2,855
Views
24
CrossRef citations to date
0
Altmetric
Research Paper

Differences in the fast muscle methylome provide insight into sex-specific epigenetic regulation of growth in Nile tilapia during early stages of domestication

ORCID Icon, , & ORCID Icon
Pages 818-836 | Received 30 Dec 2018, Accepted 04 May 2019, Published online: 25 May 2019

References

  • Johnston IA. Environment and plasticity of myogenesis in teleost fish. J Exp Biol. 2006;209(12):2249–2264.
  • van der Most JP, de Jong B, Parmentier KH, et al. Trade-off between growth and immune function: a meta-analysis of selection experiments. Funct Ecol. 2010;25(1):74–80.
  • Hagen Ø, Fernandes JMO, Solberg C, et al. Expression of growth-related genes in muscle during fasting and refeeding of juvenile Atlantic halibut, Hippoglossus hippoglossus L. Comp Biochem Physiol Part B Biochem Mol Biol. 2009;152(1):47–53.
  • Valente LMP, Moutou KA, Lec C, et al. What determines growth potential and juvenile quality of farmed fish species? Rev Aquac. 2013;5:S168–S193.
  • Nagasawa K, Giannetto A, Fernandes JMO. Photoperiod influences growth and MLL (mixed-lineage leukaemia) expression in Atlantic cod. PLoS One. 2012;7:5.
  • Biro PA, Abrahams MV, Post JR, et al. Behavioural trade-offs between growth and mortality explain evolution of submaximal growth rates. J Anim Ecol. 2006;75(5):1165–1171.
  • Runemark A, Brydegaard M, Svensson EI. Does relaxed predation drive phenotypic divergence among insular populations? J Evol Biol. 2014;27(8):1676–1690.
  • Huntingford FA. Implications of domestication and rearing conditions for the behaviour of cultivated fishes. J Fish Biol. 2004;65(s1):122–142.
  • Elsbeth McPhee M. Generations in captivity increases behavioral variance: considerations for captive breeding and reintroduction programs. Biol Conserv. 2004;115(1):71–77.
  • Teletchea F, Fontaine P. Levels of domestication in fish: implications for the sustainable future of aquaculture. Fish Fisheries. 2014;15(2):181–195.
  • Solberg MF, Ø S, Nilsen F, et al. Does domestication cause changes in growth reaction norms? A study of farmed, wild and hybrid Atlantic salmon families exposed to environmental stress. PLoS One. 2013;8(1):e54469.
  • Christie MR, Marine ML, Fox SE, et al. A single generation of domestication heritably alters the expression of hundreds of genes. Nat Commun. 2016;7. DOI:10.1038/ncomms10676.
  • Swain DP, Riddell BE, Murray CB. Morphological differences between hatchery and wild populations of coho salmon (Oncorhynchus kisutch): environmental versus genetic origin. Can J Fisheries Aquat Sci. 1991;48(9):1783–1791.
  • Barros SP, Offenbacher S. Epigenetics: connecting environment and genotype to phenotype and disease. J Dent Res. 2009;88(5):400–408.
  • Le Luyer J, Laporte M, Beacham TD, et al. Parallel epigenetic modifications induced by hatchery rearing in a Pacific salmon. Proc Natl Acad Sci USA. 2017;114(49):12964–12969.
  • Sun LX, Wang YY, Zhao Y, et al. Global DNA methylation changes in Nile Tilapia gonads during high temperature-induced masculinization. PLoS One. 2016;11(8):e0158483.
  • Wan ZY, Xia JH, Lin G, et al. Genome-wide methylation analysis identified sexually dimorphic methylated regions in hybrid tilapia. Sci Rep. 2016;6:35903.
  • Burgerhout E, Mommens M, Johnsen H, et al. Genetic background and embryonic temperature affect DNA methylation and expression of myogenin and muscle development in Atlantic salmon (Salmo salar). PLoS One. 2017;12(6):e0179918.
  • Campos C, Valente LM, Conceicao LE, et al. Temperature affects methylation of the myogenin putative promoter, its expression and muscle cellularity in Senegalese sole larvae. Epigenetics. 2013 Apr;8(4):389–397. PubMed PMID: 23538611; PubMed Central PMCID: PMCPMC3674048. eng.
  • Moghadam H, Mørkøre T, Robinson N. Epigenetics-potential for programming fish for aquaculture? J Mar Sci Eng. 2015;3(2):175–192.
  • Soltan MA, Hassaan MS, Meshrf RN. Response of Nile tilapia (Oreochromis niloticus) to diet acidification: effect on growth performance and feed utilization. J Appl Aquacult. 2017;29(3–4):207–219.
  • Charo-Karisa H, Komen H, Reynolds S, et al. Genetic and environmental factors affecting growth of Nile tilapia (Oreochromis niloticus) juveniles: modelling spatial correlations between hapas. Aquaculture. 2006;255(1–4):586–596.
  • Lind CE, Safari A, Agyakwah SK, et al. Differences in sexual size dimorphism among farmed tilapia species and strains undergoing genetic improvement for body weight. Aquacult Rep. 2015;1:20–27.
  • Byun SK, An TH, Son MJ, et al. HDAC11 inhibits myoblast differentiation through repression of MyoD-dependent transcription. Mol Cells. 2017;40(9):667–676.
  • Yu T, Luo G, Zhang L, et al. Leptin promotes proliferation and inhibits differentiation in porcine skeletal myoblasts. Biosci Biotechnol Biochem. 2008;72(1):13–21.
  • Yang JT, Rando TA, Mohler WA, et al. Genetic analysis of α4 integrin functions in the development of mouse skeletal muscle. J Cell Biol. 1996;135(3):829–835.
  • Mendias CL, Gumucio JP, Davis ME, et al. Transforming growth factor-beta induces skeletal muscle atrophy and fibrosis through the induction of atrogin-1 and scleraxis. Muscle Nerve. 2012;45(1):55–59.
  • Obara K, Ohsumi Y. Atg14: a key player in orchestrating autophagy. Int J Cell Biol. 2011;2011:713435.
  • Seiliez I, Gutierrez J, Salmerón C, et al. An in vivo and in vitro assessment of autophagy-related gene expression in muscle of rainbow trout (Oncorhynchus mykiss). Comp Biochem Physiol Part B Biochem Mol Biol. 2010;157(3):258–266.
  • Giorgetti E, Yu Z, Chua Jason P, et al. Rescue of metabolic alterations in AR113Q skeletal muscle by peripheral androgen receptor gene silencing. Cell Rep. 2016;17(1):125–136.
  • Marino JS, Tausch BJ, Dearth CL, et al. β2-Integrins contribute to skeletal muscle hypertrophy in mice. Am J Physiol Cell Physiol. 2008;295(4):C1026–C1036.
  • Mourikis P, Tajbakhsh S. Distinct contextual roles for Notch signalling in skeletal muscle stem cells. BMC Dev Biol. 2014;14(1):2.
  • Bae G-U, Yang Y-J, Jiang G, et al. Neogenin regulates skeletal myofiber size and focal adhesion kinase and extracellular signal-regulated kinase activities in vivo and in vitro. Mol Biol Cell. 2009;20(23):4920–4931.
  • Tran P, Ho SM, Kim BG, et al. TGF-β-activated kinase 1 (TAK1) and apoptosis signal-regulating kinase 1 (ASK1) interact with the promyogenic receptor Cdo to promote myogenic differentiation via activation of p38MAPK pathway. J Biol Chem. 2012;287(15):11602–11615.
  • Rangwala SM, Wang X, Calvo JA, et al. Estrogen-related receptor gamma is a key regulator of muscle mitochondrial activity and oxidative capacity. J Biol Chem. 2010;285(29):22619–226129.
  • Li Z, Gilbert JA, Zhang Y, et al. An HMGA2-IGF2BP2 axis regulates myoblast proliferation and myogenesis. Dev Cell. 2012;23(6):1176–1188.
  • Huang H, Weng H, Sun W, et al. Recognition of RNA N 6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–295.
  • Okada C, Yamashita E, Lee SJ, et al. A high-resolution structure of the pre-microRNA nuclear export machinery. Science. 2009;326(5957):1275–1279.
  • Lee GE, Kim JH, Taylor M, et al. DNA methyltransferase 1-associated protein (DMAP1) is a co-repressor that stimulates DNA methylation globally and locally at sites of double strand break repair. J Biol Chem. 2010;285(48):37630–37640.
  • Kirsh O, Seeler J-S, Pichler A, et al. The SUMO E3 ligase RanBP2 promotes modification of the HDAC4 deacetylase. Embo J. 2002;21(11):2682–2691.
  • Sanchez R, Zhou -M-M. The PHD finger: a versatile epigenome reader. Trends Biochem Sci. 2011;36(7):364–372.
  • Jallow Z, Jacobi UG, Weeks DL, et al. Specialized and redundant roles of TBP and a vertebrate-specific TBP paralog in embryonic gene regulation in Xenopus. Proc Natl Acad Sci USA. 2004;101(37):13525–13530.
  • Liebermann DA, Tront JS, Sha X, et al. Gadd45 stress sensors in malignancy and leukemia. Crit Rev Oncog. 2011;16(1–2):129–140.
  • Hays TT, Patsalos A, Hammers D, et al. A novel role of growth differentiation factor 3 for the regulation of muscle metabolism. Diabetes. 2018;67:1.
  • Ahmetov II, Rogozkin VA. The role of PGC-1α in the regulation of skeletal muscle metabolism. Human Physiol. 2013;39(4):441–449.
  • Redfield A, Nieman MT, Knudsen KA. Cadherins Promote Skeletal Muscle Differentiation in Three-dimensional Cultures. J Cell Biol. 1997;138(6):1323.
  • Yin F, Herring BP. GATA-6 can act as a positive or negative regulator of smooth muscle-specific gene expression. J Biol Chem. 2005;280(6):4745–4752.
  • Chen X, Wang Z, Tang S, et al. Genome-wide mapping of DNA methylation in Nile tilapia. Hydrobiologia. 2016;791(1):1–11.
  • Chatterjee A, Stockwell PA, Horsfield JA, et al. Base-resolution DNA methylation landscape of zebrafish brain and liver. Genom Data. 2014;2:342–344.
  • Martinez-Arguelles DB, Lee S, Papadopoulos V. In silico analysis identifies novel restriction enzyme combinations that expand reduced representation bisulfite sequencing CpG coverage. BMC Res Notes. 2014;7(1):534.
  • Jones PA. The DNA methylation paradox. Trends Genet. 1999;15(1):34–37.
  • Brenet F, Moh M, Funk P, et al. DNA methylation of the first exon is tightly linked to transcriptional silencing. PLoS One. 2011;6(1):e14524.
  • Shukla S, Kavak E, Gregory M, et al. CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature. 2011;479(7371):74–79.
  • Maunakea AK, Nagarajan RP, Bilenky M, et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 2010;466(7303):253–257.
  • Engreitz JM, Haines JE, Perez EM, et al. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature. 2016;539(7629):452–455.
  • Chen J, Fan Z, Tan D, et al. A review of genetic advances related to sex control and manipulation in tilapia. J World Aquacult Soc. 2018;49(2):277–291.
  • Schreiber S, Focken U, Becker K. Individually reared female Nile tilapia (Oreochrornis niloticus) can grow faster than males. J Appl Ichthyol. 2007;14(1–2):43–47.
  • Yanginlar C, Logie C. HDAC11 is a regulator of diverse immune functions. BBA-Gene Regul Mech. 2018;1861(1):54–59.
  • Hirose K, Ito T, Fukawa K, et al. Evaluation of effects of multiple candidate genes (lep, lepr, mc4r, pik3c3, and vrtn) on production traits in Duroc pigs. Anim Sci J. 2014;85(3):198–206.
  • Hao Y, Cui Y, Gu X. Genome-wide DNA methylation profiles changes associated with constant heat stress in pigs as measured by bisulfite sequencing. Sci Rep. 2016;6:27507.
  • Guerra B, Fuentes T, Delgado-Guerra S, et al. Gender dimorphism in skeletal muscle leptin receptors, serum leptin and insulin sensitivity. PLoS One. 2008;3(10):e3466.
  • Meadows E, Cho J-H, Flynn JM, et al. Myogenin regulates a distinct genetic program in adult muscle stem cells. Dev Biol. 2008;322(2):406–414.
  • Xiong X, Tao R, DePinho RA, et al. The autophagy-related gene 14 (atg14) is regulated by forkhead box O transcription factors and circadian rhythms and plays a critical role in hepatic autophagy and lipid metabolism. J Biol Chem. 2012;287(46):39107–39114.
  • Ruszkowska A, Ruszkowski M, Dauter Z, et al. Structural insights into the RNA methyltransferase domain of METTL16. Sci Rep. 2018;8(1):5311.
  • Kudou K, Komatsu T, Nogami J, et al. The requirement of Mettl3-promoted MyoD mRNA maintenance in proliferative myoblasts for skeletal muscle differentiation. Open Biol. 2017;7(9):170119.
  • Printen JA, Brady MJ, Saltiel AR. PTG, a protein phosphatase 1-binding protein with a role in glycogen metabolism. Science. 1997;275(5305):1475–1478.
  • Rönn T, Volkov P, Davegårdh C, et al. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS Genet. 2013;9(6):e1003572.
  • Huang CW, Li YH, Hu SY, et al. Differential expression patterns of growth-related microRNAs in the skeletal muscle of Nile tilapia (Oreochromis niloticus). J Anim Sci. 2012;90(12):4266–4279.
  • Pinhal D, Bovolenta LA, Moxon S, et al. Genome-wide microRNA screening in Nile tilapia reveals pervasive isomiRs’ transcription, sex-biased arm switching and increasing complexity of expression throughout development. Sci Rep. 2018;8(1):8248.
  • Keller IS, Bayer T, Salzburger W, et al. Effects of parental care on resource allocation into immune defense and buccal microbiota in mouthbrooding cichlid fishes. Evolution. 2018;72(5):1109–1123.
  • Van Wassenbergh S, Potes NZ, Adriaens D. Hydrodynamic drag constrains head enlargement for mouthbrooding in cichlids. J Royal Soc Interface. 2015;12:109.
  • O’Connor CM, Reardon EE, Chapman LJ. Shorter gills in mouth-brooding females of the cichlid Pseudocrenilabrus multicolor. Copeia. 2012;2012(3):382–388.
  • Corrie LW-C, Chapman LJ, Reardon EE. Brood protection at a cost: mouth brooding under hypoxia in an African cichlid. Environ Biol Fishes. 2008;82(1):41–49.
  • Tremblay A, Simoneau JA, Bouchard C. Impact of exercise intensity on body fatness and skeletal muscle metabolism. Metabolism. 1994;43(7):814–818.
  • Niehrs C, Schäfer A. Active DNA demethylation by Gadd45 and DNA repair. Trends Cell Biol. 2012;22(4):220–227.
  • Ishida K, Yuge Y, Hanaoka M, et al. Gadd45g regulates dental epithelial cell proliferation through p38 MAPK-mediated p21 expression. Genes Cells. 2013;18(8):660–671.
  • Shen Y, Ma K, Liu F, et al. Characterization of two novel gadd45a genes in hybrid tilapia and their responses to the infection of Streptococcus agalactiae. Fish Shellfish Immunol. 2016;54:276–281.
  • Ying J, Srivastava G, Hsieh WS, et al. The stress-responsive gene gadd45g is a functional tumor suppressor, with its response to environmental stresses frequently disrupted epigenetically in multiple tumors. Clin Cancer Res. 2005;11(18):6442–6449.
  • Tamura RE, de Vasconcellos JF, Sarkar D, et al. GADD45 proteins: central players in tumorigenesis. Curr Mol Med. 2012;12(5):634–651.
  • Ebert SM, Dyle MC, Kunkel SD, et al. Stress-induced skeletal muscle Gadd45a expression reprograms myonuclei and causes muscle atrophy. J Biol Chem. 2012;287(33):27290–27301.
  • Bruemmer D, Yin F, Liu J, et al. Regulation of the growth arrest and DNA damage-inducible gene 45 (gadd45) by peroxisome proliferator-activated receptor gamma in vascular smooth muscle cells. Circ Res. 2003;93(4):e38–47.
  • Varga T, Mounier R, Patsalos A, et al. Macrophage PPARγ, a lipid activated transcription factor controls the growth factor GDF3 and skeletal muscle regeneration. Immunity. 2016;45(5):1038–1051.
  • Kelstrup L, Hjort L, Houshmand-Oeregaard A, et al. Gene expression and DNA methylation of PPARGC1A in muscle and adipose tissue from adult offspring of women with diabetes in pregnancy. Diabetes. 2016;65(10):2900–2910.
  • Benton CR, Wright DC, Bonen A. PGC-1alpha-mediated regulation of gene expression and metabolism: implications for nutrition and exercise prescriptions. Appl Physiol Nutr Metab. 2008;33(5):682–843.
  • Handschin C, Spiegelman BM. Peroxisome proliferator-activated receptor gamma coactivator 1 coactivators, energy homeostasis, and metabolism. Endocr Rev. 2006;27(7):728–735.
  • Barres R, Osler ME, Yan J, et al. Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density. Cell Metab. 2009;10(3):189–198.
  • Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnetjournal. 2011;17(1):10–12.
  • Krueger F, Andrews SR. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics. 2011;27(11):1571–1572.
  • Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9. DOI:10.1038/nmeth.1923
  • Akalin A, Kormaksson M, Li S, et al. methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles. Genome Biol. 2012;13(10):R87–R87.
  • Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14(2):178–192.
  • Krzywinski M, Schein J, Birol İ, et al. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19(9):1639–1645.
  • Heinz S, Benner C, Spann N, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell. 2010;38(4):576–589.
  • Rebhan M, Chalifa-Caspi V, Prilusky J, et al. GeneCards: integrating information about genes, proteins and diseases. Trends Genet. 1997;13(4):163.
  • Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57.