Next Article in Journal
Antiproliferative, Antimicrobial and Antiviral Activity of β-Aryl-δ-iodo-γ-lactones, Their Effect on Cellular Oxidative Stress Markers and Biological Membranes
Next Article in Special Issue
Lipid Profile of Activated Macrophages and Contribution of Group V Phospholipase A2
Previous Article in Journal
Interrogating Host Antiviral Environments Driven by Nuclear DNA Sensing: A Multiomic Perspective
Previous Article in Special Issue
Sphingomyelinases and Liver Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Representative GIIA Phospholipase A2 Activates Preadipocytes to Produce Inflammatory Mediators Implicated in Obesity Development

1
Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil
2
Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica
3
Faculdade de Ciências Farmacêuticas de Riberão Preto, Universidade de São Paulo, Ribeirão Preto 14040-903, SP, Brazil
*
Authors to whom correspondence should be addressed.
Biomolecules 2020, 10(12), 1593; https://doi.org/10.3390/biom10121593
Submission received: 11 September 2020 / Revised: 15 November 2020 / Accepted: 18 November 2020 / Published: 24 November 2020
(This article belongs to the Special Issue Phospholipases: From Structure to Biological Function)

Abstract

:
Adipose tissue secretes proinflammatory mediators which promote systemic and adipose tissue inflammation seen in obesity. Group IIA (GIIA)-secreted phospholipase A2 (sPLA2) enzymes are found to be elevated in plasma and adipose tissue from obese patients and are active during inflammation, generating proinflammatory mediators, including prostaglandin E2 (PGE2). PGE2 exerts anti-lipolytic actions and increases triacylglycerol levels in adipose tissue. However, the inflammatory actions of GIIA sPLA2s in adipose tissue cells and mechanisms leading to increased PGE2 levels in these cells are unclear. This study investigates the ability of a representative GIIA sPLA2, MT-III, to activate proinflammatory responses in preadipocytes, focusing on the biosynthesis of prostaglandins, adipocytokines and mechanisms involved in these effects. Our results showed that MT-III induced biosynthesis of PGE2, PGI2, MCP-1, IL-6 and gene expression of leptin and adiponectin in preadipocytes. The MT-III-induced PGE2 biosynthesis was dependent on cytosolic PLA2 (cPLA2)-α, cyclooxygenases (COX)-1 and COX-2 pathways and regulated by a positive loop via the EP4 receptor. Moreover, MT-III upregulated COX-2 and microsomal prostaglandin synthase (mPGES)-1 protein expression. MCP-1 biosynthesis induced by MT-III was dependent on the EP4 receptor, while IL-6 biosynthesis was dependent on EP3 receptor engagement by PGE2. These data highlight preadipocytes as targets for GIIA sPLA2s and provide insight into the roles played by this group of sPLA2s in obesity.

1. Introduction

Obesity is a chronic low-grade inflammatory condition in which adipose tissue serves as the source of inflammatory mediators. In obesity and associated diseases, such as diabetes and cardiovascular disease, high plasma and tissue activities of secreted phospholipase A2 (sPLA2) enzymes, especially group IIA (GIIA) sPLA2s, have been demonstrated [1,2].
Phospholipases A2s (PLA2s) are lipolytic enzymes with important physiological functions, including cell membrane remodelling and lipid metabolism. These enzymes are classified according to their cellular localization as either intracellular PLA2 (iPLA2) enzymes with high molecular weight or sPLA2s, enzymes with low molecular weight. sPLA2s hydrolyze glycerophospholipids at the sn-2 position of the glycerol backbone, releasing fatty acids and lysophospholipids in a calcium-dependent manner. sPLA2s are classified into 11 groups and possess, as a common motif, a conserved His-Asp catalytic dyad. Group IIA sPLA2 comprises mammalian sPLA2s found in the inflammatory fluid of mammals and sPLA2s from Viperidae snake venoms. Besides their role in cell membrane physiology, mammalian group IIA sPLA2 are known as important autocrine and paracrine players in inflammatory processes by releasing fatty acids from cell membranes leading to production of pro-inflammatory mediators such as leukotrienes and prostaglandins [3,4,5]. Their role in metabolic diseases, such as obesity, has also been shown [1,4,6]. It is known that inhibition of sPLA2s, using pharmacological intervention, reduced lipid mediator’s synthesis and inflammatory parameters linked to obesity. In this sense, prostaglandin E2 (PGE2) is the most abundant lipid mediator produced by the body. This mediator is constitutively produced in all tissues by the cyclooxygenases (COX) enzymatic system and terminal PGE-synthases [7,8]. PGE2 is a powerful molecule carrying multiple biological effects, which are mediated by four subtypes of G protein-coupled receptors, named EP1, EP2, EP3 and EP4, depending on the tissue or cell type [7,8]. PGE2 is recognized as an important mediator of inflammation, pain and fever. In addition, PGE2 plays important roles in the regulation of proliferation and cell differentiation, and exerts anti-lipolytic actions and increases triacylglycerol levels in adipose tissue cells, contributing to lipid accumulation in these cells [9,10]. However, the molecular mechanisms triggered by GIIA sPLA2s that lead to the biosynthesis of PGE2 by adipose tissue cells are poorly known.
Preadipocytes correspond to a greater cellular fraction present in white adipose tissue and contribute significantly to the production and secretion of inflammatory mediators, such as PGE2 and adipokines, involved in the pathogenesis of obesity [11,12,13,14]. It has been shown that preadipocytes are target cells for a variety of inflammatory factors secreted by macrophages, which are the main cells involved in establishing an inflammatory environment in adipose tissue. In addition, when compared to mature adipocytes, preadipocytes are more responsive to inflammatory stimuli, as they offer a greater activation of the transcription nuclear factor kappa B (NF-kB) and related protein kinases [15]. Therefore, these cells may be used as a cell model for the understanding of the inductors and mechanisms involved in the development of inflammatory processes linked to obesity.
Myotoxin-III (MT-III) is a representative GIIA sPLA2 isolated from Bothrops asper snake venom that shares functional and structural similarities with mammalian pro-inflammatory sPLA2s of the same group [16,17,18]. MT-III is known to trigger inflammatory events in both in vivo and in vitro experimental models. Our group has previously shown that MT-III activates macrophages’ functions and induces the accumulation of lipids into these cells [19]. In addition, this enzyme is able to upregulate the differentiation of macrophages into foam cells [20], which are closely associated with diseases linked to lipid imbalance, including obesity [21,22]. On these bases, in this study, the ability of MT-III to activate proinflammatory responses in preadipocytes focusing on the biosynthesis of lipid mediators, cytokines and adipokines and the mechanisms involved in this process were investigated. In this study, we show for the first time that preadipocytes are target cells for the action of MT-III, a representative GIIA sPLA2, which triggers inflammatory pathways implicated in the development of obesity. The effect of MT-III involves the biosynthesis of PGE2, MCP-1 and IL-6 and gene expression of leptin and adiponectin. PGE2 biosynthesis is dependent upon the activation of cytosolic PLA2 (cPLA2)-α, COX-1, COX-2 and mPGES-1 pathways. EP3 and EP4 receptors play key roles in the release of PGE2 and cytokines.

2. Materials and Methods

2.1. Chemicals and Reagents

(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) MTT and L-glutamine were obtained from USB (Cleveland, OH, USA). Mouse mAb anti-β-actin was purchased from Sigma-Aldrich (St. Louis, MO, USA). The PGE2 enzyme immunoassay kit, Valeryl Salicylate, compounds NS-398, AH6809, AH23848, SC-19220, L-798106 and polyclonal antibodies against COX-1, COX-2, mPGES-1 and the EP4 receptor were purchased from Cayman Chemical Company (Ann Arbor, MI, USA). Pyrrolidine-2 (Pyr-2) was purchased from Calbiochem-Novabiochem Corp. (La Jolla, CA, USA). Secondary antibodies, anti-mouse and anti-rabbit, conjugated to HRP and nitrocellulose membrane, were obtained from GE Healthcare (Buckinghamshire, UK). The Cytometric Bead Assay (CBA) kit was purchased from BD Bioscience (San Jose, CA, USA). Gentamicin was purchased from Schering-Plough (Whitehouse Station, NJ, USA), DMSO from Amresco (Solon, OH, USA) and Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum and real-time polymerase chain reaction (PCR) assay kit from Life Technologies (São Paulo, SP, Brazil).

2.2. Phospholipase A2 (PLA2)

Aspartate-49 sPLA2, named MT-III (Uniprot accession no.: P20474), from B. asper venom was purified by ion-exchange chromatography on CM Sephadex C-25 using a KCl gradient from 0 to 0.75 M at pH 7.0 as described [23], followed by RP-HPLC on a semipreparative C8 column (Vydac; 106,250 mm, 5 mm particle size), eluted at a flow rate of 2.5 mL/min with a gradient of acetonitrile (0–70%, containing 0.1% trifluoroacetic acid) over 30 min. Homogeneity was verified by SDS-PAGE, run under reducing conditions, in which a single band of 14 kDa was observed. The complete amino acid sequence of this enzyme has been described previously [23,24]. The absence of endotoxin contamination in the MT-III batches used was demonstrated by a quantitative LAL test [25], which revealed undetectable levels of endotoxin (0.125 EU/mL).

2.3. Cytotoxicity Assay

The cytotoxicity of MT-III and toward the 3T3-L1 preadipocyte was evaluated using the MTT assay previously described [19]. In brief, 4 × 103 preadipocytes per well in DMEM, supplemented with 40 μg/mL gentamicin sulfate and 2 mM L-glutamine, were plated in 96-well plates and incubated with MT-III (0.4 μM), COX inhibitors or PGE2 antagonist receptors, diluted in medium or with the same volume of medium alone (control) for 1, 3, 6, 12, 24 and 48 h at 37 °C in a humidified atmosphere (5% CO2). MTT (5 mg/mL) was dissolved in PBS and filtered for sterilization and removal of insoluble residues. Stock MTT solution (10% in culture medium) was added to all wells in each assay, and plates were incubated for 3 h at 37 °C. Dimethyl sulfoxide (DMSO) (100 μL) was added to all wells and mixed thoroughly for 30 min, at room temperature. Absorbances were then recorded in a microtiter plate reader, at 540 nm. Results were expressed as percentages of viable cells, considering control cells incubated with medium alone as 100% viable.

2.4. 3T3-L1 Cell Culture and Stimulation

3T3-L1 preadipocytes obtained from the American Type Culture Collection were cultured as described [26]. Cells were processed according to the experimental protocol, in which 5 × 103 preadipocytes per well were seeded in 12-wells culture plates and maintained in culture medium for 48 h before stimulation. Preadipocytes were serum-starved in DMEM with 1% (v/v) gentamicin sulfate supplemented with 1% (v/v) L-glutamine for 18 h prior to all treatments. Cellular homogenates were used for the Western blotting analysis of COX-1, COX-2, EP1–EP4 receptors and mPGES-1 protein expression, and supernatants of each treatment were used to measure lipid mediators PGE2, PGI2, LTB4 and TXA2 by Enzyme Immunoassay (EIA) and cytokines MCP-1, IL-6, IL-10, IL-12, TNF-α and INF-γ by CBA. Cells were stimulated with MT-III (0.4 mM) diluted in DMEM (serum free) or DMEM alone (control) for selected periods of time and maintained at 37 °C in a humidified atmosphere (5% CO2). To investigate the mechanism involved in the PGE2 and cytokine biosynthesis, selective inhibitors or antagonists were used at concentrations previously tested: 10 μM valeryl salicylate (COX-1 inhibitor) and NS-398 (COX-2 inhibitor); 10 μM SC-19220 (EP1 receptor antagonist); AH6809 (EP2 receptor antagonist) and AH23848 (EP4 receptor antagonist); and 1 μM L-798106 (EP3 receptor antagonist) [27,28,29,30,31]. All of the stock solutions were prepared in DMSO and stored at −20 °C. Aliquots were diluted in DMEM immediately before use. DMSO concentration was always lower than 1%. The viability of cells treated with inhibitors or antagonists was evaluated with MTT assay. No significant changes in cell viability were registered with any of the above agents or the vehicle at the concentrations used (data not shown).

2.5. Western Blotting

COX-1, COX-2, EP1–EP4 receptors and mPGES-1 protein expression from homogenate cells were detected by Western blotting. Briefly, MT-III-stimulated and non-stimulated cells were lysed with 100 mL of a sample buffer (0.5 M Tris-HCl, pH 6.8, 20% SDS, 1% glycerol, 1 M β-mercaptoethanol, 0.1% bromophenol blue) and boiled for 10 min. Samples were resolved by SDS-PAGE on 10% bis-acrylamide gels overlaid with a 5% stacking gel. Proteins were then transferred to nitrocellulose membranes using a Mini Trans-Blot (Bio-Rad Laboratories, Richmond, CA, USA). Membranes were blocked for 1 h with 5% albumin in Tris-buffered saline (20 mM Tris, 100 mM NaCl and 0.5% Tween 20, pH 7.2) and incubated overnight with primary antibodies against COX-1 and COX-2; EP1, EP2, EP3, and EP4 receptors; and mPGES-1 (1:500 dilution) or β-actin (1:3000 dilution) for 1 h at room temperature. Membranes were then washed and incubated with the appropriate secondary antibody conjugated to horseradish peroxidase. Immunoreactive bands were detected by the entry-level peroxidase substrate for enhanced chemiluminescence, according to the instructions of the manufacturer (GE Healthcare). Band densities were quantified with an ImageQuant LAS 4000 mini densitometer (GE Healthcare) using the image analysis software ImageQuant TL (GE Healthcare).

2.6. Eicosanoid and Cytokines Quantification

PGE2, PGI2, LTB4 and TXA2 were measured using an EIA kit, while cytokines (MCP-1, IL-6, IL-10, IL-12, TNF-α, INF-γ) were quantified using a CBA kit from supernatants of preadipocytes incubated with each treatment. Kits were used according to the instructions of the manufacturer.

2.7. Adipocytokines Expression by Quantitative Real-Time PCR

Quantitative polymerase chain reaction was performed as described [32]. Briefly, the total RNA from preadipocytes, incubated MT-III or DMEM alone (control) for 1, 3 and 6 h was isolated using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and reverse transcription with oligo (dT) priming was performed from 2 μg of total RNA using Superscript III (Invitrogen, Carlsbad, CA, USA). The relative expression of each transcript was determined by quantitative real-time PCR in an ABI 7000 Sequence Detection System (Applied Biosystems, Forrest City, CA, USA). Each well of the 96-well reaction plate contained a total volume of 25 µL of Power SYBR Green PCR Master Mix (Applied Biosystems). The threshold cycle (Ct) was used to determine the relative expression level of each gene by normalizing to the Ct of Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH). The method of delta–delta cycle threshold (ddCt) was used to calculate the relative fold change of each gene. Data are represented as mean + SEM.

2.8. Statistical Analysis

Data are expressed as mean ± SEM (n = 4). Multiple comparisons among groups were performed using the one-way ANOVA and, as a post-test, the Bonferroni test. Differences between experimental groups were considered significant for p-values < 0.05. All statistical tests were performed using Prism version 5 software (GraphPad, San Diego, CA, USA).

3. Results

3.1. MT-III Induces the Release of Lipid Mediators by Preadipocytes

Lipid mediators are involved in lipid abnormalities and contribute to the triggering of inflammatory processes in adipose tissue [33]. Therefore, we investigated the ability of MT-III to induce the release of lipid mediators linked to inflammatory processes, such as PGE2, PGI2, TXA2 and LTB4, by cultured preadipocytes. From preliminary studies (data not shown), the submaximal concentration of 0.4 µM of MT-III was chosen for these studies as it would allow potential inhibition or exacerbation of its effects by drug treatment to be detected. As shown in Figure 1, the incubation of preadipocytes with MT-III induced a significant release of PGE2 (A) from 1 to 24 h and of PGI2 (B) from 12 to 48 h when compared with controls. However, the incubation of cells with MT-III did not alter TXA2 (C) or LTB4 (D) levels in any of the time periods evaluated. These results indicate the ability of MT-III to activate preadipocytes for the production of PGE2 and PGI2.

3.2. MT-III-Induced Release of PGE2 Is Dependent on COX-1 and COX-2 in Preadipocytes

COX-1 and COX-2 are enzymes responsible for the metabolization of arachidonic acid–generating prostanoids, such as PGE2 [8,34]. In order to verify the mechanism involved in the MT-III-induced biosynthesis of PGE2, we investigated the participation of COX-1 and COX-2 in this effect. As seen in Figure 2A, preadipocytes incubated with MT-III, in the presence of vehicle (DMSO), showed a significant release of PGE2 after 6 h when compared with controls. Preadipocytes treated either with COX-1 inhibitor (valerylsalicylate) or COX-2 inhibitor (NS-398) before the MT-III stimulus showed a reduction in PGE2 release which was statistically significant when compared to the positive control. Treatment of cells with both valerylsalicylate and NS-398 compounds abolished the MT-III-induced release of PGE2 when compared to the positive control. These results indicate that COX-1 and COX-2 are key enzymes involved in the release of PGE2, induced by MT-III, in preadipocytes. Having shown that both COX isoforms participate in the signalling pathway triggered by MT-III that leads to PGE2 production, we next investigated whether MT-III is able to upregulate the protein expression of COX-1 and COX-2 in preadipocytes. Our results show that preadipocytes constitutively expressed both isoforms of COX. Figure 2B,C show that COX-1 protein expression did not differ significantly between control cells and cells treated with MT-III. However, the protein expression of COX-2 was higher in cells incubated with the phospholipase A2 after 6 and 12 h (Figure 2D,E). Therefore, although the COX-2 isoform is constitutively expressed by preadipocytes [35], our results show that MT-III upregulates the protein expression of COX-2 but not COX-1 in preadipocytes.

3.3. MT-III Upregulates Protein Expression of mPGES-1 by Preadipocytes

An inducible synthase responsible for the terminal synthesis of PGE2, mPGES-1 is upregulated in inflammatory conditions [36,37]. Based on this, we evaluated the ability of MT-III to upregulate the protein expression of this enzyme in preadipocytes. Our results show that MT-III upregulated the protein expression of mPGES-1 after 1 h of stimulation when compared with the control (Figure 3). The phospholipase A2 did not alter the protein expression of mPGES-1 at other time intervals evaluated. These results demonstrate that MT-III induces protein expression of mPGES-1 in preadipocytes.

3.4. MT-III-Induced Release of PGE2 Is Dependent on Cytosolic PLA2-α in Preadipocytes

It is known that sPLA2s cross-talk with cPLA2 for the synthesis of inflammatory mediators [38,39,40]. Based on this, we investigated the participation of cPLA2-α in the release of PGE2 induced by MT-III. Our results show that preadipocytes incubated with MT-III, in the presence of vehicle (positive cotrol), showed a significant release of PGE2 after 3 h when compared with respective control. However, pre-treatment of cells with cPLA2-α inhibitor (Pyr-2) abrogated the MT-III-induced release of PGE2 when compared to the positive control (Figure 4). These results indicate that the MT-III-induced production of PGE2 is dependent on cPLA2-α in preadipocytes.

3.5. MT-III-Induced Release of PGE2 Is Dependent on the EP4 Receptor in Preadipocytes

PGE2 exerts its effects through activation of four subtypes of G protein-coupled receptors, named EP1, EP2, EP3 and EP4, and these receptors are able to regulate PGE2 biosynthesis [34,37,41]. It is known that the activation of the EP4 receptor by PGE2 may lead to the increased expression of key enzymes of biosynthesis cascade of this prostaglandin, such as COX-2 and mPGES-1 [8,41,42]. Therefore, we investigated whether PGE2 biosynthesis, induced by MT-III, was dependent on the activation of these receptors. As shown in Figure 5A, the incubation of preadipocytes with DMEM plus vehicle or antagonists did not cause a significant release of PGE2 after 6 h of incubation. Pre-treatment of cells with DMEM plus vehicle followed by incubation with MT-III (0.4 μM), for the same time period, induced a significant increase in PGE2 release, relative to baseline control. However, pre-treatment of cells with antagonists of EP1 (SC-19220), EP2 (AH6809) or EP3 (L-798106) receptors did not alter MT-III-induced PGE2 release when compared with controls. In contrast, pre-treatment of cells with the EP4 receptor antagonist (AH23848) abolished the MT-III-induced release of PGE2 when compared to the positive control. To better understand the involvement of the EP receptors in the effects induced by MT-III, we next analysed the protein expression of these receptors in preadipocytes stimulated with MT-III and in control cells incubated with culture medium alone. Our results show that there was no alteration in the protein expression of EP receptors in preadipocytes incubated with MT-III (0.4 μM) in any of the time periods evaluated when compared with controls (Figure 5B–I). These results indicate that PGE2 biosynthesis, induced by MT-III, in preadipocytes is dependent on the engagement of the EP4 receptor by PGE2, but not on increased protein expression of this receptor.

3.6. MT-III Induces Release of Inflammatory Cytokines by Preadipocytes

Inflammatory cytokines are found in high levels in obesity inflammatory processes and contribute to the development and maintenance of this inflammatory state [43,44,45]. On these bases, we investigated the capacity of MT-III to induce the release of the inflammatory cytokines MCP-1, IL-6, IL-10, IL-12, TNF-α and IFN-γ by preadipocytes. Figure 6A shows that MT-III induced significant release of MCP-1 from 30 min up to 24 h of incubation when compared with controls. In addition, MT-III induced significant release of IL-6 after 12 h of incubation when compared to the respective controls (Figure 6B). However, MT-III did not alter the release of IL-10, IL-12, TNF-α or IFN-γ (data not shown). In this sense, these results evidence the capacity of MT-III to induce the release of MCP-1 and IL-6 by 3T3-L1 in preadipocytes.

3.7. EP3 and EP4 Receptors Participate in the MT-III-Induced Release of IL-6 and MCP-1 by Preadipocytes

Previous studies have shown that EP3 and EP4 PGE2 receptors regulate the release of proinflammatory cytokines [46,47]. Therefore, we investigated the participation of EP3 and EP4 receptors in the MT-III-induced release of IL-6 and MCP-1, respectively, by preadipocytes. Figure 7A shows that the stimulation of preadipocytes with MT-III, in the presence of vehicle, significantly increased MCP-1 release after 24 h when compared with the control. Pre-treatment of preadipocytes with the EP4 antagonist (AH238481) significantly reduced the release of MCP-1 in cells stimulated with MT-III in comparison with the positive control. Similarly, pre-treatment of cells with the EP3 antagonist (L-798106) reduced the MT-III-induced release of IL-6 after 12 h, which was significant in comparison with the positive control (Figure 7B). These results indicate that EP3 and EP4 receptors participate in the release of IL-6 and MCP-1, respectively, in preadipocytes stimulated with MT-III.

3.8. MT-III Upregulates Gene Expression of Adipokines in Preadipocytes

Adipokines are produced by white adipose tissue and are involved in a wide variety of physiological and pathological processes. Proinflammatory adipokines contribute to the development and maintenance of the inflammatory state in obese individuals [35,48,49]. In light of this, we investigated the ability of MT-III to induce gene expression of the adipokines leptin, resistin and adiponectin by preadipocytes. As demonstrated in Figure 8A, preadipocytes incubated with MT-III showed a significant increase in the gene expression of leptin from 1 to 3 h when compared with controls. In addition, preadipocytes incubated with MT-III showed a significant increase in the gene expression of adiponectin after 3 h (Figure 8B). However, the phospholipase A2 did not affect the gene expression of resistin in any of the time periods evaluated (Figure 8C). These results indicate that preadipocytes can respond to MT-III with the production of leptin and adiponectin but not resistin.

4. Discussion

Levels of sPLA2 are elevated in the serum of obese patients as well as in inflamed fat tissue [1,2,50,51]. Previous studies have implicated sPLA2s in metabolic diseases, including obesity [1,4,6]. However, the direct effects and mechanisms triggered by this class of enzymes on adipose tissue cells are not completely known. We herein report the ability of MT-III, a representative GIIA sPLA2, to activate proinflammatory pathways in preadipocytes.
Prostanoids are produced from the metabolism of arachidonic acid by the cyclooxygenases system (COX-1 and COX-2) and are implicated in events related to the development of obesity, including inflammation and the differentiation of preadipocytes into mature adipocytes [33,52]. Our results demonstrate that MT-III induced an early and sustained release of PGE2, followed by a late release of PGI2. Taking into account the marked biosynthesis of PGE2 in preadipocytes stimulated by MT-III and the contribution of this mediator to the inflammation process in the adipose tissue, the mechanisms involved in PGE2 biosynthesis, induced by MT-III, were investigated. Our findings with a pharmacological approach indicated that PGE2 production induced by MT-III is dependent upon the activation of COX-1 and COX-2 in preadipocytes. As an additional mechanism, MT-III upregulated COX-2 protein expression, but not COX-1 protein expression in preadipocytes. To our knowledge, this is the first demonstration that a GIIA sPLA2 directly activates PGE2 biosynthesis in preadipocytes, the precursor cells of mature adipocytes. Furthermore, our data evidence preadipocytes as target cells for GIIA sPLA2 action.
PGE2 synthases, including mPGES-1, mPGES-2 and cPGES, participate in the terminal step of PGE2 biosynthesis by converting PGH2 into PGE2 [53]. In contrast to mPGES-2 and cPGES, the mPGES-1 isoform is upregulated in inflammatory conditions [54,55]. During obesity, upregulation of mPGES-1 expression has been shown in the adipose tissue during adipogenic processes [56]. Accordingly, we found that MT-III increased the expression of mPGES-1 in preadipocytes. The early release of PGE2 correlated with mPGES-1 expression, indicating the participation of this terminal synthase in PGE2 biosynthetic cascade triggered by MT-III, probably via functional coupling of COX-1 and mPGES-1 in the early stage of PLA2 stimulation. These data reinforce the ability of GIIA sPLA2s to activate mechanisms in preadipocytes that contribute to the development of obesity.
It is now well recognized that mammalian GIIA sPLA2s do not exert their biological actions through their catalytic mechanism alone [57]. Several reports evidence that human GIIA sPLA2s lead to eicosanoid production by means other than by directly providing arachidonic acid (AA) through catalysis. Among the non-catalytic mechanisms described is the crosstalk between mammalian GIIA sPLA2 and cPLA2 [58,59,60,61,62]. Several lines of evidence point out that the high AA specificity of cPLA2-α and the lack of fatty acid selectivity in sPLA2s can be combined to achieve specific cellular responses [38,39,40]. In this context, our finding that inhibition of the cytosolic (cPLA2)-α by compound Pyr-2 abrogated the release of PGE2 induced by MT-III indicates that the cPLA2-α is a crucial partner for the effect triggered by MT-III in preadipocytes. This finding is in line with our previous data showing that MT-III increased phosphorylation of cPLA2-α at Ser505, a hallmark of cPLA2-alpha activation, in human monocytes [63]. Furthermore, although MT-III has the ability to release arachidonic acid from membrane phosphatidylcholine [63], our results evidence that the catalytic activity of MT-III does not play a role in production of PGE2 in preadipocytes. A similar mechanism is widely accepted for mammalian GIIA sPLA2s [57].
We further extended our knowledge of the mechanisms involved in the generation of PGE2 induced by MT-III by focusing on the participation of the EP4 receptor, which was shown to regulate the expression of key enzymes involved in PGE2 biosynthesis, including COX-2 and PGESm-1 [2,41,52,64,65,66]. Our results, showing that EP4 antagonism by compound AH23848 abolished PGE2 release induced by MT-III, indicate a critical role of this receptor in the effect of MT-III. These results strongly suggest that engagement of the EP4 receptor by PGE2 triggers a positive feedback loop regulating the biosynthetic cascade of this mediator in preadipocytes stimulated by MT-III. Activation of this positive loop likely contributes to increased levels of PGE2 observed throughout the period of stimulation with MT-III. In accordance with our data, studies using siRNA for knockdown EP4 gene in macrophages have shown reduced COX-2 expression upon stimuli by lipopolysaccharide [64]. In addition, our data evidenced a late release of PGI2, which is considered a biomarker of adipocyte differentiation [67,68], in cells stimulated by MT-III. This suggests the involvement of GIIA sPLA2s in the differentiation of preadipocytes. Although not investigated in this study, this hypothesis is currently being investigated in our laboratory.
Development of inflammation in the adipose tissue involves an early migration of leukocytes, mainly monocytes, into this tissue, followed by the secretion of several pro-inflammatory mediators by these cells, including cytokines, thus establishing an inflammatory environment [69,70,71,72]. Our findings showing a long-lasting release of MCP-1 in preadipocytes stimulated by MT-III strongly suggest that GIIA sPLA2s are implicated in the infiltration of monocytes and macrophages into adipose tissue and contribute to an inflammatory response in this tissue since MCP-1 is the key chemoattractant for monocytes during inflammatory conditions [70,72,73].
In addition, the release of IL-6 seen in preadipocytes stimulated by MT-III may contribute to the establishment of an inflammatory environment in the adipose tissue. These findings are in accordance with previous reports that levels of IL-6 are elevated in inflamed adipose tissue of obese patients that was associated with the induction of insulin resistance [74,75,76]. Furthermore, using pharmacological interference, we found that the MT-III-induced release of MCP-1 and IL-6 was dependent on EP4 or EP3 activation, respectively, in preadipocytes stimulated by MT-III. In view of previous evidence that the engagement of distinct EP receptors by PGE2 triggers signalling pathways linked to the biosynthesis of proinflammatory cytokines [77,78], our findings indicate that PGE2 biosynthesis, induced by MT-III, is an essential step for the activation of proinflammatory pathways linked to cytokine production in preadipocytes stimulated by the phospholipase A2.
Adipose tissue produces specific cytokines known as adipokines, which are pivotal mediators that maintain a low-grade inflammation, which characterizes obesity [79,80]. These mediators have been described as exerting autocrine and paracrine effects and regulating appetite and satiety, glucose and lipid metabolism, blood pressure regulation, inflammation and immune functions [81,82,83]. Accordingly, we found that MT-III upregulated the expression of the adipokines leptin and adiponectin in preadipocytes. Previous reports have demonstrated that leptin is able to stimulate the production of proinflammatory cytokines by macrophages and expression of adhesion molecules by endothelial cells, thus contributing to the development of the inflammatory process in the adipose tissue [84]. Therefore, this mediator may be critical for the inflammatory effects triggered by MT-III in adipose tissue by promoting key inflammatory events. Moreover, in light of the modulatory effects of adiponectin in biological systems and inflammation [85], our findings suggest that this mediator may control the inflammatory response induced by MT-III leading to a low-grade inflammation environment, which characterizes obesity. In contrast, MT-III did not affect resistin expression in preadipocytes. This may be due to the predominance of mature adipocytes over preadipocytes for the production of this mediator [86]. Although the mechanisms related to the release of adipokines by MT-III have not been presently investigated, participation of PGE2 and MCP-1 in the expression of leptin can be suggested since PGE2 and MCP-1 have been described as activators of signalling pathways leading to leptin biosynthesis [87,88]. To the best of our knowledge, this is the first demonstration that a GIIA PLA2 has the ability to induce the expression of adipokines in preadipocytes.

5. Conclusions

In this study, we demonstrate for the first time the ability of a representative GIIA phospholipase A2, MT-III, to directly activate preadipocytes to release PGE2 and the critical role of this mediator, acting via receptors EP3 and EP4, in inflammatory responses induced by this sPLA2. MT-III also induced release of PGI2, MCP-1 and IL-6 but not TNF-α, INF-γ, IL-12 or IL-10, and upregulated the expression of leptin and adiponectin. The MT-III-induced PGE2 biosynthesis was dependent on the activation of cPLA2-α, COX-1 and COX-2 pathways and positively regulated by the EP4 receptor. As an additional mechanism, MT-III upregulated COX-2 and mPGES-1 protein expression. MCP-1 biosynthesis induced by this sPLA2 was dependent on the activation of the EP4 receptor, while IL-6 biosynthesis was dependent on the EP3 receptor in preadipocytes. Taken together, these findings provide evidence of a new target cell of the action of GIIA sPLA2s, extending the knowledge of the effect of this class of enzymes in the adipose tissue (Scheme 1) given new insights into the roles of GIIA sPLA2s in obesity and associated disorders.

Author Contributions

Conceptualization: E.L. and C.T.; methodology, E.L., P.M., R.M.M.; formal analysis, E.L., P.M., R.M.M.; investigation, E.L., P.M., R.M.M.; B.L. isolated and purified MT-III; writing—original draft preparation, E.L., P.M., R.M.M. and C.T.; writing—review and editing, E.L., P.M., R.M.M. and C.T.; supervision, C.T.; project administration, S.V.S. and C.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), grant numbers 2011/23236-4, 2015/24701-3, 2017/197339.

Acknowledgments

The authors thank Renata Hage do Amaral Hernandez for providing technical assistance.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Garces, F.; López, F.; Niño, C.; Fernandez, A.; Chacin, L.; Hurt-Camejo, E.; Camejo, G.; Apitz-Castro, R. High Plasma Phospholipase A2 Activity, Inflammation Markers, and LDL Alterations in Obesity with or Without Type 2 Diabetes. Obesity 2010, 18, 2023–2029. [Google Scholar] [CrossRef] [PubMed]
  2. Dutour, A.; Achard, V.; Sell, H.; Naour, N.; Collart, F.; Gaborit, B.; Silaghi, A.; Eckel, J.; Alessi, M.-C.; Henegar, C.; et al. Secretory Type II Phospholipase A2 Is Produced and Secreted by Epicardial Adipose Tissue and Overexpressed in Patients with Coronary Artery Disease. J. Clin. Endocrinol. Metab. 2010, 95, 963–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Murakami, M.; Taketomi, Y.; Miki, Y.; Sato, H.; Yamamoto, K.; Lambeau, G. Emerging roles of secreted phospholipase A2 enzymes: The 3rd edition. Biochimie 2014, 107, 105–113. [Google Scholar] [CrossRef] [PubMed]
  4. Murakami, M.; Taketomi, Y.; Sato, H.; Yamamoto, K. Secreted phospholipase A2 revisited. J. Biochem. 2011, 150, 233–255. [Google Scholar] [CrossRef] [Green Version]
  5. Murakami, M.; Taketomi, Y. Secreted phospholipase A 2 and mast cells. Allergol. Int. 2015, 64, 4–10. [Google Scholar] [CrossRef] [Green Version]
  6. Iyer, A.; Lim, J.; Poudyal, H.; Reid, R.C.; Suen, J.Y.; Webster, J.; Prins, J.B.; Whitehead, J.P.; Fairlie, D.P.; Brown, L. An Inhibitor of Phospholipase A2 Group IIA Modulates Adipocyte Signaling and Protects Against Diet-Induced Metabolic Syndrome in Rats. Diabetes 2012, 61, 2320–2329. [Google Scholar] [CrossRef] [Green Version]
  7. Jacob, P.J.; Manju, S.L.; Ethiraj, K.R.; Elias, G. Safer anti-inflammatory therapy through dual COX-2/5-LOX inhibitors: A structure-based approach. Eur. J. Pharm. Sci. 2018, 121, 356–381. [Google Scholar] [CrossRef]
  8. Park, J.Y.; Pillinger, M.H.; Abramson, S.B. Prostaglandin E2 synthesis and secretion: The role of PGE2 synthases. Clin. Immunol. 2006, 119, 229–240. [Google Scholar] [CrossRef]
  9. Pérez, S.; Aspichueta, P.; Ochoa, B.; Chico, Y.; Ochoa, B. The 2-series prostaglandins suppress VLDL secretion in an inflammatory condition-dependent manner in primary rat hepatocytes. Biochim. Et Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2006, 1761, 160–171. [Google Scholar] [CrossRef]
  10. Enomoto, N.; Ikejima, K.; Yamashina, S.; Enomoto, A.; Nishiura, T.; Nishimura, T.; Brenner, D.A.; Schemmer, P.; Bradford, B.U.; Rivera, C.A.; et al. Kupffer cell-derived prostaglandin E2is involved in alcohol-induced fat accumulation in rat liver. Am. J. Physiol. Liver Physiol. 2000, 279, G100–G106. [Google Scholar] [CrossRef] [Green Version]
  11. O’Hara, A.; Lim, F.-L.; Mazzatti, D.J.; Trayhurn, P. Stimulation of inflammatory gene expression in human preadipocytes by macrophage-conditioned medium: Upregulation of IL-6 production by macrophage-derived IL-1β. Mol. Cell. Endocrinol. 2012, 349, 239–247. [Google Scholar] [CrossRef] [PubMed]
  12. Wood, I.; Trayhurn, P. Signalling role of adipose tissue: Adipokines and inflammation in obesity. Biochem. Soc. Trans. 2005, 33, 1078. [Google Scholar] [CrossRef]
  13. Mafra, D.; Mafra, D. Adipokines in obesity. Clin. Chim. Acta 2013, 419, 87–94. [Google Scholar] [CrossRef]
  14. Karastergiou, K.; Mohamed-Ali, V. The autocrine and paracrine roles of adipokines. Mol. Cell. Endocrinol. 2010, 318, 69–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chung, S.; LaPoint, K.; Martinez, K.; Kennedy, A.; Sandberg, M.B.; McIntosh, M.K. Preadipocytes Mediate Lipopolysaccharide-Induced Inflammation and Insulin Resistance in Primary Cultures of Newly Differentiated Human Adipocytes. Endocrinology 2006, 147, 5340–5351. [Google Scholar] [CrossRef] [PubMed]
  16. Davidson, F.F.; Dennis, E.A. Evolutionary relationships and implications for the regulation of phospholipase A2 from snake venom to human secreted forms. J. Mol. Evol. 1990, 31, 228–238. [Google Scholar] [CrossRef] [PubMed]
  17. Gutiérrez, J.M.; Lomonte, B. Phospholipase A2 myotoxins from Bothrops snake venoms. Toxicon 1995, 33, 1405–1424. [Google Scholar] [CrossRef]
  18. Schaloske, R.H.; Dennis, E.A. The phospholipase A2 superfamily and its group numbering system. Biochim. Et Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2006, 1761, 1246–1259. [Google Scholar] [CrossRef]
  19. Leiguez, E.; Zuliani, J.P.; Cianciarullo, A.M.; Fernandes, C.M.; Gutiérrez, J.M.; Teixeira, C.D.F.P. A group IIA-secreted phospholipase A2 from snake venom induces lipid body formation in macrophages: The roles of intracellular phospholipases A2 and distinct signaling pathways. J. Leukoc. Biol. 2011, 90, 155–166. [Google Scholar] [CrossRef]
  20. Leiguez, E.; Giannotti, K.C.; Viana, M.D.N.; Matsubara, M.H.; Fernandes, C.M.; Gutiérrez, J.M.; Lomonte, B.; Teixeira, C. A Snake Venom-Secreted Phospholipase A2Induces Foam Cell Formation Depending on the Activation of Factors Involved in Lipid Homeostasis. Mediat. Inflamm. 2018, 2018, 1–13. [Google Scholar] [CrossRef] [Green Version]
  21. Xie, Z.; Wang, X.; Liu, X.; Du, H.; Sun, C.; Shao, X.; Tian, J.; Gu, X.; Wang, H.; Tian, J.; et al. Adipose-Derived Exosomes Exert Proatherogenic Effects by Regulating Macrophage Foam Cell Formation and Polarization. J. Am. Heart Assoc. 2018, 7, e007442. [Google Scholar] [CrossRef] [PubMed]
  22. Shapiro, H.; Pecht, T.; Shaco-Levy, R.; Harman-Boehm, I.; Kirshtein, B.; Kuperman, Y.; Chen, A.; Blüher, M.; Shai, I.; Rudich, A. Adipose Tissue Foam Cells Are Present in Human Obesity. J. Clin. Endocrinol. Metab. 2013, 98, 1173–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kaiser, I.I.; Gutiérrez, J.M.; Plummer, D.; Aird, S.D.; Odell, G.V. The amino acid sequence of a myotoxic phospholipase from the venom of Bothrops asper. Arch. Biochem. Biophys. 1990, 278, 319–325. [Google Scholar] [CrossRef]
  24. Díaz-Oreiro, C.; Gutiérrez, J.M. Chemical modification of histidine and lysine residues of myotoxic phospholipases A2 isolated from Bothrops asper and Bothrops godmani snake venoms: Effects on enzymatic and pharmacological properties. Toxicon 1997, 35, 241–252. [Google Scholar] [CrossRef]
  25. Takayama, K.; Mitchell, D.H.; Din, Z.Z.; Mukerjee, P.; Li, C.; Coleman, D.L. Monomeric Re lipopolysaccharide from Escherichia coli is more active than the aggregated form in the Limulus amebocyte lysate assay and in inducing Egr-1 mRNA in murine peritoneal macrophages. J. Biol. Chem. 1994, 269, 2241–2244. [Google Scholar]
  26. Dordevic, A.L.; Konstantopoulos, N.; Cameron-Smith, D. 3T3-L1 Preadipocytes Exhibit Heightened Monocyte-Chemoattractant Protein-1 Response to Acute Fatty Acid Exposure. PLoS ONE 2014, 9, e99382. [Google Scholar] [CrossRef] [Green Version]
  27. Chang, Y.-H.; Lee, S.T.; Lin, W.-W. Effects of cannabinoids on LPS-stimulated inflammatory mediator release from macrophages: Involvement of eicosanoids. J. Cell. Biochem. 2001, 81, 715–723. [Google Scholar] [CrossRef]
  28. Choi, H.C.; Kim, H.S.; Lee, K.Y.; Chang, K.C.; Kang, Y.J. NS-398, a selective COX-2 inhibitor, inhibits proliferation of IL-1β-stimulated vascular smooth muscle cells by induction of HO-1. Biochem. Biophys. Res. Commun. 2008, 376, 753–757. [Google Scholar] [CrossRef]
  29. Lin, Y.-S.; Hsieh, M.; Lee, Y.-J.; Liu, K.-L.; Lin, T.-H. AH23848 accelerates inducible nitric oxide synthase degradation through attenuation of cAMP signaling in glomerular mesangial cells. Nitric Oxide 2008, 18, 93–104. [Google Scholar] [CrossRef]
  30. Lin, C.-C.; Lin, W.-N.; Wang, W.-J.; Sun, C.-C.; Tung, W.-H.; Wang, H.-H.; Yang, C.-M. Functional coupling expression of COX-2 and cPLA2 induced by ATP in rat vascular smooth muscle cells: Role of ERK1/2, p38 MAPK, and NF-κB. Cardiovasc. Res. 2009, 82, 522–531. [Google Scholar] [CrossRef] [Green Version]
  31. Chen, L.; Miao, Y.; Zhang, Y.; Dou, D.; Liu, L.; Tian, X.; Yang, G.; Pu, D.; Zhang, X.; Kang, J.; et al. Inactivation of the E-Prostanoid 3 Receptor Attenuates the Angiotensin II Pressor Response via Decreasing Arterial Contractility. Arter. Thromb. Vasc. Biol. 2012, 32, 3024–3032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ratchford, A.M.; Esguerra, C.R.; Moley, K.H. Decreased Oocyte-Granulosa Cell Gap Junction Communication and Connexin Expression in a Type 1 Diabetic Mouse Model. Mol. Endocrinol. 2008, 22, 2643–2654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Iyer, A.; Fairlie, D.P.; Prins, J.B.; Hammock, B.D.; Brown, L. Inflammatory lipid mediators in adipocyte function and obesity. Nat. Rev. Endocrinol. 2010, 6, 71–82. [Google Scholar] [CrossRef] [PubMed]
  34. Kawahara, K.; Hohjoh, H.; Inazumi, T.; Tsuchiya, S.; Sugimoto, Y. Prostaglandin E2-induced inflammation: Relevance of prostaglandin E receptors. Biochim. Et Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2015, 1851, 414–421. [Google Scholar] [CrossRef] [PubMed]
  35. Yan, H.; Kermouni, A.; Abdel-Hafez, M.; Lau, D.C. Role of cyclooxygenases COX-1 and COX-2 in modulating adipogenesis in 3T3-L1 cells. J. Lipid Res. 2003, 44, 424–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Yang, G.; Chen, L. An Update of Microsomal Prostaglandin E Synthase-1 and PGE2Receptors in Cardiovascular Health and Diseases. Oxid. Med. Cell. Longev. 2016, 2016, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. McCoy, J.M.; Wicks, J.R.; Audoly, L.P. The role of prostaglandin E2 receptors in the pathogenesis of rheumatoid arthritis. J. Clin. Investig. 2002, 110, 651–658. [Google Scholar] [CrossRef]
  38. Dennis, E.A.; Cao, J.; Hsu, Y.-H.; Magrioti, V.; Kokotos, G. Phospholipase A2Enzymes: Physical Structure, Biological Function, Disease Implication, Chemical Inhibition, and Therapeutic Intervention. Chem. Rev. 2011, 111, 6130–6185. [Google Scholar] [CrossRef] [Green Version]
  39. Balsinde, J.; Winstead, M.V.; A Dennis, E. Phospholipase A2 regulation of arachidonic acid mobilization. FEBS Lett. 2002, 531, 2–6. [Google Scholar] [CrossRef]
  40. Lambeau, G.; Gelb, M.H. Biochemistry and Physiology of Mammalian Secreted Phospholipases A2. Annu. Rev. Biochem. 2008, 77, 495–520. [Google Scholar] [CrossRef] [Green Version]
  41. Viana, M.N.; Leiguez, E.; Gutiérrez, J.M.; Rucavado, A.; Markus, R.P.; Marçola, M.; Teixeira, C.; Fernandes, C.M. A representative metalloprotease induces PGE2 synthesis in fibroblast-like synoviocytes via the NF-κB/COX-2 pathway with amplification by IL-1β and the EP4 receptor. Sci. Rep. 2020, 10, 3269. [Google Scholar] [CrossRef] [PubMed]
  42. Fantuzzi, G. Adipose tissue, adipokines, and inflammation. J. Allergy Clin. Immunol. 2005, 115, 911–919. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, T.; He, C. Pro-inflammatory cytokines: The link between obesity and osteoarthritis. Cytokine Growth Factor Rev. 2018, 44, 38–50. [Google Scholar] [CrossRef] [PubMed]
  44. Stolarczyk, E. Adipose tissue inflammation in obesity: A metabolic or immune response? Curr. Opin. Pharmacol. 2017, 37, 35–40. [Google Scholar] [CrossRef]
  45. Yamane, H.; Sugimoto, Y.; Tanaka, S.; Ichikawa, A. Prostaglandin E2 Receptors, EP2 and EP4, Differentially Modulate TNF-α and IL-6 Production Induced by Lipopolysaccharide in Mouse Peritoneal Neutrophils. Biochem. Biophys. Res. Commun. 2000, 278, 224–228. [Google Scholar] [CrossRef]
  46. Li, X.; Ellman, M.; Muddasani, P.; Wang, J.H.-C.; Cs-Szabo, G.; Van Wijnen, A.J.; Im, H.-J. Prostaglandin E2and its cognate EP receptors control human adult articular cartilage homeostasis and are linked to the pathophysiology of osteoarthritis. Arthritis Rheum. 2009, 60, 513–523. [Google Scholar] [CrossRef] [Green Version]
  47. Tilg, H.; Moschen, A.R. Adipocytokines: Mediators linking adipose tissue, inflammation and immunity. Nat. Rev. Immunol. 2006, 6, 772–783. [Google Scholar] [CrossRef]
  48. Fasshauer, M.; Blüher, M. Adipokines in health and disease. Trends Pharmacol. Sci. 2015, 36, 461–470. [Google Scholar] [CrossRef]
  49. Golia, E.; Limongelli, G.; Natale, F.; Fimiani, F.; Maddaloni, V.; Russo, P.E.; Riegler, L.; Bianchi, R.; Crisci, M.; Di Palma, G.; et al. Adipose tissue and vascular inflammation in coronary artery disease. World J. Cardiol. 2014, 6, 539. [Google Scholar] [CrossRef]
  50. Paradis, M.-E.; Hogue, M.-O.; Mauger, J.-F.; Couillard, C.; Couture, P.; Bergeron, N.; Lamarche, B. Visceral adipose tissue accumulation, secretory phospholipase A2-IIA and atherogenecity of LDL. Int. J. Obes. 2006, 30, 1615–1622. [Google Scholar] [CrossRef] [Green Version]
  51. Hui, D.Y. Phospholipase A2 enzymes in metabolic and cardiovascular diseases. Curr. Opin. Lipidol. 2012, 23, 235–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Rahman, M.S. Prostacyclin: A major prostaglandin in the regulation of adipose tissue development. J. Cell. Physiol. 2018, 234, 3254–3262. [Google Scholar] [CrossRef] [PubMed]
  53. Maione, F.; Casillo, G.M.; Raucci, F.; Iqbal, A.J.; Mascolo, N. The functional link between microsomal prostaglandin E synthase-1 (mPGES-1) and peroxisome proliferator-activated receptor γ (PPARγ) in the onset of inflammation. Pharmacol. Res. 2020, 157, 104807. [Google Scholar] [CrossRef] [PubMed]
  54. Ikeda-Matsuo, Y. The Role of mPGES-1 in Inflammatory Brain Diseases. Biol. Pharm. Bull. 2017, 40, 557–563. [Google Scholar] [CrossRef] [Green Version]
  55. Samuelsson, B.; Morgenstern, R.; Jakobsson, P. Membrane Prostaglandin E Synthase-1: A Novel. Pharmacol. Rev. 2007, 59, 207–224. [Google Scholar] [CrossRef]
  56. Michaud, A.; Lacroix-Pepin, N.; Pelletier, M.; Daris, M.; Biertho, L.; Fortier, M.A.; Tchernof, A. Expression of Genes Related to Prostaglandin Synthesis or Signaling in Human Subcutaneous and Omental Adipose Tissue: Depot Differences and Modulation by Adipogenesis. Mediat. Inflamm. 2014, 2014, 1–13. [Google Scholar] [CrossRef] [Green Version]
  57. Kim, R.R.; Chen, Z.; Mann, T.J.; Bastard, K.; Scott, K.F.; Church, W.B. Structural and Functional Aspects of Targeting the Secreted Human Group IIA Phospholipase A2. Molecules 2020, 25, 4459. [Google Scholar] [CrossRef]
  58. Suga, H.; Murakami, M.; Kudo, I.; Inoue, K. Participation in cellular prostaglandin synthesis of type-II phospholipase A2 secreted and anchored on cell-surface heparan sulfate proteoglycan. JBIC J. Biol. Inorg. Chem. 1993, 218, 807–813. [Google Scholar] [CrossRef]
  59. Lee, L.K.; Bryant, K.J.; Bouveret, R.; Lei, P.-W.; Duff, A.P.; Harrop, S.J.; Huang, E.P.; Harvey, R.P.; Gelb, M.H.; Gray, P.P.; et al. Selective Inhibition of Human Group IIA-secreted Phospholipase A2 (hGIIA) Signaling Reveals Arachidonic Acid Metabolism Is Associated with Colocalization of hGIIA to Vimentin in Rheumatoid Synoviocytes. J. Biol. Chem. 2013, 288, 15269–15279. [Google Scholar] [CrossRef] [Green Version]
  60. Perlson, E.; Michaelevski, I.; Kowalsman, N.; Ben-Yaakov, K.; Shaked, M.; Seger, R.; Eisenstein, M.; Fainzilber, M. Vimentin Binding to Phosphorylated Erk Sterically Hinders Enzymatic Dephosphorylation of the Kinase. J. Mol. Biol. 2006, 364, 938–944. [Google Scholar] [CrossRef]
  61. Sales, T.A.; Marcussi, S.; Da Cunha, E.F.; Kuca, K.; Kuca, K. Can Inhibitors of Snake Venom Phospholipases A2 Lead to New Insights into Anti-Inflammatory Therapy in Humans? A Theoretical Study. Toxins 2017, 9, 341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Murakami, M.; Kambe, T.; Shimbara, S.; Yamamoto, S.; Kuwata, H.; Kudo, I. Functional Association of Type IIA Secretory Phospholipase A2with the Glycosylphosphatidylinositol-anchored Heparan Sulfate Proteoglycan in the Cyclooxygenase-2-mediated Delayed Prostanoid-biosynthetic Pathway. J. Biol. Chem. 1999, 274, 29927–29936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Rodríguez, J.P.; Leiguez, E.; Guijas, C.; Lomonte, B.; Gutiérrez, J.M.; Teixeira, C.; Balboa, M.; Balsinde, J. A Lipidomic Perspective of the Action of Group IIA Secreted Phospholipase A2 on Human Monocytes: Lipid Droplet Biogenesis and Activation of Cytosolic Phospholipase A2α. Biomolecules 2020, 10, 891. [Google Scholar] [CrossRef] [PubMed]
  64. Khan, K.M.F.; Kothari, P.; Du, B.; Dannenberg, A.J.; Falcone, D.J. Matrix Metalloproteinase-Dependent Microsomal Prostaglandin E Synthase-1 Expression in Macrophages: Role of TNF-α and the EP4 Prostanoid Receptor. J. Immunol. 2012, 188, 1970–1980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kojima, F.; Naraba, H.; Sasaki, Y.; Beppu, M.; Aoki, H.; Kawai, S. Prostaglandin E2is an enhancer of interleukin-1β-induced expression of membrane-associated prostaglandin E synthase in rheumatoid synovial fibroblasts. Arthritis Rheum. 2003, 48, 2819–2828. [Google Scholar] [CrossRef]
  66. Khan, H.; Rengasamy, K.R.; Pervaiz, A.; Nabavi, S.M.; Atanasov, A.G.; Kamal, M.A.; Perviaz, A. Plant-derived mPGES-1 inhibitors or suppressors: A new emerging trend in the search for small molecules to combat inflammation. Eur. J. Med. Chem. 2018, 153, 2–28. [Google Scholar] [CrossRef]
  67. Rahman, M.S.; Khan, F.; Syeda, P.K.; Nishimura, K.; Jisaka, M.; Nagaya, T.; Shono, F.; Yokota, K. Endogenous synthesis of prostacyclin was positively regulated during the maturation phase of cultured adipocytes. Cytotechnology 2013, 66, 635–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Darimont, C.; Vassaux, G.; Ailhaud, G.; Negrel, R. Differentiation of preadipose cells: Paracrine role of prostacyclin upon stimulation of adipose cells by angiotensin-II. Endocrinology 1994, 135, 2030–2036. [Google Scholar] [CrossRef]
  69. Weinstock, A.; Silva, H.M.; Moore, K.J.; Schmidt, A.M.; Fisher, E.A. Leukocyte Heterogeneity in Adipose Tissue, Including in Obesity. Circ. Res. 2020, 126, 1590–1612. [Google Scholar] [CrossRef]
  70. Mraz, M.; Haluzik, M. The role of adipose tissue immune cells in obesity and low-grade inflammation. J. Endocrinol. 2014, 222, R113–R127. [Google Scholar] [CrossRef] [Green Version]
  71. Russo, L.; Lumeng, C.N. Properties and functions of adipose tissue macrophages in obesity. Immunology 2018, 155, 407–417. [Google Scholar] [CrossRef] [PubMed]
  72. Crop, M.; Baan, C.; Korevaar, S.; Ijzermans, J.; Pescatori, M.; Stubbs, A.; Van Ijcken, W.; Dahlke, M.; Eggenhofer, E.; Weimar, W.; et al. Inflammatory conditions affect gene expression and function of human adipose tissue-derived mesenchymal stem cells. Clin. Exp. Immunol. 2010, 162, 474–486. [Google Scholar] [CrossRef] [PubMed]
  73. Christiansen, T.; Richelsen, B.; Bruun, J.M. Monocyte chemoattractant protein-1 is produced in isolated adipocytes, associated with adiposity and reduced after weight loss in morbid obese subjects. Int. J. Obes. 2005, 29, 146–150. [Google Scholar] [CrossRef] [Green Version]
  74. Hagman, E.; Besor, O.; Hershkop, K.; Santoro, N.; Pierpont, B.; Mata, M.; Caprio, S.; Weiss, R. Relation of the degree of obesity in childhood to adipose tissue insulin resistance. Acta Diabetol. 2019, 56, 219–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Bastard, J.-P.; Maachi, M.; Lagathu, C.; Kim, M.J.; Caron, M.; Vidal, H.; Capeau, J.; Fève, B. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur. Cytokine Netw. 2006, 17, 4–12. [Google Scholar] [PubMed]
  76. Eder, K.; Baffy, N.; Falus, A.; Fulop, A.K. The major inflammatory mediator interleukin-6 and obesity. Inflamm. Res. 2009, 58, 727–736. [Google Scholar] [CrossRef] [PubMed]
  77. Józefowski, S.; Bobek, M.; Marcinkiewicz, J. Exogenous but not endogenous prostanoids regulate cytokine secretion from murine bone marrow dendritic cells: EP2, DP, and IP but not EP1, EP3, and FP prostanoid receptors are involved. Int. Immunopharmacol. 2003, 3, 865–878. [Google Scholar] [CrossRef]
  78. Jiang, J.; Dingledine, R. Role of Prostaglandin Receptor EP2 in the Regulations of Cancer Cell Proliferation, Invasion, and Inflammation. J. Pharmacol. Exp. Ther. 2013, 344, 360–367. [Google Scholar] [CrossRef] [Green Version]
  79. Sam, S.; Mazzone, T. Adipose tissue changes in obesity and the impact on metabolic function. Transl. Res. 2014, 164, 284–292. [Google Scholar] [CrossRef]
  80. Kershaw, E.E.; Flier, J.S. Adipose Tissue as an Endocrine Organ. J. Clin. Endocrinol. Metab. 2004, 89, 2548–2556. [Google Scholar] [CrossRef]
  81. Lago, F.; Diéguez, C.; Gómez-Reino, J.; Gualillo, O. Adipokines as emerging mediators of immune response and inflammation. Nat. Clin. Pract. Rheumatol. 2007, 3, 716–724. [Google Scholar] [CrossRef] [PubMed]
  82. Lago, F.; Gómez, R.; Gómez-Reino, J.J.; Dieguez, C.; Gualillo, O. Adipokines as novel modulators of lipid metabolism. Trends Biochem. Sci. 2009, 34, 500–510. [Google Scholar] [CrossRef] [PubMed]
  83. Wozniak, S.E.; Gee, L.L.; Wachtel, M.S.; Frezza, E.E. Adipose Tissue: The New Endocrine Organ? A Review Article. Dig. Dis. Sci. 2009, 54, 1847–1856. [Google Scholar] [CrossRef] [PubMed]
  84. Fernández-Riejos, P.; Najib, S.; Santos-Alvarez, J.; Martín-Romero, C.; Pérez-Pérez, A.; González-Yanes, C.; Sánchez-Margalet, V. Role of Leptin in the Activation of Immune Cells. Mediat. Inflamm. 2010, 2010, 1–8. [Google Scholar] [CrossRef] [PubMed]
  85. Balistreri, C.R.; Caruso, C.; Candore, G. The Role of Adipose Tissue and Adipokines in Obesity-Related Inflammatory Diseases. Mediat. Inflamm. 2010, 2010, 802078. [Google Scholar] [CrossRef]
  86. Schäffler, A.; Schölmerich, J.; Salzberger, B. Adipose tissue as an immunological organ: Toll-like receptors, C1q/TNFs and CTRPs. Trends Immunol. 2007, 28, 393–399. [Google Scholar] [CrossRef]
  87. Fain, J.N.; Bahouth, S.W. Regulation of Leptin Release by Mammalian Adipose Tissue. Biochem. Biophys. Res. Commun. 2000, 274, 571–575. [Google Scholar] [CrossRef]
  88. Powell, K. The two faces of fat. Nat. Cell Biol. 2007, 447, 525–527. [Google Scholar] [CrossRef]
Figure 1. MT-III induces production of PGE2, PGI2, TXA2 and LTB4 by 3T3-L1 preadipocytes. Cells were incubated with MT-III (0.4 µM) or DMEM (control) for 1 to 48 h. Bar graphs show the MT-III-induced release of PGE2 (A), PGI2 (B), TXA2 (C) and LTB4 (D) by preadipocytes. Concentrations were quantified in culture supernatants by EIA commercial kit. Results are expressed as mean ± SEM from 3 independent experiments. * p < 0.05 as compared with control group (two-way ANOVA and Bonferroni posttest).
Figure 1. MT-III induces production of PGE2, PGI2, TXA2 and LTB4 by 3T3-L1 preadipocytes. Cells were incubated with MT-III (0.4 µM) or DMEM (control) for 1 to 48 h. Bar graphs show the MT-III-induced release of PGE2 (A), PGI2 (B), TXA2 (C) and LTB4 (D) by preadipocytes. Concentrations were quantified in culture supernatants by EIA commercial kit. Results are expressed as mean ± SEM from 3 independent experiments. * p < 0.05 as compared with control group (two-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g001
Figure 2. MT-III activates COX-1 and COX-2 pathways for release of PGE2 by 3T3-L1 preadipocytes. (A) Cells were incubated with either valerylsalicylate (VSA) (10 µM), or NS-398 (10 µM), or both for 1 h, followed by incubation with MT-III (0.4 µM) for 6 h. PGE2 concentrations were quantified in culture supernatants by EIA commercial kit. (BE) 3T3-L1 preadipocytes were incubated with MT-III (0.4 µM) or DMEM (control) for 1 up to 48 h. (B) Western blotting of COX-1 and β-actin (loading control) showing immunoreactive bands. (D) Western blotting of COX-2 and β-actin (loading control) showing immunoreactive bands. Densitometric analysis of immunoreactive (C) COX-1 and (E) COX-2 bands. Density data (in arbitrary units) were normalized with those of β-actin. Results are expressed as mean ± SEM from 3 independent experiments. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (two-way ANOVA and Bonferroni posttest).
Figure 2. MT-III activates COX-1 and COX-2 pathways for release of PGE2 by 3T3-L1 preadipocytes. (A) Cells were incubated with either valerylsalicylate (VSA) (10 µM), or NS-398 (10 µM), or both for 1 h, followed by incubation with MT-III (0.4 µM) for 6 h. PGE2 concentrations were quantified in culture supernatants by EIA commercial kit. (BE) 3T3-L1 preadipocytes were incubated with MT-III (0.4 µM) or DMEM (control) for 1 up to 48 h. (B) Western blotting of COX-1 and β-actin (loading control) showing immunoreactive bands. (D) Western blotting of COX-2 and β-actin (loading control) showing immunoreactive bands. Densitometric analysis of immunoreactive (C) COX-1 and (E) COX-2 bands. Density data (in arbitrary units) were normalized with those of β-actin. Results are expressed as mean ± SEM from 3 independent experiments. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (two-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g002
Figure 3. MT-III upregulates protein expression of mPGES-1 in 3T3-L1 preadipocyte. Cells were incubated with MT-III (0.4 µM) or DMEM (control) for 1 up to 12 h. (A) Western blotting of mPGES-1 and β-actin (loading control) showing immunoreactive bands. (B) Densitometric analysis of immunoreactive mPGES-1 bands. Density data (in arbitrary units) were normalized with those of β-actin. Results are expressed as mean ± SEM from 3 experiments. * p < 0.05 as compared with the control group (two-way ANOVA and Bonferroni posttest).
Figure 3. MT-III upregulates protein expression of mPGES-1 in 3T3-L1 preadipocyte. Cells were incubated with MT-III (0.4 µM) or DMEM (control) for 1 up to 12 h. (A) Western blotting of mPGES-1 and β-actin (loading control) showing immunoreactive bands. (B) Densitometric analysis of immunoreactive mPGES-1 bands. Density data (in arbitrary units) were normalized with those of β-actin. Results are expressed as mean ± SEM from 3 experiments. * p < 0.05 as compared with the control group (two-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g003
Figure 4. MT-III-induced PGE2 release is dependent on cPLA2-α in 3T3-L1 preadipocytes. Cells were incubated with Pyr-2 (1 μM) for 1 h followed by incubation with MT-III (0.4 µM) for 3 h. PGE2 concentrations were quantified in culture supernatants by EIA commercial kit. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (two-way ANOVA and Bonferroni posttest).
Figure 4. MT-III-induced PGE2 release is dependent on cPLA2-α in 3T3-L1 preadipocytes. Cells were incubated with Pyr-2 (1 μM) for 1 h followed by incubation with MT-III (0.4 µM) for 3 h. PGE2 concentrations were quantified in culture supernatants by EIA commercial kit. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (two-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g004
Figure 5. EP4 receptor participates in MT-III-induced PGE2 biosynthesis in 3T3-L1 preadipocytes. (A) Preadipocytes were incubated with SC-19220 (10 μM), AH6809 (10 μM), L-798106 (1 μM) or AH23848 (10 μM) for 1 h followed by incubation with MT-III (0.4 µM) for 6 h. PGE2 concentrations were quantified in culture supernatants by EIA commercial kit. (BI) 3T3-L1 cells were incubated with MT-III (0.4 µM) or DMEM (control) for 1 up to 48 h. (B,D,F,H) Western blotting of EP1, EP2, EP3 and EP4 receptors, respectively, and β-actin (loading control), showing immunoreactive bands. (C,E,G,I) Densitometric analysis of immunoreactive bands for EP1, EP2, EP3 and EP4 receptors, respectively. Results are expressed as mean ± SEM from 3 independent experiments. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (one-way ANOVA and Bonferroni posttest in (A) and two-way ANOVA and Bonferroni posttest in (C,E,G,I)).
Figure 5. EP4 receptor participates in MT-III-induced PGE2 biosynthesis in 3T3-L1 preadipocytes. (A) Preadipocytes were incubated with SC-19220 (10 μM), AH6809 (10 μM), L-798106 (1 μM) or AH23848 (10 μM) for 1 h followed by incubation with MT-III (0.4 µM) for 6 h. PGE2 concentrations were quantified in culture supernatants by EIA commercial kit. (BI) 3T3-L1 cells were incubated with MT-III (0.4 µM) or DMEM (control) for 1 up to 48 h. (B,D,F,H) Western blotting of EP1, EP2, EP3 and EP4 receptors, respectively, and β-actin (loading control), showing immunoreactive bands. (C,E,G,I) Densitometric analysis of immunoreactive bands for EP1, EP2, EP3 and EP4 receptors, respectively. Results are expressed as mean ± SEM from 3 independent experiments. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (one-way ANOVA and Bonferroni posttest in (A) and two-way ANOVA and Bonferroni posttest in (C,E,G,I)).
Biomolecules 10 01593 g005aBiomolecules 10 01593 g005b
Figure 6. MT-III induces MCP-1 and IL-6 production by 3T3-L1 preadipocytes. Cells were incubated with MT-III (0.4 μM), or DMEM (control) for ½ up to 48 h. Bar graphs show concentrations of (A) MCP-1 and (B) IL-6 released by cells incubated with MT-III. Cytokines concentrations were quantified in culture supernatants by Cytometric Bead Array (CBA). Results are expressed as mean ± SEM from 5 experiments. * p < 0.05 as compared with control group (two-way ANOVA and Bonferroni posttest).
Figure 6. MT-III induces MCP-1 and IL-6 production by 3T3-L1 preadipocytes. Cells were incubated with MT-III (0.4 μM), or DMEM (control) for ½ up to 48 h. Bar graphs show concentrations of (A) MCP-1 and (B) IL-6 released by cells incubated with MT-III. Cytokines concentrations were quantified in culture supernatants by Cytometric Bead Array (CBA). Results are expressed as mean ± SEM from 5 experiments. * p < 0.05 as compared with control group (two-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g006
Figure 7. EP3 and EP4 receptors participate in the MT-III-induced release of IL-6 and MCP-1, respectively, by 3T3-L1 preadipocytes. Cells were incubated with AH23848 (10 μM) or L-798106 (1 μM) or vehicle for 1 h followed by incubation with MT-III (0,4 μM) for 12 or 24 h. Graphs show participation of EP4 receptor in the MT-III-induced release of MCP-1 (A) and participation of the EP3 receptor in the MT-III-induced release of IL-6 (B). Concentration of cytokines were quantified from culture supernatants by Cytometric Bead Array (CBA). Results are expressed as mean ± SEM from 5 experiments. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (one-way ANOVA and Bonferroni posttest).
Figure 7. EP3 and EP4 receptors participate in the MT-III-induced release of IL-6 and MCP-1, respectively, by 3T3-L1 preadipocytes. Cells were incubated with AH23848 (10 μM) or L-798106 (1 μM) or vehicle for 1 h followed by incubation with MT-III (0,4 μM) for 12 or 24 h. Graphs show participation of EP4 receptor in the MT-III-induced release of MCP-1 (A) and participation of the EP3 receptor in the MT-III-induced release of IL-6 (B). Concentration of cytokines were quantified from culture supernatants by Cytometric Bead Array (CBA). Results are expressed as mean ± SEM from 5 experiments. * p < 0.05 as compared with control group and # p < 0.05 as compared with MT-III group (one-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g007
Figure 8. MT-III upregulates gene expression of leptin and adiponectin by 3T3-L1 preadipocytes. Cells were incubated with MT-III (0.4 μM) or DMEM (control) for 1, 3 or 6 h. Graphs show gene expression of leptin (A), adiponectin (B) and resistin (C) in the presence of MT-III. Concentrations of adipokines were quantified in cell lysates by qPCR. Results are expressed as mean ± SEM from 5 experiments. * p < 0.05 as compared with the control group (two-way ANOVA and Bonferroni posttest).
Figure 8. MT-III upregulates gene expression of leptin and adiponectin by 3T3-L1 preadipocytes. Cells were incubated with MT-III (0.4 μM) or DMEM (control) for 1, 3 or 6 h. Graphs show gene expression of leptin (A), adiponectin (B) and resistin (C) in the presence of MT-III. Concentrations of adipokines were quantified in cell lysates by qPCR. Results are expressed as mean ± SEM from 5 experiments. * p < 0.05 as compared with the control group (two-way ANOVA and Bonferroni posttest).
Biomolecules 10 01593 g008
Scheme 1. Proinflammatory pathways activated by MT-III, a GIIA snake venom sPLA2, in 3T3-L1 preadipocytes. (1) MT-III stimulates preadipocytes to release PGE2 and (2) PGI2; (3) PGE2 release induced by MT-III is dependent on cPLA2-α, (4) COX-1, COX-2, mPGES-1 and (5) EP4 receptor, which triggers a positive loop for PGE2 production; (6) MT-III up-regulates COX-2 and mPGES-1, key enzymes involved in PGE biosynthesis. Moreover, MT-III induces the release of (7) MCP-1, dependent on the EP4 receptor, and (8) IL-6, dependent on the EP3 receptor. Furthermore, (9) MT-III up-regulates leptin and adiponectin gene expression
Scheme 1. Proinflammatory pathways activated by MT-III, a GIIA snake venom sPLA2, in 3T3-L1 preadipocytes. (1) MT-III stimulates preadipocytes to release PGE2 and (2) PGI2; (3) PGE2 release induced by MT-III is dependent on cPLA2-α, (4) COX-1, COX-2, mPGES-1 and (5) EP4 receptor, which triggers a positive loop for PGE2 production; (6) MT-III up-regulates COX-2 and mPGES-1, key enzymes involved in PGE biosynthesis. Moreover, MT-III induces the release of (7) MCP-1, dependent on the EP4 receptor, and (8) IL-6, dependent on the EP3 receptor. Furthermore, (9) MT-III up-regulates leptin and adiponectin gene expression
Biomolecules 10 01593 sch001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Leiguez, E.; Motta, P.; Maia Marques, R.; Lomonte, B.; Sampaio, S.V.; Teixeira, C. A Representative GIIA Phospholipase A2 Activates Preadipocytes to Produce Inflammatory Mediators Implicated in Obesity Development. Biomolecules 2020, 10, 1593. https://doi.org/10.3390/biom10121593

AMA Style

Leiguez E, Motta P, Maia Marques R, Lomonte B, Sampaio SV, Teixeira C. A Representative GIIA Phospholipase A2 Activates Preadipocytes to Produce Inflammatory Mediators Implicated in Obesity Development. Biomolecules. 2020; 10(12):1593. https://doi.org/10.3390/biom10121593

Chicago/Turabian Style

Leiguez, Elbio, Priscila Motta, Rodrigo Maia Marques, Bruno Lomonte, Suely Vilela Sampaio, and Catarina Teixeira. 2020. "A Representative GIIA Phospholipase A2 Activates Preadipocytes to Produce Inflammatory Mediators Implicated in Obesity Development" Biomolecules 10, no. 12: 1593. https://doi.org/10.3390/biom10121593

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop