Skip to main content
PPAR Research logoLink to PPAR Research
. 2007 Dec 6;2008:745804. doi: 10.1155/2008/745804

PPAR-γ in the Cardiovascular System

Sheng Zhong Duan 1, Christine Y Ivashchenko 1, Michael G Usher 1, Richard M Mortensen 1, 2, 3,2,3,*
PMCID: PMC2225467  PMID: 18288291

Abstract

Peroxisome proliferator-activated receptor-γ (PPAR-γ), an essential transcriptional mediator of adipogenesis, lipid metabolism, insulin sensitivity, and glucose homeostasis, is increasingly recognized as a key player in inflammatory cells and in cardiovascular diseases (CVD) such as hypertension, cardiac hypertrophy, congestive heart failure, and atherosclerosis. PPAR-γ agonists, the thiazolidinediones (TZDs), increase insulin sensitivity, lower blood glucose, decrease circulating free fatty acids and triglycerides, lower blood pressure, reduce inflammatory markers, and reduce atherosclerosis in insulin-resistant patients and animal models. Human genetic studies on PPAR-γ have revealed that functional changes in this nuclear receptor are associated with CVD. Recent controversial clinical studies raise the question of deleterious action of PPAR-γ agonists on the cardiovascular system. These complex interactions of metabolic responsive factors and cardiovascular disease promise to be important areas of focus for the future.

1. INTRODUCTION

Cardiovascular disease (CVD) is the major cause of morbidity and mortality in developed countries [1]. Searching for the underlying risk factors has revealed that a cluster of contributors is often present simultaneously. This risk factor clustering, most notably the core trio of insulin resistance, dyslipidemia, and hypertension, has been called by a number of different names including metabolic syndrome (MetS), insulin resistance syndrome, the deadly quartet, and Syndrome X [17]. Although somewhat controversial, the usefulness of clustering this syndrome remains clear. The mechanistic connections among the trio are not completely understood.

A major focus is to understand the biological and molecular mechanisms underlying this syndrome and to develop better treatment. One class of molecules that are proposed to be important in the etiology of MetS is the nutrient-sensing nuclear transcription factors, peroxisome proliferator-activated receptors (PPARs), and the related liver X receptors (LXRs) [7]. Among these nuclear receptors, PPAR-γ is of intense interest, not only because its ligands, thiazolidinediones (TZDs), are clinically used for T2DM, but also because it may be a nexus that connects metabolic disorders to CVD [1, 49]. In addition to its important roles in insulin sensitivity and glucose homeostasis, PPAR-γ is also associated with CVD such as coronary heart disease, atherosclerosis, and stroke [7, 10]. MetS is also linked to cardiac hypertrophy because populations with MetS have higher prevalence of cardiac hypertrophy [1114]. However, action of PPAR-γ agonists is not only of metabolism in insulin responsive tissues, but also more directly in the inflammatory, cardiac, and vascular cells. The components of MetS are common risk factors to CVD [1]. In this review, we will focus on PPAR-γ in the cardiovascular (CV) system, including its expression, gain, and loss of function, and the mechanisms by which it functions in cardiovascular cells.

2. PPAR-γ GENE AND ITS EXPRESSION OF CV-RELEVANT TISSUES

PPAR-γ is the most extensively studied PPAR, even though the cloning of this receptor came four years later than that of PPAR-α [15]. The PPAR-γ gene extends over more than 100 kb of genomic DNA. It includes six common coding exons: one exon for the N-terminal A/B domain, two exons for the DNA binding domain, with each one encoding one of the two zinc fingers, one exon for the hinge region, and two exons for the ligand binding domain in the C-terminal region [16, 17]. There are two major splice isoforms in the mouse, PPAR-γ1 and PPAR-γ2, whereas at least two other isoforms, PPAR-γ3 and PPAR-γ4, have also been identified in other species including humans [16]. PPAR-γ1 is encoded by eight exons, comprising two γ1-specific exons, A1 and A2, that constitute the 5-untranslated region, and the six coding exons that are common to both γ1 and γ2 mRNAs. The PPAR-γ1 protein consists of 477 amino acids [16]. The PPAR-γ2 mRNA is composed of seven exons, the additional one, exon B, comprising the γ2 5-untranslated region and an additional N-terminal amino acid sequence specific for γ2. As a result, PPAR-γ2 is a larger protein, consisting of 505 amino acids [16].

The function PPAR-γ was initially recognized in adipose tissue [18], although its expression was first identified in other tissues [15]. It is well expressed in cardiovascular-system-relevant tissues such as heart, endothelium, vascular smooth muscle, kidney, and macrophages [10, 1923]. PPAR-γ2 is mainly expressed in adipocytes while PPAR-γ1 is more widely expressed [23].

3. PPAR-γ LIGANDS

Natural ligands —

Several polyunsaturated fatty acids and their metabolites have been identified as PPAR-γ ligands although no ligand has clearly been identified as a critical physiologic ligand. Endogenous ligands including 15-deoxy-Δ12,14 prostaglandin J2 (15-d-PGJ2) [24]. 15-d-PGJ2 had been one of the most promising candidates for the endogenous PPAR-γ ligand. It binds to PPAR-γ with a dissociation constant (K d) in the low-micromolar range and can activate PPAR-γ target genes at concentrations at or near the K d [25]. However, it has never been definitively proven to exist in vivo, nor are its effects that are specific to PPAR-γ [25]. Other natural ligands of PPAR-γ include 9- and 13-hydroxyoctadecadienoic acid (HODE), which are components of oxidized low-density lipoprotein [26], and 12- and 15-hydroxyeicosatetraenoic acid (HETE) [27]. Some researchers have argued that fatty acids are the important natural ligands although they are fairly low affinity ligands. A recent class of high-affinity ligands, the nitrolipids, has been identified, but their physiologic function and the role of PPAR-γ in their effects have not yet been fully delineated [28].

Synthetic ligands —

TZDs, or “glitazones,” are a class of pharmaceutical compounds used clinically as insulin sensitizers in patients with T2DM [17]. The first clinically used agent in this class, troglitazone (Rezulin), was removed from the market because of rare but life-threatening hepatic toxicity. Fortunately, its successors, rosiglitazone (Avandia), and pioglitazone (Actos), have not been linked to this side effect [23]. More than 15 million prescriptions for these TZDs are dispensed annually in the United States alone. However, adverse effects such as edema and weight gain have been problematic [23]. Another side effect of TZDs in animals is cardiac hypertrophy, which has limited the approved doses of these drugs for clinical use [2933]. More recently, increased risks of myocardial infarction and possibly death from cardiovascular causes have been reported to be associated with rosiglitazone (Avandia) treatment [34], although the result is controversial [35].

TZDs' “on-target” and “off-target” effects —

Compelling evidence has shown that PPAR-γ is the main target of TZDs. PPAR-γ mediates the insulin sensitizing effects of TZDs in fat, skeletal muscle, and liver [3639]. TZDs' effects on fluid retention and weight gain are also dependent on PPAR-γ [40, 41]. However, several studies demonstrate that some of TZDs' effects are independent of PPAR-γ or “off-target.” In macrophages, it has been recognized that although TZDs modulate lipid metabolism through PPAR-γ, some of TZDs' anti-inflammatory effects are independent of it [42] although only at higher doses. Some of the antiproliferative effects of TZDs in embryonic stem cells [43] or cancer cell lines [44, 45] are independent of PPAR-γ. Further, PPAR-γ in skeletal muscle and liver may not be mediating TZDs' insulin sensitizing effects under different conditions or in different models [38, 39, 46]. Loss-of-function studies have provided additional insight on the possible “off-target” effects of TZDs.

Understanding which TZD effects are PPAR-γ independent is an important issue for designing more specific PPAR-γ agonists with fewer side effects. TZDs induce cardiac hypertrophy in animals [2933] independent of cardiac PPAR-γ [47]. TZDs also increase the incidence of congestive heart failure [48] presumably due to fluid retention caused by PPAR-γ activation in the kidney [40, 41]. Myocardial infarction incidence is increased in meta-analysis of clinical trials [34], but it is not known whether this side effect is mediated by PPAR-γ or whether this finding will be confirmed in a prospective study [35]. Further, defining the role of PPAR-γ in these effects would provide guidance for the design of the next generation of TZDs.

4. GAIN AND LOSS OF PPAR-γ FUNCTION IN THE CV SYSTEM

Although originally found to be critical in adipogenesis and regulating insulin signaling, PPAR-γ is also important in CV system [16, 17, 49, 50]. Human genetic studies have revealed that PPAR-γ mutation in humans can result in either gain-of-function or loss-of-function [51]. In animals, gain-of-function studies of PPAR-γ have mostly utilized agonists, particularly synthetic ones (TZDs); Loss-of-function studies have used knockdown or knockout and transgenic mouse models of mutant PPAR-γ, which are powerful tools to study physiological mechanisms. The outcome of these approaches in studying PPAR-γ in CV system has been fruitful and sometimes surprising.

Human mutations —

Pro12Ala mutation is a loss-of-function mutation and has been reported to be associated with not only increased protection against insulin resistance and type-2 diabetes [5256], but also a decreased incidence of myocardial infraction [57] and lower diastolic blood pressure [58]. These cardiovascular effects are likely independent of metabolic impact of this mutation [57, 58].

Pro467Leu, Val290Met, Phe388Leu, and Arg425Cys are all loss-of-function mutations (dominant negative) and have been associated with partial lipodystrophy, insulin resistance, diabetes, and hypertension [5962], although it is not known whether the elevated blood pressure is due to impaired insulin sensitivity.

C161T mutation is a silent polymorphism and has been reported to be associated with reduction in coronary artery disease, likely independent of obesity and of lipid abnormalities, possibly through direct effects on local vascular wall, implicating the protective role of PPAR-γ in atherogenesis [63].

However, ligand binding domain mutants of PPAR-γ with dominant negative actions have been shown to be promiscuous, stimulate associations with nuclear receptor corepressor (N-CoR) and silencing mediator of retinoid and thyroid receptors (SMRT), and inhibit activities of all three wild-type PPARs [64]. These less specific properties of the mutants need to be explored in the human mutations to determine which effects on metabolic syndrome are mediated through PPAR-γ or the PPARs, PPAR-α or PPAR-δ.

Germline gene inactivation —

Homozygous germline PPAR-γ knockout mice die at E10 due to defects in trophoblast [65, 66]. Heterozygous mutations are viable, have less adipose tissue, and are more insulin sensitive than wild-type counterparts [67, 68]. This illustrates the complex response to gene dosage of PPAR-γ.

PPAR-γ and hypertension —

PPAR-γ agonists can lower blood pressure and this effect may be at least partially independent of their insulin-sensitizing effects [17, 49, 50]. Dominant negative mutation of PPAR-γ (Pro467Leu) in mice results in hypertension and fat redistribution but not insulin resistance or diabetes seen in the same mutation in humans [69]. Another line of dominant negative PPAR-γ mutant mice (Leu466Ala) have hypertension (female only) and insulin resistance [70]. As discussed above, dominant negative PPAR-γ mutants can be more promiscuous, inhibiting activity of other PPARs, so we cannot at this point conclude that these mutants strictly act by altering only PPAR-γ activity [64]. Therefore, comparison to loss of function mutants and knockouts is critical.

Embryonic lethality of the germline PPAR-γ knockout has been rescued by breeding Mox2-Cre, in which Cre recombinase is expressed in epiblast-derived tissue but not other tissues, to floxed PPAR-γ mice [71]. The generalized PPAR-γ knockout mice have lipodystrophy and insulin resistance as expected. Surprisingly, they have hypotension rather than hypertension. This is paradoxical because PPAR-γ agonists lower blood pressure [7274]. Knockout and agonist having the same phenotype may be resolved by testing the hypothesis that PPAR-γ suppresses certain key gene expression to control blood pressure and that both agonist and knockout can relieve the suppression. Further, the phenotypes in the generalized PPAR-γ knockout mice suggest that hypertension is separable from lipodystrophy or insulin resistance, even though they are highly associated in humans [59, 61] and in A-ZIP mice [71]. Mechanistically, the vasculature from these generalized PPAR-γ knockout mice has defects in both relaxation and contraction, contributing to the hypotension.

It has not been completely determined whether the hypotension phenotype seen in the generalized PPAR-γ knockout mice is attributable to PPAR-γ deficiency in vascular endothelium or smooth muscle or both. Endothelium-specific PPAR-γ knockout mice are reported to be not having any phenotype at baseline, although this study only used tail cuff to measure the blood pressure [75]. When fed with high fat diet, they have higher blood pressure and heart rate than their wild type control mice. Rosiglitazone does not affect the diet-induced hypertension in these knockout mice, although the decrease of blood pressure typically seen in treated wild-type control mice was not reported [75]. Smooth muscle-specific PPAR-γ knockout mouse model may clarify the role of smooth muscle PPAR-γ in blood pressure regulation.

The kidney is an important organ in controlling blood pressure and PPAR-γ is expressed in this organ, although PPAR-γ deficiency in collecting duct does not alter blood pressure in mice [40, 41]. The knockout does block weight gain, fluid retention, and blood volume expansion caused by TZDs [40, 41]. These results indicate that PPAR-γ deficiency in collecting duct is unlikely to contribute to the hypotension phenotype seen in the generalized PPAR-γ knockout mice [71].

PPAR-γ and cardiac hypertrophy —

PPAR-γ agonists have been shown to inhibit hypertrophy of cultured neonatal rat ventricular cardiomyocytes induced by mechanical stress or angiotensin II, and cardiac hypertrophy induced by aortic constriction in rats and mice [7678]. The inhibition on hypertrophy was accompanied by the inhibition on expression of embryonic genes, including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), skeletal α-Actin, as well as that of endothelin-1 that can induce cardiac hypertrophy [7678]. Aortic constriction causes more profound cardiac hypertrophy in heterozygous PPAR-γ knockout mice than wild-type controls, further indicating the involvement of PPAR-γ in cardiac growth [77]. Nuclear Factor-Kappa Beta (NF-κB) pathway is at least partially mediating the inhibition on hypertrophic growth in vitro [76]. More recently, pioglitazone has been reported to inhibit the gene expression of inflammatory cytokines such as interleukin (IL)-1β, IL-6 [79], suggesting the possible involvement of PPAR-γ's anti-inflammatory activity in cardiac hypertrophy. Paradoxically, TZDs also induce cardiac hypertrophy in mice, rats, and dogs [3033].

The cardiomyocyte-specific PPAR-γ knockout mice have age-progressive cardiac hypertrophy with preserved systolic cardiac function [47]. Rosiglitazone induce cardiac hypertrophy in both knockout mice and wild-type littermate control mice, demonstrating that this TZD's hypertrophic effect is at least partially independent of PPAR-γ in cardiomyocytes [47]. Whether the cardiac hypertrophy caused by rosiglitazone occurs through PPAR-γ independent effects in cardiomyocytes, PPAR-γ in nonmyocyte cells, or blood volume expansion [33, 48], remains to be further determined. Studying these knockout mice under pathological stimulations such as pressure overload may yield more meaningful results to understand the function of PPAR-γ in the heart.

Rosiglitazone and cardiomyocyte-specific PPAR-γ knockout activate distinctly different hypertrophic pathways [47]. Rosiglitazone increases phosphorylation of both p38 mitogen-activated protein kinase (p38-MAPK) and extracellular signal-related kinase (ERK) 1/2 in the heart. The activation of p38-MAPK is independent of cardiomyocyte PPAR-γ and that of ERK1/2 is dependent. The activation of either ERKs or p38-MAPK is sufficient to induce hypertrophy [80, 81] and may therefore contribute to the cardiac hypertrophy induced by rosiglitazone.

The cardiomyocyte-specific PPAR-γ knockout mouse hearts were found to have increased expression of cardiac embryonic genes ANP and β-myosin heavy chain (β-MHC), and elevated NF-κB activity. Embryonic gene expression in adult hearts is a one of the characteristics of pathological cardiac hypertrophy [82, 83]. NF-κB is both necessary and sufficient for hypertrophic growth of cardiomyocytes [84]. Therefore, NF-κB activation is likely to be part of the mechanisms that PPAR-γ deficiency in cardiomyocytes induces cardiac hypertrophy. However, the interaction between these two transcription factors needs to be further characterized.

Another cardiomyocyte-specific PPAR-γ knockout mouse line also has progressive cardiac hypertrophy and accompanied elevation of cardiac gene expression (ANP and skeletal α-actin) [85]. However, these mice also have dilated cardiomyopathy, heart failure, and mitochondrial oxidative damage. Increased myocardial superoxide content, instead of NF-κB activation, seems to be mediating the severe cardiac phenotype [85]. Similarly, when different floxed PPAR-γ mice were used to delete this receptor in skeletal muscle specifically, one mouse line [37] had more severe phenotypes than the other [46]. It is not clear whether these phenotypic differences are because of the different genetic design for the deletion or purely genetic background differences.

PPAR-γ, inflammation, and atherosclerosis —

PPAR-γ is not only expressed in macrophages, endothelial cells, and smooth muscle cells in normal vasculature [10, 1923], but also in atherosclerotic lesions [86, 87]. PPAR-γ agonists reduce atherosclerosis in human patients and animal models [8892] even though there were concerns that these compounds could be proatherogenic because they may promote the macrophages uptake of lipids and speed the foam cell formation [26]. These antiatherogenic effects can be independent of their beneficial effects on metabolism [93, 94]. More direct antiatherogenic function of PPAR-γ was demonstrated by the result that transplantation of PPAR-γ deficient bone marrow to low-density lipoprotein (LDL) receptor null mice led to a significant increase in atherosclerosis [95]. The beneficial effects are largely attributable to PPAR-γ' s anti-inflammation activity and its role in modulating lipid homeostasis in macrophages.

Vascular inflammation has been increasingly appreciated as an important factor in the pathogenesis of atherosclerosis [9699]. The importance of macrophage PPAR-γ in CVD has begun to be appreciated since foam cells in atherosclerotic lesions were found to have high level of PPAR-γ expression [86, 87]. PPAR-γ activation decreases inflammatory cytokines (e.g., tumor necrosis factor-α, IL-6, and IL-1β) produced by macrophages [100, 101]. By inducing the expression of LXR-α and ATP-binding cassette A1, PPAR-γ activation promotes cholesterol efflux from macrophages resulting in inhibition of foam cell formation [95]. Consistently, macrophage-specific PPAR-γ knockout mice have reduced basal cholesterol efflux, most likely because of decreased expression of lipoprotein lipase, scavenger receptor CD36, LXR-α, and ATP-binding cassette G1 [21]. More profound effects on macrophages by PPAR-γ are also possible since it has been recently shown that PPAR-γ controls alternative activation of macrophages and can thereby improve insulin resistance [102]. It is likely that this effect on differentiation of macrophages is also important in effects of CVD.

Endothelial cells play a key role in the inflammatory process of vasculature responding to injuries [9699]. TZDs have been shown to reduce superoxide generation and inhibit the expression of vascular cell adhesion molecule-1, intercellular cell adhesion molecule-1, and lectinlike oxidized LDL receptor, and hence inhibit inflammation in endothelial cells [103106], suggesting an important role of endothelial PPAR-γ in the development of atherosclerosis. The existing endothelium-specific PPAR-γ knockout mice [75] and generalized PPAR-γ knockout mice [71] can be useful tools to study the function of endothelial PPAR-γ in atherosclerosis.

Different cell types are reported to have different mechanisms for PPAR-γ to inhibit inflammation. In intestinal Caco-2 cells, PPAR-γ inhibits inflammation by binding to NF-κB and facilitating its nuclear export [107]. In macrophages, PPAR-γ prevents corepressor complex N-CoR dissociation from the promoters of NF-κB responsive inflammatory gene inducible nitric oxide synthase and therefore represses its expression [108]. It remains to be determined whether PPAR-γ in endothelial or other vascular cells uses one of these pathways or an entirely different mechanism.

The growth and movement of vascular smooth muscle cell within neointima is one of the key steps leading to the formation of atherosclerotic plaque [96]. PPAR-γ agonists have been shown to block the proliferation and increase the apoptosis of vascular smooth muscle cells, suggesting more beneficial effects of PPAR-γ activation in vasculature [109, 110].

PPAR-γ and cardiac remodeling —

Cardiac remodeling after ischemic injury is one of the major causes that lead to heart failure [111, 112]. The remodeling process is characterized by myocyte hypertrophy and cardiac fibrosis [111, 112]. PPAR-γ agonists attenuate this remodeling process after ischemia in experimental animals [113]. Recent in vitro studies on PPAR-γ in cardiac fibroblasts, a major source of fibrillar collagens that lead to fibrosis [111, 112], have revealed more mechanistic insights.

Pioglitazone reduces cell growth, synthesis of collagen type I, and expression of matrix metalloproteinase-1 in cardiac fibroblasts undergone anoxia-reoxygenation or treated with angiotensin II, likely through inhibition of reactive oxygen species generation and NF-κB activation [114, 115]. Brain natriuretic peptide has been implicated in these effects [116]. In cultured cardiac fibroblasts, PPAR-γ agonists induce the expression of vascular endothelial growth factor, a crucial player in the infarcted/ischemic heart, further indicating the beneficial effects of PPAR-γ agonists in cardiac remodeling [117]. However, all of these studies are based upon gain-of-function results. Further investigation using loss-of-function studies would advance our understanding the role of PPAR-γ in cardiac fibroblasts and cardiac remodeling and provide more therapeutic guidance.

PPAR-γ in cardiovascular side effects of TZDs —

Despite the obviously beneficial effects that TZDs have in CV system [118], these compounds have some cardiovascular side effects that are dangerous to be overlooked. As mentioned above, TZDs induce cardiac hypertrophy in animals, a limitation to the dosages in their clinic use.

Congestive heart failure remains to be one of the major contraindications to the clinical use of TZDs [48]. This is presumed to be secondary to the fluid retention caused by activation of PPAR-γ in the kidney, likely in the collecting duct [40, 41]. Collecting duct knockouts of PPAR-γ is able to excrete salt loads more easily although there is no end effect on blood pressure on normal salt diets [40, 41]. PPAR-γ knockout blocked the effect of TZD on mRNA expression of the sodium channel ENaC-γ although the baseline level in the knockout was higher [40].

One recent report regarding the association between rosiglitazone treatment and significantly increased risk in myocardial infarction as well as an increased risk with borderline significance in death from cardiovascular causes has brought a lot of attention to the safety of this drug [34]. The findings were based on limited access to the original data, and meta-analysis used to reach the conclusions is always considered less convincing than a large prospective trial designed to assess the outcome of interest. Such a prospective study is indeed ongoing and the investigators performed an interim analysis and found that rosiglitazone was not significantly associated with increased risk of myocardial infarction and death from cardiovascular causes, although the findings were inconclusive because of the incompleteness of the study [35]. One side effect of rosiglitazone this interim analysis did confirm is the increased incidence of heart failure [35].

Although the findings need to be confirmed, the possible adverse effects of rosiglitazone in myocardial infarction and death from cardiovascular causes are worrisome due to the fact that diabetic patients are already at higher risk for cardiovascular diseases. The mechanisms of the possible adverse effects are uncertain, and could involve myocardial as well as vascular changes. Pioglitazone, another member in the same TZD class, does not seem to have these side effects [119]. In comparison to rosiglitazone, pioglitazone appears to have more beneficial effects on lipid profile [120], which may be one of the contributors to these side effects. However, the exact mechanisms and molecular basis are yet to be explored. In order to ultimately understand this drug and help new drug design, it is critical to address questions such as whether PPAR-γ is mediating these effects of rosiglitazone and whether heart (cardiomyocytes, cardiac fibroblasts, endothelial cells, or smooth muscle cells) is the direct target.

The cardiovascular phenotypes of these gain- or loss-of function studies are summarized in Table 1.

Table 1.

Cardiovascular phenotypes in gain and loss of PPAR-γ function.

CH MI BP CAD AS
TZDs Agonism Gain of function * [3033, 47] ** [34] [17, 49, 50] [8791]
Pro12Ala Human mutation Loss of function [57] [58]
Pro467Leu Human mutation Loss of function [59, 60]
Val290Met Human mutation Loss of function [60]
Phe388Leu Human mutation Loss of function [61]
Arg425Cys Human mutation Loss of function [62]
C161T Human mutation Loss of function [63]
Pro467Leu Mouse mutation Loss of function [69]
Leu466Ala Mouse mutation Loss of function [70]
Generalized KO Transgenic mouse Loss of function [71] [71]
Cardiac KO Transgenic mouse Loss of function [47]
Endothelial KO Transgenic mouse Loss of function [75]
Collecting duct KO Transgenic mouse Loss of function [40, 41]

TZDs: thiazolidinediones; KO: knockout; CH: cardiac hypertrophy;

MI: myocardial infarction; BP: blood pressure; CAD: coronary artery disease; AS: atherosclerosis

*: in animals only; **: rosiglitazone only

Numbers in square brackets are the reference numbers.

5. CONCLUSIONS

PPAR-γ is now firmly established as an important player in cardiovascular diseases. Understanding the mechanisms of PPAR-γ action in heart and vascular cells where action on NF-κB appears to be important in controlling growth and inflammation may lead to improved targeting of the PPAR-γ activity in these cells. The interactions of PPAR-γ with other nuclear transcription factors which have partially overlapping effects such as the PPAR-α, PPAR-δ, and LXR will likely reveal a complex control system of inflammatory and growth responses to nutrient signaling.

ACKNOWLEDGMENT

This work was funded in part by National Heart, Lung, and Blood Institute R01HL070902 and R01HL083201.

References

  • 1.Kahn R, Buse J, Ferrannini E, Stern M. The metabolic syndrome: time for a critical appraisal: joint statement from the american diabetes association and the european association for the study of diabetes. Diabetes Care. 2005;28(9):2289–2304. doi: 10.2337/diacare.28.9.2289. [DOI] [PubMed] [Google Scholar]
  • 2.Kaplan NM. The deadly quartet. upper-body obesity, glucose intolerance, hypertriglyceridemia, and hypertension. Archives of Internal Medicine. 1989;149(7):1514–1520. doi: 10.1001/archinte.149.7.1514. [DOI] [PubMed] [Google Scholar]
  • 3.DeFronzo RA, Ferrannini E. Insulin resistance: a multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care. 1991;14(3):173–194. doi: 10.2337/diacare.14.3.173. [DOI] [PubMed] [Google Scholar]
  • 4.Zimmet P, Magliano D, Matsuzawa Y, et al. The metabolic syndrome: a global public health problem and a new definition. Journal of Atherosclerosis and Thrombosis. 2005;12(6):295–300. doi: 10.5551/jat.12.295. [DOI] [PubMed] [Google Scholar]
  • 5.Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet. 2005;365(9468):1415–1428. doi: 10.1016/S0140-6736(05)66378-7. [DOI] [PubMed] [Google Scholar]
  • 6.Shaw DI, Hall WL, Williams CM. Metabolic syndrome: what is it and what are the implications. The Proceedings of the Nutrition Society. 2005;64(3):349–357. doi: 10.1079/pns2005442. [DOI] [PubMed] [Google Scholar]
  • 7.Shulman AI, Mangelsdorf DJ. Retinoid X receptor heterodimers in the metabolic syndrome. New England Journal of Medicine. 2005;353(6):604–615. doi: 10.1056/NEJMra043590. [DOI] [PubMed] [Google Scholar]
  • 8.Bishop-Bailey D, Wray J. Peroxisome proliferator-activated receptors: a critical review on endogenous pathways for ligand generation. Prostaglandins and Other Lipid Mediators. 2003;71(1-2):1–22. doi: 10.1016/s0090-6980(03)00003-0. [DOI] [PubMed] [Google Scholar]
  • 9.Walcher D, Marx N. Insulin resistance and cardiovascular disease: the role of PPARgamma activators beyond their anti-diabetic action. Diabetes Vascular Disease Research. 2004;1(2):76–81. doi: 10.3132/dvdr.2004.011. [DOI] [PubMed] [Google Scholar]
  • 10.Bishop-Bailey D. Peroxisome proliferator-activated receptors in the cardiovascular system. British Journal of Pharmacology. 2000;129(5):823–834. doi: 10.1038/sj.bjp.0703149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Mulè G, Cottone S, Mongiovì R, et al. Influence of the metabolic syndrome on aortic stiffness in never treated hypertensive patients. Nutrition, Metabolism, and Cardiovascular Diseases. 2006;16(1):54–59. doi: 10.1016/j.numecd.2005.03.005. [DOI] [PubMed] [Google Scholar]
  • 12.Mulè G, Nardi E, Cottone S, et al. Influence of metabolic syndrome on hypertension-related target organ damage. Journal of Internal Medicine. 2005;257(6):503–513. doi: 10.1111/j.1365-2796.2005.01493.x. [DOI] [PubMed] [Google Scholar]
  • 13.Chinali M, Devereux RB, Howard BV, et al. Comparison of cardiac structure and function in american indians with and without the metabolic syndrome (the strong heart study) American Journal of Cardiology. 2004;93(1):40–44. doi: 10.1016/j.amjcard.2003.09.009. [DOI] [PubMed] [Google Scholar]
  • 14.Lind L, Andersson P-E, Andren B, et al. Left ventricular hypertrophy in hypertension is associated with the insulin resistance metabolic syndrome. Journal of Hypertension. 1995;13(4):433–438. [PubMed] [Google Scholar]
  • 15.Kliewer SA, Forman BM, Blumberg B, et al. Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. Proceedings of The National Academy of Sciences of the United States of America. 1994;91(15):7355–7359. doi: 10.1073/pnas.91.15.7355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocrine Reviews. 1999;20(5):649–688. doi: 10.1210/edrv.20.5.0380. [DOI] [PubMed] [Google Scholar]
  • 17.Willson TM, Lambert MH, Kliewer SA. Peroxisome proliferator-activated receptor gamma and metabolic disease. Annual Review of Biochemistry. 2001;70:341–367. doi: 10.1146/annurev.biochem.70.1.341. [DOI] [PubMed] [Google Scholar]
  • 18.Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell. 1994;79(7):1147–1156. doi: 10.1016/0092-8674(94)90006-x. [DOI] [PubMed] [Google Scholar]
  • 19.Fajas L, Auboeuf D, Raspe E, et al. The organization,promoter analysis, and expression of the human PPARgamma gene. The Journal of Biological Chemistry. 1997;272(30):18779–18789. doi: 10.1074/jbc.272.30.18779. [DOI] [PubMed] [Google Scholar]
  • 20.Vidal-Puig AJ, Considine RV, Jimenez-Liñan M, et al. Peroxisome proliferator-activated receptor gene expression in human tissues: effects of obesity, weight loss, and regulation by insulin and glucocorticoids. Journal of Clinical Investigation. 1997;99(10):2416–2422. doi: 10.1172/JCI119424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Akiyama TE, Sakai S, Lambert G, et al. Conditional disruption of the peroxisome proliferator-activated receptor gamma gene in mice results in lowered expression of ABCA1, ABCG1, and apoE in macrophages and reduced cholesterol efflux. Molecular and Cellular Biology. 2002;22(8):2607–2619. doi: 10.1128/MCB.22.8.2607-2619.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Braissant O, Foufelle F, Scotto C, et al. Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology. 1996;137(1):354–366. doi: 10.1210/endo.137.1.8536636. [DOI] [PubMed] [Google Scholar]
  • 23.Rangwala SM, Lazar MA. Peroxisome proliferator-activated receptor gamma in diabetes and metabolism. Trends in Pharmacological Sciences. 2004;25(6):331–336. doi: 10.1016/j.tips.2004.03.012. [DOI] [PubMed] [Google Scholar]
  • 24.Forman BM, Tontonoz P, Chen J, et al. 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell. 1995;83(5):803–812. doi: 10.1016/0092-8674(95)90193-0. [DOI] [PubMed] [Google Scholar]
  • 25.Rosen ED, Spiegelman BM. PPARgamma: a nuclear regulator of metabolism, differentiation, and cell growth. The Journal of Biological Chemistry. 2001;276(41):37731–37734. doi: 10.1074/jbc.R100034200. [DOI] [PubMed] [Google Scholar]
  • 26.Nagy L, Tontonoz P, Alvarez JG, et al. Oxidized LDL regulates macrophage gene expression through ligand activation of PPARgamma. Cell. 1998;93(2):229–240. doi: 10.1016/s0092-8674(00)81574-3. [DOI] [PubMed] [Google Scholar]
  • 27.Huang JT, Welch JS, Ricote M, et al. Interleukin-4-dependent production of PPAR-gamma ligands in macrophages by 12/15-lipoxygenase. Nature. 1999;400(6742):378–382. doi: 10.1038/22572. [DOI] [PubMed] [Google Scholar]
  • 28.Schopfer FJ, Lin Y, Baker PR, et al. Nitrolinoleic acid: an endogenous peroxisome proliferator-activated receptor gamma ligand. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(7):2340–2345. doi: 10.1073/pnas.0408384102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wu L, Wang R, De Champlain J, Wilson TW. Beneficial and deleterious effects of rosiglitazone on hypertension development in spontaneously hypertensive rats. American Journal of Hypertension. 2004;17(9):749–756. doi: 10.1016/j.amjhyper.2004.04.010. [DOI] [PubMed] [Google Scholar]
  • 30.Pickavance LC, Tadayyon M, Widdowson PS, et al. Therapeutic index for rosiglitazone in dietary obese rats: separation of efficacy and haemodilution. British Journal of Pharmacology. 1999;128(7):1570–1576. doi: 10.1038/sj.bjp.0702932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Actos (pioglitazone hydrochloride) [package insert] Lincolnshire, Ill, USA: Takeda Pharmaceuticals America; 2003. [Google Scholar]
  • 32.Avandia (rosiglitazone maleate) [package insert] Research Triangle Park, NC: GlaxoSmithKline Pharmaceuticals; 2002. [Google Scholar]
  • 33.Arakawa K, Ishihara T, Aoto M, et al. An antidiabetic thiazolidinedione induces eccentric cardiac hypertrophy by cardiac volume overload in rats. Clinical and Experimental Pharmacology and Physiology. 2004;31(1-2):8–13. doi: 10.1111/j.1440-1681.2004.03954.x. [DOI] [PubMed] [Google Scholar]
  • 34.Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. New England Journal of Medicine. 2007;356(24):2457–2471. doi: 10.1056/NEJMoa072761. [DOI] [PubMed] [Google Scholar]
  • 35.Home PD, Pocock SJ, Beck-Nielsen H, et al. Rosiglitazone evaluated for cardiovascular outcomes—an interim analysis. New England Journal of Medicine. 2007;357(1):28–38. doi: 10.1056/NEJMoa073394. [DOI] [PubMed] [Google Scholar]
  • 36.He W, Barak Y, Hevener A, et al. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(26):15712–15717. doi: 10.1073/pnas.2536828100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hevener AL, He W, Barak Y, et al. Muscle-specific Pparg deletion causes insulin resistance. Nature Medicine. 2003;9(12):1491–1497. doi: 10.1038/nm956. [DOI] [PubMed] [Google Scholar]
  • 38.Gavrilova O, Haluzik M, Matsusue K, et al. Liver peroxisome proliferator-activated receptor gamma contributes to hepatic steatosis, triglyceride clearance, and regulation of body fat mass. Journal of Biological Chemistry. 2003;278(36):34268–34276. doi: 10.1074/jbc.M300043200. [DOI] [PubMed] [Google Scholar]
  • 39.Matsusue K, Haluzik M, Lambert G, et al. Liver-specific disruption of PPARgamma in leptin-deficient mice improves fatty liver but aggravates diabetic phenotypes. Journal of Clinical Investigation. 2003;111(5):737–747. doi: 10.1172/JCI17223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Guan Y, Hao C, Cha DR, et al. Thiazolidinediones expand body fluid volume through PPARgamma stimulation of ENaC-mediated renal salt absorption. Nature Medicine. 2005;11(8):861–866. doi: 10.1038/nm1278. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang H, Zhang A, Kohan DE, et al. Collecting duct-specific deletion of peroxisome proliferator-activated receptor gamma blocks thiazolidinedione-induced fluid retention. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(26):9406–9411. doi: 10.1073/pnas.0501744102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chawla A, Barak Y, Nagy L, et al. PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation. Nature Medicine. 2001;7(1):48–52. doi: 10.1038/83336. [DOI] [PubMed] [Google Scholar]
  • 43.Palakurthi SS, Aktas H, Grubissich LM, et al. Anticancer effects of thiazolidinediones are independent of peroxisome proliferator-activated receptor gamma and mediated by inhibition of translation initiation. Cancer Research. 2001;61(16):6213–6218. [PubMed] [Google Scholar]
  • 44.Kim Y, Suh N, Sporn M, Reed JC. An inducible pathway for degradation of FLIP protein sensitizes tumor cells to TRAIL-induced apoptosis. Journal of Biological Chemistry. 2002;277(25):22320–22329. doi: 10.1074/jbc.M202458200. [DOI] [PubMed] [Google Scholar]
  • 45.Abe A, Kiriyama Y, Hirano M, et al. Troglitazone suppresses cell growth of KU812 cells independently of PPARgamma. European Journal of Pharmacology. 2002;436(1-2):7–13. doi: 10.1016/s0014-2999(01)01577-1. [DOI] [PubMed] [Google Scholar]
  • 46.Norris AW, Chen L, Fisher SJ, et al. Muscle-specific PPARgamma-deficient mice develop increased adiposity and insulin resistance but respond to thiazolidinediones. Journal of Clinical Investigation. 2003;112(4):608–618. doi: 10.1172/JCI17305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Duan SZ, Ivashchenko CY, Russell MW, et al. Cardiomyocyte-specffic knockout and agonist of peroxisome proliferator-activated receptor-gamma both induce cardiac hypertrophy in mice. Circulation Research. 2005;97(4):372–379. doi: 10.1161/01.RES.0000179226.34112.6d. [DOI] [PubMed] [Google Scholar]
  • 48.Nesto RW, Bell D, Bonow RO, et al. Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the american heart association and american diabetes association. Circulation. 2003;108(23):2941–2948. doi: 10.1161/01.CIR.0000103683.99399.7E. [DOI] [PubMed] [Google Scholar]
  • 49.Berger JP, Akiyama TE, Meinke PT. PPARs: Therapeutic targets for metabolic disease. Trends in Pharmacological Sciences. 2005;26(5):244–251. doi: 10.1016/j.tips.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 50.Lehrke M, Lazar MA. The many faces of PPARgamma. Cell. 2005;123(6):993–999. doi: 10.1016/j.cell.2005.11.026. [DOI] [PubMed] [Google Scholar]
  • 51.Knouff C, Auwerx J. Peroxisome proliferator-activated receptor-gamma calls for activation in moderation: lessons from genetics and pharmacology. Endocrine Reviews. 2004;25(6):899–918. doi: 10.1210/er.2003-0036. [DOI] [PubMed] [Google Scholar]
  • 52.Deeb SS, Fajas L, Nemoto M, et al. A Pro12Ala substitution in PPARgamma2 associated with decreased receptor activity, lower body mass index and improved insulin sensitivity. Nature Genetics. 1998;20(3):284–287. doi: 10.1038/3099. [DOI] [PubMed] [Google Scholar]
  • 53.Altshuler D, Hirschhorn JN, Klannemark M, et al. The common PPARgamma Pro12Ala polymorphism is associated with decreased risk of type 2 diabetes. Nature Genetics. 2000;26(1):76–80. doi: 10.1038/79216. [DOI] [PubMed] [Google Scholar]
  • 54.Pihlajamaki J, Miettinen R, Valve R, et al. The Pro12A1a substitution in the peroxisome proliferator activated receptor gamma 2 is associated with an insulin-sensitive phenotype in families with familial combined hyperlipidemia and in nondiabetic elderly subjects with dyslipidemia. Atherosclerosis. 2000;151(2):567–574. doi: 10.1016/s0021-9150(99)00433-5. [DOI] [PubMed] [Google Scholar]
  • 55.Mori H, Ikegami H, Kawaguchi Y, et al. The Pro12 Ala substitution in PPAR-gamma is associated with resistance to development of diabetes in the general population: possible involvement in impairment of insulin secretion in individuals with type 2 diabetes. Diabetes. 2001;50(4):891–894. doi: 10.2337/diabetes.50.4.891. [DOI] [PubMed] [Google Scholar]
  • 56.Lindi VI, Uusitupa MI, Lindstrom J, et al. Association of the Pro12Ala polymorphism in the PPAR—gamma2 gene with 3-year incidence of type 2 diabetes and body weight change in the finnish diabetes prevention study. Diabetes. 2002;51(8):2581–2586. doi: 10.2337/diabetes.51.8.2581. [DOI] [PubMed] [Google Scholar]
  • 57.Ridker PM, Cook NR, Cheng S, et al. Alanine for proline substitution in the peroxisome proliferator-activated receptor gamma-2 (PPARG2) gene and the risk of incident myocardial infarction. Arteriosclerosis, Thrombosis, and Vascular Biology. 2003;23(5):859–863. doi: 10.1161/01.ATV.0000068680.19521.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ostgren CJ, Lindblad U, Melander O, et al. Peroxisome proliferator-activated receptor-gammaPro12Ala polymorphism and the association with blood pressure in type 2 diabetes: skaraborg hypertension and diabetes project. Journal of Hypertension. 2003;21(9):1657–1662. doi: 10.1097/01.hjh.0000084734.53355.0d. [DOI] [PubMed] [Google Scholar]
  • 59.Savage DB, Tan GD, Acerini CL, et al. Human metabolic syndrome resulting from dominant-negative mutations in the nuclear receptor peroxisome proliferator-activated receptor-gamma. Diabetes. 2003;52(4):910–917. doi: 10.2337/diabetes.52.4.910. [DOI] [PubMed] [Google Scholar]
  • 60.Barroso I, Gurnell M, Crowley VE, et al. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature. 1999;402(6764):880–883. doi: 10.1038/47254. [DOI] [PubMed] [Google Scholar]
  • 61.Hegele RA, Cao H, Frankowski C, Mathews ST, Leff T. PPARG F388L, a transactivation-deficient mutant, in familial partial lipodystrophy. Diabetes. 2002;51(12):3586–3590. doi: 10.2337/diabetes.51.12.3586. [DOI] [PubMed] [Google Scholar]
  • 62.Agarwal AK, Garg A. A novel heterozygous mutation in peroxisome proliferator-activated receptor-γ gene in a patient with familial partial lipodystrophy. Journal of Clinical Endocrinology and Metabolism. 2002;87(1):408–411. doi: 10.1210/jcem.87.1.8290. [DOI] [PubMed] [Google Scholar]
  • 63.Wang XL, Oosterhof J, Duarte N. Peroxisome proliferator-activated receptor γ C161T polymorphism and coronary artery disease. Cardiovascular Research. 1999;44(3):588–594. doi: 10.1016/s0008-6363(99)00256-4. [DOI] [PubMed] [Google Scholar]
  • 64.Semple RK, Meirhaeghe A, Vidal-Puig AJ, et al. A dominant negative human peroxisome proliferator-activated receptor (PPAR)α is a constitutive transcriptional corepressor and inhibits signaling through all PPAR isoforms. Endocrinology. 2005;146(4):1871–1882. doi: 10.1210/en.2004-1405. [DOI] [PubMed] [Google Scholar]
  • 65.Barak Y, Nelson MC, Ong ES, et al. PPARγ is required for placental, cardiac, and adipose tissue development. Molecular Cell. 1999;4(4):585–595. doi: 10.1016/s1097-2765(00)80209-9. [DOI] [PubMed] [Google Scholar]
  • 66.Kubota N, Terauchi Y, Miki H, et al. PPARγ mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance. Molecular Cell. 1999;4(4):597–609. doi: 10.1016/s1097-2765(00)80210-5. [DOI] [PubMed] [Google Scholar]
  • 67.Miles PDG, Barak Y, He W, Evans RM, Olefsky JM. Improved insulin-sensitivity in mice heterozygous for PPAR-γ deficiency. Journal of Clinical Investigation. 2000;105(3):287–292. doi: 10.1172/JCI8538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yamauchi T, Kamon J, Waki H, et al. The mechanisms by which both heterozygous peroxisome proliferator-activated receptor γ (PPARγ) deficiency and PPARγ agonist improve insulin resistance. The Journal of Biological Chemistry. 2001;276(44):41245–41254. doi: 10.1074/jbc.M103241200. [DOI] [PubMed] [Google Scholar]
  • 69.Tsai Y-S, Kim H-J, Takahashi N, et al. Hypertension and abnormal fat distribution but not insulin resistance in mice with P465L PPARγ . Journal of Clinical Investigation. 2004;114(2):240–249. doi: 10.1172/JCI20964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Freedman BD, Lee E-J, Park Y, Jameson JL. A dominant negative peroxisome proliferator-activated receptor-γ knock-in mouse exhibits features of the metabolic syndrome. Journal of Biological Chemistry. 2005;280(17):17118–17125. doi: 10.1074/jbc.M407539200. [DOI] [PubMed] [Google Scholar]
  • 71.Sheng ZD, Ivashchenko CY, Whitesall SE, et al. Hypotension, lipodystrophy, and insulin resistance in generalized PPARγ-deficient mice rescued from embryonic lethality. Journal of Clinical Investigation. 2007;117(3):812–822. doi: 10.1172/JCI28859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.St John Sutton M, Rendell M, Dandona P, et al. A comparison of the effects of rosiglitazone and glyburide on cardiovascular function and glycemic control in patients with type 2 diabetes. Diabetes Care. 2002;25(11):2058–2064. doi: 10.2337/diacare.25.11.2058. [DOI] [PubMed] [Google Scholar]
  • 73.Atkins KB, Northcott CA, Watts SW, Brosius FC. Effects of PPAR-γ ligands on vascular smooth muscle marker expression in hypertensive and normal arteries. American Journal of Physiology—Heart and Circulatory Physiology. 2005;288(1 57-1):H235–H243. doi: 10.1152/ajpheart.00643.2004. [DOI] [PubMed] [Google Scholar]
  • 74.Diep QN, Mabrouk ME, Cohn JS, et al. Structure, endothelial function, cell growth, and inflammation in blood vessels of angiotensin II-infused rats: role of peroxisome proliferator-activated receptor-γ . Circulation. 2002;105(19):2296–2302. doi: 10.1161/01.cir.0000016049.86468.23. [DOI] [PubMed] [Google Scholar]
  • 75.Nicol CJ, Adachi M, Akiyama TE, Gonzalez FJ. PPARγ in endothelial cells influences high fat diet-induced hypertension. American Journal of Hypertension. 2005;18(4):549–556. doi: 10.1016/j.amjhyper.2004.10.032. [DOI] [PubMed] [Google Scholar]
  • 76.Yamamoto K, Ohki R, Lee RT, Ikeda U, Shimada K. Peroxisome proliferator-activated receptor γ activators inhibit cardiac hypertrophy in cardiac myocytes. Circulation. 2001;104(14):1670–1675. doi: 10.1161/hc4001.097186. [DOI] [PubMed] [Google Scholar]
  • 77.Asakawa M, Takano H, Nagai T, et al. Peroxisome proliferator-activated receptor γ plays a critical role in inhibition of cardiac hypertrophy in vitro and in vivo. Circulation. 2002;105(10):1240–1246. doi: 10.1161/hc1002.105225. [DOI] [PubMed] [Google Scholar]
  • 78.Sakai S, Miyauchi T, Irukayama-Tomobe Y, Ogata T, Goto K, Yamaguchi I. Peroxisome proliferator-activated receptor-γ activators inhibit endothelin-1-related cardiac hypertrophy in rats. Clinical Science. 2002;103(48):16–20. doi: 10.1042/CS103S016S. [DOI] [PubMed] [Google Scholar]
  • 79.Ye P, Yang W, Wu S-M, Sheng L. Effect of pioglitazone on the expression of inflammatory cytokines in attenuating rat cardiomyocyte hypertrophy. Methods and Findings in Experimental and Clinical Pharmacology. 2006;28(10):691–696. doi: 10.1358/mf.2006.28.10.1037500. [DOI] [PubMed] [Google Scholar]
  • 80.Bueno OF, De Windt LJ, Tymitz KM, et al. The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO Journal. 2000;19(23):6341–6350. doi: 10.1093/emboj/19.23.6341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang Y, Huang S, Sah VP, et al. Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. Journal of Biological Chemistry. 1998;273(4):2161–2168. doi: 10.1074/jbc.273.4.2161. [DOI] [PubMed] [Google Scholar]
  • 82.Frey N, Olson EN. Cardiac hypertrophy: the good, the bad, and the ugly. Annual Review of Physiology. 2003;65:45–79. doi: 10.1146/annurev.physiol.65.092101.142243. [DOI] [PubMed] [Google Scholar]
  • 83.Hoshijima M, Chien KR. Mixed signals in heart failure: Cancer rules. Journal of Clinical Investigation. 2002;109(7):849–855. doi: 10.1172/JCI15380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Purcell NH, Tang G, Yu C, Mercurio F, DiDonato JA, Lin A. Activation of NF-κB is required for hypertrophic growth of primary rat neonatal ventricular cardiomyocytes. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(12):6668–6673. doi: 10.1073/pnas.111155798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ding G, Fu M, Qin Q, et al. Cardiac peroxisome proliferator-activated receptor gamma is essential in protecting cardiomyocytes from oxidative damage. Cardiovascular Research. 2007;76(2):269–279. doi: 10.1016/j.cardiores.2007.06.027. [DOI] [PubMed] [Google Scholar]
  • 86.Marx N, Sukhova G, Murphy C, Libby P, Plutzky J. Macrophages in human atheroma contain PPARgamma: differentiation-dependent peroxisomal proliferator-activated receptor gamma(PPARgamma) expression and reduction of MMP-9 activity through PPARgamma activation in mononuclear phagocytes in vitro. The American Journal of Pathology. 1998;153(1):17–23. doi: 10.1016/s0002-9440(10)65540-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ricote M, Huang J, Fajas L, et al. Expression of the peroxisome proliferator-activated receptor gamma (PPARgamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(13):7614–7619. doi: 10.1073/pnas.95.13.7614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass CK. Peroxisome proliferator-activated receptor γ ligands inhibit development of atherosclerosis in LDL receptor-deficient mice. Journal of Clinical Investigation. 2000;106(4):523–531. doi: 10.1172/JCI10370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Minamikawa J, Tanaka S, Yamauchi M, Inoue D, Koshiyama H. Potent inhibitory effect of troglitazone on carotid arterial wall thickness in type 2 diabetes. Journal of Clinical Endocrinology and Metabolism. 1998;83(5):1818–1820. doi: 10.1210/jcem.83.5.4932. [DOI] [PubMed] [Google Scholar]
  • 90.Chen Z, Ishibashi S, Perrey S, et al. Troglitazone inhibits atherosclerosis in apolipoprotein E-knockout mice: Pleiotropic effects on CD36 expression and HDL. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21(3):372–377. doi: 10.1161/01.atv.21.3.372. [DOI] [PubMed] [Google Scholar]
  • 91.Collins AR, Meehan WP, Kintscher U, et al. Troglitazone inhibits formation of early atherosclerotic lesions in diabetic and nondiabetic low density lipoprotein receptor-deficient mice. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21(3):365–371. doi: 10.1161/01.atv.21.3.365. [DOI] [PubMed] [Google Scholar]
  • 92.Marfella R, D'Amico M, Esposito K, et al. The ubiquitin-proteasome system and inflammatory activity in diabetic atherosclerotic plaques: effects of rosiglitazone treatment. Diabetes. 2006;55(3):622–632. doi: 10.2337/diabetes.55.03.06.db05-0832. [DOI] [PubMed] [Google Scholar]
  • 93.Calkin AC, Forbes JM, Smith CM, et al. Rosiglitazone attenuates atherosclerosis in a model of insulin insufficiency independent of its metabolic effects. Arteriosclerosis, Thrombosis, and Vascular Biology. 2005;25(9):1903–1909. doi: 10.1161/01.ATV.0000177813.99577.6b. [DOI] [PubMed] [Google Scholar]
  • 94.Levi Z, Shaish A, Yacov N, et al. Rosiglitazone (PPARγ-agonist) attenuates atherogenesis with no effect on hyperglycaemia in a combined diabetes-atherosclerosis mouse model. Diabetes, Obesity and Metabolism. 2003;5(1):45–50. doi: 10.1046/j.1463-1326.2003.00240.x. [DOI] [PubMed] [Google Scholar]
  • 95.Chawla A, Boisvert WA, Lee C-H, et al. A PPARγ-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Molecular Cell. 2001;7(1):161–171. doi: 10.1016/s1097-2765(01)00164-2. [DOI] [PubMed] [Google Scholar]
  • 96.Ross R. Atherosclerosis—an inflammatory disease. The New England Journal of Medicine. 1999;340(2):115–126. doi: 10.1056/NEJM199901143400207. [DOI] [PubMed] [Google Scholar]
  • 97.Libby P. Inflammation in atherosclerosis. Nature. 2002;420(6917):868–874. doi: 10.1038/nature01323. [DOI] [PubMed] [Google Scholar]
  • 98.Lusis AJ. Atherosclerosis. Nature. 2000;407(6801):233–241. doi: 10.1038/35025203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Glass CK, Witztum JL. Atherosclerosis: the road ahead. Cell. 2001;104(4):503–516. doi: 10.1016/s0092-8674(01)00238-0. [DOI] [PubMed] [Google Scholar]
  • 100.Jiang C, Ting AT, Seed B. PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998;391(6662):82–86. doi: 10.1038/34184. [DOI] [PubMed] [Google Scholar]
  • 101.Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-γ is a negative regulator of macrophage activation. Nature. 1998;391(6662):79–82. doi: 10.1038/34178. [DOI] [PubMed] [Google Scholar]
  • 102.Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, et al. Macrophage-specific PPARγ controls alternative activation and improves insulin resistance. Nature. 2007;447(7148):1116–1120. doi: 10.1038/nature05894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sasaki M, Jordan P, Welbourne T, et al. Troglitazone, a PPAR-γ activator prevents endothelial cell adhesion molecule expression and lymphocyte adhesion mediated by TNF-α . BMC Physiology. 2005;5:3. doi: 10.1186/1472-6793-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Imamoto E, Yoshida N, Uchiyama K, et al. Inhibitory effect of pioglitazone on expression of adhesion molecules on neutrophils and endothelial cells. BioFactors. 2004;20(1):37–47. doi: 10.1002/biof.5520200104. [DOI] [PubMed] [Google Scholar]
  • 105.Mehta JL, Hu B, Chen J, Li D. Pioglitazone inhibits LOX-1 expression in human coronary artery endothelial cells by reducing intracellular superoxide radical generation. Arteriosclerosis, Thrombosis, and Vascular Biology. 2003;23(12):2203–2208. doi: 10.1161/01.ATV.0000094411.98127.5F. [DOI] [PubMed] [Google Scholar]
  • 106.Pasceri V, Wu HD, Willerson JT, Yeh ETH. Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor-γ activators. Circulation. 2000;101(3):235–238. doi: 10.1161/01.cir.101.3.235. [DOI] [PubMed] [Google Scholar]
  • 107.Kelly D, Campbell JI, King TP, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-γ and RelA. Nature Immunology. 2004;5(1):104–112. doi: 10.1038/ni1018. [DOI] [PubMed] [Google Scholar]
  • 108.Pascual G, Fong AL, Ogawa S, et al. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-γ . Nature. 2005;437(7059):759–763. doi: 10.1038/nature03988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bruemmer D, Yin F, Liu J, et al. Peroxisome proliferator-activated receptor γ inhibits expression of minichromosome maintenance proteins in vascular smooth muscle cells. Molecular Endocrinology. 2003;17(6):1005–1018. doi: 10.1210/me.2002-0410. [DOI] [PubMed] [Google Scholar]
  • 110.Bruemmer D, Yin F, Liu J, et al. Regulation of the growth arrest and DNA damage-inducible gene 45 (GADD45) by peroxisome proliferator-activated receptor γ in vascular smooth muscle cells. Circulation Research. 2003;93(4):e38–e47. doi: 10.1161/01.RES.0000088344.15288.E6. [DOI] [PubMed] [Google Scholar]
  • 111.Pfeffer MA, Braunwald E. Ventricular remodeling after myocardial infarction: experimental observations and clinical implications. Circulation. 1990;81(4):1161–1172. doi: 10.1161/01.cir.81.4.1161. [DOI] [PubMed] [Google Scholar]
  • 112.Swynghedauw B. Molecular mechanisms of myocardial remodeling. Physiological Reviews. 1999;79(1):215–262. doi: 10.1152/physrev.1999.79.1.215. [DOI] [PubMed] [Google Scholar]
  • 113.Shiomi T, Tsutsui H, Hayashidani S, et al. Pioglitazone, a peroxisome proliferator-activated receptor-γ agonist, attenuates left ventricular remodeling and failure after experimental myocardial infarction. Circulation. 2002;106(24):3126–3132. doi: 10.1161/01.cir.0000039346.31538.2c. [DOI] [PubMed] [Google Scholar]
  • 114.Chen K, Li D, Zhang X, Hermonat PL, Mehta JL. Anoxia-reoxygenation stimulates collagen type-1 and MMP-1 expression in cardiac fibroblasts: modulation by the PPAR-γ ligand pioglitazone. Journal of Cardiovascular Pharmacology. 2004;44(6):682–687. doi: 10.1097/00005344-200412000-00010. [DOI] [PubMed] [Google Scholar]
  • 115.Chen K, Chen J, Li D, Zhang X, Mehta JL. Angiotensin II regulation of collagen type I expression in cardiac fibroblasts: Modulation by PPAR-γ ligand pioglitazone. Hypertension. 2004;44(5):655–661. doi: 10.1161/01.HYP.0000144400.49062.6b. [DOI] [PubMed] [Google Scholar]
  • 116.Makino N, Sugano M, Satoh S, Oyama J, Maeda T. Peroxisome proliferator-activated receptor-γ ligands attenuate brain natriuretic peptide production and affect remodeling in cardiac fibroblasts in reoxygenation after hypoxia. Cell Biochemistry and Biophysics. 2006;44(1):65–71. doi: 10.1385/CBB:44:1:065. [DOI] [PubMed] [Google Scholar]
  • 117.Chintalgattu V, Harris GS, Akula SM, Katwa LC. PPAR-γ agonists induce the expression of VEGF and its receptors in cultured cardiac myofibroblasts. Cardiovascular Research. 2007;74(1):140–150. doi: 10.1016/j.cardiores.2007.01.010. [DOI] [PubMed] [Google Scholar]
  • 118.Taylor AM, McNamara CA. Are thiazolidinediones good or bad for the heart? Current Diabetes Reports. 2006;6(5):378–383. doi: 10.1007/s11892-006-0009-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Dormandy JA, Charbonnel B, Eckland DJ, et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial in macroVascular Events): a randomised controlled trial. Lancet. 2005;366(9493):1279–1289. doi: 10.1016/S0140-6736(05)67528-9. [DOI] [PubMed] [Google Scholar]
  • 120.Goldberg RB, Kendall DM, Deeg MA, Jacober SJ. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care. 2005;28(7):1547–1554. doi: 10.2337/diacare.28.7.1547. [DOI] [PubMed] [Google Scholar]

Articles from PPAR Research are provided here courtesy of Wiley

RESOURCES