DOI QR코드

DOI QR Code

Screening of key miRNAs related with the differentiation of subcutaneous adipocytes and the validation of miR-133a-3p functional significance in goats

  • Xin, Li (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University) ;
  • Hao, Zhang (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University) ;
  • Yong, Wang (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University) ;
  • Yanyan, Li (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University) ;
  • Youli, Wang (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University) ;
  • Jiangjiang, Zhu (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University) ;
  • Yaqiu, Lin (Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Protection and Utilization of Ministry of Education/Sichuan Province, Southwest Minzu University)
  • Received : 2022.03.25
  • Accepted : 2022.06.03
  • Published : 2023.01.01

Abstract

Objective: Adipocyte differentiation is regulated by a variety of functional genes and noncoding RNAs. However, the role of miRNAs in lipid deposition of goat white adipose tissue is still unclear. Therefore, this study revealed the miRNA expression profile in goat subcutaneous adipocytes by sRNA-seq. Methods: The miRNA expressed in goat subcutaneous preadipocytes and the mature adipocytes were sequenced by sRNA-seq. The differentially expressed miRNAs (DEm) were screened and gene ontology (GO) and Kyoto encyclopedia for genes and genomes (KEGG) analyses were performed. Gain-of-function and loss-of-function combined with oil red O staining, Bodipy staining, and quantitative reverse-transcription polymerase chain reaction (qPCR) were utilized to determine the effect of miR-133a-3p on adipocyte differentiation. Results: A total of 218 DEm were screened out. The target genes of these DEm were significantly enriched in GO items such as biological regulation and in KEGG terms such as FAK signaling pathway and MAPK signaling pathway. qPCR verified that the expression trend of miRNA was consistent with miRNA-seq. The gain-of-function or loss-of-function of miR-133a-3p showed that it promoted or inhibited the accumulation of lipid droplets, and CCAAT enhancer binding protein α (C/EBPα) and C/EBPβ were extremely significantly up-regulated or down-regulated respectively (p<0.01), the loss-of-function also led to a significant down-regulation of peroxisome proliferator activated receptor gamma (PPARγ) (p<0.01). Conclusion: This study successfully identified miRNAs expression patterns in goat subcutaneous adipocytes, and functional identification indicates that miR-133a-3p is a positive regulator of the differentiation process of goat subcutaneous adipocytes. Our results lay the foundation for the molecular mechanism of lipid deposition in meat-source goats from the perspective of miRNA.

Keywords

Acknowledgement

We want to affectionately thank Prof. Bai Wenlin for his assistance in the revision of this article.

References

  1. Ripoll G, Alcalde MJ, Arguello A, Cordoba MG, Panea B. Effect of rearing system on the straight and branched fatty acids of goat milk and meat of suckling kids. Foods 2020;9:471. https://doi.org/10.3390/foods9040471
  2. Watkins PJ, Frank D. Heptadecanoic acid as an indicator of BCFA content in sheep fat. Meat Sci 2019;151:33-5. https://doi.org/10.1016/j.meatsci.2019.01.005
  3. Gregoire FM, Smas CM, Sul HS. Understanding adipocyte differentiation. Physiol Rev 1998;78:783-809. https://doi.org/10.1152/physrev.1998.78.3.783
  4. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281-97. https://doi.org/10.1016/s0092-8674(04)00045-5
  5. Ambros V. The functions of animal microRNAs. Nature 2004;431:350-5. https://doi.org/10.1038/nature02871
  6. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 2014;15:509-24. https://doi.org/10.1038/nrm3838
  7. Sun YM, Qin J, Liu SG, et al. PDGFRα Regulated by miR34a and FoxO1 Promotes Adipogenesis in Porcine Intramuscular Preadipocytes through Erk Signaling Pathway. Int J Mol Sci 2017;18:2424. https://doi.org/10.3390/ijms18112424
  8. Chen L, Song J, Cui J, et al. microRNAs regulate adipocyte differentiation. Cell Biol Int 2013;37:533-46. https://doi.org/10.1002/cbin.10063
  9. Hu X, Tang J, Hu X, et al. MiR-27b impairs adipocyte differentiation of human adipose tissue-derived mesenchymal stem cells by targeting LPL. Cell Physiol Biochem 2018;47:545-55. https://doi.org/10.1159/000489988
  10. Qi R, Wang J, Wang Q, et al. MicroRNA-425 controls lipogenesis and lipolysis in adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2019;1864:744-55. https://doi.org/10.1016/j.bbalip.2019.02.007
  11. Zhang M, Li DH, Li F, et al. Integrated analysis of MiRNA and genes associated with meat quality reveals that gga-mir140-5p affects intramuscular fat deposition in chickens. Cell Physiol Biochem 2018;46:2421-33. https://doi.org/10.1159/000489649
  12. Hu Y, Wang K, He X, Chiang DY, Prins JF, Liu J. A probabilistic framework for aligning paired-end RNA-seq data. Bioinformatics 2010;26:1950-7. https://doi.org/10.1093/bioinformatics/btq336
  13. Li X, Zhang H, Wang Y, et al. RNA-seq analysis reveals the positive role of KLF5 in the differentiation of subcutaneous adipocyte in goats. Gene 2022;808:145969. https://doi.org/10.1016/j.gene.2021.145969
  14. Peng Y, Yu S, Li H, Xiang H, Peng J, Jiang S. MicroRNAs: emerging roles in adipogenesis and obesity. Cell Signal 2014;26:1888-96. https://doi.org/10.1016/j.cellsig.2014.05.006
  15. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet 2009;10:57-63. https://doi.org/10.1038/nrg2484
  16. Bakhtiarizadeh MR, Salehi A, Alamouti AA, Abdollahi-Arpanahi R, Salami SA. Deep transcriptome analysis using RNA-Seq suggests novel insights into molecular aspects of fat-tail metabolism in sheep. Sci Rep 2019;9:9203. https://doi.org/10.1038/s41598-019-45665-3
  17. Zhang Y, Yu B, Yu J, et al. Butyrate promotes slow-twitch myofiber formation and mitochondrial biogenesis in finishing pigs via inducing specific microRNAs and PGC-1α expression. J Anim Sci 2019;97:3180-92. https://doi.org/10.1093/jas/skz187
  18. Wang L, Zhang N, Pan HP, Wang Z, Cao ZY. MiR-499-5p contributes to hepatic insulin resistance by suppressing PTEN. Cell Physiol Biochem 2015;36:2357-65. https://doi.org/10.1159/000430198
  19. Liu W, Bi P, Shan T, et al. miR-133a regulates adipocyte browning in vivo. PLoS Genet 2013;9:e1003626. https://doi.org/10.1371/journal.pgen.1003626
  20. Liu H, Wang T, Chen X, et al. Inhibition of miR-499-5p expression improves nonalcoholic fatty liver disease. Ann Hum Genet 2020 Jan 20 [Epub]. https://doi.org/10.1111/ahg.12374
  21. Y oussef EM, Elfiky AM, BanglySoliman, Abu-Shahba N, Elhefnawi MM. Expression profiling and analysis of some miRNAs in subcutaneous white adipose tissue during development of obesity. Genes Nutr 2020;15:8. https://doi.org/10.1186/s12263-020-00666-0
  22. Ning X, Liu S, Qiu Y, et al. Expression profiles and biological roles of miR-196a in swine. Genes (Basel) 2016;7:5. https://doi.org/10.3390/genes7020005
  23. Mori M, Nakagami H, Rodriguez-Araujo G, Nimura K, Kaneda Y. Essential role for miR-196a in brown adipogenesis of white fat progenitor cells. PLoS Biol 2012;10:e1001314. https://doi.org/10.1371/journal.pbio.1001314
  24. Hou Y, Fu L, Li J, et al. Transcriptome analysis of potential miRNA involved in adipogenic differentiation of C2C12 myoblasts. Lipids 2018;53:375-86. https://doi.org/10.1002/lipd.12032
  25. Li JJ, Xie D. Cleavage of focal adhesion kinase (FAK) is essential in adipocyte differentiation. Biochem Biophys Res Commun 2007;357:648-54. https://doi.org/10.1016/j.bbrc.2007.03.184
  26. Xiong Y, Wang YX, Xu Q, et al. LKB1 regulates goat intramuscular adipogenesis through focal adhesion pathway. Front Physiol 2021;12:755598. https://doi.org/10.3389/fphys.2021.755598
  27. Sakaue H, Ogawa W, Nakamura T, Mori T, Nakamura K, Kasuga M. Role of MAPK phosphatase-1 (MKP-1) in adipocyte differentiation. J Biol Chem 2004;279:39951-7. https://doi.org/10.1074/jbc.M407353200
  28. Wu W, Zhang J, Zhao C, Sun Y, Pang W, Yang G. CTRP6 regulates porcine adipocyte proliferation and differentiation by the AdipoR1/MAPK signaling pathway. J Agric Food Chem 2017;65:5512-22. https://doi.org/10.1021/acs.jafc.7b00594
  29. Wang Q, Li D, Cao G, et al. IL-27 signalling promotes adipocyte thermogenesis and energy expenditure. Nature 2021;600:314-8. https://doi.org/10.1038/s41586-021-04127-5
  30. Okla M, Kim J, Koehler K, Chung S. Dietary factors promoting brown and beige fat development and thermogenesis. Adv Nutr 2017;8:473-83. https://doi.org/10.3945/an.116.014332
  31. Sakamoto T, Takahashi N, Goto T, Kawada T. Dietary factors evoke thermogenesis in adipose tissues. Obes Res Clin Pract 2014;8:e533-9. https://doi.org/10.1016/j.orcp.2013.12.002
  32. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell 2014;156:20-44. https://doi.org/10.1016/j.cell.2013.12.012
  33. Zhang Y, Li R, Meng Y, et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes 2014;63:514-25. https://doi.org/10.2337/db13-1106
  34. Song Z, Wang Y, Zhang F, Yao F, Sun C. Calcium signaling pathways: key pathways in the regulation of obesity. Int J Mol Sci 2019;20:2768. https://doi.org/10.3390/ijms20112768
  35. He YH, He Y, Liao XL, et al. The calcium-sensing receptor promotes adipocyte differentiation and adipogenesis through PPARγ pathway. Mol Cell Biochem 2012;361:321-8. https://doi.org/10.1007/s11010-011-1118-5
  36. 36 Xiao J, Bai XQ, Liao L, et al. Hydrogen sulfide inhibits PCSK9 expression through the PI3K/Akt-SREBP-2 signaling pathway to influence lipid metabolism in HepG2 cells. Int J Mol Med 2019;43:2055-63. https://doi.org/10.3892/ijmm.2019.4118
  37. Song BQ, Chi Y, Li X, et al. Inhibition of notch signaling promotes the adipogenic differentiation of mesenchymal stem cells through autophagy activation and PTEN-PI3K/AKT/mTOR pathway. Cell Physiol Biochem 2015;36:1991-2002. https://doi.org/10.1159/000430167
  38. Lee JH, Jung HA, Kang MJ, Choi JS, Kim GD. Fucosterol, isolated from Ecklonia stolonifera, inhibits adipogenesis through modulation of FoxO1 pathway in 3T3-L1 adipocytes. J Pharm Pharmacol 2017;69:325-33. https://doi.org/10.1111/jphp.12684
  39. Krycer JR, Quek LE, Francis D, et al. Insulin signaling requires glucose to promote lipid anabolism in adipocytes. J Biol Chem 2020;295:13250-66. https://doi.org/10.1074/jbc.RA120.014907
  40. Shan T, Liu J, Wu W, Xu Z, Wang Y. Roles of notch signaling in adipocyte progenitor cells and mature adipocytes. J Cell Physiol 2017;232:1258-61. https://doi.org/10.1002/jcp.25697
  41. Bluthgen N, Legewie S. Systems analysis of MAPK signal transduction. Essays Biochem 2008;45:95-108. https://doi.org/10.1042/bse045095
  42. Han HJ, Lee YJ. Insulin stimulates Ca2+ uptake via PKC, cAMP, and p38 MAPK in mouse embryonic stem cells. Life Sci 2005;76:2903-19. https://doi.org/10.1016/j.lfs.2004.10.060
  43. Oliveira MS, Rheinheimer J, Moehlecke M, et al. UCP2, IL18, and miR-133a-3p are dysregulated in subcutaneous adipose tissue of patients with obesity. Mol Cell Endocrinol 2020;509:110805. https://doi.org/10.1016/j.mce.2020.110805