Abstract

Objective Decreased fatty acid desaturase 1 (Fads1) activity results in the accumulation of saturated fat, which contributes to the development of nonalcoholic steatohepatitis (NASH). The aim of this study was to determine whether hepatic overexpression of Fads1 attenuates a dietary high-fat high-fructose (HFHF)-induced metabolic phenotype. Methods Hepatocyte-specific overexpression of Fads1 was achieved via the tail vein injection of recombinant adeno-associated virus (AAV) serotype 8 containing an albumin promoter with rat Fads1 sequence. Blank vectors served as controls. Male weanling Sprague-Dawley rats were assigned to five groups and fed with chow diet, low-fat high-fructose diet with AAV8 blank control (LFHF-AAV8-Blank), LFHF-AAV8-Fads1, high-fat high-fructose diet with AAV8 blank control (HFHF-AAV8-Blank), or HFHF-AAV8-Fads1 (n=8 rats/group) for 8 weeks. Metabolic phenotypes were characterized, including body weight, level of hepatic steatosis, glucose and insulin tolerance tests (GTT, ITT). Liver, skeletal muscle and adipose tissue insulin signaling were examined by western blot. Results Hepatocyte-specific overexpression of Fads1 significantly decreased body weight gain and improved glucose tolerance, but not hepatic steatosis, in LFHF-fed rats. These effects were not evident in HFHF-fed rats. On the contrary, white adipose tissue weight was markedly reduced in HFHF-AAV8-Fads1 rats compared to HFHF-AAV8-blank control rats, and to a lesser extent in LFHF rats. Both LFHF-AAV8-Fads1 and HFHF-AAV8-Fads1 rats displayed decreased fasting plasma triglycerides and increased plasma free fatty acids compared to their AAV8-blank control rats, and this was associated with upregulation of adipose tissue hormone sensitive lipase (HSL). While phosphorylated Akt (p-Akt) expression was downregulated, phosphorylated glycogen synthase kinase 3β (GSK-3β) was upregulated by both LFHF and HFHF diet in skeletal muscle, these alterations were not evident in the liver and adipose tissue. Moreover, decreased expression of p-Akt in skeletal muscle was partially restored in HFHF-AAV8-Fads1 rats, but not in LFHF-AAV8-Fads1 rats. Conclusion Our data demonstrated that hepatocyte-specific overexpression of Fads1 leads to the improvement of metabolic phenotypes in a diet- and tissue-specific manner, and this was associated with improved insulin signaling in skeletal muscle. The underlying mechanism(s) need further investigation.

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