+ All Categories
Home > Documents > Lysophospholipid Receptors (Signaling and Biochemistry) || Regulation of the Nuclear Hormone...

Lysophospholipid Receptors (Signaling and Biochemistry) || Regulation of the Nuclear Hormone...

Date post: 03-Dec-2016
Category:
Upload: wouter
View: 213 times
Download: 1 times
Share this document with a friend
24
349 CHAPTER 16 Regulation of the Nuclear Hormone Receptor PPARγ by Endogenous Lysophosphatidic Acids (LPAs) RYOKO TSUKAHARA, TAMOTSU TSUKAHARA, and GABOR TIGYI 16.1. INTRODUCTION 16.1.1. Lysophosphatidic Acid (LPA) in Vascular Biology LPA is produced in serum after the activation of multiple biochemical path- ways linked to platelet activation (1–4). The concentration of LPA in plasma is in the nanomolar range, whereas it can reach concentrations as high as 10 μM in serum after blood clotting (5–7). LPA production in blood requires autotaxin (ATX), which is a secreted lysophospholipase D that generates LPA from lysophosphatidylcholine (LPC) and lysophosphatidylserine (LPS) (8, 9). LPA plays an important role in vascular development (10). ATX-deficient mice die at approximately embryonic day 9.5 with severe vessel development defects in the yolk sac and enlarged embryonic blood vessels (10, 11). Mice that overexpress ATX have elevated plasma LPA levels and show bleeding diathesis, whereas ATX +/heterozygous mice have almost half the plasma LPA levels and are prone to thrombosis (12). LPA promotes proliferation and migration of vascular smooth muscle cells (VSMCs) (13–15). LPA also increases endothelial permeability (16–18) and induces E-selectin, vascular cell adhesion molecule-1, and vascular endothelial growth factor-C expression in human endothelial cells (19, 20). Hayashi et al. reported that LPA promotes VSMC dedifferentiation from the contractile phenotype to a secretory phenotype and stimulates VSMC signaling pathways in vitro (14). Lysophospholipid Receptors: Signaling and Biochemistry, First Edition. Edited by Jerold Chun, Timothy Hla, Sara Spiegel, and Wouter Moolenaar. © 2013 John Wiley & Sons, Inc. Published 2013 by John Wiley & Sons, Inc.
Transcript

349

CHAPTER 16

Regulation of the Nuclear Hormone Receptor PPARγ by Endogenous Lysophosphatidic Acids (LPAs)RYOKO TSUKAHARA, TAMOTSU TSUKAHARA, and GABOR TIGYI

16.1. INTRODUCTION

16.1.1. Lysophosphatidic Acid (LPA) in Vascular Biology

LPA is produced in serum after the activation of multiple biochemical path-ways linked to platelet activation (1–4). The concentration of LPA in plasma is in the nanomolar range, whereas it can reach concentrations as high as 10 μM in serum after blood clotting (5–7). LPA production in blood requires autotaxin (ATX), which is a secreted lysophospholipase D that generates LPA from lysophosphatidylcholine (LPC) and lysophosphatidylserine (LPS) (8, 9).

LPA plays an important role in vascular development (10). ATX-deficient mice die at approximately embryonic day 9.5 with severe vessel development defects in the yolk sac and enlarged embryonic blood vessels (10, 11). Mice that overexpress ATX have elevated plasma LPA levels and show bleeding diathesis, whereas ATX+/− heterozygous mice have almost half the plasma LPA levels and are prone to thrombosis (12).

LPA promotes proliferation and migration of vascular smooth muscle cells (VSMCs) (13–15). LPA also increases endothelial permeability (16–18) and induces E-selectin, vascular cell adhesion molecule-1, and vascular endothelial growth factor-C expression in human endothelial cells (19, 20). Hayashi et al. reported that LPA promotes VSMC dedifferentiation from the contractile phenotype to a secretory phenotype and stimulates VSMC signaling pathways in vitro (14).

Lysophospholipid Receptors: Signaling and Biochemistry, First Edition. Edited by Jerold Chun, Timothy Hla, Sara Spiegel, and Wouter Moolenaar.© 2013 John Wiley & Sons, Inc. Published 2013 by John Wiley & Sons, Inc.

350 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

Many of the cellular responses elicited by LPA, including platelet activa-tion (21–23), endothelial cell activation (21), proliferation, migration, and phenotypic modulation of VSMCs (13–15), can potentially be involved in neo-intima formation. Oxidative modification of low-density lipoprotein (LDL) is considered to be an early event in arterial wall remodeling leading to athero-sclerosis (24, 25), and uptake and oxidation of LDL in the arterial wall is an important mechanism in the pathogenesis of atherosclerosis (26). It has been shown that alkyl-LPA is formed during mild oxidation of LDL (mox-LDL) (21, 27). In addition, the lipid-rich core of human atherosclerotic plaques, which accumulates oxidized lipids including mox-LDL, contains several species of acyl- and alkyl-LPA (21). LPAs accumulated in human atheroscle-rotic plaques have the potential to activate platelets and to initiate thrombus formation upon plaque rupture (3, 22). As a result of plaque rupture, LPA becomes exposed to circulating platelets, initiating activation, which in turn may contribute to the induction of thrombosis, leading to myocardial infarc-tion and stroke. A clinical study has shown that the serum LPA level is signifi-cantly elevated in patients with acute myocardial infarction (28).

16.1.2. Neointima Formation Induced by LPA

Atherosclerosis is the leading cause of death and cardiovascular morbidity in developed countries. Neointimal lesions are characterized by the accumulation of cells within the arterial wall and are an initial step in the pathogenesis of atherosclerosis, which ultimately leads to the ischemic syndromes of the heart and stroke (29, 30).

Yoshida et al. first reported that LPA species containing unsaturated fatty acyl groups 16:1, 18:1, and 18:2 induced neointima formation when injected intralumenally into the rat carotid artery, whereas saturated acyl-LPA species with the same number of carbons were inactive (31). This model comes close to the pathophysiological response seen in humans when mechanical injury is not the cause of the arterial wall remodeling leading to neointimal lesions. In Yoshida’s model, LPA was injected through the external carotid artery into a ligated section of the common carotid artery (CCA) that was rinsed free of blood and maintained close to the mean arterial perfusion pressure. There was no mechanical injury or removal of endothelial cells in the CCA. A brief 1-hour exposure to unsaturated but not to saturated species of LPA caused neointima development. Our group found that the LPA-elicited neointima was not mediated by the LPA G protein-coupled receptors (GPCRs) LPA1 and LPA2, which are the major LPA receptor subtypes expressed in the vessel wall (32). A peroxisome proliferator-activating receptor γ (PPARγ)-specific inhibitor, GW9662, abolished the LPA-induced neointima formation, suggest-ing that arterial wall remodeling elicited by LPA is a PPARγ-mediated response (33). Conversely, the PPARγ-specific agonist rosiglitazone (ROSI, Fig. 16.1) also elicited neointima formation in the same model. Genetic evi-dence also supports the role of PPARγ in arterial wall remodeling. When

INTRODUCTION 351

conditional knockout PPARγ−/− mice targeted to the endothelial cells, VSMCs, and cells of the macrophage/monocyte lineage were exposed to alkyl-LPA (Fig. 16.1) or ROSI, neither elicited neointima formation (32). It is important to note that in a carotid injury model, ROSI diminished the size of the neo-intima (34). The opposite effect of ROSI and LPA in the injury model versus the noninjury model indicates differences in the mechanism underlying vas-cular wall remodeling in the two models (32, 34). These results, combined with the observation that GW9662 abolished neointima in response to ROSI or alkyl-LPA (33), suggest that PPARγ is required for LPA-induced neointima formation in the absence of vascular wall injury.

Subramanian et al. reported that LPA1 and LPA3 GPCRs play an important role in injury-induced neointima formation (35). Using a wire injury model, these authors showed that neointima formation was inhibited by the LPA1 and LPA3 inhibitor Ki16425, and that vascular wall remodeling induced by LPA was inhibited by short-term knockdown of either LPA1 or LPA3 with siRNA (35). In sharp contrast with the findings of Subramanian et al., LPA1

−/− mice in our noninjury model developed neointimal lesions in response to alkyl-LPA stimulation. Moreover, although LPA1 and LPA3 GPCRs are respectively 12 and 100 times less sensitive to alkyl-LPA than LPA (36), alkyl-LPA is more potent than LPA in inducing neointima formation (33). They also observed that Mac-2 positive cells were accumulated in the neointimal area after vas-cular injury (35); in contrast, our noninjury model showed very few macro-phages in the neointimal region induced by alkyl-LPA or ROSI (32), suggesting that the cellular elements leading to neointima formation are different between the two models.

Figure 16.1. Structures of the lysophosphatidic acids, ROSI, and PIO.

352 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

Although some clinical studies in diabetic patients have shown a reduction in carotid artery wall thickness after treatment with thiazolidinediones (TZDs) (37, 38), other studies suggested that ROSI increases the risk of myocardial infarction and death from cardiovascular causes (39–41), indicating that TZD therapy may actually increase the risk of cardiovascular events. In 2010, the U.S. Food and Drug Administration restricted the use of ROSI to patients with type 2 diabetes due to the potential for cardiovascular ischemic risks, including heart attack.

16.1.3. PPARγ: An Intracellular LPA Receptor

Only unsaturated acyl-LPA species induce neointima formation; saturated acyl-LPA species are inactive (31, 33), indicating the need for an unsaturated fatty acid in the activation of the mechanism that underlies the formation of neointima. The structure–activity relationship of neointima induction by LPA does not match that of the known LPA GPCRs because saturated LPA species activate the LPA GPCRs (33, 36). However, the structure–activity relationship of PPARγ activation by LPA species matches that of the neointima response in vivo (33). ROSI and the unsaturated acyl-LPA species all activated the peroxisome activator response element-containing acyl-coenzyme A oxidase-luciferase (PPRE-Acox-Luc) reporter construct, whereas all saturated species, 2,3-cyclic phosphatidic acid (CPA, Fig. 16.1), and the related lipid mediator sphingosine-1-phosphate were inactive (42).

PPARs are members of the nuclear hormone receptor superfamily, many of which function as lipid-activated transcription factors (43). There are three PPAR isoforms that include PPARα, β/δ, and γ. PPARγ plays an important role in regulating lipid metabolism, glucose homeostasis, cell differentiation, and motility (44–46). PPARγ has two isoforms, PPARγ1 and PPARγ2. PPARγ2 differs from PPARγ1 only by the addition of 30 amino acids at the N-terminus, caused by differential promoter usage and alternative splicing (47). PPARγ1 is expressed ubiquitously in almost all tissues, whereas PPARγ2 is highly expressed only in the adipose tissue (48). Genetic deletion of PPARγ1 causes embryonic mortality (49). In contrast, deletion of PPARγ2 causes minimal alterations in lipid metabo-lism (50). PPARγ heterodimerizes with the retinoid X receptor α (RXRα), and it is the ligand-binding domain (LBD) of PPARγ that interacts with the agonists, including LPA (51). The PPARγ-RXRα heterodimer binds the peroxisome proliferator response element (PPRE) in the promoter region of the target genes. In the absence of ligands, the corepressors nuclear receptor co-repressor 1 (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) (52–54) bind to the heterodimer to suppress target gene activation. Upon ligand binding, PPARγ undergoes a conformational change that facili-tates the dissociation of the corepressors and recruits the p300 coactivator and the PGC-1α coactivator (55, 56), resulting in target gene transcription (57).

A number of putative physiological agonists of PPARγ have been identified, including 15d-PGJ2 (58), oxidatively modified fatty acids (59–61), select

INTRODUCTION 353

species of LPA and alkyl-LPA (33, 42, 62), oxidized phospholipids (63), and nitrated fatty acids (61). Among these ligands, alkyl-LPA stands out with an equilibrium binding constant of 60 nM (42) that is similar to that of the TZD class of synthetic agonists. The TZD class of antidiabetics, including ROSI, pioglitazone (PIO), and troglitazone, are full agonists of PPARγ (64–67).

However, despite the beneficial effects of PPARγ on glucose and lipid homeostasis, excessive PPARγ activity can be deleterious. PPARγ agonists promote adipocytic differentiation of 3T3-L1 cells and also stimulate the uptake of LDL by macrophages, leading to foam cell formation in the arterial wall (68). Our results suggest that LPA-mediated activation of PPARγ can also contribute to vascular wall pathologies.

PPARγ plays an important role in the cardiovascular system. PPARγ is expressed in all cell types of the vessel wall (69–71), as well as in macrophages and histiocytes (72). In human atherosclerotic plaques and neointimal lesions, PPARγ expression is upregulated in VSMCs (27), endothelial cells (69), and macrophages (72). PPARγ expression is also elevated in neointimal lesions after mechanical injury to the endothelium (27).

16.1.4. CPA: An Endogenous Antagonist of PPARγ

CPA, an analog of LPA with a five-atom ring linking the phosphate to two of the glycerol carbons (Fig. 16.1), is found in diverse organisms from slime mold to humans (73); its functions are largely unknown. The concentration of CPA in human serum is estimated to be ∼10 nM, ∼100-fold lower than that of LPA (74, 75). Although CPA is structurally similar to LPA, it shows several unique actions. CPA inhibits cell proliferation (76), induces actin stress fiber formation (76), promotes differentiation and survival of cultured embryonic hippocampal neurons (77), inhibits LPA-induced platelet aggregation (78), and suppresses cancer cell invasion and metastasis in vitro and in vivo (79–81). Our group showed that CPA negatively regulates PPARγ functions by stabilizing the SMRT–PPARγ complex (82). We showed that CPA is generated intracellularly by phospholipase D2 (PLD2) (82). 5-Fluoro-2-indolyl des-chlorohalopemide (FIPI) is an inhibitor of both PLD1 and PLD2 isozymes (83). FIPI inhibits PLD2-mediated CPA production in human peripheral mononuclear cells stimulated with LPS (82). We also demonstrated that activation of PLD2-mediated CPA production or topical application of CPA together with PPARγ agonists prevents neointima formation, adipocytic differentiation, lipid accu-mulation, and the upregulation of PPARγ target gene transcription in mouse macrophages (82). These findings support our hypothesis that CPA is an endog-enous antagonist of PPARγ.

In the following sections, we describe some of the fundamental methodolo-gies that we and others have used in probing the actions of LPA and CPA on PPARγ in vitro and in vivo.

354 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

16.2. METHODS PROBING PPARγ FUNCTION WITH LPA ANALOGS

16.2.1. Reporter Gene Assay for Ligand Activation of PPARγ

The reporter gene assay is a sensitive method for monitoring ligand-induced gene expression. The determination of PPARγ activation in cells transiently transfected with the PPRE-ACox-Luc or TK-MH100-Luc reporter gene con-struct has been reported previously (42, 82) (Fig. 16.2a, b). We used the B103 rat neuroblastoma cell line because this cell line lacks LPA GPCRs LPA1, LPA2, and LPA3 and expresses very low levels of endogenous PPARγ, making it an ideal low-background cell type for transfection studies. B103 cells, 3.0 × 104 per well, were plated to 96-well plates the day before transfection. Using Lipo-fectAMINE 2000, the cells were transfected with 125 ng of the reporter plasmid (PPRE-ACox-Luc or TK-MH100-Luc), 62.5 ng of pcDNA3.1-PPARγ or pCMX-Gal4-PPARγ, and 12.5 ng of SV40-β-galactosidase (Promega, Madison, WI), the latter to monitor transfection efficiency. Twenty hours post-transfection, 10 μM ROSI (ALEXIS Biochemicals, San Diego, CA), alkyl-LPA 18:1, or CPA 18:1 (Avanti Polar Lipids, Alabaster, AL) dissolved in 1% dimethyl sulfoxide (DMSO) and mixed with Opti-MEM I (Invitrogen, Grand Island, NY) and 1% fetal bovine serum (FBS) were applied to the cells for 20 hours. Luciferase and β-galactosidase activities were measured with the Steady-Glo® luciferase assay system (Promega) and the Galacto-Light Plus™ system (Applied Biosystems), respectively. Samples were run in quadruplicate, and the means ± SE were calculated. Representative data are shown in Figure 16.2c.

16.2.2. Competition Ligand-Binding Assay to PPARγ

PPARγ consists of an LBD and a DNA-binding domain (DBD) (Fig. 16.3). To test whether a lysophospholipid interacts with the PPARγ LBD, competition ligand-binding assays were performed (Fig. 16.4a). Hexahistidine (His6) epitope-tagged PPARγ LBD fusion protein or empty vector control containing the His6 and thrombin recognition site was expressed in BL-21 (DE3) E. coli cells (Invi-trogen). Transformed BL-21 cells were induced using 0.3 mM isopropyl 1-β-D-galactopyranoside (Fisher Scientific, Waltham, MA) for 12 hours at 25°C and were collected by centrifugation. The PPARγ LBD was extracted with lysis buffer (50 mM HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid) –KOH, pH 6.8, 200 mM NaCl, 5 mM dithiothreitol [DTT], 1 mM phenylmeth-anesulfonyl fluoride, 0.5% Triton X-100, and 15% glycerol) and centrifuged at 12,000 × g for 20 minutes. The supernatant (1 mL) was incubated with 50 μL of TALON metal affinity resin (BD Biosciences, San Jose, CA) at 4°C in the lysis buffer for 1 hour. The resin was washed five times with wash buffer (50 mM HEPES-KOH, pH 6.8, 200 mM NaCl, 5 mM DTT, 15% glycerol, and 5 mM imidazole) and eluted with 150 mM imidazole in wash buffer. The purity of the PPARγ LBD was determined using sodium dodecyl sulfate (SDS) polyacryl-amide gel electrophoresis followed by Coomassie Blue staining and Western blot analysis using an antibody to PPARγ (sc-7196, Santa Cruz Biotechnology,

METHODS PROBING PPARγ FUNCTION WITH LPA ANALOGS 355

Santa Cruz, CA). [32P]alkyl-LPA 18:1 was synthesized from 1-O-octadecenyl-sn-glycerol 18:1 using recombinant 1,2-diacylglycerol kinase (Calbiochem, Darmstadt, Germany). 1-O-Octadecenyl-sn-glycerol 18:1 was solubilized in 20 μL of an octyl-β-glucoside/cardiolipin solution (7.5% octyl-β-glucoside and 5 mM cardiolipin in 0.5% Triton X-100) by sonication in a bath sonicator (Branson, Danbury, CT) for 30 seconds. 1-O-Octadecenyl-sn-glycerol 18:1/octyl-β-glucoside/cardiolipin solution was mixed with 50 μL of reaction buffer

Figure 16.2. CPA inhibits PPARγ-dependent gene expression. (a, b) Schematic diagram of the reporter gene assay using PPARγ and its reporter plasmids. PPARγ agonists induce luciferase transcription, whereas the PPARγ antagonist inhibits PPARγ agonist-induced reporter gene transcription. (c) CPA suppresses ROSI-induced PPARγ-dependent reporter gene activation in B103 cells. B103 cells (3.0 × 104) were transfected with reporter plasmid (PPRE-ACox-Luc or TK-MH100-Luc), pcDNA3.1-PPARγ or pCMX-Gal4-PPARγ, and SV40-β-galactosidase. After transfection, the cells were exposed to 10 μM CPA with or without ROSI (10 μM) for 20 h, and luciferase activities were measured. Data represent mean ± SEM; n = 4. (See color insert.)

356 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

Figure 16.3. The domain structure of PPARγ1 and PPAR γ2. Human PPARγ1 and PPAR γ2 proteins are 53 and 57 kDa, respectively. The two PPARγ isoforms differ only 30 amino acids at the N-terminus. Domains C and E represent DNA-binding domain (DBD) and ligand-binding domain (LBD), respectively.

DNA-bindingdomain (DBD)

30 aa

N-terminaldomain

C-terminaldomain

Hingeregion

A/B C D E F

A/B C D E F

Ligand-bindingdomain (LBD)

PPARg 1

PPARg 2

Figure 16.4. CPA is a high-affinity ligand of PPARγ. (a) Schematic diagram of competi-tive ligand-binding assay using LPA, alkyl-LPA, ROSI, and CPA. (b) Competitive displacement of 5 nM [3H]-ROSI from PPARγ-LBD was determined using 2.5 μM cold ROSI, LPA 18:1, or CPA. Data are mean ± SEM; **p < 0.01.

METHODS PROBING PPARγ FUNCTION WITH LPA ANALOGS 357

(100 mM imidazole HCl, pH 6.6, 100 mM NaCl, 25 mM MgCl2, and 2 mM EGTA), 2 μL of 100 mM DTT (freshly prepared), 10 μL of diluted 1,2-diacylglycerol kinase, and water to a total volume of 90 μL. The reaction was started by the addition of 10 μL of [γ-32P]ATP (10 mCi/mL; specific activity, 111TBq/nmol; PerkinElmer, Waltham, MA) and incubated at 25°C for 1 hour. Lipids were extracted by a modification of the method of Bligh and Dyer (84). NaCl (1 M) was added to bring the aqueous volume to 0.8 mL, and lipids were extracted with 3 mL of chloroform/methanol (1:2, v/v). One milliliter of chloro-form and 1 mL 1 M NaCl were added and the phases were separated by cen-trifugation (3000 × g, 5 minutes). The lower phase was collected and dried under nitrogen. The extract was dissolved in 50 μL chloroform/methanol (1:2, v/v) and was spotted on a Silica Gel 60 (Merck, Whitehouse Station, NJ) thin-layer chro-matography plate. Plates were developed with chloroform/methanol/acetic acid (65:15:5, v/v) and subjected to autoradiography. The radioactive spot corre-sponding to alkyl-LPA 18:1 was scraped off from the plate and eluted using chloroform/methanol (1:2, v/v). The radioligand bound to the PPARγ LBD fusion protein was precipitated using 36% (w/v) polyethylene glycol 8000 (Fisher Scientific) in the presence of 3.3% (w/v) γ-globulin (Sigma-Aldrich, St. Louis, MO), collected on DEAE-81 filter disks (Whatman) and quantified by liquid scintillation counting. Nonspecific binding was determined in the presence of 10 μM cold alkyl-LPA. For competition binding assays, 1 μg of His6-PPARγ LBD protein was incubated in 200 μL of binding buffer in the presence of 5 nM [3H]ROSI or 5 nM [32P]alkyl-LPA with or without 2.5 μM cold compounds at 18°C for 1 hour. The radioactive ligand-bound His6-PPARγ LBD was collected on DEAE-81 filter disks. The disks were washed three times with wash buffer (50 mM HEPES, pH 6.8, 100 mM NaCl, 5 mM DTT), and bound radioactivity was quantified by scintillation counting. An example of LPA and CPA competition with [3H]ROSI binding to PPARγ is shown in Figure 16.4b.

16.2.3. PPARγ-Corepressor Two-Hybrid Assay in Mammalian Cells

This assay was performed to show that CPA stabilizes SMRT corepressor binding to PPARγ (Fig. 16.5a). CV-1 cells (African green monkey kidney cell line) were grown in Dulbecco’s Modified Eagle’s Medium (DMEM) contain-ing 10% FBS supplemented with 100 IU/mL penicillin G and 100 μg/mL streptomycin. The cells were plated to 96-well plates at a density of 1.0 × 104 cells per well in DMEM containing 10% FBS. The next day, the cells were transiently transfected with 71 ng UAS-Luc, 14.3 ng Gal4-SMRT, 14.3 ng pVP16-PPARγ2, and 10 ng pSV-β-galactosidase (Promega) using Lipo-fectamine™ LTX (Invitrogen) and Opti-MEM® (Invitrogen) for 20 hours. The transfected cells were pretreated with 10 nM, 100 nM, 1 μM, and 10 μM of CPA 18:1 or cyclic glycerophosphate (CGP, Fig. 16.1; Avanti Polar Lipids), which is the alkyl ether analog of CPA 18:1, for 30 minutes, followed by treat-ment with 1 nM ROSI for 6 hours. Luciferase and β-galactosidase activities were measured with the Steady-Glo luciferase assay system (Promega) and

358 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

the Galacto-Light Plus system (Applied Biosystems), respectively. Samples were run in quintuplicate and the mean ± standard error of the mean (SEM) was calculated. As shown in Figure 16.5b, CP A 18:1 and CGP 18:1 dose dependently stabilized the PPARγ–SMRT complex and prevented ROSI-induced dissociation of the complex (*p < 0.05; **p < 0.01).

16.2.4. Ligand-Induced Neointima Model in the Rat Carotid Artery

To examine the regulatory role of CPA in PPARγ-dependent pathophysiologi-cal responses, a noninjury infusion model was used in rats. The animal proce-dures were approved by the Institutional Animal Care and Use Committee of the University of Tennessee Health Science Center. Adult male Sprague-Dawley rats weighing 250–300 g were anesthetized with ketamine (80 mg/kg) and xylazine (5 mg/kg). After anesthesia, the forelimbs and incisors were fixed

Figure 16.5. CPA inhibits PPARγ activation and stabilizes binding of PPARγ corepres-sor SMRT. (a) Schematic diagram of a two-hybrid assay using full length of SMRT and PPARγ2. (b) CPA 18:1 and CGP 18:1 dose dependently inhibited SMRT release from PPARγ induced by ROSI. CV-1 cells were transfected with UAS-Luc, pECE-Gal4-SMRT, pVP16-PPARγ2, and pSV-b-gal. After transfection, the cells were treated with CPA 18:1 or CGP 18:1 for 30 minutes, followed by 1 nM ROSI for 6 h, and luciferase activities were measured. Data are mean ± SEM; n = 4; *p < 0.05, **p < 0.01, represen-tative of three transfections.

METHODS PROBING PPARγ FUNCTION WITH LPA ANALOGS 359

to the surgical board with tape. The hair around the neck was shaved, and the skin was disinfected with povidone-iodine and 70% isopropyl alcohol. The surgery was carried out under a dissecting microscope. The right side of the carotid artery was surgically exposed through a midline incision. The proximal sides of the CCA and internal carotid artery (ICA) were ligated using vessel clips (85 gauge pressure, BRI, Silver Spring, MD). The distal end of the external carotid artery (ECA) was tied with 5.0 surgical sutures (Harvard, Holliston, MA); subsequently, another 5.0 surgical suture was passed through the proximal end of the ECA and a loose loop was made. A PE-10 catheter (Becton Dickinson, Franklin Lakes, NJ) was connected to a servo-controlled peristaltic pump (Model PS-200; Living Systems Instrumentation, ST. Albans, VT) to maintain the intra-arterial pressure at 100 mmHg. The other end of the PE-10 catheter was inserted into the ECA through an arteriotomy without reaching into the CCA. The loose loop was tightened on the ECA to keep the catheter in place. The clip occluding the CCA was temporarily released, and the vessel was rinsed with a retrograde injection of 50 μL phosphate-buffered saline (PBS) to remove residual blood. The CCA was clipped again, and a treatment solution either 0.1% DMSO (vehicle), 10 μM alkyl-LPA, 10 μM ROSI, 10 μM PIO, 10 nM insulin, 750 nM FIPI, or a combination of these compounds was applied. The delivery pressure of the solutions was maintained at 100 mmHg using the peristaltic pump.

In some experiments, ROSI was applied outside the carotid artery in a pluronic gel. Pluronic F-127 (Sigma-Aldrich) was dissolved in PBS (25% w/v) and maintained at 4°C until use. At the time of compound administration via the PE-10 catheter, 100 μL of the pluronic gel containing 100 μM ROSI and 0.1% DMSO was topically applied around the adventitia of the CCA for 1 hour. The control group received the pluronic gel without ligand.

For cotreatment with FIPI, the inhibitor was applied for 30 minutes as a pretreatment and was also included during the application of the PPARγ ago-nists. After a 60-minute incubation, the ligation was loosened to withdraw the catheter and the loop was tied up again to ligate the ECA. The vessel clips on the CCA and ICA were released to restore blood flow. The wound was closed with a 4.0 surgical suture. After the surgery, the rats were kept on a warming pad to avoid hypothermia during recovery and were administered Buprenex (Reckitt Benckiser Healthcare, Hull, UK) (0.05 mg/kg). Three weeks after the surgery, transcardial perfusion was performed with 4% buffered paraformal-dehyde (pH 7.4), and the CCAs were dissected and postfixed in 4% buffered paraformaldehyde (pH 7.4). The dissected CCAs were embedded in paraffin, and 5-μm-thick sections were cut, deparaffinized, and stained with Masson’s trichrome stain (Allan Scientific, Kalamazoo, MI). Neointima formation was assessed using light microscopy and digital images were collected. To evaluate neointima progression, the intima-to-media ratio was measured. The intima (area between the endothelium and the internal elastic lamina) and media (area between the external and internal elastic lamina) were measured using Image J software (version 1.42) (NIH Bethesda, MD), and intima-to-media ratios were calculated.

360 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

As shown in Figure 16.6, neointima formation induced by alkyl-LPA was attenuated by insulin due to the production of CPA through PLD2 activation by insulin (Fig. 16.6b, c, e) (82). The protective effect of insulin was blocked by the PLD inhibitor FIPI because the CPA production was not blocked by the inhibition of PLD2 activity (Fig. 16.6d, e).

Another TZD analog PIO was also tested in the noninjury neointima model. However, the magnitude was significantly less than that induced by ROSI (Fig. 16.7b, c, e). This might be one of the reasons that ROSI is associ-ated with higher cardiovascular risks. ROSI applied to the outside of CCAs with pluronic gel also induced albeit much less neointima formation (Fig. 16.7d, e), suggesting that ROSI diffused toward the inside of the vessel and promoted arterial wall thickness.

16.2.5. Immunohistochemical Staining of the Cellular Elements of Ligand-Induced Neointima

Bone marrow-derived vascular progenitor cells (VPCs) have been suggested to contribute to neointima formation in injury-induced models (85, 86). To assess whether VPCs are recruited to form neointima in response to alkyl-LPA or ROSI treatment in the noninjury model, immunohistochemical staining was performed using antibodies to the VSMC marker α smooth muscle actin (αSMA) and the VPC markers, CD34 and c-kit. Ten-micrometer-thick cryosec-tions of the rat CCAs were double stained for αSMA and either CD34 or c-kit to identify VPCs in the neointimal regions. After fixation and rehydration, cryo-sections were incubated in blocking solution containing 5% normal goat serum (Vector Laboratories, Burlingame, CA) and 5% normal horse serum (Vector Laboratories) in PBS (pH 7.4) at room temperature for 1 hour. Anti-αSMA antibody (mouse monoclonal [clone 1A4], 1:400 dilution; Abcam, Cambridge, MA) and either anti-CD34 (goat polyclonal [sc7045], 1:100 dilution; Santa Cruz Biotechnology) or anti-c-kit (rabbit polyclonal [sc168], 1:100 dilution; Santa Cruz Biotechnology) diluted in blocking solution were applied to the slides and incubated in a humidified chamber at 37°C for 2 hours. After washing with PBS, the slides were incubated with fluorescein-conjugated antimouse IgG (1:200 dilution, Vector Laboratories) and either Alexa Fluor 568 antigoat IgG (1:100, Invitrogen) or biotinylated antirabbit IgG (1:500 dilution, Vector Laboratories) dissolved in blocking solution at room temperature for 1 hour, followed by incubation with rhodamine-avidin D (1:1000, Vector Laboratories) at room temperature for 30 minutes. The slides were mounted with VECTASHIELD with 4′,6-diamidino-2-phenylindole (DAPI) (Vector Laboratories) and visual-ized using a Nikon (Melville, NY) Eclipse 80i fluorescence microscope.

Since CD34 is also an endothelial cell marker, the endothelial cell layer was CD34 positive in DMSO-treated CCA (Fig. 16.8b). The neointimal layer elic-ited by alkyl-LPA expressed strong αSMA staining (Fig. 16.8d); however, CD34-positive cells were barely detected in the neointima region (Fig. 16.8e) and no double-positive cells were observed (Fig. 16.8f). Similarly, very few

METHODS PROBING PPARγ FUNCTION WITH LPA ANALOGS 361

Figure 16.6. PLD inhibitor FIPI inhibits the protective effect of insulin on vascular wall remodeling. (a–d) Carotid arteries of anesthetized adult rats were treated with DMSO (vehicle) (a), 10 μM alkyl-LPA (b), 10 μM alkyl-LPA and 10 nM insulin (c), or 10 μM alkyl-LPA, 10 nM insulin, and 750 nM FIPI (d) for 1 hour. Three weeks after the treatment, the CCAs were dissected and the sections were stained with Masson’s trichrome stain. Scale bars represent 200 μm. (e) Quantification of intima/media ratios. Data are mean ± SEM; n = 5; **p < 0.01.

362 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

Figure 16.7. PIO is less effective than ROSI eliciting neointima and ROSI application to the adventitia induces less neointima compared to lumenal infusion. (a–d) Carotid arteries of anesthetized adult rats were infused with vehicle (a), 10 μM PIO (b), or 10 μM ROSI (c) for 1 hour. ROSI (100 μM) was applied outside of the vessel using 25% pluronic gel for 1 hour (d). The sections were stained with Masson’s trichrome stain. Scale bars represent 200 μm. (e) Quantification of intima/media ratios. Data are the mean ± SEM; n = 5; **p < 0.01.

METHODS PROBING PPARγ FUNCTION WITH LPA ANALOGS 363

c-kit-positive cells were seen in alkyl-LPA-induced neointima (Fig. 16.9e) and no double-positive cells were detected (Fig. 16.9f). These results suggest that most of the cells in the neointima induced by alkyl-LPA do not express the markers of VPCs but show αSMA positivity.

It has been shown that LPA promotes VSMC proliferation (13–15). To confirm whether alkyl-LPA induction causes VSMC proliferation leading to neointima, immunohistochemical staining was performed for the proliferation

Figure 16.8. Immunohistochemical staining for αSMA and CD34 in the neointima induced by alkyl-LPA. Rat carotid arteries 2 weeks after treatment with DMSO (a–c) and 10 μM alkyl-LPA (d–f) were double stained for αSMA (green; a, d) and CD34 (red; b, e). The cells were intensively stained with anti-αSMA in the media and neo-intima layers (a, d). Anti-CD34 stained endothelial cell layers (b, e), but positive cells were barely detected in the neointima (e). No CD34 and αSMA double-stained cells were seen in the merged image (f). The area between the white arrows marks the neointima (d–f). Scale bars: 200 μm.

364 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

marker ki-67. After deparaffinization and rehydration of the rat CCA sections, heat-mediated antigen retrieval was carried out using citrate-based antigen unmasking solution (Vector Laboratories) for 10 minutes on formalin-fixed tissue slides. After three washes in PBS, the slides were blocked with the block-ing solution described above at room temperature for 1 hour. αSMA antibody (rabbit polyclonal [ab5694], 1:400 dilution; Abcam) and ki-67 antibody (mouse

Figure 16.9. Immunohistochemical staining for αSMA and c-kit in the neointima induced by alkyl-LPA. Two weeks after injection with DMSO (a–c) and 10 μM alkyl-LPA (d–f), rat CCAs were double stained for αSMA (green; a, d) and c-kit (red; b, e). Anti-αSMA staining showed intensive immunoreactivity in the neointimal layers (d); however, anti-c-kit staining showed no staining in the neointima region (e). Double staining for αSMA and c-kit staining showed no double-positive cells (f). The area between the white arrows marks the neointima (d–f). The yellow arrows point to c-kit-positive cells. Scale bars: 200 μm. (See color insert.)

SUMMARY 365

monoclonal [clone MIB-5], 1:50 dilution; Dako, Carpinteria, CA) diluted in blocking solution were applied to the slides and incubated at 4°C overnight. The slides were incubated with biotinylated goat antirabbit IgG (1:500 dilu-tion, Vector Laboratories) and fluorescein-conjugated antimouse IgG (1:200 dilution, Vector Laboratories) for 30 minutes, followed by incubation with rhodamine-avidin D (1:1000, Vector Laboratories) at room temperature for 30 minutes. As shown in Figure 16.10, most cells in the neointima intensely express αSMA (Fig. 16.10d), which is characteristic of the VSMC lineage. However, there were scarce ki-67-positive proliferating cells in the neointima lesions (Fig. 16.10d). Taken together, the αSMA-positive cells are recruited rather than proliferated locally in the neointima.

16.3. SUMMARY

In this chapter, we have focused on arterial wall responses mediated by PPARγ modulated by LPAs. We have also described some of the fundamental methods used to study the regulation of PPARγ by endogenous LPA analogs.

Figure 16.10. Ki-67 expression in the neointima after alkyl-LPA treatment. αSMA immunostaining was strongly detected in the vessel wall 2 weeks after incubation with DMSO (a) and alkyl-LPA (c); however, anti-ki-67 staining showed no positive cells (b) and very few positive cells in the neointima (d). The area between the white arrows marks the neointima (d). The yellow arrows point to ki-67-positive cells. Scale bars: 200 μm.

366 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

CPA is a high-affinity ligand and an endogenous negative regulator of PPARγ, unlike unsaturated acyl species of LPA and alkyl-LPA, which have agonist properties (Fig. 16.11). CPA binds to a site within the LBD and inhibits PPARγ activation (Fig. 16.11). CPA binding to PPARγ stabilizes the binding of the corepressor SMRT. LPA and alkyl-LPA induce neointima formation through the activation of PPARγ, whereas CPA inhibits PPARγ-mediated arterial wall remodeling in the noninjury model. In addition, physiological stimuli of PLD2 are effective in blocking PPARγ-dependent vascular remodeling.

αSMA was strongly expressed in the neointima layers induced by alkyl-LPA; however, very few CD34 and c-kit-positive cells were seen in the alkyl-LPA-elicited neointima. Moreover, ki-67-positive cells in the neointimal region were scarcely seen, suggesting that the accumulation of αSMA-positive cells in the neointima is most likely due to migration of VSMCs.

These observations detailed in this chapter were intended to point to the intracellular signaling role of LPA and natural analogs. Thus, LPAs fulfill a dual role as mediators through the activation of cell surface GPCRs and as second messengers intracellularly through the activation/inhibition of PPARγ. Overall, our findings may provide a novel mechanism for the regulation of PPARγ and they open novel therapeutic opportunities. Further clarification of the PLD2–CPA axis could lead to the possibility of synthesizing novel medi-cines modulating PPARγ.

Figure 16.11. Schematic diagram of the PPARγ signaling. LPA, alkyl-LPA, PIO, and ROSI activate PPARγ and promote downstream signals, whereas CPA negatively regu-lates PPARγ. CPA stabilizes PPARγ–SMRT corepressor complex and inhibits PPARγ-mediated downstream signaling transduction. (See color insert.)

REFERENCES 367

REFERENCES

1. Bolen AL, Naren AP, Yarlagadda S, Beranova-Giorgianni S, Chen L, et al. 2011. The phospholipase A1 activity of lysophospholipase A-I links platelet activation to LPA production during blood coagulation. J Lipid Res 52:958–970.

2. Tigyi G, Parrill AL. 2003. Molecular mechanisms of lysophosphatidic acid action. Prog Lipid Res 42:498–526.

3. Spector AA. 2003. Plaque rupture, lysophosphatidic acid, and thrombosis. Circula-tion 108:641–643.

4. Sano T, Baker D, Virag T, Wada A, Yatomi Y, et al. 2002. Multiple mechanisms linked to platelet activation result in lysophosphatidic acid and sphingosine 1-phosphate generation in blood. J Biol Chem 277:21197–21206.

5. Baker DL, Desiderio DM, Miller DD, Tolley B, Tigyi GJ. 2001. Direct quantitative analysis of lysophosphatidic acid molecular species by stable isotope dilution elec-trospray ionization liquid chromatography-mass spectrometry. Anal Biochem 292:287–295.

6. Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, et al. 2002. Serum lysophosphatidic acid is produced through diverse phospholipase pathways. J Biol Chem 277:48737–48744.

7. Baker DL, Morrison P, Miller B, Riely CA, Tolley B, et al. 2002. Plasma lysophos-phatidic acid concentration and ovarian cancer. JAMA 287:3081–3082.

8. Tokumura A, Majima E, Kariya Y, Tominaga K, Kogure K, et al. 2002. Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as autotaxin, a multifunctional phosphodiesterase. J Biol Chem 277:39436–39442.

9. Umezu-Goto M, Kishi Y, Taira A, Hama K, Dohmae N, et al. 2002. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lyso-phosphatidic acid production. J Cell Biol 158:227–233.

10. van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, et al. 2006. Autotaxin, a secreted lysophospholipase D, is essential for blood vessel formation during development. Mol Cell Biol 26:5015–5522.

11. Tanaka M, Okudaira S, Kishi Y, Ohkawa R, Iseki S, et al. 2006. Autotaxin stabilizes blood vessels and is required for embryonic vasculature by producing lysophos-phatidic acid. J Biol Chem 281:25822–25830.

12. Pamuklar Z, Federico L, Liu S, Umezu-Goto M, Dong A, et al. 2009. Autotaxin/lysopholipase D and lysophosphatidic acid regulate murine hemostasis and throm-bosis. J Biol Chem 284:7385–7394.

13. Siess W. 2002. Athero- and thrombogenic actions of lysophosphatidic acid and sphingosine-1-phosphate. Biochim Biophys Acta 1582:204–215.

14. Hayashi K, Takahashi M, Nishida W, Yoshida K, Ohkawa Y, et al. 2001. Phenotypic modulation of vascular smooth muscle cells induced by unsaturated lysophospha-tidic acids. Circ Res 89:251–258.

15. Kim J, Keys JR, Eckhart AD. 2006. Vascular smooth muscle migration and prolif-eration in response to lysophosphatidic acid (LPA) is mediated by LPA receptors coupling to Gq. Cell Signal 18:1695–1701.

368 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

16. Schulze C, Smales C, Rubin LL, Staddon JM. 1997. Lysophosphatidic acid increases tight junction permeability in cultured brain endothelial cells. J Neurochem 68:991–1000.

17. van Nieuw Amerongen GP, Vermeer MA, van Hinsbergh VW. 2000. Role of RhoA and Rho kinase in lysophosphatidic acid-induced endothelial barrier dysfunction. Arterioscler Thromb Vasc Biol 20:E127–E133.

18. Alexander JS, Patton WF, Christman BW, Cuiper LL, Haselton FR. 1998. Platelet-derived lysophosphatidic acid decreases endothelial permeability in vitro. Am J Physiol 274:H115–H122.

19. Rizza C, Leitinger N, Yue J, Fischer DJ, Wang DA, et al. 1999. Lysophosphatidic acid as a regulator of endothelial/leukocyte interaction. Lab Invest 79:1227–1235.

20. Lin CI, Chen CN, Huang MT, Lee SJ, Lin CH, et al. 2008. Lysophosphatidic acid upregulates vascular endothelial growth factor-C and tube formation in human endothelial cells through LPA(1/3), COX-2, and NF-kappaB activation- and EGFR transactivation-dependent mechanisms. Cell Signal 20:1804–1814.

21. Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, et al. 1999. Lysophosphatidic acid mediates the rapid activation of platelets and endothelial cells by mildly oxidized low density lipoprotein and accumulates in human atherosclerotic lesions. Proc Natl Acad Sci U S A 96:6931–6936.

22. Rother E, Brandl R, Baker DL, Goyal P, Gebhard H, et al. 2003. Subtype-selective antagonists of lysophosphatidic acid receptors inhibit platelet activation triggered by the lipid core of atherosclerotic plaques. Circulation 108:741–747.

23. Haseruck N, Erl W, Pandey D, Tigyi G, Ohlmann P, et al. 2004. The plaque lipid lysophosphatidic acid stimulates platelet activation and platelet-monocyte aggre-gate formation in whole blood: involvement of P2Y1 and P2Y12 receptors. Blood 103:2585–2592.

24. Stocker R, Keaney JF, Jr. 2004. Role of oxidative modifications in atherosclerosis. Physiol Rev 84:1381–1478.

25. Asmis R, Begley JG, Jelk J, Everson WV. 2005. Lipoprotein aggregation protects human monocyte-derived macrophages from OxLDL-induced cytotoxicity. J Lipid Res 46:1124–1132.

26. Meydani M. 2001. Vitamin E and atherosclerosis: beyond prevention of LDL oxida-tion. J Nutr 131:366S–368S.

27. Law RE, Goetze S, Xi XP, Jackson S, Kawano Y, et al. 2000. Expression and func-tion of PPARgamma in rat and human vascular smooth muscle cells. Circulation 101:1311–1318.

28. Chen X, Yang XY, Wang ND, Ding C, Yang YJ, et al. 2003. Serum lysophosphatidic acid concentrations measured by dot immunogold filtration assay in patients with acute myocardial infarction. Scand J Clin Lab Invest 63:497–503.

29. Ross R. 1999. Atherosclerosis is an inflammatory disease. Am Heart J 138:S419–S420.

30. Baim D, Brady TJ, Casscells SW, Dunne M, Fayad Z, et al. 2002. Thoughts on the role of the healing professions and the events of September 11, 2001. Circulation 105:1509–1510.

31. Yoshida K, Nishida W, Hayashi K, Ohkawa Y, Ogawa A, et al. 2003. Vascular remod-eling induced by naturally occurring unsaturated lysophosphatidic acid in vivo. Circulation 108:1746–1752.

REFERENCES 369

32. Cheng Y, Makarova N, Tsukahara R, Guo H, Shuyu E, et al. 2009. Lysophosphatidic acid-induced arterial wall remodeling: requirement of PPARgamma but not LPA1 or LPA2 GPCR. Cell Signal 21:1874–1884.

33. Zhang C, Baker DL, Yasuda S, Makarova N, Balazs L, et al. 2004. Lysophosphatidic acid induces neointima formation through PPARgamma activation. J Exp Med 199:763–774.

34. Lim S, Jin CJ, Kim M, Chung SS, Park HS, et al. 2006. PPARgamma gene transfer sustains apoptosis, inhibits vascular smooth muscle cell proliferation, and reduces neointima formation after balloon injury in rats. Arterioscler Thromb Vasc Biol 26:808–813.

35. Subramanian P, Karshovska E, Reinhard P, Megens RT, Zhou Z, et al. 2010. Lyso-phosphatidic acid receptors LPA1 and LPA3 promote CXCL12-mediated smooth muscle progenitor cell recruitment in neointima formation. Circ Res 107:96–105.

36. Fujiwara Y, Sardar V, Tokumura A, Baker D, Murakami-Murofushi K, et al. 2005. Identification of residues responsible for ligand recognition and regioisomeric selectivity of lysophosphatidic acid receptors expressed in mammalian cells. J Biol Chem 280:35038–35050.

37. Minamikawa J, Tanaka S, Yamauchi M, Inoue D, Koshiyama H. 1998. Potent inhibi-tory effect of troglitazone on carotid arterial wall thickness in type 2 diabetes. J Clin Endocrinol Metab 83:1818–1820.

38. Haffner SM, Greenberg AS, Weston WM, Chen H, Williams K, Freed MI. 2002. Effect of rosiglitazone treatment on nontraditional markers of cardiovascular disease in patients with type 2 diabetes mellitus. Circulation 106:679–684.

39. Nissen SE, Wolski K. 2007. Effect of rosiglitazone on the risk of myocardial infarc-tion and death from cardiovascular causes. N Engl J Med 356:2457–2471.

40. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, et al. 2005. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care 28:1547–1554.

41. Nissen SE, Wolski K. 2010. Rosiglitazone revisited: an updated meta-analysis of risk for myocardial infarction and cardiovascular mortality. Arch Intern Med 170:1191–1201.

42. Tsukahara T, Tsukahara R, Yasuda S, Makarova N, Valentine WJ, et al. 2006. Dif-ferent residues mediate recognition of 1-O-oleyllysophosphatidic acid and rosigli-tazone in the ligand binding domain of peroxisome proliferator-activated receptor gamma. J Biol Chem 281:3398–3407.

43. Tontonoz P, Spiegelman BM. 2008. Fat and beyond: the diverse biology of PPAR-gamma. Annu Rev Biochem 77:289–312.

44. Evans RM. 2005. The nuclear receptor superfamily: a Rosetta Stone for physiol-ogy. Mol Endocrinol 19:1429–1438.

45. Duval C, Chinetti G, Trottein F, Fruchart JC, Staels B. 2002. The role of PPARs in atherosclerosis. Trends Mol Med 8:422–430.

46. Kiec-Wilk B, Dembinska-Kiec A, Olszanecka A, Bodzioch M, Kawecka-Jaszcz K. 2005. The selected pathophysiological aspects of PPARs activation. J Physiol Phar-macol 56:149–162.

47. Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, et al. 1997. The orga-nization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem 272:18779–18789.

370 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

48. Lehrke M, Lazar MA. 2005. The many faces of PPARgamma. Cell 123:993–999.49. Asami-Miyagishi R, Iseki S, Usui M, Uchida K, Kubo H, Morita I. 2004. Expression

and function of PPARgamma in rat placental development. Biochem Biophys Res Commun 315:497–501.

50. Medina-Gomez G, Virtue S, Lelliott C, Boiani R, Campbell M, et al. 2005. The link between nutritional status and insulin sensitivity is dependent on the adipocyte-specific peroxisome proliferator-activated receptor-gamma2 isoform. Diabetes 54:1706–1716.

51. Weatherman RV, Fletterick RJ, Scanlan TS. 1999. Nuclear-receptor ligands and ligand-binding domains. Annu Rev Biochem 68:559–581.

52. Yu C, Markan K, Temple KA, Deplewski D, Brady MJ, Cohen RN. 2005. The nuclear receptor corepressors NCoR and SMRT decrease peroxisome proliferator-activated receptor gamma transcriptional activity and repress 3T3-L1 adipogenesis. J Biol Chem 280:13600–13605.

53. Dowell P, Ishmael JE, Avram D, Peterson VJ, Nevrivy DJ, Leid M. 1999. Identifica-tion of nuclear receptor corepressor as a peroxisome proliferator-activated recep-tor alpha interacting protein. J Biol Chem 274:15901–15907.

54. Chen JD, Evans RM. 1995. A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377:454–457.

55. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, et al. 1998. Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature 395:137–143.

56. Yu S, Reddy JK. 2007. Transcription coactivators for peroxisome proliferator-activated receptors. Biochim Biophys Acta 1771:936–951.

57. Lala DS, Mukherjee R, Schulman IG, Koch SS, Dardashti LJ, et al. 1996. Activation of specific RXR heterodimers by an antagonist of RXR homodimers. Nature 383:450–453.

58. Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM. 1995. 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell 83:803–812.

59. Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM. 1998. Oxidized LDL regulates macrophage gene expression through ligand activation of PPARgamma. Cell 93:229–240.

60. Baker PR, Lin Y, Schopfer FJ, Woodcock SR, Groeger AL, et al. 2005. Fatty acid transduction of nitric oxide signaling: multiple nitrated unsaturated fatty acid deriv-atives exist in human blood and urine and serve as endogenous peroxisome proliferator-activated receptor ligands. J Biol Chem 280:42464–42475.

61. Li Y, Zhang J, Schopfer FJ, Martynowski D, Garcia-Barrio MT, et al. 2008. Molecu-lar recognition of nitrated fatty acids by PPAR gamma. Nat Struct Mol Biol 15:865–867.

62. McIntyre TM, Pontsler AV, Silva AR, St Hilaire A, Xu Y, et al. 2003. Identification of an intracellular receptor for lysophosphatidic acid (LPA): LPA is a transcellular PPARgamma agonist. Proc Natl Acad Sci U S A 100:131–136.

63. Davies SS, Pontsler AV, Marathe GK, Harrison KA, Murphy RC, et al. 2001. Oxi-dized alkyl phospholipids are specific, high affinity peroxisome proliferator-activated receptor gamma ligands and agonists. J Biol Chem 276:16015–16023.

REFERENCES 371

64. Froberg K, Andersen LB. 2005. Mini review: physical activity and fitness and its relations to cardiovascular disease risk factors in children. Int J Obes 29 Suppl 2:S34–S39.

65. Lautamaki R, Airaksinen KE, Seppanen M, Toikka J, Luotolahti M, et al. 2005. Rosiglitazone improves myocardial glucose uptake in patients with type 2 diabetes and coronary artery disease: a 16-week randomized, double-blind, placebo-controlled study. Diabetes 54:2787–2794.

66. Negro R, Mangieri T, Dazzi D, Pezzarossa A, Hassan H. 2005. Rosiglitazone effects on blood pressure and metabolic parameters in nondipper diabetic patients. Dia-betes Res Clin Pract 70:20–25.

67. Sarafidis PA, Lasaridis AN, Nilsson PM, Mouslech TF, Hitoglou-Makedou AD, et al. 2005. The effect of rosiglitazone on novel atherosclerotic risk factors in patients with type 2 diabetes mellitus and hypertension. An open-label observational study. Metabolism 54:1236–1242.

68. Moore KJ, Rosen ED, Fitzgerald ML, Randow F, Andersson LP, et al. 2001. The role of PPAR-gamma in macrophage differentiation and cholesterol uptake. Nat Med 7:41–47.

69. Delerive P, Martin-Nizard F, Chinetti G, Trottein F, Fruchart JC, et al. 1999. Peroxi-some proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the acti-vator protein-1 signaling pathway. Circ Res 85:394–402.

70. Marx N, Schonbeck U, Lazar MA, Libby P, Plutzky J. 1998. Peroxisome proliferator-activated receptor gamma activators inhibit gene expression and migration in human vascular smooth muscle cells. Circ Res 83:1097–1103.

71. Rangwala SM, Lazar MA. 2004. Peroxisome proliferator-activated receptor gamma in diabetes and metabolism. Trends Pharmacol Sci 25:331–336.

72. Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM. 1998. PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 93:241–252.

73. Murakami-Murofushi K, Mukai M, Kobayashi S, Kobayashi T, Tigyi G, Murofushi H. 2000. A novel lipid mediator, cyclic phosphatidic acid (cPA), and its biological functions. Ann N Y Acad Sci 905:319–321.

74. Kobayashi T, Tanaka-Ishii R, Taguchi R, Ikezawa H, Murakami-Murofushi K. 1999. Existence of a bioactive lipid, cyclic phosphatidic acid, bound to human serum albumin. Life Sci 65:2185–2191.

75. Eichholtz T, Jalink K, Fahrenfort I, Moolenaar WH. 1993. The bioactive phospho-lipid lysophosphatidic acid is released from activated platelets. Biochem J 291(Pt 3):677–680.

76. Fischer DJ, Liliom K, Guo Z, Nusser N, Virag T, et al. 1998. Naturally occurring analogs of lysophosphatidic acid elicit different cellular responses through selective activation of multiple receptor subtypes. Mol Pharmacol 54:979–988.

77. Fujiwara Y, Sebok A, Meakin S, Kobayashi T, Murakami-Murofushi K, Tigyi G. 2003. Cyclic phosphatidic acid elicits neurotrophin-like actions in embryonic hip-pocampal neurons. J Neurochem 87:1272–1283.

78. Gueguen G, Gaige B, Grevy JM, Rogalle P, Bellan J, et al. 1999. Structure-activity analysis of the effects of lysophosphatidic acid on platelet aggregation. Biochemis-try 38:8440–8450.

372 REGULATION OF NUCLEAR HORMONE RECEPTOR PPARγ BY ENDOGENOUS LPAS

79. Mukai M, Imamura F, Ayaki M, Shinkai K, Iwasaki T, et al. 1999. Inhibition of tumor invasion and metastasis by a novel lysophosphatidic acid (cyclic LPA). Int J Cancer 81:918–922.

80. Ishihara R, Tatsuta M, Iishi H, Baba M, Uedo N, et al. 2004. Attenuation by cyclic phosphatidic acid of peritoneal metastasis of azoxymethane-induced intestinal cancers in Wistar rats. Int J Cancer 110:188–193.

81. Uchiyama A, Mukai M, Fujiwara Y, Kobayashi S, Kawai N, et al. 2007. Inhibition of transcellular tumor cell migration and metastasis by novel carba-derivatives of cyclic phosphatidic acid. Biochim Biophys Acta 1771:103–112.

82. Tsukahara T, Tsukahara R, Fujiwara Y, Yue J, Cheng Y, et al. 2010. Phospholipase D2-dependent inhibition of the nuclear hormone receptor PPARgamma by cyclic phosphatidic acid. Mol Cell 39:421–432.

83. Su W, Yeku O, Olepu S, Genna A, Park JS, et al. 2009. 5-Fluoro-2-indolyl des-chlorohalopemide (FIPI), a phospholipase D pharmacological inhibitor that alters cell spreading and inhibits chemotaxis. Mol Pharmacol 75:437–446.

84. Bligh EG, Dyer WJ. 1959. A rapid method of total lipid extraction and purification. Can J Biochem Physiol 37:911–917.

85. Saiura A, Sata M, Hirata Y, Nagai R, Makuuchi M. 2001. Circulating smooth muscle progenitor cells contribute to atherosclerosis. Nat Med 7:382–383.

86. Zernecke A, Schober A, Bot I, von Hundelshausen P, Liehn EA, et al. 2005. SDF-1alpha/CXCR4 axis is instrumental in neointimal hyperplasia and recruitment of smooth muscle progenitor cells. Circ Res 96:784–791.


Recommended