Elsevier

Biochemical Pharmacology

Volume 93, Issue 4, 15 February 2015, Pages 482-495
Biochemical Pharmacology

A platelet-activating factor (PAF) receptor deficiency exacerbates diet-induced obesity but PAF/PAF receptor signaling does not contribute to the development of obesity-induced chronic inflammation

https://doi.org/10.1016/j.bcp.2014.12.022Get rights and content

Abstract

Platelet-activating factor (PAF) is a well-known phospholipid that mediates acute inflammatory responses. In the present study, we investigated whether PAF/PAF receptor signaling contributed to chronic inflammation in the white adipose tissue (WAT) of PAF receptor-knockout (PAFR-KO) mice. Body and epididymal WAT weights were higher in PAFR-KO mice fed a high-fat diet (HFD) than in wild-type (WT) mice. TNF-α mRNA expression levels in epididymal WAT and the infiltration of CD11c-positive macrophages into epididymal WAT, which led to chronic inflammation, were also elevated in HFD-fed PAFR-KO mice. HFD-fed PAFR-KO mice had higher levels of fasting serum glucose than HFD-fed WT mice as well as impaired glucose tolerance. Although PAF receptor signaling up-regulated the expression of TNF-α and lipopolysaccharide induced the expression of acyl-CoA:lysophosphatidylcholine acyltransferase 2 (LPCAT2) mRNA in bone marrow-derived macrophages, no significant differences were observed in the expression of LPCAT2 mRNA and PAF levels in epididymal WAT between HFD-fed mice and normal diet-fed mice. In addition to our previous finding in which energy expenditure in PAF receptor (PAFR)-deficient mice was low due to impaired brown adipose tissue function, the present study demonstrated that PAF/PAF receptor signaling up-regulated the expression of Ucp1 mRNA, which is essential for cellular thermogenesis, in 3T3-L1 adipocytes. We concluded that the marked accumulation of abdominal fat due to HFD feeding led to more severe chronic inflammation in WAT, which is associated with glucose metabolism disorders, in PAFR-KO mice than in WT mice, and PAF/PAF receptor signaling may regulate energy expenditure and adiposity.

Introduction

Obesity is an emerging global medical issue that has long been considered as one of the most prominent risk factors for metabolic syndrome, including insulin resistance, hypertension, and hyperlipidemia [1]. Recent extensive studies have shown that insulin resistance in obesity can be attributed to chronic low-grade inflammation in adipose tissue [2]. As obesity develops, neutrophils, macrophages, and T cells infiltrate into expanding adipose tissues, and pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β are released from these infiltrating immune cells [3], [4], [5]. These secreted pro-inflammatory cytokines have been shown to suppress insulin signaling and evoke insulin resistance [6]. Therefore, many factors that are associated with inflammatory responses are known to regulate chronic inflammation and insulin resistance [7].

Signaling lipids such as prostaglandins, eicosanoids, phosphoinositides, sphingolipids, and fatty acids control cellular processes, and also play important roles in immune responses. These lipid mediators are produced by lipid-modifying enzymes and exert their biological effects by binding to cognate receptors. Recent studies demonstrated that these lipid-associated enzymes and receptors regulated obesity-induced insulin resistance and indicated that signaling lipids also play crucial roles in insulin resistance [8], [9]. Platelet-activating factor (PAF, 1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholine) is a bioactive phospholipid that was first identified as a substance that aggregates platelets during anaphylactic shock in rabbits [10]. However, recent studies found that PAF mediated a broad range of biological actions including immune and non-immune responses. PAF is enzymatically synthesized by two distinct pathways: a de novo pathway and remodeling pathway. PAF is synthesized from 1-O-alkyl-2-acetyl-sn-glycerol in the de novo pathway. In the remodeling pathway, lyso-PAF (1-O-alkyl-sn-glycero-3-phosphocholine) is generated from 1-O-alkyl-2 acyl-sn-glycero-3-phosphocholine by phospholipase A2 (PLA2), and cytosolic PLA2α (cPLA2α) plays a major role in the production of PAF by inflammatory cells [11]. PAF is synthesized through the remodeling pathway under inflammatory conditions, and PAF is then biosynthesized from lyso-PAF by lysophosphatidylcholine acyltransferase 2 (LPCAT2) [12]. PAF was previously shown to exert its bioactive effects by binding to its G-protein-coupled seven-transmembrane receptor [13], [14]. The PAF receptor couples with Gαi and Gαq proteins, which regulate the concentrations of cyclic AMP and Ca2+, respectively. PAF has well-characterized biological actions, such as the stimulation of inflammatory cells, including platelets and neutrophils, promotion of leukocyte chemotaxis, regulation of pro-inflammatory cytokines, and smooth muscle contraction, and has been implicated in many inflammatory diseases [15], [16], [17], [18], [19], [20].

We previously reported that adiposity and weight gain were increased with age in PAF receptor-knockout (PAFR-KO) mice, and, although these mice developed severe obesity, abnormal liver weights were not observed [21]. The epididymal white adipose tissue (WAT) of PAFR-KO mice at 24 weeks was susceptible to chronic inflammation (the infiltration of macrophages and production of proinflammatory adipokines) earlier than that of the wild-type (WT) littermates; however, PAFR-KO mice did not exhibit metabolic disorders such as hyperglycemia [21]. We here investigated the role of PAF receptor signaling in obesity-induced chronic inflammation in WAT using high-fat diet (HFD)-fed mice. Inflammatory cytokine mRNA levels and the infiltration of classically activated macrophages in the WAT were markedly higher in HFD-fed PAFR-KO mice than in HFD-fed WT mice in vivo, even though PAF receptor signaling promoted inflammatory phenotypes in the in vitro experiments. Furthermore, fasting serum levels of glucose were higher and glucose tolerance was more severely impaired in HFD-fed PAFR-KO mice than in HFD-fed WT mice. In the present study, we demonstrated for the first time that PAFR-KO mice fed a HFD developed chronic inflammation and glucose metabolism disorders in vivo.

Section snippets

Reagents

Lipopolysaccharide (LPS) from Escherichia coli O55:B5 was purchased from Difco Laboratories (Detroit, MI). 1-O-Hexadecyl-2-(N-methylcarbamyl)-sn-glycero-3-phosphocholine (c-PAF) was purchased from Sigma–Aldrich (St. Louis, MO). IL-4 was purchased from R&D Systems (Minneapolis, MN).

Animals

PAFR-KO and WT mice were originally obtained from Drs. Satoshi Ishii and Takao Shimizu, as described previously [22]. Mice were housed under specific pathogen-free conditions at the University of Shizuoka and given ad

The PAF receptor, cPLA2α, and LPCAT2 were highly expressed in WAT

In the first series of experiments, we evaluated the expression of genes encoding the PAF receptor (Ptafr), cPLA2α, LPCAT2, and LPCAT1 in adipose tissue. Whole lung tissue was used as a positive control [11]. The expression levels of PAF receptor mRNA were similar between brown adipose tissue and whole lung tissue or were higher in WAT. PAF receptor mRNA levels were markedly upregulated in epididymal WAT (Fig. 1A). The PAF synthetic enzymes, cPLA2α and LPCAT2, were also highly expressed in both

Discussion

In the present study, we generated obesity model mice at a young age with glucose metabolism disorders; a deficiency in the PAF receptor in 20-week-old mice fed a HFD for 12 weeks resulted in more severe obesity, higher levels of serum glucose, and less glucose tolerance than WT mice (Fig. 3, Fig. 9). Body weight with age and adiposity were higher in ND-fed PAFR-KO mice than in ND-fed WT littermates, and PAFR-KO mice did not exhibit metabolic disorders such as mild hyperglycemia or fatty liver

Acknowledgments

This work was supported in part by a Grant-in-Aid for Young Scientists (B) (26860041, to MY) and Grant-in-Aid for challenging Exploratory Research (26670032, to JS) from the JSPS KAKENHI and by the Sasakawa Scientific Research Grant from The Japan Science Society (to MY). We gratefully acknowledge Ryu Shou, Yoshiki Hattori, and Masayuki Miyatake for their excellent technical assistance.

References (41)

  • S.P. Weisberg et al.

    Obesity is associated with macrophage accumulation in adipose tissue

    J Clin Invest

    (2003)
  • S. Nishimura et al.

    CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity

    Nat Med

    (2009)
  • M. Yuan et al.

    Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta

    Science

    (2001)
  • O. Osborn et al.

    The cellular and signaling networks linking the immune system and metabolism in disease

    Nat Med

    (2011)
  • M.P. Wymann et al.

    Lipid signalling in disease

    Nat Rev Mol Cell Biol

    (2008)
  • A. Iyer et al.

    Inflammatory lipid mediators in adipocyte function and obesity

    Nat Rev Endocrinol

    (2010)
  • J. Benveniste et al.

    Leukocyte-dependent histamine release from rabbit platelets. The role of IgE, basophils, and a platelet-activating factor

    J Exp Med

    (1972)
  • T. Shimizu

    Lipid mediators in health and disease: enzymes and receptors as therapeutic targets for the regulation of immunity and inflammation

    Annu Rev Pharmacol Toxicol

    (2009)
  • S. Ishii et al.

    Impaired anaphylactic responses with intact sensitivity to endotoxin in mice lacking a platelet-activating factor receptor

    J Exp Med

    (1998)
  • T. Nagase et al.

    Platelet-activating factor mediates acid-induced lung injury in genetically engineered mice

    J Clin Invest

    (1999)
  • Cited by (0)

    1

    Masakazu Matsui and Ryoko Higa contributed equally to this study.

    View full text