Inhibitions of FASN suppress triglyceride synthesis via the control of malonyl-CoA in goat mammary epithelial cells

2017 ◽  
Vol 57 (8) ◽  
pp. 1624 ◽  
Author(s):  
J. Luo ◽  
J. J. Zhu ◽  
Y. T. Sun ◽  
H. B. Shi ◽  
J. Li

Fatty acid synthase (FASN) is the key enzyme for de novo fatty acid synthesis from acetyl-CoA and malonyl-CoA. All the steps involved in fatty acid synthesis by FASN have been clearly defined in monogastrics and ruminants. However, there are no data on the mechanism of how FASN affects triglyceride synthesis. Inhibition of FASN in goat mammary epithelial cells by C75, a synthetic inhibitor of FASN activity, and shRNA markedly suppressed the accumulation of triglyceride in goat mammary epithelial cells. Meanwhile, C75 treatment significantly reduced the relative content of monounsaturated fatty acids (C16:1 and C18:1). Corresponding to the suppression of lipid accumulation, both of C75 and shRNA also decreased the mRNA expression of GPAM, AGPAT6 and DGAT2, all of which are related to triglyceride synthesis. The fact that treatment of malonyl-CoA decreased the expression of these genes is consistent with the results of shRNA treatment. Furthermore, the supplement of malonyl-CoA enhanced the suppression on GPAM, AGPAT6, LPIN1, DGAT1 and DGAT2. The results underscore the role of malonyl-CoA in inhibition of FASN in regulating triglyceride synthesis in goat mammary epithelial cells.

Animals ◽  
2020 ◽  
Vol 10 (6) ◽  
pp. 930
Author(s):  
Jiangtao Huang ◽  
Bangguo Dai ◽  
Hexuan Qu ◽  
Yuling Zhong ◽  
Yue Ma ◽  
...  

EGF acts as a ligand of the EGF receptor (EGFR) to activate the EGFR-mediated signaling pathways and is involved in the regulation of cell physiology. However, the roles of EGFR mediated signaling pathways in the regulation of lipid metabolism in goat mammary epithelial cells (GMECs) are poorly understood. To evaluate the impact of EGF on GMECs, the triglyceride (TG) content and lipid droplet were detected, using TG assay and immunofluorescence. Further, expression of lipogenic genes, the protein kinase B (Akt), phospholipase C-γ1 (PLC-γ1) and extracellular signal-regulated kinases (ERK)1/2 signaling pathways were measured by real-time polymerase chain reaction and Western blot, respectively. The results showed that the mRNA expression of EGFR gene was significantly upregulated in lactating goat mammary gland tissues compared to non-lactation period (p < 0.05). TG contents in EGF-treated GMECs were significantly increased (p < 0.05), and an increase of lipid droplets was also detected. In vitro studies demonstrated that the mRNA levels of lipogenesis-related FASN, ACC, SCD1, LXRa, LXRb and SP1 genes were positively correlated to the mRNA level of EGFR gene shown by gene overexpression and silencing (p < 0.05). The phosphorylations of Akt, ERK1/2 and PLC-γ1 in GMECs were greatly upregulated in the presence of EGF, and specific inhibitors were capable of blocking the phosphorylation of Akt, ERK1/2 and PLC-γ1. Compared with EGF-treated GMECs, the mRNA levels of FASN, ACC and SCD1 were significantly decreased in GMECs co-treated with PLC-γ1 and Akt inhibitor and EGF (p < 0.05), and TG content was also dropped significantly. These observations implied that EGFR plays an important role in regulating de novo fatty acid synthesis in GMECs, mainly mediated by Akt and PLC-γ1 signaling pathways.


2020 ◽  
Vol 98 (3) ◽  
Author(s):  
Huibin Tian ◽  
Jun Luo ◽  
Hengbo Shi ◽  
Xiaoying Chen ◽  
Jiao Wu ◽  
...  

Abstract A key member of the nuclear receptor superfamily is the peroxisome proliferator-activated receptor alpha (PPARA) isoform, which in nonruminants is closely associated with fatty acid oxidation. Whether PPARA plays a role in milk fatty acid synthesis in ruminants is unknown. The main objective of the present study was to use primary goat mammary epithelial cells (GMEC) to activate PPARA via the agonist WY-14643 (WY) or to silence it via transfection of small-interfering RNA (siRNA). Three copies of the peroxisome proliferator-activated receptor response element (PPRE) contained in a luciferase reporter vector were transfected into GMEC followed by incubation with WY at 0, 10, 20, 30, 50, or 100 µM. A dose of 50 µM WY was most effective at activating PPRE without influencing PPARA mRNA abundance. Transfecting siRNA targeting PPARA decreased its mRNA abundance to 20% and protein level to 50% of basal levels. Use of WY upregulated FASN, SCD1, ACSL1, DGAT1, FABP4, and CD36 (1.1-, 1.5-, 2-, 1.4-, 1.5-, and 5-fold, respectively), but downregulated DGAT2 and PGC1A (−20% and −40%, respectively) abundance. In contrast, triacylglycerol concentration decreased and the content and desaturation index of C16:1 and C18:1 increased. Thus, activation of PPARA via WY appeared to channel fatty acids away from esterification. Knockdown of PPARA via siRNA downregulated ACACA, SCD1, AGPAT6, CD36, HSL, and SREBF1 (−43%, −67%, −16%, −56%, −26%, and −29%, respectively), but upregulated ACSL1, DGAT2, FABP3, and PGC1A (2-, 1.4-, 1.3-, and 2.5-fold, respectively) mRNA abundance. A decrease in the content and desaturation index of C16:1 and C18:1 coupled with an increase in triacylglycerol content accompanied those effects at the mRNA level. Overall, data suggest that PPARA could promote the synthesis of MUFA in GMEC through its effects on mRNA abundance of genes related to fatty acid synthesis, oxidation, transport, and triacylglycerol synthesis.


Sign in / Sign up

Export Citation Format

Share Document