Meat Quality of Holstein Steers [J]. Scientia Agricultura Sinica, 2021, 54(1): 203-212.
[5] ZHU XingHao,CHEN Qing,SHAO BingHao,GUO YuJun,ZHANG XiangLi,DU PengFei,ZHU Yao,HUANG YanQun,CHEN Wen. Effect of the Heterozygous Sex-Linked Dwarf Gene on Fat Deposition in Normal Type Chickens [J]. Scientia Agricultura Sinica, 2021, 54(1): 213-223.
[6] LI XueRu,SHI XiXiong,WANG JianZhong,ZHANG PanGao,TIAN Zhu,HAN Ling. Effect of Nitric Oxide Synthetase Inhibitor on Yak Meat Quality During Post-Mortem Aging [J]. Scientia Agricultura Sinica, 2020, 53(8): 1617-1626.
[7] CHEN LiJing,CHEN Zhuo,LI Na,SUN YaWei,LI HongBo,SONG WenWen,ZHANG Yang,YAO Gang. Comparison of the Carcass and Beef Quality Traits with the Expression of the Lipid Metabolism Related Genes Between Xinjiang Brown Cattle and Angus Beef Cattle [J]. Scientia Agricultura Sinica, 2020, 53(22): 4700-4709.
[8] ZHAO YangYang,LI Yan,HAN YongSheng,WANG XiaoLing,LI JianGuo,GAO YanXia,CAO YuFeng,LI QiuFeng. The Slaughter Characteristics and Meat Quality of Holstein Bulls Grain-Fed Under Different Dietary Crude Protein Levels [J]. Scientia Agricultura Sinica, 2020, 53(2): 431-439.
[9] SHI Lei, LI YunLei, SUN YanYan, CHEN JiLan. Research Progress on the Regulatory Mechanism of Lighting Schedule Affecting the Reproduction Performance of Chickens [J]. Scientia Agricultura Sinica, 2018, 51(16): 3191-3200.
[10] ZHU Hong-long, YANG Jie, XU Xiao-bo, PAN Xiao-qing,QIN Feng, LI Jian, XU Ye-fei, ZHOU Xiao-yun, GU Hong-ru. Effects of Rearing Environment on Physiological and Behavioural Responses of Pigs to Pre-Slaughter Handing, Carcass Straits, and Meat Quality [J]. Scientia Agricultura Sinica, 2016, 49(22): 4441-4450.
[11] ZHOU Qin-fei, HE Zhi-fei, LI Hong-jun. Effect of Cage Type on Slaughter Performance and Meat Quality Change During Postmortem Aging of Meat Rabbit [J]. Scientia Agricultura Sinica, 2016, 49(12): 2430-2436.
[12] XING Huan, LUAN Su-jun, SUN Yong-bo, SA Ren-na, ZHANG Hong-fu. Effect of Ammonia in Broiler Houses on the Antioxidant Activity and Meat Quality of Broiler [J]. Scientia Agricultura Sinica, 2015, 48(21): 4347-4357.
[13] HUANG Jin-yu, JIAO Jin-zhen, RAN Tao, TANG Shao-xun, KANG Jin-he, TAN Zhi-liang. Study on the Developmental Changes of Meat and Antioxidant Capacity of Grazing and House-Feeding Goats [J]. Scientia Agricultura Sinica, 2015, 48(14): 2827-2838.
[14] GUO Xin, HUANG Feng, ZHANG Chun-jiang, HU Hong-hai, CHEN Wen-bo, ZHANG Hong. Effects of Pressure Varied Static Brining on Pork Quality [J]. Scientia Agricultura Sinica, 2015, 48(11): 2229-2240.
[15] XUE Shan, HE Zhi-Fei, LI Hong-Jun. Variation of Muscle Glycogen of IRA Rabbit After Slaughter and Its Correlation with Changes of Rabbit Meat Quality [J]. Scientia Agricultura Sinica, 2014, 47(4): 814-822.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!