Browse > Article
http://dx.doi.org/10.5713/ajas.18.0807

Integrated mRNA and miRNA profile expression in livers of Jinhua and Landrace pigs  

Huang, Minjie (Department of Animal Genetics and Breeding, College of Animal Science, Zhejiang University)
Chen, Lixing (Department of Animal Genetics and Breeding, College of Animal Science, Zhejiang University)
Shen, Yifei (Department of Animal Genetics and Breeding, College of Animal Science, Zhejiang University)
Chen, Jiucheng (Department of Animal Genetics and Breeding, College of Animal Science, Zhejiang University)
Guo, Xiaoling (Department of Animal Genetics and Breeding, College of Animal Science, Zhejiang University)
Xu, Ningying (Department of Animal Genetics and Breeding, College of Animal Science, Zhejiang University)
Publication Information
Asian-Australasian Journal of Animal Sciences / v.32, no.10, 2019 , pp. 1483-1490 More about this Journal
Abstract
Objective: To explore the molecular mechanisms of fat metabolism and deposition in pigs, an experiment was conducted to identify hepatic mRNAs and miRNAs expression and determine the potential interaction of them in two phenotypically extreme pig breeds. Methods: mRNA and miRNA profiling of liver from 70-day Jinhua (JH) and Landrace (LD) pigs were performed using RNA sequencing. Blood samples were taken to detect results of serum biochemistry. Bioinformatics analysis were applied to construct differentially expressed miRNA-mRNA network. Results: Serum total triiodothyronine and total thyroxine were significantly lower in Jinhua pigs, but the content of serum total cholesterol (TCH) and low-density lipoprotein cholesterol were strikingly higher. A total of 467 differentially expressed genes (DEGs) and 35 differentially expressed miRNAs (DE miRNAs) were identified between JH and LD groups. Gene ontology analysis suggested that DEGs were involved in oxidation-reduction, lipid biosynthetic and lipid metabolism process. Interaction network of DEGs and DE miRNAs were constructed, according to target prediction results. Conclusion: We generated transcriptome and miRNAome profiles of liver from JH and LD pig breeds which represent distinguishing phenotypes of growth and metabolism. The potential miRNA-mRNA interaction networks may provide a comprehensive understanding in the mechanism of lipid metabolism. These results serve as a basis for further investigation on biological functions of miRNAs in the porcine liver.
Keywords
mRNA; miRNA; Liver; Pig; RNA-seq;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Zappaterra M, Deserti M, Mazza R, Braglia S, Zambonelli P, Davoli R. A gene and protein expression study on four porcine genes related to intramuscular fat deposition. Meat Sci 2016;121:27-32. https://doi.org/10.1016/j.meatsci.2016.05.007   DOI
2 Yongdong P, Shulong Y, Huanan L, Hong X, Jian P, Siwen J. MicroRNAs: emerging roles in adipogenesis and obesity. Cell Signal 2014;26:1888-96. https://doi.org/10.1016/j.cellsig.2014.05.006   DOI
3 Miller ER, Ullrey DE. The pig as a model for human nutrition. Annu Rev Nutr 1987;7:361-82. https://doi.org/10.1146/annurev. nu.07.070187.002045   DOI
4 Ting W, Zhenhai Z, Zhangqin Y, et al. Distinctive genes determine different intramuscular fat and muscle fiber ratios of the longissimus dorsi muscles in Jinhua and landrace pigs. PLoS One 2013;8:e53181. https://doi.org/10.1371/journal.pone.0053181   DOI
5 Chen P, Baas TJ, Mabry JW, Koehler KJ. Genetic correlations between lean growth and litter traits in U.S. Yorkshire, Duroc, Hampshire, and Landrace pigs. J Anim Sci 2003;81:1700-5. https://doi.org/10.2527/2003.8171700x   DOI
6 Mckenna LB, Schug J, Vourekas A, et al. MicroRNAs control intestinal epithelial differentiation, architecture, and barrier function. Gastroenterology 2010;139:1654-64. https://doi.org/10.1053/j.gastro.2010.07.040   DOI
7 Meng SX, Yi-Yang HU, Feng Q. Application of microRNA in prevention and treatment of nonalcoholic fatty liver disease with traditional Chinese medicine. Chin J Tradit Chin Med Pharm 2014;29:806-9.
8 Christine E, Scott D, Murray SF, et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab 2006;3:87-98. https://doi.org/10.1016/j.cmet.2006.01.005   DOI
9 Sharda T, Kalpana G. miR-122 is a unique molecule with great potential in diagnosis, prognosis of liver disease, and therapy both as miRNA mimic and antimir. Curr Gene Ther 2015;15:142-50. https://doi.org/10.2174/1566523214666141224095610   DOI
10 Peng Y, Xiang H, Chen C, et al. MiR-224 impairs adipocyte early differentiation and regulates fatty acid metabolism. Int J Biochem Cell B 2013;45:1585-93. https://doi.org/10.1016/j.biocel.2013.04.029   DOI
11 Eun Kyung L, Jeong LM, Kotb A, et al. miR-130 suppresses adipogenesis by inhibiting peroxisome proliferator-activated receptor gamma expression. Mol Cell Biol 2011;31:626-38. https://doi.org/10.1128/MCB.00894-10   DOI
12 Qun L, Zhanguo G, Alarcon RM, Jianping Y, Zhong Y. A role of miR-27 in the regulation of adipogenesis. FEBS J 2009;276:2348-58.   DOI
13 Spurlock ME, Gabler NK. The development of porcine models of obesity and the metabolic syndrome. J Nutr 2008;138:397-402. https://doi.org/10.1093/jn/138.2.397   DOI
14 Shen Y, Mao H, Huang M, et al. Long noncoding RNA and mRNA expression profiles in the thyroid gland of two phenotypically extreme pig breeds using ribo-zero RNA sequencing. Genes 2016;7:34. https://doi.org/10.3390/genes7070034   DOI
15 Xuguang Z, Sheue-Yann C. New insights into regulation of lipid metabolism by thyroid hormone. Curr Opin Endocrinol Diabetes Obes 2010;17:408-13. https://doi.org/10.1097/MED.0b013e32833d6d46   DOI
16 Etherton TD, Louveau I, SoRensen MT, Chaudhuri S. Mechanisms by which somatotropin decreases adipose tissue growth. Am J Clin Nutr 1993;58(Suppl 2):287S-95S.   DOI
17 Uygun A, Kadayifci A, Yesilova Z, et al. Serum leptin levels in patients with nonalcoholic steatohepatitis. Am J Gastroenterol 2000;95:3584-9. https://doi.org/10.1111/j.1572-0241.2000.03297.x   DOI
18 Chegary M, Ht B, Ruiter JP, et al. Mitochondrial long chain fatty acid $\beta$-oxidation in man and mouse. Biochim Biophys Acta 2009;1791:806-15. https://doi.org/10.1016/j.bbalip.2009.05.006   DOI
19 Newberry EP, Xie Y, Kennedy SM, Luo J, Davidson NO. Protection against Western diet-induced obesity and hepatic steatosis in liver fatty acid-binding protein knockout mice. Hepatology 2006;44:1191-205. https://doi.org/10.1002/hep.21369   DOI
20 Wong WM, Gerry AB, Putt W, et al. Common variants of apolipoprotein A-IV differ in their ability to inhibit low density lipoprotein oxidation. Atherosclerosis 2007;192:266-74. https://doi.org/10.1016/j.atherosclerosis.2006.07.017   DOI
21 Rafael K, Moeller MJ. The next level of complexity: post-transcriptional regulation by microRNAs. Kidney Int 2011;80:692-3. https://doi.org/10.1038/ki.2011.175   DOI
22 Pan S, Zheng Y, Zhao R, Yang X. MicroRNA-130b and microRNA-374b mediate the effect of maternal dietary protein on offspring lipid metabolism in meishan pigs. Br J Nutr 2013;109:1731-8. https://doi.org/10.1017/S0007114512003728   DOI
23 Dimitrios I, Konstantinos D, Yaeko H, Goldberg IJ, Zannis VI. MicroRNA-370 controls the expression of microRNA-122 and Cpt1alpha and affects lipid metabolism. J Lipid Res 2010;51:1513-23. https://doi.org 10.1194/jlr.M004812   DOI
24 Jing-Xia L, Dawei Z, Xunwei X, et al. Eaf1 and Eaf2 negatively regulate canonical Wnt/$\beta$-catenin signaling. Development 2013;140:1067-78. https://doi.org/10.1242/dev.086157   DOI
25 Kang MH, Zhang LH, Wijesekara N, et al. Regulation of ABCA1 Protein Expression and Function in Hepatic and Pancreatic Islet Cells by miR-145. Arterioscler Thromb Vasc Biol 2013;33:2724-32. https://doi.org/10.1161/ATVBAHA.113.302004   DOI
26 Ting F, Sunmi S, Sunge C, et al. MicroRNA 34a inhibits beige and brown fat formation in obesity in part by suppressing adipocyte fibroblast growth factor 21 signaling and SIRT1 function. Mol Cell Biol 2014;34:4130-42. https://doi.org/10.1128/MCB.00596-14   DOI
27 Jian Huang LZ, Lianping Xing, Di Chen. MicroRNA-204 regulates Runx2 protein expression and mesenchymal progenitor cell differentiation. Stem Cells 2010;28:357-64. https://doi.org/10.1002/stem.288
28 Xiao L, Li H, Yang G. Sequential expression of Wnt/$\beta$-catenin signal pathway related genes and adipocyte transcription factors during porcine adipose tissue development. Chin J Biotechnol 2008;24:746-53. https://doi.org/10.1016/S1872-2075(08)60039-4   DOI
29 Suh JM, Gao X, Mckay J, Mckay R, Salo Z, Graff JM. Hedgehog signaling plays a conserved role in inhibiting fat formation. Cell Metab 2006;3:25-34. https://doi.org/10.1016/j.cmet.2005.11.012   DOI
30 Riancho JA, Vazquez L, Garcia-Perez MA, et al. Association of ACACB polymorphisms with obesity and diabetes. Mol Genet Metab 2011;104:670-6. https://doi.org/10.1016/j.ymgme.2011.08.013   DOI