Submit or Track your Manuscript LOG-IN

Microarray and Bioinformatics Analysis of Differentially-Expressed MicroRNAs in Wnt/Β-Catenin Signaling-Activated Bovine Skeletal Muscle Satellite Cells

Microarray and Bioinformatics Analysis of Differentially-Expressed MicroRNAs in Wnt/Β-Catenin Signaling-Activated Bovine Skeletal Muscle Satellite Cells

Ming-Yan Shi2, Xu-Liang Yan1, Ping-Ping Han1, Yu-Jie Tang1 and Hui-Xia Li1*

1College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China 
2College of Life Science, Luoyang Normal University, Luoyang, 471934, China
 
Xu-Liang Yan and Ming-Yan Shi are co-first authors.

*      Corresponding author: lihuixia@njau.edu.cn

ABSTRACT

Skeletal muscle satellite cells (SMSC) are a population of multipotent cells that exist not only during animal infancy, but are also present in the adult mammalian body. Previous studies have indicated that Wnt/β-catenin signaling is involved in the regulation of multipotent cell differentiation and is essential for the regulation of adipogenesis. However, the differentially-expressed microRNAs (miRNAs) involved in SMSC adipogenesis induction, and the role of these miRNAs in Wnt/β-catenin signaling, has received little attention, particularly in livestock. In the present study, bovine skeletal muscle satellite cells (bSMSCs) were cultured and the Wnt/β-catenin signaling pathway was activated by LiCl. Investigation of the patterns of differentially-expressed miRNAs and analysis of bSMSCs target genes were then carried out using microarray and bioinformatics. The data revealed significant changes (>2-fold) in 29 out of 783 differentially expressed miRNAs, 12 of which were up-regulated and 17 were down-regulated. Bioinformatics analysis showed that 24 of these 29 miRNAs were involved in fat metabolism, cell differentiation and the development of the immune system in 315 potential target genes, while 39 target genes were associated with adipogenesis and metabolism, immune responses, and stem cell differentiation. Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes database showed that these differentially-expressed miRNAs were involved in several pathways and were target genes that could regulate SMSC differentiation into adipocytes. Our study provided an overview of the expression profile of miRNAs, and demonstrated interactions between miRNAs and target genes, which improve our understanding of the important role that miRNAs play in SMSC adipogenic differentiation.

To share on other social networks, click on any share button. What are these?

Pakistan Journal of Zoology

April

Pakistan J. Zool., Vol. 56, Iss. 2, pp. 503-1000

Featuring

Click here for more

Subscribe Today

Receive free updates on new articles, opportunities and benefits


Subscribe Unsubscribe