Abstract
Obesity results in increased macrophage recruitment to adipose tissue that promotes a chronic low-grade inflammatory state linked to increased fatty acid efflux from adipocytes. Activated macrophages produce a variety of pro-inflammatory lipids such as leukotriene C4 (LTC4) and 5-, 12-, and 15-hydroxyeicosatetraenoic acid (HETE) suggesting the hypothesis that fatty acids may stimulate eicosanoid synthesis. To assess if eicosanoid production increases with obesity, adipose tissue of leptin deficient ob/ob mice was analyzed. In ob/ob mice, LTC4 and 12-HETE levels increased in the visceral (but not subcutaneous) adipose depot while the 5-HETE levels decreased and 15-HETE abundance was unchanged. Since macrophages produce the majority of inflammatory molecules in adipose tissue, treatment of RAW264.7 or primary peritoneal macrophages with free fatty acids led to increased secretion of LTC4 and 5-HETE, but not 12- or 15-HETE. Fatty acid binding proteins (FABPs) facilitate the intracellular trafficking of fatty acids and other hydrophobic ligands and in vitro stabilize the LTC4 precursor leukotriene A4 (LTA4) from non-enzymatic hydrolysis. Consistent with a role for FABPs in LTC4 synthesis, treatment of macrophages with HTS01037, a specific FABP inhibitor, resulted in a marked decrease in both basal and fatty acid-stimulated LTC4 secretion but no change in 5-HETE production or 5-lipoxygenase expression. These results indicate that the products of adipocyte lipolysis may stimulate the 5-lipoxygenase pathway leading to FABP-dependent production of LTC4 and contribute to the insulin resistant state.
Keywords: macrophage, fatty acids, leukotrienes, inflammation, LTC4, fatty acid binding protein
1. Introduction
Obesity-induced insulin resistance is characterized by increased basal lipolysis resulting in elevated levels of circulating free fatty acids and a state of chronic low-grade local inflammation [1–4]. A hallmark of the inflammatory state is infiltration of macrophages into adipose tissue resulting in release of a variety of pro-inflammatory molecules that affect insulin action and systemic energy metabolism [5–7]. Most studies examining inflammatory processes in adipose tissue have focused on the production and signaling effects of pro-inflammatory cytokines such as interleukin 6, tumor necrosis factor α, and monocyte chemoattractant protein-1 (MCP-1) [8–12]. However, the role of pro-inflammatory lipid mediators has received less attention.
Pro-inflammatory lipid mediators result from enzymatic oxygenation and/or cyclization of arachidonic acid, that is present in abundance at the sn-2 position of phospholipids in many cell types including macrophages and other immune cells. Arachidonic acid is metabolized by the cyclooxygenase (COX) and lipoxygenase (LOX) enzymes resulting in formation of prostaglandins and leukotrienes, respectively [13, 14]. Leukotriene production is dependent on calcium-induced translocation of cytosolic phospholipase A2 (cPLA2) and 5-lipoxygenase (5LO) to the perinuclear membrane resulting in liberation of arachidonic acid and formation of leukotriene A4 (LTA4) [15]. LTA4 is metabolized by leukotriene A4 hydrolase or leukotriene C4 synthase (LTC4S) to produce leukotriene B4 (LTB4) or leukotriene C4 (LTC4), respectively. Under some circumstances leukotriene synthesis may occur via a transcellular mechanism where formation may take place in a cell distinct from that producing LTA4 [16]. LTB4 and LTC4 are secreted and LTC4 undergoes extracellular proteolytic processing to leukotriene D4 (LTD4) and leukotriene E4 (LTE4) [15].
Leukotriene production is an important component of the immune response. After synthesis and secretion from the cell, leukotrienes bind to cell surface G-protein coupled receptors on target cells and elicit a variety of biological outputs [17–19]. LTB4 promotes chemotaxis and activation of various leukocytes [20, 21]. For example, LTB4 stimulates MCP-1 synthesis by macrophages leading to monocyte recruitment [22]. Cysteinyl leukotriene (LTC4, LTD4, and LTE4) bioactivity has been characterized primarily in bronchial diseases such as asthma and other chronic inflammatory disorders of the lung where LTC4 and its metabolites LTD4 and LTE4 act as potent bronchoconstrictors, promote airway remodeling, mucus secretion and vascular permeability [19, 23–25].
While the role of leukotrienes in chronic lung inflammation is well characterized, a role for leukotrienes in adipose tissue inflammation is not well understood [26]. In the chronic inflammatory state observed in obesity-induced insulin resistance, eicosanoid biology may play a role in the onset and maintenance of the inflammatory response. Indeed, several recent reports focus on the role of hydroxyeicosatetraenoic acids (HETEs) in adipose tissue suggesting their importance in obesity related disorders. Lipoxygenases catalyze synthesis of HETEs via an oxygenation reaction, resulting in monohydroxylated products. Studies have shown that the 12/15-lipoxygenase product 12S-hydroxyeicosatetraenoic acid (12S-HETE) is associated with obesity and insulin resistance [27–29]. Moreover, disruption of the receptor for LTB4 renders experimental mice resistant to obesity-linked insulin resistance and reduces macrophage infiltration of adipose tissue [30]. Targeted deletion of 5-lipoxygenase leads to increased adiposity but protects mice from insulin resistance [31]. Finally, pharmacologic inhibition of 5-lipoxygenase activating protein (FLAP) results in reduced inflammation and improved insulin sensitivity in high-fat-fed mice [32].
Fatty acid binding proteins (FABPs), a family of 15 kDa polypeptides, bind fatty acids and other hydrophobic molecules in a ligand-binding cavity to facilitate solubilization and intracellular transport [33, 34]. The two major FABPs in adipose tissue, adipocyte FABP (AFABP) and epithelial FABP (EFABP) are expressed in both adipocytes and macrophages [35]. While AFABP is more abundantly expressed in adipocytes compared to EFABP the opposite is true in macrophages. In addition, low levels of the heart FABP (HFABP) is expressed in both adipocytes and macrophages. Targeted disruption of AFABP and/or EFABP results in reduced inflammation and protection against obesity-induced insulin resistance in mouse models [36]. While the exact mechanisms of this protective effect are unknown, FABPs increase the half-life of the unstable epoxide-containing LTA4, the precursor of LTB4 and LTC4, up to 20-fold in vitro [37]. This property suggests that FABPs in general may play a crucial role in leukotriene biosynthesis by stabilizing LTA4 against hydrolysis, resulting in increased inflammatory signaling.
Based on increased adipose tissue inflammation observed in the obese, insulin resistant state, we hypothesized that inflammatory eicosanoid levels are increased in adipose tissue of ob/ob mice. In conjunction, we hypothesized that macrophages produce eicosanoids in a fatty acid-dependent manner. Finally, considering the stabilizing effect of FABPs on LTA4, we hypothesized that absence or inhibition of FABPs block leukotriene production by decreasing overall availability of LTA4 for conversion to more stable leukotrienes. To that end, we used targeted lipidomic profiling of eicosanoids and found that visceral adipose depots from ob/ob mice contain elevated levels of inflammatory LTC4 and 12-HETE relative to control mice. We demonstrate that fatty acid treatment of macrophages increased LTC4 and 5-HETE levels and that inhibition of FABPs significantly abrogated the fatty acid-dependent LTC4 production indicating that LTC4 formation is dependent on FABPs.
2. Materials and Methods
2.1. Materials
PGE2, PGD2, PGE2-d4, LTB4, LTC4, LTD4, LTE4, LTC4-d5, 5S-HETE, 12S-HETE, 15S-HETE, and15S-HETE-d8 were obtained from Cayman Chemical (Ann Arbor, MI). Fluo4-AM and red blood cell lysing buffer was purchased from Sigma-Aldrich (St. Louis, MO). Palmitate (16:0), stearate (18:0), palmitoleate (16:1n-7), oleate (18:1n-9), linoleate (18:2n-6), and α-linolenate (18:3n-3) were obtained from Nu-Chek Prep, Inc. (Elysian, MN). Strata-X solid phase extraction cartridges (200 mg/3mL) were purchased from Phenomenex (Torrance, CA). HTS01037 was a kind gift provided by Maybridge Ltd, UK. cPLA2 (sc-438) and 5LO (sc-20785) antibodies were purchased from Santa Cruz Biotechnology.
2.2. Animals
C57Bl/6J and Lepob (ob/ob) mice were purchased from Jackson Laboratories (Bar Harbor, ME). The mice were sacrificed and tissues harvested at 12 weeks of age and blood glucose levels were measured immediately using a one-touch ultra glucose meter. All procedures were approved by the University of Minnesota Institutional Animal Care and Use Committee.
2.3. Primary peritoneal macrophage isolation
Female C57Bl/6J mice were injected with 2 mL 4% thioglycollate to induce macrophage infiltration into the peritoneal cavity. After four days, animals were sacrificed using carbon dioxide asphyxiation and macrophages harvested by peritoneal lavage with sterile phosphate-buffered saline with 3% fetal bovine serum (FBS). The lavage fluid was centrifuged at 1500 rpm at 4° C for 10 minutes and the resultant cell pellet was resuspended in red blood cell lysing buffer and incubated for 15 minutes on ice. The cells were centrifuged at 1500 rpm for 10 minutes and then resuspended in RPMI 1640 medium supplemented with 10% FBS. Cells were plated in 6- or 12-well plates at a density of 1 × 106 cells per well, incubated overnight and used the next day.
2.4 Stromal Vascular and Primary Adipocyte Preparation
Murine epididymal fat pads were minced and digested with Krebs-Ringer Hepes (KRH) buffer (3.8 mM Hepes, 3.2 mM CaCl2, 2 mM sodium pyruvate, 1.2 mM MgSO4, 4.7 mM KCl, 120 mM NaCl, 5 mM NaHCO3, 1× glutamax, 1% BSA fatty acid free, 5 mM glucose, pH 7.4) containing 1 mg/ml collagenase (type II, Sigma) for 1 h at 37° C with constant gentle agitation. Digested tissue was filtered through a 100 μm mesh nylon cell strainer and centrifuged at 500 × g for 10 min at room temperature. Adipocytes were recovered from the floating cell layer and stromal vascular cells from the pellet. Separated stromal vascular and adipocyte fractions were each incubated for 2 h in KRH and media collected for eicosanoid analysis describe in section 2.6.
2.5. Fatty acid stimulated eicosanoid synthesis
RAW264.7 macrophages were cultured in DMEM containing 10% FBS prior to treatment with free fatty acids. RAW264.7 or primary peritoneal macrophages were washed twice prior to treatment with fatty acids, 20 μM fatty acid complexed to 5 μM BSA (4:1 ratio) in Krebs-Ringer Hepes (KRH) buffer containing 4 mM calcium. After 2h the culture media were collected for eicosanoid analysis. For time course experiments, primary peritoneal macrophages were incubated with free fatty acids and culture media were collected after 1, 2, and 4h. For dose-response experiments, RAW264.7 macrophages were treated with 10, 15, 20, or 25 μM palmitate complexed to 5 μM BSA for 2h before culture media were collected. Palmitate concentrations are reported as free palmitate when complexed to BSA at the indicated ratios as calculated according to Richieri et al, [38]. For studies with HTS01037, primary peritoneal macrophages were incubated with the FABP inhibitor for 18 h, washed and incubated with fresh medium containing HTS01037 plus fatty acids coupled to BSA. The medium was collected after 1 h and LTC4 quantitated by LC-MS/MS.
2.6. Lipidomic analyses of eicosanoids
Internal standards (4 μL of 250 nM LTC4-d5, PGE2-d4, and 15S-HETE-d8) were added to culture media immediately following harvest. Samples were vortexed briefly and loaded on Strata-X columns conditioned with 4 mL methanol and equilibrated with 4 mL water. The columns were washed with 4 mL water and eicosanoids eluted with 4 mL methanol. The eluate was dried under nitrogen and resuspended in 25 μL of methanol for LC-MS/MS analysis. For tissue analyses, samples were homogenized in 100mM sodium acetate pH 3.9, 500 μM diethylenetriaminepentaacetic acid and 250 μM butylated hydroxytoluene containing internal standards and centrifuged at 3800 rpm for 10 minutes. The aqueous phase was transferred to Strata-X solid phase extraction cartridges and eicosanoids recovered.
LC-MS/MS analyses for cell culture experiments were performed using an Agilent 1100 HPLC coupled to an AB/Sciex API 4000QTrap mass spectrometer with methods similar to Buczynski et al. [39]. Analyses of adipose tissues were performed using a Shimamdzu Prominence UPLC coupled to an AB/Sciex API 5500QTrap. Chromatographic separations were achieved using a 2.1 × 100 mm Agilent Zorbax Eclipse C18 column with a 3.5 micron particle size using a gradient elution. Solvent A consisted of 70:30:0.1 water:acetonitrile:acetic acid and solvent B consisted of 50:50 acetonitrile:isopropanol. A gradient was employed at a flow rate of 400 μL/minute (0–1 min 0% B, 1–10 min, 0–100% B, 10–15 min 100% B, 15–16 min 100-0% B, 16–23 min 0% B).
Mass spectrometry analyses were performed using electrospray ionization with an ionspray voltage of −4500V and a curtain gas flow rate of 10 L/min at a source temperature of 600° C. All analytes were detected using multiple reaction monitoring (MRM) conditions listed in Table 1 and quantified by stable isotope dilution.
Table 1.
Multiple Reaction Monitoring (MRM) Conditions
| Analyte | MRM 1 (m/z) | MRM 2 (m/z) |
|---|---|---|
| LTB4 | 335.3/195.0 | 335.3/203.0 |
| LTC4 | 624.5/272.0 | 624.5/254.2 |
| LTD4 | 495.0/177.0 | 495.0/143.0 |
| LTE4 | 438.2/333.0 | 438.2/351.2 |
| 5S-HETE | 319.4/115.0 | 319.4/203.1 |
| 12S-HETE | 319.4/179.1 | 319.4/203.1 |
| 15S-HETE | 319.4/175.0 | 319.4/203.1 |
| 5,6-diHETE | 335.3/115.0 | 335.2/219.0 |
| PGE2 | 351.2/271.0 | 351.2/189.1 |
| PGD2 | 351.2/271.0 | 351.2/189.1 |
| PGE2-d4 | 355.2/275.2 | 355.2/193.2 |
| LTC4-d5 | 629.5/272.0 | 629.5/254.2 |
| 15S-HETE-d8 | 327.4/226.2 | 327.4/309.5 |
2.7. Quantitative RT-PCR
Expression of mRNAs was measured by quantitative RT-PCR. Total RNA was isolated from cells using Trizol (Invitrogen) and cDNA was made using iScript cDNA synthesis kit (BioRad). Relative transcript measurements were performed with a MyiQ detection system (BioRad) using relevant primers and iQ SYBRgreen Supermix. Data was normalized to TFIIE. Primers: cPLA2 Forward: 5'-CCTTTATGGCTCCTGACCTATTTG-3', cPLA2 Reverse: 5'-CATTACTCACGATGTGCTTTGCT-3', LTC4S Forward: 5'-AGCCCTGTGCGGACTGTTCTAC-3', LTC4S Reverse: 5'-GCATCTGGAGCCATCTGAAGAG-3', TFIIE Forward: 5'-CAAGGCTTTAGGGGACCAGATAC-3', TFIIE Reverse: 5'-CATCCATTGACTCCACAGTGACAC-3'
2.8. Immunoblotting
Cells were lysed in RIPA buffer, sonicated and centrifuged at 14000 rpm for 10 min. Proteins were subjected to SDS-PAGE, transferred to PVDF membranes and incubated with the indicated primary antibodies. Membranes were incubated with secondary antibody conjugated to LI-COR IRDye for 45 minutes and visualized using LI-COR Odyssey infrared imager (LI-COR biotechnologies, Lincoln, NE).
2.9. Statistical analysis
Data are presented as mean ± S.E. Statistical comparisons were performed pairwise by Student's t-test with statistical significance defined by p<0.05.
3. Results
3.1. LTC4 and 12-HETE are elevated in adipose tissue from ob/ob mice
To evaluate the eicosanoid profile in adipose tissue of lean and obese mice, we utilized male C57Bl/6J or leptin deficient ob/ob mice maintained on a normal chow diet. At 12 weeks of age, ob/ob mice had significantly increased body weight compared to lean mice as would be expected (Fig. 1A). After a 4-hour fast, serum glucose levels were significantly elevated in ob/ob mice compared to controls (Fig. 1B). The increase in body weight was accompanied by an increase in epididymal fat pad weight (Fig. 1C), as well as increased inguinal adipose tissue weight (Fig. 1D). Eicosanoid levels in visceral and subcutaneous adipose tissue, represented by the epididymal and inguinal depots respectively, were measured by targeted lipidomic profiling in ob/ob and control mice (a representative chromatogram of synthetic standards is displayed in Fig. 2A). LTC4 levels in epididymal adipose tissue were increased in ob/ob animals compared to control mice (Fig. 2B). Similarly, LTC4 levels were elevated in inguinal adipose tissue of ob/ob mice compared to control mice (Fig. 2C). This result agrees with previously data establishing that epididymal adipose tissue, as well as subcutaneous adipose tissue, from obese mice fed a high-fat diet produce increased levels of cysteinyl leukotrienes compared to mice fed a normal chow diet [40]. Furthermore, levels of 5-HETE were decreased, 12-HETE levels were increased and 15-HETE were unaltered in epididymal adipose tissue from ob/ob mice versus control mice (Fig. 2D). Prostaglandins were not quantified, as chromatograms from adipose tissue preparations exhibited a more complex multiple reaction monitoring signature resulting in an inability to adequately resolve PGE2 and PGD2 likely due to the presence of other isomers such as isoprostanes [41]. In addition, LTB4, 6-trans-LTB4, 5,6-diHETE, LTD4 and LTE4 were not detected in adipose tissue from chow fed C57BL/6J or ob/ob mice. These data indicate that in conditions where fatty acids are elevated, LTC4 and 12-HETE production are also increased in adipose tissue.
Figure 1. Characterization of ob/ob and C57Bl/6J mice.
A) Body weight, B) fasted glucose levels, C) epididymal and D) inguinal white adipose tissue weights of 12-week old control and ob/ob mice. n=18 for control and n=12 for ob/ob animals.
Figure 2. Eicosanoid levels in adipose tissue of ob/ob and C57Bl/6J mice.
A) Chromatogram of synthetic standards of eicosanoids measured via LC-MS/MS. LTC4 levels in B) epididymal and C) inguinal white adipose tissue of 12-week old ob/ob or control C57Bl/6J mice profiled via LC-MS/MS. D) Levels of HETEs in epididymal white adipose tissue. n=3–12 for control and n=6–12 for ob/ob.
3.2 Eicosanoid production by primary adipocytes and stromal vascular fraction
Adipose tissue is comprised of a variety of cell types including adipocytes, preadipocytes, endothelial cells, macrophages and other immune cells such as T cells and dendritic cells [6, 42, 43]. Although adipocytes are capable of secreting leukotrienes, cells within the stromal vascular fraction produce the majority of leukotrienes in visceral adipose tissue [40]. Furthermore, in respiratory disorders such as asthma and chronic obstructive pulmonary disease (COPD), immune cells are the primary producers of eicosanoids [44, 45]. To further characterize the types of cells that produce eicosanoids, adipocytes and stromal vascular (SV) cells were separated by collagenase digestion and eicosanoids evaluated. The SV fraction produced LTB4, LTC4, and LTE4, while primary adipocytes produced only LTC4 (Fig. 3A). The presence of LTB4 and LTE4 in the isolated SV fraction, but not whole tissue, may be due to LTA4 metabolism via LTA4 hydrolase and gamma-glutamyl transpeptidase and dipeptidases, respectively [15]. In addition to greater leukotriene production in the SV fraction, 12-HETE was also found at elevated levels in the SV fraction. 5-HETE was produced in equivalent amounts by the SV and adipocyte fractions while 15-HETE was only produced by adipocytes (Fig. 3B). Finally, PGE2 was produced at greater levels by SV cells while PGD2 was produced equivalently by both SV cells and adipocytes (Fig. 3C).
Figure 3. Eicosanoid production by primary adipocytes and stromal vascular fraction.

Minced adipose tissue was collagenase digested and separated into primary adipocyte and stromal vascular fractions and A) leukotrienes, B) HETEs and C) prostaglandins were measured by LC-MS/MS. n=3 for stromal vascular fractions, while n=2 for adipocyte fractions.
3.3. Fatty acid-dependent production of eicosanoids in macrophages
As immune cells were the primary produces of a number of eicosanoids such as leukotrienes and 12-HETE, macrophages may produce pro-inflammatory lipids in a fatty acid-dependent manner. To test the hypothesis that macrophages in adipose tissue carry out fatty acid-dependent eicosanoid synthesis we evaluated the capacity of RAW264.7 macrophages and primary peritoneal macrophages to produce leukotrienes and other eicosanoids. Treatment of RAW264.7 macrophages with palmitate led to a dose-dependent increase in LTC4 secreted into the medium (Fig. 4A). Furthermore, treatment with palmitate, stearate, oleate, or linoleate for 2h led to a significant increase in LTC4 production as compared to a fatty acid-free control, while palmitoleate showed a trend toward an increase (Fig. 4B). LTC4 was not found intracellularly in any cell type examined (results not shown). LTB4, LTD4 and LTE4 were not detected in media from RAW264.7 macrophages under any treatment condition. PGD2 and PGE2 were detected in all analyses, but levels were not altered by fatty acid treatment. Furthermore, 5-HETE, 12-HETE, and 15-HETE were not detected under any condition using RAW264.7 macrophages.
Figure 4. Quantitation of eicosanoids in RAW264.7 macrophages.
A) Dose-dependent palmitate-induced LTC4 production by RAW264.7 macrophages was quantified by LC-MS/MS (n=4 at each concentration). B) RAW264.7 macrophages were treated with fatty acids for 2h and LTC4 levels were measured by LC-MS/MS (n=4).
Treatment of primary peritoneal macrophages with palmitate, stearate, palmitoleate, oleate, or linoleate led to dose-dependent changes in eicosanoid production. All fatty acids resulted in significantly greater LTC4 levels than fatty acid-free control (Fig. 5A). Most of the change was observed within the first hour of treatment. PGE2 levels were also elevated in response to all fatty acids, but this happened at a slower time scale compared to LTC4 and in most cases PGE2 was only significantly increased after four hours of fatty acid stimulation (Fig. 5B). Fatty acid treatments also lead to greater levels of 5-HETE (Fig. 5C). In general, these changes were observed within the first hour of treatment. Finally, treatment with fatty acids did not result in increased production of 12-HETE, with the exception of oleate, after 4h treatment (Fig. 5D). LTD4, LTE4, and LTB4 were not detected under any treatment condition, and 15-HETE levels did not differ from control under any conditions (data not shown). Although cultured and primary macrophages both produce LTC4 in a fatty acid-dependent manner, primary adipocytes do not. Adipocytes are capable of producing LTC4, but the levels were not altered in response to palmitate stimulation (results not shown).
Figure 5. Fatty acid-induced eicosanoid synthesis in primary peritoneal macrophages.
Primary peritoneal macrophages were treated with 20 μM of the indicated fatty acid complexed to 5μM BSA and the production of A) LTC4, B) PGE2, C) 5-HETE, and D) 12-HETE measured by LC-MS/MS after 1, 2, and 4 hour incubations (n=4).
3.4. FABPs play a role in LTC4 production in macrophages
To further characterize the mechanism of fatty acid-dependent eicosanoid formation, we evaluated the role of FABPs in eicosanoid production. FABPs stabilize LTA4, the unstable precursor of the biologically active leukotrienes [37]. Since macrophages express AFABP, EFABP and HFABP, and all FABPs have been shown to stabilize LTA4 in vitro, we utilized a pharmacologic approach towards blocking FABP function. To characterize FABP-dependent eicosanoid production we utilized a specific FABP inhibitor, HTS01037, that binds avidly to all FABP isoforms [46]. Pre-treatment of primary peritoneal macrophages with HTS01037 prior to fatty acid stimulation resulted in reduced LTC4 levels in a time (Fig. 6A) and concentration (Fig. 6B) dependent manner. Furthermore, pretreatment with HTS01037 resulted in blunted fatty acid-dependent LTC4 production (Fig. 6C). Although LTC4 was consistently reduced upon HTS01037 treatment, the basal abundance of other eicosanoids (5-HETE, 12-HETE and 15-HETE) was not lower and the level of 12-HETE was increased (Fig. 6D).
Figure 6. HTS01037 treatment of macrophages.
To evaluate inhibition of eicosanoid production by HTS01037, primary peritoneal macrophages were; A) treated with 20 μM HTS01037 (HTS) for the indicated times and then evaluated for LTC4 secretion into the cell culture medium during the final 60 min (n=3), B) treated with the indicated levels of HTS01037 for 18 h and then analyzed for LTC4 secretion during the final 60 min (n=3), C) incubated for 18h with HTS01037 prior to 2h fatty acid treatment and analysis of LTC4 (n=6) or D) HETEs (n=6). E) RT-PCR analysis of cPLA2 and 5LO in primary peritoneal macrophages after 18 h HTS01037 treatment (n=6). F) cPLA2 and 5LO protein levels in primary peritoneal macrophages were analyzed by immunoblotting after 18 h HTS01037 treatment (n=3). * denotes significant difference from 0h or 0 μM, ** indicates significant difference from 0 and 1h or 0 and 0.2 μM and *** denotes significant difference from 0, 1, and 2h. #, below the limit of quantification.
HTS01037 may affect LTC4 production directly by inhibiting FABP lipid binding resulting in release and destabilization of LTA4. It is also possible that HTS01037 indirectly reduced LTC4 production by affecting the expression of enzymes involved in LTC4 production. To assess if HTS01037 altered the expression of enzymes in the leukotriene pathway, quantitative RT-PCR was performed. cPLA2 and LTC4S mRNA expression were not changed in response to HTS01037 treatment (Fig. 6E). Furthermore, western blot analysis confirmed that 5LO and cPLA2 protein levels were unaltered upon stimulation with HTS01037 (Fig. 6F).
4. Discussion
Inflammation is often characterized by formation of potent bioactive eicosanoids, such as leukotrienes that act on G-protein coupled receptors to initiate a signaling cascade linked to altered cellular function [19]. In the lung, cysteinyl leukotrienes contribute significantly to bronchoconstriction and vascular permeability in asthma and other pulmonary inflammatory diseases. To that end, antagonists of leukotriene receptors are used to counteract the pathology of these diseases [17, 24, 47]. Obesity has been characterized in part by a chronic low-grade inflammatory state yet the involvement of leukotrienes in adipose biology has not been fully delineated. The present study was undertaken to evaluate eicosanoid synthesis in adipose tissue and in particular, fatty acid-dependent eicosanoid synthesis by macrophages. In this context, fatty acids are not substrates for eicosanoid synthesis but rather function as regulatory ligands that stimulate macrophage signaling and elicit the formation and secretion of eicosanoids.
In this study, we evaluated the levels of eicosanoids in visceral and subcutaneous adipose tissue of control or ob/ob mice. Ob/ob mice on the C57BL/6J genetic background have been characterized as a model of obesity-induced insulin resistance [48]. LTC4 levels were increased in both the epididymal and inguinal adipose depot in ob/ob mice. We found 5-HETE levels reduced, 12-HETE levels increased and 15-HETE levels unaltered in the epididymal white adipose tissue from ob/ob mice. A previous study by Gonzalez-Periz and colleagues also reported levels of a number of eicosanoids by LCMS/MS analysis [49]. However, our study differs in that cysteinyl leukotrienes were analyzed in addition to a representative sampling of HETEs and prostaglandins. Based on the finding that LTC4 levels differed in ob/ob vs control mice, further experiments were designed to examine fatty acid dependent LTC4 synthesis. An additional intriguing possibility is that since the levels of both LTC4 and 12-HETE are increased in adipose tissue from obese models that the two lipids are somehow linked or that one leads to the production of the other.
To address which cell types in adipose tissue were responsible for eicosanoid synthesis, we digested adipose tissue and separated the stromal vascular fraction from adipocytes. Profiling of eicosanoids in separated cells revealed that the SV fraction was responsible for the majority of leukotriene, 12-HETE, and PGE2 production. Interestingly, the eicosanoid profile of the SV fraction was quite different from whole tissue. Specifically, LTB4 and LTE4 were not detected in whole tissue homogenate, but were produced by the SV fraction. Leukotriene synthesis and metabolism occurs, in part, via transcellular mechanisms [16, 50]. It is likely that the unique composition of a collagenase digested SV fraction allows LTA4 and LTC4 access to enzymes that convert them to LTB4 and LTE4, respectively, while this access may be restricted in whole tissue where vascular cells are morphologically separated from macrophages and other immune cells. Further study would provide greater mechanistic evidence as to the nature of the differences between whole tissue and collagenase digested SV fractions. However, the results of this experiment support the hypothesis that leukotrienes are produced in greater quantity by immune cells as opposed to adipocytes.
To further characterize immune cell production of eicosanoids, we evaluated fatty acid-dependent eicosanoid production in macrophages. The fatty acids chosen for this study represent the five most abundant fatty acids found in serum of humans and animals consuming a western high-fat diet. Interestingly, saturated, monounsaturated, and polyunsaturated fatty acids were all capable of inducing LTC4 synthesis in RAW264.7 macrophages. A major characteristic of obesity is increased lipolysis resulting in increased levels of free fatty acids [51, 52]. The free fatty acid composition of adipose tissue and serum mirrors the fatty acid composition of the diet ingested [53, 54]. These results suggest that the major fatty acids comprising a western high-fat diet are all capable of inducing LTC4 production in macrophages. High-fat diets, consisting of 60% calories from fat derived from lard, are commonly used to induce the metabolic syndrome in mouse models. This diet is high in both saturated and monounsaturated fatty acids and induces pro-inflammatory effects in vivo [55]. The data presented herein suggest that these pro-inflammatory events may, at least in part, be mediated through LTC4 production. However, RAW264.7 macrophages did not produce 5-HETE, 12-HETE, or 15-HETE. Previous work has indicated that immortalized and primary macrophages display different eicosanoid production and metabolism [56]. Due to this heterogeneity, we also evaluated the eicosanoid profiles in response to fatty acids in primary murine peritoneal macrophages.
In primary peritoneal macrophages, fatty acid treatment revealed a more complex eicosanoid profile than in immortalized murine cell lines. All fatty acid treatments led to increased production of LTC4, PGE2, and 5-HETE. Interestingly, the time course of formation varied for each eicosanoid. LTC4 and 5-HETE levels were increased relatively quickly as measured at the 1h time point. These results are consistent with previous studies showing that 5-HETE and LTC4 production occur in an acute manner in response to sustained increases in calcium flux [39]. In contrast, PGE2 levels were not significantly increased until 4h. Increased PGE2 production relies primarily on increased expression of COX-2, resulting in a slower onset of stimulation-induced PGE2 production. Extant literature suggests that both LTC4 and free fatty acids induce COX-2 expression and subsequent PGE2 production [57, 58]. From our data, it is unclear whether LTC4 formation induces PGE2 production or if increased PGE2 is a result of fatty acid treatment. Interestingly, we did not observe a fatty acid-dependent increase in 12-HETE levels in macrophages indicating that adipocytes may be responsible for production of this inflammatory mediator [27]. It is important to note that the time course of fatty acid-dependent leukotriene production is slower than previously described. Other studies have relied heavily on calcium ionophores such as A23187 or ADP to induce a rapid increase in calcium that results in increased leukotriene production within minutes, or even seconds [59]. In contrast, fatty acid treatment results in a sustained increase in calcium flux that occurs much more slowly than ionophore treatment, accounting for the delayed increase in leukotriene production (results not shown). Taken together, our results suggest a primary role for macrophages in adipose tissue LTC4 production, although other cells found in the stromal vascular fraction may also contribute.
LTA4 is a highly unstable molecule in aqueous solutions due to an epoxide moiety susceptible to spontaneous water hydrolysis, resulting in formation of 5,6-diHETE or 5,12-diHETE [60]. Based on the observation that FABPs can stabilize LTA4, we examined the role of FABPs in leukotriene formation. Pharmacologic inhibition of FABPs using HTS01037 reduces lipolysis in adipocytes and reduces inflammatory cytokine production in macrophages [46]. Macrophages express EFABP > AFABP >> HFABP as assessed using microarray analysis and mono-specific antibodies directed towards each FABP isoform (unpublished). The use of HTS01037 does not allow us to distinguish which, if any, isoform of FABP expressed in macrophages plays the major role in leukotriene production. HTS01037 binds to AFABP inside the ligand-binding cavity in a manner similar to other ligands, coordinating the carboxyl group to Arg126 and Tyr128. Such residues are conserved in both EFABP and HFABP. HTS01037 treatment of macrophages ablated LTC4 levels in the media in a time and concentration-dependent manner. Interestingly, LTC4 levels transiently increased in response to HTS01037 but decreased at longer time points. Furthermore, LTC4 production in response to fatty acid treatment was blocked by pretreatment with HTS01037 indicating that FABPs play a crucial role in leukotriene production. While HTS01037 can block LTA4 binding to FABPs, it is also possible that HTS01037 has indirect effects on the expression of fatty acid-dependent transcription factors such as PPARγ and PPARδ. Such changes in gene expression could affect other steps in the leukotriene production pathway. However, mRNA and protein levels of enzymes in the leukotriene synthesizing machinery were not altered upon HTS01037 stimulation (Figure 6).
Macrophages express a variety of free fatty acid receptors including TLR4, CD36 and GPR120 [39, 61, 62]. Toll-like receptor 4 (TLR4) plays a role in the inflammatory response induced by invading pathogens and saturated fatty acids such as palmitate and stearate resulting in the activation of nuclear factor κB (NF-κB) and subsequent formation of pro-inflammatory cytokines and prostaglandins via increased expression of cyclooxygenase-2 [63, 64]. TLR4 is incapable of binding fatty acids directly but recent data identified Fetuin-A as an endogenous ligand of TLR4 and as a necessary component for activation of TLR4 in response to fatty acids [65]. Fetuin-A is produced and secreted from the liver and would therefore not be available in our primary or cultured macrophages. However, it remains to be evaluated if cell types other than hepatocytes can secrete Fetuin-A or an unidentified protein with a similar function or if the effect that has been attributed to TLR4 activation by fatty acids in culture models in fact represents activation of another fatty acid receptor. Macrophages abundantly express the class B scavenger receptor CD36 [66, 67]. CD36 facilitates uptake of long chain fatty acids but is also involved in transduction of signaling events [61, 66, 68–70]. CD36 knockout mice have increased insulin sensitivity, improved insulin signaling and reduced inflammation within the adipose tissue [69]. CD36 contributes to inflammation by promoting macrophage infiltration into adipose tissue and pro-inflammatory cytokine production as well as production of prostaglandins [66]. GPR120 selectively binds omega-3 fatty acids such as α-linolenate and docosahexaenoate [71]. GPR120 signaling mediated by n-3 fatty acids leads to blockade of NF-κB activation by preventing TAK1-dependent phosphorylation of IKKβ resulting in an attenuated inflammatory response [62]. While our studies herein do not address which cell surface fatty acid receptor mediates eicosanoid production, future studies will focus on the signaling cascades activated by free fatty acids.
In sum, our study shows that free fatty acids induce production of pro-inflammatory LTC4 by macrophages and that the genetically obese mouse model, ob/ob, contains elevated levels of LTC4 and 12-HETE in adipose tissue. This production of LTC4 is dependent on the presence of FABPs, presumably via stabilization of LTA4. Previous studies examining the role of lipid metabolites in obesity-induced inflammation and insulin resistance have been limited to examination of 12-HETE [28, 29]. Our results provide evidence that other lipid metabolites such as LTC4 (and LTB4) could be potential mediators of obesity-induced inflammation and may be linked to obesity-linked insulin resistance. Consistent with this, Mothe-Satney et al. [72] have shown that mice deficient in 5-lipoxygenase exhibit reduced levels of inflammatory macrophages and T cells. Moreover, Claria et al. [73] have recently demonstrated that human adipose tissue produces a variety of lipid mediators (including resolvins and leukotrienes) implying that such molecules may play a significant role in insulin resistance in humans. Future studies will focus on the molecular actions of such signaling lipids.
Highlights
Adipose tissue from obese mice contains elevated levels of LTC4 and 12S-HETE.
Macrophages produce LTC4 and 5S-HETE in response to fatty acid stimulation.
Inhibition of Fatty Acid Binding Proteins (FABPs) block LTC4 production.
Acknowledgements
This work was supported by the National Institutes of Health grants DK053189 and AES MN-70-043 to DAB, F32 DK091004 to EKL and NIH DK050456 (The Minnesota Obesity Center). KH was supported by the Cargill Fellowship in Systems Biology and a Doctoral Dissertation Fellowship from the University of Minnesota. We thank Jonathan Marchant for advise on calcium studies and the members of the Bernlohr laboratory for their comments and suggestions in preparation of this manuscript. We also thank the staff of the University of Minnesota Center for Mass Spectrometry and Proteomics and the Minnesota Supercomputing Institute for their assistance.
Abbreviations
- LT
leukotriene
- FABP
fatty acid binding protein
- AFABP
adipocyte fatty acid binding protein
- EFABP
epithelial fatty acid binding protein
- HFABP
heart fatty acid binding protein
- HETE
hydroxyeicosatetraenoic acid
- PG
prostaglandin
- MCP-1
monocyte chemoattractant protein-1
- SV fraction
stromal vascular fraction
- MRM
multiple reaction monitoring
- cPLA2
cytosolic phospholipase A2
- 5LO
5-lipoxygenase
- LTC4S
leukotriene C4 synthase
- COX-2
cyclooxygenase 2
- PPAR
peroxisome proliferator-activated receptor
- TLR4
Toll-like receptor 4
- NF-κB
nuclear factor κB
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–1830. doi: 10.1172/JCI19451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Savage DB, Petersen KF, Shulman GI. Mechanisms of insulin resistance in humans and possible links with inflammation. Hypertension. 2005;45:828–833. doi: 10.1161/01.HYP.0000163475.04421.e4. [DOI] [PubMed] [Google Scholar]
- [3].Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. 2006;116:1793–1801. doi: 10.1172/JCI29069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246. doi: 10.1146/annurev-physiol-021909-135846. [DOI] [PubMed] [Google Scholar]
- [5].Berg AH, Scherer PE. Adipose tissue, inflammation, and cardiovascular disease. Circ Res. 2005;96:939–949. doi: 10.1161/01.RES.0000163635.62927.34. [DOI] [PubMed] [Google Scholar]
- [6].Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808. doi: 10.1172/JCI19246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Patsouris D, Li PP, Thapar D, Chapman J, Olefsky JM, Neels JG. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 2008;8:301–309. doi: 10.1016/j.cmet.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259:87–91. doi: 10.1126/science.7678183. [DOI] [PubMed] [Google Scholar]
- [9].Fain JN. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm. 2006;74:443–477. doi: 10.1016/S0083-6729(06)74018-3. [DOI] [PubMed] [Google Scholar]
- [10].Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, Kitazawa S, Miyachi H, Maeda S, Egashira K, Kasuga M. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494–1505. doi: 10.1172/JCI26498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Bruun JM, Lihn AS, Pedersen SB, Richelsen B. Monocyte chemoattractant protein-1 release is higher in visceral than subcutaneous human adipose tissue (AT): implication of macrophages resident in the AT. J Clin Endocrinol Metab. 2005;90:2282–2289. doi: 10.1210/jc.2004-1696. [DOI] [PubMed] [Google Scholar]
- [12].Di Gregorio GB, Yao-Borengasser A, Rasouli N, Varma V, Lu T, Miles LM, Ranganathan G, Peterson CA, McGehee RE, Kern PA. Expression of CD68 and macrophage chemoattractant protein-1 genes in human adipose and muscle tissues: association with cytokine expression, insulin resistance, and reduction by pioglitazone. Diabetes. 2005;54:2305–2313. doi: 10.2337/diabetes.54.8.2305. [DOI] [PubMed] [Google Scholar]
- [13].Leslie CC. Macrophage phospholipase A2 specific for sn-2-arachidonic acid. Methods Enzymol. 1990;187:216–225. doi: 10.1016/0076-6879(90)87026-y. [DOI] [PubMed] [Google Scholar]
- [14].Burke JE, Dennis EA. Phospholipase A2 structure/function, mechanism, and signaling. J Lipid Res. 2009;50(Suppl):S237–242. doi: 10.1194/jlr.R800033-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Murphy RC, Gijon MA. Biosynthesis and metabolism of leukotrienes. Biochem J. 2007;405:379–395. doi: 10.1042/BJ20070289. [DOI] [PubMed] [Google Scholar]
- [16].Sala A, Folco G, Murphy RC. Transcellular biosynthesis of eicosanoids. Pharmacological reports : PR. 2010;62:503–510. doi: 10.1016/s1734-1140(10)70306-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Capra V, Thompson MD, Sala A, Cole DE, Folco G, Rovati GE. Cysteinyl-leukotrienes and their receptors in asthma and other inflammatory diseases: critical update and emerging trends. Med Res Rev. 2007;27:469–527. doi: 10.1002/med.20071. [DOI] [PubMed] [Google Scholar]
- [18].Busse WW. Leukotrienes and inflammation. Am J Respir Crit Care Med. 1998;157:S210–213. [PubMed] [Google Scholar]
- [19].Peters-Golden M, Henderson WR., Jr. Leukotrienes. N Engl J Med. 2007;357:1841–1854. doi: 10.1056/NEJMra071371. [DOI] [PubMed] [Google Scholar]
- [20].Crooks SW, Stockley RA. Leukotriene B4. Int J Biochem Cell Biol. 1998;30:173–178. doi: 10.1016/s1357-2725(97)00123-4. [DOI] [PubMed] [Google Scholar]
- [21].Costa MF, de Souza-Martins R, de Souza MC, Benjamim CF, Piva B, Diaz BL, Peters-Golden M, Henriques MG, Canetti C, Penido C. Leukotriene B4 mediates gammadelta T lymphocyte migration in response to diverse stimuli. J Leukoc Biol. 2010;87:323–332. doi: 10.1189/jlb.0809563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Matsukawa A, Hogaboam CM, Lukacs NW, Lincoln PM, Strieter RM, Kunkel SL. Endogenous monocyte chemoattractant protein-1 (MCP-1) protects mice in a model of acute septic peritonitis: cross-talk between MCP-1 and leukotriene B4. J Immunol. 1999;163:6148–6154. [PubMed] [Google Scholar]
- [23].Samuelsson B. Leukotrienes: mediators of immediate hypersensitivity reactions and inflammation. Science. 1983;220:568–575. doi: 10.1126/science.6301011. [DOI] [PubMed] [Google Scholar]
- [24].Hallstrand TS, Henderson WR., Jr. Role of leukotrienes in exercise-induced bronchoconstriction. Curr Allergy Asthma Rep. 2009;9:18–25. doi: 10.1007/s11882-009-0003-8. [DOI] [PubMed] [Google Scholar]
- [25].Mehrotra AK, Henderson WR., Jr. The role of leukotrienes in airway remodeling. Curr Mol Med. 2009;9:383–391. doi: 10.2174/156652409787847209. [DOI] [PubMed] [Google Scholar]
- [26].Martinez-Clemente M, Claria J, Titos E. The 5-lipoxygenase/leukotriene pathway in obesity, insulin resistance, and fatty liver disease. Curr Opin Clin Nutr Metab Care. 2011;14:347–353. doi: 10.1097/MCO.0b013e32834777fa. [DOI] [PubMed] [Google Scholar]
- [27].Chakrabarti SK, Cole BK, Wen Y, Keller SR, Nadler JL. 12/15-lipoxygenase products induce inflammation and impair insulin signaling in 3T3-L1 adipocytes. Obesity (Silver Spring) 2009;17:1657–1663. doi: 10.1038/oby.2009.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Chakrabarti SK, Wen Y, Dobrian AD, Cole BK, Ma Q, Pei H, Williams MD, Bevard MH, Vandenhoff GE, Keller SR, Gu J, Nadler JL. Evidence for activation of inflammatory lipoxygenase pathways in visceral adipose tissue of obese Zucker rats. Am J Physiol Endocrinol Metab. 2011;300:E175–187. doi: 10.1152/ajpendo.00203.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Dobrian AD, Lieb DC, Ma Q, Lindsay JW, Cole BK, Ma K, Chakrabarti SK, Kuhn NS, Wohlgemuth SD, Fontana M, Nadler JL. Differential expression and localization of 12/15 lipoxygenases in adipose tissue in human obese subjects. Biochem Biophys Res Commun. 2010;403:485–490. doi: 10.1016/j.bbrc.2010.11.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Spite M, Hellmann J, Tang Y, Mathis SP, Kosuri M, Bhatnagar A, Jala VR, Haribabu B. Deficiency of the leukotriene B4 receptor, BLT-1, protects against systemic insulin resistance in diet-induced obesity. J Immunol. 2011;187:1942–1949. doi: 10.4049/jimmunol.1100196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Mehrabian M, Schulthess FT, Nebohacova M, Castellani LW, Zhou Z, Hartiala J, Oberholzer J, Lusis AJ, Maedler K, Allayee H. Identification of ALOX5 as a gene regulating adiposity and pancreatic function. Diabetologia. 2008;51:978–988. doi: 10.1007/s00125-008-1002-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Horrillo R, Gonzalez-Periz A, Martinez-Clemente M, Lopez-Parra M, Ferre N, Titos E, Moran-Salvador E, Deulofeu R, Arroyo V, Claria J. 5-lipoxygenase activating protein signals adipose tissue inflammation and lipid dysfunction in experimental obesity. J Immunol. 2010;184:3978–3987. doi: 10.4049/jimmunol.0901355. [DOI] [PubMed] [Google Scholar]
- [33].Storch J, Corsico B. The emerging functions and mechanisms of mammalian fatty acid-binding proteins. Annu Rev Nutr. 2008;28:73–95. doi: 10.1146/annurev.nutr.27.061406.093710. [DOI] [PubMed] [Google Scholar]
- [34].Furuhashi M, Hotamisligil GS. Fatty acid-binding proteins: role in metabolic diseases and potential as drug targets. Nat Rev Drug Discov. 2008;7:489–503. doi: 10.1038/nrd2589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Makowski L, Boord JB, Maeda K, Babaev VR, Uysal KT, Morgan MA, Parker RA, Suttles J, Fazio S, Hotamisligil GS, Linton MF. Lack of macrophage fatty-acid-binding protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis. Nat Med. 2001;7:699–705. doi: 10.1038/89076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Hotamisligil GS, Johnson RS, Distel RJ, Ellis R, Papaioannou VE, Spiegelman BM. Uncoupling of obesity from insulin resistance through a targeted mutation in aP2, the adipocyte fatty acid binding protein. Science. 1996;274:1377–1379. doi: 10.1126/science.274.5291.1377. [DOI] [PubMed] [Google Scholar]
- [37].Dickinson Zimmer JS, Voelker DR, Bernlohr DA, Murphy RC. Stabilization of leukotriene A4 by epithelial fatty acid-binding protein in the rat basophilic leukemia cell. J Biol Chem. 2004;279:7420–7426. doi: 10.1074/jbc.M311404200. [DOI] [PubMed] [Google Scholar]
- [38].Richieri GV, Anel A, Kleinfeld AM. Interactions of long-chain fatty acids and albumin: determination of free fatty acid levels using the fluorescent probe ADIFAB. Biochemistry. 1993;32:7574–7580. doi: 10.1021/bi00080a032. [DOI] [PubMed] [Google Scholar]
- [39].Buczynski MW, Stephens DL, Bowers-Gentry RC, Grkovich A, Deems RA, Dennis EA. TLR-4 and sustained calcium agonists synergistically produce eicosanoids independent of protein synthesis in RAW264.7 cells. J Biol Chem. 2007;282:22834–22847. doi: 10.1074/jbc.M701831200. [DOI] [PubMed] [Google Scholar]
- [40].Mothe-Satney I, Filloux C, Amghar H, Pons C, Bourlier V, Galitzky J, Grimaldi PA, Feral CC, Bouloumie A, Van Obberghen E, Neels JG. Adipocytes secrete leukotrienes: contribution to obesity-associated inflammation and insulin resistance in mice. Diabetes. 2012;61:2311–2319. doi: 10.2337/db11-1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Hetu PO, Riendeau D. Down-regulation of microsomal prostaglandin E2 synthase-1 in adipose tissue by high-fat feeding. Obesity (Silver Spring, Md.) 2007;15:60–68. doi: 10.1038/oby.2007.514. [DOI] [PubMed] [Google Scholar]
- [42].Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond) 2008;32:451–463. doi: 10.1038/sj.ijo.0803744. [DOI] [PubMed] [Google Scholar]
- [43].Anderson EK, Gutierrez DA, Hasty AH. Adipose tissue recruitment of leukocytes. Curr Opin Lipidol. 2010;21:172–177. doi: 10.1097/MOL.0b013e3283393867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Busse WW, Lemanske RF., Jr. Asthma. N Engl J Med. 2001;344:350–362. doi: 10.1056/NEJM200102013440507. [DOI] [PubMed] [Google Scholar]
- [45].Montuschi P, Kharitonov SA, Ciabattoni G, Barnes PJ. Exhaled leukotrienes and prostaglandins in COPD. Thorax. 2003;58:585–588. doi: 10.1136/thorax.58.7.585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Hertzel AV, Hellberg K, Reynolds JM, Kruse AC, Juhlmann BE, Smith AJ, Sanders MA, Ohlendorf DH, Suttles J, Bernlohr DA. Identification and characterization of a small molecule inhibitor of Fatty Acid binding proteins. J Med Chem. 2009;52:6024–6031. doi: 10.1021/jm900720m. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Reiss TF, Altman LC, Chervinsky P, Bewtra A, Stricker WE, Noonan GP, Kundu S, Zhang J. Effects of montelukast (MK-0476), a new potent cysteinyl leukotriene (LTD4) receptor antagonist, in patients with chronic asthma. J Allergy Clin Immunol. 1996;98:528–534. doi: 10.1016/s0091-6749(96)70086-6. [DOI] [PubMed] [Google Scholar]
- [48].Lindstrom P. The physiology of obese-hyperglycemic mice [ob/ob mice] TheScientificWorldJournal. 2007;7:666–685. doi: 10.1100/tsw.2007.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Gonzalez-Periz A, Horrillo R, Ferre N, Gronert K, Dong B, Moran-Salvador E, Titos E, Martinez-Clemente M, Lopez-Parra M, Arroyo V, Claria J. Obesity-induced insulin resistance and hepatic steatosis are alleviated by omega-3 fatty acids: a role for resolvins and protectins. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2009;23:1946–1957. doi: 10.1096/fj.08-125674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Zarini S, Gijon MA, Ransome AE, Murphy RC, Sala A. Transcellular biosynthesis of cysteinyl leukotrienes in vivo during mouse peritoneal inflammation. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:8296–8301. doi: 10.1073/pnas.0903851106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Engfeldt P, Arner P. Lipolysis in human adipocytes, effects of cell size, age and of regional differences. Horm Metab Res Suppl. 1988;19:26–29. [PubMed] [Google Scholar]
- [52].Wueest S, Rapold RA, Rytka JM, Schoenle EJ, Konrad D. Basal lipolysis, not the degree of insulin resistance, differentiates large from small isolated adipocytes in high-fat fed mice. Diabetologia. 2009;52:541–546. doi: 10.1007/s00125-008-1223-5. [DOI] [PubMed] [Google Scholar]
- [53].Field CJ, Angel A, Clandinin MT. Relationship of diet to the fatty acid composition of human adipose tissue structural and stored lipids. Am J Clin Nutr. 1985;42:1206–1220. doi: 10.1093/ajcn/42.6.1206. [DOI] [PubMed] [Google Scholar]
- [54].Raatz SK, Bibus D, Thomas W, Kris-Etherton P. Total fat intake modifies plasma fatty acid composition in humans. J Nutr. 2001;131:231–234. doi: 10.1093/jn/131.2.231. [DOI] [PubMed] [Google Scholar]
- [55].Gallou-Kabani C, Vige A, Gross MS, Rabes JP, Boileau C, Larue-Achagiotis C, Tome D, Jais JP, Junien C. C57BL/6J and A/J mice fed a high-fat diet delineate components of metabolic syndrome. Obesity (Silver Spring) 2007;15:1996–2005. doi: 10.1038/oby.2007.238. [DOI] [PubMed] [Google Scholar]
- [56].Norris PC, Reichart D, Dumlao DS, Glass CK, Dennis EA. Specificity of eicosanoid production depends on the TLR-4-stimulated macrophage phenotype. J Leukoc Biol. 2011 doi: 10.1189/jlb.0311153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Rossi A, Acquaviva AM, Iuliano F, Di Paola R, Cuzzocrea S, Sautebin L. Up-regulation of prostaglandin biosynthesis by leukotriene C4 in elicited mice peritoneal macrophages activated with lipopolysaccharide/interferon-{gamma} J Leukoc Biol. 2005;78:985–991. doi: 10.1189/jlb.1004619. [DOI] [PubMed] [Google Scholar]
- [58].Kadotani A, Tsuchiya Y, Hatakeyama H, Katagiri H, Kanzaki M. Different impacts of saturated and unsaturated free fatty acids on COX-2 expression in C(2)C(12) myotubes. Am J Physiol Endocrinol Metab. 2009;297:E1291–1303. doi: 10.1152/ajpendo.00293.2009. [DOI] [PubMed] [Google Scholar]
- [59].Norris PC, Dennis EA. Omega-3 fatty acids cause dramatic changes in TLR4 and purinergic eicosanoid signaling. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:8517–8522. doi: 10.1073/pnas.1200189109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Borgeat P, Samuelsson B. Arachidonic acid metabolism in polymorphonuclear leukocytes: unstable intermediate in formation of dihydroxy acids. Proceedings of the National Academy of Sciences of the United States of America. 1979;76:3213–3217. doi: 10.1073/pnas.76.7.3213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Silverstein RL, Febbraio M. CD36, a scavenger receptor involved in immunity, metabolism, angiogenesis, and behavior. Sci Signal. 2009;2:re3. doi: 10.1126/scisignal.272re3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, Olefsky JM. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. 2010;142:687–698. doi: 10.1016/j.cell.2010.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Lee JY, Plakidas A, Lee WH, Heikkinen A, Chanmugam P, Bray G, Hwang DH. Differential modulation of Toll-like receptors by fatty acids: preferential inhibition by n-3 polyunsaturated fatty acids. J Lipid Res. 2003;44:479–486. doi: 10.1194/jlr.M200361-JLR200. [DOI] [PubMed] [Google Scholar]
- [64].Scott T, Owens MD. Thrombocytes respond to lipopolysaccharide through Toll-like receptor-4, and MAP kinase and NF-kappaB pathways leading to expression of interleukin-6 and cyclooxygenase-2 with production of prostaglandin E2. Molecular immunology. 2008;45:1001–1008. doi: 10.1016/j.molimm.2007.07.035. [DOI] [PubMed] [Google Scholar]
- [65].Pal D, Dasgupta S, Kundu R, Maitra S, Das G, Mukhopadhyay S, Ray S, Majumdar SS, Bhattacharya S. Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med. doi: 10.1038/nm.2851. [DOI] [PubMed] [Google Scholar]
- [66].Kuda O, Jenkins CM, Skinner JR, Moon SH, Su X, Gross RW, Abumrad NA. CD36 protein is involved in store-operated calcium flux, phospholipase A2 activation, and production of prostaglandin E2. The Journal of biological chemistry. 2011;286:17785–17795. doi: 10.1074/jbc.M111.232975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Pacheco P, Vieira-de-Abreu A, Gomes RN, Barbosa-Lima G, Wermelinger LB, Maya-Monteiro CM, Silva AR, Bozza MT, Castro-Faria-Neto HC, Bandeira-Melo C, Bozza PT. Monocyte chemoattractant protein-1/CC chemokine ligand 2 controls microtubule-driven biogenesis and leukotriene B4-synthesizing function of macrophage lipid bodies elicited by innate immune response. J Immunol. 2007;179:8500–8508. doi: 10.4049/jimmunol.179.12.8500. [DOI] [PubMed] [Google Scholar]
- [68].Su X, Abumrad NA. Cellular fatty acid uptake: a pathway under construction. Trends Endocrinol Metab. 2009;20:72–77. doi: 10.1016/j.tem.2008.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Kennedy DJ, Kuchibhotla S, Westfall KM, Silverstein RL, Morton RE, Febbraio M. A CD36-dependent pathway enhances macrophage and adipose tissue inflammation and impairs insulin signalling. Cardiovasc Res. 2011;89:604–613. doi: 10.1093/cvr/cvq360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Martin C, Chevrot M, Poirier H, Passilly-Degrace P, Niot I, Besnard P. CD36 as a lipid sensor. Physiol Behav. 2011;105:36–42. doi: 10.1016/j.physbeh.2011.02.029. [DOI] [PubMed] [Google Scholar]
- [71].Hirasawa A, Tsumaya K, Awaji T, Katsuma S, Adachi T, Yamada M, Sugimoto Y, Miyazaki S, Tsujimoto G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med. 2005;11:90–94. doi: 10.1038/nm1168. [DOI] [PubMed] [Google Scholar]
- [72].Mothe-Satney I, Filloux C, Amghar H, Pons C, Bourlier V, Galitzky J, Grimaldi PA, Féral CC, Bouloumié A, Van Obberghen E, Neels JG. Adipocytes Secrete Leukotrienes; Contribution to Obesity-Associated Inflammation and Insulin Resistance in Mice. Diabetes. 2012;61:2311–2319. doi: 10.2337/db11-1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Clària J, Nguyen BT, Madenci A, Ozaki CK, Serhan CN. Diversity of lipid mediators in human adipose tissue. Am J Physiol. Cell Physiol. doi: 10.1152/ajpcell.00351.2012. doi:10.1152/ajpcell.00351. [DOI] [PMC free article] [PubMed] [Google Scholar]





