Elsevier

Cytokine

Volume 41, Issue 2, February 2008, Pages 174-181
Cytokine

TNFα is a potent inducer of platelet-activating factor synthesis in adipocytes but not in preadipocytes. Differential regulation by PI3K

https://doi.org/10.1016/j.cyto.2007.11.008Get rights and content

Abstract

Tumour necrosis factor alpha (TNFα) induces platelet-activating factor (PAF) synthesis in many inflammatory cells. Here, we investigate the possibility that TNFα stimulates PAF synthesis in rat adipocytes and preadipocytes and that phosphoinositide 3-kinase (PI3K) and extracellular signal-regulated kinase 1/2 (ERK1/2) are implicated in this process. Primary cultures were incubated with [3H]lyso-PAF and stimulated by TNFα in the presence or absence of wortmannin. We found that, although both cultures synthesized PAF at a similar basal rate, TNFα-induced PAF synthesis in adipocytes was 7-fold higher than in preadipocytes. This suggested a maturation of PAF–TNFα interrelationship during adipocyte differentiation. Wortmannin enhanced TNFα-dependent PAF synthesis in adipocytes but not in preadipocytes, indicating the negative control by PI3K in mature cells. PAF increase was due to the regulation of its biosynthesis since PAF-acetylhydrolase (PAF-AH) activity was TNFα- and wortmannin-independent. Our hypothesis is that PAF mediates TNFα inflammatory effects in both adipocytes and preadipocytes and that this pathway is enhanced during adipocyte differentiation, a mechanism which is highly active during the development of obesity.

Introduction

White Adipose Tissue (WAT),1 besides its traditional role in energy storage, is now well documented as a secretory and endocrine organ, which actively participates in the regulation of various physiological and pathological processes including immunity and inflammation [1]. Apart from adipocytes, which comprise the main cell type, WAT also contains stromovascular and immune cells, as well as connective tissue matrix and nerve tissue. WAT secretes biologically active proteins, collectively known as adipokines, derived from both adipocytes and the non-adipocyte fraction of the adipose tissue [2]. Several of these adipokines are known to be associated with inflammation as well as with obesity or metabolic syndrome [1], [3], [4]. Among them, tumour necrosis factor α (TNFα), which is oversecreted from WAT of obese individuals [5], has been suggested to be responsible for metabolic disturbances associated with obesity, mainly with insulin resistance [6].

TNFα is a proinflammatory cytokine implicated in a number of cell functions such as differentiation, proliferation and apoptosis [7]. Many cell types, including adipocytes, express and secrete TNFα, which acts in a paracrine, autocrine and even endocrine manner via two distinct receptors: TNFR1 and TNFR2 [8], [9], [10]. The downstream targets of TNFα, however, are not fully elucidated, although TNFα has been shown to activate phosphoinositide 3-kinase (PI3K) [11], [12], [13] as well as members of the mitogen-activated protein kinase (MAPK) family [14], [15], [16]. In addition, the implication of lipid mediators, such as platelet-activating factor (PAF), has been reported [17], [18], [19]. PAF, which is also a mediator of inflammation, is synthesized in many cell types by de novo or remodeling pathways [20]. Upon stimulation, PAF is synthesized mainly through the remodeling pathway which involves phospholipase A2 (PLA2) action on membrane glyceryl ethers and the consequent acetylation of the lyso-product at the sn-2 position by PAF-acetyltransferase [21]. Interestingly, in this pathway, extracellular signal-regulated kinase (ERK) is implicated; it has been shown that, in human neutrophils, ERK participates in PLA2 activation which is the initial step of PAF synthesis [22]. PAF mediates a number of biological activities of TNFα, including TNFα-induced lethal hepatitis and the impairment of skeletal muscle contractility [17], [18].

In adipocytes, TNFα has multiple effects, such as regulation of gene expression [12], [23], downregulation of adipocyte specific proteins [24], regulation of adipocyte number and size [8], [25], induction of insulin resistance [26] and stimulation of lipolysis [14], [27], [28]. There is evidence that PI3K as well as ERK1/2 are involved in some of these effects [11], [12], [14], [28].

Our research group has already shown the ability of mature adipocytes to synthesize PAF under stimulation and the presence of PAF-acetylhydrolase (PAF-AH) activity in these cells. This enzyme catalyzes the hydrolysis of the acetyl ester at the sn-2 position of PAF and converts it into its inactive metabolite, lyso-PAF, thus regulating PAF levels in a number of cells [29]. In the present study we investigate the possibility that TNFα stimulates PAF biosynthesis in adipocytes. We are also interested in whether a similar effect exists in preadipocytes. This would suggest that PAF, a potent inducer of inflammation, is recruited by TNFα to exert its inflammatory effects in adipose tissue. Our interest is also focused on the possible implication of PI3K and ERK1/2 in this process.

Section snippets

Materials

Cell culture media and reagents were purchased from Biochrom. Collagenase (from Clostridium histolyticum, type VII), TNFα (rat recombinant) and wortmannin were purchased from Sigma. 1-O-Hexadecyl-2-[3H]acetyl-sn-glycero-3-phosphocholine ([3H]acetyl-PAF, specific radioactivity 6 Ci/mmol) and 1-O-hexadecyl-2-sn-glycero-3-phosphocholine ([3H]lyso-PAF, specific radioactivity 58.5 Ci/mmol) were purchased from DuPont NEN. Solid phase extraction (SPE) columns were from IST. Tris–HCl was from Bio-Rad.

PAF synthesis from lyso-PAF in adipocytes and preadipocytes

In vivo PAF synthesis from lyso-PAF was studied using radioactive [3H]lyso-PAF and acetyl-CoA as substrates. In order to separate the synthesized PAF from the remaining non-esterified lyso-PAF a modified TLC method was used. The developing solvent consisted of chloroform–methanol–acetic acid–water (50:25:10:4). The efficiency of this separation method was first demonstrated by developing standard radioactive [3H]PAF and [3H]lyso-PAF on the same plate. After the development, the plate was

Discussion

Our results present evidence that TNFα is a potent inducer of PAF biosynthesis in the main cell type of white adipose tissue, namely adipocytes. TNFα also induces PAF synthesis in preadipocytes, but interestingly, the effect of TNFα on PAF production was 7-fold stronger in mature adipocytes. This difference reflects a maturation of TNFα–PAF interrelationship during the differentiation of preadipocytes into adipocytes, a process which is highly active during the development of obesity. Moreover,

Acknowledgments

This paper is part of the 03ED386 research project, implemented within the framework of the “Reinforcement Programme of Human Research Manpower” (PENED) and co-financed by National and Community Funds (25% from the Greek Ministry of Development-General Secretariat of Research and Technology and 75% from E.U.-European Social Fund.

References (55)

  • H. Ruan et al.

    Insulin resistance in adipose tissue: direct and indirect effects of tumor necrosis factor-α

    Cytokine Growth Factor Rev

    (2003)
  • J.K. Sethi et al.

    The role of TNFα in adipocyte metabolism

    Semin Cell Dev Biol

    (1999)
  • K. Karasawa et al.

    Plasma platelet activating factor-acetylhydrolase (PAF-AH)

    Prog Lipid Res

    (2003)
  • M. Rodbell

    Metabolism of isolated fat cells

    J Biol Chem

    (1964)
  • H. Ruan et al.

    Standard isolation of primary adipose cells from mouse epididymal fat pads induces inflammatory mediators and down-regulates adipocyte genes

    J Biol Chem

    (2003)
  • J. Folch et al.

    A simple method for the separation and purification of total lipids from animal tissue

    J Biol Chem

    (1957)
  • A. Chroni et al.

    Characterization of a platelet activating factor acetylhydrolase from rat adipocyte

    Life Sci

    (2000)
  • O.H. Lowry et al.

    Protein measurement with the Folin phenol reagent

    J Biol Chem

    (1951)
  • P. Schling et al.

    Expression of tumor necrosis factor alpha and its receptors during cellular differentiation

    Cytokine

    (2006)
  • B. Feve et al.

    Les relations entre obésité, inflammation et insulinoresistance

    C R Biol

    (2006)
  • E. Ramos et al.

    Is obesity an inflammatory disease?

    Surgery

    (2003)
  • P.A. Permana et al.

    Macrophage-secreted factors induce adipocyte inflammation and insulin resistance

    Biochem Biophys Res Commun

    (2006)
  • K. Doi et al.

    Attenuation of folic acid-induced renal inflammatory injury in platelet-activating factor receptor-deficient mice

    Am J Pathol

    (2006)
  • T. Asano et al.

    p110β is up-regulated during differentiation of 3T3-L1 cells and contributes to the highly insulin-responsive glucose transport activity

    J Biol Chem

    (2000)
  • T. Pederson et al.

    Regulation of proteins involved in insulin signaling pathways in differentiating human adipocytes

    Biochem Biophys Res Commun

    (2000)
  • E.E. Kershaw et al.

    Adipose tissue as an endocrine organ

    J Clin Endocrinol Metab

    (2004)
  • K. Esposito et al.

    Role of adipokines in the obesity-inflammation relationship: the effect of fat removal

    Plast Reconstr Surg

    (2006)
  • Cited by (10)

    • Mechanisms underlying fat pad remodeling induced by fasting: role of PAF receptor

      2020, Nutrition
      Citation Excerpt :

      PAFR is expressed in several cells including endothelium, leukocytes, adipocytes, and stromal vascular cells [10,23]. Gountopoulou et al. [24] demonstrated that the incubation of adipocyte in vitro with PAF increased TNF-α production, indicating the association between this lipid mediator with the metabolism of adipose tissue [24]. Based on the assumption that inflammation may modulate lipolysis rate, we argue whether during fasting the inflammatory response would be important to maintain the proper fat mobilization.

    • Significance of lipid mediators in corneal injury and repair

      2010, Journal of Lipid Research
      Citation Excerpt :

      The results suggest either: a) the existence of two independent pathways controlling apoptosis, or b) the involvement of TNFα in increasing PAF synthesis, which amplifies the apoptotic response to TNFα. With respect to the second possibility, there are reports that TNFα in other cells stimulates PAF synthesis (121–123); however, we are more inclined to favor the first possibility because our experiments with LAU-0901 showed no effect on apoptosis induced by TNFα (118). PAF is also an important player in corneal neovascularization, or angiogenesis, which is a condition that causes loss of corneal transparency.

    View all citing articles on Scopus
    View full text