Advance Publication by-J-STAGE Circulation Journal REVIEW Official Journal of the Japanese Circulation Society http://www.j-circ.or.jp Pleiotropic Effects of Peroxisome Proliferator-Activated Receptor γ and δ in Vascular Diseases Wai San Cheang, BSc; Xi Fang, BSc; Xiao Yu Tian, PhD

Peroxisome proliferator-activated receptors gamma (PPARγ) and delta (PPARδ) are nuclear receptors that have significant physiological effects on glucose and lipid metabolism. Experimental studies in animal models of meta- bolic disease have demonstrated their effects on improving lipid profile, insulin sensitivity, and reducing inflamma- tory responses. PPARγ and -δ are also expressed in the vasculature and their beneficial effects have been examined in various models such as atherosclerosis, hypertension, diabetic vascular complications, etc. using pharmacological ligands or genetic tools including viral vectors and transgenic mice. These studies sug- gest that PPARγ and δ are antiinflammatory, antiatherogenic, antioxidant, and antifibrotic against vascular diseases. Several signaling pathways, effector molecules, as well as coactivators/repressors have been identified as respon- sible for the protective effects of PPARγ and -δ in the vasculature. We discuss the pleiotropic effect of PPARγ and δ in vascular dysfunction, including atherosclerosis, hypertension, vascular remodeling, vascular injury, and dia- betic vasculopathy, in various animal models, and the major underlying mechanisms. We also compare the pheno- types of several endothelial cell/vascular smooth muscle-specific PPARγ and -δ knockout and overexpressing transgenic mice in various disease models, and the implications underlying the functional importance of vascular PPARγ and δ in regulating whole-body homeostasis.

Key Words: Atherosclerosis; Endothelial function; Peroxisome proliferator-activated receptors; Vasculature

eroxisome proliferator-activated receptors (PPARs), leading to improved insulin sensitivity.6 PPARγ is also impli- including PPARα (NR1C1), PPARβ/δ (NR1C2) and cated in the regulation of lipid metabolism,7 as well as the P PPARγ (NR1C3), belong to the nuclear receptor su- maturation of monocytes/macrophages and the control of in- perfamily that functions as transcription factors to regulate flammatory reactions.8 In both humans and mice, loss-of-func- cellular differentiation, development and metabolism (carbohy- tion mutation of PPARγ is associated with insulin resistance and drate, lipid and protein). PPARα is predominantly expressed in .9,10 In addition, targeted deletion of PPARγ in skeletal cells with high rates of fatty acid catabolism such as those found muscle, macrophages, and adipose tissue suggests that PPARγ in liver, heart, kidney and skeletal muscle.1 PPARγ is mainly in these tissues regulates whole-body glucose homeostasis.11–13 associated with adipose tissue, playing a role in various physi- The function of PPARδ was first examined in globally ological and pathophysiological events, including adipocyte PPARδ-deficient mice and later studied using the recently differentiation2 and insulin sensitivity. PPARδ is abundantly developed synthetic PPARδ ligands. A high-affinity PPARδ and ubiquitously expressed at much higher levels than PPARγ ligand, GW501516, can reduce weight gain and reduce tri- and PPARα.3 Upon binding of its ligands, PPARs heterodimer- glycerides, and increase high-density lipoprotein (HDL) -cho- ize with the retinoid receptor (RXR) and bind to peroxisome lesterol in obese mice by increasing peripheral fatty acid ca- proliferator hormone responsive elements in the promoter re- tabolism.14,15 PPARδ also regulates glucose homeostasis. gion of the target genes to activate/repress transcription.4 En- GW501516 also lowers the plasma insulin level and improves dogenous ligands for the PPARs include free fatty acids and glucose tolerance and insulin sensitivity in diet-induced obese eicosanoids, and synthetic ligands for PPARγ and PPARδ have mice.16 Collectively, these findings suggest that PPARδ could been developed, targeting diabetes and dyslipidemia.5 be a potential therapeutic target for combating and The biological functions of PPARγ and PPARδ have been insulin resistance. extensively studied in regard to metabolism. PPARγ activation In addition to a regulatory function in glucose and lipid enhances the lipid storage capacity of the adipose mass, and metabolism, PPARs are expressed in the cardiovascular sys- also increases the number of small, insulin-sensitive adipocytes, tem and their functions there have been shown to be most

Received June 7, 2013; revised manuscript received August 30, 2013; accepted September 19, 2013; released online October 10, 2013 Institute of Vascular Medicine and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong (W.S.C., X.Y.T.); Institute of Cardiovascular Science, Peking University Health Science Center, Beijing (X.F.), China; and Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX (X.Y.T.), USA Mailing address: Xiao Yu Tian, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave. Houston TX 77030 USA. E-mail: [email protected] ISSN-1346-9843 doi: 10.1253/circj.CJ-13-0647 All rights are reserved to the Japanese Circulation Society. For permissions, please e-mail: [email protected] Advance Publication by-J-STAGE CHEANG WS et al. likely independent of their metabolic actions.17,18 PPARγ and inhibits the response of VSMC to inflammatory stimuli. Tumor PPARδ are expressed in endothelial cells (ECs) and vascular necrosis factor alpha (TNFα)-induced expression of chemo- smooth muscle cells (VSMCs). Here, we will focus on the role kines VCAM-1, MCP-1, and CX3CL1 can all be inhibited by of PPARγ and PPARδ in vascular biology beyond their regu- PPARγ activation in cultured VSMCs through inhibition of latory effects on glucose and lipid metabolism, and discuss NF-κB.33 Interleukin (IL)-1β-induced gene expression of COX2 their potential clinical implications. and IL-6 is inhibited by PPARγ activation through inhibition of DNA binding of NF-κB and C/EBP, thereby enhancing the 34,35 γ antiinflammatory effect of PPARγ. PPARγ agonists also Role of PPAR in ECs and VSMCs in Vitro suppress several growth factors, including platelet-derived PPARγ Activation in ECs growth factor (PDGF), basic fibroblast growth factor, throm- Inflammation PPARγ reduces the activation and inflam- bin, insulin, and AngII.36,37 Secondly, data show that inhibition mation of ECs by suppressing the expression of several proin- of PPARγ activity by point mutation directly promotes VSMC flammatory cytokines and chemokines through inhibition of the proliferation and migration.38 TZDs and the endogenous ligand nuclear factor kappa B (NF-κB), AP-1, protein kinase C (PKC) 15-d-PGJ2 also directly inhibit VSMC proliferation by decreas- signaling pathways, and oxidative stress components.19–21 Ei- ing the degradation of p27 (Kip1), an inhibitor of both CDK ther PPARγ agonist (TZDs) or overexpres- and phosphorylation of Rb, and thus prevent G1/S phase transi- sion of the constitutively active PPARγ can inhibit the expres- tion, causing cell cycle arrest.39 PPARγ activation inhibits sion of proinflammatory adhesion molecules and chemokines, VSMC migration through suppression of Ets-1 and matrix including intercellular adhesion molecule (ICAM-1), vascular metalloproteinase 9 (MMP9).40,41 PPARγ activation also sup- cell adhesion molecule (VCAM-1), E-selectin and monocyte presses telomerase activity and mini-chromosome maintenance chemotactic protein (MCP-1) in human ECs.19,20,22,23 In addi- proteins expression, thus regulating the replication and lifespan tion, TZDs inhibits the expressions of NADPH oxidase Nox-1, of VSMCs.42 In addition, and pro- 2 and 4, reducing the production of reactive oxygen species mote apoptosis of VSMC in a PPARγ-dependent manner via (ROS) and subsequent EC activation.24 Interestingly, PPARγ upregulation of growth arrest and DNA damage inducible gene can be also activated by laminar shear stress, which exerts an- 45 (GADD45), as well as p53 expression.43,44 tiinflammatory and antiatherogenic effects on ECs.25 Vasoreactivity The direct effect of PPARγ on VSMCs to Vasoreactivity Apart from antiinflammatory effects, PPARγ modulate vascular tone is associated with signaling pathways also modulates vascular tone through antagonizing the produc- related to AngII-induced vasoconstriction, including AT1R, tion or activation of vasoconstrictors, such as angiotensin II Ca2+ influx and Rho kinase activity. PPARγ inhibits AT1R and endothelin-1, while promoting vasodilatation, mainly by gene transcription by interaction with Sp1, thus reducing Sp1 increasing the production of vasodilating nitric oxide (NO).26 binding to the GC-box-related sequence in the AT1R promoter PPARγ agonist treatment increases NO release in human um- in VSMCs.45 PPARγ also regulates the expression of RGS5 by bilical vein ECs by enhancing the interaction of Hsp90 and binding to its promoter. RGS5 regulates PKC and BKCa chan- endothelial NO synthase (eNOS), resulting in increased eNOS nel activity in VSMCs, which is responsible for maintaining phosphorylation and activation, and the subsequent production myogenic tone in resistance arteries.46 In contrast, PPARγ li- of NO.27 In addition, PPARγ activation also enhances NO gands inhibit the Rho kinase pathway in VSMCs by upregulat- bioavailability by reducing endothelial oxidative stress.24 Con- ing SHP-2, which dephosphorylates Vav, a GTP/GDP exchange versely, NO can activate PPARγ in ECs through a p38-MAPK factor that upregulates Rho kinase activity.47 Furthermore, in dependent mechanism, suggesting a positive feedback loop large vessels, loss of PPARγ also leads to increased RhoA and between PPARγ and NO.28 Rho kinase activity and hypertension, because PPARγ directly Apart from enhancing vasodilation directly, PPARγ also controls expression of RhoBTB1 within the Cullin-3-RhoBTB1 antagonize potent vasoconstrictor endothelin-1 (ET-1). PPARγ complex that induces RhoA degradation.48 agonists also suppress the expression and secretion of ET-1 in 29 ECs by inhibiting the AP-1 transcriptional pathway. Apart In Vivo Effect of PPARγ Activation in from inducing vasoconstriction through binding to the ET-A receptor on VSMCs, ET-1 can also induce vasodilatation by Vascular Diseases (Figure 1) binding to the ET-B receptor (ETBR) on ECs and activating Phenotype of EC/VSMC-Specific PPARγ Knockout and eNOS, an effect more pronounced in resistance arteries. Impor- Mutation Mice tantly, PPARγ directly binds to the ETBR promoter and up- Disruption of endothelial PPARγ accelerates diet-induced ath- regulates ETBR, thus suppressing ET-1 induced vasoconstric- erogenesis in low-density lipoprotein receptor (LDLr)–/– mice.49 tion.30 In addition, PPARγ also antagonizes the vasoconstriction EC-specific PPARγ knockout (KO) mice also have exacerbated induced by angiotensin II (AngII). PPARγ activation inhibits lipopolysaccharide (LPS) -induced pulmonary inflammation AngII-induced vasoconstriction by increasing NO production. and injury as shown by increased infiltration of inflammatory Interestingly, 2 angiotensin-receptor blockers (ARBs), namely cells, edema, ROS and proinflammatory cytokine production.50 and irbesartan, are also partial PPARγ agonists, Interestingly, mice with Tie2-Cre mediated EC-specific PPARγ thus some of the vasoprotective effects of telmisartan, and ir- deletion show significant dyslipidemia and lack of response to besartan, are dependent on activation of PPARγ, as demon- the free fatty acid (FFA) and triglyceride (TG) lowering effects strated by us and others.31,32 of the PPARγ agonist rosiglitazone, which indicates endothe- lial PPARγ integrates metabolic and vascular responses and Function of PPARγ in VSMCs may contribute to the in vivo effects of PPARγ agonists on Inflammation and Vascular Remodeling PPARγ activa- lipid metabolism, possibly because metabolic organs, includ- tion inhibits VSMC proliferation and migration through sev- ing muscle, fat and liver, are highly vascularized and micro- eral mechanisms, including direct inhibition of the production vascular ECs control fatty acid transport and uptake, TG me- of growth factors and proinflammatory cytokines, or regulation tabolism, and LDL function as well.51 These lines of evidence of genes controlling the cell cycle. Firstly, PPARγ activation concur with the in vitro data supporting antiinflammatory and Advance Publication by-J-STAGE PPARγ, PPARδ and Vascular Diseases

Figure 1. Schematic of the signaling pathways and cellular effects of peroxisome proliferator-activated receptor gamma (PPARγ). It can be activated by thiazolidinediones (TZDs) and endogenous ligands (L). Activation of PPARγ in endothelial cells (ECs) causes ETBR upregulation and nitric oxide (NO) release, while also inhibiting chemokines and adhesion molecule expression by repress- ing transcriptional activity of NF-κB/AP-1. In vascular smooth muscle cells (VSMCs), PPARγ activation inhibits contractile compo- nents, proliferation, and migration. eNOS, endothelial nitric oxide synthase; COX, cyclooxygenase; GADD, growth arrest and DNA damage inducible gene; ICAM, intercellular adhesion molecule; IL, interleukin; LDL, low-density lipoprotein; MCP, monocyte chemotactic protein; MMP, matrix metalloproteinase; PDGF, platelet-derived growth factor; PPRE, peroxisome proliferator hormone responsive element; NF-κB, nuclear factor kappa B; VCAM, vascular cell adhesion molecule.

anti-atherogenic functions of endothelial PPARγ, and a poten- cytes, causing impaired intravascular thermoregulation, endo- tial contributory role in lipid metabolism. thelial dysfunction, and exaggerated atherosclerosis.57 Together with the evidence from in vitro studies of VSMC, PPARγ also regulates vasomotor and inflammatory responses Effect of PPARγ Activation in Diabetic Vascular Function in the vasculature in vivo. Contraction of mouse aorta is reduced Evidence suggests that treatment with TZDs improves vascu- and vasodilatation is enhanced in response to β-adrenergic lar function in diabetic mice and humans. A 12-week treat- agonists in the aorta from VSMC-specific PPARγ KO mice, ment with rosiglitazone reduced plasma levels of C-reactive inducing a phenotype of biphasic response in blood pressure to protein (CRP) and increased the median coronary flow veloc- norepinephrine.52 However, overexpressing DN-PPARγ also ity reserve, which is an indicator of coronary endothelial func- caused increased Rho kinase activity in another study.48 It is tion, in patients with diabetes and coronary artery disease.58 thus possible that PPARγ interacts with different proteins in treatment improved eNOS activity and enhanced conduit and resistance arteries in response to vasoactive stim- the vasodilatory response to acetylcholine in diabetic mice.59 uli to maintain vascular tone and blood pressure. In addition, Telmisartan, a partial PPARγ agonist, also protected against PPARγ has a clear role against vascular remodeling. Inducible diabetic vasculopathy in a mouse model of obesity and type 2 PPARγ KO in VSMCs exacerbated AngII-induced vascular diabetes, through normalizing of vascular PPARγ downregu- remodeling. Endothelium-dependent relaxation to acetylcho- lation in the diabetic mice.60 However, the in vivo effect of line was reduced in VSMC-specific PPARγ KO mice and fur- PPARγ activation by TZDs on endothelial function may be an ther impaired by AngII infusion, revealing the protective role indirect effect of improved glucose tolerance, insulin sensitiv- of VSMC PPARγ in AngII-induced vascular injury.53 Loss of ity, and increased vasoprotective adiponectin production in PPARγ in VSMC promotes transplantation-associated vascular diabetic mice.61 Rosiglitazone was withdrawn from the market lesion formation through increased VCAM-1 expression.54,55 because of increased risks of myocardial infarction or other Deletion of PPARγ in VSMCs also accelerates vascular calci- cardiovascular events based on several meta-analyses from fication in LDLr–/– mice, mediated through the secreted friz- randomized trials and Medicare databases.62,63 Such effects zled-related protein-2, which functions as a Wnt5a antagonist.56 may be related to some unknown genes that might be induced Interestingly, VSMC-specific PPARγ KO mice (SM22α-Cre) or repressed by either PPARγ or its coactivators/repressors show an unexpected phenotype of loss of perivascular adipo- in the transcriptional complex, which is yet to be discovered. Advance Publication by-J-STAGE CHEANG WS et al.

Moreover, in experimental settings, the effects of PPARγ on the in all types of cells, including ECs and VSMCs.80,81 In ECs, vascular system are only observed over a short period in rodent PPARδ activation inhibits inflammation, oxidative stress, and studies aiming at understanding the biology of PPARγ in vas- apoptosis; while stimulating angiogenesis. In human ECs, the culature, so we cannot exclude the possibility of any unknown PPARδ ligands, L-165041, GW0742 and GW501516, attenu- causes that are responsible for the adverse effect of PPARγ on ate inflammatory responses by reducing the translocation of the cardiovascular outcomes in type 2 diabetic patients. NF-κB and the subsequent expression of VCAM-1, ICAM-1, E-selectin, MCP-1 and growth-regulated oncogene alpha Role of PPARγ in Atherosclerosis (GROα).82–84 Apart from its ability to suppress proinflamma- The antiatherogenic effects of PPARγ agonists have been dem- tory adhesion molecules and chemokines, GW0742 also pro- onstrated in LDLr–/–, and ApoE–/– mouse models, and hu- tects ECs against oxidative stress by upregulation of antioxi- mans.64 Li et al demonstrated that rosiglitazone or GW7845 dant enzymes, including superoxide dismutase-1, catalase and reduced the development of atherosclerosis in male LDLr–/– thioredoxin reductase.85 Importantly, an endogenous ligand mice.65 The atheroprotective effect of troglitazone was ob- for PPARδ is prostacyclin (PGI2), which can be released by served in ApoE–/– mice,66 and also in high fructose- or high the endothelium. Besides its vasodilatory and antiaggregating fat-fed mice.67 Adenoviral overexpression of PPARγ1 can in- functions, prostacyclin also enhances the proangiogenic func- duce caveolin-1 expression and enhance macrophage choles- tion of human and mouse endothelial progenitor cells through terol efflux, thus attenuate atherosclerosis in ApoE–/– mice.68 PPARδ.86,87 Prostacyclin and L-165041 both can prevent H2O2- Likewise, inhibition of PPARγ exaggerates atherosclerosis. induced apoptosis in human ECs by inducing the binding of Bone marrow transplantation from PPARγ null mice to LDLr–/– PPARδ to the 14-3-3α promoter, which augments proapoptotic mice resulted in an increase in atheroma formation.69 Bone Bcl-2 protein Bad sequestration and reduces Bad translocation marrow reconstitution with macrophage-specific KO of PPARγ to mitochondria to prevent initiation of the death signal.88 also exacerbated atherogenesis, by increasing macrophage re- cruitment.70 In addition to its ability to reduce plaque forma- PPARδ in VSMCs tion, PPARγ also promotes plaque stability. Rosiglitazone can Similar to PPARγ, PPARδ also exerts an antiproliferative effect modify plaque composition and decrease the number of buried on VSMCs. PPARδ activation blocks cell cycle progression fibrous caps to stabilize vulnerable plaques in ApoE–/– mice.71 and reduces VSMC proliferation. L-165041 inhibits PDGF- Pioglitazone reduces MMP expression and macrophage ac- induced VSMC proliferation and neointimal formation after tivity in carotid plaques from ApoE–/– mice, confirming the carotid injury in rats by inhibiting cyclin D1 and cyclin-depen- plaque-stabilizing effect of PPARγ activation.72 Such effects dent kinase 4 (CDK4) expression and decreasing the phos- are also verified using adenovirus-mediated overexpression of phorylation of the retinoblastoma tumor suppressor protein,89 PPARγ in ApoE–/– mice, including reduced lipid deposition, while GW501516 reduces IL-1β-induced proliferation and mi- and reduced macrophage number, increased collagen and SMCs gration of rat aortic VSMCs through increase expression of p53 in the plaques, accompanied by decreased MMP-9, tissue fac- and p21.90 PPARδ activation also induces cell cycle inhibitors tor, MCP-1.73 Therefore, PPARγ is atheroprotective during p21 and p27 in rat aortic VSMCs.91 In addition, GW501516 different stages of plaque formation by enhancing cholesterol exerts antiinflammatory92 and antiapoptotic effects93 in murine efflux, reducing macrophage number, reducing extracellular aortic VSMCs, through its direct target gene TGFβ1. matrix reaction, and stabilizing vulnerable plaques. In Vivo Effect of PPARδ Activation in Effect of PPARγ Against Hypertension Genetic analysis shows that 2 dominant negative mutations in Vascular Diseases (Figure 2) PPARγ are associated with severe hypertension in humans,9 Effect of PPARδ in Metabolic Disease indicating an important role of PPARγ in blood pressure regula- Existing evidence indicates that PPARδ activation exerts ben- tion. PPARγ agonists lower blood pressure in diabetic mice.74–76 eficial effects on lipid and glucose metabolism in mouse mod- We showed that rosiglitazone attenuated ET-1-induced vaso- els of obesity and diabetes. GW501516 or overexpression of constriction and enhanced the ET-1-mediated vasodilating ef- PPARδ reduces weight gain and circulating TG level, while fect through upregulation of ETBR expression in mouse aortas increasing HDL in high fat diet-induced obese mice, db/db, and and mesenteric arteries, which may contribute to the regulation ob/ob mice through stimulating peripheral fatty acid catabo- of vascular tone in hypertension by PPARγ agonists.30 We also lism.14–16,94 In obese mice, administration of GW501516 low- showed that PPARγ ameliorates hyperreactivity of the pulmo- ers the plasma insulin level and improves insulin sensitivity by nary artery and thus inhibits vascular remodeling in rat models enhancing glucose consumption and fatty acid synthesis in the of pulmonary arterial hypertension. PPARγ activation inhibits liver.15,16 Both GW0742 treatment and hepatic overexpression 5-HT 2B receptor expression by inhibiting 5-HT-induced AP- of PPARδ rescue hepatic steatosis by inhibiting lipogenesis in 1/c-jun binding to AP-1 responsive elements in the 5-HT2BR obese diabetic mice.95 promoter, thus suppressing the effect of 5-HT on the pulmo- nary vasculature.77 Pioglitazone also attenuates vascular fibro- PPARδ Against Endothelial Dysfunction sis and improves vascular function in spontaneously hyperten- GW0742 and L-165041 at higher concentrations (>1 μmol/L) sive rats78 through increasing COX-2-derived prostaglandin I2 are reported to directly induce endothelium-dependent relax- (PGI2) production, reducing oxidative stress, leading to in- ation, eNOS phosphorylation and NO production in rat aorta, creased NO bioavailability and thus to improved vasodilation through activation of the phosphatidylinositol 3-kinase (PI3K)/ in resistance arteries.79 Akt pathway.96 Administration of GW0742 in vivo restores endothelial function in streptozotocin-induced type 1 diabetic rats by increasing NO bioavailability, as well as decreasing Role of PPARδ in ECs and VSMCs in Vitro NADPH oxidase-driven superoxide production.97 Moreover, PPARδ in ECs our recent study demonstrated that another PPARδ agonist, PPARβ/δ (NR1C2) is abundantly and ubiquitously expressed GW501516, restores endothelial dysfunction in the aorta and Advance Publication by-J-STAGE PPARγ, PPARδ and Vascular Diseases

Figure 2. Schematic showing the effect of proliferator-activated receptor delta (PPARδ) in the vasculature. Upon activation in endothelial cells (ECs), PPARδ also exhibits antiinflammatory effects through targeting NF-κB. PPARδ activation also upregulates antioxidant enzymes, and activates the PI3K pathway to enhance nitric oxide (NO) production. In vascular smooth muscle cells (VSMCs), PPARδ activation inhibits cell proliferation by repressing cell cycle components. eNOS, endothelial nitric oxide synthase; GROα, growth-regulated oncogene alpha; HDL, high-density lipoprotein; ICAM, intercellular adhesion molecule; IL, interleukin; LDL, low-density lipoprotein; PPRE, peroxisome proliferator hormone responsive element; VCAM, vascular cell adhesion molecule.

mesenteric resistance arteries of diabetic db/db mice and diet- of PPARδ ameliorates the dyslipidemia that is recognized as induced obese mice through enhancing PI3K/Akt signaling with the major contributor to atherosclerosis susceptibility. In line subsequent increases in eNOS activity and NO production.98 with the evidence from in vitro studies of ECs and VSMCs, PPARδ agonists were found to reduce atherosclerotic lesion PPARδ Against Hypertension area through antiinflammatory mechanisms in both ApoE–/– The effect of PPARδ ligands in hypertension has also been and LDLr–/– mice.103–105 PPARδ activation suppresses the ex- examined. In hypertensive rats, administration of GW0742 pression of ICAM-1 and MCP-1 in the mouse aorta and TNFα decreases systolic blood pressure, mesenteric vascular hyper- expression in macrophages, thus reducing atherosclerosis in trophy, and endothelial dysfunction, accompanied by an in- LDLr–/– mice.105 Notably, selective deletion of PPARδ in mac- crease in eNOS activity and decreased NADPH oxidase activ- rophages attenuates atherosclerosis in LDLr–/– mice.106 This ity and expression of proinflammatory and proatherogenic unexpected observation can be explained by the physical inter- genes, which is similar to the effect of PPARγ.99 Additionally, action between PPARδ and Bcl-6. Bcl-6 is not sequestered in GW501516 and L-165041 enhance the recovery of blood flow the absence of PPARδ and thus suppresses the transcription of in mice after hind limb ischemia and stimulate the prolifera- proatherogenic genes such as MCP-1. tion of endothelial progenitor cells by acting on the PI3K/Akt Recently, the effects of several PPARδ or dual PPARα/δ pathway.100 In general, PPARδ activation is beneficial for agonists have been examined in small-scale clinical trials. vascular function. PPARδ agonist, MBX-8025, improved the lipid profile and re- duced the CRP level in overweight dyslipidemia subjects.107,108 PPARδ Against Atherosclerosis Similar effects were also observed with GW501516 in subjects Various lines of evidence show that PPARδ may retard the with .101,109,110 Dual PPARα/δ agonist, development and progression of atherosclerosis through mod- GFT505, improves both lipid and glucose metabolism in pa- ulating lipid metabolism in different tissues and exerting an tients with central obesity, dyslipidemia, and impaired glucose antiinflammatory effect through macrophages. PPARδ agonists metabolism.101,111 Although no adverse effect has been found normalize the levels of HDL, LDL and TG in obese and dia- in these clinical trials, GW501516 may have increased the betic animal models as well as in obese men.14–16,94,101 Impor- incidence of adenoma in a mouse model of colorectal can- tantly, PPARδ acts as sensor for very low-density lipoprotein cer.112,113 The safety of PPARδ needs to be further evaluated (VLDL), regulating TG homeostasis locally in peripheral tis- before it can be used to treat metabolic disease. sues such as macrophages and vessel walls.102 The activation Advance Publication by-J-STAGE CHEANG WS et al.

metabolic syndrome. Proc Natl Acad Sci USA 2003; 100: 15924 – Conclusions 15929. The antiinflammatory and antiatherogenic effects of PPARγ 17. Wang N, Yin R, Liu Y, Mao G, Xi F. Role of peroxisome prolifer- ator-activated receptor-gamma in atherosclerosis: An update. Circ J and PPARδ have been clearly demonstrated in vivo and in 2011; 75: 528 – 535. vitro. Mechanistic studies have also revealed important inter- 18. Takano H, Komuro I. Peroxisome proliferator-activated receptor mediate transcription factors and target genes that are respon- gamma and cardiovascular diseases. Circ J 2009; 73: 214 – 220. sible for the vasoprotective effects. However, there are still 19. Marx N, Mach F, Sauty A, Leung JH, Sarafi MN, Ransohoff RM, et al. Peroxisome proliferator-activated receptor-gamma activators missing steps to be identified, such as coactivators and repres- inhibit IFN-gamma-induced expression of the T cell-active CXC sors of PPARγ and PPARδ, and epigenetic modifiers, as well chemokines IP-10, Mig, and I-TAC in human endothelial cells. J as intermediate metabolites that may activate certain enzymes Immunol 2000; 164: 6503 – 6508. or induce post-translational modification. Exploring the inter- 20. Wang N, Verna L, Chen NG, Chen J, Li H, Forman BM, et al. Con- stitutive activation of peroxisome proliferator-activated receptor- action of these novel targets with PPARγ and PPARδ, espe- gamma suppresses pro-inflammatory adhesion molecules in human cially in vivo, in the context of diabetes and atherosclerosis vascular endothelial cells. J Biol Chem 2002; 277: 34176 – 34181. may provide more insights. In addition, more detailed analysis 21. Verrier E, Wang L, Wadham C, Albanese N, Hahn C, Gamble JR, of the metabolism of the whole organism under the conditions et al. PPARgamma agonists ameliorate endothelial cell activation via inhibition of diacylglycerol-protein kinase c signaling pathway: of EC-specific loss-of-function and gain-of-function will be Role of diacylglycerol kinase. Circ Res 2004; 94: 1515 – 1522. important in revealing the potential role of endothelial PPARγ 22. Pasceri V, Wu HD, Willerson JT, Yeh ET. Modulation of vascular and PPARδ in metabolic disease. These data may provide us inflammation in vitro and in vivo by peroxisome proliferator-acti- with more clues on how to safely regulate PPARγ and PPARδ vated receptor-gamma activators. Circulation 2000; 101: 235 – 238. activity in vivo to treat cardiovascular diseases. 23. Jackson SM, Parhami F, Xi XP, Berliner JA, Hsueh WA, Law RE, et al. Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell inter- References action. Arterioscler Thromb Vasc Biol 1999; 19: 2094 – 2104. 1. Braissant O, Foufelle F, Scotto C, Dauca M, Wahli W. Differential 24. Hwang J, Kleinhenz DJ, Lassegue B, Griendling KK, Dikalov S, expression of peroxisome proliferator-activated receptors (PPARs): Hart CM. Peroxisome proliferator-activated receptor-gamma li- Tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. gands regulate endothelial membrane superoxide production. Am J Endocrinology 1996; 137: 354 – 366. Physiol Cell Physiol 2005; 288: C899 – C905. 2. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in 25. Qin X, Tian J, Zhang P, Fan Y, Chen L, Guan Y, et al. Laminar shear fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. stress up-regulates the expression of stearoyl-CoA desaturase-1 in Cell 1994; 79: 1147 – 1156. vascular endothelial cells. Cardiovasc Res 2007; 74: 506 – 514. 3. Kliewer SA, Umesono K, Mangelsdorf DJ, Evans RM. Retinoid X 26. Ikejima H, Imanishi T, Tsujioka H, Kuroi A, Muragaki Y, Mochizuki receptor interacts with nuclear receptors in retinoic acid, thyroid S, et al. Effect of pioglitazone on nitroglycerin-induced impairment hormone and vitamin D3 signalling. Nature 1992; 355: 446 – 449. of nitric oxide bioavailability by a catheter-type nitric oxide sensor. 4. Gearing KL, Gottlicher M, Teboul M, Widmark E, Gustafsson JA. Circ J 2008; 72: 998 – 1002. Interaction of the peroxisome-proliferator-activated receptor and 27. Polikandriotis JA, Mazzella LJ, Rupnow HL, Hart CM. Peroxisome retinoid X receptor. Proc Natl Acad Sci USA 1993; 90: 1440 – 1444. proliferator-activated receptor gamma ligands stimulate endothe- 5. Vamecq J, Latruffe N. Medical significance of peroxisome prolif- lial nitric oxide production through distinct peroxisome prolifera- erator-activated receptors. Lancet 1999; 354: 141 – 148. tor-activated receptor gamma-dependent mechanisms. Arterioscler 6. Yamauchi T, Waki H, Kamon J, Murakami K, Motojima K, Komeda Thromb Vasc Biol 2005; 25: 1810 – 1816. K, et al. Inhibition of rxr and PPARgamma ameliorates diet-induced 28. Ptasinska A, Wang S, Zhang J, Wesley RA, Danner RL. Nitric oxide obesity and . J Clin Invest 2001; 108: 1001 – 1013. activation of peroxisome proliferator-activated receptor gamma 7. Rosen ED, Spiegelman BM. Ppargamma: A nuclear regulator of through a p38 MAPK signaling pathway. FASEB J 2007; 21: 950 – metabolism, differentiation, and cell growth. J Biol Chem 2001; 276: 961. 37731 – 37734. 29. Delerive P, Martin-Nizard F, Chinetti G, Trottein F, Fruchart JC, 8. Moore KJ, Rosen ED, Fitzgerald ML, Randow F, Andersson LP, Najib J, et al. Peroxisome proliferator-activated receptor activators Altshuler D, et al. The role of PPAR-gamma in macrophage differ- inhibit thrombin-induced endothelin-1 production in human vascu- entiation and cholesterol uptake. Nat Med 2001; 7: 41 – 47. lar endothelial cells by inhibiting the activator protein-1 signaling 9. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos pathway. Circ Res 1999; 85: 394 – 402. MA, et al. Dominant negative mutations in human PPARgamma 30. Tian J, Wong WT, Tian XY, Zhang P, Huang Y, Wang N. Rosigli- associated with severe insulin resistance, diabetes mellitus and hy- tazone attenuates endothelin-1-induced vasoconstriction by upregu- pertension. Nature 1999; 402: 880 – 883. lating endothelial expression of endothelin B receptor. Hypertension 10. Matsui J, Terauchi Y, Kubota N, Takamoto I, Eto K, Yamashita T, 2010; 56: 129 – 135. et al. Pioglitazone reduces islet triglyceride content and restores 31. Yuen CY, Wong WT, Tian XY, Wong SL, Lau CW, Yu J, et al. impaired glucose-stimulated insulin secretion in heterozygous per- Telmisartan inhibits vasoconstriction via PPARgamma-dependent oxisome proliferator-activated receptor-gamma-deficient mice on a expression and activation of endothelial nitric oxide synthase. Car- high-fat diet. Diabetes 2004; 53: 2844 – 2854. diovasc Res 2011; 90: 122 – 129. 11. Kanda T, Brown JD, Orasanu G, Vogel S, Gonzalez FJ, Sartoretto 32. Horiuchi M, Iwanami J, Mogi M. Regulation of angiotensin II re- J, et al. PPARgamma in the endothelium regulates metabolic re- ceptors beyond the classical pathway. Clin Sci (Lond) 2012; 123: sponses to high-fat diet in mice. J Clin Invest 2009; 119: 110 – 124. 193 – 203. 12. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, 33. Park KG, Lee KM, Chang YC, Magae J, Ando K, Kim KB, et al. Subramanian V, Mukundan L, et al. Macrophage-specific PPAR- The ascochlorin derivative, AS-6, inhibits TNF-alpha-induced ad- gamma controls alternative activation and improves insulin resis- hesion molecule and chemokine expression in rat vascular smooth tance. Nature 2007; 447: 1116 – 1120. muscle cells. Life Sci 2006; 80: 120 – 126. 13. He W, Barak Y, Hevener A, Olson P, Liao D, Le J, et al. Adipose- 34. Martín A, Pérez-Girón JV, Hernanz R, Palacios R, Briones AM, specific peroxisome proliferator-activated receptor gamma knock- Fortuño A, et al. Peroxisome proliferator-activated receptor-γ activa- out causes insulin resistance in fat and liver but not in muscle. Proc tion reduces cyclooxygenase-2 expression in vascular smooth mus- Natl Acad Sci USA 2003; 100: 15712 – 15717. cle cells from hypertensive rats by interfering with oxidative stress. 14. Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, et al. Peroxi- J Hypertens 2012; 30: 315 – 326. some-proliferator-activated receptor delta activates fat metabolism 35. Takata Y, Kitami Y, Yang ZH, Nakamura M, Okura T, Hiwada K. to prevent obesity. Cell 2003; 113: 159 – 170. Vascular inflammation is negatively autoregulated by interaction 15. Lee CH, Olson P, Hevener A, Mehl I, Chong LW, Olefsky JM, et between CCAAT/enhancer-binding protein-delta and peroxisome al. PPARdelta regulates glucose metabolism and insulin sensitivity. proliferator-activated receptor-gamma. Circ Res 2002; 91: 427 – 433. Proc Natl Acad Sci USA 2006; 103: 3444 – 3449. 36. Marx N, Schonbeck U, Lazar MA, Libby P, Plutzky J. Peroxisome 16. Tanaka T, Yamamoto J, Iwasaki S, Asaba H, Hamura H, Ikeda Y, proliferator-activated receptor gamma activators inhibit gene ex- et al. Activation of peroxisome proliferator-activated receptor delta pression and migration in human vascular smooth muscle cells. induces fatty acid beta-oxidation in skeletal muscle and attenuates Circ Res 1998; 83: 1097 – 1103. Advance Publication by-J-STAGE PPARγ, PPARδ and Vascular Diseases

37. Law RE, Meehan WP, Xi XP, Graf K, Wuthrich DA, Coats W, et al. 57. Chang L, Villacorta L, Li R, Hamblin M, Xu W, Dou C, et al. Loss Troglitazone inhibits vascular smooth muscle cell growth and inti- of perivascular adipose tissue on peroxisome proliferator-activated mal hyperplasia. J Clin Invest 1996; 98: 1897 – 1905. receptor-γ deletion in smooth muscle cells impairs intravascular 38. Meredith D, Panchatcharam M, Miriyala S, Tsai YS, Morris AJ, thermoregulation and enhances atherosclerosis. Circulation 2012; Maeda N, et al. Dominant-negative loss of PPARgamma function 126: 1067 – 1078. enhances smooth muscle cell proliferation, migration, and vascular 58. Yu J, Zhang Z, Li Z, Feng X, He L, Liu S, et al. Peroxisome prolif- remodeling. Arterioscler Thromb Vasc Biol 2009; 29: 465 – 471. erator-activated receptor-gamma(PPARgamma) agonist improves 39. Wakino S, Kintscher U, Kim S, Yin F, Hsueh WA, Law RE. Per- coronary artery endothelial function in diabetic patients with coro- oxisome proliferator-activated receptor gamma ligands inhibit reti- nary artery disease. J Int Med Res 2010; 38: 86 – 94. noblastoma phosphorylation and G1--> s transition in vascular 59. Huang PH, Sata M, Nishimatsu H, Sumi M, Hirata Y, Nagai R. smooth muscle cells. J Biol Chem 2000; 275: 22435 – 22441. Pioglitazone ameliorates endothelial dysfunction and restores isch- 40. Lee CS, Kwon YW, Yang HM, Kim SH, Kim TY, Hur J, et al. New emia-induced angiogenesis in diabetic mice. Biomed Pharmaco- mechanism of rosiglitazone to reduce neointimal hyperplasia: Ac- ther 2008; 62: 46 – 52. tivation of glycogen synthase kinase-3beta followed by inhibition 60. Toyama K, Nakamura T, Kataoka K, Yasuda O, Fukuda M, Tokutomi of MMP-9. Arterioscler Thromb Vasc Biol 2009; 29: 472 – 479. Y, et al. Telmisartan protects against diabetic vascular complications 41. Goetze S, Kintscher U, Kim S, Meehan WP, Kaneshiro K, Collins in a mouse model of obesity and type 2 diabetes, partially through AR, et al. Peroxisome proliferator-activated receptor-gamma ligands peroxisome proliferator activated receptor-gamma-dependent activ- inhibit nuclear but not cytosolic extracellular signal-regulated kinase/ ity. Biochem Biophys Res Commun 2011; 410: 508 – 513. mitogen-activated protein kinase-regulated steps in vascular smooth 61. Wong Wing T, Tian Xiao Y, Xu A, Yu J, Lau Chi W, Hoo Ruby muscle cell migration. J Cardiovasc Pharmacol 2001; 38: 909 – 921. LC, et al. Adiponectin is required for PPARγ-mediated improve- 42. Ogawa D, Nomiyama T, Nakamachi T, Heywood EB, Stone JF, ment of endothelial function in diabetic mice. Cell Metabolism 2011; Berger JP, et al. Activation of peroxisome proliferator-activated 14: 104 – 115. receptor gamma suppresses telomerase activity in vascular smooth 62. Graham DJ, Ouellet-Hellstrom R, MaCurdy TE, Ali F, Sholley C, muscle cells. Circ Res 2006; 98: e50 – e59. Worrall C, et al. Risk of acute myocardial infarction, stroke, heart 43. Bruemmer D, Yin F, Liu J, Berger JP, Sakai T, Blaschke F, et al. failure, and death in elderly medicare patients treated with rosigli- Regulation of the growth arrest and DNA damage-inducible gene 45 tazone or pioglitazone. JAMA 2010; 304: 411 – 418. (GADD45) by peroxisome proliferator-activated receptor gamma in 63. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocar- vascular smooth muscle cells. Circ Res 2003; 93: e38 – e47. dial infarction and death from cardiovascular causes. N Engl J Med 44. Okura T, Nakamura M, Takata Y, Watanabe S, Kitami Y, Hiwada 2007; 356: 2457 – 2471. K. Troglitazone induces apoptosis via the p53 and GADD45 path- 64. Lim S, Despres JP, Koh KK. Prevention of atherosclerosis in over- way in vascular smooth muscle cells. Eur J Pharmacol 2000; 407: weight/obese patients in need of novel multi-targeted approaches. 227 – 235. Circ J 2011; 75: 1019 – 1027. 45. Sugawara A, Takeuchi K, Uruno A, Ikeda Y, Arima S, Kudo M, et 65. Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass al. Transcriptional suppression of type 1 angiotensin II receptor gene CK. Peroxisome proliferator-activated receptor gamma ligands in- expression by peroxisome proliferator-activated receptor-gamma in hibit development of atherosclerosis in LDL receptor-deficient vascular smooth muscle cells. Endocrinology 2001; 142: 3125 – 3134. mice. J Clin Invest 2000; 106: 523 – 531. 46. Ketsawatsomkron P, Lorca RA, Keen HL, Weatherford ET, Liu X, 66. Chen Z, Ishibashi S, Perrey S, Osuga J, Gotoda T, Kitamine T, et Pelham CJ, et al. PPARgamma regulates resistance vessel tone al. Troglitazone inhibits atherosclerosis in apolipoprotein E-knock- through a mechanism involving RGS5-mediated control of protein out mice: Pleiotropic effects on CD36 expression and HDL. Arte- kinase C and BKCa channel activity. Circ Res 2012; 111: 1446 – rioscler Thromb Vasc Biol 2001; 21: 372 – 377. 1458. 67. Collins AR, Meehan WP, Kintscher U, Jackson S, Wakino S, Noh 47. Wakino S, Hayashi K, Kanda T, Tatematsu S, Homma K, Yoshioka G, et al. Troglitazone inhibits formation of early atherosclerotic K, et al. Peroxisome proliferator-activated receptor gamma ligands lesions in diabetic and nondiabetic low density lipoprotein receptor- inhibit Rho/Rho kinase pathway by inducing protein tyrosine phos- deficient mice. Arterioscler Thromb Vasc Biol 2001; 21: 365 – 371. phatase SHP-2. Circ Res 2004; 95: e45 – e55. 68. Hu Q, Zhang XJ, Liu CX, Wang XP, Zhang Y. Ppargamma1-in- 48. Pelham CJ, Ketsawatsomkron P, Groh S, Grobe JL, de Lange WJ, duced caveolin-1 enhances cholesterol efflux and attenuates athero- Ibeawuchi SR, et al. Cullin-3 regulates vascular smooth muscle func- sclerosis in apolipoprotein E-deficient mice. J Vasc Res 2010; 47: tion and arterial blood pressure via PPARgamma and RhoA/Rho- 69 – 79. kinase. Cell Metab 2012; 16: 462 – 472. 69. Chawla A, Boisvert WA, Lee CH, Laffitte BA, Barak Y, Joseph SB, 49. Qu A, Shah YM, Manna SK, Gonzalez FJ. Disruption of endothe- et al. A PPAR gamma-LXR-ABCA1 pathway in macrophages is lial peroxisome proliferator-activated receptor accelerates diet-in- involved in cholesterol efflux and atherogenesis. Mol Cell 2001; 7: duced atherogenesis in LDL receptor-null mice. Arterioscler Thromb 161 – 171. Vasc Biol 2011; 32: 65 – 73. 70. Babaev VR, Yancey PG, Ryzhov SV, Kon V, Breyer MD, Magnuson 50. Reddy AT, Lakshmi SP, Kleinhenz JM, Sutliff RL, Hart CM, Reddy MA, et al. Conditional knockout of macrophage PPARgamma in- RC. Endothelial cell peroxisome proliferator-activated receptor re- creases atherosclerosis in c57bl/6 and low-density lipoprotein recep- duces endotoxemic pulmonary inflammation and injury. J Clin Im- tor-deficient mice. Arterioscler Thromb Vasc Biol 2005; 25: 1647 – munol 2012; 189: 5411 – 5420. 1653. 51. Kanda T, Brown JD, Orasanu G, Vogel S, Gonzalez FJ, Sartoretto 71. Zhou M, Xu H, Pan L, Wen J, Liao W, Chen K. Rosiglitazone J, et al. Pparγ in the endothelium regulates metabolic responses to promotes atherosclerotic plaque stability in fat-fed ApoE-knockout high-fat diet in mice. J Clin Invest 2008; 119: 110 – 124. mice. Eur J Pharmacol 2008; 590: 297 – 302. 52. Chang L, Villacorta L, Zhang J, Garcia-Barrio MT, Yang K, Hamblin 72. Chang K, Francis SA, Aikawa E, Figueiredo JL, Kohler RH, M, et al. Vascular smooth muscle cell-selective peroxisome prolif- McCarthy JR, et al. Pioglitazone suppresses inflammation in vivo erator-activated receptor-gamma deletion leads to hypotension. in murine carotid atherosclerosis: Novel detection by dual-target Circulation 2009; 119: 2161 – 2169 fluorescence molecular imaging. Arterioscler Thromb Vasc Biol 53. Marchesi C, Rehman A, Rautureau Y, Kasal DA, Briet M, Leibowitz 2010; 30: 1933 – 1939. A, et al. Protective role of vascular smooth muscle cell PPARγ in 73. Hu Q, Zhang XJ, Zhang C, Zhao YX, He H, Liu CX, et al. Peroxi- angiotensin II-induced vascular disease. Cardiovasc Res 2012; 97: some proliferator-activated receptor-gamma1 gene therapy attenu- 562 – 570. ates atherosclerosis and stabilizes plaques in apolipoprotein E-defi- 54. Hamblin M, Chang L, Zhang H, Yang K, Zhang J, Chen YE. Vas- cient mice. Hum Gene Ther 2008; 19: 287 – 299. cular smooth muscle cell peroxisome proliferator-activated receptor-γ 74. Walker AB, Chattington PD, Buckingham RE, Williams G. The mediates pioglitazone-reduced vascular lesion formation. Arterio- rosiglitazone (BRL-49653) lowers blood pressure scler Thromb Vasc Biol 2010; 31: 352 – 359. and protects against impairment of endothelial function in zucker 55. Subramanian V, Golledge J, Ijaz T, Bruemmer D, Daugherty A. fatty rats. Diabetes 1999; 48: 1448 – 1453. Pioglitazone-induced reductions in atherosclerosis occur via smooth 75. Diep QN, El Mabrouk M, Cohn JS, Endemann D, Amiri F, Virdis muscle cell-specific interaction with PPAR.Circ Res 2010; 107: A, et al. Structure, endothelial function, cell growth, and inflamma- 953 – 958. tion in blood vessels of angiotensin II-infused rats: Role of peroxi- 56. Woldt E, Terrand J, Mlih M, Matz RL, Bruban V, Coudane F, et al. some proliferator-activated receptor-gamma. Circulation 2002; 105: The nuclear hormone receptor PPARγ counteracts vascular calcifica- 2296 – 2302. tion by inhibiting wnt5a signalling in vascular smooth muscle cells. 76. Ryan MJ, Didion SP, Mathur S, Faraci FM, Sigmund CD. PPAR Nat Commun 2012; 3: 1077. (gamma) agonist rosiglitazone improves vascular function and low- Advance Publication by-J-STAGE CHEANG WS et al.

ers blood pressure in hypertensive transgenic mice. Hypertension 1053. 2004; 43: 661 – 666. 95. Qin X, Xie X, Fan Y, Tian J, Guan Y, Wang X, et al. Peroxisome 77. Liu Y, Tian XY, Mao G, Fang X, Fung ML, Shyy JY, et al. Peroxi- proliferator-activated receptor-delta induces insulin-induced gene-1 some proliferator-activated receptor-gamma ameliorates pulmonary and suppresses hepatic lipogenesis in obese diabetic mice. Hepatol- arterial hypertension by inhibiting 5-hydroxytryptamine 2b recep- ogy 2008; 48: 432 – 441. tor. Hypertension 2012; 60: 1471 – 1478. 96. Jimenez R, Sanchez M, Zarzuelo MJ, Romero M, Quintela AM, 78. Gao D, Ning N, Hao G, Niu X. Pioglitazone attenuates vascular Lopez-Sepulveda R, et al. Endothelium-dependent vasodilator ef- fibrosis in spontaneously hypertensive rats. PPAR Res 2012; 2012: fects of peroxisome proliferator-activated receptor beta agonists via 1 – 7. the phosphatidyl-inositol-3 kinase-Akt pathway. J Pharmacol Exp 79. Hernanz R, Martín Á, Pérez-Girón JV, Palacios R, Briones AM, Ther 2010; 332: 554 – 561. Miguel M, et al. Pioglitazone treatment increases COX-2-derived 97. Quintela AM, Jimenez R, Gomez-Guzman M, Zarzuelo MJ, Galindo prostacyclin production and reduces oxidative stress in hyperten- P, Sanchez M, et al. Activation of peroxisome proliferator-activated sive rats: Role in vascular function. Br J Pharmacol 2012; 166: receptor-beta/-delta (PPARbeta/delta) prevents endothelial dys- 1303 – 1319. function in type 1 diabetic rats. Free Radic Biol Med 2012; 53: 80. Piqueras L, Reynolds AR, Hodivala-Dilke KM, Alfranca A, Redondo 730 – 741. JM, Hatae T, et al. Activation of PPARbeta/delta induces endothe- 98. Tian XY, Wong WT, Wang NP, Lu Y, Cheang WS, Liu J, et al. lial cell proliferation and angiogenesis. Arterioscler Thromb Vasc PPAR delta activation protects endothelial function in diabetic Biol 2007; 27: 63 – 69. mice. Diabetes 2012; 61: 3285 – 3293. 81. Zhang J, Fu M, Zhu X, Xiao Y, Mou Y, Zheng H, et al. Peroxisome 99. Zarzuelo MJ, Jimenez R, Galindo P, Sanchez M, Nieto A, Romero proliferator-activated receptor delta is up-regulated during vascular M, et al. Antihypertensive effects of peroxisome proliferator-acti- lesion formation and promotes post-confluent cell proliferation in vated receptor-beta activation in spontaneously hypertensive rats. vascular smooth muscle cells. J Biol Chem 2002; 277: 11505 – Hypertension 2011; 58: 733 – 743. 11512. 100. Han JK, Lee HS, Yang HM, Hur J, Jun SI, Kim JY, et al. Peroxi- 82. Piqueras L, Sanz MJ, Perretti M, Morcillo E, Norling L, Mitchell some proliferator-activated receptor-delta agonist enhances vascu- JA, et al. Activation of PPARbeta/delta inhibits leukocyte recruit- logenesis by regulating endothelial progenitor cells through genom- ment, cell adhesion molecule expression, and chemokine release. J ic and nongenomic activations of the phosphatidylinositol 3-kinase/ Leukoc Biol 2009; 86: 115 – 122. Akt pathway. Circulation 2008; 118: 1021 – 1033. 83. Rival Y, Beneteau N, Taillandier T, Pezet M, Dupont-Passelaigue 101. Riserus U, Sprecher D, Johnson T, Olson E, Hirschberg S, Liu A, E, Patoiseau JF, et al. PPARalpha and PPARdelta activators in- et al. Activation of peroxisome proliferator-activated receptor (PPAR) hibit cytokine-induced nuclear translocation of NF-kappaB and delta promotes reversal of multiple metabolic abnormalities, re- expression of VCAM-1 in EAhy926 endothelial cells. Eur J Phar- duces oxidative stress, and increases fatty acid oxidation in moder- macol 2002; 435: 143 – 151. ately obese men. Diabetes 2008; 57: 332 – 339. 84. Liang YJ, Liu YC, Chen CY, Lai LP, Shyu KG, Juang SJ, et al. 102. Chawla A, Lee CH, Barak Y, He WM, Rosenfeld J, Liao D, et al. Comparison of PPARdelta and PPARgamma in inhibiting the pro- PPAR gamma is a very low-density lipoprotein sensor in macro- inflammatory effects of C-reactive protein in endothelial cells. Int phages. Proc Natl Acad Sci USA 2003; 100: 1268 – 1273. J Cardiol 2010; 143: 361 – 367. 103. Takata Y, Liu J, Yin F, Collins AR, Lyon CJ, Lee CH, et al. PPAR 85. Fan Y, Wang Y, Tang Z, Zhang H, Qin X, Zhu Y, et al. Suppression delta-mediated antiinflammatory mechanisms inhibit angiotensin of pro-inflammatory adhesion molecules by PPAR-delta in human II-accelerated atherosclerosis. Proc Natl Acad Sci USA 2008; 105: vascular endothelial cells. Arterioscler Thromb Vasc Biol 2008; 28: 4277 – 4282. 315 – 321. 104. Barish GD, Atkins AR, Downes M, Olson P, Chong LW, Nelson 86. He T, Lu T, d’Uscio LV, Lam CF, Lee HC, Katusic ZS. Angio- M, et al. PPARdelta regulates multiple proinflammatory pathways genic function of prostacyclin biosynthesis in human endothelial to suppress atherosclerosis. Proc Natl Acad Sci USA 2008; 105: progenitor cells. Circ Res 2008; 103: 80 – 88. 4271 – 4276. 87. Aburakawa Y, Kawabe J, Okada M, Yamauchi A, Asanome A, 105. Graham TL, Mookherjee C, Suckling KE, Palmer CNA, Patel L. Kabara M, et al. Prostacyclin stimulated integrin-dependent angio- The PPAR delta agonist GW0742X reduces atherosclerosis in genic effects of endothelial progenitor cells and mediated potent LDLr(-/-) mice. Atherosclerosis 2005; 181: 29 – 37. circulation recovery in ischemic hind limb model. Circ J 2013; 77: 106. Lee CH, Chawla A, Urbiztondo N, Liao D, Boisvert WA, Evans 1053 – 1062. RM. Transcriptional repression of atherogenic inflammation: Mod- 88. Liou JY, Lee S, Ghelani D, Matijevic-Aleksic N, Wu KK. Protec- ulation by PPAR delta. Science 2003; 302: 453 – 457. tion of endothelial survival by peroxisome proliferator-activated 107. Choi YJ, Roberts BK, Wang X, Geaney JC, Naim S, Wojnoonski receptor-delta mediated 14-3-3 upregulation. Arterioscl Throm K, et al. Effects of the PPAR-delta agonist MBX-8025 on athero- Vasc Biol 2006; 26: 1481 – 1487. genic dyslipidemia. Atherosclerosis 2012; 220: 470 – 476. 89. Lim HJ, Lee S, Park JH, Lee KS, Choi HE, Chung KS, et al. PPAR 108. Bays HE, Schwartz S, Littlejohn T 3rd, Kerzner B, Krauss RM, delta agonist l-165041 inhibits rat vascular smooth muscle cell Karpf DB, et al. MBX-8025, a novel peroxisome proliferator recep- proliferation and migration via inhibition of cell cycle. Atheroscle- tor-delta agonist: Lipid and other metabolic effects in dyslipidemic rosis 2009; 202: 446 – 454. overweight patients treated with and without atorvastatin. J Clin 90. Kim HJ, Kim MY, Hwang JS, Lee JH, Chang KC, Kim JH, et al. Endocrinol Metab 2011; 96: 2889 – 2897. PPARdelta inhibits IL-1beta-stimulated proliferation and migration 109. Olson EJ, Pearce GL, Jones NP, Sprecher DL. Lipid effects of of vascular smooth muscle cells via up-regulation of IL-1ra. Cell peroxisome proliferator-activated receptor-delta agonist GW501516 Mol Life Sci 2010; 67: 2119 – 2130. in subjects with low high-density lipoprotein cholesterol: Charac- 91. Sue YM, Chung CP, Lin H, Chou Y, Jen CY, Li HF, et al. teristics of metabolic syndrome. Arterioscler Thromb Vasc Biol PPARdelta-mediated p21/p27 induction via increased CREB-bind- 2012; 32: 2289 – 2294. ing protein nuclear translocation in beraprost-induced antiprolifera- 110. Ooi EM, Watts GF, Sprecher DL, Chan DC, Barrett PH. Mechanism tion of murine aortic smooth muscle cells. Am J Physiol Cell of action of a peroxisome proliferator-activated receptor (PPAR)- Physiol 2009; 297: C321 – C329. delta agonist on lipoprotein metabolism in dyslipidemic subjects with 92. Kim HJ, Ham SA, Kim SU, Hwang JY, Kim JH, Chang KC, et al. central obesity. J Clin Endocrinol Metab 2011; 96: E1568 – E1576. Transforming growth factor-beta 1 is a molecular target for the 111. Cariou B, Zair Y, Staels B, Bruckert E. Effects of the new dual peroxisome proliferator-activated receptor delta. Circ Res 2008; PPAR alpha/delta agonist GFT505 on lipid and glucose homeosta- 102: 193 – 200. sis in abdominally obese patients with combined dyslipidemia or 93. Kim HJ, Kim MY, Jin H, Kim HJ, Kang SS, Kim HJ, et al. Peroxi- impaired glucose metabolism. Diabetes Care 2011; 34: 2008 – 2014. some proliferator-activated receptor delta regulates extracellular 112. Pollock CB, Rodriguez O, Martin PL, Albanese C, Li X, Kopelovich matrix and apoptosis of vascular smooth muscle cells through the L, et al. Induction of metastatic gastric cancer by peroxisome pro- activation of transforming growth factor-beta 1/smad3. Circ Res liferator-activated receptordelta activation. PPAR Res 2010; 2010: 2009; 105: 16 – 24. 571783. 94. Leibowitz MD, Ardecky RJ, Boehm MF, Broderick CL, Carfagna 113. Gupta RA, Wang D, Katkuri S, Wang H, Dey SK, DuBois RN. MA, Crombie DL, et al. Biological characterization of a heterodi- Activation of nuclear hormone receptor peroxisome proliferator- mer-selective retinoid X receptor modulator: Potential benefits for activated receptor-delta accelerates intestinal adenoma growth. Nat the treatment of type 2 diabetes. Endocrinology 2006; 147: 1044 – Med 2004; 10: 245 – 247.