Abstract

BackgroundLivestock production aims to provide meats of high and consistent eating quality. Insufficient intramuscular (IM) fat and excessive subcutaneous (SC) fat are paramount pork quality challenges. IM fat and SC fat, which are modulated by the adipogenesis of IM and SC adipocytes, play key roles in pork quality. Galectin-12 (LGALS12) was proven to be an important regulator of fat deposition in porcine. However, the current knowledge of the transcriptome-wide role of LGALS12 in adipocytes is still limited. This study was aimed to discover the different regulatory mechanisms of LGALS12 in porcine IM and SC adipocyte.ResultsThe siRNA-mediated knockdown of the expression of LGALS12 identified 1075 and 3016 differentially expressed genes (DEGs) in IM and SC adipocytes, respectively. Among these, 585 were up- and 490 were downregulated in the IM adipocytes, while 2186 were up- and 830 were downregulated in the SC adipocytes. Moreover, 418 DGEs were observed only in the IM adipocytes, 2359 DGEs only in the SC adipocytes, and 657 DGEs in both types of adipocytes. According to Gene Ontology (GO) analysis, DEGs in both IM and SC adipocytes were mainly enriched in categories related to lipids or fat cell differentiation. Pathway analysis of the DEGs revealed 88 changed signaling pathways in the IM adipocytes and 86 in the SC adipocytes. The signaling pathways present in only one type of adipocyte were identified from among the top 50 signaling pathways in each type of adipocyte. Four signaling pathways, encompassing PI3K-AKT, cardiac muscle contraction, fatty acid metabolism and Ras, were significantly enriched in the IM adipocytes. On the other hand, four different signaling pathways, encompassing TNF, WNT, cGMP-PKG and NF-kappa B, were greatly enriched in the SC ones. The pathway changes were confirmed by chemical inhibition assays.ConclusionsOur data reveals that LGALS12 knockdown alters the expression of numerous genes involved in key biological processes in the development of adipocytes. These observations provide a global view of the role of LGALS12 in porcine IM and SC adipocytes; thus, improving our understanding of the regulatory mechanisms by which this gene acts in fat development.

Highlights

  • Livestock production aims to provide meats of high and consistent eating quality

  • Our previous study indicated that the intracellular triglyceride content of SC preadipocytes increased more dramatically than that of IM preadipocytes during cell differentiation, which was accompanied by lower expression levels of genes related to lipid metabolism such as Peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer binding protein-α (C/EBPα), Adipose Triglyceride Lipase (ATGL), Hormone-sensitive lipase (HSL), Lipoprotein Lipase (LPL), Fatty acid binding protein 4 (aP2) and Fatty acid synthetase (FAS), in IM adipocytes [5]

  • RNA sequencing technology (RNA-Seq) investigation of the effects of silencing LGALS12 expression in adipocytes The expression levels of LGALS12 determined by qRTPCR in LGALS12-siRNA treated intramuscular (IM) and subcutaneous (SC) adipocytes were about 50% lower than those of cells with negative control group (NC)-siRNA treatment (n = 3, P < 0.01) (Additional file 1: Figure S1), which indicated that LGALS12-siRNA effectively interfered with the expression of LGALS12

Read more

Summary

Introduction

Livestock production aims to provide meats of high and consistent eating quality. Insufficient intramuscular (IM) fat and excessive subcutaneous (SC) fat are paramount pork quality challenges. This study was aimed to discover the different regulatory mechanisms of LGALS12 in porcine IM and SC adipocyte. The intramuscular (IM) fat content is considered a crucial indicator of porcine meat quality, while subcutaneous (SC) fat affects the lean meat percentage of the carcass [1]. To meet the consumers’ increasing demands for high-quality pork, a main goal of breeding is to improve IM fat and to decrease the SC fat content [2]. Our previous study indicated that the intracellular triglyceride content of SC preadipocytes increased more dramatically than that of IM preadipocytes during cell differentiation, which was accompanied by lower expression levels of genes related to lipid metabolism such as PPARγ, C/EBPα, ATGL, HSL, LPL, aP2 and FAS, in IM adipocytes [5]. The expression of genes and proteins that participate in cell growth, such as insulin-like growth factor II (IGF-II) and prohibitin-1, was higher in IM than in SC adipocytes [6]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call