期刊论文详细信息
BMC Biotechnology
AAV2-mediated follistatin overexpression induces ovine primary myoblasts proliferation
Mahmood Nazari2  Fatemeh Salabi1  Li Zhang2  Fuping Zhao2  Caihong Wei2  Lixin Du2 
[1] Transgenic and Stem Cell Core, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, People’s Republic of China
[2] National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, People’s Republic of China
关键词: Proliferation;    Myoblasts;    Adeno-associated virus;    Over-expression;    Follistatin;   
Others  :  1084511
DOI  :  10.1186/s12896-014-0087-7
 received in 2014-03-10, accepted in 2014-10-06,  发布年份 2014
PDF
【 摘 要 】

Background

Follistatin (FST) has been shown to bind to some TGF-β family members and can function as a potent myostatin (MSTN) antagonist. Recent studies have revealed that over-expression of FST by adeno-associated viruses increases muscle growth in mice, humans and nonhuman primates. In the present study, to determine the effect of FST on ovine primary myoblast (OPM) proliferation, FST was over-expressed using an adeno-associated virus serotype 2 (AAV 2) vector.

Results

Western blot results showed that AAV induced the expression of FST protein in transduced OPM cells. Real-time quantitative PCR results indicated that over-expression of FST resulted in a dramatic increase in Akt I and CDK2 expression and a decrease in p21 expression. Moreover, cell cycle analysis confirmed that FST down-regulated p21, a CDK inhibitor, and increased the level of CDK2 expression in OPM cells. Hence, follistatin positively regulated the G1 to S progression. Our results showed that FST induced proliferation through a down-regulation of p21, as only the p21 expression level was down-regulated as a result of FST over-expression in myoblasts, whereas no change was observed in the level of p57 expression.

Conclusions

These results expanded our understanding of the regulation mechanism of FST in ovine primary myoblasts. Our results provide the first evidence that the AAV viral system can be used for gene transfer in ovine myoblast cells. Moreover, the results showed that an AAV vector can successfully induce the expression of FST in OPM cells in vitro. These findings demonstrated that FST over-expression induces proliferation through a down-regulation of the p21 gene under proliferating conditions.

【 授权许可】

   
2014 Nazari et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150113162201655.pdf 2165KB PDF download
Figure 6. 31KB Image download
Figure 5. 96KB Image download
20140822214619140.pdf 27KB PDF download
Figure 3. 61KB Image download
Figure 2. 166KB Image download
Figure 1. 67KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 5.

Figure 6.

【 参考文献 】
  • [1]Honaramooz A, Megee S, Zeng W, Destrempes M, Overton S: Adeno-associated virus (AAV)-mediated transduction of male germ line stem cells results in transgene transmission after germ cell transplantation. FASEB J 2008, 22:374-382. http://www.ncbi.nlm.nih.gov/pubmed/17873102
  • [2]Naldini L, Blomer P, Gallay D, Ory R, Mulligan FH, Gage I, Verma M, Trono D: In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 1996, 272:263-267. http://www.ncbi.nlm.nih.gov/pubmed/8602510
  • [3]Ragot T, Vincent N, Chafey P, Vigne E, Gilgenkrantz H, Couton D: Efficient adenovirus-mediated transfer of a human minidystrophin gene to skeletal muscle of mdx mice. Nature 1993, 361:647-650. http://www.nature.com/nature/journal/v361/n6413/abs/361647a0.html
  • [4]Russell DW, Kay MA: Adeno-associated virus vectors and hematology. Blood 1999, 94:864-874. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3739711/
  • [5]Srivastava A: Obstacles to human hematopoietic stem cell transduction by recombinant adeno-associated virus 2 vectors. J Cell Biochem 2002, 38(Suppl):39-45. http://www.ncbi.nlm.nih.gov/pubmed/12046848
  • [6]Flotte TR, Barraza X, Solow R, Afione SA, Carter BJ, Guggino WB: An improved system for packaging recombinant adenoassociated virus vectors capable of in vivo transduction. Gene Ther 1995, 2:29-37. http://www.ncbi.nlm.nih.gov/pubmed/7712331
  • [7]Flotte TR, Carter BJ: Adeno-associated virus vectors for gene therapy. Gene Ther 1995, 2:357-362. http://www.ncbi.nlm.nih.gov/pubmed/7584109
  • [8]Xiao X, Li J, Samulski RJ: Efficient long-term gene transfer into muscle tissue of immunocompetent mice by adeno-associated virus vector. J Virol 1996, 70:8098-8108. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC190884/pdf/708098.pdf
  • [9]Haidet AM, Rizo L, Handy C, Umapathi P, Eagle A, Shilling C: Long-term enhancement of skeletal muscle mass and strength by single gene administration of myostatin inhibitors. Proc Natl Acad Sci U S A 2008, 105:4318-4322. http://www.ncbi.nlm.nih.gov/pubmed/18334646
  • [10]Kota J, Handy CR, Haidet AM, Montgomery CL, Eagle A, Mendell JR, Kaspar BK: Follistatin gene delivery enhances muscle growth and strength in nonhuman primates. Sci Transl Med 2010, 1(Suppl 6):1-17. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2852878/
  • [11]Amthor H, Nicholas G, McKinnell I, Kemp CF, Sharma M, Kambadur R, Patel K: Follistatin complexes myostatin and antagonizes myostatin-mediated inhibition of myogenesis. Dev Biol 2004, 270:19-30. http://www.ncbi.nlm.nih.gov/pubmed/15136138
  • [12]Lee SJ: Sprinting without myostatin: a genetic determinant of athletic prowess. Trends Genet 2007, 23:475-477. http://www.sciencedirect.com/science/article/pii/S0168952507002697
  • [13]Han DS, Huang HP, Wang TG, Hung MY: Transcription activation of myostatin by trichostatin A in differentiated C2C12 myocytes via ASK1-MKK3/4/6-JNK and p38 mitogen-activated protein kinase pathways. J Cell Biochem 2010, 111:564-573. http://onlinelibrary.wiley.com/doi/10.1002/jcb.22740/pdf
  • [14]Philip B, Lu Z, Gao Y: Regulation of GDF-8 signaling by the p38 MAPK. Cell Signal 2005, 17:365-375. http://www.sciencedirect.com/science/article/pii/S0898656804001603
  • [15]Trendelenburg AU, Meyer A, Rohner D, Boyle J, Hatakeyama S, Glass DJ: Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Cell Physiol 2009, 296:C1258-C1270. http://ajpcell.physiology.org/content/296/6/C1258
  • [16]McPherron AC, Lawler AM, Lee SJ: Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature 1997, 387:83-90. http://www.ncbi.nlm.nih.gov/pubmed/9139826
  • [17]Grobet L, Martin LJ, Poncelet D, Pirottin D: A deletion in the bovine myostatin gene causes the double muscled phenotype in cattle. Nat Genet 1997, 17:71-74. http://www.nature.com/ng/journal/v17/n1/pdf/ng0997-71.pdf
  • [18]Wagner KR, McPherron AC, Winik N, Lee SJ: Loss of myostatin attenuates severity of muscular dystrophy in mdx mice. Ann Neurol 2002, 52:832-836. http://www.ncbi.nlm.nih.gov/pubmed/12447939
  • [19]Zhang N, Zhang X, Liu M, Tan L: Ovine Follistatin gene expression and functional analysis of Follistatin domains. Chin J Biotech 2010, 26:1050-1056. http://www.ncbi.nlm.nih.gov/pubmed/21090108
  • [20]Benabdallah BF, Bouchentouf M, Rousseau J, Tremblay JP: Overexpression of Follistatin in human myoblasts increases their proliferation and differentiation, and improves the graft success in SCID mice. Cell Transplant 2009, 18:709-718. http://www.ncbi.nlm.nih.gov/pubmed/19520047
  • [21]Keutmann HT, Schneyer AL, Sidis Y: The role of Follistatin domains in Follistatin biological action. Mol Endocrinol 2004, 18:228-240. http://www.ncbi.nlm.nih.gov/pubmed/14563935
  • [22]Sharma M, Kambadur R, Matthews KG, Somers WG, Devlin GP: Myostatin, a transforming growth factor-beta superfamily member, is expressed in heart muscle and is upregulated in cardiomyocytes after infarct. J Cell Physio 1999, 180:1-9. http://www.ncbi.nlm.nih.gov/pubmed/10362012
  • [23]Thomas M, Langley B, Berry C, Sharma M, Kirk S, Bass J, Kambadur R: Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast proliferation. J Biol Chem 2000, 275:40235-40243. http://www.jbc.org/content/275/51/40235.long
  • [24]Li ZB, Kollias HD, Wagner KR: Myostatin directly regulates skeletal muscle fibrosis. J Biol Chem 2008, 283:19371-19378. http://www.ncbi.nlm.nih.gov/pubmed/18453534
  • [25]Zhu X, Hadhazy M, Wehling M, Tidball JG, McNally EM: Dominant negative myostatin produces hypertrophy without hyperplasia in muscle. FEBS Lett 2000, 474:71-75. http://www.ncbi.nlm.nih.gov/pubmed/10828454
  • [26]Taylor WE, Bhasin S, Artaza J, Byhower F, Azam M: Myostatin inhibits cell proliferation and protein synthesis in C2C12 muscle cells. Am J Physiol Endocrinol Metab 2001, 280:E221-E228. http://www.ncbi.nlm.nih.gov/pubmed/11158924
  • [27][http://www.cshlpress.com/pdf/sample/2013/MC4/MC4FM.pdf] webcite Sambrook J, Michael RG: Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press; 2012. .
  • [28]Hembree JR, Hathaway MR, Dayton WR: Isolation and culture of fetal porcine myogenic cells and the effect of insulin, IGF-1, and sera on protein turnover in porcine myotube cultures. J Anim Sci 1991, 69:3241-3250. http://www.journalofanimalscience.org/content/69/8/3241.full.pdf
  • [29]Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C (T)) Method. Methods 2001, 25:402-408. http://www.sciencedirect.com/science/article/pii/S1046202301912629
  文献评价指标  
  下载次数:53次 浏览次数:11次