Next Article in Journal
A20 Promotes Ripoptosome Formation and TNF-Induced Apoptosis via cIAPs Regulation and NIK Stabilization in Keratinocytes
Next Article in Special Issue
Vascular Lipidomic Profiling of Potential Endogenous Fatty Acid PPAR Ligands Reveals the Coronary Artery as Major Producer of CYP450-Derived Epoxy Fatty Acids
Previous Article in Journal
Cycloastragenol as an Exogenous Enhancer of Chondrogenic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. A Morphological Study
Previous Article in Special Issue
Vascular PPARβ/δ Promotes Tumor Angiogenesis and Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

PPAR-Mediated Toxicology and Applied Pharmacology

1
School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
2
Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
*
Authors to whom correspondence should be addressed.
Cells 2020, 9(2), 352; https://doi.org/10.3390/cells9020352
Submission received: 5 January 2020 / Revised: 26 January 2020 / Accepted: 30 January 2020 / Published: 3 February 2020
(This article belongs to the Special Issue The Role of PPARs in Disease)

Abstract

:
Peroxisome proliferator-activated receptors (PPARs), members of the nuclear hormone receptor family, attract wide attention as promising therapeutic targets for the treatment of multiple diseases, and their target selective ligands were also intensively developed for pharmacological agents such as the approved drugs fibrates and thiazolidinediones (TZDs). Despite their potent pharmacological activities, PPARs are reported to be involved in agent- and pollutant-induced multiple organ toxicity or protective effects against toxicity. A better understanding of the protective and the detrimental role of PPARs will help to preserve efficacy of the PPAR modulators but diminish adverse effects. The present review summarizes and critiques current findings related to PPAR-mediated types of toxicity and protective effects against toxicity for a systematic understanding of PPARs in toxicology and applied pharmacology.

Graphical Abstract

1. Introduction

Peroxisome proliferator-activated receptors (PPARs), a group of nuclear hormone receptors, are composed of three isoforms which were identified as PPARα, PPARγ, and PPARβ. Each is encoded by distinct genes and has different targeting ligands, tissue distribution, and biological activities. PPAR family proteins, like other nuclear receptors, have three main functional segments, activation function 1 (AF1) and the conserved DNA-binding domain (DBD), the hinge region, and the ligand-binding domain (LBD) and AF2. The variable N-terminal regulatory AF1 domain binds co-regulators and the conserved DBD, which can bind to the peroxisome proliferator response elements (PPREs). The mobile hinge region links DBD and the conserved LBD in the middle. LBD and the variable C-terminal AF2 domain form a large ligand binding pocket [1,2]. Due to the large LBD pocket, PPARs have the capacity to bind various compounds, including endogenous or synthetic ligands and xenobiotic chemicals. In pharmacology, the ligands of PPARs are classified into full agonists, partial agonists, neutral antagonists, and inverse agonists. Recently, we summarized the 84 types of PPAR synthetic ligands for the treatment of various diseases in current clinical drug applications [3]. The LBD contains a C-terminal AF2 motif that is a ligand-dependent activation region [4]. Under physiological conditions, PPARs bind with co-repressors and form heterodimers with retinoid X receptor (RXR) [5]. In response to ligand activation, the protein conformation is changed and stabilized, which leads to dissociation of co-repressors and the recruitment of transcription co-activators and DNA-binding cofactors. This complex regulates transcription of target genes by binding specific DNA sequences, called peroxisome proliferator response elements (PPREs), on promoter regions of target genes [6,7].
PPAR activated genes play critical roles in fatty acid transportation and catabolism, glucose metabolism, adipogenesis, thermogenesis, cholesterol transportation and biosynthesis, and anti-inflammatory response [4,8]. Because of their broad-spectrum biological activities, PPARs arouse much attention, and they are studied intensively. Accumulated studies show that activation of PPARs has unique pharmacological effects on cardiovascular function, neurodegeneration, inflammation, cancer, fertility, and reproduction, and it is well established for managing dyslipidemia, diabetes, insulin resistance, and metabolic syndrome, which stimulates researchers to persistently develop more new drugs targeting PPARs [9,10]. Some PPAR agonists are approved as clinical agents such as thiazolidinediones, fibrates, and glitazars, for the treatment of diabetes, dyslipidemia, and diabetes-associated complications, respectively.
Despite the multiple biological activities of PPARs, several studies and clinical cases indicated that PPARs mediate various adverse effects of drugs, especially PPAR ligands or xenobiotic chemical-induced toxicity in different systems. Thiazolidinediones (TZDs), one class of PPARγ agonists, can cause fluid retention, heart failure, and hepatotoxicity [11,12]. Glitazones, another form of PPARγ ligand, were reported to cause peripheral edema, congestive heart failure, and body weight gain. Gemfibrozil, as a valuable agent to coronary heart disease, was shown to induce tumorigenesis, muscle weakness, and liver hypertrophy [13]. The detailed information and adverse reactions or toxicity of the 18 approved clinical agents that target PPARs are summarized in Table 1. Because of the high prevalence of tumorigenesis in PPAR activation by synthetic compounds, the Food and Drug Administration (FDA) requires that any PPAR agonists undergo a two-year rodent carcinogenicity study before being tested in clinical trials [13]. Moreover, PPARs were shown to be involved in pollutant-induced toxicity in the cardiovascular system, liver, reproductive and developmental system, gastrointestinal tract, muscle, and nervous system.
Based on the above information, this review is focused on the reports of PPAR activation-mediated toxicity and protective effects to date, aiming to provide an overview of studies evaluating the toxic role of PPARs in various systems and the molecular mechanisms of PPAR-elicited toxicity.

2. PPARs in Cardiotoxicity

Considering the high expression level of PPARs in cardiac muscles and their strong implication in metabolic disorders and endocrine disruption, interference with PPARs can affect metabolic homeostasis and development of the cardiovascular system.
Cardiac edemas and the impairment of cardiac development were observed in marine medaka larvae fish exposed to perfluorooctane sulfonate (PFOS) by interfering PPARα and PPARβ [14,15]. Perfluorooctanoic acid (PFOA) exposure-induced right-ventricular wall thinning elevation in chicken embryos is also likely due to PPARα [16]. After exposure to di-ethyl-hexylphthalate (DEHP), changes in the metabolic profile via the PPARα pathway can be detected in rat cardiomyocytes [17]. Triclocarban (TCC) is a high-performance broad-spectrum fungicide, which can induce cardiac metabolic alterations in mice by suppression of PPARα messenger RNA (mRNA) expression and other enzymes involved in energy and lipid metabolism. A further study found TCC directly interacted with the active site of PPARα in both mice and human tissues [18]. Exposure to airborne particulate matter is positively correlated with cardiorespiratory mortality [19]. Some studies showed that heart abnormal energy metabolism caused by seasonal ambient fine particles (PM2.5) was related to PPARα-regulated fatty-acid and glucose transporters. Unmanaged heart abnormal energy metabolism eventually leads to cardiac damage and heart failure [20]. Considerable research suggested that PPARs play pivotal roles in myocardial energy dysfunction. Energy substrate utilization showed a marked shift from fatty acid to glucose and lactate and cardiac hypertrophy in PPARα−/− hearts [21]. PPARα-null hearts with decreased contractile and metabolic remodeling were rescued by enhancing myocardial glucose transportation and utilization [21].
Furthermore, PPARγ inhibits cardiac growth and embryonic gene expression and decreases nuclear factor kappa B (NF-κB) activity in mice [22]. Cardiomyocyte-specific PPARγ knockout mice were more susceptible to cardiac hypertrophy with systolic cardiac function [22]. CKD-501, a new selective PPARγ agonist, induced heart toxicity in db/db mice by PPARγ-dependent mechanism [23]. Rosiglitazone leads to cardiac hypertrophy partially independent of cardiomyocyte PPARγ [22]. Another study indicated that rosiglitazone caused oxidative stress-induced mitochondrial dysfunction via PPARγ-independent pathways in mouse hearts [24]. Anna et al. reported that atorvastatin ameliorated cardiac hypertrophy by improving the protein expression of PPARα and PPARβ, which regulated the gene expression involved in fatty acid metabolism and avoided NF-κB activation by reducing the protein–protein interaction between PPARs and p65 [25]. Moreover, atorvastatin reduced the paraquat-induced cardiotoxicity via the PPARγ pathway [26]. Hesperidin, a flavanone glycoside and a known PPARγ ligand, improved cardiac hypertrophy by improving cardiac hemodynamics, as well as inhibiting oxidative stress and apoptosis through increasing PPARγ expression [27]. Piperine, a phenolic component of black pepper, attenuated cardiac fibrosis via PPARγ activation and the inhibition of protein kinase B (AKT) / glycogen synthase kinase 3 β (GSK3β) [28]. Interestingly, the regulation of PPARγ by pioglitazone suppressed cardiac hypertrophy as indicated by decreased heart/body weight ratio, wall thickness, and myocyte diameter [29], but the effect of pioglitazone on limiting myocardial infarct size was a PPARγ-independent event [30]. Epoxyeicosatrienoic acids (EET), a primary arachidonic acid metabolite, blocked tumor necrosis factor α (TNFα)-induced cardiotoxicity by reducing inflammation via upregulation of PPARγ expression [31].
Some dual PPARα/γ agonists such as tesaglitazar display an increased risk of cardiovascular events. Treatment with tesaglitazar in mice caused cardiac dysfunction associated with low mitochondrial abundance [32]. In addition, tesaglitazar increased acetylation of proliferator-activated receptor gamma coactivator 1α (PGC1α) and decreased the expression of sirtuin 1 (SIRT1), which was associated with competition between PPARα and PPARγ. LY510929, another dual PPARα/γ agonist, was shown to cause left-ventricular hypertrophy in rats [33]. However, aleglitazar inhibited hyperglycemia-induced cardiomyocyte apoptosis by activation of both PPARα and PPARγ [34,35]. Activation of PPARβ signaling mediated docosahexaenoic acid (DHA), and its metabolites elicited cytotoxicity in H9c2 cells via the de novo formation of ceramide [36]. Doxorubicin (DOX) caused a remarkable decrease in cardiac dP/dT and cardiac output by inhibition of PPARβ expression in rats [37].
PPARβ plays an important role in angiogenesis and cancers. Activation of PPARβ in blood vessels promotes tumor vascularization and the progression of different cancer cell types through direct activation of platelet-derived growth factor receptor beta (PDGFRβ), platelet-derived growth factor subunit B (PDGFB), and the c-Kit [38]. Figure 1 summarizes regulation of PPARs in cardiotoxicity.

3. PPARs in Hepatotoxicity

In the liver, PPARs play indispensable roles in fatty-acid and glucose metabolism, and they supply energy to peripheral tissues. Numerous studies reported that xenobiotic chemicals and environmental contaminants disrupted the normal liver homeostasis by activating PPAR subtypes that are highly expressed in hepatocytes, especially PPARα. Indeed, PPARα was recognized as a target for pollutants, which could interact with the similar nuclear receptors and subsequently induce metabolic disorders.
Phthalates, common plasticizers in nearly all plastic consumer goods, are defined as PPAR modulators [6]. Accumulative studies showed that phthalates activated PPARα and other lipid-activated nuclear receptors in the liver, which induced metabolic disruption and endocrine disorders. The exposure concentration of phthalate metabolites such as DEHP and mono (2-ethylhexyl) phthalate (MEHP) positively correlated with insulin resistance and abdominal obesity in American male adults [39,40,41]. Di-n-butyl-di-(4-chlorobenzohydroxamato) tin (DBDCT), an organotin with high antitumor activity, was also demonstrated to induce notable toxicity in rat liver tissue via the PPAR signaling pathway [42]. DBDCT treatment aroused acute and focal necrosis and Kupffer cell hyperplasia in rat liver. The decreased expression levels of cluster of differentiation 36 (CD36), fatty acid binding protein 4 (FABP4), enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase (EHHADH), acetyl-CoA acyltransferase 1 (ACAA1), phosphoenolpyruvate carboxykinase (PEPCK), PPARα, and PPARγ in DBDCT-treated liver tissue were indicated by proteomics. Furthermore, the toxic effect was alleviated by PPARγ blocking agent T0070907 [42,43]. Additionally, organotins, the major components of agricultural fungicides and pesticides, were documented to exert similar functions as PPARγ and PPARβ ligands, which promote weight gain and increase fat storage by target gene induction in liver [44]. For example, tributyltin chloride (TBT) enhanced adipogenesis and adipocyte differentiation by directly stimulating downstream transcription of PPARγ in liver and adipose tissues. In mouse models, uterus exposure to TBT disrupted hepatic architecture and caused liver steatosis by increasing lipid accumulation and adipocyte maturation [40,45]. Thus, PPARs play an important role in contaminant-induced toxicity.
The hepatotoxicity of PPARα ligands was rarely documented. Few PPARα ligands are proven to be hepatotoxicants. Fenofibrate exerts only a minimal increase of alanine aminotransferase and aspartate aminotransferase [6,46]. In contrast to hepatotoxicity mediated by PPARs, PPAR ligands also display some protective effect against hepatotoxicity. PPARα ligand activation was proven to prevent acute liver toxicity induced by alcohol, carbon tetrachloride (CCl4), acetaminophen, chloroform, thioacetamide, and bromobenzene due to the induction of fatty acid catabolism and anti-inflammatory properties [47,48,49]. PPARα agonists showed a reversal of fatty liver in mice even with continued ethanol consumption [50]. PPARγ agonist troglitazone and rosiglitazone are reported to induce mild liver toxicity in patients that might be PPARγ-independent because of the low expression level of PPARγ in the liver [12]. Despite the hepatotoxicity of PPARγ activation, PPARγ ligand treatment attenuated fibrogenesis by inhibiting the activation of hepatic stellate cells (HSCs) [51,52]. PPARγ ligands exhibited a suppressive effect on the expression of fibrogenic genes including collagen and α-smooth muscle actin. PPARβ activation by L-165041 enhanced the HSC proliferation and fibrogenic gene expression, and it exacerbated CCl4-induced liver fibrotic progression [53]. PPARα, PPARγ, and PPARβ display different roles in hepatotoxicity. Activation of PPARα prevents acute liver toxicity. Activation of PPARγ induces mild liver toxicity but attenuates liver fibrogenesis. Activation of PPARβ promotes the progression of liver fibrosis.
Numerous studies reported that hepatocarcinogenesis was the major toxicity induced by PPARα activation [54,55]. Unmanaged peroxisomal proliferation and hepatomegaly observed in fibrate-treated livers can ultimately lead to hepatocellular carcinoma [56]. The hepatocarcinogenesis by PPARα activation was fully investigated over 30 years. The main target of PPARα is the liver, which induces pleiotropic impacts such as hypertrophy and hyperplasia [57,58]. These unmanaged responses cause hepatocellular carcinomas in rodents. The mechanisms remain elucidated. Some studies propose that PPARα-mediated DNA replication, proliferation, and suppressed apoptosis result in PPARα agonist-induced hepatocarcinogenesis [59]. Actually, the effect of PPARα on hepatocarcinogenesis varies among different species. In human, an increased risk of liver cancer of fibrates is not yet reported. This might be due to no significant peroxisome proliferation induced by hypolipidemic agents [60] and less expression of PPARα in patient livers compared to rodent liver. Although humans show resistance to the adverse effect of PPARα-induced hepatocarcinogenesis, vigilance is still required to develop new agents.

4. PPARs in Gastrointestinal Toxicity

As indicated by emerging evidence, PPARs and their ligands also play an important role in the regulation of immune and inflammatory reactions in the gastrointestinal (GI) system.
In view of modulation of several target genes involved in metabolic processes and immune response in the GI tract, PPARs and their ligands became a research hotspot in gastroenterology [61]. Accumulative evidence showed that inflammatory bowel diseases (IBDs) and colon cancer (CC), two important GI diseases, are related to PPARs and their ligands [62,63]. PPAR agonists might serve as a new effective pharmacotherapy for IBDs and CC. PPARα mediated the anti-inflammatory effect of glucocorticoid (GC) in a chemical-induced colitis mouse model [64]. More recently, it was shown that PPARα activation diminished the therapeutic effects of rSj16 in dextran sulfate sodium (DSS)-induced colitis mice, indicating that the PPARα signaling pathway plays a crucial role in DSS-induced colitis progression [65].
With the high expression in GI tract mucosa, especially in the intestine and colon [66,67,68], PPARγ is closely related to GI injury and inflammatory response. The inflammatory reaction is the common pathological process of many GI diseases and trauma. Once the homeostasis of GI is disrupted by exogenous factors or endogenous metabolites and shifts to the pro-inflammatory state, the pro-inflammatory cytokines such as TNF-α, interleukin 1β (IL-1β), IL-6 are liberated by the hyperactive immune cells. Transcription factor NF-κB is one of the most important regulatory mechanisms of immune and inflammatory responses mediated by PPARs and their ligands in the GI tract. In colon, PPARγ downregulated NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways, which subsequently inhibited the mucosal production of inflammatory cytokines [69]. Furthermore, in intestinal cells, activation of PPARγ resulted in decreased expression of intercellular adhesion molecule 1 (ICAM-1) and TNF-α [70], which are downstream targets of NF-kB [71]. Treatment with troglitazone attenuated colitis induced by intrarectal administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) [69]. PPARγ could function as an endogenous anti-inflammatory pathway in a murine model of intestinal ischemia–reperfusion (I/R) injury. Activation of PPARγ by its agonist BRL-49653 had a protective effect on intestinal acute I/R injury [70]. However, the protective activity of BRL-49653 was abolished in PPARγ-deficient mice. In the investigation of the prevention and treatment of radiation-induced intestinal damage, accumulating evidence supported that the administration of PPARγ agonists alleviated radiation-induced intestinal toxicity. PPARγ agonists were shown to reverse radiation-induced apoptosis and inflammation and to exert radio-protective effects on healthy bowel upon irradiation [72,73]. Further research of acute intestinal injury reported that PPARγ agonist rosiglitazone reduced the expression of the fibrotic marker transforming growth factor β (TGFβ) and phosphorylation of the p65 subunit of NF-kB triggered by pro-inflammatory cytokine TNF-α [72]. In ulcerative colitis research, promoting the nuclear localization of PPARγ weakened the activity of NF-κB signaling in both rectal tissues from dextran sulfate sodium (DSS)-induced mice and lipopolysaccharide (LPS)-stimulated macrophages [74]. PPARs inhibited the expression of macrophage-related inflammatory mediators and macrophage infiltration in the acute irradiation intestinal damage [75]. Compared with wild-type mice, PPARγ-deficient mice showed significantly severe damage after an I/R injury procedure, which indicated the anti-inflammatory and protective role of PPARγ in GI damage [70]. These studies indicated the role of PPARγ in suppression of NF-kB activation and inflammatory response in intestinal tissues.
Additionally, several reports indicated that PPARs and their ligands could lead to carcinogenesis by affecting the metabolism of glucose and lipids. Intestinal PPARα exhibited a protective effect against colon carcinogenesis by inhibiting methylation of P21 and P27 [76]. Human colorectal tumors also show lower levels of PPARα compared to normal tissue [76]. PPARγ synthetic activator rosiglitazone has a radio-sensitizing effect on human bowel cancer cells [72]. PPARγ was reported to be associated with colorectal cancer via insulin and inflammatory mechanisms [77,78]. On the other hand, PPARγ was shown to be expressed in human colonic mucosa and cancer. The ability of PPARγ activation to decrease cyclooxygenase-2 (COX-2) expression and induce apoptosis suggests that the PPARγ pathway might be a tumor suppressor in humans [79]. Another study reports that 8% of primary colorectal tumors harbor function-dead mutations in one allele of the PPARγ gene and emphasizes the potential role of this receptor as a therapeutic target for cancer or in designing a mouse colon cancer model [80]. The treatment of colon cancer by suppressing the methylation of PPARγ promoter and enhancing PPARγ expression is also underway, because the hyper-methylation of promoter regions can induce PPARγ gene silence. Moreover, the risk of radiation-induced intestinal toxicity in methylated patients was also increased compared with un-methylated patients [81]. Furthermore, PPARβ is induced in intestinal stem cells and progenitor cells in high-fat diet-treated mice and enhances stemness and tumorigenicity of intestine [82]. Arachidonic acid derivative prostaglandin E2 (PGE2), which is a biologically active lipid, increases cell survival and improves intestinal adenoma formation by indirectly activating PPARβ via the phosphatidylinositol-3 kinase (PI3K)/Akt signaling pathway [83]. The activation of PPARβ also upregulates COX-2, which is a key activator for colon cancer cells [59].
We summarize the regulatory mechanism of PPARs in gastrointestinal toxicity in Figure 2. A better understanding of the role of PPARs in the GI system will help to develop novel pharmacotherapy against colon carcinogenesis and diminish intestinal toxicity.

5. PPARs in Reproductive and Developmental Toxicity

Three isoforms of PPARs were found in the reproductive system including the hypothalamus, pituitary, testis, ovary, uterus, adrenal, and mammary glands. Numerous studies showed that PPARs play a role in the normal reproductive and developmental functions, and abnormal regulation of PPARs by exposure to endogenous or exogenous compounds might lead to physiological dysfunction in reproductive system [84,85,86]. Thus, the research on reproductive and developmental disorders focuses on PPARs and their modulators.
Triptolide is a major active compound in Chinese herb Tripterygium wilfordii multiglycoside, and it is widely used for treatment of autoimmune diseases and nephrotic syndrome [87]. However, we previously reported that triptolide causes mitochondrial damage and dysregulates fatty-acid metabolism by upregulating expression and nuclear translocation of PPARα in mouse sertoli cells [88]. A metabolomics study revealed that triptolide caused impairment of spermatogenesis accompanied by abnormal lipid and energy metabolism in male mice through downregulation of PPARs [89]. Different concentrations and times of triptolide exposure led to the different behaviors of PPARs. These findings support that PPARs are key mediators in triptolide-induced reproduction toxicity.
Phthalates, which activate PPARs, have a remarkable effect on fertility rates, ovulation, development of the male reproductive tract, spermatogenesis, and teratogenesis [6]. Early exposure to phthalates influenced perinatal and postnatal cardiometabolic programming [90]. DEHP, a phthalate ester, is commonly used in industry as a plasticizer, which activates PPARα to regulate the expression of downstream target genes. DEHP treatment had no remarkable effect on body, liver, and ovary weight in female dams (F0) and offspring (F1) in either wild-type or PPARα-knockout mice. However, it suppressed the expression of ovarian estrogen receptor α, and the repression of ovarian estrogen receptor α expression by DEHP was lost in PPARα-knockout mice [91]. PPARα transcription is related to fertility impairment in female mice exposed to high doses of DEHP (500 mg/kg of body weight per day) [92]. Moreover, it was reported that MEHP, a principle active metabolite of DEHP, decreased the activity and production of aromatase, which converted testosterone to estradiol in ovarian granulosa cells by activating PPARα and PPARγ [93]. Benzo [a]pyrene (B [a]P) is a ubiquitous environmental contaminant, and the combination of B [a]P and DEHP induced ovotoxicity in female rats and suppressed sex hormone secretion via the PPAR-mediated signaling pathway [94].
Dehydroepiandrosterone (3c-hydroxy-5-androsten-17-one, DHEA) is a ligand of PPARα, and it also stimulates the production of PPARα. Some clinical studies showed that dietary supplementation of DHEA reversed the oocyte quality in mice and aged women [95,96]. Additionally, reduced DHEA and loss of function of PPARα result in the decreased follicle quality associated with the changes of fatty-acid metabolism, transport, and mitochondrial function. Perfluorooctanoic acid (PFOA), a synthetic perfluorinated compound (PFC) which is widely distributed, significantly inhibited mammary gland growth in mice through activation of PPARα, and this effect was reversed by supplementation with exogenous estrogen or progesterone [97]. Moreover, perfluorooctane sulfonate (PFOS) is a product of metabolic degradation of PFCs and has an estrogenic activity and endocrine-disruptive properties in the marine medaka embryos, partially through the regulation of PPARs.
Additionally, 15-deoxy-delta12,14-prostaglandin J2 (15dPGJ2), which is converted by arachidonic acid via successive dehydration and isomerization, acts as an endogenous ligand of PPARγ via direct covalent binding, and it plays a key role in lipid homeostasis [98,99]. Kurtz and colleagues found that 15dPGJ2 partially restored the mRNA expression of oxidizing enzymes including acyl-CoA 1 (ACO) and carnitine palmitoyltransferase 1 (CPT1) in the lungs of male fetuses from diabetic rats, but this effect was not observed in female fetuses [100]. Moreover, it was reported that 15dPGJ2 modulated lipid metabolism and nitric oxide production in diabetes-induced placental dysfunction partially through the PPAR pathway [101]. Trichloroethylene (TCE) reduced fertilizability of oocyte and its ability to bind sperm plasma membrane proteins in rats [102]. A systematic evaluation of TCE showed that TCE could cause cardiac defects in humans when the exposure is during a sensitive period of fetal development [103]. Tributyltin chloride (TBT) activates all three types of PPARs. TBT has effects on reproductive function and induces abnormal mammary gland fat accumulation by increasing PPARγ expression [104,105]. TZDs (e.g., pioglitazone, rosiglitazone, and troglitazone) activate PPARγ to regulate the transcription of genes responsible for glucose and lipid metabolism. TZDs clinically sensitize peripheral insulin in patients with type 2 diabetes by regulating glucose and lipid metabolism [106,107]. Oral administration of rosiglitazone 4 mg once a day for three months improves hyperandrogenemia, insulin resistance, lipidemia, C-reactive protein levels, ovarian volume, and follicle number in patients with polycystic ovary syndrome (PCOS) [108]. Rosiglitazone exhibited significant protective effects on metabolic, hormonal, and morphological features of PCOS. Significant changes were also observed in the isovaleryl carnitine levels and lipid oxidation rates after pioglitazone treatment [109]. Rosiglitazone significantly improved oocyte quality in diet-induced obesity (DIO) mice, indicating the positive effect of PPARγ on ovarian function [110]. Rosiglitazone affects steroidogenesis in porcine ovarian follicles by stimulating PPARγ [111,112]. In vivo experiments demonstrated that fenofibrate inhibited ovarian estrogen synthesis [113]. A review concluded that clofibrate and gemfibrozil caused atypical changes in maternal and fetal liver during pregnancy, but there was no direct evidence of developmental toxicity or teratogenicity of clofibrate and gemfibrozil [6]. Irbesartan (IRB) is one of the most widely used angiotensin type 1 (AT1) receptor blockers (ARBs) with PPARγ agonistic activity. Rats treated with IRB showed an increase in estradiol and follicle-stimulating hormone levels, which subsequently ameliorated ovarian dysfunction [114]. These studies indicate that activation of PPARγ signaling protects ovarian function.
Genistein (49,5,7-trihydroxyisoflavone, GEN), a kind of isoflavones derived from soybeans, was investigated for its antioxidant, anticancer, and anti-inflammatory activities [115]. It is a natural ligand of PPARs, and it can improve the development and metabolism of chick embryos through the activation of PPARs [116,117]. Prostacyclin (PGI2) activated its nuclear receptor PPARβ to accelerate blastocyst hatching in mice [118]. These studies suggest that the activation of PPARs is involved in toxicant-induced reproductive toxicity.
The anti-tumor effects of PPAR agonists were documented. Rosiglitazone and troglitazone, both PPARγ activators, showed inhibitory effects on pituitary adenoma cells in mice and human, and they were considered to be a new oral drug for the treatment of pituitary tumors [119]. Moreover, troglitazone treatment stabilized the prostate-specific antigen levels in patients with advanced prostate cancer clinically by upregulating E-cadherin and glutathione peroxidase 3 [120]. Rosiglitazone showed an inhibitory effect on proliferation of primary human prostate cancer cells [121]. However, the activation of PPARβ by selective agonist GW501516 was reported to stimulate proliferation of human breast and prostate cancer cells which are responsive to sexual hormones [122]. PPARβ activation by GW501516 increased cyclin-dependent kinase 2 (CDK2) and vascular endothelial growth factor α (VEGFα) expression, indicating the improved cell proliferation and angiogenesis. This study suggested the possibility of PPARβ antagonists in treating breast and prostate cancer.

6. Other Systemic Toxicity and Protective Effects Mediated by PPARs

Fibrates, PPARα synthetic ligands, were developed for treatment of hyperlipidemia in the clinic, such as fenofibrate, bezafibrate, ciprofibrate, and so on [123,124,125]. However, muscle weakness, muscle pain, and even rhabdomyolysis were observed during their application [6]. Different fibrates lead to different degrees of myopathy, and that might be due to different mechanisms. The underlying mechanism is still unclear. Some studies reported that PPARα activation in skeletal muscle transactivated the genes encoding muscle proteases, and the increased expression of skeletal muscle proteases led to severe myopathy [126,127]. The muscle toxicity might result from the blood concentration of the drug, because remarkably higher incidence occurs in patients with kidney failure or hypoalbuminemia [128]. Moreover, Motojma et al. proposed that the increase in pyruvate dehydrokinase isoenzyme4 (PDK4) and the decrease in serum triglyceride (TG) level mediated by PPARα in skeletal muscle caused the degradation of protein in muscle, ultimately resulting in myopathy and even rhabdomyolysis [129]. Due to the low incidence of rhabdomyolysis, no drug was withdrawn from the market because of the muscular toxicity.
In contrast to the adverse effect mediated by PPARs, PPARs also exert protective effects against nephrotoxicity and neuron injury.
Diabetic kidney disease is one complication of type 2 diabetes. PPARα and PPARγ are famous targets for treating diabetes, especially PPARγ. Increasing studies indicated that PPARs play important roles in kidney physiology and pathology. In most cases, PPARγ serves as a therapeutic target for treating nephrotoxicity. PPARγ-null mice showed spontaneous diabetic nephropathy. PPARγ knockout mice exhibited kidney hypertrophy accompanied by increased glucosuria, albuminuria, renal fibrosis, and mesangial expansion [130,131].
PPARs also play key roles in regulating brain self-repair. Central nervous system diseases, neuron injury, and cell death are closely related to neuroinflammation [132,133]. Lovastatin (LOV) can protect vulnerable oligodendrocytes in a mouse model of multiple sclerosis (MS) by inhibiting guanosine triphosphate (GTP)-binding proteins, small Rho GTPases, via a PPARα-dependent mechanism [134]. Healthy oligodendrocytes are essential for the synaptic survival of MS neurons. PPARα activation increases the seizure threshold and controls the seizure frequency [134]. The high expression of PPARα in the brain region also prevents nicotine-induced neuronal damage by regulating tyrosine kinases and phosphokinases in neuronal current. It decreases the frequency of seizures caused by the activation of nicotine receptors in vertebral neurons [135]. Animal model studies showed that fenofibrate prevented convulsions caused by dysregulation of neurotransmitters [136]. Substantia nigra has high-density microglia which show two polarization states, M1 and M2, which have pro-inflammatory or anti-inflammatory effects, respectively [132,137]. Therefore, inhibiting the activation of M1 microglia and promoting the activation of M2 microglia are beneficial to central system diseases. In the condition of inflammation, M1 microglia are activated and release pro-inflammatory factors and neurotoxic substances, such as cytokines, reactive oxygen species, prostaglandins, and complements, which aggravate inflammatory injury [138]. Recent studies showed that PPARs (mainly PPARγ) regulate microglia-mediated inflammation in Parkinson’s disease (PD) and other neurodegenerative diseases [138,139,140]. Pioglitazone, a PPARγ ligand, was shown to inhibit the activation and secretion of glial cells by activating PPARγ [141]. Pioglitazone also inhibits the degeneration of dopamine neurons, which induces inflammation and promotes neuron death [141]. Rosiglitazone has a protective effect on neurotoxin 1-methy-4-phenyl-1,2,3,4,6-tetrahydropyidine (MPTP)-induced PD mouse model via upregulation of M2 phenotypic-related anti-inflammatory factors and the downregulation of M1 phenotypic-related pro-inflammatory factors [142]. A recent study found that PPARα/γ dual agonist MHY908 protects dopamine neurons from MPTP-induced loss in PD mice by reducing neuroinflammation and microglia activation [141]. Moreover, L-165041, a PPARβ agonist, can inhibit the radiation-induced inflammation in microglia by inhibition of the NF-kB signaling pathway [143]. At present, Alzheimer’s disease is also considered to be a neuroinflammatory disease and is characterized by abnormal accumulation of β-amyloid (Aβ). Under the condition of Aβ accumulation, M1 microglia were activated, resulting in neuronal injury and apoptosis [144]. It was shown that adiponectin can activate M2 microglia and enhance the clearance of Aβ by activating the PPARγ signaling pathway.

7. Conclusions

Better understanding of the role of PPARs in toxicology and pharmacology and the underlying molecular basis is necessary for PPARs-related clinical drug discovery and development. Unfortunately, there are limited studies reviewing the integrated network of relationships in these aspects. Lots of PPAR ligands have beneficial effects on applied pharmacology, but they are also accompanied by various toxicities. Here, we mainly summarized the regulation of PPARs in toxicology and protection against toxicity in various systems, such as cardiotoxicity, hepatotoxicity, gastrointestinal toxicity, and reproductive and developmental toxicity (Figure 3). We hope that a comprehensive understanding of PPAR-mediated toxicology and applied pharmacology will contribute to the safety of PPAR-targeted therapies in the future.

Author Contributions

Conceptualization, P.X. and Y.X.; writing—original draft preparation, Y.X., P.X., Y.Z., S.Z. and Y.L.; writing—review and editing, Y.X., P.X. and Z.H.; supervision, P.X. and Z.H.; project administration, P.X. and Z.H.; funding acquisition, Z.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (Grant No. 81773992), the General Projects in the Natural Science Foundation of Guangdong Province (Grant No. 2018A0303130170), and the National Engineering and Technology Research Center for New drug Druggability Evaluation (Seed Program of Guangdong Province, 2017B090903004). All support is gratefully acknowledged.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Grygiel-Gorniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications—A review. Nutr. J. 2014, 13, 17. [Google Scholar] [CrossRef] [Green Version]
  2. Lagana, A.S.; Vitale, S.G.; Nigro, A.; Sofo, V.; Salmeri, F.M.; Rossetti, P.; Rapisarda, A.M.; La Vignera, S.; Condorelli, R.A.; Rizzo, G.; et al. Pleiotropic Actions of Peroxisome Proliferator-Activated Receptors (PPARs) in Dysregulated Metabolic Homeostasis, Inflammation and Cancer: Current Evidence and Future Perspectives. Int. J. Mol. Sci. 2016, 17, 999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hong, F.; Xu, P.; Zhai, Y. The Opportunities and Challenges of Peroxisome Proliferator-Activated Receptors Ligands in Clinical Drug Discovery and Development. Int. J. Mol. Sci. 2018, 19, 2189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Berger, J.; Wagner, J.A. Physiological and therapeutic roles of peroxisome proliferator-activated receptors. Diabetes Technol. Ther. 2002, 4, 163–174. [Google Scholar] [CrossRef] [PubMed]
  5. Amber-Vitos, O.; Chaturvedi, N.; Nachliel, E.; Gutman, M.; Tsfadia, Y. The effect of regulating molecules on the structure of the PPAR-RXR complex. Biochim. Biophys. Acta 2016, 1861, 1852–1863. [Google Scholar] [CrossRef]
  6. Peraza, M.A.; Burdick, A.D.; Marin, H.E.; Gonzalez, F.J.; Peters, J.M. The toxicology of ligands for peroxisome proliferator-activated receptors (PPAR). Toxicol. Sci. 2006, 90, 269–295. [Google Scholar] [CrossRef] [Green Version]
  7. Corrales, P.; Vidal-Puig, A.; Medina-Gomez, G. PPARs and Metabolic Disorders Associated with Challenged Adipose Tissue Plasticity. Int. J. Mol. Sci. 2018, 19, 2124. [Google Scholar] [CrossRef] [Green Version]
  8. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef]
  9. Hong, F.; Pan, S.; Guo, Y.; Xu, P.; Zhai, Y. PPARs as Nuclear Receptors for Nutrient and Energy Metabolism. Molecules 2019, 24, 2545. [Google Scholar] [CrossRef] [Green Version]
  10. Boeckmans, J.; Natale, A.; Rombaut, M.; Buyl, K.; Rogiers, V.; De Kock, J.; Vanhaecke, T.; Rodrigues, M.R. Anti-NASH Drug Development Hitches a Lift on PPAR Agonism. Cells 2019, 9, 37. [Google Scholar] [CrossRef] [Green Version]
  11. Nakamura, T.; Funahashi, T.; Yamashita, S.; Nishida, M.; Nishida, Y.; Takahashi, M.; Hotta, K.; Kuriyama, H.; Kihara, S.; Ohuchi, N.; et al. Thiazolidinedione derivative improves fat distribution and multiple risk factors in subjects with visceral fat accumulation--double-blind placebo-controlled trial. Diabetes Res. Clin. Pract. 2001, 54, 181–190. [Google Scholar] [CrossRef]
  12. Watkins, P.B.; Whitcomb, R.W. Hepatic dysfunction associated with troglitazone. N. Engl. J. Med. 1998, 338, 916–917. [Google Scholar] [CrossRef] [PubMed]
  13. Rubenstrunk, A.; Hanf, R.; Hum, D.W.; Fruchart, J.C.; Staels, B. Safety issues and prospects for future generations of PPAR modulators. Biochim. Biophys. Acta 2007, 1771, 1065–1081. [Google Scholar] [CrossRef] [PubMed]
  14. Fang, C.; Wu, X.; Huang, Q.; Liao, Y.; Liu, L.; Qiu, L.; Shen, H.; Dong, S. PFOS elicits transcriptional responses of the ER, AHR and PPAR pathways in Oryzias melastigma in a stage-specific manner. Aquat. Toxicol. 2012, 106, 9–19. [Google Scholar] [CrossRef]
  15. Huang, Q.; Fang, C.; Wu, X.; Fan, J.; Dong, S. Perfluorooctane sulfonate impairs the cardiac development of a marine medaka (Oryzias melastigma). Aquat. Toxicol. 2011, 105, 71–77. [Google Scholar] [CrossRef]
  16. Zhao, M.; Jiang, Q.; Geng, M.; Zhu, L.; Xia, Y.; Khanal, A.; Wang, C. The role of PPAR alpha in perfluorooctanoic acid induced developmental cardiotoxicity and l-carnitine mediated protection-Results of in ovo gene silencing. Environ. Toxicol. Pharmacol. 2017, 56, 136–144. [Google Scholar] [CrossRef]
  17. Posnack, N.G.; Swift, L.M.; Kay, M.W.; Lee, N.H.; Sarvazyan, N. Phthalate exposure changes the metabolic profile of cardiac muscle cells. Environ. Health Perspect. 2012, 120, 1243–1251. [Google Scholar] [CrossRef]
  18. Xie, W.; Zhang, W.; Ren, J.; Li, W.; Zhou, L.; Cui, Y.; Chen, H.; Yu, W.; Zhuang, X.; Zhang, Z.; et al. Metabonomics Indicates Inhibition of Fatty Acid Synthesis, beta-Oxidation, and Tricarboxylic Acid Cycle in Triclocarban-Induced Cardiac Metabolic Alterations in Male Mice. J. Agric. Food Chem. 2018, 66, 1533–1542. [Google Scholar] [CrossRef]
  19. Ebenstein, A.; Fan, M.; Greenstone, M.; He, G.; Zhou, M. New evidence on the impact of sustained exposure to air pollution on life expectancy from China’s Huai River Policy. Proc. Natl. Acad. Sci. USA 2017, 114, 10384–10389. [Google Scholar] [CrossRef] [Green Version]
  20. Jin, X.; Xue, B.; Ahmed, R.Z.; Ding, G.; Li, Z. Fine particles cause the abnormality of cardiac ATP levels via PPARa-mediated utilization of fatty acid and glucose using in vivo and in vitro models. Environ. Pollut. 2019, 249, 286–294. [Google Scholar] [CrossRef]
  21. Luptak, I.; Balschi, J.A.; Xing, Y.; Leone, T.C.; Kelly, D.P.; Tian, R. Decreased contractile and metabolic reserve in peroxisome proliferator-activated receptor-alpha-null hearts can be rescued by increasing glucose transport and utilization. Circulation 2005, 112, 2339–2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Duan, S.Z.; Ivashchenko, C.Y.; Russell, M.W.; Milstone, D.S.; Mortensen, R.M. Cardiomyocyte-specific knockout and agonist of peroxisome proliferator-activated receptor-γ both induce cardiac hypertrophy in mice. Circ. Res. 2005, 97, 372–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Yang, H.I.; Kim, W.S.; Kim, D.H.; Kang, J.S. Histopathological Evaluation of Heart Toxicity of a Novel Selective PPAR-γ Agonists CKD-501 in db/db Mice. Biomol. Ther. 2013, 21, 84–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. He, H.; Tao, H.; Xiong, H.; Duan, S.Z.; McGowan, F.X., Jr.; Mortensen, R.M.; Balschi, J.A. Rosiglitazone causes cardiotoxicity via peroxisome proliferator-activated receptor gamma-independent mitochondrial oxidative stress in mouse hearts. Toxicol. Sci. 2014, 138, 468–481. [Google Scholar] [CrossRef] [Green Version]
  25. Planavila, A.; Laguna, J.C.; Vazquez-Carrera, M. Atorvastatin improves peroxisome proliferator-activated receptor signaling in cardiac hypertrophy by preventing nuclear factor-kappa B activation. Biochim. Biophys. Acta 2005, 1687, 76–83. [Google Scholar] [CrossRef]
  26. Malekinejad, M.; Masoumi Verki, M.; Khoramjouy, M.; Alenabi, A.; Hallaj-Salahipour, M.; Malekinejad, H. Cardioprotective Effects of Atorvastatin Are Mediated Through PPARgamma in Paraquat-Exposed Rats. J. Cardiovasc. Pharmacol. 2019, 74, 400–408. [Google Scholar] [CrossRef]
  27. Bhargava, P.; Verma, V.K.; Malik, S.; Khan, S.I.; Bhatia, J.; Arya, D.S. Hesperidin regresses cardiac hypertrophy by virtue of PPAR-γ agonistic, anti-inflammatory, antiapoptotic, and antioxidant properties. J. Biochem. Mol. Toxicol. 2019, 33, e22283. [Google Scholar] [CrossRef]
  28. Ma, Z.G.; Yuan, Y.P.; Zhang, X.; Xu, S.C.; Wang, S.S.; Tang, Q.Z. Piperine Attenuates Pathological Cardiac Fibrosis via PPAR-γ/AKT Pathways. EBioMedicine 2017, 18, 179–187. [Google Scholar] [CrossRef] [Green Version]
  29. Asakawa, M.; Takano, H.; Nagai, T.; Uozumi, H.; Hasegawa, H.; Kubota, N.; Saito, T.; Masuda, Y.; Kadowaki, T.; Komuro, I. Peroxisome proliferator-activated receptor gamma plays a critical role in inhibition of cardiac hypertrophy in vitro and in vivo. Circulation 2002, 105, 1240–1246. [Google Scholar] [CrossRef] [Green Version]
  30. Birnbaum, Y.; Long, B.; Qian, J.; Perez-Polo, J.R.; Ye, Y. Pioglitazone limits myocardial infarct size, activates Akt, and upregulates cPLA2 and COX-2 in a PPAR-γ-independent manner. Basic Res. Cardiol. 2011, 106, 431–446. [Google Scholar] [CrossRef]
  31. Zhao, G.; Wang, J.; Xu, X.; Jing, Y.; Tu, L.; Li, X.; Chen, C.; Cianflone, K.; Wang, P.; Dackor, R.T.; et al. Epoxyeicosatrienoic acids protect rat hearts against tumor necrosis factor-alpha-induced injury. J. Lipid Res. 2012, 53, 456–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kalliora, C.; Kyriazis, I.D.; Oka, S.I.; Lieu, M.J.; Yue, Y.; Area-Gomez, E.; Pol, C.J.; Tian, Y.; Mizushima, W.; Chin, A.; et al. Dual peroxisome-proliferator-activated-receptor-alpha/gamma activation inhibits SIRT1-PGC1alpha axis and causes cardiac dysfunction. JCI Insight 2019, 5, 129556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Engle, S.K.; Solter, P.F.; Credille, K.M.; Bull, C.M.; Adams, S.; Berna, M.J.; Schultze, A.E.; Rothstein, E.C.; Cockman, M.D.; Pritt, M.L.; et al. Detection of left ventricular hypertrophy in rats administered a peroxisome proliferator-activated receptor alpha/gamma dual agonist using natriuretic peptides and imaging. Toxicol. Sci. 2010, 114, 183–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Chen, Y.; Chen, H.; Birnbaum, Y.; Nanhwan, M.K.; Bajaj, M.; Ye, Y.; Qian, J. Aleglitazar, a dual peroxisome proliferator-activated receptor-alpha and -gamma agonist, protects cardiomyocytes against the adverse effects of hyperglycaemia. Diab. Vasc. Dis. Res. 2017, 14, 152–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Khuchua, Z.; Glukhov, A.I.; Strauss, A.W.; Javadov, S. Elucidating the Beneficial Role of PPAR Agonists in Cardiac Diseases. Int. J. Mol. Sci. 2018, 19, 3464. [Google Scholar] [CrossRef] [Green Version]
  36. Samokhvalov, V.; Zlobine, I.; Jamieson, K.L.; Jurasz, P.; Chen, C.; Lee, K.S.; Hammock, B.D.; Seubert, J.M. PPARdelta signaling mediates the cytotoxicity of DHA in H9c2 cells. Toxicol. Lett. 2015, 232, 10–20. [Google Scholar] [CrossRef] [Green Version]
  37. Chen, Z.C.; Chen, L.J.; Cheng, J.T. Doxorubicin-Induced Cardiac Toxicity Is Mediated by Lowering of Peroxisome Proliferator-Activated Receptor delta Expression in Rats. PPAR Res. 2013, 2013, 456042. [Google Scholar] [CrossRef] [Green Version]
  38. Wagner, K.D.; Du, S.; Martin, L.; Leccia, N.; Michiels, J.F.; Wagner, N. Vascular PPARbeta/delta Promotes Tumor Angiogenesis and Progression. Cells 2019, 8, 1623. [Google Scholar] [CrossRef] [Green Version]
  39. Ito, Y.; Nakajima, T. PPARalpha- and DEHP-Induced Cancers. PPAR Res. 2008, 2008, 759716. [Google Scholar] [CrossRef] [Green Version]
  40. Casals-Casas, C.; Feige, J.N.; Desvergne, B. Interference of pollutants with PPARs: Endocrine disruption meets metabolism. Int. J. Obes. (Lond.) 2008, 32, S53–S61. [Google Scholar] [CrossRef] [Green Version]
  41. Stahlhut, R.W.; van Wijngaarden, E.; Dye, T.D.; Cook, S.; Swan, S.H. Concentrations of urinary phthalate metabolites are associated with increased waist circumference and insuslin resistance in adult U.S. males. Environ. Health Perspect. 2007, 115, 876–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Li, Y.; Liu, X.; Niu, L.; Li, Q. Proteomics Analysis Reveals an Important Role for the PPAR Signaling Pathway in DBDCT-Induced Hepatotoxicity Mechanisms. Molecules 2017, 22, 1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Qiao, X.; Li, Y.; Mai, J.; Ji, X.; Li, Q. Effect of Dibutyltin Dilaurate on Triglyceride Metabolism through the Inhibition of the mTOR Pathway in Human HL7702 Liver Cells. Molecules 2018, 23, 1654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Upham, J.; Acott, P.D.; O’Regan, P.; Sinal, C.J.; Crocker, J.F.; Geldenhuys, L.; Murphy, M.G. The pesticide adjuvant, Toximul, alters hepatic metabolism through effects on downstream targets of PPARalpha. Biochim. Biophys. Acta 2007, 1772, 1057–1064. [Google Scholar] [CrossRef] [Green Version]
  45. Grun, F.; Watanabe, H.; Zamanian, Z.; Maeda, L.; Arima, K.; Cubacha, R.; Gardiner, D.M.; Kanno, J.; Iguchi, T.; Blumberg, B. Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates. Mol. Endocrinol. 2006, 20, 2141–2155. [Google Scholar] [CrossRef]
  46. Jeng, L.B.; Velmurugan, B.K.; Hsu, H.H.; Wen, S.Y.; Shen, C.Y.; Lin, C.H.; Lin, Y.M.; Chen, R.J.; Kuo, W.W.; Huang, C.Y. Fenofibrate induced PPAR alpha expression was attenuated by oestrogen receptor alpha overexpression in Hep3B cells. Environ. Toxicol. 2018, 33, 234–247. [Google Scholar] [CrossRef]
  47. Nakajima, T.; Kamijo, Y.; Tanaka, N.; Sugiyama, E.; Tanaka, E.; Kiyosawa, K.; Fukushima, Y.; Peters, J.M.; Gonzalez, F.J.; Aoyama, T. Peroxisome proliferator-activated receptor alpha protects against alcohol-induced liver damage. Hepatology 2004, 40, 972–980. [Google Scholar] [CrossRef]
  48. Anderson, S.P.; Howroyd, P.; Liu, J.; Qian, X.; Bahnemann, R.; Swanson, C.; Kwak, M.K.; Kensler, T.W.; Corton, J.C. The transcriptional response to a peroxisome proliferator-activated receptor alpha agonist includes increased expression of proteome maintenance genes. J. Biol. Chem. 2004, 279, 52390–52398. [Google Scholar] [CrossRef] [Green Version]
  49. Shankar, K.; Vaidya, V.S.; Corton, J.C.; Bucci, T.J.; Liu, J.; Waalkes, M.P.; Mehendale, H.M. Activation of PPAR-alpha in streptozotocin-induced diabetes is essential for resistance against acetaminophen toxicity. FASEB J. 2003, 17, 1748–1750. [Google Scholar] [CrossRef]
  50. You, M.; Crabb, D.W. Recent advances in alcoholic liver disease II. Minireview: Molecular mechanisms of alcoholic fatty liver. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 287, G1–G6. [Google Scholar] [CrossRef] [Green Version]
  51. Kon, K.; Ikejima, K.; Hirose, M.; Yoshikawa, M.; Enomoto, N.; Kitamura, T.; Takei, Y.; Sato, N. Pioglitazone prevents early-phase hepatic fibrogenesis caused by carbon tetrachloride. Biochem. Bioph. Res. Commun. 2002, 291, 55–61. [Google Scholar] [CrossRef] [PubMed]
  52. Choi, J.H.; Kim, S.M.; Lee, G.H.; Jin, S.W.; Lee, H.S.; Chung, Y.C.; Jeong, H.G. Platyconic Acid A, Platycodi Radix-Derived Saponin, Suppresses TGF-1-induced Activation of Hepatic Stellate Cells via Blocking SMAD and Activating the PPAR Signaling Pathway. Cells 2019, 8, 1544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hellemans, K.; Michalik, L.; Dittie, A.; Knorr, A.; Rombouts, K.; De Jong, J.; Heirman, C.; Quartier, E.; Schuit, F.; Wahli, W.; et al. Peroxisome proliferator-activated receptor-beta signaling contributes to enhanced proliferation of hepatic stellate cells. Gastroenterology 2003, 124, 184–201. [Google Scholar] [CrossRef] [PubMed]
  54. Hays, T.; Rusyn, I.; Burns, A.M.; Kennett, M.J.; Ward, J.M.; Gonzalez, F.J.; Peters, J.M. Role of peroxisome proliferator-activated receptor-alpha (PPARalpha) in bezafibrate-induced hepatocarcinogenesis and cholestasis. Carcinogenesis 2005, 26, 219–227. [Google Scholar] [CrossRef] [Green Version]
  55. Corton, J.C.; Peters, J.M.; Klaunig, J.E. The PPARalpha-dependent rodent liver tumor response is not relevant to humans: Addressing misconceptions. Arch. Toxicol. 2018, 92, 83–119. [Google Scholar] [CrossRef] [Green Version]
  56. Reddy, J.K.; Chu, R. Peroxisome proliferator-induced pleiotropic responses: Pursuit of a phenomenon. Ann. N. Y. Acad. Sci. 1996, 804, 176–201. [Google Scholar] [CrossRef]
  57. Werle-Schneider, G.; Wolfelschneider, A.; von Brevern, M.C.; Scheel, J.; Storck, T.; Muller, D.; Glockner, R.; Bartsch, H.; Bartelmann, M. Gene expression profiles in rat liver slices exposed to hepatocarcinogenic enzyme inducers, peroxisome proliferators, and 17alpha-ethinylestradiol. Int. J. Toxicol. 2006, 25, 379–395. [Google Scholar] [CrossRef]
  58. Corton, J.C.; Lapinskas, P.J.; Gonzalez, F.J. Central role of PPARalpha in the mechanism of action of hepatocarcinogenic peroxisome proliferators. Mutat. Res. 2000, 448, 139–151. [Google Scholar] [CrossRef]
  59. Michalik, L.; Desvergne, B.; Wahli, W. Peroxisome-proliferator-activated receptors and cancers: Complex stories. Nat. Rev. Cancer 2004, 4, 61–70. [Google Scholar] [CrossRef]
  60. Boitier, E.; Gautier, J.C.; Roberts, R. Advances in understanding the regulation of apoptosis and mitosis by peroxisome-proliferator activated receptors in pre-clinical models: Relevance for human health and disease. Comp. Hepatol. 2003, 2, 3. [Google Scholar] [CrossRef]
  61. Clark, R.B.; Bishop-Bailey, D.; Estrada-Hernandez, T.; Hla, T.; Puddington, L.; Padula, S.J. The nuclear receptor PPAR gamma and immunoregulation: PPAR gamma mediates inhibition of helper T cell responses. J. Immunol. 2000, 164, 1364–1371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Mirza, A.Z.; Althagafi, I.I.; Shamshad, H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur. J. Med. Chem. 2019, 166, 502–513. [Google Scholar] [CrossRef] [PubMed]
  63. Kong, R.; Luo, H.; Wang, N.; Li, J.; Xu, S.; Chen, K.; Feng, J.; Wu, L.; Li, S.; Liu, T.; et al. Portulaca Extract Attenuates Development of Dextran Sulfate Sodium Induced Colitis in Mice through Activation of PPARgamma. PPAR Res. 2018, 2018, 6079101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Riccardi, L.; Mazzon, E.; Bruscoli, S.; Esposito, E.; Crisafulli, C.; Di Paola, R.; Caminiti, R.; Riccardi, C.; Cuzzocrea, S. Peroxisome proliferator-activated receptor-alpha modulates the anti-inflammatory effect of glucocorticoids in a model of inflammatory bowel disease in mice. Shock 2009, 31, 308–316. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, L.; Xie, H.; Xu, L.; Liao, Q.; Wan, S.; Yu, Z.; Lin, D.; Zhang, B.; Lv, Z.; Wu, Z.; et al. rSj16 Protects against DSS-Induced Colitis by Inhibiting the PPAR-alpha Signaling Pathway. Theranostics 2017, 7, 3446–3460. [Google Scholar] [CrossRef] [PubMed]
  66. Auboeuf, D.; Rieusset, J.; Fajas, L.; Vallier, P.; Frering, V.; Riou, J.P.; 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-alpha in humans: No alteration in adipose tissue of obese and NIDDM patients. Diabetes 1997, 46, 1319–1327. [Google Scholar] [CrossRef]
  67. Zieleniak, A.; Wojcik, M.; Wozniak, L.A. Structure and physiological functions of the human peroxisome proliferator-activated receptor gamma. Arch. Immunol. Ther. Exp. 2008, 56, 331–345. [Google Scholar] [CrossRef]
  68. Da Silva, S.; Keita, A.V.; Mohlin, S.; Pahlman, S.; Theodorou, V.; Pahlman, I.; Mattson, J.P.; Soderholm, J.D. A Novel Topical PPARgamma Agonist Induces PPARgamma Activity in Ulcerative Colitis Mucosa and Prevents and Reverses Inflammation in Induced Colitis Models. Inflamm. Bowel Dis. 2018, 24, 792–805. [Google Scholar] [CrossRef]
  69. Desreumaux, P.; Dubuquoy, L.; Nutten, S.; Peuchmaur, M.; Englaro, W.; Schoonjans, K.; Derijard, B.; Desvergne, B.; Wahli, W.; Chambon, P.; et al. Attenuation of colon inflammation through activators of the retinoid X receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma) heterodimer. A basis for new therapeutic strategies. J. Exp. Med. 2001, 193, 827–838. [Google Scholar] [CrossRef] [Green Version]
  70. Nakajima, A.; Wada, K.; Miki, H.; Kubota, N.; Nakajima, N.; Terauchi, Y.; Ohnishi, S.; Saubermann, L.J.; Kadowaki, T.; Blumberg, R.S.; et al. Endogenous PPAR gamma mediates anti-inflammatory activity in murine ischemia-reperfusion injury. Gastroenterology 2001, 120, 460–469. [Google Scholar] [CrossRef]
  71. Barnes, P.J.; Karin, M. Nuclear factor-kappaB: A pivotal transcription factor in chronic inflammatory diseases. N. Engl. J. Med. 1997, 336, 1066–1071. [Google Scholar] [CrossRef] [PubMed]
  72. Mangoni, M.; Sottili, M.; Gerini, C.; Desideri, I.; Bastida, C.; Pallotta, S.; Castiglione, F.; Bonomo, P.; Meattini, I.; Greto, D.; et al. A PPAR-γ agonist protects from radiation-induced intestinal toxicity. United Eur. Gastroenterol J. 2017, 5, 218–226. [Google Scholar] [CrossRef] [PubMed]
  73. Linard, C.; Souidi, M. PPARs in Irradiation-Induced Gastrointestinal Toxicity. PPAR Res. 2010, 2010, 528327. [Google Scholar] [CrossRef] [PubMed]
  74. Tian, X.; Peng, Z.; Luo, S.; Zhang, S.; Li, B.; Zhou, C.; Fan, H. Aesculin protects against DSS-Induced colitis though activating PPARgamma and inhibiting NF-small ka, CyrillicB pathway. Eur. J. Pharmacol. 2019, 857, 172453. [Google Scholar] [CrossRef] [PubMed]
  75. Linard, C.; Gremy, O.; Benderitter, M. Reduction of peroxisome proliferation-activated receptor gamma expression by gamma-irradiation as a mechanism contributing to inflammatory response in rat colon: Modulation by the 5-aminosalicylic acid agonist. J. Pharmacol. Exp. Ther. 2008, 324, 911–920. [Google Scholar] [CrossRef] [PubMed]
  76. Luo, Y.; Xie, C.; Brocker, C.N.; Fan, J.; Wu, X.; Feng, L.; Wang, Q.; Zhao, J.; Lu, D.; Tandon, M.; et al. Intestinal PPARalpha Protects Against Colon Carcinogenesis via Regulation of Methyltransferases DNMT1 and PRMT6. Gastroenterology 2019, 157, 744–759 e4. [Google Scholar] [CrossRef] [PubMed]
  77. Slattery, M.L.; Curtin, K.; Wolff, R.; Ma, K.N.; Sweeney, C.; Murtaugh, M.; Potter, J.D.; Levin, T.R.; Samowitz, W. PPARgamma and colon and rectal cancer: Associations with specific tumor mutations, aspirin, ibuprofen and insulin-related genes (United States). Cancer Causes Control. 2006, 17, 239–249. [Google Scholar] [CrossRef]
  78. Sabatino, L.; Ziccardi, P.; Cerchia, C.; Muccillo, L.; Piemontese, L.; Loiodice, F.; Colantuoni, V.; Lupo, A.; Lavecchia, A. Chiral phenoxyacetic acid analogues inhibit colon cancer cell proliferation acting as PPARgamma partial agonists. Sci. Rep. 2019, 9, 5434. [Google Scholar] [CrossRef] [Green Version]
  79. Yang, W.L.; Frucht, H. Activation of the PPAR pathway induces apoptosis and COX-2 inhibition in HT-29 human colon cancer cells. Carcinogenesis 2001, 22, 1379–1383. [Google Scholar] [CrossRef] [Green Version]
  80. Sarraf, P.; Mueller, E.; Jones, D.; King, F.J.; DeAngelo, D.J.; Partridge, J.B.; Holden, S.A.; Chen, L.B.; Singer, S.; Fletcher, C.; et al. Differentiation and reversal of malignant changes in colon cancer through PPARgamma. Nat. Med. 1998, 4, 1046–1052. [Google Scholar] [CrossRef]
  81. Lian, B.; Ren, Y.; Zhang, H.; Lin, T.; Wang, Y. An adenosine derivative (IFC-305) reduced the risk of radiation-induced intestinal toxicity in the treatment of colon cancer by suppressing the methylation of PPAR-r promoter. Biomed. Pharmacother. 2019, 118, 109202. [Google Scholar] [CrossRef] [PubMed]
  82. Beyaz, S.; Mana, M.D.; Roper, J.; Kedrin, D.; Saadatpour, A.; Hong, S.J.; Bauer-Rowe, K.E.; Xifaras, M.E.; Akkad, A.; Arias, E.; et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 2016, 531, 53–58. [Google Scholar] [CrossRef] [PubMed]
  83. You, M.; Yuan, S.; Shi, J.; Hou, Y. PPARdelta signaling regulates colorectal cancer. Curr. Pharm. Des. 2015, 21, 2956–2959. [Google Scholar] [CrossRef] [PubMed]
  84. Vitti, M.; Di Emidio, G.; Di Carlo, M.; Carta, G.; Antonosante, A.; Artini, P.G.; Cimini, A.; Tatone, C.; Benedetti, E. Peroxisome Proliferator-Activated Receptors in Female Reproduction and Fertility. PPAR Res. 2016, 2016, 4612306. [Google Scholar] [CrossRef]
  85. Froment, P.; Gizard, F.; Defever, D.; Staels, B.; Dupont, J.; Monget, P. Peroxisome proliferator-activated receptors in reproductive tissues: From gametogenesis to parturition. J. Endocrinol. 2006, 189, 199–209. [Google Scholar] [CrossRef] [Green Version]
  86. Huang, Q.; Chen, Q. Mediating Roles of PPARs in the Effects of Environmental Chemicals on Sex Steroids. PPAR Res. 2017, 2017, 3203161. [Google Scholar] [CrossRef]
  87. Ziaei, S.; Halaby, R. Immunosuppressive, anti-inflammatory and anti-cancer properties of triptolide: A mini review. Avicenna J. Phytomed. 2016, 6, 149–164. [Google Scholar]
  88. Cheng, Y.; Chen, G.; Wang, L.; Kong, J.; Pan, J.; Xi, Y.; Shen, F.; Huang, Z. Triptolide-induced mitochondrial damage dysregulates fatty acid metabolism in mouse sertoli cells. Toxicol. Lett. 2018, 292, 136–150. [Google Scholar] [CrossRef]
  89. Ma, B.; Qi, H.; Li, J.; Xu, H.; Chi, B.; Zhu, J.; Yu, L.; An, G.; Zhang, Q. Triptolide disrupts fatty acids and peroxisome proliferator-activated receptor (PPAR) levels in male mice testes followed by testicular injury: A GC-MS based metabolomics study. Toxicology 2015, 336, 84–95. [Google Scholar] [CrossRef]
  90. Philips, E.M.; Jaddoe, V.W.V.; Trasande, L. Effects of early exposure to phthalates and bisphenols on cardiometabolic outcomes in pregnancy and childhood. Reprod. Toxicol. 2017, 68, 105–118. [Google Scholar] [CrossRef] [Green Version]
  91. Kawano, M.; Qin, X.Y.; Yoshida, M.; Fukuda, T.; Nansai, H.; Hayashi, Y.; Nakajima, T.; Sone, H. Peroxisome proliferator-activated receptor alpha mediates di-(2-ethylhexyl) phthalate transgenerational repression of ovarian Esr1 expression in female mice. Toxicol. Lett. 2014, 228, 235–240. [Google Scholar] [CrossRef] [PubMed]
  92. Schmidt, J.S.; Schaedlich, K.; Fiandanese, N.; Pocar, P.; Fischer, B. Effects of di(2-ethylhexyl) phthalate (DEHP) on female fertility and adipogenesis in C3H/N mice. Environ. Health Perspect. 2012, 120, 1123–1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lovekamp-Swan, T.; Jetten, A.M.; Davis, B.J. Dual activation of PPARalpha and PPARgamma by mono-(2-ethylhexyl) phthalate in rat ovarian granulosa cells. Mol. Cell Endocrinol. 2003, 201, 133–141. [Google Scholar] [CrossRef]
  94. Xu, C.; Chen, J.A.; Qiu, Z.; Zhao, Q.; Luo, J.; Yang, L.; Zeng, H.; Huang, Y.; Zhang, L.; Cao, J.; et al. Ovotoxicity and PPAR-mediated aromatase downregulation in female Sprague-Dawley rats following combined oral exposure to benzo [a]pyrene and di-(2-ethylhexyl) phthalate. Toxicol. Lett. 2010, 199, 323–332. [Google Scholar] [CrossRef]
  95. Ford, J.H. Reduced quality and accelerated follicle loss with female reproductive aging—Does decline in theca dehydroepiandrosterone (DHEA) underlie the problem? J. Biomed. Sci. 2013, 20, 93. [Google Scholar] [CrossRef] [Green Version]
  96. Eftekhari Moghadam, A.R.; Saki, G.; Hemadi, M.; Mazaheri, Z.; Khodadadi, A. Effect of dehydroepiandrosterone on meiotic spindle structure and oocyte quality in mice. Iran. J. Basic Med. Sci. 2018, 21, 1020–1025. [Google Scholar]
  97. Zhao, Y.; Tan, Y.S.; Strynar, M.J.; Perez, G.; Haslam, S.Z.; Yang, C. Perfluorooctanoic acid effects on ovaries mediate its inhibition of peripubertal mammary gland development in Balb/c and C57Bl/6 mice. Reprod. Toxicol. 2012, 33, 563–576. [Google Scholar] [CrossRef] [Green Version]
  98. Soares, A.F.; Nosjean, O.; Cozzone, D.; D’Orazio, D.; Becchi, M.; Guichardant, M.; Ferry, G.; Boutin, J.A.; Lagarde, M.; Geloen, A. Covalent binding of 15-deoxy-delta12,14-prostaglandin J2 to PPARgamma. Biochem. Biophys. Res. Commun. 2005, 337, 521–525. [Google Scholar] [CrossRef]
  99. Li, J.; Guo, C.; Wu, J. 15-Deoxy-(12,14)-Prostaglandin J2 (15d-PGJ2), an Endogenous Ligand of PPAR-γ: Function and Mechanism. PPAR Res. 2019, 2019, 7242030. [Google Scholar] [CrossRef] [Green Version]
  100. Kurtz, M.; Capobianco, E.; Careaga, V.; Martinez, N.; Mazzucco, M.B.; Maier, M.; Jawerbaum, A. Peroxisome proliferator-activated receptor ligands regulate lipid content, metabolism, and composition in fetal lungs of diabetic rats. J. Endocrinol. 2014, 220, 345–359. [Google Scholar] [CrossRef] [Green Version]
  101. Kaplan, J.M.; Cook, J.A.; Hake, P.W.; O’Connor, M.; Burroughs, T.J.; Zingarelli, B. 15-Deoxy-delta(12,14)-prostaglandin J(2) (15D-PGJ(2)), a peroxisome proliferator activated receptor gamma ligand, reduces tissue leukosequestration and mortality in endotoxic shock. Shock 2005, 24, 59–65. [Google Scholar] [CrossRef] [PubMed]
  102. Berger, T.; Horner, C.M. In vivo exposure of female rats to toxicants may affect oocyte quality. Reprod. Toxicol. 2003, 17, 273–281. [Google Scholar] [CrossRef]
  103. Makris, S.L.; Scott, C.S.; Fox, J.; Knudsen, T.B.; Hotchkiss, A.K.; Arzuaga, X.; Euling, S.Y.; Powers, C.M.; Jinot, J.; Hogan, K.A.; et al. A systematic evaluation of the potential effects of trichloroethylene exposure on cardiac development. Reprod. Toxicol. 2016, 65, 321–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. da Costa, C.S.; Miranda-Alves, L.; La Merrill, M.A.; Silva, I.V.; Graceli, J.B. The tributyltin leads to obesogenic mammary gland abnormalities in adult female rats. Toxicol. Lett. 2019, 307, 59–71. [Google Scholar] [CrossRef]
  105. Crawford, K.A.; Clark, B.W.; Heiger-Bernays, W.J.; Karchner, S.I.; Hahn, M.E.; Nacci, D.E.; Schlezinger, J.J. Tributyltin disrupts fin development in Fundulus heteroclitus from both PCB-sensitive and resistant populations: Investigations of potential interactions between AHR and PPARgamma. Aquat. Toxicol. 2020, 218, 105334. [Google Scholar] [CrossRef]
  106. Hsueh, W.A.; Law, R. The central role of fat and effect of peroxisome proliferator-activated receptor-gamma on progression of insulin resistance and cardiovascular disease. Am. J. Cardiol. 2003, 92, 3J–9J. [Google Scholar] [CrossRef]
  107. Dennis, J.M.; Henley, W.E.; Weedon, M.N.; Lonergan, M.; Rodgers, L.R.; Jones, A.G.; Hamilton, W.T.; Sattar, N.; Janmohamed, S.; Holman, R.R.; et al. Sex and BMI Alter the Benefits and Risks of Sulfonylureas and Thiazolidinediones in Type 2 Diabetes: A Framework for Evaluating Stratification Using Routine Clinical and Individual Trial Data. Diabetes Care 2018, 41, 1844–1853. [Google Scholar] [CrossRef] [Green Version]
  108. Mohiyiddeen, L.; Watson, A.J.; Apostolopoulos, N.V.; Berry, R.; Alexandraki, K.I.; Jude, E.B. Effects of low-dose metformin and rosiglitazone on biochemical, clinical, metabolic and biophysical outcomes in polycystic ovary syndrome. J. Obstet. Gynaecol. 2013, 33, 165–170. [Google Scholar] [CrossRef]
  109. Vigerust, N.F.; Bohov, P.; Bjorndal, B.; Seifert, R.; Nygard, O.; Svardal, A.; Glintborg, D.; Berge, R.K.; Gaster, M. Free carnitine and acylcarnitines in obese patients with polycystic ovary syndrome and effects of pioglitazone treatment. Fertil. Steril. 2012, 98, 1620–1626 e1. [Google Scholar] [CrossRef]
  110. Minge, C.E.; Bennett, B.D.; Norman, R.J.; Robker, R.L. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone reverses the adverse effects of diet-induced obesity on oocyte quality. Endocrinology 2008, 149, 2646–2656. [Google Scholar] [CrossRef] [Green Version]
  111. Rak-Mardyla, A.; Karpeta, A. Rosiglitazone stimulates peroxisome proliferator-activated receptor gamma expression and directly affects in vitro steroidogenesis in porcine ovarian follicles. Theriogenology 2014, 82, 1–9. [Google Scholar] [CrossRef] [PubMed]
  112. Kurzynska, A.; Bogacki, M.; Chojnowska, K.; Bogacka, I. Peroxisome proliferator activated receptor ligands affect progesterone and 17beta-estradiol secretion by porcine corpus luteum during early pregnancy. J. Physiol. Pharmacol. 2014, 65, 709–717. [Google Scholar]
  113. Toda, K.; Okada, T.; Miyaura, C.; Saibara, T. Fenofibrate, a ligand for PPARalpha, inhibits aromatase cytochrome P450 expression in the ovary of mouse. J. Lipid Res. 2003, 44, 265–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Abdel-Raheem, I.T.; Omran, G.A.; Katary, M.A. Irbesartan, an angiotensin II receptor antagonist, with selective PPAR-γ-modulating activity improves function and structure of chemotherapy-damaged ovaries in rats. Fundam. Clin. Pharmacol. 2015, 29, 286–298. [Google Scholar] [CrossRef] [PubMed]
  115. Polkowski, K.; Mazurek, A.P. Biological properties of genistein. A review of in vitro and in vivo data. Acta Pol. Pharm. 2000, 57, 135–155. [Google Scholar] [PubMed]
  116. Lv, Z.; Fan, H.; Zhang, B.; Ning, C.; Xing, K.; Guo, Y. Dietary genistein supplementation in laying broiler breeder hens alters the development and metabolism of offspring embryos as revealed by hepatic transcriptome analysis. FASEB J. 2018, 32, 4214–4228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Wang, L.; Waltenberger, B.; Pferschy-Wenzig, E.M.; Blunder, M.; Liu, X.; Malainer, C.; Blazevic, T.; Schwaiger, S.; Rollinger, J.M.; Heiss, E.H.; et al. Natural product agonists of peroxisome proliferator-activated receptor gamma (PPARgamma): A review. Biochem. Pharmacol. 2014, 92, 73–89. [Google Scholar] [CrossRef] [Green Version]
  118. Kang, H.J.; Hwang, S.J.; Yoon, J.A.; Jun, J.H.; Lim, H.J.; Yoon, T.K.; Song, H. Activation of peroxisome proliferators-activated receptor delta (PPARdelta) promotes blastocyst hatching in mice. Mol. Hum. Reprod. 2011, 17, 653–660. [Google Scholar] [CrossRef] [Green Version]
  119. Heaney, A.P.; Fernando, M.; Melmed, S. PPAR-γ receptor ligands: Novel therapy for pituitary adenomas. J. Clin. Investig. 2003, 111, 1381–1388. [Google Scholar] [CrossRef]
  120. Chang, S.N.; Lee, J.M.; Oh, H.; Kim, U.; Ryu, B.; Park, J.H. Troglitazone inhibits the migration and invasion of PC-3 human prostate cancer cells by upregulating E-cadherin and glutathione peroxidase 3. Oncol. Lett. 2018, 16, 5482–5488. [Google Scholar] [CrossRef] [Green Version]
  121. Xu, Y.; Iyengar, S.; Roberts, R.L.; Shappell, S.B.; Peehl, D.M. Primary culture model of peroxisome proliferator-activated receptor gamma activity in prostate cancer cells. J. Cell Physiol. 2003, 196, 131–143. [Google Scholar] [CrossRef] [PubMed]
  122. Stephen, R.L.; Gustafsson, M.C.; Jarvis, M.; Tatoud, R.; Marshall, B.R.; Knight, D.; Ehrenborg, E.; Harris, A.L.; Wolf, C.R.; Palmer, C.N. Activation of peroxisome proliferator-activated receptor delta stimulates the proliferation of human breast and prostate cancer cell lines. Cancer Res. 2004, 64, 3162–3170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Xu, P.; Zhai, Y.; Wang, J. The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis. Int. J. Mol. Sci. 2018, 19, 1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Jahansouz, C.; Xu, H.; Hertzel, A.V.; Kizy, S.; Steen, K.A.; Foncea, R.; Serrot, F.J.; Kvalheim, N.; Luthra, G.; Ewing, K.; et al. Partitioning of adipose lipid metabolism by altered expression and function of PPAR isoforms after bariatric surgery. Int. J. Obes. 2018, 42, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Wang, J.; Xu, P.; Xie, X.; Li, J.; Zhang, J.; Wang, J.; Hong, F.; Li, J.; Zhang, Y.; Song, Y.; et al. DBZ (Danshensu Bingpian Zhi), a Novel Natural Compound Derivative, Attenuates Atherosclerosis in Apolipoprotein E-Deficient Mice. J. Am. Heart Assoc. 2017, 6, e006297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Levak-Frank, S.; Radner, H.; Walsh, A.; Stollberger, R.; Knipping, G.; Hoefler, G.; Sattler, W.; Weinstock, P.H.; Breslow, J.L.; Zechner, R. Muscle-specific overexpression of lipoprotein lipase causes a severe myopathy characterized by proliferation of mitochondria and peroxisomes in transgenic mice. J. Clin. Investig. 1995, 96, 976–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Hoefler, G.; Noehammer, C.; Levak-Frank, S.; el-Shabrawi, Y.; Schauer, S.; Zechner, R.; Radner, H. Muscle-specific overexpression of human lipoprotein lipase in mice causes increased intracellular free fatty acids and induction of peroxisomal enzymes. Biochimie 1997, 79, 163–168. [Google Scholar] [CrossRef]
  128. Hodel, C. Myopathy and rhabdomyolysis with lipid-lowering drugs. Toxicol. Lett. 2002, 128, 159–168. [Google Scholar] [CrossRef]
  129. Motojima, K.; Seto, K. Fibrates and statins rapidly and synergistically induce pyruvate dehydrogenase kinase 4 mRNA in the liver and muscles of mice. Biol. Pharm. Bull. 2003, 26, 954–958. [Google Scholar] [CrossRef] [Green Version]
  130. Toffoli, B.; Gilardi, F.; Winkler, C.; Soderberg, M.; Kowalczuk, L.; Arsenijevic, Y.; Bamberg, K.; Bonny, O.; Desvergne, B. Nephropathy in Pparg-null mice highlights PPARgamma systemic activities in metabolism and in the immune system. PLoS ONE 2017, 12, e0171474. [Google Scholar] [CrossRef] [Green Version]
  131. Corrales, P.; Izquierdo-Lahuerta, A.; Medina-Gomez, G. Maintenance of Kidney Metabolic Homeostasis by PPAR Gamma. Int. J. Mol. Sci. 2018, 19, 2063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Lee, Y.; Lee, S.; Chang, S.C.; Lee, J. Significant roles of neuroinflammation in Parkinson’s disease: Therapeutic targets for PD prevention. Arch. Pharm Res. 2019, 42, 416–425. [Google Scholar] [CrossRef] [PubMed]
  133. D’Angelo, M.; Antonosante, A.; Castelli, V.; Catanesi, M.; Moorthy, N.; Iannotta, D.; Cimini, A.; Benedetti, E. PPARs and Energy Metabolism Adaptation during Neurogenesis and Neuronal Maturation. Int. J. Mol. Sci. 2018, 19, 1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Paintlia, A.S.; Paintlia, M.K.; Singh, A.K.; Singh, I. Modulation of Rho-Rock signaling pathway protects oligodendrocytes against cytokine toxicity via PPAR-alpha-dependent mechanism. Glia 2013, 61, 1500–1517. [Google Scholar] [CrossRef] [Green Version]
  135. Panlilio, L.V.; Justinova, Z.; Mascia, P.; Pistis, M.; Luchicchi, A.; Lecca, S.; Barnes, C.; Redhi, G.H.; Adair, J.; Heishman, S.J.; et al. Novel use of a lipid-lowering fibrate medication to prevent nicotine reward and relapse: Preclinical findings. Neuropsychopharmacology 2012, 37, 1838–1847. [Google Scholar] [CrossRef] [Green Version]
  136. Porta, N.; Vallee, L.; Lecointe, C.; Bouchaert, E.; Staels, B.; Bordet, R.; Auvin, S. Fenofibrate, a peroxisome proliferator-activated receptor-alpha agonist, exerts anticonvulsive properties. Epilepsia 2009, 50, 943–948. [Google Scholar] [CrossRef]
  137. Wang, G.; Zhou, Y.; Wang, Y.; Li, D.; Liu, J.; Zhang, F. Age-Associated Dopaminergic Neuron Loss and Midbrain Glia Cell Phenotypic Polarization. Neuroscience 2019, 415, 89–96. [Google Scholar] [CrossRef]
  138. Loane, D.J.; Byrnes, K.R. Role of microglia in neurotrauma. Neurotherapeutics 2010, 7, 366–377. [Google Scholar] [CrossRef]
  139. Villapol, S. Roles of Peroxisome Proliferator-Activated Receptor Gamma on Brain and Peripheral Inflammation. Cell Mol. Neurobiol. 2018, 38, 121–132. [Google Scholar] [CrossRef]
  140. Le Menn, G.; Neels, J.G. Regulation of Immune Cell Function by PPARs and the Connection with Metabolic and Neurodegenerative Diseases. Int. J. Mol. Sci. 2018, 19, 1575. [Google Scholar] [CrossRef] [Green Version]
  141. Dehmer, T.; Heneka, M.T.; Sastre, M.; Dichgans, J.; Schulz, J.B. Protection by pioglitazone in the MPTP model of Parkinson’s disease correlates with I kappa B alpha induction and block of NF kappa B and iNOS activation. J. Neurochem. 2004, 88, 494–501. [Google Scholar] [CrossRef] [PubMed]
  142. Breidert, T.; Callebert, J.; Heneka, M.T.; Landreth, G.; Launay, J.M.; Hirsch, E.C. Protective action of the peroxisome proliferator-activated receptor-gamma agonist pioglitazone in a mouse model of Parkinson’s disease. J. Neurochem. 2002, 82, 615–624. [Google Scholar] [CrossRef] [PubMed]
  143. Schnegg, C.I.; Kooshki, M.; Hsu, F.C.; Sui, G.; Robbins, M.E. PPARdelta prevents radiation-induced proinflammatory responses in microglia via transrepression of NF-kappaB and inhibition of the PKCalpha/MEK1/2/ERK1/2/AP-1 pathway. Free Radic. Biol. Med. 2012, 52, 1734–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Song, J.; Choi, S.M.; Kim, B.C. Adiponectin Regulates the Polarization and Function of Microglia via PPAR-γ Signaling under Amyloid β Toxicity. Front. Cell Neurosci. 2017, 11, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The regulation of PPARs in cardiotoxicity.
Figure 1. The regulation of PPARs in cardiotoxicity.
Cells 09 00352 g001
Figure 2. The regulation by PPARs in gastrointestinal toxicity.
Figure 2. The regulation by PPARs in gastrointestinal toxicity.
Cells 09 00352 g002
Figure 3. Concept map of the PPARs in various systemic toxicities.
Figure 3. Concept map of the PPARs in various systemic toxicities.
Cells 09 00352 g003
Table 1. The information and adverse reactions or toxicity of peroxisome proliferator-activated receptor (PPAR) targets related to 18 approved clinical drugs.
Table 1. The information and adverse reactions or toxicity of peroxisome proliferator-activated receptor (PPAR) targets related to 18 approved clinical drugs.
Generic Name
(Brand Name)
Type of PPAR AgonistMolecular Weight and Molecular FormulaStructureCompanyIndicationsAdverse Reaction or Toxicity
Rosiglitazone maleate
(Avandia)
PPARγ agonist473.5
C22H23N3O7S
Cells 09 00352 i001GlaxoSmithKlineDiabetesHeadache, cough, cold symptoms, and back pain
Pioglitazone hydrochloride
(Actos)
PPARγ agonist392.898
C19H21ClN2O3S
Cells 09 00352 i002Takeda/LillyDiabetesCold or flu-like symptoms, headache, gradual weight gain, muscle pain, back pain, tooth problems, and mouth pain
Lobeglitazone sulfate
(Duvie)
Dual PPARα/γ agonist578.61
C24H26N4O9S2
Cells 09 00352 i003Chong Kun DangDiabetesEdema and weight gain
Alogliptin benzoate/pioglitazone hydrochloride (Oseni)Dipeptidyl peptidase IV inhibitor/PPARγ agonist461.519(C25H27N5O4)/392.898 (C19H21ClN2O3S) Cells 09 00352 i004TakedaDiabetesUpper respiratory tract infection, bone fracture, headache, nasopharyngitis, and pharyngitis
Glimepiride/pioglitazone hydrochloride
(Duetact)
Sulfonylurea receptor modulator/PPARγ agonist490.62(C24H34N4O5S)/392.898 (C19H21ClN2O3S) Cells 09 00352 i005TakedaDiabetesCongestive heart failure, hypoglycemia, edema, fractures, and hemolytic anemia
Pioglitazone hydrochloride/metformin hydrochloride
(Actoplus Met)
PPARγ agonist/ adenosine monophosphate-activated protein kinase (AMPK) activator392.898 (C19H21ClN2O3S)/165.6(C4H12ClN5) Cells 09 00352 i006TakedaDiabetesHeadache, nausea, vomiting, stomach upset, diarrhea, weakness, sore throat, muscle pain, weight gain, tooth problems, a metallic taste in the mouth, and sneezing, runny nose, cough, or other signs of a cold
Rosiglitazone maleate/metformin hydrochloride
(Avandamet)
PPARγ agonist; AMPK activator473.5(C22H23N3O7S)/165.6(C4H12ClN5) Cells 09 00352 i007GlaxoSmithKlineDiabetesLactic acidosis, cardiac failure, adverse cardiovascular events, edema, weight gain, hepatic effects, macular edema, fractures, hematologic effects, and ovulation
Glimepiride/rosiglitazone maleate (Avandaryl)Sulfonylurea receptor modulator/PPARγ agonist490.62 (C24H34N4O5S)/473.5(C22H23N3O7S) Cells 09 00352 i008GlaxoSmithKlineDiabetesCardiac failure with rosiglitazone, major adverse cardiovascular events, hypoglycemia, edema, weight gain, hepatic effects, macular edema, fractures, hypersensitivity reactions, hematologic effects, hemolytic anemia, and increased risk of cardiovascular mortality for sulfonylurea drugs
Clofibrate
(Atromid-S)
PPARα agonist242.699
C12H15ClO3
Cells 09 00352 i009PfizerHyperlipidemia Hypertriglyceridemia HypercholesterolemiaCommon: diarrhea, nausea
Rare: abnormal heart rhythm, acute inflammation of the pancreas, anemia, angina, gallstones, kidney failure, and low levels of white blood cells
Fenofibrate
(Antara)
PPARα agonist360.834
C20H21ClO4
Cells 09 00352 i010AbbvieHypercholesterolemia HypertriglyceridemiaCommon: abnormal liver tests including aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and headache
Rare: high blood pressure, dizziness, itching, nausea, upset stomach, constipation, diarrhea, urinary tract infections, muscle pain, kidney problems, and respiratory tract infections
Choline fenofibrate
(Fenofibric Acid)
PPARα agonist421.918
C22H28ClNO5
Cells 09 00352 i011AbbvieHyperlipidemiaDiarrhea, dyspepsia, nasopharyngitis, sinusitis, upper respiratory tract infection, arthralgia, myalgia, pain in extremities, dizziness
Bezafibrate
(Bezalip)
PPARα agonist361.822
C19H20ClNO4
Cells 09 00352 i012Roche DiagnosticsHypertriglyceridemia hypercholesterolemia mixed hyperlipidemiaStomach upset, stomach pain, gas, or nausea may occur in the first several days; itchy skin, redness, headache, and dizziness
Gemfibrozil
(Lopid)
PPARα agonist250.338
C15H22O3
Cells 09 00352 i013PfizerHyperlipidemia
Ischemic heart disorder
Stomach upset, stomach/abdominal pain, nausea, vomiting, diarrhea, constipation, rash, dizziness, headache, changes in the way things taste, muscle pain
Ciprofibrate
(Lipanor)
PPARα agonist289.152
C13H14Cl2O3
Cells 09 00352 i014Sanofi-AventisHyperlipidemiaHair loss, balding, headache, balance problems, feeling dizzy, drowsiness or fatigue, feeling sick (nausea) or being sick (vomiting), diarrhea, indigestion or stomach pains, muscle pains
Pemafibrate
(Parmodia)
PPARα agonist490.556
C28H30N2O6
Cells 09 00352 i015KowaDyslipidemiaCholelithiasis (upper abdominal pain, fever) and diabetes mellitus (dry mouth, excess intake of fluid, excessive urination, fatigue)
Pravastatin sodium/fenofibrate
(Pravafenix)
3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibitor/PPARα agonist446.5(C23H35NaO7)/360.83(C20H21ClO4) Cells 09 00352 i016Laboratoires SMBMixed hyperlipidemia
Coronary heart disease
Abdominal distension (bloating), abdominal pain (stomach ache), constipation, diarrhea, dry mouth, dyspepsia (heartburn), eructation (belching), flatulence (gas), nausea (feeling sick), abdominal discomfort, vomiting, and raised blood levels of liver enzymes
Fenofibrate/simvastatin
(Cholib)
PPARα agonist/
HMGCR inhibitor
360.834 (C20H21ClO4)/418.57(C25H38O5) Cells 09 00352 i017MylanMixed hyperlipidemiaRaised blood creatinine levels, upper-respiratory-tract infection (colds), increased blood platelet counts, gastroenteritis (diarrhea and vomiting) and increased levels of alanine aminotransferase
Saroglitazar
(Lipaglyn)
PPARα/γ agonist439.57
C25H29NO4S
Cells 09 00352 i018Zydus CadilaDiabetic dyslipidemiaAsthenia, gastritis, chest discomfort, peripheral edema, dizziness, and tremors

Share and Cite

MDPI and ACS Style

Xi, Y.; Zhang, Y.; Zhu, S.; Luo, Y.; Xu, P.; Huang, Z. PPAR-Mediated Toxicology and Applied Pharmacology. Cells 2020, 9, 352. https://doi.org/10.3390/cells9020352

AMA Style

Xi Y, Zhang Y, Zhu S, Luo Y, Xu P, Huang Z. PPAR-Mediated Toxicology and Applied Pharmacology. Cells. 2020; 9(2):352. https://doi.org/10.3390/cells9020352

Chicago/Turabian Style

Xi, Yue, Yunhui Zhang, Sirui Zhu, Yuping Luo, Pengfei Xu, and Zhiying Huang. 2020. "PPAR-Mediated Toxicology and Applied Pharmacology" Cells 9, no. 2: 352. https://doi.org/10.3390/cells9020352

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop