Abstract

Metabolic syndrome is characterized by visceral adiposity, insulin resistance, high triglyceride (TG)- and low high-density lipoprotein cholesterol-levels, hypertension, and diabetes—all of which often cause cardiovascular and cerebrovascular diseases. It remains unclear, however, why visceral adiposity but not subcutaneous adiposity causes insulin resistance and other pathological situations. Lipoprotein lipase (LPL) catalyzes hydrolysis of TG in plasma lipoproteins. In the present study, we investigated whether the effects of angiotensin II (AngII) on TG metabolism are mediated through an effect on LPL expression. Adipose tissues were divided into visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) for comparison. AngII accelerated LPL expression in SAT but, on the contrary, suppressed its expression in VAT. In both SAT and VAT, AngII signaled through the same type 1 receptor. In SAT, AngII increased LPL expression via c-Src and p38 MAPK signaling. In VAT, however, AngII reduced LPL expression via the Gq class of G proteins and the subsequent phospholipase C β4 (PLCβ4), protein kinase C β1, nuclear factor κB, and inducible nitric oxide synthase signaling pathways. PLCβ4 small interfering RNA experiments showed that PLCβ4 expression is important for the AngII-induced LPL reduction in VAT, in which PLCβ4 expression increases in the evening and falls at night. Interestingly, PLCβ4 expression in VAT decreased with fasting, while AngII did not decrease LPL expression in VAT in a fasting state. In conclusion, AngII reduces LPL expression through PLCβ4, the expression of which is regulated by feeding in VAT, whereas AngII increases LPL expression in SAT. The different effects of AngII on LPL expression and, hence, TG metabolism in VAT and SAT may partly explain their different contributions to the development of metabolic syndrome.

Highlights

  • The triglyceride (TG) lipase gene subfamily is comprised of three evolutionarily related lipases, i.e., lipoprotein lipase (LPL), hepatic lipase, and endothelial lipase, and plays a central role in plasma lipoprotein metabolism and homeostasis

  • We examined whether modification of the expression and secretion of LPL by angiotensin II (AngII) depends on adipocytes or fibrocytes in adipose tissues

  • A third of phospholipase C β4 (PLCβ4) protein was still expressed by the small interfering RNA (siRNA) (Fig 7A), the reduction in LPL protein and mRNA expression induced by AngII was almost completely reversed by the PLCβ4 siRNA, while this siRNA did not appreciably affect the phorbol 12-myristate 13-acetate (PMA)-induced suppression of LPL expression (Fig 7B and 7C). These results suggest that the AngII-induced inhibition of LPL expression is mediated by PLCβ4 and, raise the possibility that the difference in the expression level of PLCβ4 between subcutaneous and visceral adipocytes may cause the different effects of AngII on LPL expression in adipose tissues

Read more

Summary

Introduction

The triglyceride (TG) lipase gene subfamily is comprised of three evolutionarily related lipases, i.e., lipoprotein lipase (LPL), hepatic lipase, and endothelial lipase, and plays a central role in plasma lipoprotein metabolism and homeostasis. A recent study using 1511 individuals in the MESA (Multi-Ethnic Study of Atherosclerosis) with adiposity assessment by computed tomography (CT) suggested that visceral adiposity is essential to assessing cardiometabolic risk, regardless of age, race, and body mass index [7]. It is not fully understood, why visceral adipose tissue (VAT) but not subcutaneous adipose tissue (SAT) brings about insulin resistance and related events [4,5,6,7]

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