期刊论文详细信息
BMC Genomics
Characterization of a novel chicken muscle disorder through differential gene expression and pathway analysis using RNA-sequencing
Behnam Abasht2  William R Lee1  Weixuan Fu2  Erin M Brannick2  Marie F Mutryn2 
[1]Maple Leaf Farms, Leesburg, IN, USA
[2]Department of Animal and Food Sciences, University of Delaware, 531 South College Ave, Newark 19716, DE, USA
关键词: Skeletal muscle;    RNA-sequencing;    Pectoralis major;    Myopathy;    Broiler;    Wooden Breast;    Myodegeneration;    Chicken;   
Others  :  1203908
DOI  :  10.1186/s12864-015-1623-0
 received in 2014-08-04, accepted in 2015-05-06,  发布年份 2015
PDF
【 摘 要 】

Background

Improvements in poultry production within the past 50 years have led to increased muscle yield and growth rate, which may be contributing to an increased rate and development of new muscle disorders in chickens. Previously reported muscle disorders and conditions are generally associated with poor meat quality traits and have a significant negative economic impact on the poultry industry. Recently, a novel myopathy phenotype has emerged which is characterized by palpably “hard” or tough breast muscle. The objective of this study is to identify the underlying biological mechanisms that contribute to this emerging muscle disorder colloquially referred to as “Wooden Breast”, through the use of RNA-sequencing technology.

Methods

We constructed cDNA libraries from five affected and six unaffected breast muscle samples from a line of commercial broiler chickens. After paired-end sequencing of samples using the Illumina Hiseq platform, we used Tophat to align the resulting sequence reads to the chicken reference genome and then used Cufflinks to find significant changes in gene transcript expression between each group. By comparing our gene list to previously published histology findings on this disorder and using Ingenuity Pathways Analysis (IPA®), we aim to develop a characteristic gene expression profile for this novel disorder through analyzing genes, gene families, and predicted biological pathways.

Results

Over 1500 genes were differentially expressed between affected and unaffected birds. There was an average of approximately 98 million reads per sample, across all samples. Results from the IPA analysis suggested “Diseases and Disorders” such as connective tissue disorders, “Molecular and Cellular Functions” such as cellular assembly and organization, cellular function and maintenance, and cellular movement, “Physiological System Development and Function” such as tissue development, and embryonic development, and “Top Canonical Pathways” such as, coagulation system, axonal guidance signaling, and acute phase response signaling, are associated with the Wooden Breast disease.

Conclusions

There is convincing evidence by RNA-seq analysis to support localized hypoxia, oxidative stress, increased intracellular calcium, as well as the possible presence of muscle fiber-type switching, as key features of Wooden Breast Disease, which are supported by reported microscopic lesions of the disease.

【 授权许可】

   
2015 Mutryn et al.; licensee BioMed Central.

【 预 览 】
附件列表
Files Size Format View
20150523023706446.pdf 1801KB PDF download
Fig. 3. 55KB Image download
Fig. 2. 64KB Image download
Fig. 1. 82KB Image download
【 图 表 】

Fig. 1.

Fig. 2.

Fig. 3.

【 参考文献 】
  • [1]Bianchi M, Petracci M, Franchini A, Cavani C. The occurrence of deep pectoral myopathy in roaster chickens. Poult Sci. 2006; 85:1843-6.
  • [2]Bilgili SF, Hess JB. Green Muscle Disease in broilers increasing. World Poult. 2002; 18:42-3.
  • [3]Bilgili SF, Hess JB. Green Muscle Disease: Reducing the Incidence in Broiler Flocks. Ross Tech. 2008; 8:48.
  • [4]Kuttappan VA, Lee YS, Erf GF, Meullenet J-FC, McKee SR, Owens CM. Consumer acceptance of visual appearance of broiler breast meat with varying degrees of white striping. Poult Sci. 2012;1240–1247.
  • [5]Lesiów T, Kijowski J. Impact of PSE and DFD Meat On Poultry Processing – A Review. Polish J Food Nutr Sci. 2003; 12/53:3-8.
  • [6]Sihvo H-K, Immonen K, Puolanne E. Myodegeneration with fibrosis and regeneration in the pectoralis major muscle of broilers. Vet Pathol. 2014; 51:619-23.
  • [7]Owens CM. Identifying quality defects in poultry processing. Watt Poult USA. 2014.42-50. December
  • [8]Bilgili SF. Broiler Chicken Myopathies: II. Woody Breast? Worthw Oper Guidel Suggest. 2013.1. April
  • [9]Kuttappan VA, Shivaprasad H, Shaw DP, Valentine BA, Hargis BM, Clark FD et al.. IMMUNOLOGY, HEALTH, AND DISEASE Pathological changes associated with white striping in broiler breast muscles. Poult Sci. 2013; 92:331-8.
  • [10]Guetchom B, Venne D, Chenier S, Chorfi Y: Effect of extra dietary vitamin E on preventing nutritional myopathy in broiler chickens. J Appl Poult Res. 2012;21(3):548–55.
  • [11]Bains BS, Watson ARA. Nutritional Myopathy- A cause of ataxia in broiler chickens. N Z Vet J. 1978; 26:31-2.
  • [12]Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009; 10:57-63.
  • [13]Fast QC [http://www.bioinformatics.babraham.ac.uk/projects/fastqc/]
  • [14]Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR et al.. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012; 7:562-78.
  • [15]Cufflinks [http://cole-trapnell-lab.github.io/cufflinks/]
  • [16]Ingenuity Systems [http:/www.ingenuity.com]
  • [17]Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL et al.. Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol. 2008; 26:317-25.
  • [18]Periasamy M, Reed TD, Liu LH, Ji Y, Loukianov E, Paul RJ et al.. Impaired cardiac performance in heterozygous mice with a null mutation in the sarco(endo)plasmic reticulum Ca2 + −ATPase isoform 2 (SERCA2) gene. J Biol Chem. 1999; 274:2556-62.
  • [19]Prasad V, Boivin GP, Miller ML, Liu LH, Erwin CR, Warner BW et al.. Haploinsufficiency of Atp2a2, encoding the sarco(endo)plasmic reticulum Ca2 + −ATPase isoform 2 Ca2+ pump, predisposes mice to squamous cell tumors via a novel mode of cancer susceptibility. Cancer Res. 2005; 65:8655-61.
  • [20]Mitchell MA. Muscle Abnormalities- Pathophysiological mechanisms. In: Poult Meat Sci. 5th ed. Richardson RI, Mead GC, editors. CAB International, Oxon, UK; 1999: p.65-98.
  • [21]Kuo TH, Kim HR, Zhu L, Yu Y, Lin HM, Tsang W. Modulation of endoplasmic reticulum calcium pump by Bcl-2. Oncogene. 1998; 17:1903-10.
  • [22]Kaprielian Z, Fambrough DM. Expression of fast and slow isoforms of the Ca2 + −ATPase in developing chick skeletal muscle. Dev Biol. 1987; 124:490-503.
  • [23]Gailly P, Boland B, Himpens B, Casteels R, Gillis JM. Critical evaluation of cytosolic calcium determination in resting muscle fibres from normal and dystrophic (mdx) mice. Cell Calcium. 1993; 14:473-83.
  • [24]Mahon M. Muscle Abnormalities- Morphological Aspects. In: Poult Meat Sci. 5th ed. Richardson RI, Mead GC, editors. CAB International, Oxon, UK; 1999: p.19-64.
  • [25]Gissel H: The role of Ca2+ in muscle cell damage. Ann N Y Acad Sci. 2005;1066:166–80.
  • [26]Jackson MJ, Jones DA, Edwards RH. Experimental skeletal muscle damage: the nature of the calcium-activated degenerative processes. Eur J Clin Invest. 1984; 14:369-74.
  • [27]Sandercock DA, Mitchell MA. Myopathy in broiler chickens: a role for Ca(2+)-activated phospholipase A2? Poult Sci. 2003; 82:1307-12.
  • [28]Shaw K. Environmental Cues Like Hypoxia Can Trigger Gene Expression and Cancer Development. Nat Educ. 2008; 1:198.
  • [29]Semenza GL. HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol. 2000; 88:1474-80.
  • [30]Brahimi-Horn MC, Bellot G, Pouysségur J. Hypoxia and energetic tumour metabolism. Curr Opin Genet Dev. 2011; 21:67-72.
  • [31]Urbani L, Piccoli M, Franzin C, Pozzobon M, de Coppi P. Hypoxia Increases Mouse Satellite Cell Clone Proliferation Maintaining both In Vitro and In Vivo Heterogeneity and Myogenic Potential. PLoS One. 2012;7.
  • [32]Van der Slot AJ, Zuurmond A-M, Bardoel AFJ, Wijmenga C, Pruijs HEH, Sillence DO et al.. Identification of PLOD2 as telopeptide lysyl hydroxylase, an important enzyme in fibrosis. J Biol Chem. 2003; 278:40967-72.
  • [33]Gilkes DM, Bajpai S, Chaturvedi P, Wirtz D, Semenza GL. Hypoxia-inducible Factor 1 (HIF-1) Promotes Extracellular Matrix Remodeling under Hypoxic Conditions by Inducing P4HA1, P4HA2, and PLOD2 Expression in Fibroblasts. J Biol Chem. 2013; 288:10819-29.
  • [34]Macpherson LJ, Dubin AE, Evans MJ, Marr F, Schultz PG, Cravatt BF et al.. Noxious compounds activate TRPA1 ion channels through covalent modification of cysteines. Nature. 2007; 445:541-5.
  • [35]Bautista DM, Jordt SE, Nikai T, Tsuruda PR, Read AJ, Poblete J et al.. TRPA1 Mediates the Inflammatory Actions of Environmental Irritants and Proalgesic Agents. Cell. 2006; 124:1269-82.
  • [36]Hatano N, Itoh Y, Suzuki H, Muraki Y, Hayashi H, Onozaki K, Wood IC, Beech DJ, Muraki K: Hypoxia-inducible Factor-1 (HIF1) Switches on Transient Receptor Potential Ankyrin Repeat 1 (TRPA1) Gene Expression via a Hypoxia Response Element-like Motif to Modulate Cytokine Release. J Biol Chem. 2012;287(38):31962–72.
  • [37]Ciraci C, Tuggle CK, Wannemuehler MJ, Nettleton D, Lamont SJ. Unique genome-wide transcriptome profiles of chicken macrophages exposed to Salmonella-derived endotoxin. BMC Genomics. 2010; 11:545.
  • [38]Tubak V, Határvölgyi E, Krenács L, Korpos E, Kúsz E, Duda E et al.. Expression of immunoregulatory tumor necrosis factor-like molecule TL1A in chicken chondrocyte differentiation. Can J Vet Res. 2009; 73:34-8.
  • [39]Sato H, Kida Y, Mai M, Endo Y, Sasaki T, Tanaka J et al.. Expression of genes encoding type IV collagen-degrading metalloproteinases and tissue inhibitors of metalloproteinases in various human tumor cells. Oncogene. 1992; 7:77-83.
  • [40]Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P. MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: Differential regulation by inflammatory cytokines. Blood. 2007; 109:4055-63.
  • [41]Milkiewicz M, Haas TL. Effect of mechanical stretch on HIF-1{alpha} and MMP-2 expression in capillaries isolated from overloaded skeletal muscles: laser capture microdissection study. Am J Physiol Heart Circ Physiol. 2005; 289:H1315-20.
  • [42]Jackson WM, Aragon AB, Onodera J, Koehler SM, Ji Y, Bulken-Hoover JD et al.. Cytokine expression in muscle following traumatic injury. J Orthop Res. 2011; 29:1613-20.
  • [43]Cui H, Darmanin S, Natsuisaka M, Kondo T, Asaka M, Shindoh M et al.. Enhanced expression of asparagine synthetase under glucose-deprived conditions protects pancreatic cancer cells from apoptosis induced by glucose deprivation and cisplatin. Cancer Res. 2007; 67:3345-55.
  • [44]Zoll J, Ponsot E, Dufour S, Doutreleau S, Ventura-Clapier R, Vogt M et al.. Exercise Training in Normobaric Hypoxia in Endurance Runners. III Muscular Adjustments of Selected Gene Transcripts Volume. 2006; 100:1258-66.
  • [45]Zimmerman U-J, Wang P, Zhang X, Bogdanovich S, Forster R. Anti-oxidative response of carbonic anhydrase III in skeletal muscle. IUBMB Life. 2004; 56:343-7.
  • [46]Minchenko A, Leshchinsky I, Opentanova I, Sang N, Srinivas V, Armstead V et al.. Hypoxia-inducible factor-1-mediated expression of the 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) gene. Its possible role in the Warburg effect. J Biol Chem. 2002; 277:6183-7.
  • [47]Kronqvist P, Kawaguchi N, Albrechtsen R, Xu X, Schrøder HD, Moghadaszadeh B et al.. ADAM12 alleviates the skeletal muscle pathology in mdx dystrophic mice. Am J Pathol. 2002; 161:1535-40.
  • [48]Lopes-Ferreira M, Núñez J, Rucavado A, Farsky SHP, Lomonte B, Angulo Y et al.. Skeletal muscle necrosis and regeneration after injection of Thalassophryne nattereri (niquim) fish venom in mice. Int J Exp Pathol. 2001; 82:55-64.
  • [49]Rodrigues CAV, Diogo MM, da Silva CL, Cabral JMS. Hypoxia enhances proliferation of mouse embryonic stem cell-derived neural stem cells. Biotechnol Bioeng. 2010; 106:260-70.
  • [50]Yin H, Zhang S, Gilbert ER, Siegel PB, Zhu Q, Wong E a. Expression profiles of muscle genes in postnatal skeletal muscle in lines of chickens divergently selected for high and low body weight. Poult Sci. 2014; 93:147-54.
  • [51]Li Y, Yang X, Ni Y, Decuypere E, Buyse J, Everaert N et al.. Early-age feed restriction affects viability and gene expression of satellite cells isolated from the gastrocnemius muscle of broiler chicks. J Anim Sci Biotechnol. 2012; 3:33.
  • [52]Halevy O, Geyra A, Barak M, Uni Z, Sklan D. Early posthatch starvation decreases satellite cell proliferation and skeletal muscle growth in chicks. J Nutr. 2000; 130:858-64.
  • [53]Kuang S, Chargé SB, Seale P, Huh M, Rudnicki MA. Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis. J Cell Biol. 2006; 172:103-13.
  • [54]Powers SK, Duarte J, Kavazis AN, Talbert EE. Reactive oxygen species are signalling molecules for skeletal muscle adaptation. Exp Physiol. 2010; 95:1-9.
  • [55]Balaban RS, Nemoto S, Finkel T: Mitochondria, oxidants, and aging. Cell. 2005;120(4):483–95.
  • [56]Allen DG, Lamb GD, Westerblad H. Impaired calcium release during fatigue. J Appl Physiol. 2008; 104:296-305.
  • [57]Stephanou A, Latchman DS: Transcriptional modulation of heat-shock protein gene expression. Biochem Res Int. 2011;2011.
  • [58]Kalmar B, Greensmith L: Induction of heat shock proteins for protection against oxidative stress. Adv Drug Deliv Rev. 2009;61(4):310–18.
  • [59]Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000; 408:239-247.
  • [60]Lezoualc’h F, Engert S, Berning B, Behl C. Corticotropin-releasing hormone-mediated neuroprotection against oxidative stress is associated with the increased release of non-amyloidogenic amyloid beta precursor protein and with the suppression of nuclear factor-kappaB. Mol Endocrinol. 2000; 14:147-159.
  • [61]Karalis K, Muglia LJ, Bae D, Hilderbrand H, Majzoub JA. CRH and the immune system. In J Neuroimmunol Volume. 1997; 72:131-136.
  • [62]Fischer J, Bosse A, Pallauf J. Effect of selenium deficiency on the antioxidative status and muscle damage in growing turkeys. Arch Anim Nutr. 2008; 62:485-497.
  • [63]Brown RG, Sweeny PR, Moran ET. Collagen levels in tissues from selenium deficient ducks. Comp Biochem Physiol A Comp Physiol. 1982; 72:383-389.
  • [64]Lehnert SA, Byrne KA, Reverter A, Nattrass GS, Greenwood PL, Wang YH et al.. Gene Expression Profiling of Bovine Skeletal Muscle in Response to and during Recovery from Chronic and Severe Undernutrition. 2006.
  • [65]Terrill JR, Radley-Crabb HG, Iwasaki T, Lemckert FA, Arthur PG, Grounds MD: Oxidative stress and pathology in muscular dystrophies: Focus on protein thiol oxidation and dysferlinopathies. FEBS J. 2013;280(17):4149–64.
  • [66]Dudkiewicz M, Szczepińska T, Grynberg M, Pawłowski K. A novel protein kinase-like domain in a selenoprotein, widespread in the tree of life. PLoS One. 2012; 7: Article ID e32138
  • [67]Huang J-Q, Li D-L, Zhao H, Sun L-H, Xia X-J, Wang K-N et al.. The selenium deficiency disease exudative diathesis in chicks is associated with downregulation of seven common selenoprotein genes in liver and muscle. J Nutr. 2011; 141:1605-10.
  • [68]Peng D, Belkhiri A, Hu T, Chaturvedi R, Asim M, Wilson KT, Zaika A, El-Rifai W: Glutathione peroxidase 7 protects against oxidative DNA damage in oesophageal cells. Gut. 2012;61(9):1250–60.
  • [69]Young B, Lowe JS, Stevens A, Heath JW. Wheater’s Functional Histology: A Text and Colour Atlas. Fifth. Elsevier, Philadelphia, PA; 2006.
  • [70]Kranen RW, van Kuppevelt TH, Goedhart HA, Veerkamp CH, Lambooy E, Veerkamp JH. Hemoglobin and myoglobin content in muscles of broiler chickens. Poult Sci. 1999; 78:467-476.
  • [71]Blessing MH, Müller G. Myoglobin concentration in the chicken, especially in the gizzard (a biochemical, light and electron microscopic study). Comp Biochem Physiol A Comp Physiol. 1974; 47:535-540.
  • [72]Fleming BK, Froning GW, Yang TS. Heme Pigment Levels in Chicken Broilers Chilled in Ice Slush and Air 1. Poult Sci. 1991; 70:2197-2200.
  • [73]Pette D, Staron RS. Transitions of muscle fiber phenotypic profiles. Histochem Cell Biol. 2001; 115:359-372.
  • [74]Malila Y, Tempelman RJ, Sporer KRB, Ernst CW, Velleman SG, Reed KM et al.. Differential gene expression between normal and pale, soft, and exudative turkey meat. Poult Sci. 2013; 92:1621-33.
  • [75]Mullen AJ, Barton PJR. Structural characterization of the human fast skeletal muscle troponin I gene (TNNI2). Gene. 2000; 242:313-320.
  • [76]Yang H, Xu ZY, Lei MG, Li FE, Deng CY, Xiong YZ et al.. Association of 3 polymorphisms in porcine troponin I genes (TNNI1 and TNNI2) with meat quality traits. J Appl Genet. 2010; 51:51-57.
  • [77]Frey N, Barrientos T, Shelton JM, Frank D, Rütten H, Gehring D et al.. Mice lacking calsarcin-1 are sensitized to calcineurin signaling and show accelerated cardiomyopathy in response to pathological biomechanical stress. Nat Med. 2004; 10:1336-1343.
  • [78]Braun T, Gautel M. Transcriptional mechanisms regulating skeletal muscle differentiation, growth and homeostasis. Nat Rev Mol Cell Biol. 2011; 12:349-361.
  • [79]Chen Z, Zhao T-J, Li J, Gao Y-S, Meng F-G, Yan Y-B et al.. Slow skeletal muscle myosin-binding protein-C (MyBPC1) mediates recruitment of muscle-type creatine kinase (CK) to myosin. Biochem J. 2011; 436:437-445.
  • [80]Jordan T, Jiang H, Li H, DiMario JX. Inhibition of ryanodine receptor 1 in fast skeletal muscle fibers induces a fast-to-slow muscle fiber type transition. J Cell Sci. 2004; 117:6175-6183.
  • [81]Fulle S, Protasi F, Di Tano G, Pietrangelo T, Beltramin A, Boncompagni S et al.. The contribution of reactive oxygen species to sarcopenia and muscle ageing. Exp Gerontol. 2004; 39:17-24.
  • [82]Fujii J, Otsu K, Zorzato F, de Leon S, Khanna V, Weiler J et al.. Identification of a mutation in porcine ryanodine receptor associated with malignant hyperthermia. 1991.
  • [83]Fiege M, Wappler F, Weisshorn R, Gerbershagen MU, Menge M, Schulte Am Esch J. Induction of malignant hyperthermia in susceptible swine by 3,4-methylenedioxymethamphetamine (“ecstasy”). Anesthesiology. 2003; 99:1132-1136.
  • [84]Jordan T, Jiang H, Li H, DiMario JX. Regulation of skeletal muscle fiber type and slow myosin heavy chain 2 gene expression by inositol trisphosphate receptor 1. J Cell Sci. 2005; 118:2295-2302.
  • [85]DiMario JX. Protein kinase C signaling controls skeletal muscle fiber types. Exp Cell Res. 2001; 263:23-32.
  • [86]Jordan T, Li J, Jiang H, DiMario JX. Repression of slow myosin heavy chain 2 gene expression in fast skeletal muscle fibers by muscarinic acetylcholine receptor and G(alpha)q signaling. J Cell Biol. 2003; 162:843-850.
  • [87]Jackson SP, Green RD, Miller MF. Phenotypic characterization of rambouillet sheep expressing the Callipyge gene.1. Inheritance of the condition and production characteristics. J Anim Sci. 1997; 75:14-18.
  • [88]Vuocolo T, Byrne K, White J, McWilliam S, Reverter A, Cockett NE et al.. Identification of a gene network contributing to hypertrophy in callipyge skeletal muscle. Physiol Genomics. 2007; 28:253-272.
  • [89]Hershberger RE, Parks SB, Kushner JD, Li D, Ludwigsen S, Jakobs P et al.. Coding sequence mutations identified in MYH7, TNNT2, SCN5A, CSRP3, LBD3, and TCAP from 313 patients with familial or idiopathic dilated cardiomyopathy. Clin Transl Sci. 2008; 1:21-26.
  • [90]Xu X, Qiu H, Du ZQ, Fan B, Rothschild MF, Yuan F et al.. Porcine CSRP3: Polymorphism and association analyses with meat quality traits and comparative analyses with CSRP1 and CSRP2. Mol Biol Rep. 2010; 37:451-459.
  • [91]Arber S, Halder G, Caroni P. Muscle LIM protein, a novel essential regulator of myogenesis, promotes myogenic differentiation. Cell. 1994; 79:221-231.
  • [92]Barash IA, Mathew L, Lahey M, Greaser ML, Lieber RL. Muscle LIM protein plays both structural and functional roles in skeletal muscle. Am J Physiol Cell Physiol. 2005; 289:C1312-C1320.
  • [93]Kostek MC, Chen Y-W, Cuthbertson DJ, Shi R, Fedele MJ, Esser KA et al.. Gene expression responses over 24 h to lengthening and shortening contractions in human muscle: major changes in CSRP3, MUSTN1, SIX1, and FBXO32. Physiol Genomics. 2007; 31:42-52.
  • [94]Schneider AG, Sultan KR, Pette D. Muscle LIM protein: expressed in slow muscle and induced in fast muscle by enhanced contractile activity. Am J Physiol. 1999; 276:C900-C906.
  • [95]Heath E, Tahri D, Andermarcher E, Schofield P, Fleming S, Boulter CA. Abnormal skeletal and cardiac development, cardiomyopathy, muscle atrophy and cataracts in mice with a targeted disruption of the Nov (Ccn3) gene. BMC Dev Biol. 2008; 8:18.
  • [96]Chen CC, Lau LF: Functions and mechanisms of action of CCN matricellular proteins. Int J Biochem Cell Biol. 2009;41(4):771–83.
  • [97]Sabourin LA, Rudnicki MA. The molecular regulation of myogenesis. Clin Genet. 2000; 57:16-25.
  • [98]Manara MC, Perbal B, Benini S, Strammiello R, Cerisano V, Perdichizzi S et al.. The expression of ccn3(nov) gene in musculoskeletal tumors. Am J Pathol. 2002; 160:849-859.
  • [99]Lin CG, Leu S-J, Chen N, Tebeau CM, Lin S-X, Yeung C-Y et al.. CCN3 (NOV) is a novel angiogenic regulator of the CCN protein family. J Biol Chem. 2003; 278:24200-24208.
  • [100]Liu C, Gersch RP, Hawke TJ, Hadjiargyrou M. Silencing of Mustn1 inhibits myogenic fusion and differentiation. Am J Physiol Cell Physiol. 2010; 298:C1100-C1108.
  • [101]Zheng Q, Zhang Y, Chen Y, Yang N, Wang X-J, Zhu D. Systematic identification of genes involved in divergent skeletal muscle growth rates of broiler and layer chickens. BMC Genomics. 2009; 10:87.
  • [102]Hall A. Rho GTPases and the Actin Cytoskeleton. 1998.
  • [103]Bishop AL, Hall A. Rho GTPases and their effector proteins. Biochem J. 2000; 348(Pt 2):241-55.
  • [104]Bassères DS, Tizzei EV, Duarte AAS, Costa FF, Saad STO. ARHGAP10, a novel human gene coding for a potentially cytoskeletal Rho-GTPase activating protein. Biochem Biophys Res Commun. 2002; 294:579-85.
  • [105]Huard J, Li Y, Fu FH. Muscle injuries and repair: current trends in research. J Bone Joint Surg Am. 2002; 84-A:822-832.
  • [106]Sheehan SM, Allen RE. Skeletal muscle satellite cell proliferation in response to members of the fibroblast growth factor family and hepatocyte growth factor. J Cell Physiol. 1999; 181:499-506.
  • [107]Mitchell P, Steenstrup T, Hannon K. Expression of fibroblast growth factor family during postnatal skeletal muscle hypertrophy. J Appl Physiol. 1999; 86:313-319.
  • [108]Smyth AB, O’ Neill E, Smith DM. Functional properties of muscle proteins in processes poultry products. In: Poult Meat Sci. 5th ed. Richardson RI, Mead GC, editors. CAB International, Oxon, UK; 1999: p.377-396.
  • [109]Rayment I, Holden HM, Whittaker M, Yohn CB, Lorenz M, Holmes KC et al.. Structure of the actin-myosin complex and its implications for muscle contraction. Science. 1993; 261:58-65.
  • [110]Do M-KQ, Sato Y, Shimizu N, Suzuki T, Shono J -i., Mizunoya W, Nakamura M, Ikeuchi Y, Anderson JE, Tatsumi R: Growth factor regulation of neural chemorepellent Sema3A expression in satellite cell cultures. AJP Cell Physiol. 2011;301(5)C1270–C1279.
  • [111]Levi M, Keller TT, Van Gorp E, Ten Cate H: Infection and inflammation and the coagulation system. Cardiovasc Res. 2003;60(1):26–39.
  • [112]Emery AE, Burt D. Intracellular calcium and pathogenesis and antenatal diagnosis of Duchenne muscular dystrophy. Br Med J. 1980; 280:355-357.
  • [113]Byrd SK. Alterations in the sarcoplasmic reticulum: a possible link to exercise-induced muscle damage. Med Sci Sport Exerc. 1992; 24:531-536.
  • [114]Tay JS, Lai PS, Low PS, Lee WL, Gan GC. Pathogenesis of Duchenne muscular dystrophy: the calcium hypothesis revisited. J Paediatr Child Health. 1992; 28:291-293.
  • [115]Oberc MA, Engel WK. Ultrastructural localization of calcium in normal and abnormal skeletal muscle. Lab Invest. 1977; 36:566-577.
  • [116]MacRae VE, Mahon M, Gilpin S, Sandercock DA, Hunter RR, Mitchell MA. A comparison of breast muscle characteristics in three broiler great-grandparent lines. Poult Sci. 2007; 86:382-385.
  • [117]VanVleet JF, Valentine BA. Muscles and Tendons. Jubb. 2007.
  • [118]Gordon GB, Barcza MA, Bush ME. Lipid Accumulation in Hypoxic Tissue Culture Cells. Am J Pathol. 1977; 88:663-678.
  • [119]Chabowski A, Górski J, Calles-Escandon J, Tandon NN, Bonen A. Hypoxia-induced fatty acid transporter translocation increases fatty acid transport and contributes to lipid accumulation in the heart. FEBS Lett. 2006; 580:3617-23.
  • [120]Boström P, Magnusson B, Svensson P-A, Wiklund O, Borén J, Carlsson LMS et al.. Hypoxia converts human macrophages into triglyceride-loaded foam cells. Arterioscler Thromb Vasc Biol. 2006; 26:1871-6.
  • [121]Lien RJ, Bilgili SF, Hess JB, Joiner KS. Induction of deep pectoral myopathy in broiler chickens via encouraged wing flapping. J Appl Poult Res. 2012; 21:556-562.
  • [122]Whitehead NP, Yeung EW, Allen DG. Muscle damage in mdx (dystrophic) mice: role of calcium and reactive oxygen species. Clin Exp Pharmacol Physiol. 2006; 33:657-662.
  • [123]Han R. Muscle membrane repair and inflammatory attack in dysferlinopathy. Skelet Muscle. 2011; 1:10.
  • [124]Tidball JG. Inflammatory processes in muscle injury and repair. Am J Physiol Regul Integr Comp Physiol. 2005; 288:R345-R353.
  • [125]Brown NS, Bicknell R. Hypoxia and oxidative stress in breast cancer. Oxidative stress: its effects on the growth, metastatic potential and response to therapy of breast cancer. Breast Cancer Res. 2001; 3:323-7.
  • [126]Li YP, Atkins CM, Sweatt JD, Reid MB. Mitochondria mediate tumor necrosis factor-alpha/NF-kappaB signaling in skeletal muscle myotubes. Antioxid Redox Signal. 1999; 1:97-104.
  • [127]Tidball JG, Rinaldi C: Immunological Response to Muscle Injury. In Muscle Fundam Biol Mech Dis Vol 2. Edited by Griendling KK, Kitsis RN, Stull JT. London: Academic Press; 2012:899–920.
  文献评价指标  
  下载次数:0次 浏览次数:13次