Skip to main content
Erschienen in: European Journal of Nutrition 3/2016

12.06.2015 | Original Contribution

Low-protein diet fed to crossbred sows during pregnancy and lactation enhances myostatin gene expression through epigenetic regulation in skeletal muscle of weaning piglets

verfasst von: Yimin Jia, Guichao Gao, Haogang Song, Demin Cai, Xiaojing Yang, Ruqian Zhao

Erschienen in: European Journal of Nutrition | Ausgabe 3/2016

Einloggen, um Zugang zu erhalten

Abstract

Purpose

This study was aimed to investigate the effects of a maternal low-protein diet on transcriptional regulation of the myostatin (MSTN) gene in skeletal muscle of weaning piglets.

Methods

Sows were fed either a standard-protein (SP, 15 and 18 % crude protein) or a low-protein (LP, 50 % protein level of SP) diet throughout pregnancy and lactation. Longissimus dorsi muscle was sampled from male piglets at 28 days of age. The mRNA was determined by RT-PCR, and protein was measured by Western blot. Chromatin immunoprecipitation assay was used to determine the binding of transcription factors and histone H3 modifications on the MSTN gene promoter.

Results

The maternal LP diet significantly decreased body weight and average daily gain (P < 0.05), which was associated with significantly lower plasma concentration of urea nitrogen and total protein (P < 0.05), as well as decreased muscle RNA content (P < 0.05). MSTN mRNA (P < 0.05) was significantly increased, together with enhanced (P < 0.05) mRNA and protein expression of forkhead box class O family member protein 3 (FoxO3), and a tendency of an increase (P = 0.10) in glucocorticoid receptor (GR) mRNA in the muscle of LP piglets. Furthermore, the binding of both FoxO3 and GR to the MSTN gene promoter was significantly higher (P < 0.05) in muscle of LP piglets, together with significantly enriched (P < 0.05) gene activation markers, H3K9Ac and H3K4me3.

Conclusion

These results indicate that MSTN mediates maternal LP diet-induced growth retardation, through epigenetic regulation involving FoxO3 and GR binding to its promoter.
Literatur
1.
Zurück zum Zitat Joulia D, Bernardi H, Garandel V, Rabenoelina F, Vernus B, Cabello G (2003) Mechanisms involved in the inhibition of myoblast proliferation and differentiation by myostatin. Exp Cell Res 286:263–275CrossRef Joulia D, Bernardi H, Garandel V, Rabenoelina F, Vernus B, Cabello G (2003) Mechanisms involved in the inhibition of myoblast proliferation and differentiation by myostatin. Exp Cell Res 286:263–275CrossRef
2.
Zurück zum Zitat Welle S, Burgess K, Thornton CA, Tawil R (2009) Relation between extent of myostatin depletion and muscle growth in mature mice. Am J Physiol Endocrinol Metab 297:E935–E940CrossRef Welle S, Burgess K, Thornton CA, Tawil R (2009) Relation between extent of myostatin depletion and muscle growth in mature mice. Am J Physiol Endocrinol Metab 297:E935–E940CrossRef
3.
Zurück zum Zitat Wang Q, McPherron AC (2012) Myostatin inhibition induces muscle fibre hypertrophy prior to satellite cell activation. J Physiol 590:2151–2165CrossRef Wang Q, McPherron AC (2012) Myostatin inhibition induces muscle fibre hypertrophy prior to satellite cell activation. J Physiol 590:2151–2165CrossRef
4.
Zurück zum Zitat Hennebry A, Berry C, Siriett V, O’Callaghan P, Chau L, Watson T, Sharma M, Kambadur R (2009) Myostatin regulates fiber-type composition of skeletal muscle by regulating MEF2 and MyoD gene expression. Am J Physiol Cell Physiol 296:C525–C534CrossRef Hennebry A, Berry C, Siriett V, O’Callaghan P, Chau L, Watson T, Sharma M, Kambadur R (2009) Myostatin regulates fiber-type composition of skeletal muscle by regulating MEF2 and MyoD gene expression. Am J Physiol Cell Physiol 296:C525–C534CrossRef
5.
Zurück zum Zitat Reisz-Porszasz S, Bhasin S, Artaza JN, Shen R, Sinha-Hikim I, Hogue A, Fielder TJ, Gonzalez-Cadavid NF (2003) Lower skeletal muscle mass in male transgenic mice with muscle-specific overexpression of myostatin. Am J Physiol Endocrinol Metab 285:E876–E888CrossRef Reisz-Porszasz S, Bhasin S, Artaza JN, Shen R, Sinha-Hikim I, Hogue A, Fielder TJ, Gonzalez-Cadavid NF (2003) Lower skeletal muscle mass in male transgenic mice with muscle-specific overexpression of myostatin. Am J Physiol Endocrinol Metab 285:E876–E888CrossRef
6.
Zurück zum Zitat Zhu X, Topouzis S, Liang LF, Stotish RL (2004) Myostatin signaling through Smad2, Smad3 and Smad4 is regulated by the inhibitory Smad7 by a negative feedback mechanism. Cytokine 26:262–272CrossRef Zhu X, Topouzis S, Liang LF, Stotish RL (2004) Myostatin signaling through Smad2, Smad3 and Smad4 is regulated by the inhibitory Smad7 by a negative feedback mechanism. Cytokine 26:262–272CrossRef
7.
Zurück zum Zitat Yang W, Chen Y, Zhang Y, Wang X, Yang N, Zhu D (2006) Extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway is involved in myostatin-regulated differentiation repression. Cancer Res 66:1320–1326CrossRef Yang W, Chen Y, Zhang Y, Wang X, Yang N, Zhu D (2006) Extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway is involved in myostatin-regulated differentiation repression. Cancer Res 66:1320–1326CrossRef
8.
Zurück zum Zitat Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117:399–412CrossRef Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117:399–412CrossRef
9.
Zurück zum Zitat Peiris HN, Ponnampalam AP, Osepchook CC, Mitchell MD, Green MP (2010) Placental expression of myostatin and follistatin-like-3 protein in a model of developmental programming. Am J Physiol Endocrinol Metab 298:E854–E861CrossRef Peiris HN, Ponnampalam AP, Osepchook CC, Mitchell MD, Green MP (2010) Placental expression of myostatin and follistatin-like-3 protein in a model of developmental programming. Am J Physiol Endocrinol Metab 298:E854–E861CrossRef
10.
Zurück zum Zitat Reed SA, Raja JS, Hoffman ML, Zinn SA, Govoni KE (2014) Poor maternal nutrition inhibits muscle development in ovine offspring. J Anim Sci Biotechnol 5(1):43CrossRef Reed SA, Raja JS, Hoffman ML, Zinn SA, Govoni KE (2014) Poor maternal nutrition inhibits muscle development in ovine offspring. J Anim Sci Biotechnol 5(1):43CrossRef
11.
Zurück zum Zitat Liu X, Wang J, Li R, Yang X, Sun Q, Albrecht E, Zhao R (2011) Maternal dietary protein affects transcriptional regulation of myostatin gene distinctively at weaning and finishing stages in skeletal muscle of Meishan pigs. Epigenetics 6:899–907CrossRef Liu X, Wang J, Li R, Yang X, Sun Q, Albrecht E, Zhao R (2011) Maternal dietary protein affects transcriptional regulation of myostatin gene distinctively at weaning and finishing stages in skeletal muscle of Meishan pigs. Epigenetics 6:899–907CrossRef
12.
Zurück zum Zitat McMillen IC, Robinson JS (2005) Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev 85:571–633CrossRef McMillen IC, Robinson JS (2005) Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev 85:571–633CrossRef
13.
Zurück zum Zitat Jones PA (2012) Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13:484–492CrossRef Jones PA (2012) Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13:484–492CrossRef
14.
Zurück zum Zitat Shahbazian MD, Grunstein M (2007) Functions of site-specific histone acetylation and deacetylation. Annu Rev Biochem 76:75–100CrossRef Shahbazian MD, Grunstein M (2007) Functions of site-specific histone acetylation and deacetylation. Annu Rev Biochem 76:75–100CrossRef
15.
Zurück zum Zitat Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, Paylor RE, Lubin FD (2010) Histone methylation regulates memory formation. J Neurosci 30:3589–3599CrossRef Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, Paylor RE, Lubin FD (2010) Histone methylation regulates memory formation. J Neurosci 30:3589–3599CrossRef
16.
Zurück zum Zitat Huang Y, Greene E, Murray Stewart T, Goodwin AC, Baylin SB, Woster PM, Casero RA Jr (2007) Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc Natl Acad Sci USA 104:8023–8028CrossRef Huang Y, Greene E, Murray Stewart T, Goodwin AC, Baylin SB, Woster PM, Casero RA Jr (2007) Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc Natl Acad Sci USA 104:8023–8028CrossRef
17.
Zurück zum Zitat Bettini ML, Pan F, Bettini M, Finkelstein D, Rehg JE, Floess S, Bell BD, Ziegler SF, Huehn J, Pardoll DM, Vignali DA (2012) Loss of epigenetic modification driven by the Foxp3 transcription factor leads to regulatory T cell insufficiency. Immunity 36:717–730CrossRef Bettini ML, Pan F, Bettini M, Finkelstein D, Rehg JE, Floess S, Bell BD, Ziegler SF, Huehn J, Pardoll DM, Vignali DA (2012) Loss of epigenetic modification driven by the Foxp3 transcription factor leads to regulatory T cell insufficiency. Immunity 36:717–730CrossRef
18.
Zurück zum Zitat Watson ML, Baehr LM, Reichardt HM, Tuckermann JP, Bodine SC, Furlow JD (2012) A cell-autonomous role for the glucocorticoid receptor in skeletal muscle atrophy induced by systemic glucocorticoid exposure. Am J Physiol Endocrinol Metab 302:E1210–E1220CrossRef Watson ML, Baehr LM, Reichardt HM, Tuckermann JP, Bodine SC, Furlow JD (2012) A cell-autonomous role for the glucocorticoid receptor in skeletal muscle atrophy induced by systemic glucocorticoid exposure. Am J Physiol Endocrinol Metab 302:E1210–E1220CrossRef
19.
Zurück zum Zitat Allen DL, Unterman TG (2007) Regulation of myostatin expression and myoblast differentiation by FoxO and SMAD transcription factors. Am J Physiol Cell Physiol 292:C188–C199CrossRef Allen DL, Unterman TG (2007) Regulation of myostatin expression and myoblast differentiation by FoxO and SMAD transcription factors. Am J Physiol Cell Physiol 292:C188–C199CrossRef
20.
Zurück zum Zitat Johnson LR, Chandler AM (1973) RNA and DNA of gastric and duodenal mucosa in antrectomized and gastrin-treated rats. Am J Physio 224:937–940 Johnson LR, Chandler AM (1973) RNA and DNA of gastric and duodenal mucosa in antrectomized and gastrin-treated rats. Am J Physio 224:937–940
21.
Zurück zum Zitat Dwyer CM, Stickland NC, Fletcher JM (1994) The influence of maternal nutrition on muscle fiber number development in the porcine fetus and on subsequent postnatal growth. J Anim Sci 72:911–917 Dwyer CM, Stickland NC, Fletcher JM (1994) The influence of maternal nutrition on muscle fiber number development in the porcine fetus and on subsequent postnatal growth. J Anim Sci 72:911–917
22.
Zurück zum Zitat Zambrano E, Bautista CJ, Deas M, Martinez-Samayoa PM, Gonzalez-Zamorano M, Ledesma H, Morales J, Larrea F, Nathanielsz PW (2006) A low maternal protein diet during pregnancy and lactation has sex- and window of exposure-specific effects on offspring growth and food intake, glucose metabolism and serum leptin in the rat. J Physiol 571:221–230CrossRef Zambrano E, Bautista CJ, Deas M, Martinez-Samayoa PM, Gonzalez-Zamorano M, Ledesma H, Morales J, Larrea F, Nathanielsz PW (2006) A low maternal protein diet during pregnancy and lactation has sex- and window of exposure-specific effects on offspring growth and food intake, glucose metabolism and serum leptin in the rat. J Physiol 571:221–230CrossRef
23.
Zurück zum Zitat Addis T, Barrett E, Poo LJ, Yuen DW (1947) The relation between the serum urea concentration and the protein consumption of normal individuals. J Clin Invest 26:869–874CrossRef Addis T, Barrett E, Poo LJ, Yuen DW (1947) The relation between the serum urea concentration and the protein consumption of normal individuals. J Clin Invest 26:869–874CrossRef
24.
Zurück zum Zitat Wu G, Pond WG, Ott T, Bazer FW (1998) Maternal dietary protein deficiency decreases amino acid concentrations in fetal plasma and allantoic fluid of pigs. J Nutr 128:894–902 Wu G, Pond WG, Ott T, Bazer FW (1998) Maternal dietary protein deficiency decreases amino acid concentrations in fetal plasma and allantoic fluid of pigs. J Nutr 128:894–902
25.
Zurück zum Zitat Millward DJ, Garlick PJ, James WP, Nnanyelugo DO, Ryatt JS (1973) Relationship between protein synthesis and RNA content in skeletal muscle. Nature 241:204–205CrossRef Millward DJ, Garlick PJ, James WP, Nnanyelugo DO, Ryatt JS (1973) Relationship between protein synthesis and RNA content in skeletal muscle. Nature 241:204–205CrossRef
26.
Zurück zum Zitat Schakman O, Kalista S, Barbe C, Loumaye A, Thissen JP (2013) Glucocorticoid-induced skeletal muscle atrophy. Int J Biochem Cell Biol 45:2163–2172CrossRef Schakman O, Kalista S, Barbe C, Loumaye A, Thissen JP (2013) Glucocorticoid-induced skeletal muscle atrophy. Int J Biochem Cell Biol 45:2163–2172CrossRef
27.
Zurück zum Zitat Ma K, Mallidis C, Artaza J, Taylor W, Gonzalez-Cadavid N, Bhasin S (2001) Characterization of 5′-regulatory region of human myostatin gene: regulation by dexamethasone in vitro. Am J Physiol Endocrinol Metab 281:E1128–E1136 Ma K, Mallidis C, Artaza J, Taylor W, Gonzalez-Cadavid N, Bhasin S (2001) Characterization of 5′-regulatory region of human myostatin gene: regulation by dexamethasone in vitro. Am J Physiol Endocrinol Metab 281:E1128–E1136
28.
Zurück zum Zitat Ma K, Mallidis C, Bhasin S, Mahabadi V, Artaza J, Gonzalez-Cadavid N, Arias J, Salehian B (2003) Glucocorticoid-induced skeletal muscle atrophy is associated with upregulation of myostatin gene expression. Am J Physiol Endocrinol Metab 285:E363–E371CrossRef Ma K, Mallidis C, Bhasin S, Mahabadi V, Artaza J, Gonzalez-Cadavid N, Arias J, Salehian B (2003) Glucocorticoid-induced skeletal muscle atrophy is associated with upregulation of myostatin gene expression. Am J Physiol Endocrinol Metab 285:E363–E371CrossRef
29.
Zurück zum Zitat Du R, An XR, Chen YF, Qin J (2007) Some motifs were important for myostatin transcriptional regulation in sheep (Ovis aries). J Biochem Mol Biol 40:547–553CrossRef Du R, An XR, Chen YF, Qin J (2007) Some motifs were important for myostatin transcriptional regulation in sheep (Ovis aries). J Biochem Mol Biol 40:547–553CrossRef
30.
Zurück zum Zitat Qin J, Du R, Yang YQ, Zhang HQ, Li Q, Liu L, Guan H, Hou J, An XR (2013) Dexamethasone-induced skeletal muscle atrophy was associated with upregulation of myostatin promoter activity. Res Vet Sci 94:84–89CrossRef Qin J, Du R, Yang YQ, Zhang HQ, Li Q, Liu L, Guan H, Hou J, An XR (2013) Dexamethasone-induced skeletal muscle atrophy was associated with upregulation of myostatin promoter activity. Res Vet Sci 94:84–89CrossRef
31.
Zurück zum Zitat Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M (2007) FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6:458–471CrossRef Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M (2007) FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6:458–471CrossRef
32.
Zurück zum Zitat Han DS, Huang HP, Wang TG, Hung MY, Ke JY, Chang KT, Chang HY, Ho YP, Hsieh WY, Yang WS (2010) 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 111:564–573CrossRef Han DS, Huang HP, Wang TG, Hung MY, Ke JY, Chang KT, Chang HY, Ho YP, Hsieh WY, Yang WS (2010) 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 111:564–573CrossRef
Metadaten
Titel
Low-protein diet fed to crossbred sows during pregnancy and lactation enhances myostatin gene expression through epigenetic regulation in skeletal muscle of weaning piglets
verfasst von
Yimin Jia
Guichao Gao
Haogang Song
Demin Cai
Xiaojing Yang
Ruqian Zhao
Publikationsdatum
12.06.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Nutrition / Ausgabe 3/2016
Print ISSN: 1436-6207
Elektronische ISSN: 1436-6215
DOI
https://doi.org/10.1007/s00394-015-0949-3

Weitere Artikel der Ausgabe 3/2016

European Journal of Nutrition 3/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.