Resveratrol stimulates microRNA expression during differentiation of bovine primary myoblasts

  • Dan Hao Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China; and Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
  • Xiao Wang Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens Lyngby, Denmark
  • Xiaogang Wang Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
  • Bo Thomsen Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
  • Kaixing Qu Yunnan Academy of Grassland and Animal Science, Kunming, Yunnan, China
  • Xianyong Lan Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
  • Yongzhen Huang Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
  • Chuzhao Lei Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
  • Bizhi Huang Yunnan Academy of Grassland and Animal Science, Kunming, Yunnan, China
  • Hong Chen Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
Keywords: Biomarker; Cattle skeletal cell; MicroRNA; Polyphenol resveratrol treatment

Abstract

Background: Resveratrol (RSV), a phenolic compound, is present in many human dietary sources, such as peanuts, peanut butter, grapes skin, and grape wine. RSV has been widely known for its benefits on human health. Beef from cattle skeletal muscle is one of the main sources of protein for human consumption. Previous studies have also found that pork and chicken qualities are influenced by the feed supplementation with RSV. In addition, our previous study demonstrated the RSV effects on bovine myoblast differentiation using messenger RNA (mRNA) data. In this study, we mainly focused on the influences of RSV on microRNA (miRNA) expression.

Method: We used 20 μM RSV to treat primary bovine myoblasts and extracted RNA for miRNA sequencing. After quality control and alignment for clean reads, we conducted quantification and analysis of differentially expressed (DE) miRNAs in the case (RSV-treated) group versus control (non-RSV treated) group. Next, we predicted the target genes for the DE miRNAs and analyzed them for the enrichments of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways.

Results: Finally, we identified 93 DE miRNAs (adjusted P-value < 0.05), of them 44 were upregulated and 49 were downregulated. Bta-miR-34c was the most significantly upregulated miRNA. In silico, prediction results indicated 1,869 target genes for the 93 DE miRNAs. GO enrichment analysis for the genes targeted by DE miRNAs revealed two significant GO terms (adjusted P-value < 0.05), in which the most significant one was stereocilium (GO:0032420). KEGG enrichment analysis showed five significant pathways, and the top significant KEGG pathway was the insulin signaling pathway (bta04910) (adjusted P-value < 0.05).

Conclusions: This study provided an improved understanding of effects of RSV on primary bovine myoblast differentiation through the miRNA modulations. The results suggested that RSV could promote differentiation of primary bovine myoblast by stimulating the miRNA expressions. The target genes of DE miRNAs were significantly enriched in the insulin signaling pathway, thus potentially contributing to improving muscle leanness by increasing the energy metabolism.

Downloads

Download data is not yet available.

References


  1. Ambros V. The functions of animal microRNAs. Nature 2004; 431: 350–5. doi: 10.1038/nature02871

  2. Izzotti A, Cartiglia C, Steele VE, De Flora S. MicroRNAs as targets for dietary and pharmacological inhibitors of mutagenesis and carcinogenesis. Mutat Res Rev Mutat Res 2012; 751(2): 287–303. doi: 10.1016/j.mrrev.2012.05.004

  3. Kumar A, Butt NA, Levenson AS. Natural epigenetic-modifying molecules in medical therapy. In: Tollefsbol TO, ed. Medical epigenetics. Birmingham, AL: Mica Haley; 2016, pp. 747–98. doi: 10.1016/B978-0-12-803239-8.00039-9

  4. Raza SHA, Kaster N, Khan R, Abdelnour SA, El-hack MEA, Khafaga AF, et al. The Rple of microRNAs in muscle tissue development in beef cattle. Genes 2020; 11: 295. doi: 10.3390/genes11030295

  5. Venkatadri R, Muni T, Iyer AKV, Yakisich JS, Azad N. Role of apoptosis-related miRNAs in resveratrol-induced breast cancer cell death. Cell Death Dis 2016; 7: e2104. doi: 10.1038/cddis.2016.6

  6. Fagone E, Conte E, Gili E, Fruciano M, Pistorio MP, Lo Furno D, et al. Resveratrol inhibits transforming growth factor-β-induced proliferation and differentiation of ex vivo human lung fibroblasts into myofibroblasts through ERK/Akt inhibition and PTEN restoration. Exp Lung Res 2011; 37(3): 162–74. doi: 10.3109/01902148.2010.524722

  7. Leong CW, Wong CH, Lao SC, Leong EC, Lao IF, Law PTW, et al. Effect of resveratrol on proliferation and differentiation of embryonic cardiomyoblasts. Biochem Biophys Res Commun 2007; 360(1): 173–80. doi: 10.1016/j.bbrc.2007.06.025

  8. Meng Q, Sun S, Bai Y, Luo Z, Li Z, Shi B, et al. Effects of dietary resveratrol supplementation in sows on antioxidative status, myofiber characteristic and meat quality of offspring. Meat Sci 2020; 167: 108176. doi: 10.1016/j.meatsci.2020.108176

  9. Zhang C, Luo J, Yu B, Zheng P, Huang Z, Mao X, et al. Dietary resveratrol supplementation improves meat quality of finishing pigs through changing muscle fiber characteristics and antioxidative status. Meat Sci 2015; 102: 15–21. doi: 10.1016/j.meatsci.2014.11.014

  10. Kaminski J, Lançon A, Aires V, Limagne E, Tili E, Michaille JJ, et al. Resveratrol initiates differentiation of mouse skeletal muscle-derived C2C12 myoblasts. Biochem Pharmacol 2012; 84(10): 1251–9. doi: 10.1016/j.bcp.2012.08.023

  11. Joseph AM, Malamo AG, Silvestre J, Wawrzyniak N, Carey-Love S, Nguyen LMD, et al. Short-term caloric restriction, resveratrol, or combined treatment regimens initiated in late-life alter mitochondrial protein expression profiles in a fiber-type specific manner in aged animals. Exp Gerontol 2013; 48(9): 858–68. doi: 10.1016/j.exger.2013.05.061

  12. Miyake M, Takahashi H, Kitagawa E. AMPK activation by AICAR inhibits myogenic differentiation and myostatin expression in Cattle. Cell Tissue Res 2012; 349(2): 615–23. doi: 10.1007/s00441-012-1422-8

  13. Hao D, Wang X, Wang X, Thomsen B, Kadarmideen HN, Lan X, et al. Transcriptomic changes in bovine skeletal muscle cells after resveratrol treatment. Gene 2020; 754: 144849. doi: 10.1016/j.gene.2020.144849

  14. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 2009; 10: R25. doi: 10.1186/gb-2009-10-3-r25

  15. Friedländer MR, MacKowiak SD, Li N, Chen W, Rajewsky N. MiRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades. Nucl Acids Res 2012; 40(1): 37–52. doi: 10.1093/nar/gkr688

  16. Zhou L, Chen J, Li Z, Li X, Hu X, Huang Y, et al. Integrated profiling of microRNAs and mRNAs: microRNAs located on Xq27.3 associate with clear cell renal cell carcinoma. PLoS One 2010; 5(12): e15224. doi: 10.1371/journal.pone.0015224

  17. Anders S, Huber W. Differential expression analysis for sequence count data. Genome Biol 2010; 11: R106. doi: 10.1186/gb-2010-11-10-r106

  18. Enright AJ, John B, Gaul U, Tuschl T, Sander C, Marks DS. MicroRNA targets in drosophila. Genome Biol 2003; 5(1): R1. doi: 10.1186/gb-2003-5-1-r1

  19. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 2012; 16(5): 284–7. doi: 10.1089/omi.2011.0118

  20. Lançon A, Kaminski J, Tili E, Michaille JJ, Latruffe N. Control of microRNA expression as a new way for resveratrol to deliver its beneficial effects. J Agricult Food Chem 2012; 60(36): 8783–9. doi: 10.1021/jf301479v

  21. Bosutti A, Degens H. The impact of resveratrol and hydrogen peroxide on muscle cell plasticity shows a dose-dependent interaction. Sci Rep 2015; 5: 8093. doi: 10.1038/srep08093

  22. Latruffe N, Lançon A, Frazzi R, Aires V, Delmas D, Michaille JJ, et al. Exploring new ways of regulation by resveratrol involving miRNAs, with emphasis on inflammation. Ann N Y Acad Sci 2015; 1348(1): 97–106. doi: 10.1111/nyas.12819

  23. Kaminski J, Lançon A, Tili E, Aires V, Demarquoy J, Lizard G, et al. Dietary resveratrol modulates metabolic functions in skeletal muscle cells. J Food Drug Anal 2012; 20: 398–401. doi: 10.38212/2224-6614.2098

  24. Bartoň L, Bureš D, Řehák D, Kott T, Makovický P. Tissue-specific fatty acid composition, cellularity, and gene expression in diverse cattle breeds. Animal 2021; 15(1): 100025. doi: 10.1016/j.animal.2020.100025

  25. Park SJ, Beak SH, Jung DJS, Kim SY, Jeong IH, Piao MY, et al. Genetic, management, and nutritional factors affecting intramuscular fat deposition in beef cattle – a review. Asian-Australas J Anim Sci 2018; 31(7): 1043–61. doi: 10.5713/ajas.18.0310

  26. Roudbari Z, Coort SL, Kutmon M, Eijssen L, Melius J, Sadkowski T, et al. Identification of biological pathways contributing to marbling in skeletal muscle to improve beef cattle breeding. Front Genet 2020; 10: 1370. doi: 10.3109/10799893.2015.1030412

  27. Mei C, Wang H, Liao Q, Khan R, Raza SHA, Zhao C, et al. Genome-wide analysis reveals the effects of artificial selection on production and meat quality traits in Qinchuan cattle. Genomics 2019; 111(6): 1201–8. doi: 10.1016/j.ygeno.2018.09.021

  28. Botti SCCF, Santos AO, Dias I, Degasperi FT, Irazusta SP. Extraction, characterization and quantification of resveratrol from the inemaking pomace: emphasis on the vacuum drying process. Rev Brasil Aplicações Vácuo 2018; 37: 79–86. doi: 10.17563/rbav.v37i2.1095

  29. Zhao JX, Li Q, Zhang RX, Liu WZ, Ren YS, Zhang CX, et al. Effect of dietary grape pomace on growth performance, meat quality and antioxidant activity in ram lambs. Animal Feed Sci Technol 2018; 236: 76–85. doi: 10.1016/j.anifeedsci.2017.12.004

  30. Zhang C, Zhao X, Wang L, Yang L, Chen X, Geng Z. Resveratrol beneficially affects meat quality of heat-stressed broilers which is associated with changes in muscle antioxidant status. Animal Sci J 2017; 88(10): 1569–74. doi: 10.1111/asj.12812

  31. Zhang C, Wang L, Zhao XH, Chen XY, Yang L, Geng ZY. Dietary resveratrol supplementation prevents transport-stress-impaired meat quality of broilers through maintaining muscle energy metabolism and antioxidant status. Poultry Sci 2017; 96(7): 2219–25. doi: 10.3382/ps/pex004

  32. Cheng K, Yu C, Li Z, Li S, Yan E, Song Z, et al. Resveratrol improves meat quality, muscular antioxidant capacity, lipid metabolism and fiber type composition of intrauterine growth retarded pigs. Meat Sci 2020; 170: 108237. doi: 10.1016/j.meatsci.2020.108237

  33. Bekhit AED, Geesink GH, Ilian MA, Morton JD, Bickerstaffe R. The effects of natural antioxidants on oxidative processes and metmyoglobin reducing activity in beef patties. Food Chem 2003; 81(2): 175–87. doi: 10.1016/S0308-8146(02)00410-7

  34. Alway SE, McCrory JL, Kearcher K, Vickers A, Frear B, Gilleland DL, et al. Resveratrol enhances exercise-induced cellular and functional adaptations of skeletal muscle in older men and women. J Gerontol A Biol Sci Med Sci 2017; 72(12): 1595–606. doi: 10.1093/gerona/glx089

  35. Sun J, Li M, Li Z, Xue J, Lan X, Zhang C, et al. Identification and profiling of conserved and novel microRNAs from Chinese Qinchuan bovine longissimus thoracis. BMC Genomics 2013; 14: 42. doi: 10.1186/1471-2164-14-42

  36. Muroya S, Taniguchi M, Shibata M, Oe M, Ojima K, Nakajima I, et al. Profiling of differentially expressed microRNA and the bioinformatic target gene analyses in bovine fast- and slow-type muscles by massively parallel sequencing. J Animal Sci 2013; 91(1): 90–103. doi: 10.2527/jas.2012-5371

  37. Van Der Deen M, Taipaleenmäki H, Zhang Y, Teplyuk NM, Gupta A, Cinghu S, et al. MicroRNA-34c inversely couples the biological functions of the runt-related transcription factor RUNX2 and the tumor suppressor p53 in osteosarcoma. J Biol Chem 2013; 288: 21307–19. doi: 10.1074/jbc.M112.445890

  38. Wei J, Shi Y, Zheng L, Zhou B, Inose H, Wang J, et al. miR-34s inhibit osteoblast proliferation and differentiation in the mouse by targeting SATB2. J Cell Biol 2012; 197(4): 509–21. doi: 10.1083/jcb.201201057

  39. Wang M, Liu C, Su Y, Zhang K, Zhang Y, Chen M, et al. miRNA-34c inhibits myoblasts proliferation by targeting YY1. Cell Cycle 2017; 16(18): 1661–72. doi: 10.1080/15384101.2017.1281479

  40. Hou L, Xu J, Li H, Ou J, Jiao Y, Hu C, et al. MIR-34c represses muscle development by forming a regulatory loop with Notch1. Sci Rep 2017; 7: 9346. doi: 10.1038/s41598-017-09688-y

  41. Hayes J, Peruzzi PP, Lawler S. MicroRNAs in cancer: biomarkers, functions and therapy. Trends Mol Med 2014; 20(8): 460–9. doi: 10.1016/j.molmed.2014.06.005

  42. Adhami M, Haghdoost AA, Sadeghi B, Malekpour Afshar R. Candidate miRNAs in human breast cancer biomarkers: a systematic review. Breast Cancer 2018; 25: 198–205. doi: 10.1007/s12282-017-0814-8

  43. Pockar S, Globocnik Petrovic M, Peterlin B, Vidovic Valentincic N. MiRNA as biomarker for uveitis – a systematic review of the literature. Gene 2019; 696: 162–75. doi: 10.1016/j.gene.2019.02.004

  44. Aryani A, Denecke B. In vitro application of ribonucleases: comparison of the effects on mRNA and miRNA stability. BMC Res Notes 2015; 8(164): 1–9. doi: 10.1186/s13104-015-1114-z

  45. Tavallaie R, De Almeida SRM, Gooding JJ. Toward biosensors for the detection of circulating microRNA as a cancer biomarker: an overview of the challenges and successes. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2015; 7(4): 580–29. doi: 10.1002/wnan.1324

  46. Kitzmann M, Fernandez A. Crosstalk between cell cycle regulators and the myogenic factor MyoD in skeletal myoblasts. Cell Mol Life Sci 2001; 58(4): 571–9. doi: 10.1007/PL00000882

  47. Luo W, Nie Q, Zhang X. MicroRNAs involved in skeletal muscle differentiation. J Genet Genomics 2013; 40(3): 107–16. doi: 10.1016/j.jgg.2013.02.002

  48. Chakraborty C, Doss CGP, Bandyopadhyay S, Agoramoorthy G. Influence of miRNA in insulin signaling pathway and insulin resistance: micro-molecules with a major role in type-2 diabetes. Wiley Interdiscip Rev RNA 2014; 5(5): 697–712. doi: 10.1002/wrna.1240

  49. Lin X, Smagghe G. Roles of the insulin signaling pathway in insect development and organ growth. Peptides 2019; 122: 169923. doi: 10.1016/j.peptides.2018.02.001

  50. Huang J, Feng X, Zhu R, Guo D, Wei Y, Cao X, et al. Comparative transcriptome analysis reveals that PCK1 is a potential gene affecting IMF deposition in buffalo. BMC Genomics 2020; 21: 710. doi: 10.1186/s12864-020-07120-w

  51. Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res 2015; 35(6): 600–4. doi: 10.3109/10799893.2015.1030412

  52. Byun HR, Kim DK, Koh JY. Obesity and downregulated hypothalamic leptin receptors in male metallothionein-3-null mice. Neurobiol Dis 2011; 44(1): 125–32. doi: 10.1016/j.nbd.2011.06.012

  53. Sun Y, Wang P, Zheng H, Smith RG. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proceed Natl Acad Sci U S A 2004; 101(13): 4679–84. doi: 10.1073/pnas.0305930101

  54. Pan WW, Myers MG. Leptin and the maintenance of elevated body weight. Nat Rev Neurosci 2018; 19: 95–105. doi: 10.1038/nrn.2017.168

  55. Vislovukh A, Kratassiouk G, Porto E, Gralievska N, Beldiman C, Pinna G, et al. Proto-oncogenic isoform A2 of eukaryotic translation elongation factor eEF1 is a target of miR-663 and miR-744. Br J Cancer 2013; 108: 2304–11. doi: 10.1038/bjc.2013.243

  56. Farooqi AA, Khalid S, Ahmad A. Regulation of cell signaling pathways and miRNAs by resveratrol in different cancers. Int J Mol Sci 2018; 19(3): 652. doi: 10.3390/ijms19030652

  57. Holton TA, Vijayakumar V, Khaldi N. Bioinformatics: current perspectives and future directions for food and nutritional research facilitated by a food-wiki database. Trends Food Sci Technol 2013; 80: 342–7. doi: 10.1016/j.tifs.2013.08.009

  58. Hocquette JF, Lehnert S, Barendse W, Cassar-Malek I, Picard B. Recent advances in cattle functional genomics and their application to beef quality. Animal 2007; 1(1): 159–73. doi: 10.1017/S1751731107658042

  59. Chen Z, Chu S, Xu X, Jiang J, Wang W, Shen H, et al. Analysis of longissimus muscle quality characteristics and associations with DNA methylation status in cattle. Genes Genomics 2019; 41: 1147–63. doi: 10.1007/s13258-019-00844-4

  60. Magee DA, Spillane C, Berkowicz EW, Sikora KM, Machugh DE. Imprinted loci in domestic livestock species as epigenomic targets for artificial selection of complex traits. Animal Genet 2014; 45(Suppl 1): 25–39. doi: 10.1111/age.12168

  61. Wang X, Kadarmideen HN. Metabolomics analyses in high-low feed efficient dairy cows reveal novel biochemical mechanisms and predictive biomarkers. Metabolites 2019; 9(7): 151. doi: 10.3390/metabo9070151

  62. Novais FJ, Pires PRL, Alexandre PA, Dromms RA, Iglesias AH, Ferraz JBS, et al. Identification of a metabolomic signature associated with feed efficiency in beef cattle. BMC Genomics 2019; 20(8): 1–10. doi: 10.1186/s12864-018-5406-2

  63. Desdouits M, de Graaf M, Strubbia S, Oude Munnink BB, Kroneman A, Le Guyader FS, et al. Novel opportunities for NGS-based one health surveillance of foodborne viruses. One Health Outlook 2020; 2(14): 1–8. doi: 10.1186/s42522-020-00015-6

  64. Zhang D, Cheng X, Sun D, Ding S, Cai P, Yuan L, et al. AdditiveChem: a comprehensive bioinformatics knowledge-base for food additive chemicals. Food Chem 2020; 308: 125519. doi: 10.1016/j.foodchem.2019.125519

Published
2021-03-18
How to Cite
Hao D., Wang X., Wang X., Thomsen B., Qu K., Lan X., Huang Y., Lei C., Huang B., & Chen H. (2021). Resveratrol stimulates microRNA expression during differentiation of bovine primary myoblasts. Food & Nutrition Research, 65. https://doi.org/10.29219/fnr.v65.5453
Section
Original Articles