Skip to main content

Main menu

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Subject Collections
    • JASN Podcasts
    • Archives
    • Saved Searches
    • ASN Meeting Abstracts
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Subscriptions
  • More
    • About JASN
    • Alerts
    • Advertising
    • Editorial Fellowship Program
    • Feedback
    • Reprints
    • Impact Factor
  • ASN Kidney News
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
American Society of Nephrology
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Advertisement
American Society of Nephrology

Advanced Search

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Subject Collections
    • JASN Podcasts
    • Archives
    • Saved Searches
    • ASN Meeting Abstracts
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Subscriptions
  • More
    • About JASN
    • Alerts
    • Advertising
    • Editorial Fellowship Program
    • Feedback
    • Reprints
    • Impact Factor
  • ASN Kidney News
  • Follow JASN on Twitter
  • Visit ASN on Facebook
  • Follow JASN on RSS
  • Community Forum
Obesity: What Does It Have to Do with Kidney Disease?
You have accessRestricted Access

Peroxisome Proliferator-Activated Receptor Family and Its Relationship to Renal Complications of the Metabolic Syndrome

Youfei Guan
JASN November 2004, 15 (11) 2801-2815; DOI: https://doi.org/10.1097/01.ASN.0000139067.83419.46
Youfei Guan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data Supps
  • Info & Metrics
  • View PDF
Loading

Abstract

Peroxisome proliferator-activated receptors (PPAR) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors. Three PPAR isoforms, designated PPARα, -β/δ, and -γ, have been identified and attracted enormous attention as a result of the key role that these receptors play in regulating adipogenesis, lipid metabolism, insulin sensitivity, inflammation, and BP. Growing evidence points to a causative relationship between PPAR activity and the metabolic syndrome, including insulin resistance, glucose intolerance or type 2 diabetes, obesity, dyslipidemia, hypertension, atherosclerosis, and albuminuria. Importantly, both PPAR-α activators, such as the fibric acid class of hypolipidemic drugs, and PPAR-γ agonists, including antidiabetic thiazolidinediones, have been proved to be effective for improving diverse aspects of the metabolic syndrome. All three PPAR isoforms seem to play important roles in the development of diabetic nephropathy in type 2 diabetes. Accumulating data suggesting that PPAR may serve as potential therapeutic targets for treating the metabolic syndrome and its related renal complications have begun to emerge. This article reviews the literature pertaining to the action, ligand selectivity, and physiologic role of PPAR. Particular emphasis is placed on their pathogenic roles in the metabolic syndrome and the therapeutic utility of PPAR modulators in the treatment of diabetic nephropathy.

Peroxisome proliferator-activated receptors (PPAR) are nuclear hormone-activated receptors and transcription factors. To date, three different PPAR subtypes have been cloned and characterized: PPAR-α, PPAR-β/δ, and PPAR-γ (1–4⇓⇓⇓). The ligands for PPAR have been demonstrated to include structurally diverse compounds that vary from industrial chemicals and pharmaceutical drugs to endogenous fatty acids. These ligands can induce enormous molecular and cellular changes, including peroxisome proliferation, adipogenesis, β-oxidation enhancement, and cell-cycle regulation. After a decade of intense study, much has been learned regarding the molecular mechanisms by which PPAR activation results in its biologic consequences. PPAR have been shown to be critical factors in regulating diverse biologic processes, including lipid metabolism, adipogenesis, insulin sensitivity, immune response, and cell growth and differentiation (1–4⇓⇓⇓) and participate in the pathogenesis of a cluster of human diseases designated the metabolic syndrome, which includes insulin resistance, glucose intolerance, obesity, dyslipidemia, hypertension, atherosclerosis, and microalbuminuria (5–7⇓⇓). Importantly, the fibrate class of PPAR-α agonists including fenofibrate and clofibrate are clinically proven lipid-lowering drugs (8), whereas the thiazolidinedione (TZD) class of PPAR-γ ligands such as rosiglitazone (Avandia) and pioglitazone (Actos) have recently been introduced into clinical practice for treating hyperglycemia and insulin resistance in patients with type 2 diabetes (9). Increasing evidence also points to the therapeutic potential of PPAR-β/δ activators for improving insulin sensitivity and dyslipidemia (10). In addition, PPAR ligands have been considered as potential therapeutic agents for treatment of hypertension, atherosclerosis, and diabetic nephropathy (1,11⇓). This review examines the current knowledge relating to the action, ligand selectivity, and physiologic role of the PPAR family and then discusses the current understanding of the pathogenic roles of PPAR in the metabolic syndrome, with particular focus on the therapeutic potentials of PPAR modulators in the treatment of diabetic nephropathy.

PPAR Are Ligand-Activated Nuclear Receptors and Transcription Factors

PPAR as Nuclear Receptors

PPAR was originally identified by Isseman and Green (12) after screening the liver cDNA library with a cDNA sequence located in the highly conserved C domain of nuclear hormone receptors. The predicted encoded protein shares functional domains with other nuclear hormone receptors (Figure 1). These investigators further demonstrated that chemicals that act as peroxisome proliferators were potent ligands for this new nuclear receptor, hence its designation as PPAR-α. Since this initial identification, two additional PPAR isoforms with high sequence identity were identified and designated PPAR-β/δ and PPAR-γ. These three PPAR constitute the 1C group of the nuclear hormone receptor superfamily that consists of 48 nuclear receptor (NR) members. Similar to other nuclear receptors, PPARs including PPAR-α (NR 1C1), PPAR-β/δ (NR 1C2), and PPAR-γ (NR 1C3) contain four major functional domains: the N-terminal ligand-independent transactivation domain (A/B domain), the DNA binding domain (BDB or C domain), the co-factor docking domain (D domain), and the C-terminal E/F domain including ligand binding domain (LBD) and the ligand-dependent transactivation domain (AF2 domain; Figure 1) (13). Although PPARs exhibit high homology at the amino acid level and share very similar structure, they are differentially expressed in the body and display different ligand selectivity and biologic actions. Gene mapping studies demonstrated that these three PPAR isoforms are encoded by distinct genes mapping to human chromosome 22, 6, and 3, respectively (3). To date, three splice variants of the PPAR-γ isoform, designated PPAR-γ1, PPAR-γ2, and PPAR-γ3, have also been identified. These arise from differential splicing and alternative promoter usage of the PPAR-γ gene, sharing the six 3′ coding exons but differing only in the 5′ exons. PPAR-γ1 and -γ3 share identical protein sequence (PPAR-γ1 protein). In contrast, because the 5′ exon of the PPAR-γ2 variant contains an additional translation initial site, PPAR-γ2 gene gives rise to a protein with 30 additional amino acids in the N-terminal, which are not present in PPAR-γ1 (14,15⇓). Different promoter usage and protein sequence result in distinct gene expression pattern and biologic function of PPAR-γ1 and PPAR-γ3 (15).

Figure1
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 1. Schematic representation of the domain structure of human peroxisome proliferator-activated receptor-α (PPAR-α), -β/δ, and -γ. The number inside each domain corresponds to amino acid sequence identity of human PPAR-β/δ and -γ relative to PPAR-α. The numbers above the boxes indicate amino acid positions. DBD, DNA binding domain; LBD, ligand binding domain.

Ligands for PPAR Isoforms

The divergent amino acid sequence in the LBD of the three PPAR isoforms is thought to provide the molecular basis for ligand selectivity. On the basis of the elucidated crystal structure, a large ligand-binding pocket (1300 Å) exists in all three PPAR isoforms, allowing diverse and structurally distinct compounds access to the LBD (13) and enabling PPAR to sense a broad range of endogenous substances, including fatty acids and their metabolites (16). Thus far, a variety of endogenous and exogenous compounds, including industrial chemicals such as herbicides and plasticizers as well as synthetic pharmaceutical agents including hypolipidemic fibrates (e.g., fenofibrate, clofibrate) and antidiabetic TZD, have been shown to bind to and activate PPAR (17). Although many fatty acids are capable of activating all three PPAR isoforms (1,18⇓), some preference for specific fatty acids by each PPAR has been demonstrated. Fatty acids and their derivatives, including 8-S-hydroxyeicosatetraenoic acid, the arachidonic acid lipoxygenase metabolite LTB4, and arachidonate monooxygenase metabolite epoxyeicosatrienoic acids, have been shown to potently activate PPAR-α (1,17,19⇓⇓). Synthetic lipid-lowering drug fibrates are also potent ligands for activating PPAR-α. Endogenous arachidonic acid cyclooxygenase metabolite prostacyclin, the linoleic acid 15-lipoxygenase-1 product 13-S-hydroxyoctadecadienoic acid, and synthetic compounds including L-165041 and GW2433 have been found to be selective PPAR-β/δ ligands (20,21⇓). Naturally occurring PPAR-γ ligands including 15-deoxy-Δ (12,14⇓)-prostaglandin J2 (22) and oxidized metabolites of linoleic acid 9-hydroxy- and 13-hydroxy-octadecadienoic acids have been identified (23,24⇓). Furthermore, synthetic antidiabetic TZD including rosiglitazone (Avandia) and pioglitazone (ACTOS) are potent PPAR-γ selective agonists and have been very effective in improving glycemic control via insulin sensitization. The development and clinical use of PPAR ligands in the past decade have greatly advanced our understanding of the physiologic and pathophysiologic roles of PPAR and therapeutic implications of modulating these receptors.

Mode of PPAR Action

Upon binding their cognate ligands, the transcriptional activity of PPAR is altered. A conformational change in the PPAR/retinoid X receptor-α (RXR-α) dimer allows the heterodimer to bind to PPAR-response elements (PPRE) to activate gene transcription. PPRE generally consist of a direct repeat of hexameric core recognition elements spaced by 1 bp (DR1, 5′AGGTCANAGGTCA-3′) located in the promoter regions of target genes (Figure 2) (1). After activation of the PPAR/RXR heterodimer at the PPRE, the PPAR/RXR-α complex can recruit diverse nuclear receptor co-factors that modulate transcriptional activity of PPAR and RXR-α receptor heterodimer. These coactivators include cAMP response element-binding protein, PPAR-γ coactivators, cAMP response element-binding protein binding protein, and steroid receptor coactivator-1. Co-repressors such as nuclear receptor co-repressor and silencing mediator of retinoid acid and thyroid hormone receptor can modulate the transcriptional activity of PPAR by remodeling chromatin and establishing physical contacts with transcription initiation machinery (3,25⇓). Therefore, multiple mechanisms are involved in controlling the transcription of PPAR target genes in a given cell or tissue. The expression level of PPAR receptors, the chemical properties and local concentrations of PPAR-specific ligands, and the availability of these co-factors all contribute to the biologic effect of PPAR activation or inactivation.

Figure2
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 2. Molecular mechanism of action of PPAR-α, -β/δ, and -γ. The PPAR isoforms form heterodimers with retinoid X receptor-α (RXR-α) in the presence of their ligands. The resultant heterodimer binds to PPAR-response elements (PPRE) in the promoter regions of PPAR-driven genes through DNA-binding domains of PPAR and RXR-α. Coactivators and co-repressors are essential for modulating the transcription of PPAR target genes, which are involved in many biologic processes closely related to the metabolic syndrome, including insulin sensitization, adipogenesis, and lipid metabolism.

Tissue Expression of PPAR

PPAR-α, PPAR-β/δ, and PPAR-γ are differentially expressed among tissues (2,26–28⇓⇓⇓). In general, PPAR-α is highly expressed in tissues that possess high mitochondrial and β-oxidation activity, including liver, renal cortex, intestine mucosa, and heart. Lower expression of PPAR-α is also observed in several other tissues. PPAR-γ is highly enriched in adipose tissue, but lower expression levels have also been reported in urinary bladder, intestine, kidney, spleen, adrenal, heart, liver, lung, brain, and vasculature. Unlike PPAR-α and PPAR-γ, PPAR-β/δ seems to be ubiquitously expressed at low levels in almost every tissue examined. In the kidney, PPAR-α is highly abundant in the proximal tubules and medullary thick ascending limbs with much lower levels in glomerular mesangial cells (26,29⇓), whereas PPAR-γ is primarily expressed in the distal medullary collecting ducts and to a lesser extent in glomeruli and renal microvasculature (26,30⇓). In the kidney, PPAR-β/δ seems to be diffusely expressed in renal cortex and medulla, including medullary interstitial and stromal cells (26). The differential tissue distribution and intrarenal localization of these three PPAR isoforms is consistent with the possibility that they play distinct roles in these tissues, including the kidney. Because the target genes of PPAR-α, -β/δ, and -γ in these tissues are mainly involved in adipogenesis, lipid metabolism, insulin sensitivity, glucose homeostasis, and cell growth and differentiation, PPARs are likely candidate targets for modulating the metabolic syndrome.

PPAR and the Metabolic Syndrome and Its Cardiovascular Complications

The Metabolic Syndrome

The term metabolic syndrome (also referred to as syndrome X) was first introduced by Reaven in 1988 to describe the concurrent existence of atherogenic risk factors and underlying insulin resistance (7,31⇓). The definition was then refined by the World Health Organization in 1997 to refer to a specific clustering of disorders associated with increased risk for cardiovascular disease and related mortality (32). According to the current unifying definition, key elements of the metabolic syndrome include insulin resistance (hyperinsulinemia), abnormal glucose metabolism (impaired glucose intolerance or type 2 diabetes), hypertension, atherogenic dyslipidemia (low HDL cholesterol or high triglycerides), central obesity, hyperuricemia, microalbuminuria, and hypercoagulation state (increased fibrinogen and plasminogen activator inhibitor-1 (Figure 3). For clinical diagnosis of the metabolic syndrome, there are currently two similar diagnostic criteria. The World Health Organization definition requires at least one of three major features, including type 2 diabetes, impaired glucose tolerance, and insulin resistance, plus at least two of the minor features, including hypertension, obesity, hypertriglyceridemia, and microalbuminuria (32) (Table 1). The National Cholesterol Education Program Adult Treatment Panel III Guidelines require three of five clinical criteria, including abdominal adiposity, hypertriglyceridemia, low HDL, hypertension, and fasting hyperglycemia, to make a diagnosis (33). Using either diagnostic criteria, the prevalence of the metabolic syndrome currently exceeds 20% of individuals at least 20 yr of age and 40% of the population over the age of 40 yr, affecting ∼47 million Americans (5,34⇓). A substantial proportion of the U.S. population is at risk for the major adverse health consequence of the metabolic syndrome: cardiovascular disease. There is a threefold increase in risk for coronary artery disease and stroke in patients with the syndrome (35). Therapy directed at individual features of the metabolic syndrome therefore should have a dramatic impact on lowering morbidity and mortality from cardiovascular disease.

Figure3
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 3. Key features of the metabolic syndrome or the insulin-resistant syndrome (see text for details). Note that insulin resistance is believed to be a central element in the development of the metabolic syndrome.

View this table:
  • View inline
  • View popup

Table 1. Diagnostic criteria for metabolic syndrome

Although multiple factors contribute to the metabolic syndrome, insulin resistance seems to be a central pathophysiologic process behind the metabolic syndrome (33). Insulin resistance is a well-established and major risk factor for development of type 2 diabetes. In fact, the severity of insulin resistance is one of the strongest predictors of type 2 diabetes (36). Up to 75% of individuals with type 2 diabetes also meet the diagnostic criteria for the metabolic syndrome, with insulin resistance, obesity, hypertension, dyslipidemia, and microalbuminuria (37). It has also become clear that excessive intra-abdominal obesity is associated with insulin resistance, hyperinsulinemia, glucose intolerance, and atherogenic lipid profile (5,38⇓), and an epidemic of obesity derived presumably from physical inactivity and excessive food intake contributes to the worldwide increased incidence of type 2 diabetes (39). Hypertension is another common feature of the metabolic syndrome, occurring with a prevalence of 50% in insulin-resistant patients. It has been proposed that insulin resistance is associated with impaired insulin-mediated vasodilation, contributing to hypertension (40). Finally, atherogenic dyslipidemia is a common finding in patients with type 2 diabetes, particularly in nondiabetic insulin-resistant individuals (41). Although the causality behind the association between insulin resistance and dyslipidemia remains largely controversial, lipid-lowering pharmacologic agents have been shown to produce marked reductions in cardiovascular disease events in patients with diabetes (42,43⇓).

PPAR-α and the Metabolic Syndrome

Accumulating evidence demonstrates that PPAR-α is an important modulator of the metabolic syndrome and may be a therapeutic target for treating some of its features, especially cardiovascular complications. PPAR-α has been identified as a key regulator of the genes involved in fatty acid oxidation, which occurs in mitochondria, peroxisomes, and microsomes in the liver (44). Transcription and protein levels of critical enzymes in β-oxidation and ω-oxidation pathways are direct targets of PPAR-α, including acyl CoA oxidase, carnitine palmitoyl transferase I, mitochondrial hydroxymethylglutaryl-CoA synthase, and cytochrome P450 4A enzymes (CYP4A) (1). By increasing the expression of these genes, PPAR-α ligands significantly activate hepatic fatty acid oxidation, whereas genetic inactivation of the PPAR-α gene results in massive accumulation of lipids in the liver, severe hypoketonemia, hypoglycemia, hypothermia, and elevated plasma free fatty acid levels (45). These data clearly indicate that PPAR-α is a key factor in governing metabolic adaptation to increased fatty acids.

PPAR-α also plays a critical role in lipid metabolism. Its known target genes are involved in almost all aspects of lipid metabolism, including uptake, binding, and oxidation of fatty acids; lipoprotein assembly; and lipid transport (1,46⇓). Synthetic PPAR-α ligands, such as gemfibrozil, fenofibrate, and clofibrate, have been used in clinical practice as hypolipidemic agents for >3 decades. Fibrates increase the synthesis of HDL through several PPAR target genes, including apolipoprotein A-I, apolipoprotein A-II, lipoprotein lipase, adenosine triphosphate-binding cassette transporter-1, and scavenger receptor class B type I and CLA-1, thereby enhancing the HDL cholesterol protective effect and leading to major clinical benefits. PPAR-α activation also increases fatty acid uptake, decreases triglyceride level, and promotes triglyceride-VLDL lipolysis (46–48⇓⇓). Taken together, it seems that PPAR-α may be a lipid sensor and can exert beneficial metabolic effects on lipid metabolism.

Altered PPAR-α has also been implicated in the pathogenesis of obesity and insulin resistance (49). Activation of PPAR-α reduces weight gain in rodents, and inactivation of PPAR-α results in a late onset of obese phenotype (50,51⇓). Treatment of PPAR-α null mice with a high-fat diet leads to a more dramatic increase in body weight (52), further suggesting that PPAR-α may be involved in the genesis of obesity. Evidence has recently emerged suggesting that PPAR-α is an important regulator of insulin sensitivity. Treatment with PPAR-α activators dramatically improved insulin resistance and glycemic control in type 2 diabetic db/db mice and OLETF rats (53–55⇓⇓). Similarly, the PPAR-α agonist bezafibrate markedly improved glucose intolerance and insulin resistance in a lipoatrophic diabetic patient (56). More important, Koh et al. (54) recently reported that the PPAR-α agonist fenofibrate prevents the development of diabetes in insulin-resistant obese OLETF rats. Although the downstream mechanisms underlying these observations are not clear, they are consistent with the idea that PPAR-α plays a critical role in regulating insulin sensitivity in vivo and that its activation may lead to the delay of onset of type 2 diabetes.

PPAR-α has also been implicated in BP regulation and vascular inflammation. PPAR-α is expressed in both vascular endothelial cells and smooth muscle cells (57,58⇓), where it inhibits vascular inflammation, oxidative stress, and cell growth and migration through blocking NF-κB, TGF-β/Smad, and mitogen-activated protein kinase (MAPK) pathways (59,60⇓). In vivo studies further demonstrate that PPAR-α agonists can significantly reduce angiotensin II (Ang II)–induced hypertension in rats, possibly by improving endothelial dysfunction (61). Alternatively, effects of PPAR-α activation on renal salt transport seem likely. PPAR-α has been found to co-localize with arachidonate CYP450 4A enzymes in the renal proximal tubule, and P450 4A monooxygenases are known PPAR-α target genes. Knockout cyp4A14 mice exhibit androgen-dependent hypertension (62), whereas 4A10−/− mice develop salt-sensitive hypertension (Dr. J. Capdevila, personal communication). Thus, regulation of CYP4A by PPAR-α ligands could play an important role in sodium homeostasis and BP regulation (63–65⇓⇓).

Finally, fibrate PPAR-α activators have been reported to potently reduce atherosclerosis both in apoE−/− mice and in human ApoAI transgenic apoE−/− mice (66). More important, fibrate treatment of patients who exhibit more than three features characteristic of the metabolic syndrome (diabetes, glucose intolerance or high fasting insulin, hypertension, obesity, and high triglycerides or low HDL cholesterol) was associated with a significant 35% risk reduction in the rate of coronary artery disease death, nonfatal myocardial infarction, or definite stroke (43,67,68⇓⇓). These data support the concept that fibrate PPAR-α agonists may be particularly effective agents for the cardiovascular complications of the metabolic syndrome.

In summary, PPAR-α participates in many biologic events, including lipid metabolism, energy homeostasis, inflammation, BP regulation, and insulin sensitization. It therefore may be an important factor in the pathogenesis of the metabolic syndrome. Activation of PPAR-α favors fatty acid oxidation and improves insulin resistance. Modulators of PPAR-α activity could provide useful therapeutic agents for treating some features of the metabolic syndrome and preventing the onset of type 2 diabetes.

PPAR-β/δ and the Metabolic Syndrome

Although the existence of PPAR-β/δ has been recognized for several years, its biologic role and medical relevance have just recently come under investigation. It has been found that PPAR-β/δ action encompasses a panoply of physiologic and pathophysiologic activities, including effects on reproduction, mast cell immunity, bone formation, skin and brain development, wound healing, and tumorigenesis (1). Similar to the other two isoforms of the PPAR family, emerging evidence suggests that PPAR-β/δ may be a pivotal factor in metabolic control. Early studies pointing to a possible role for PPAR-β/δ in adipogenesis showed overexpression of PPAR-β/δ in NIH 3T3 fibroblast–induced PPAR-γ2 expression. In 3T3-L1 preadipocytes, which express endogenous PPAR-γ, a PPAR-β/δ selective agonist L-165041 markedly promoted mitotic clonal expansion with only minor terminal differentiation (69). Therefore, the activation of PPAR-β/δ may be involved in adipogenesis via promoting proliferation of adipocyte precursor cells. A role for PPAR-β/δ in the pathogenesis of obesity has also been suggested on the basis of a study in which PPAR-β/δ null mice exhibited reduced fat mass (70). Nevertheless, no reduction of fat mass is observed in adipose-specific ablation of PPAR-β/δ mice (71). More recently, Evans and colleagues (72) further demonstrated that adipocyte-specific PPAR-β/δ transgenic mice exhibit a lean phenotype with increased fatty acid oxidation and energy uncoupling in fat tissue. In addition, these mice are resistant to high-fat diet–induced and leptin receptor mutation–induced obesity. It seems that PPAR-β/δ may serve as a regulator of fat catabolism and is a potential therapeutic target for treating obesity and its associated disorders.

Other studies using PPAR-β/δ selective agonists point to key roles for PPAR-β/δ in lipid metabolism. Using obese diabetic db/db mice as a model, Leibowitz et al. (10) examined the effect of a PPAR-β/δ–specific agonist L-165041 on plasma lipid profile. L-165041 treatment significantly increases HDL cholesterol levels, possibly associated with a decreased lipoprotein lipase activity in the white adipose tissue. Confirmatory results were recently obtained with a more potent and selective PPAR-β/δ agonist, GW501516 (Ki = 1.1 ± 0.1 nM) in insulin-resistant middle-aged obese rhesus monkeys (73). The results showed that GW501516 causes a dramatic dose-dependent increase in serum HDL cholesterol and a reduction in LDL and triglycerides, suggesting that activation of PPAR-β/δ is associated with a less atherogenic lipid profile.

PPAR-β/δ receptor may also be critically involved in insulin resistance. In a primate model of the metabolic syndrome, the PPAR-β/δ selective agonist GW501516 dose-dependently lowered plasma insulin levels, without adverse effects on glycemic control (73). Similarly, in ob/ob mice, a model of the metabolic syndrome, a PPAR-β/δ–specific agonist markedly improved glucose tolerance and insulin resistance (74). Although the underlying mechanism is unclear, activation of PPAR-β/δ in skeletal muscle, which has a significant role in insulin sensitivity, has been proposed to account for the beneficial metabolic effects of PPAR-β/δ agonists on lipid profile and insulin resistance, possibly as a result of increased fatty acid catabolism, cholesterol efflux, energy expenditure (74,75⇓), and oxidative capability (76) in the muscle.

Finally, PPAR-β/δ has recently been proposed as a potential target for modulating foam cell and macrophage activation in atherosclerosis. In vitro studies suggested that PPAR-β/δ activation in cultured macrophage results in increased expression of the reverse cholesterol transporter ATP-binding cassette A1 and enhances efflux of cholesterol (73). PPAR-β/δ also participates in cellular VLDL sensing and mediates VLDL-triglyceride–driven transcription events in macrophage (77). VLDL-triglyceride treatment results in triglyceride accumulation and the induction of adipocyte phenotype, which can be blocked by disruption of the PPAR-β/δ gene. These new observations suggest that agonists for PPAR-β/δ may be effective agents to reverse cholesterol deposition in foam cells in atherotic lesions and therefore decrease cardiovascular disease associated with the metabolic syndrome. It is interesting that a very recent study showed that PPARβ/δ−/− bone marrow transplantation leads to a drastic reduction in atherotic lesions compared with PPAR-β/δ+/+ bone marrow transplantation in γ-irradiated LDLR−/− mice that were fed a high-fat diet, likely as a result of disassociation of PPAR-β/δ from anti-inflammatory transcriptional repressors including BCL-6 in macrophages (78). Thus, these findings suggest that both PPAR-β/δ modulators (agonists or antagonists) may have therapeutic potential in attenuating inflammation and slowing the progression of atherosclerosis. Taken together, PPAR-β/δ is a critical player in the pathogenesis of the metabolic syndrome, and its ligands may provide useful agents for treating dyslipidemia, obesity, insulin resistance, and atherosclerosis.

PPAR-γ and the Metabolic Syndrome

Studies conducted during past decade have led to an explosion in our understanding of the role of PPAR-γ in the metabolic syndrome. PPAR-γ has been implicated in almost all aspects of the cluster of metabolic disorders, including obesity, insulin resistance, dyslipidemia, inflammation, and hypertension. Much of this evidence derives from the increasing clinical use of synthetic TZD ligands of PPAR-γ as a new class of insulin-sensitizing agents in treating type 2 diabetes. PPAR-γ may also provide an attractive therapeutic target in the treatment of obesity, hypertension, and atherosclerosis.

The first exciting discovery about PPAR-γ was the demonstration of the key role that it plays in adipogenesis, i.e., fat cell differentiation. PPAR-γ is especially abundant in adipose tissues and particularly important during adipocyte differentiation (79). Many genes involved in fatty acid transport and metabolism are transcriptionally regulated by PPAR-γ, including adipocyte fatty acid binding protein, phosphoenolpyruvate carboxykinase, acyl-CoA synthase, fatty acid transport protein (FATP), fatty acid translocase (CD36), and lipoprotein lipase (2). In preadipocytes, TZD PPAR-γ ligands alone are sufficient to induce terminal differentiation, and forced ectopic expression of PPAR-γ in fibroblasts drives adipogenesis in the presence of PPAR-γ agonists (80). In contrast, inhibition of endogenous PPAR-γ activity by a dominant-negative PPAR-γ mutant completely blocked TZD-induced preadipocyte differentiation (81). In vivo studies also support a role for PPAR-γ in fat formation. Dysfunction of PPAR-γ has also been found to have significant effects on body fat mass. PPAR-γ null mice completely lack adipose tissues, consistent with experiments showing that PPAR-γ null embryonic stem cells fail to contribute to development of fat tissues (82). Similarly, a gain of function of human PPAR-γ mutant (Pro115Gln) causes severe obesity in patients (83), whereas patients with the common Pro12Ala PPAR-γ variant that displays lower activity than wild-type are associated with lower body mass and decreased risk for type 2 diabetes (84). Collectively, these data demonstrate that PPAR-γ is a key transcription factor in adipogenesis, and overactive PPAR-γ may contribute to the pathogenesis of obesity. Although some studies indicate that TZD drugs induce a redistribution of fat away from the central compartment to the periphery (85), caution should be taken when TZD drugs are administered to patients with type 2 diabetes. The safety of these drugs for long-term usage in larger patient populations remains to be established in future clinical studies.

As discussed above, insulin resistance is a central feature of the metabolic syndrome (86). It is generally believed that the development of insulin resistance is an early event in the development and progression of type 2 diabetes. Importantly, loss-of-function mutation of PPAR-γ results in severe insulin resistance in humans (87). That synthetic TZD PPAR-γ agonists, including troglitazone (no longer available in clinical practice), rosiglitazone, and pioglitazone, effectively improve insulin sensitivity and lower blood glucose level in patients with type 2 diabetes is consistent with the possibility that PPAR-γ activation improves insulin resistance (88). In support of this, ligands of RXR-α, the obligate partner of PPAR-γ, also lower glucose in a manner that is additive to PPAR-γ agonists (89,90⇓). In addition, TZD PPAR-γ agonists have recently been found to prevent the onset of type 2 diabetes in insulin-resistant OLETF rats (54), perhaps by improving peripheral insulin action and exerting beneficial effects on pancreatic β-cells. These data support a model whereby increased PPAR-γ activity enhances insulin sensitivity. However, recent studies using both PPAR-γ antagonists and heterozygous PPAR-γ–deficient mice (PPARγ+/−) paint a more complex picture. Unexpectedly, PPARγ+/− mice presumed to have reduced PPAR-γ activity exhibit enhanced rather than reduced insulin sensitivity as well as reduced weight gain after a high-fat diet (91,92⇓). In a separate study using selective PPAR-γ antagonists, both PPAR-γ and RXR-α inhibitors ameliorated high-fat diet–induced obesity and insulin resistance in mice (93). Taken together, these findings suggest insulin sensitivity depends on an optimal range of endogenous PPAR-γ activity, although the underlying mechanisms remain largely uncharacterized.

One possible mechanism by which PPAR-γ increases insulin sensitivity relates to adipocyte-derived signaling molecules or adipocytokines. Several groups have reported that altered levels of adipocytokines, including TNF-α, IL-6, leptin, resistin, and adiponectin, may contribute to the favorable effects of TZD (94–100⇓⇓⇓⇓⇓⇓). For instance, neutralizing antibodies to resistin and recombinant adiponectin dramatically improve insulin sensitivity in db/db type 2 diabetic mice or high-fat diet–fed mice (95,97⇓). Increased TNF-α, leptin, and resistin levels and decreased adiponectin expression in adipose tissues are associated with the development of insulin resistance (99,101⇓). Conversely, reduced TNF-α, leptin, and resistin production and an increase in adiponectin synthesis were evident after TZD treatment (99). Increased glucose uptake in adipose tissue, reduction in free fatty acid burden on liver and muscle, a decrease in free fatty acids release from adipocyte, and an increase in energy expenditure also have been suggested to contribute the beneficial effect of TZD PPAR-γ agonists on insulin resistance (102).

Atherogenic dyslipidemia including elevated plasma triglycerides and reduced HDL cholesterol is a common component of the metabolic syndrome. Because PPAR-γ has been proved to be pivotal in controlling adipogenesis and fatty acid metabolism and loss-of-function mutation of PPAR-γ causes elevated triglycerides and low HDL levels in humans (87,103⇓), TZD PPAR-γ activators are expected to improve dyslipidemia in insulin-resistant individuals. In fact, significant and sustained increases in HDL cholesterol and decreased free fatty acid levels and total cholesterol to HDL cholesterol ratio have been observed in people who were treated with troglitazone and pioglitazone (104–106⇓⇓). Although TZD causes a slight increase in LDL cholesterol, they produce an increase in the level of relative large and less atherogenic buoyant LDL particles and a decrease in the number of small but more atherogenic dense LDL particles (107,108⇓). Furthermore, both TZD rosiglitazone and pioglitazone have been shown to be effective and well tolerated when used in combination with statins (107,109⇓), likely adding beneficial effects on dyslipidemia in patients with the metabolic syndrome.

PPAR-γ activity has also been suggested to play an important role in BP regulation. TZDs have been found to significantly lower BP in both diabetic animals and humans (110–114⇓⇓⇓⇓). It is generally thought that the antihypertensive effect of TZD is a result of improved insulin sensitivity, because enhanced insulin sensitivity has been found to be associated with lower BP in both diabetic animals (115–118⇓⇓⇓) and patients (119). Evidence has also emerged suggesting that direct vascular actions of PPAR-γ may also play a role. PPAR-γ has been detected in both endothelium and vascular smooth muscle cells (120–124⇓⇓⇓⇓). Studies on 1K/1C and Ang II–induced hypertensive rats and human renin/angiotensinogen transgenic mice, hypertensive models not associated with insulin resistance, show that TZD treatment significantly lowers BP (125–127⇓⇓), arguing for direct vascular action of TZD. Although the underlying mechanisms remain unclear, inhibition of vasoconstrictors such as endothelin-1 and stimulation of vasodilators including prostacyclin and nitric oxide have been proposed to play a role (128–132⇓⇓⇓⇓). PPAR-γ may also directly affect vascular smooth muscle cell tone through downregulation of Ang II receptor 1, thereby decreasing vascular contractility (133). It is also worth mentioning that TZD treatment frequently results in sodium and water retention, possibly via activating PPAR-γ in renal microvasculature and collecting duct (26,30,134,135⇓⇓⇓). The long-term consequences of TZD-associated fluid retention on BP remain to be determined. Taken together, PPAR-γ activity regulates BP through multiple mechanisms, and its ligands might provide adjunct antihypertensive agents in the metabolic syndrome. Caution must be applied when using these agents in patients who are prone to fluid retention and/or congestive heart failure.

Finally, PPAR-γ has been implicated in almost all of the pathologic processes that contribute atherosclerosis, including foam cell formation, inflammation, and cell proliferation (11). PPAR-γ promotes adipogenesis raised the worrisome possibility that PPAR-γ might be proatherogenic in patients who receive TZD treatment. However, recent data from animal studies and clinical trials indicate that TZD PPAR-γ ligands actually consistently decrease atherosclerotic lesion formation, independent of changes in circulating lipid levels, BP, and glycemic control (136–138⇓⇓). The underlying beneficial mechanisms may be via increased cholesterol efflux from macrophages by enhanced expression of the ABAC1 gene, as a consequence of activating the PPAR-γ–LXR–ABAC1 pathway (139,140⇓). Thus, although PPAR-γ is involved in both influx (oxidized LDL-CD36 pathway) and efflux (LXR-ABAC1 pathway) of cholesterol in macrophages, the net effect seems to be removal of cholesterol from macrophages, thereby blocking foam cell formation. It has been long known that PPAR-γ has anti-inflammatory effects on monocytes. PPAR-γ activation can reduce cytokine (TNF-α, IL-1, and IL-6) production (141), probably by inhibiting the activity of proinflammatory transcription factors such as NF-κB, AP-1, and STAT (142). PPAR-γ also reduces vascular smooth muscle cell proliferation, increases monocyte apoptosis, and suppresses metalloproteinase-9 expression in atherotic plaques (121,122,143,144⇓⇓⇓). In addition, PPAR-γ agonists may indirectly suppress systemic production of a proinflammatory milieu mainly by inhibiting TNF-α, plasminogen activator inhibitor-1, and IL-6 expression in adipose tissue (145,146⇓). Elevated levels of HDL cholesterol and reduced triglyceride levels may also contribute to the beneficial effect of PPAR-γ agonists in atherosclerosis (147). On the basis of these data, it seems likely that TZD PPAR-γ agonists will have beneficial effects on atherosclerosis and provide a promising therapy for the metabolic syndrome and its cardiovascular complications.

Taken together, all three PPAR isoforms play critical regulatory roles in a variety of biologic processes closely related to the metabolic syndrome, including adipogenesis, lipid metabolism, energy metabolism, insulin sensitivity, inflammation, and cell growth and differentiation. Modulators including both agonists and antagonists for the three PPAR isoforms may serve as potential therapeutic drugs in the treatment of the metabolic syndrome and delay the onset of type 2 diabetes as well.

PPAR and Renal Complications of the Metabolic Syndrome

With worldwide epidemic of the metabolic syndrome, renal complications related to each feature of this cluster of metabolic abnormalities have become a serious health concern. The primary therapy for treating the metabolic syndrome remains a multifactorial process that includes the physiologic approaches of lifestyle change, diet control, and weight loss and pharmacologic interventions including aspirin, anti-ischemic agents, antihypertensive drugs, lipid-lowering therapy, and insulin sensitizers (5,6⇓). All three PPAR isoforms have been identified as therapeutic targets in the treatment of the metabolic syndrome (see above). The fibrate class of PPAR-α agonists including clofibrate and fenofibrate and the TZD class of PPAR-γ activators including rosiglitazone and pioglitazone have been used effectively in clinical practice in treating insulin resistance, atherogenic dyslipidemia, obesity, hypertension, and atherosclerosis. Recently, increasing evidence has also suggested a therapeutic potential for PPAR activators in renal complications of the metabolic syndrome, especially diabetic nephropathy (1,17⇓). The potential therapeutic role of PPAR-α, PPAR-β/δ, and PPAR-γ in diabetic nephropathy is discussed below.

PPAR-α and Diabetic Nephropathy

Although it has been well established that PPAR-α is abundant in the kidney, the role of PPAR-α in renal physiology and diabetic nephropathy is just emerging. Given the high level in renal proximal tubules (26,148,149⇓⇓), PPAR-α has been implicated in the metabolic control of the kidney to maintain a sustained balance of energy production and expenditure (150). Activation of PPAR-α by clofibrate significantly induced expression of β-oxidation enzymes in renal cortex, including long-chain acyl-CoA dehydrogenase, medium-chain acyl-CoA dehydrogenase, and acyl-CoA oxidase (151), suggesting that renal PPAR-α might play a major role in triggering the fatty acid utilization and adaptive response to dietary lipids by the kidney. This idea is further supported by a recent study in which fasting (24 h)-induced upregulation of pyruvate dehydrogenase kinases was blunted in PPAR-α–deficient mice, indicating that PPAR-α is important for renal adaptation to starvation (152).

Although PPAR-α induction in response to fasting and hyperlipidemia seems beneficial, the effect of PPAR-α in diabetic nephropathy remains uncertain. Cardiac overexpression of PPAR-α results in increased fatty acid oxidation, elevated lipid droplets, and worsened cardiomyopathy, suggesting that PPAR-α activation may be harmful (153). These findings raise a potential therapeutic concern for patients who have diabetic nephropathy and receive oral fibrate treatment, because a marked increase in PPAR-α gene expression has been evident in diabetic kidney (154). However, clinical evidence suggests a beneficial effect of fibrate treatment on type 2 diabetes and diabetic nephropathy (155,156⇓). In normotensive patients with non–insulin-dependent diabetes, effective treatment of dyslipidemia by a PPAR-α activator gemfibrozil for 12 mo was associated with significant stabilization of urine albumin excretion (156). In line with these clinical observations, recent studies showed that fibrate treatment improves insulin resistance and glycemic control (54,157⇓) and reduces diabetic renal complications (55) in genetic rodent models of type 2 diabetes. In db/db type 2 diabetic mice, treatment with fenofibrate markedly lowers urinary albumin excretion and improves glomerular mesangial expansion (55).

To date, multiple mechanisms have been implicated in the favorable renal effect of PPAR-α activators. In addition to the systemic effect, direct renal action may play a role. One possible mechanism relates to the effect of PPAR-α activators on the mesangial matrix production induced by TGF-β signaling pathway. Because glomerular mesangial cells express PPAR-α (158) and clofibrate directly inhibits oxidant stress–induced TGF-β1 expression in these cells (159), it has been proposed that PPAR-α activators may exert direct effects on mesangial cells where PPAR-α activation blocks TGF-β signaling pathway, thereby attenuating glomerular matrix production. In agreement with this, a recent study demonstrated that fenofibrate downregulates TGF-β1 and TGF-β signaling receptor II expression and decreases collagen IV deposition in the diabetic glomeruli (55). In addition, there is evidence that starved PPAR-α null mice develop increased albuminuria and exhibit albumin accumulation in the proximal tubules (160). Thus, PPAR-α activity may facilitate albumin reabsorption and degradation in this nephron segment (160). This mechanism may contribute to the beneficial effect of PPAR-α agonists on albuminuria in type 2 diabetes. Taken together, PPAR-α is important in the pathogenesis of diabetic nephropathy and may be a reasonable therapeutic target for treating diabetic renal complications.

PPAR-β/δ and Diabetic Nephropathy

Although PPAR-β/δ seems to be abundant in the kidney (26,161⇓) and ubiquitously expressed along the nephron (26), the role of this PPAR isotype in the kidney remains poorly understood. Recently, a study by Escher et al. (161) raised a possibility that PPAR-β/δ may contribute to renal metabolic adaptation to fasting and refeeding. In their study, after an overnight fast, PPAR-β/δ mRNA in the kidney was dramatically downregulated and was rapidly restored to control levels upon refeeding. Moreover, this tight nutritional regulation is independent of the presence of PPAR-α, because similar regulation of PPAR-β/δ is observed in PPAR-α knockout mice. In cultured renal medullary interstitial cells, abundant and active PPAR-β/δ has been observed. Overexpression of PPAR-β/δ can protect cultured medullary interstitial cells from hypertonicity-induced cell death, suggesting that PPAR-β/δ is an important survival factor for medullary interstitial cells in the hypertonic condition in renal medulla (162). Given the role that PPAR-β/δ plays in the metabolic syndrome and its renal expression (see above), it is anticipated that this receptor may also be involved in the pathogenesis of diabetic nephropathy. Additional studies using PPAR-β/δ–specific agonists or PPAR-β/δ gene targeting mice should help to clarify the involvement of PPAR-β/δ in this setting.

PPAR-γ and Diabetic Nephropathy

In contrast to PPAR-α and PPAR-β/δ, PPAR-γ has a well-established therapeutic role for treating type 2 diabetes (9,163⇓). Favorable renal effects have also been seen, including both indirect systemic effect and direct renal effects. TZD therapy has been associated with marked reduction in microalbuminuria in patients with type 2 diabetes (114,164,165⇓⇓). When compared with other oral hypoglycemic agents, including metformin, glyburide, and glibenclamide, all TZD PPAR-γ agonists (troglitazone, rosiglitazone, and pioglitazone) exhibit similar glycemic control but seem to provide superior renal protection in humans with type 2 diabetes (166–168⇓⇓). In keeping with the observations that PPAR-γ is present in glomerular mesangial cells (29,169⇓), these data are consistent with direct renal action of TZD in diabetic kidney. Moreover, TZD PPAR-γ agonists have been shown to improve diabetic nephropathy in animal models of both type 1 and type 2 diabetes, further supporting the idea that systemic effect and local action both account for the beneficial renal effect of TZD in type 2 diabetes (167,170–174⇓⇓⇓⇓⇓). For example, troglitazone therapy significantly decreased urinary albumin excretion, reduced glomerular hyperfiltration, ameliorated mesangial expansion, and inhibited renal matrix protein and TGF-β expression in streptozotocin-induced type 1 and Zucker type 2 diabetic rats (172,175⇓). In a separate study, Baylis et al. (173) demonstrated that rosiglitazone treatment reduced albuminuria, improved GFR, and normalized glomerulosclerosis and tubulointerstitial fibrosis in obese type 2 diabetic rats. Finally, the possibility that direct renal effects mediate favorable action of TZD in diabetic nephropathy is strengthened by its presence in renal glomeruli and cultured mesangial cells (29,30,169,176,177⇓⇓⇓⇓) and by the fact that troglitazone is effective in slowing the progression of glomerular fibrosis in a non–insulin-resistant 5/6 nephrectomized rat model (178). Treatment of cultured mesangial cells with PPAR-γ ligands inhibits cell growth and promotes cell differentiation (30,177⇓). Activation of PPAR-γ has also been found to markedly block advanced glycosylation end products–induced mitogen-activated protein kinase activity (179) and high glucose–stimulated vascular endothelial cell growth factor expression (180), consistent with the inhibitory effect of PPAR-γ on cell proliferation of mesangial cells. Importantly, PPAR-γ activators can also suppress high glucose– and TGF-β–induced collagen I and fibronectin production in these cells (176,181⇓). Collectively, these data suggest that PPAR-γ may reverse the phenotypic change of mesangial cells, induce cell growth arrest, and decrease extracellular matrix accumulation in the diabetic condition. In addition to mesangial cells, PPAR-γ expression was found recently to be expressed in the proximal tubular cells (182,183⇓) and upregulated in the presence of high glucose (183). Activation of PPAR-γ induces the G1 phase of cell-cycle arrest, and suppresses high glucose–induced AP-1 activity and MCP-1 expression in the proximal tubular cell line HK2 cells (183). These findings suggest that PPAR-γ activators have potential antiproliferative and anti-inflammatory effects in renal proximal tubular cells and may be beneficial in slowing the development of diabetic tubulointerstitial fibrosis. In summary, PPAR-γ agonists can improve urine albumin excretion and slow the progression of glomerulosclerosis in animal models. These desirable renal effects make PPAR-γ a promising target for treating glomerular fibrotic diseases, especially diabetic nephropathy.

Conclusions

We are faced with an epidemic of global proportions in type 2 diabetes. Diabetic nephropathy related to the increased incidence of type 2 diabetes has become a dominant health concern. The metabolic syndrome and associated insulin resistance are important contributors to the pathogenesis of both diabetes and its complications. In the last decade, numerous efforts have been directed toward understanding the underlying mechanisms and developing novel therapeutic agents targeting the metabolic syndrome. PPARs comprise a subfamily of nuclear receptor and transcription factors and have been proved to play a critical role in modulating insulin resistance, hypertension, dyslipidemia, obesity, hypertension, and inflammation. Given the close relationship between PPAR activity and the metabolic syndrome, PPAR agonists and antagonists are promising therapeutic agents for diseases including type 2 diabetes, obesity, hypertension, hyperlipidemia, and atherosclerosis. Fibrate PPAR-α agonists and TZD PPAR-γ agonists have already been used successfully as clinically effective hypolipidemic drugs and insulin sensitizers. PPAR-β/δ agonists may provide additional insulin and lipid modulators via effects on skeletal muscle. In addition, there is increasing evidence suggesting that all three PPAR isoforms contribute to the metabolic control of renal function and are involved in the pathogenesis of diabetic nephropathy. PPAR-α agonists and PPAR-γ agonists both may provide therapeutic options for nephropathy in type 2 diabetes. Because each PPAR isoform plays a distinct role in the pathogenesis of the metabolic syndrome, recent effort has been focused on developing newer agents either with more selective activity or possessing dual- or pan-PPAR activating properties. Such novel drugs hold great promise in the treatment of the metabolic syndrome and its renal complications.

Acknowledgments

This work was supported by grants from that National Institute of Diabetes and Digestive and Kidney Diseases (DK-065074-01 to Youfei Guan [Y.G.], DK-37097 to Matthew Breyer) and the American Heart Association (AHA160200B to Y.G.).

I thank Dr. Matthew Breyer for helpful discussion and critical reading of this manuscript. I also thank many colleagues and collaborators for contributions to our research described in this review.

  • © 2004 American Society of Nephrology

References

  1. ↵
    Guan Y, Breyer MD: Peroxisome proliferator-activated receptors (PPARs): Novel therapeutic targets in renal disease. Kidney Int 60: 14–30, 2001
    OpenUrlCrossRefPubMed
  2. ↵
    Fajas L, Debril MB, Auwerx J: Peroxisome proliferator-activated receptor-gamma: From adipogenesis to carcinogenesis. J Mol Endocrinol 27: 1–9, 2001
    OpenUrlAbstract
  3. ↵
    Desvergene B, Wahli W: Peroxisome proliferator-activated receptor: Nuclear control of metabolism. Endocr Rev 20: 649–688, 1999
    OpenUrlCrossRefPubMed
  4. ↵
    Willson TM, Lambert MH, Kliewer SA: Peroxisome proliferator-activated receptor gamma and metabolic disease. Annu Rev Biochem 70: 341–367, 2001
    OpenUrlCrossRefPubMed
  5. ↵
    Scott CL: Diagnosis, prevention, and intervention for the metabolic syndrome. Am J Cardiol 92: 35i–42i, 2003
    OpenUrlCrossRefPubMed
  6. ↵
    Ginsberg HN: Treatment for patients with the metabolic syndrome. Am J Cardiol 91: 29E–39E, 2003
    OpenUrlCrossRefPubMed
  7. ↵
    Gurnell M, Savage DB, Chatterjee VK, O’Rahilly S: The metabolic syndrome: Peroxisome proliferator-activated receptor gamma and its therapeutic modulation. J Clin Endocrinol Metab 88: 2412–2421, 2003
    OpenUrlCrossRefPubMed
  8. ↵
    Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E, Fruchart J-C: Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation 98: 2088–2093, 1998
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Jones AB: Peroxisome proliferator-activated receptor (PPAR) modulators: Diabetes and beyond. Med Res Rev 21: 540–552, 2001
    OpenUrlCrossRefPubMed
  10. ↵
    Leibowitz MD, Fievet C, Hennuyer N, Peinado-Onsurbe J, Duez H, Bergera J, Cullinan CA, Sparrow CP, Baffic J, Berger GD, Santini C, Marquis RW, Tolman RL, Smith RG, Moller DE, Auwerx J: Activation of PPARΔ alters lipid metabolism in db/db mice. FEBS Lett 473: 333–336, 2000
    OpenUrlCrossRefPubMed
  11. ↵
    Hsueh WA, Bruemmer D: Peroxisome proliferator-activated receptor gamma: Implications for cardiovascular disease. Hypertension 43: 297–305, 2004
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Issemann I, Green S: Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347: 645–650, 1990
    OpenUrlCrossRefPubMed
  13. ↵
    Moras D, Gronemeyer H: The nuclear receptor ligand-binding domain: Structure and function. Curr Opin Cell Biol 3: 384–391, 1998
    OpenUrl
  14. ↵
    Fajas L, Auboeuf D, Raspé E, Schoonjans K, Lefebvre A-M, Saladin R, Najib J, Laville M, Fruchart J-C, Deeb S, Vidal-Puig A, Flier J, Briggs MR, Staels B, Vidal H, Auwerx J: The organization, promoter analysis, and expression of the human PPARγ gene. J Biol Chem 272: 18779–18789, 1997
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Fajas L, Fruchart JC, Auwerx J: PPARγ3 mRNA: A distinct PPARγ mRNA subtype transcribed from an independent promoter. FEBS Lett 438: 55–60, 1998
    OpenUrlCrossRefPubMed
  16. ↵
    Ricote M, Valledor AF, Glass CK: Decoding transcriptional programs regulated by PPARs and LXRs in the macrophage: Effects on lipid homeostasis, inflammation, and atherosclerosis. Arterioscler Thromb Vasc Biol 24: 230–239, 2004
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Guan Y, Breyer MD: Targeting peroxisome proliferator-activated receptors (PPARs) in kidney and urologic disease. Minerva Urol Nefrol 54: 65–79, 2002
    OpenUrlPubMed
  18. ↵
    Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, Lehmann JM: Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors α and γ. Proc Natl Acad Sci U S A 94: 4318–4323, 1997
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Cowart LA, Wei S, Hsu MH, Johnson EF, Krishna MU, Falck JR, Capdevila JH: The CYP4A isoforms hydroxylate epoxyeicosatrienoic acids to form high affinity peroxisome proliferator-activated receptor ligands. J Biol Chem 277: 35105–35112, 2002
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Brown PJ, Smith-Oliver TA, Charifson PS, Tomkinson NC, Fivush AM, Sternbach DD, Wade LE, Orband-Miller L, Parks DJ, Blanchard SG, Kliewer SA, Lehmann JM, Willson TM: Identification of peroxisome proliferator-activated receptor ligands from a biased chemical library. Chem Biol 4: 909–918, 1997
    OpenUrlCrossRefPubMed
  21. ↵
    Shureiqi I, Jiang W, Zuo X, Wu Y, Stimmel JB, Leesnitzer LM, Morris JS, Fan HZ, Fischer SM, Lippman SM: The 15-lipoxygenase-1 product 13-S-hydroxyoctadecadienoic acid down-regulates PPAR-delta to induce apoptosis in colorectal cancer cells. Proc Natl Acad Sci U S A 100: 9968–9973, 2003
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Forman B, Tontonoz P, Chen J, Brun R, Spiegelman B, Evans R: 15-Deoxy-Δ12,14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR-gamma. Cell 83: 803–812, 1995
    OpenUrlCrossRefPubMed
  23. ↵
    Nagy L, Tontonoz P, Alvarez JGA, Chen H, Evans RA: Oxidized LDL regulates macrophage gene expression through ligand activation of PPARγ. Cell 93: 229–240, 1998
    OpenUrlCrossRefPubMed
  24. ↵
    Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM: PPARγ promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 93: 241–252, 1998
    OpenUrlCrossRefPubMed
  25. ↵
    Qi C, Zhu Y, Reddy JK: Peroxisome proliferator-activated receptors, coactivators, and downstream targets. Cell Biochem Biophys 32: 187–204, 2000
    OpenUrlCrossRefPubMed
  26. ↵
    Guan Y, Zhang Y, Davis L, Breyer MD: Expression of peroxisome proliferator-activated receptors in urinary tract of rabbits and humans. Am J Physiol 273: F1013–F1022, 1997
    OpenUrlPubMed
  27. ↵
    Auboeuf D, Rieusset J, Fajas L, Vallier P, Frering V, Riou JP, Staels B, Auwerx J, Laville M, Vidal H: Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-α in humans. No alteration in adipose tissue of obese and NIDDM patients. Diabetes 46: 1319–1327, 1997
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Mukherjee R, Noonan LJD, McDonnell DP: Human and rat peroxisome proliferator activated receptors (PPARs) demonstrate similar tissue distribution but different responsiveness to PPAR activators. J Steroid Biochem Mol Biol 51: 157–166, 1994
    OpenUrlCrossRefPubMed
  29. ↵
    Ruan XZ, Moorhead JF, Fernando R, Wheeler DC, Powis SH, Varghese Z: PPAR agonists protect mesangial cells from interleukin 1β-induced intracellular lipid accumulation by activating the ABCA1 cholesterol efflux pathway. J Am Soc Nephrol 14: 593–600, 2003
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Guan Y, Zhang Y, Schneider A, Davis L, Breyer RM, Breyer MD: Peroxisome proliferator-activated receptor-gamma activity is associated with renal microvasculature. Am J Physiol Renal Physiol 281: F1036–F1046, 2001
    OpenUrlCrossRefPubMed
  31. ↵
    Reaven GM: Banting lecture 1988. Role of insulin resistance in human disease. Diabetes 37: 1595–1607, 1988
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Alberti KG, Zimmet PZ: Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: Diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med 15: 539–553, 1998
    OpenUrlCrossRefPubMed
  33. ↵
    Kereiakes DJ, Willerson JT: Metabolic syndrome epidemic. Circulation 108: 1552–1553, 2003
    OpenUrlFREE Full Text
  34. ↵
    Ford ES, Giles WH, Dietz WH: Prevalence of the metabolic syndrome among US adults: Findings from the third National Health and Nutrition Examination Survey. JAMA 287: 356–359, 2002
    OpenUrlCrossRefPubMed
  35. ↵
    Isomaa B, Almgren P, Tuomi T, Forsen B, Lahti K, Nissen M, Taskinen MR, Groop L: Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care 24: 683–689, 2001
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Henry RR: Insulin resistance: From predisposing factor to therapeutic target in type 2 diabetes. Clin Ther 25 [Suppl B]: B47–B63, 2003
    OpenUrlCrossRefPubMed
  37. ↵
    Stolar MW, Chilton RJ: Type 2 diabetes, cardiovascular risk, and the link to insulin resistance. Clin Ther 25 [Suppl B]: B4–B31, 2003
    OpenUrlCrossRefPubMed
  38. ↵
    Lebovitz HE: The relationship of obesity to the metabolic syndrome. Int J Clin Pract Suppl 134: 18–27, 2003
    OpenUrlPubMed
  39. ↵
    Williams G: Obesity and type 2 diabetes: A conflict of interests? Int J Obes Relat Metab Disord 23 [Suppl 7]: S2–S4, 1999
    OpenUrlPubMed
  40. ↵
    Stumvoll M, Haring H: Insulin resistance and insulin sensitizers. Horm Res 55 [Suppl 2]: 3–13, 2001
    OpenUrlCrossRefPubMed
  41. ↵
    Siegel RD, Cupples A, Schaefer EJ, Wilson PW: Lipoproteins, apolipoproteins, and low-density lipoprotein size among diabetics in the Framingham offspring study. Metabolism 45: 1267–1272, 1996
    OpenUrlCrossRefPubMed
  42. ↵
    Pyorala K, Pedersen TR, Kjekshus J, Faergeman O, Olsson AG, Thorgeirsson G: Cholesterol lowering with simvastatin improves prognosis of diabetic patients with coronary heart disease. A subgroup analysis of the Scandinavian Simvastatin Survival Study (4S). Diabetes Care 20: 614–620, 1997
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB, Faas FH, Linares E, Schaefer EJ, Schectman G, Wilt TJ, Wittes J: Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med 341: 410–418, 1999
    OpenUrlCrossRefPubMed
  44. ↵
    Reddy JK: Nonalcoholic steatosis and steatohepatitis. III. Peroxisomal beta-oxidation, PPAR alpha, and steatohepatitis. Am J Physiol Gastrointest Liver Physiol 281: G1333–G1339, 2001
    OpenUrlPubMed
  45. ↵
    Leone TC, Weinheimer CJ, Kelly DP: A critical role for the peroxisome proliferator-activated receptor alpha (PPARα) in the cellular fasting response: The PPARα-null mouse as a model of fatty acid oxidation disorders. Proc Natl Acad Sci U S A 96: 7473–7478, 1999
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Neve BP, Fruchart JC, Staels B: Role of the peroxisome proliferator-activated receptors (PPAR) in atherosclerosis. Biochem Pharmacol 60: 1245–1250, 2000
    OpenUrlCrossRefPubMed
  47. ↵
    Fruchart JC: Peroxisome proliferator-activated receptor-alpha activation and high-density lipoprotein metabolism. Am J Cardiol 88: 24N–29N, 2001
    OpenUrlCrossRefPubMed
  48. ↵
    Motojima K, Passilly P, Peters JM, Gonzalez FJ, Latruffe N: Expression of putative fatty acid transporter gene are regulated by peroxisome proliferator-activated receptor α and γ activators in a tissue- and inducer-specific manner. J Biol Chem 273: 16710–16714, 1998
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Seedorf U, Assmann G: The role of PPAR alpha in obesity. Nutr Metab Cardiovasc Dis 11: 189–194, 2001
    OpenUrlPubMed
  50. ↵
    Costet P, Legendre C, More J, Edgar A, Galtier P, Pineau T: Peroxisome proliferator-activated receptor alpha-isoform deficiency leads to progressive dyslipidemia with sexually dimorphic obesity and steatosis. J Biol Chem 273: 29577–29585, 1998
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Vazquez M, Merlos M, Adzet T, Laguna JC: Decreased susceptibility to copper-induced oxidation of rat-lipoproteins after fibrate treatment: Influence of fatty acid composition. Br J Pharmacol 117: 1155–1162, 1996
    OpenUrlPubMed
  52. ↵
    Kim BH, Won YS, Kim EY, Yoon M, Nam KT, Oh GT, Kim DY: Phenotype of peroxisome proliferator-activated receptor-alpha(PPARα)deficient mice on mixed background fed high fat diet. J Vet Sci 4: 239–244, 2003
    OpenUrlPubMed
  53. ↵
    Aasum E, Belke DD, Severson DL, Riemersma RA, Cooper M, Andreassen M, Larsen TS: Cardiac function and metabolism in type 2 diabetic mice after treatment with BM 17.0744, a novel PPAR-alpha activator. Am J Physiol Heart Circ Physiol 283: H949–H957, 2002
    OpenUrlCrossRefPubMed
  54. ↵
    Koh EH, Kim MS, Park JY, Kim HS, Youn JY, Park HS, Youn JH, Lee KU: Peroxisome proliferator-activated receptor (PPAR)-alpha activation prevents diabetes in OLETF rats: Comparison with PPAR-gamma activation. Diabetes 52: 2331–2337, 2003
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Park CW, Zhang Y, Fan XF, Davis L, Capdevila JH, Guan Y, Breyer MD: A PPAR alpha agonist improves diabetic nephropathy in db/db mice. J Am Soc Nephrol 14: 393A, 2003
    OpenUrl
  56. ↵
    Panz VR, Wing JR, Raal FJ, Kedda MA, Joffe BI: Improved glucose tolerance after effective lipid-lowering therapy with bezafibrate in a patient with lipoatrophic diabetes mellitus: A putative role for Randle’s cycle in its pathogenesis? Clin Endocrinol (Oxf) 46: 365–368, 1997
    OpenUrlCrossRefPubMed
  57. ↵
    Marx N, Sukhova GK, Collins T, Libby P, Plutzky J: PPARα activators inhibit cytokine-induced vascular cell adhesion molecule-1 expression in human endothelial cells. Circulation 99: 3125–3131, 1999
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, Najib J, Maclouf J, Tedgui A: Activation of human aortic smooth muscle cells is inhibited by PPARα but not PPARγ activators. Nature 393: 790–793, 1998
    OpenUrlCrossRefPubMed
  59. ↵
    Kintscher U, Lyon C, Wakino S, Bruemmer D, Feng X, Goetze S, Graf K, Moustakas A, Staels B, Fleck E, Hsueh WA, Law RE, Diep QN, Touyz RM, Schiffrin EL: PPARα inhibits TGF-β-induced β5 integrin transcription in vascular smooth muscle cells by interacting with Smad4. Circ Res 91: e35–e44, 2002
    OpenUrlCrossRefPubMed
  60. ↵
    Diep QN, Touyz RM, Schiffrin EL: Docosahexaenoic acid, a peroxisome proliferator-activated receptor-alpha ligand, induces apoptosis in vascular smooth muscle cells by stimulation of p38 mitogen-activated protein kinase. Hypertension 36: 851–855, 2000
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Diep QN, Amiri F, Touyz RM, Cohn JS, Endemann D, Neves MF, Schiffrin EL: PPARα activator effects on Ang II-induced vascular oxidative stress and inflammation. Hypertension 40: 866–871, 2002
    OpenUrlAbstract/FREE Full Text
  62. ↵
    Holla VR, Adas F, Imig JD, Zhao X, Price E Jr, Olsen N, Kovacs WJ, Magnuson MA, Keeney DS, Breyer MD, Falck JR, Waterman MR, Capdevila JH: Alterations in the regulation of androgen-sensitive Cyp 4a monooxygenases cause hypertension. Proc Natl Acad Sci U S A 98: 5211–5216, 2001
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Capdevila JH, Falck JR, Harris RC: Cytochrome P450 and arachidonic acid bioactivation. Molecular and functional properties of the arachidonate monooxygenase. J Lipid Res 41: 163–181, 2000
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Johnson EF, Palmer CNA, Griffin KJ, Hsu M-H: Role of the peroxisome proliferator-activated receptor in cytochrome P450 4A gene regulation. FASEB J 10: 1241–1248, 1996
    OpenUrlPubMed
  65. ↵
    Roman RJ, Ma YH, Frohlich B, Markham B: Clofibrate prevents the development of hypertension in Dahl salt-sensitive rats. Hypertension 21: 985–988, 1993
    OpenUrlCrossRefPubMed
  66. ↵
    Duez H, Chao YS, Hernandez M, Torpier G, Poulain P, Mundt S, Mallat Z, Teissier E, Burton CA, Tedgui A, Fruchart JC, Fievet C, Wright SD, Staels B: Reduction of atherosclerosis by the peroxisome proliferator-activated receptor alpha agonist fenofibrate in mice. J Biol Chem 277: 48051–48057, 2002
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Rubins HB, Robins SJ: Conclusions from the VA-HIT study. Am J Cardiol 86: 543–544, 2000
    OpenUrlCrossRefPubMed
  68. ↵
    Robins SJ, Rubins HB, Faas FH, Schaefer EJ, Elam MB, Anderson JW, Collins D: Insulin resistance and cardiovascular events with low HDL cholesterol: The Veterans Affairs HDL Intervention Trial (VA-HIT). Diabetes Care 26: 1513–1517, 2003
    OpenUrlAbstract/FREE Full Text
  69. ↵
    Hansen JB, Zhang H, Rasmussen TH, Petersen RK, Flindt EN, Kristiansen K: Peroxisome proliferator-activated receptor delta (PPARΔ)-mediated regulation of preadipocyte proliferation and gene expression is dependent on cAMP signaling. J Biol Chem 276: 3175–3182, 2001
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Peters JM, Lee SS, Li W, Ward JM, Gavrilova O, Everett C, Reitman ML, Hudson LD, Gonzalez FJ: Growth, adipose, brain, and skin alterations resulting from targeted disruption of the mouse peroxisome proliferator-activated receptor beta(delta). Mol Cell Biol 20: 5119–5128, 2000
    OpenUrlAbstract/FREE Full Text
  71. ↵
    Barak Y, Liao D, He W, Ong ES, Nelson MC, Olefsky JM, Boland R, Evans RM: Effects of peroxisome proliferator-activated receptor delta on placentation, adiposity, and colorectal cancer. Proc Natl Acad Sci U S A 99: 303–308, 2002
    OpenUrlAbstract/FREE Full Text
  72. ↵
    Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, Evans RM: Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 113: 159–170, 2003
    OpenUrlCrossRefPubMed
  73. ↵
    Oliver WR Jr, Shenk JL, Snaith MR, Russell CS, Plunket KD, Bodkin NL, Lewis MC, Winegar DA, Sznaidman ML, Lambert MH, Xu HE, Sternbach DD, Kliewer SA, Hansen BC, Willson TM: A selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport. Proc Natl Acad Sci U S A 98: 5306–5311, 2001
    OpenUrlAbstract/FREE Full Text
  74. ↵
    Tanaka T, Yamamoto J, Iwasaki S, Asaba H, Hamura H, Ikeda Y, Watanabe M, Magoori K, Ioka RX, Tachibana K, Watanabe Y, Uchiyama Y, Sumi K, Iguchi H, Ito S, Doi T, Hamakubo T, Naito M, Auwerx J, Yanagisawa M, Kodama T, Sakai J: Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome. Proc Natl Acad Sci U S A 100: 15924–15929, 2003
    OpenUrlAbstract/FREE Full Text
  75. ↵
    Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, Muscat GE: The peroxisome proliferator-activated receptor beta/delta agonist, GW501516, regulates the expression of genes involved in lipid catabolism and energy uncoupling in skeletal muscle cells. Mol Endocrinol 17: 2477–2493, 2003
    OpenUrlCrossRefPubMed
  76. ↵
    Luquet S, Lopez-Soriano J, Holst D, Fredenrich A, Melki J, Rassoulzadegan M, Grimaldi PA: Peroxisome proliferator-activated receptor delta controls muscle development and oxidative capability. FASEB J 17: 2299–2301, 2003
    OpenUrlCrossRefPubMed
  77. ↵
    Chawla A, Lee CH, Barak Y, He W, Rosenfeld J, Liao D, Han J, Kang H, Evans RM: PPARΔ is a very low-density lipoprotein sensor in macrophages. Proc Natl Acad Sci U S A 100: 1268–1273, 2003
    OpenUrlAbstract/FREE Full Text
  78. ↵
    Lee CH, Chawla A, Urbiztondo N, Liao D, Boisvert WA, Evans RM, Curtiss LK: Transcriptional repression of atherogenic inflammation: modulation by PPARΔ. Science 302: 453–457, 2003
    OpenUrlAbstract/FREE Full Text
  79. ↵
    Lowell BB: PPARγ: An essential regulator of adipogenesis and modulator of fat cell function. Cell 99: 239–242, 1999
    OpenUrlCrossRefPubMed
  80. ↵
    Brun RP, Tontonoz P, Forman BM, Ellis R, Chen J, Evans RM, Spiegelman BM: Differential activation of adipogenesis by multiple PPAR isoforms. Genes Dev 10: 974–984, 1996
    OpenUrlAbstract/FREE Full Text
  81. ↵
    Gurnell M, Wentworth JM, Agostini M, Adams M, Collingwood TN, Provenzano C, Browne PO, Rajanayagam O, Burris TP, Schwabe JW, Lazar MA, Chatterjee VK: A dominant-negative peroxisome proliferator-activated receptor gamma (PPARγ) mutant is a constitutive repressor and inhibits PPARγ-mediated adipogenesis. J Biol Chem 275: 5754–5759, 2000
    OpenUrlAbstract/FREE Full Text
  82. ↵
    Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM: PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4: 611–617, 1999
    OpenUrlCrossRefPubMed
  83. ↵
    Ristow M, Muller-Wieland D, Pfeiffer A, Krone W, Kahn CR: Obesity associated with a mutation in a genetic regulator of adipocyte differentiation. N Engl J Med 339: 953–959, 1998
    OpenUrlCrossRefPubMed
  84. ↵
    Deeb SS, Fajas L, Nemoto M, Pihlajamaki J, Mykkanen L, Kuusisto J, Laakso M, Fujimoto W, Auwerx J: A Pro12Ala substitution in PPARγ2 associated with decreased receptor activity, lower body mass index and improved insulin sensitivity. Nat Genet 20: 284–287, 1998
    OpenUrlCrossRefPubMed
  85. ↵
    Zimmet P: Addressing the insulin resistance syndrome: A role for the thiazolidinediones. Trends Cardiovasc Med 12: 354–362, 2002
    OpenUrlCrossRefPubMed
  86. ↵
    Moller DE: New drug targets for type 2 diabetes and the metabolic syndrome. Nature 414: 821–827, 2001
    OpenUrlCrossRefPubMed
  87. ↵
    Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, Maslen GL, Williams TD, Lewis H, Schafer AJ, Chatterjee VK, O’Rahilly S: Dominant negative mutations in human PPARγ associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 402: 880–883, 1999
    OpenUrlCrossRefPubMed
  88. ↵
    Kumar S, Prange A, Schulze J, Lettis S, Barnett AH: Troglitazone, an insulin action enhancer, improves glycaemic control and insulin sensitivity in elderly type 2 diabetic patients. Diabet Med 15: 772–779, 1998
    OpenUrlCrossRefPubMed
  89. ↵
    Mukherjee R, Davies PJ, Crombier DL, Bischoff ED, Cesario RM, Jow L, Hamanns LG, Boehm MF, Mondon CE, Nadzan AM, Paterniti JR Jr, Heyman RA: Sensitization of diabetic and obese mice to insulin by retinoid X receptor agonists. Nature 386: 407–414, 1997
    OpenUrlCrossRefPubMed
  90. ↵
    Kliewer SA, Umesono K, Noonan DJ, Heyman RA, Evans RM: Convergence of 9-cis retinoic acid and peroxisome proliferator signalling pathways through heterodimer formation of their receptors. Nature 358: 771–774, 1992
    OpenUrlCrossRefPubMed
  91. ↵
    Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T, Eto K, Tsubamoto Y, Okuno A, Murakami K, Sekihara H, Hasegawa G, Naito M, Toyoshima Y, Tanaka S, Shiota K, Kitamura T, Fujita T, Ezaki O, Aizawa S, Kadowaki T, et al.: PPAR gamma mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance. Mol Cell 4: 597–609, 1999
    OpenUrlCrossRefPubMed
  92. ↵
    Miles PDG, Barak Y, He W, Evans RM, Olefsky JM: Improved insulin-sensitivity in mice heterozygous for PPARγ deficiency. J Clin Invest 105: 287–292, 2000
    OpenUrlCrossRefPubMed
  93. ↵
    Yamauchi T, Waki H, Kamon J, Murakami K, Motojima K, Komeda K, Miki H, Kubota N, Terauchi Y, Tsuchida A, Tsuboyama-Kasaoka N, Yamauchi N, Ide T, Hori W, Kato S, Fukayama M, Akanuma Y, Ezaki O, Itai A, Nagai R, Kimura S, Tobe K, Kagechika H, Shudo K, Kadowaki T: Inhibition of RXR and PPARγ ameliorates diet-induced obesity and type 2 diabetes. J Clin Invest 108: 1001–1013, 2001
    OpenUrlCrossRefPubMed
  94. ↵
    Shuldiner AR, Yang R, Gong DW: Resistin, obesity and insulin resistance—The emerging role of the adipocyte as an endocrine organ. N Engl J Med 345: 1345–1346, 2001
    OpenUrlCrossRefPubMed
  95. ↵
    Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA: The hormone resistin links obesity to diabetes. Nature 409: 307–312, 2001
    OpenUrlCrossRefPubMed
  96. ↵
    Savage DB, Sewter CP, Klenk ES, Segal DG, Vidal-Puig A, Considine RV, O’Rahilly S: Resistin/Fizz3 expression in relation to obesity and peroxisome proliferator-activated receptor-gamma action in humans. Diabetes 50: 2199–2202, 2001
    OpenUrlAbstract/FREE Full Text
  97. ↵
    Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T: The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7: 941–946, 2001
    OpenUrlCrossRefPubMed
  98. ↵
    Yang WS, Jeng CY, Wu TJ, Tanaka S, Funahashi T, Matsuzawa Y, Wang JP, Chen CL, Tai TY, Chuang LM: Synthetic peroxisome proliferator-activated receptor-gamma agonist, rosiglitazone, increases plasma levels of adiponectin in type 2 diabetic patients. Diabetes Care 25: 376–380, 2002
    OpenUrlAbstract/FREE Full Text
  99. ↵
    Fasshauer M, Paschke R: Regulation of adipocytokines and insulin resistance. Diabetologia 46: 1594–1603, 2003
    OpenUrlCrossRefPubMed
  100. ↵
    Combs TP, Wagner JA, Berger J, Doebber T, Wang WJ, Zhang BB, Tanen M, Berg AH, O’Rahilly S, Savage DB, Chatterjee K, Weiss S, Larson PJ, Gottesdiener KM, Gertz BJ, Charron MJ, Scherer PE, Moller DE: Induction of adipocyte complement-related protein of 30 kilodaltons by PPARγ agonists: A potential mechanism of insulin sensitization. Endocrinology 143: 998–1007, 2002
    OpenUrlCrossRefPubMed
  101. ↵
    Matsuzawa Y, Funahashi T, Kihara S, Shimomura I: Adiponectin and metabolic syndrome. Arterioscler Thromb Vasc Biol 24: 29–33, 2004
    OpenUrlAbstract/FREE Full Text
  102. ↵
    Lee CH, Olson P, Evans RM: Minireview: Lipid metabolism, metabolic diseases, and peroxisome proliferator-activated receptors. Endocrinology 144: 2201–2207, 2003
    OpenUrlCrossRefPubMed
  103. ↵
    Savage DB, Tan GD, Acerini CL, Jebb SA, Agostini M, Gurnell M, Williams RL, Umpleby AM, Thomas EL, Bell JD, Dixon AK, Dunne F, Boiani R, Cinti S, Vidal-Puig A, Karpe F, Chatterjee VK, O’Rahilly S: Human metabolic syndrome resulting from dominant-negative mutations in the nuclear receptor peroxisome proliferator-activated receptor-gamma. Diabetes 52: 910–917, 2003
    OpenUrlAbstract/FREE Full Text
  104. ↵
    Fonseca VA, Valiquett TR, Huang SM, Ghazzi MN, Whitcomb RW: Troglitazone monotherapy improves glycemic control in patients with type 2 diabetes mellitus: A randomized, controlled study. The Troglitazone Study Group. J Clin Endocrinol Metab 83: 3169–3176, 1998
    OpenUrlCrossRefPubMed
  105. ↵
    Aronoff S, Rosenblatt S, Braithwaite S, Egan JW, Mathisen AL, Schneider RL: Pioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: A 6-month randomized placebo-controlled dose-response study. The Pioglitazone 001 Study Group. Diabetes Care 23: 1605–1611, 2000
    OpenUrlAbstract/FREE Full Text
  106. ↵
    Komers R, Vrana A: Thiazolidinediones—Tools for the research of metabolic syndrome X. Physiol Res 47: 215–225, 1998
    OpenUrlPubMed
  107. ↵
    Freed MI, Ratner R, Marcovina SM, Kreider MM, Biswas N, Cohen BR, Brunzell JD: Effects of rosiglitazone alone and in combination with atorvastatin on the metabolic abnormalities in type 2 diabetes mellitus. Am J Cardiol 90: 947–952, 2002
    OpenUrlCrossRefPubMed
  108. ↵
    Tack CJ, Smits P, Demacker PN, Stalenhoef AF: Troglitazone decreases the proportion of small, dense LDL and increases the resistance of LDL to oxidation in obese subjects. Diabetes Care 21: 796–799, 1998
    OpenUrlAbstract/FREE Full Text
  109. ↵
    King AB, Armstrong DU: Lipid response to pioglitazone in diabetic patients: Clinical observations from a retrospective chart review. Diabetes Technol Ther 4: 145–151, 2002
    OpenUrlCrossRefPubMed
  110. ↵
    Inzucchi SE, Maggs DG, Spollett GR, Page SL, Rife FS, Walton V, Shulman GI: Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. N Engl J Med 338: 867–872, 1998
    OpenUrlCrossRefPubMed
  111. ↵
    Ogihara T, Rakugi H, Ikegami H, Mikami H, Masuo K: Enhancement of insulin sensitivity by troglitazone lowers blood pressure in diabetic hypertensives. Am J Hypertension 8: 316–320, 1995
    OpenUrlCrossRefPubMed
  112. ↵
    Saku K, Zhang B, Ohta T, Arakawa K: Troglitazone lowers blood pressure and enhances insulin sensitivity in Watanabe heritable hyperlipidemic rabbits. Am J Hypertens 10: 1027–1033, 1997
    OpenUrlCrossRefPubMed
  113. ↵
    Yoshimoto T, Naruse M, Nishikawa M, Naruse K, Tanabe A, Seki T, Imaki T, Demura R, Aikawa E, Demura H: Antihypertensive and vasculo- and renoprotective effects of pioglitazone in genetically obese diabetic rats. Am J Physiol 272: E989–E996, 1997
    OpenUrlPubMed
  114. ↵
    Bakris G, Viberti G, Weston WM, Heise M, Porter LE, Freed MI: Rosiglitazone reduces urinary albumin excretion in type II diabetes. J Hum Hypertens 17: 7–12, 2003
    OpenUrlCrossRefPubMed
  115. ↵
    Uchida A, Nakata T, Hatta T, Kiyama M, Kawa T, Morimoto S, Miki S, Moriguchi J, Nakamura K, Fujita H, Itoh H, Sasaki S, Takeda K, Nakagawa M: Reduction of insulin resistance attenuates the development of hypertension in sucrose-fed SHR. Life Sci 61: 455–464, 1997
    OpenUrlCrossRefPubMed
  116. ↵
    Walker AB, Chattington PD, Buckingham RE, Williams G: The thiazolidinedione rosiglitazone (BRL-49653) lowers blood pressure and protects against impairment of endothelial function in Zucker fatty rats. Diabetes 48: 1448–1453, 1999
    OpenUrlAbstract
  117. ↵
    Yoshimoto T, Naruse M, Shizume H, Naruse K, Tanabe A, Tanaka M, Tago K, Irie K, Muraki T, Demura H, Zardi L: Vasculo-protective effects of insulin sensitizing agent pioglitazone in neointimal thickening and hypertensive vascular hypertrophy. Atherosclerosis 145: 333–340, 1999
    OpenUrlCrossRefPubMed
  118. ↵
    Grinsell JW, Lardinois CK, Swislocki A, Gonzalez R, Sare JS, Michaels JR, Starich GH: Pioglitazone attenuates basal and postprandial insulin concentrations and blood pressure in the spontaneously hypertensive rat. Am J Hypertens 13: 370–375, 2000
    OpenUrlCrossRefPubMed
  119. ↵
    Sung BH, Izzo JL Jr, Dandona P, Wilson MF: Vasodilatory effects of troglitazone improve blood pressure at rest and during mental stress in type 2 diabetes mellitus. Hypertension 34: 83–88, 1999
    OpenUrlAbstract/FREE Full Text
  120. ↵
    Iijima K, Yoshizumi M, Ako J, Eto M, Kim S, Hashimoto M, Sugimoto N, Liang YQ, Sudoh N, Toba K, Ouchi Y: Expression of peroxisome proliferator-activated receptor γ (PPARγ) in rat aortic smooth muscle cells. Biochem Biophys Res Commun 247: 353–356, 1998
    OpenUrlCrossRefPubMed
  121. ↵
    Law RE, Goetze S, Xi X-P, Jackson S, Kawano Y, Demer L, Fishbein MC, Meehan WP, Hsueh WA: Expression and function of PPARγ in rat and human vascular smooth muscle cells. Circulation 101: 1311–1318, 2000
    OpenUrlAbstract/FREE Full Text
  122. ↵
    Marx N, Schönbeck U, Lazar MA, Libby P, Plutzky J: Peroxisomal proliferator-activated receptor gamma activators inhibit gene expression and migration in human vascular smooth muscle cells. Circ Res 83: 1097–1103, 1998
    OpenUrlAbstract/FREE Full Text
  123. ↵
    Marx N, Bourcier T, Sukhova GK, Libby P, Plutzky J: PPARγ activation in human endothelial cells increases plasminogen activator inhibitor type-1 expression: PPARγ as a potential mediator in vascular disease. Arterioscler Thromb Vasc Biol 19: 546–551, 1999
    OpenUrlAbstract/FREE Full Text
  124. ↵
    Satoh H, Tsukamoto Y, Hashimoto N, Togo M, Hara M, Maekawa H, Isoo N, Kimura S, Watanabe T: Thiazolidinediones suppress endothelial-1 secretion from bovine vascular endothelial cells: A new possible role of PPARγ on vascular endothelial function. Biochem Biophys Res Commun 254: 757–763, 1999
    OpenUrlCrossRefPubMed
  125. ↵
    Zhang HY, Reddy SR, Kotchen TA: Antihypertensive effect of pioglitazone is not invariably associated with increased insulin sensitivity. Hypertension 24: 106–110, 1994
    OpenUrlAbstract/FREE Full Text
  126. ↵
    Ryan MJ, Didion SP, Mathur S, Faraci FM, Sigmund CD: PPAR(gamma) agonist rosiglitazone improves vascular function and lowers blood pressure in hypertensive transgenic mice. Hypertension 43: 661–666, 2004
    OpenUrlAbstract/FREE Full Text
  127. ↵
    Diep QN, El Mabrouk M, Cohn JS, Endemann D, Amiri F, Virdis A, Neves MF, Schiffrin EL: Structure, endothelial function, cell growth, and inflammation in blood vessels of angiotensin II-infused rats: Role of peroxisome proliferator-activated receptor-gamma. Circulation 105: 2296–2302, 2002
    OpenUrlAbstract/FREE Full Text
  128. ↵
    Fujiwara T, Ohsawa T, Takahashi S, Ikeda K, Okuno A, Ushiyama S, Matsuda K, Horikoshi H: Troglitazone, a new antidiabetic agent possessing radical scavenging ability, improved decreased skin blood flow in diabetic rats. Life Sci 63: 2039–2047, 1998
    OpenUrlCrossRefPubMed
  129. ↵
    Buchanan TA, Meehan WP, Jeng YY, Yang D, Chan TM, Nadler JL, Scott S, Rude RK, Hsueh WA: Blood pressure lowering by pioglitazone. Evidence for a direct vascular effect. J Clin Invest 96: 354–360, 1995
    OpenUrlCrossRefPubMed
  130. ↵
    Ghazzi MN, Perez JE, Antonucci TK, Driscoll JH, Huang SM, Faja BW, Whitcomb RW: Cardiac and glycemic benefits of troglitazone treatment in NIDDM. Diabetes 46: 433–439, 1997
    OpenUrlAbstract/FREE Full Text
  131. ↵
    Walker AB, Naderali EK, Chattington PD, Buckingham RE, Williams G: Differential vasoactive effects of the insulin sensitizers rosiglitazone (BRL 49653) and troglitazone on human small arteries in vitro. Diabetes 47: 810–814, 1998
    OpenUrlAbstract
  132. ↵
    Dobrian AD, Schriver SD, Khraibi AA, Prewitt RL: Pioglitazone prevents hypertension and reduces oxidative stress in diet-induced obesity. Hypertension 43: 48–56, 2004
    OpenUrlAbstract/FREE Full Text
  133. ↵
    Takeda K, Ichiki T, Tokunou T, Funakoshi Y, Iino N, Hirano K, Kanaide H, Takeshita A: Peroxisome proliferator-activated receptor gamma activators downregulate angiotensin II type 1 receptor in vascular smooth muscle cells. Circulation 102: 1834–1839, 2000
    OpenUrlAbstract/FREE Full Text
  134. ↵
    Hong G, Lockhart A, Davis B, Rahmoune H, Baker S, Ye L, Thompson P, Shou Y, O’Shaughnessy K, Ronco P, Brown J: PPARγ activation enhances cell surface ENaCα via up-regulation of SGK1 in human collecting duct cells. FASEB J 17: 1966–1968, 2003
    OpenUrlCrossRefPubMed
  135. ↵
    Yang T, Michele DE, Park J, Smart AM, Lin Z, Brosius FC III, Schnermann JB, Briggs JP: Expression of peroxisomal proliferator-activated receptors and retinoid X receptors in the kidney. Am J Physiol 277: F966–F973, 1999
    OpenUrlPubMed
  136. ↵
    Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass CK: Peroxisome proliferator-activated receptor gamma ligands inhibit development of atherosclerosis in LDL receptor-deficient mice. J Clin Invest 106: 523–531, 2000
    OpenUrlCrossRefPubMed
  137. ↵
    Minamikawa J, Tanaka S, Yamauchi M, Inoue D, Koshiyama H: Potent inhibitory effect of troglitazone on carotid arterial wall thickness in type 2 diabetes. J Clin Endocrinol Metab 83: 1818–1820, 1998
    OpenUrlCrossRefPubMed
  138. ↵
    Collins AR, Meehan WP, Kintscher U, Jackson S, Wakino S, Noh G, Palinski W, Hsueh WA, Law RE: Troglitazone inhibits formation of early atherosclerotic lesions in diabetic and nondiabetic low density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol 21: 365–371, 2001
    OpenUrlAbstract/FREE Full Text
  139. ↵
    Chawla A, Boisvert WA, Lee CH, Laffitte BA, Barak Y, Joseph SB, Liao D, Nagy L, Edwards PA, Curtiss LK, Evans RM, Tontonoz P: A PPAR gamma-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell 7: 161–171, 2001
    OpenUrlCrossRefPubMed
  140. ↵
    Chinetti G, Lestavel S, Bocher V, Remaley AT, Neve B, Torra IP, Teissier E, Minnich A, Jaye M, Duverger N, Brewer HB, Fruchart JC, Clavey V, Staels B: PPAR-alpha and PPAR-gamma activators induce cholesterol removal from human macrophage foam cells through stimulation of the ABCA1 pathway. Nat Med 7: 53–58, 2001
    OpenUrlCrossRefPubMed
  141. ↵
    Jiang C, Ting AT, Seed B: PPARγ agonists inhibit production of monocyte inflammatory cytokines. Nature 391: 82–86, 1998
    OpenUrlCrossRefPubMed
  142. ↵
    Poynter ME, Daynes RA: Peroxisome proliferator-activated receptor alpha activation modulates cellular redox status, represses nuclear factor-κB signaling, and reduces inflammatory cytokine production in aging. J Biol Chem 273: 32833–32841, 1998
    OpenUrlAbstract/FREE Full Text
  143. ↵
    Chinetti G, Griglio S, Antonucci M, Torra IP, Delerive P, Majd Z, Fruchart J-C, Chapman J, Najib J, Staels B: Activation of proliferator-activator receptors α and γ induces apoptosis of human monocyte-derived macrophages. J Biol Chem 273: 25573–25580, 1998
    OpenUrlAbstract/FREE Full Text
  144. ↵
    Law RE, Meehan WP, Xi X-P, Graf K, Wuthrich DA, Coats W, Faxon D: Troglitazone inhibits vascular smooth muscle cell growth and intimal hyperplasia. J Clin Invest 98: 1897–1905, 1996
    OpenUrlCrossRefPubMed
  145. ↵
    Daynes RA, Jones DC: Emerging roles of PPARs in inflammation and immunity. Nat Rev Immunol 2: 748–759, 2002
    OpenUrlCrossRefPubMed
  146. ↵
    Delerive P, Fruchart JC, Staels B: Peroxisome proliferator-activated receptors in inflammation control. J Endocrinol 169: 453–459, 2001
    OpenUrlAbstract
  147. ↵
    Murakami K, Tobe K, Ide T, Mochizuki T, Ohashi M, Akanuma Y, Yazaki Y, Kadowaki T: A novel insulin sensitizer acts as a coligand for peroxisome proliferator-activated receptor-alpha (PPAR-alpha) and PPAR-gamma: Effect of PPAR-alpha activation on abnormal lipid metabolism in liver of Zucker fatty rats. Diabetes 47: 1841–1847, 1998
    OpenUrlAbstract
  148. ↵
    Beck F, Plummer S, Senior PV, Byrne S, Green S, Brammar WJ: The ontogeny of peroxisome-proliferator-activated receptor gene expression in the mouse and rat. Proc R Soc Lond B Biol Sci 247: 83–87, 1992
    OpenUrlCrossRefPubMed
  149. ↵
    Portilla D, Dai G, Peters JM, Gonzalez FJ, Crew MD, Proia AD: Etomoxir-induced PPARα-modulated enzymes protect during acute renal failure. Am J Physiol Renal Physiol 278: F667–F675, 2000
    OpenUrlPubMed
  150. ↵
    Portilla D: Energy metabolism and cytotoxicity. Semin Nephrol 23: 432–438, 2003
    OpenUrlCrossRefPubMed
  151. ↵
    Ouali F, Djouadi F, Merlet-Bénichou C, Bastin J: Dietary lipids regulate β-oxidation enzyme gene expression in the developing rat kidney. Am J Physiol 275: F777–F784, 1998
    OpenUrlPubMed
  152. ↵
    Sugden MC, Bulmer K, Gibbons GF, Holness MJ: Role of peroxisome proliferator-activated receptor-alpha in the mechanism underlying changes in renal pyruvate dehydrogenase kinase isoform 4 protein expression in starvation and after refeeding. Arch Biochem Biophys 395: 246–252, 2001
    OpenUrlCrossRefPubMed
  153. ↵
    Finck BN, Han X, Courtois M, Aimond F, Nerbonne JM, Kovacs A, Gross RW, Kelly DP: A critical role for PPARα-mediated lipotoxicity in the pathogenesis of diabetic cardiomyopathy: Modulation by dietary fat content. Proc Natl Acad Sci U S A 100: 1226–1231, 2003
    OpenUrlAbstract/FREE Full Text
  154. ↵
    Mishra R, Emancipator SN, Miller C, Kern T, Simonson MS: Adipose differentiation related protein and regulates of lipid homeostasis identified by gene expression profiling in murine db/db diabetic kidney. Am J Physiol Renal Physiol 286: F913–F921, 2004
    OpenUrlCrossRefPubMed
  155. ↵
    Fried LF, Orchard TJ, Kasiske BL: Effect of lipid reduction on the progression of renal disease: A meta-analysis. Kidney Int 59: 260–269, 2001
    OpenUrlCrossRefPubMed
  156. ↵
    Smulders YM, van Eeden AE, Stehouwer CD, Weijers RN, Slaats EH, Silberbusch J: Can reduction in hypertriglyceridaemia slow progression of microalbuminuria in patients with non-insulin-dependent diabetes mellitus? Eur J Clin Invest 27: 997–1002, 1997
    OpenUrlCrossRefPubMed
  157. ↵
    Kim H, Haluzik M, Asghar Z, Yau D, Joseph JW, Fernandez AM, Reitman ML, Yakar S, Stannard B, Heron-Milhavet L, Wheeler MB, LeRoith D: Peroxisome proliferator-activated receptor-alpha agonist treatment in a transgenic model of type 2 diabetes reverses the lipotoxic state and improves glucose homeostasis. Diabetes 52: 1770–1778, 2003
    OpenUrlAbstract/FREE Full Text
  158. ↵
    Scholz-Pedretti K, Gans A, Beck KF, Pfeilschifter J, Kaszkin M: Potentiation of TNF-alpha-stimulated group IIA phospholipase A(2) expression by peroxisome proliferator-activated receptor alpha activators in rat mesangial cells. J Am Soc Nephrol 13: 611–620, 2002
    OpenUrlAbstract/FREE Full Text
  159. ↵
    Wilmer WA, Dixon CL, Hebert C, Lu L, Rovin BH: PPAR-alpha ligands inhibit H2O2-mediated activation of transforming growth factor-beta1 in human mesangial cells. Antioxid Redox Signal 4: 877–884, 2002
    OpenUrlCrossRefPubMed
  160. ↵
    Kamijo Y, Hora K, Tanaka N, Usuda N, Kiyosawa K, Nakajima T, Gonzalez FJ, Aoyama T: Identification of functions of peroxisome proliferator-activated receptor alpha in proximal tubules. J Am Soc Nephrol 13: 1691–1702, 2002
    OpenUrlAbstract/FREE Full Text
  161. ↵
    Hao CM, Redha R, Morrow J, Breyer MD: Peroxisome proliferator-activated receptor delta activation promotes cell survival following hypertonic stress. J Biol Chem 277: 21341–21345, 2002
    OpenUrlAbstract/FREE Full Text
  162. ↵
    Hao CM, Redha R, Morrow J, Guan YF, Breyer MD: Hypertonic stress induces COX2 dependent prostacyclin synthesis in renal medullary interstitial cell (RMICs) and down-stream activation of PPARΔ dependent survival mechanism. J Am Soc Nephrol 12: 49A, 2001
    OpenUrl
  163. ↵
    Evans RM, Barish GD, Wang YX: PPARs and the complex journey to obesity. Nat Med 10: 355–361, 2004
    OpenUrlCrossRefPubMed
  164. ↵
    Nakamura T, Ushiyama C, Suzuki S, Shimada N, Sekizuka K, Ebihara L, Koide H: Effect of troglitazone on urinary albumin excretion and serum type IV collagen concentrations in type 2 diabetic patients with microalbuminuria or macroalbuminuria. Diabet Med 18: 308–313, 2001
    OpenUrlCrossRefPubMed
  165. ↵
    Lebovitz HE, Dole JF, Patwardhan R, Rappaport EB, Freed MI: Rosiglitazone monotherapy is effective in patients with type 2 diabetes. J Clin Endocrinol Metab 86: 280–288, 2001
    OpenUrlCrossRefPubMed
  166. ↵
    Nakamura T, Ushiyama C, Shimada N, Hayashi K, Ebihara I, Koide H: Comparative effects of pioglitazone, glibenclamide, and voglibose on urinary endothelin-1 and albumin excretion in diabetes patients. J Diabetes Complications 14: 250–254, 2000
    OpenUrlCrossRefPubMed
  167. ↵
    Imano E, Kanda T, Nakatani Y, Nishida T, Arai K, Motomura M, Kajimoto Y, Yamasaki Y, Hori M: Effect of troglitazone on microalbuminuria in patients with incipient diabetic nephropathy. Diabetes Care 21: 2135–2139, 1998
    OpenUrlAbstract/FREE Full Text
  168. ↵
    Wolffenbuttel BH, Gomis R, Squatrito S, Jones NP, Patwardhan RN: Addition of low-dose rosiglitazone to sulphonylurea therapy improves glycaemic control in type 2 diabetic patients. Diabet Med 17: 40–47, 2000
    OpenUrlCrossRefPubMed
  169. ↵
    Nicholas SB, Kawano Y, Wakino S, Collins AR, Hsueh WA: Expression and function of peroxisome proliferator-activated receptor-gamma in mesangial cells. Hypertension 37: 722–727, 2001
    OpenUrlAbstract/FREE Full Text
  170. ↵
    Fujii M, Takemura R, Yamaguchi M, Hasegawa G, Shigeta H, Nakano K, Kondo M: Troglitazone (CS-045) ameliorates albuminuria in streptozotocin-induced diabetic rats. Metabolism 46: 981–983, 1997
    OpenUrlCrossRefPubMed
  171. ↵
    Buckingham RE, Al-Barazanji KA, Toseland CDN, Slaughter M, Connor SC, West A, Bond B, Turner NC, Clapham JC: Peroxisome proliferator-activated receptor-γ agonist, rosiglitazone, protects against nephropathy and pancreatic islet abnormalities in Zucker fatty rats. Diabetes 47: 1326–1334, 1998
    OpenUrlAbstract/FREE Full Text
  172. ↵
    Isshiki K, Haneda M, Koya D, Maeda S, Sugimoto T, Kikkawa R: Thiazolidinedione compounds ameliorate glomerular dysfunction independent of their insulin-sensitizing action in diabetic rats. Diabetes 49: 1022–1032, 2000
    OpenUrlAbstract
  173. ↵
    Baylis C, Atzpodien EA, Freshour G, Engels K: Peroxisome proliferator-activated receptor [gamma] agonist provides superior renal protection versus angiotensin-converting enzyme inhibition in a rat model of type 2 diabetes with obesity. J Pharmacol Exp Ther 307: 854–860, 2003
    OpenUrlAbstract/FREE Full Text
  174. ↵
    Zhang Y, Park CW, Zheng F, Fan X, Striker G, Breyer MD, Guan Y: Endogenous PPARγ activity ameliorates diabetic nephropathy. J Am Soc Nephrol 14: 392A, 2003
    OpenUrl
  175. ↵
    McCarthy KJ, Routh RE, Shaw W, Walsh K, Welbourne TC, Johnson JH: Troglitazone halts diabetic glomerulosclerosis by blockade of mesangial expansion. Kidney Int 58: 2341–2350, 2000
    OpenUrlCrossRefPubMed
  176. ↵
    Zheng F, Fornoni A, Elliot SJ, Guan YF, Breyer MD, Striker LJ, Striker GE: Upregulation of type I collagen by TGF-β in mesangial cells is blocked by PPAR activation. Am J Physiol Renal Physiol 282: F639–F648, 2002
    OpenUrlCrossRefPubMed
  177. ↵
    Asano T, Wakisaka M, Yoshinari M, Iino K, Sonoki K, Iwase M, Fujishima M: Peroxisome proliferator-activated receptor γ1 (PPARγ1) expresses in rat mesangial cells and PPARγ agonists modulate its differentiation. Biochim Biophys Acta 1497: 148–154, 2000
    OpenUrlPubMed
  178. ↵
    Ma L, Marcantoni C, Fogo AB: Peroxisome proliferator-activated receptor-γ (PPARγ) agonist troglitazone (TGL) protects against non-diabetic glomerulosclerosis. Lab Invest 80: 176A, 2000
    OpenUrl
  179. ↵
    Chang PC, Chen TH, Chang CJ, Hou CC, Chan P, Lee HM: Advanced glycosylation end products induce inducible nitric oxide synthase (iNOS) expression via a p38 MAPK-dependent pathway. Kidney Int 65: 1664–1675, 2004
    OpenUrlCrossRefPubMed
  180. ↵
    Onozaki A, Midorikawa S, Sanada H, Hayashi Y, Baba T, Katoh T, Watanabe T: Rapid change of glucose concentration promotes mesangial cell proliferation via VEGF: Inhibitory effects of thiazolidinedione. Biochem Biophys Res Commun 317: 24–29, 2004
    OpenUrlCrossRefPubMed
  181. ↵
    Guo B, Koya D, Isono M, Sugimoto T, Kashiwagi A, Haneda M: Peroxisome proliferator-activated receptor-γ ligands inhibit TGF-β1-induced fibronectin expression in glomerular mesangial cells. Diabetes 53: 200–208, 2004
    OpenUrlAbstract/FREE Full Text
  182. ↵
    Chana RS, Lewington AJ, Brunskill NJ: Differential effects of peroxisome proliferator activated receptor-gamma (PPAR gamma) ligands in proximal tubular cells: Thiazolidinediones are partial PPAR gamma agonists. Kidney Int 65: 2081–2090, 2004
    OpenUrlCrossRefPubMed
  183. ↵
    Panchapakesan U, Pollock CA, Chen XM: The effect of high glucose and PPAR-αagonists on PPAR-αexpression and function in HK-2 cells. Am J Physiol Renal Physiol 287: F528–F534, 2004
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top

In this issue

Journal of the American Society of Nephrology: 15 (11)
Journal of the American Society of Nephrology
Vol. 15, Issue 11
1 Nov 2004
  • Table of Contents
  • Index by author
View Selected Citations (0)
Print
Download PDF
Sign up for Alerts
Email Article
Thank you for your help in sharing the high-quality science in JASN.
Enter multiple addresses on separate lines or separate them with commas.
Peroxisome Proliferator-Activated Receptor Family and Its Relationship to Renal Complications of the Metabolic Syndrome
(Your Name) has sent you a message from American Society of Nephrology
(Your Name) thought you would like to see the American Society of Nephrology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Peroxisome Proliferator-Activated Receptor Family and Its Relationship to Renal Complications of the Metabolic Syndrome
Youfei Guan
JASN Nov 2004, 15 (11) 2801-2815; DOI: 10.1097/01.ASN.0000139067.83419.46

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Peroxisome Proliferator-Activated Receptor Family and Its Relationship to Renal Complications of the Metabolic Syndrome
Youfei Guan
JASN Nov 2004, 15 (11) 2801-2815; DOI: 10.1097/01.ASN.0000139067.83419.46
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • PPAR Are Ligand-Activated Nuclear Receptors and Transcription Factors
    • PPAR and the Metabolic Syndrome and Its Cardiovascular Complications
    • PPAR and Renal Complications of the Metabolic Syndrome
    • Conclusions
    • Acknowledgments
    • References
  • Figures & Data Supps
  • Info & Metrics
  • View PDF

More in this TOC Section

  • Overview: Obesity: What Does It Have to Do with Kidney Disease?
  • The Metabolic Syndrome as a Risk Factor for Chronic Kidney Disease: More than a Fat Chance?
  • Obesity-Initiated Metabolic Syndrome and the Kidney: A Recipe for Chronic Kidney Disease?
Show more Obesity: What Does It Have to Do with Kidney Disease?

Cited By...

  • Activation of Peroxisome Proliferator-Activated Receptor {delta} Inhibits Streptozotocin-Induced Diabetic Nephropathy Through Anti-Inflammatory Mechanisms in Mice
  • Lipotoxicity in Diabetic Nephropathy: The Potential Role of Fatty Acid Oxidation
  • Role of PPAR{gamma} in renoprotection in Type 2 diabetes: molecular mechanisms and therapeutic potential
  • Peroxisome Proliferator Activated Receptor {alpha}/{gamma} Dual Agonist Tesaglitazar Attenuates Diabetic Nephropathy in db/db Mice
  • Obesity and Obesity-Initiated Metabolic Syndrome: Mechanistic Links to Chronic Kidney Disease
  • Transcriptional Regulation of Nephrin Gene by Peroxisome Proliferator-Activated Receptor-{gamma} Agonist: Molecular Mechanism of the Antiproteinuric Effect of Pioglitazone
  • Heart Failure and Nephropathy: Catastrophic and Interrelated Complications of Diabetes
  • Prevention of Progression in Diabetic Nephropathy
  • Hepatocyte Growth Factor Is a Downstream Effector that Mediates the Antifibrotic Action of Peroxisome Proliferator-Activated Receptor-{gamma} Agonists
  • Diet-induced Obesity in C57BL/6J Mice Causes Increased Renal Lipid Accumulation and Glomerulosclerosis via a Sterol Regulatory Element-binding Protein-1c-dependent Pathway
  • Overview: Obesity: What Does It Have to Do with Kidney Disease?
  • Google Scholar

Similar Articles

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Articles

  • Current Issue
  • Early Access
  • Subject Collections
  • Article Archive
  • ASN Annual Meeting Abstracts

Information for Authors

  • Submit a Manuscript
  • Author Resources
  • Editorial Fellowship Program
  • ASN Journal Policies
  • Reuse/Reprint Policy

About

  • JASN
  • ASN
  • ASN Journals
  • ASN Kidney News

Journal Information

  • About JASN
  • JASN Email Alerts
  • JASN Key Impact Information
  • JASN Podcasts
  • JASN RSS Feeds
  • Editorial Board

More Information

  • Advertise
  • ASN Podcasts
  • ASN Publications
  • Become an ASN Member
  • Feedback
  • Follow on Twitter
  • Password/Email Address Changes
  • Subscribe

© 2021 American Society of Nephrology

Print ISSN - 1046-6673 Online ISSN - 1533-3450

Powered by HighWire