DOI QR코드

DOI QR Code

Some Motifs Were Important for Myostatin Transcriptional Regulation in Sheep (Ovis aries)

  • Du, Rong (State Key Laboratory for Agrobiotechnology, College of Biological Science, China Agricultural University) ;
  • An, Xiao-Rong (State Key Laboratory for Agrobiotechnology, College of Biological Science, China Agricultural University) ;
  • Chen, Yong-Fu (State Key Laboratory for Agrobiotechnology, College of Biological Science, China Agricultural University) ;
  • Qin, Jian (College of Modern Education Technology, Shanxi Agricultural University)
  • Published : 2007.07.31

Abstract

Many motifs along the 1.2 kb myostatin promoter (MSTNpro) in sheep have been found by the MatInspecter program in our recent study. To further verify the role of the motifs and better understand the transcriptional regulation mechanism of the myostatin gene in sheep, the reporter gene EGFP (enhanced green fluorescent protein) was selected and the wild-type (W) vector MSTNPro$^W$-EGFP or motif-mutational (M) vector MSTNPro$^M$-EGFP were constructed. The transcriptional regulation activities were analyzed by detecting the fluorescence strength of EGFP in C2C12 myoblasts transfected with the vectors. The results showed that E-box (E) 3, E4, E5 and E7, particularly E3, E5 and E7, had important effects on the activity of the 1.2 kb sheep myostatin promoter. In addition, we also detected several other important motifs such as MTBF (muscle-specific Mt binding factor), MEF2 (myocyte enhancer factor 2), GRE (glucocorticoid response elements) and PRE (progesterone response elements) along the sheep myostatin promoter by the mutational analysis.

Keywords

References

  1. Akkila, W. M., Chambers, R. L., Ornatsky, O. I. and McDermott, J. C. (1997) Molecular cloning of up-regulated cytoskeletal genes from regenerating skeletal muscle: potential role of myocyte enhancer factor 2 proteins in the activation of muscleregeneration-associated genes. Biochem. J. 325, 87-93. https://doi.org/10.1042/bj3250087
  2. Apone, S. and Hauschka, S. D. (1995) Muscle gene E-box control elements. Evidence for quantitatively different transcriptional activities and the binding of distinct regulatory factors. J. Biol. Chem. 270, 21420-21427. https://doi.org/10.1074/jbc.270.36.21420
  3. Catala, F., Wanner, R., Barton, P., Cohen, A., Wright, W. and Buckingham, M. (1995) A skeletal muscle-specific enhancer regulated by factors binding to E and CArG boxes is present in the promoter of the mouse myosin light-chain 1A gene. Mol. Cell. Biol. 15, 4585-4596. https://doi.org/10.1128/MCB.15.8.4585
  4. Ceccarelli, E., McGrew, M. J., Nguyen, T., Grieshammer, U., Horgan, D., Hughes, S. H. and Rosenthal, N. (1999) An E box comprises a positional sensor for regional differences in skeletal muscle gene expression and methylation. Dev. Biol. 213, 217-229. https://doi.org/10.1006/dbio.1999.9345
  5. Du, R., Chen, Y. F., An, X. R., Yang, X. Y., Ma, Y., Zhang, L., Yuan, X. L., Chen, L. M. and Qin, J. (2005) Cloning and sequence analysis of myostatin promoter in sheep. DNA Seq. 16, 412-417. https://doi.org/10.1080/10425170500226474
  6. Goodlad, G. A. and Clark, C. M. (1991) Glucocorticoid-mediated muscle atrophy: alterations in transcriptional activity of skeletal muscle nuclei. Biochim. Biophys. Acta 1097, 166-170. https://doi.org/10.1016/0925-4439(91)90030-D
  7. Grobet, L., Martin, L. J., Poncelet, D., Pirottin, D., Brouwers, B., Riquet, J., Schoeberlein, A., Dunner, S., Menissier, F., Massabanda, J., Fries, R., Hanset, R. and Georges, M. (1997) A deletion in the bovine myostatin gene causes the doublemuscled phenotype in cattle. Nat. Genet. 17, 71-74. https://doi.org/10.1038/ng0997-71
  8. Grobet, L., Poncelet, D., Royo, L. J., Brouwers, B., Pirottin, D., Michaux, C., Menissier, F., Zanotti, M., Dunner, S. and Georges, M. (1998) Molecular definition of an allele series of mutations disrupting the myostatin function and causing double-muscling in cattle. Mamm. Genome. 9, 210-213. https://doi.org/10.1007/s003359900727
  9. Gu, Z., Zhang, Y., Shi, P., Zhang, Y. P., Zhu, D. and Li, H. (2004) Comparison of avian myostatin genes. Anim. Genet. 35, 470-472. https://doi.org/10.1111/j.1365-2052.2004.01194.x
  10. Hasselgren, P. O. (1999) Glucocorticoids and muscle catabolism. Curr. Opin. Clin. Nutr. Metab. Care. 2, 201-205. https://doi.org/10.1097/00075197-199905000-00002
  11. Kambadur, R., Sharma, M., Smith, T. P. and Bass, J. J. (1997) Mutations in myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle. Genome Res. 7, 910-916. https://doi.org/10.1101/gr.7.9.910
  12. Kerr, T., Roalson, E. H. and Rodgers, B. D. (2005) Phylogenetic analysis of the myostatin gene sub-family and the differential expression of a novel member in zebrafish. Evol. Dev. 7, 390-400. https://doi.org/10.1111/j.1525-142X.2005.05044.x
  13. Lee, S. J. and McPherron, A. C. (2001) Regulation of Myostatin activity and muscle growth. Proc. Natl. Acad. Sci. USA 98, 9306-9311. https://doi.org/10.1073/pnas.151270098
  14. Lee-Hoeflich, S. T., Zhao, X., Mehra, A. and Attisano, L. (2005) The Drosophila type II receptor, Wishful thinking, binds BMP and myoglianin to activate multiple $TGF{\alpha}$ family signaling pathways. FEBS Lett. 579, 4615-4621. https://doi.org/10.1016/j.febslet.2005.06.088
  15. Lyons, G. E., Micales, B. K., Schwarz, J., Martin, J. F. and Olson, E. N. (1995) Expression of mef2 genes in the mouse central nervous system suggests a role in neuronal maturation. J. Neurosci. 15, 5727-5738. https://doi.org/10.1523/JNEUROSCI.15-08-05727.1995
  16. Ma, K., Mallidis, C., Artaza, J., Taylor, W., Gonzalez-cadavid, N. and Bhasin, S. (2001) Characterization of 5'-regulatory region of human myostatin gene: regulation by dexamethasone in vitro. Am. J. Physiol. Endocrinol. Metab. 281, 1128-1136. https://doi.org/10.1152/ajpendo.2001.281.6.E1128
  17. Ma, K., Mallidis, C., Bhasin, S., Mahabadi, V., Artaza, J., Gonzalez-Cadavid, N., Arias, J. and Salehian, B. (2003) Glucocorticoid-induced skeletal muscle atrophy is associated with upregulation of myostatin gene expression. Am. J. Physiol. Endocrinol. Metab. 285, 363-371. https://doi.org/10.1152/ajpendo.00487.2002
  18. McPherron, A. C. and Lee, S. J. (1997b) Double muscling in cattle due to mutations in the myostatin gene. Proc. Natl. Acad. Sci. USA 94, 12457-12461. https://doi.org/10.1073/pnas.94.23.12457
  19. McPherron, A. C., Lawler, A. M. and Lee, S. J. (1997a) Regulation of skeletal muscle mass in mice by a new $TGF-{\beta}$ superfamily member. Nature 387, 83-90. https://doi.org/10.1038/387083a0
  20. Olson, E. N., Perry, M. and Schulz, R. A. (1995) Regulation of muscle differentiation by the MEF2 family of MADS box transcription factors. Dev. Biol. 172, 2-14. https://doi.org/10.1006/dbio.1995.0002
  21. Rao, M. V., Donoghue, M. J., Merlie, J. P. and Sanes, J. R. (1996) Distinct regulatory elements control muscle-specific, fiber-typeselective, and axially graded expression of a myosin light-chain gene in transgenic mice. Mol. Cell. Biol. 16, 3909-3922. https://doi.org/10.1128/MCB.16.7.3909
  22. Rios, R., Fernandez-Nocelos, S., Carneiro, I., Arce, V. M. and Devesa, J. (2004) Differential response to exogenous and endogenous myostatin in myoblasts suggests myostatin acts as an autocrine factor in vivo. Endocrinology 145, 2795-2803. https://doi.org/10.1210/en.2003-1166
  23. Roberts, S. B. and Goetz, F. W. (2001) Differential skeletal muscle expression of myostatin across teleost species, and the isolation of multiple myostatin isoforms. FEBS Lett. 491, 212-216. https://doi.org/10.1016/S0014-5793(01)02196-2
  24. Salerno, M. S., Thomas, M., Forbes, D., Watson, T., Kambadur, R. and Sharma, M. (2004) Molecular analysis of fiber typespecific expression of murine myostatin promoter. Am. J. Physiol. Cell. Physiol. 287, 1031-1040. https://doi.org/10.1152/ajpcell.00492.2003
  25. Spiller, M. P., Kambadur, R., Jeanplong, F., Thomas, M., Martyn, J. K., Bass, J. J. and Sharma, M. (2002) The myostatin gene is a downstream target gene of basic helix-loop-helix transcription factor MyoD. Mol. Cell. Biol. 22, 7066-7082. https://doi.org/10.1128/MCB.22.20.7066-7082.2002

Cited by

  1. Association of myostatin variants with growth traits of Zhikong scallop (Chlamys farreri) vol.15, pp.1, 2016, https://doi.org/10.1007/s11802-016-2633-5
  2. SNPs in minimal promoter of myostatin (GDF-8) gene and its association with body weight in broiler chicken vol.42, pp.3, 2014, https://doi.org/10.1080/09712119.2013.846859
  3. An Essential Role of Small Ubiquitin-like Modifier (SUMO)-specific Protease 2 in Myostatin Expression and Myogenesis vol.289, pp.6, 2014, https://doi.org/10.1074/jbc.M113.518282
  4. Two single nucleotide polymorphisms in the promoter of the ovine myostatin gene (MSTN) and their effect on growth and carcass muscle traits in New Zealand Romney sheep vol.133, pp.3, 2016, https://doi.org/10.1111/jbg.12171
  5. Analysis of polymorphisms in the equine MSTN gene in Polish populations of horse breeds vol.187, 2016, https://doi.org/10.1016/j.livsci.2016.03.012
  6. Structural and functional analysis of myostatin-2 promoter alleles from the marine fish Sparus aurata: Evidence for strong muscle-specific promoter activity and post-transcriptional regulation vol.361, pp.1-2, 2012, https://doi.org/10.1016/j.mce.2012.03.017
  7. Genomic cloning and promoter functional analysis of myostatin-2 in shi drum, Umbrina cirrosa: Conservation of muscle-specific promoter activity vol.164, pp.2, 2013, https://doi.org/10.1016/j.cbpb.2012.11.003
  8. A promoter polymorphism of MSTN g.−371T>A and its associations with carcass traits in Korean cattle vol.39, pp.4, 2012, https://doi.org/10.1007/s11033-011-1153-z
  9. Characterization of 5′ Upstream Region and Investigation of TTTTA Deletion in 5′ UTR of Myostatin (MSTN) Gene in Indian Goat Breeds vol.25, pp.1, 2014, https://doi.org/10.1080/10495398.2013.821994
  10. Dexamethasone-induced skeletal muscle atrophy was associated with upregulation of myostatin promoter activity vol.94, pp.1, 2013, https://doi.org/10.1016/j.rvsc.2012.07.018
  11. Single nucleotide polymorphisms in the upstream regulatory region alter the expression of myostatin vol.49, pp.6, 2013, https://doi.org/10.1007/s11626-013-9621-5
  12. Molecular Characterization of Myostatin Gene from Zhikong scallop Chlamys farreri (Jones et Preston 1904) vol.85, pp.3, 2010, https://doi.org/10.1266/ggs.85.207
  13. Analysis of the Single-Nucleotide Polymorphism in the 5′UTR and Part of Intron I of the SheepMSTNGene vol.30, pp.7, 2011, https://doi.org/10.1089/dna.2010.1153
  14. An evolutionarily conserved Myostatin proximal promoter/enhancer confers basal levels of transcription and spatial specificity in vivo vol.219, pp.9-10, 2009, https://doi.org/10.1007/s00427-009-0312-x
  15. CONVIRT: A web-based tool for transcriptional regulatory site identification using a conserved virtual chromosome vol.42, pp.12, 2009, https://doi.org/10.5483/BMBRep.2009.42.12.823
  16. Low-protein diet fed to crossbred sows during pregnancy and lactation enhances myostatin gene expression through epigenetic regulation in skeletal muscle of weaning piglets vol.55, pp.3, 2016, https://doi.org/10.1007/s00394-015-0949-3
  17. Functional analysis of pig myostatin gene promoter with some adipogenesis- and myogenesis-related factors vol.363, pp.1-2, 2012, https://doi.org/10.1007/s11010-011-1181-y
  18. The virtual element in proximal promoter of porcine myostatin is regulated by myocyte enhancer factor 2C vol.419, pp.2, 2012, https://doi.org/10.1016/j.bbrc.2012.01.135
  19. Gene Promoter vol.36, pp.12, 2017, https://doi.org/10.1089/dna.2017.3762