Fact-checked by Grok 2 weeks ago

Peroxisome proliferator-activated receptor

Peroxisome proliferator-activated receptors (PPARs) are a family of ligand-activated transcription factors that belong to the superfamily of receptors, playing crucial roles in regulating genes involved in and glucose , , , and . Discovered in the early 1990s through studies on rodent liver responses to peroxisome proliferators, PPARs function by forming heterodimers with the (RXR) and binding to specific DNA response elements to modulate target . There are three primary isoforms—PPARα, PPARβ/δ, and PPARγ—each encoded by distinct genes and exhibiting unique tissue distributions and ligand affinities. PPARα, predominantly expressed in metabolically active tissues such as the liver, heart, kidneys, and , primarily governs uptake, oxidation, and , thereby reducing circulating levels. PPARβ/δ, the most ubiquitously expressed isoform with high levels in the liver, , and , promotes , enhances expenditure, and influences , while also exhibiting properties. In contrast, PPARγ is mainly found in , macrophages, and the colon, where it drives , insulin sensitization, and storage, contributing to the regulation of and responses in immune cells. PPARs are activated by a variety of endogenous ligands, including polyunsaturated fatty acids (e.g., and ), their metabolites like eicosanoids, and oxidized , as well as synthetic compounds such as fibrates for PPARα and thiazolidinediones (TZDs) for PPARγ. These receptors have significant therapeutic implications; for instance, PPARα agonists like fibrates are used to treat by lowering triglycerides, while PPARγ agonists such as pioglitazone improve insulin sensitivity in , though they can cause side effects like fluid retention and weight gain. Additionally, in 2024, the U.S. granted accelerated approval to the PPARδ agonist seladelpar and the dual PPARα/δ agonist elafibranor for the treatment of in patients with an inadequate response to . Recent research as of 2023 has expanded their relevance to non-metabolic conditions, including neuroprotective effects in models via dual PPARβ/δ and PPARγ agonists, and potential roles in reducing in cardiac and liver diseases.

Discovery and Nomenclature

Historical Development

The development of fibrates in the represented a pivotal advance in treating . , synthesized in 1962 by Thorp and Waring at , was the first such compound introduced clinically to reduce elevated plasma and levels in patients with dyslipidemias. By the late , it had gained widespread use as a hypolipidemic agent, though its mechanism remained unclear at the time. In the , research on fibrates like in models uncovered their ability to induce marked of in hepatic , prompting the coining of the term "peroxisome proliferators" for this class of chemicals. These studies, primarily in and mice, demonstrated that such compounds upregulated enzymes involved in peroxisomal β-oxidation of , linking action to . Mid-decade experiments by Reddy and colleagues further showed that proliferators enhanced transcription of genes encoding peroxisomal oxidation enzymes in liver, suggesting a receptor-mediated process. The molecular basis for peroxisome proliferation was elucidated in 1990 when Issemann and Green cloned the first proliferator-activated receptor (PPARα) from a rat liver , identifying it as a ligand-activated in the nuclear hormone receptor superfamily. This discovery provided a receptor target for fibrates and explained their hypolipidemic effects through regulation of genes. Shortly thereafter, in 1992, Dreyer et al. cloned PPARβ (also termed PPARδ) from Xenopus laevis, expanding the family and revealing early expression patterns in frog embryos that hinted at developmental roles. The same year, Kliewer et al. demonstrated that PPARs function as obligatory heterodimers with X receptors (RXR), a critical insight into their activation and DNA-binding mechanism via peroxisome proliferator response elements. PPARγ was cloned in mammals starting in 1994, with Tontonoz et al. isolating the isoform from an cDNA library and linking it to fat cell . Human PPARγ followed in 1995, confirming its role in storage and . These early findings established PPARs as key regulators of and , setting the stage for broader physiological investigations while highlighting species-specific responses, such as pronounced effects in versus humans.

Isoforms and Nomenclature

The peroxisome proliferator-activated receptors (PPARs) comprise three main isoforms classified within the superfamily as 1 group C (NR1C): PPARα (NR1C1, encoded by the PPARA gene), PPARβ/δ (NR1C2, encoded by PPARD), and PPARγ (NR1C3, encoded by PPARG). These isoforms were originally named for their activation by peroxisome proliferators—compounds that induce proliferation in liver—reflecting early observations of their role in and responses. Over time, the evolved to emphasize their membership in the NR1C , with PPARβ/δ standardized to avoid between historical designations as either PPARβ or PPARδ. The human genes are located on distinct chromosomes: PPARA at 22q13.31, PPARD at 6p21.2, and PPARG at 3p25.2. Each isoform exhibits characteristic tissue distribution and primary functions, distinguishing their roles in metabolic regulation. PPARα is predominantly expressed in metabolically active tissues such as the liver, kidney, heart, and small intestine, where it primarily governs fatty acid β-oxidation and peroxisomal proliferation. In contrast, PPARβ/δ displays ubiquitous expression across tissues including skeletal muscle, adipose, and brain, contributing to overall energy homeostasis, mitochondrial fatty acid oxidation, and adaptive responses to fasting or exercise. PPARγ, the most adipose-enriched isoform, is highly expressed in white and brown adipose tissue, playing a central role in adipocyte differentiation (adipogenesis), lipid storage, and insulin sensitization. The PPARG gene produces two principal protein isoforms through alternative promoter usage and splicing: PPARγ1, which is broadly expressed in tissues such as adipose, liver, muscle, and immune cells, and PPARγ2, which includes an additional 30 at the and is predominantly restricted to and the intestine. These variants share overlapping DNA-binding and ligand-dependent activation properties but differ in their transcriptional potency and tissue-specific regulation.

Molecular Structure

Domain Organization

Peroxisome proliferator-activated receptors (PPARs) exhibit a conserved modular architecture typical of nuclear receptors, comprising distinct functional domains that facilitate DNA binding, ligand interaction, and transcriptional regulation. The N-terminal A/B domain harbors the ligand-independent activation function 1 (AF-1), which is involved in transcriptional activation and is subject to phosphorylation for modulation of activity. This domain is followed by the central DNA-binding domain (DBD, or C domain), a flexible hinge region (D domain), and the C-terminal ligand-binding domain (LBD, or E/F domains), which encompasses the ligand-dependent activation function 2 (AF-2). The hinge region serves as a flexible linker and docking site for coregulatory proteins, while the LBD also contains interfaces for heterodimerization with retinoid X receptor (RXR). The DBD consists of approximately 70 amino acids organized into two zinc-finger motifs that recognize specific DNA sequences known as peroxisome proliferator response elements (PPREs). These motifs enable PPAR-RXR heterodimers to bind to direct repeats of the consensus hexameric sequence AGGTCA separated by a single nucleotide (AGGTCA N AGGTCA, or DR-1 motif), thereby targeting PPAR-responsive genes. The LBD forms a globular with 12 α- surrounding a large hydrophobic - pocket, where 12 (H12) plays a critical role in AF-2 function. Upon , H12 undergoes a conformational repositioning that creates a surface for coactivator proteins, thereby facilitating transcriptional . Isoform-specific variations occur primarily in the A/B ; for instance, PPARγ2 contains an additional 30 at the compared to PPARγ1, enhancing its potential particularly in adipogenic contexts.

Ligand-Binding and Activation Mechanism

Peroxisome proliferator-activated receptors (PPARs) function as obligate heterodimers with the (RXR), a configuration essential for their transcriptional activity. This heterodimerization occurs primarily through interactions between the ligand-binding domains (LBDs) of PPAR and RXR, enabling cooperative DNA binding and ligand responsiveness. In the absence of ligand, the PPAR-RXR complex associates with corepressor proteins, such as NCoR and SMRT, which recruit histone deacetylases to maintain transcriptional repression at target gene promoters. Ligand binding to the PPAR LBD induces a conformational change that repositions helix 12 (also known as the AF-2 helix), serving as a molecular switch to disrupt corepressor interactions and expose a coactivator-binding groove. This repositioning releases corepressors, allowing recruitment of coactivators like steroid receptor coactivator-1 (SRC-1), which interact via conserved LXXLL motifs to bridge the receptor with the transcriptional machinery. The activated heterodimer then binds to peroxisome proliferator response elements (PPREs) in target gene promoters, characterized by a direct repeat spacing (DR1) consensus sequence: AGGTCA N AGGTCA, where N is any nucleotide. Coactivators such as CREB-binding protein (CBP)/p300 further facilitate chromatin remodeling through intrinsic histone acetyltransferase (HAT) activity, promoting an open chromatin structure conducive to transcription initiation. The efficiency of activation depends on and type, with full agonists eliciting maximal 12 stabilization and coactivator , while partial agonists induce intermediate conformational states with reduced transcriptional output. For instance, fibrates exhibit values of approximately 1-10 μM for PPARα activation, reflecting moderate potency that supports graded responses. Additionally, ligand-induced changes in the LBD propagate allosterically to the (DBD), enhancing DNA and heterodimer stability on PPREs. This allosteric communication ensures precise without requiring direct DBD modifications.

Expression Patterns

Tissue Distribution

Peroxisome proliferator-activated receptors (PPARs) exhibit distinct tissue-specific expression patterns across their isoforms, α, β/δ, and γ, as determined by mRNA and protein analyses in mammalian models including humans and . These patterns reflect basal distribution in adult tissues, with variations quantified through techniques such as RT-PCR and , often showing 10- to 30-fold differences in mRNA levels between high- and low-expressing organs. PPARα is highly expressed in metabolically active tissues, with the highest mRNA levels in the liver (up to 19-fold higher than in skeletal muscle), followed by the heart (7-fold higher than skeletal muscle), kidney, brown adipose tissue, and skeletal muscle. Lower expression occurs in the brain and white adipose tissue. At the cellular level, PPARα is predominantly localized in hepatocytes, cardiomyocytes, enterocytes, and proximal tubule cells of the kidney. PPARβ/δ displays a more ubiquitous expression profile across tissues, with relatively high mRNA and protein levels in the colon (particularly colonic epithelial cells), , , and adipocytes, as well as moderate levels in the liver, intestine, , lungs, , and macrophages. Quantitative assessments indicate that PPARβ/δ mRNA often exceeds that of the other isoforms by several fold in many tissues, such as the where it is the most abundant. PPARγ expression is more restricted, predominantly in white adipose tissue (10- to 30-fold higher mRNA than in liver or skeletal muscle), macrophages, and the colon (large intestine). The γ1 isoform is broadly distributed in tissues including heart, muscle, kidney, pancreas, and spleen, while γ2 is primarily in adipocytes, and γ3 in macrophages and white adipose tissue. Cellular localization is notable in adipocytes and immune cells for the γ isoforms. In most cell types, PPAR isoforms are primarily nuclear receptors, though they exhibit dynamic ligand-dependent shuttling between the nucleus and cytoplasm, with predominant nuclear retention under basal conditions. This localization is observed across neurons, astrocytes, and other cell types in tissues like the brain and peripheral nervous system.

Developmental and Pathological Expression

Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) plays a critical role in embryonic development, particularly in placental formation and skin barrier function. Targeted disruption of the PPARδ gene in mice leads to defective placentation, characterized by impaired trophoblast differentiation and vascularization, resulting in frequent embryonic lethality at mid-gestation (around embryonic day 10.5), with rare survivors. Additionally, PPARβ/δ deficiency in epidermal tissues disrupts cutaneous permeability barrier homeostasis, highlighting its essential function in skin development and wound healing processes. In contrast, PPARγ is indispensable for adipocyte differentiation during embryogenesis, where it transcriptionally regulates key genes involved in lipid accumulation and adipose tissue formation, as evidenced by PPARγ knockout mice exhibiting severe defects in white adipose tissue development alongside placental and cardiac abnormalities. In pathological conditions, PPAR expression patterns shift dynamically in response to disease progression. PPARγ mRNA and protein levels are upregulated in atherosclerotic plaques, correlating with lesion severity and potentially modulating formation and within the vascular wall. Conversely, PPARα expression is downregulated in the livers of patients and animal models with nonalcoholic (NAFLD), contributing to impaired oxidation and hepatic accumulation, as or suppresses PPARα activity through pathways like HIF-2α signaling. PPAR expression is tightly regulated by transcriptional and epigenetic mechanisms that respond to developmental cues and pathological stressors. Transcription factors such as NF-κB can repress PPARγ expression by promoting inflammatory signaling and downregulating PPAR promoters, while STAT family members, including STAT5b and STAT6, modulate PPAR activity—STAT5b inhibits PPAR-regulated transcription, whereas STAT6 facilitates PPARγ DNA binding and target gene activation. Epigenetic modifications, particularly DNA methylation at promoter regions, further control PPAR levels; for instance, hypermethylation of the PPARγ1 promoter in obesity-driven inflammation silences expression, whereas demethylation under high-fat conditions enhances PPARγ transcription in adipose tissues. During aging, PPARα expression declines in the liver, paralleling metabolic disturbances such as reduced oxidation and increased susceptibility to . This age-related downregulation in models is associated with diminished hepatic PPARα mRNA and protein levels, contributing to overall metabolic decline without altering PPARα protein stability. Such changes underscore PPARs' dynamic expression as a bridge between developmental programming and - or disease-associated dysregulation.

Physiological Functions

Regulation of Metabolism

Peroxisome proliferator-activated receptors (PPARs) serve as key transcriptional regulators of metabolic , primarily by modulating the expression of genes involved in , glucose, and . Upon activation by endogenous ligands or synthetic agonists, PPARs form heterodimers with X receptors (RXRs) and bind to proliferator response elements (PPREs) in target gene promoters, thereby coordinating adaptive responses to nutritional states such as fasting or feeding. This regulation ensures efficient fuel utilization, preventing accumulation and maintaining balance across tissues like liver, adipose, and muscle. PPARα predominantly governs hepatic lipid catabolism, particularly during when circulating s rise and activate the receptor. It induces the transcription of genes essential for β-oxidation, including ACOX1 ( oxidase 1), the rate-limiting enzyme in peroxisomal oxidation, and CPT1A (carnitine palmitoyltransferase 1A), which facilitates mitochondrial entry. This upregulation enhances the breakdown of long-chain s into , supporting ketone body production and sparing glucose for vital organs. In PPARα-deficient mice, leads to profound hepatic and due to impaired β-oxidation capacity, underscoring the isoform's critical role in lipid clearance. In contrast, PPARγ drives adipocyte differentiation and lipid storage in adipose tissue, promoting adipogenesis through the activation of genes like CEBPα and FABP4. It enhances insulin sensitivity by upregulating GLUT4 (glucose transporter 4), which increases glucose uptake in adipocytes and skeletal muscle, and ADIPOQ (adiponectin), an adipokine that improves systemic insulin signaling and suppresses hepatic gluconeogenesis. These actions redirect excess energy into fat storage, mitigating ectopic lipid deposition in non-adipose tissues. PPARγ activation thus balances lipid anabolism with insulin-mediated glucose homeostasis, preventing hyperglycemia during nutrient excess. PPARβ/δ, ubiquitously expressed, fine-tunes handling in and other oxidative tissues, particularly in response to exercise. It boosts mitochondrial uncoupling via UCP3 (uncoupling protein 3), which dissipates proton gradients to increase energy expenditure and reduce , while enhancing uptake through (fatty acid translocase), facilitating trans-membrane transport for subsequent oxidation. Exercise transiently elevates PPARβ/δ expression, promoting adaptive increases in and β-oxidation enzymes like CPT1, which improve and utilization. This isoform thus supports sustained energy production during physical activity. PPARs integrate with other transcription factors to maintain and . PPARα and PPARγ exhibit cross-talk with SREBP (sterol regulatory element-binding protein) pathways, where PPAR activation suppresses SREBP-1c-mediated to favor oxidation; similarly, interactions with LXR (liver X receptor) modulate efflux and reverse transport, preventing overload. Post-PPAR activation, β-oxidation flux rises due to elevated enzyme levels, modeled conceptually as an increase in maximum velocity (Vmax) in Michaelis-Menten : J = \frac{V_{\max} \cdot [\text{acyl-CoA}]}{K_m + [\text{acyl-CoA}]} where J is the flux, and PPAR-induced transcription amplifies Vmax through targets like CPT1A and ACOX1, enhancing overall catabolic throughput without altering substrate affinity (Km). This coordinated regulation ensures isoform-specific yet complementary control of metabolic flux, adapting to physiological demands.

Roles in Inflammation and Immunity

Peroxisome proliferator-activated receptors (PPARs) exert significant anti-inflammatory and immunomodulatory effects, particularly through their actions in immune cells and tissue repair processes. PPARγ plays a central role in macrophages by inhibiting the nuclear factor kappa B (NF-κB) pathway, which suppresses the production of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). This inhibition occurs via transrepression mechanisms, where PPARγ directly interacts with NF-κB p65, preventing its transcriptional activity. Additionally, PPARγ promotes the polarization of macrophages toward an anti-inflammatory M2 phenotype, characterized by increased expression of markers like arginase-1 (Arg-1) and CD206, which facilitates tissue resolution and repair. PPARα contributes to inflammation control primarily in the liver by suppressing the acute phase response (APR) induced by cytokines like IL-6, downregulating hepatic expression of acute phase proteins such as (Saa) and fibrinogen. This effect involves upregulation of , an endogenous inhibitor of , which sequesters in the cytoplasm and represses cytokine-driven inflammatory . PPARβ/δ, meanwhile, modulates adaptive immunity by regulating T-cell , favoring profiles that reduce pro-inflammatory production, and supports through enhanced keratinocyte migration and resistance to at injury sites. In PPARβ/δ-deficient models, wound closure is delayed due to increased keratinocyte and impaired re-epithelialization. The core mechanisms underlying these effects include transrepression via recruitment of corepressor complexes, such as NCoR/SMRT, which are stabilized by PPAR SUMOylation to block and activator protein-1 (AP-1) access to promoters. Ligand activation of PPARs also inhibits JNK signaling pathways, reducing AP-1 activity and downstream inflammatory responses in immune cells. Emerging research highlights PPARγ's role in mitigating viral-induced inflammation; for instance, PPARγ activation limits excessive storms and in models by repressing -driven responses in macrophages. These immunomodulatory functions of PPARs intersect with metabolic regulation, where lipid-derived influence both and immune balance.

Genetics

Gene Structure and Evolution

The PPAR genes, encoding the peroxisome proliferator-activated receptors (PPARα, PPARδ, and PPARγ), exhibit distinct genomic organizations in humans. The PPARA gene, located on 22q13.31, spans approximately 84 and consists of 8 s, with the coding distributed across exons 2 through 8. The PPARD gene, on 6p21.2, comprises 9 exons over about 85 , encoding a protein with modular domains typical of receptors. In contrast, the PPARG gene on 3p25.2 spans over 100 and contains 9 exons, with promoter usage and splicing; for instance, the PPARγ1 isoform uses exons A1/A2 and 1-6, while PPARγ2 initiates from an additional exon B and uses exons 1-6, resulting in a longer N-terminal activation domain. The PPAR genes belong to the ancient superfamily, which originated early in metazoan evolution through events predating bilaterian divergence. This superfamily, comprising over 50 members in s, is highly conserved across animals, with homologs identified in such as (e.g., the nuclear receptors E75, HR3, and HR38 share structural similarities in DNA- and ligand-binding domains). The three mammalian PPAR isoforms (α, δ, γ) arose from two rounds of whole-genome duplication in the ancestral lineage approximately 500 million years ago, followed by subfunctionalization that partitioned their roles in and development; these duplications expanded the NR1C subfamily from a single proto-PPAR gene in early chordates. Promoter regions of PPAR genes contain conserved regulatory elements that facilitate basal and inducible transcription. For example, the PPARA promoter includes binding sites for Sp1 transcription factors and AP-1 complexes, which drive constitutive expression in metabolically active tissues, alongside CpG islands susceptible to for epigenetic silencing. Similar motifs are present in PPARD and PPARG promoters, where Sp1 sites overlap with CpG-rich regions to integrate signals from growth factors and stress responses. Comparative genomics reveals key differences between human and PPAR loci that underlie species-specific responses to ligands. While the core structures are syntenic, variations in proliferator response elements (PPREs) within target genes—such as enhanced binding affinity in Acox1 and Cyp4a promoters—contribute to robust proliferation and hepatocarcinogenesis in mice and rats upon PPARα activation, effects absent in humans due to weaker PPRE interactions and lower receptor abundance in liver. These divergences likely stem from adaptive evolutionary pressures on , with human PPARα showing reduced efficiency for -specific PPRE motifs.

Polymorphisms and Mutations

The (PPARG) gene harbors the common (SNP) Pro12Ala (rs1801282), a missense variant in B that substitutes for at position 12 in the PPARγ2 isoform. This polymorphism is associated with a reduced risk of mellitus (T2DM), with meta-analyses indicating an 18-20% risk reduction conferred by the minor Ala allele (G) in various populations. Functionally, the Pro12Ala variant decreases PPARγ transcriptional activity by 30-50% and reduces ligand-binding affinity, leading to altered interactions with coactivators and diminished transactivation potential. Another prevalent SNP is Leu162Val (rs1800206) in the PPARA gene, which influences responses. Carriers of the Val allele exhibit elevated plasma total , low-density lipoprotein , and levels, particularly in men, contributing to interindividual variability in profiles. Rare loss-of-function mutations in PPARG, including frameshift variants, underlie familial partial type 3 (FPLD3), an autosomal dominant . These heterozygous mutations, such as those disrupting the DNA-binding or ligand-binding domains, impair PPARγ and adipogenic functions. In mouse models, PPARα null mutations generated by targeted gene disruption result in defective oxidation, with affected animals showing hepatic , , and impaired during fasting due to failure to induce target genes like MCAD and ACO. Population genetics of the PPARG Pro12Ala reveal varying frequencies, with the protective Ala occurring at approximately 10-14% in Europeans and 4-11% in East Asians, reflecting ethnic differences that may modulate susceptibility. For PPARA Leu162Val, the Val is around 7% in cohorts.

Pharmacology

Endogenous and Natural Ligands

Peroxisome proliferator-activated receptors (PPARs) are activated by a variety of endogenous ligands derived from , primarily fatty acids and their oxygenated derivatives such as eicosanoids, which serve as sensors to regulate metabolic . These ligands bind to the PPAR ligand-binding domain with generally low affinity compared to synthetic agonists, reflecting their physiological roles in responding to fluctuating levels rather than high-potency signaling. Fatty acids, including both saturated and unsaturated types, are among the most ubiquitous endogenous activators across all PPAR isoforms (α, β/δ, and γ), with binding affinities typically in the micromolar range that align with physiological concentrations during or feeding states. For PPARα, predominant in liver and involved in fatty acid oxidation, endogenous ligands include saturated and monounsaturated fatty acids such as palmitic acid and oleic acid, which bind directly and activate the receptor at concentrations relevant to lipid catabolism. Eicosanoids like leukotriene B4 (LTB4), derived from the arachidonic acid pathway via 5-lipoxygenase, exhibit higher affinity with a Kd of 60-90 nM, while prostaglandins such as PGA1, a dehydration product of PGE2, also activate PPARα with efficacy comparable to dietary fatty acids. These ligands link PPARα activation to nutrient sensing, as their biosynthesis increases during lipid mobilization, promoting genes for β-oxidation and ketogenesis. PPARα shows a relative preference for saturated fatty acids in certain contexts, such as de novo lipogenesis products, which stabilize the receptor more effectively than polyunsaturated counterparts in hepatic models. PPARβ/δ, expressed broadly in and , is activated by polyunsaturated fatty acids (PUFAs) like (DHA) from dietary omega-3 sources and endogenous arachidonic acid metabolites such as 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE), produced via 15-lipoxygenase. also serves as a for this isoform, contributing to by inducing targets like angiopoietin-like 4 (Angptl4) for regulation. These ligands, biosynthesized from membrane phospholipids during or exercise, enable PPARβ/δ to sense dietary fats and modulate uptake and oxidation without the high specificity seen in other isoforms. In contrast, PPARγ, key for and glucose metabolism in adipose and immune cells, preferentially responds to cyclopentenone prostaglandins and oxidized lipids, including 15-deoxy-Δ¹²,¹⁴-prostaglandin J2 (15d-PGJ2), a of (PGD2) via prostaglandin D synthase and non-enzymatic dehydration. 15d-PGJ2 binds with an EC₅₀ of approximately 2 μM in transcriptional assays, activating and differentiation programs at physiological levels during . Other eicosanoids like 15-hydroxyeicosatetraenoic acid (15-HETE) and 13-hydroxylinoleic acid (13-HODE), from pathways, also serve as ligands with micromolar potencies, while provides basal activation across isoforms. Natural dietary compounds such as the act as partial agonists for PPARγ, enhancing weak endogenous signals with low efficacy to support metabolic balance without full receptor stabilization. These biosynthesis pathways tie PPARγ ligands to release, underscoring their role in integrating nutrient availability with inflammatory resolution.
PPAR IsoformKey Endogenous LigandsExamples and SourcesBinding Affinity (Approximate)Biosynthesis Link
PPARαSaturated/monounsaturated fatty acids, eicosanoids, prostaglandins (dietary/endogenous), LTB4 ( via 5-LOX), PGA1 (PGE2 dehydration)Oleic acid: ~10-50 μM; LTB4: Kd 60-90 nM pathway; de novo
PPARβ/δPUFAs, hydroxy fatty acidsDHA (omega-3 from diet), 15(S)-HETE ( via 15-LOX), DHA: ~5-20 μM; 15(S)-HETE: ~10 μMMembrane phospholipid hydrolysis; dietary incorporation
PPARγOxidized prostaglandins, hydroxy lipids, fatty acids15d-PGJ2 (PGD2 metabolite), 15-HETE ( via 15-LOX), 15d-PGJ2: EC₅₀ ~2 μM; 15-HETE: ~10-30 μM/ pathways;

Synthetic Modulators

Synthetic modulators of peroxisome proliferator-activated receptors (PPARs) encompass a diverse array of designed compounds that selectively target specific isoforms or act as pan-agonists, primarily through interaction with the ligand-binding domain (LBD). These molecules typically feature an acidic head group that anchors to key residues in the LBD, facilitating helix 12 repositioning for coactivator , as observed in structures of PPARγ complexes with thiazolidinediones. This enhances binding affinity and isoform selectivity, distinguishing synthetic ligands from endogenous ones like fatty acids. Fibrates represent the primary class of synthetic for PPARα, promoting oxidation and . Fenofibrate, a widely studied , acts as a potent PPARα with high selectivity, exhibiting minimal activity on PPARγ or PPARδ at therapeutic concentrations. Similarly, Wy-14643, a prototypical , binds covalently to PPARα and induces proliferation in preclinical models, serving as a tool compound for mechanistic studies. Thiazolidinediones (TZDs) are selective PPARγ agonists developed for insulin sensitization, featuring a heterocyclic core with an acidic head for LBD engagement. Pioglitazone binds PPARγ with an of approximately 550 , showing weak cross-reactivity with PPARα ( >10 μM) and PPARδ. Rosiglitazone demonstrates higher potency for PPARγ ( ~100 ) and negligible activation of PPARα or PPARδ, underscoring its isoform selectivity in assays. For PPARβ/δ (also known as PPARδ), selective agonists include , a synthetic compound that enhances oxidation in and endurance in models. However, was found to induce cancers in preclinical studies, leading to the discontinuation of its clinical . Emerging PPAR antagonists provide tools for dissecting receptor functions and potential therapeutics. GW9662 is an irreversible, selective PPARγ antagonist that covalently modifies Cys285 in the LBD, blocking agonist-induced and tumor growth in models without affecting PPARα or PPARδ. Recent 2025 studies highlight PPARα antagonists, such as amezalpat (TPST-1120), which inhibit oxidation in cancer cells, reducing tumor growth in xenograft models of solid tumors by disrupting metabolic support for proliferation. Pan-PPAR modulators, including agonists, target multiple isoforms for broader therapeutic effects. Elafibranor functions as a PPARα/δ agonist, with preferential activation of PPARα (EC50 ~23 ) over PPARγ, modulating lipid and in non-alcoholic (NASH) models through LBD binding that stabilizes active conformations. Its structure incorporates an acidic head and flexible linker, enabling fit across the larger PPAR LBD volume for selectivity.

Clinical Significance

Therapeutic Applications

Peroxisome proliferator-activated receptor (PPAR) agonists have established therapeutic roles in managing metabolic disorders through their activation of specific receptor subtypes. Fibrates, selective PPARα agonists such as fenofibrate and , are approved for treating by lowering triglycerides and raising cholesterol levels, thereby reducing cardiovascular risk in patients with . Thiazolidinediones (TZDs), including pioglitazone and , are PPARγ agonists approved for mellitus, where they enhance insulin sensitivity, promote , and improve glycemic control. In 2024, dual PPAR agonists expanded treatment options for liver diseases. Elafibranor, a dual PPARα/δ agonist, received accelerated U.S. (FDA) approval for (PBC) in patients with inadequate response to , demonstrating significant improvements in biochemical markers of such as levels. Similarly, seladelpar, a selective PPARδ agonist, was approved for PBC by the FDA in 2024 and received conditional approval from the (EMA) in February 2025, highlighting the potential of PPAR modulation in cholestatic liver conditions. Investigational dual PPAR agonists, such as (a PPARα/γ modulator), are under evaluation for non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Phase 2 and 3 trials have shown reduces liver fat content, improves levels, and ameliorates in NAFLD patients, with doses of 2-4 mg daily yielding significant histological improvements without major adverse events. For PPARβ/δ, agonists like elafibranor are being explored beyond PBC for metabolic conditions involving muscle preservation, with preclinical evidence suggesting protection against muscle wasting in models through enhanced oxidation and mitochondrial function. Efficacy data for TZDs underscore their clinical impact, with pioglitazone typically reducing HbA1c by 1-2% in patients over 6-12 months, alongside improvements in lipid profiles. However, PPAR agonists carry risks, including fluid retention leading to and increased incidence, particularly with pioglitazone in patients with preexisting cardiac conditions; monitoring and dose adjustments are recommended to mitigate these effects. Recent advances include PPAR antagonists entering early clinical trials for . Amezalpat, a selective PPARα antagonist, received FDA orphan drug designation in 2025 for advanced and is in phase 1/2 trials, where preclinical data indicate tumor suppression via disruption of and enhancement of anti-tumor immunity; it also received orphan drug designation from the in June 2025. Similarly, FX-909, a PPARγ , is in phase 1b trials as of 2025 for urothelial , with phase 1a data showing early clinical efficacy including objective responses in patients with advanced urothelial .

Role in Diseases

Mutations in the PPARG gene, encoding PPARγ, are associated with familial partial type 3 (FPLD3), a condition characterized by partial loss of and severe metabolic disturbances including and . Loss-of-function variants in PPARγ impair differentiation and lipid storage, leading to ectopic fat accumulation and features. Similarly, PPARα deficiency accelerates the progression of non-alcoholic (NAFLD) by disrupting hepatic lipid oxidation and exacerbating and . Hepatocyte-specific deletion of PPARα in mouse models promotes NAFLD development through impaired β-oxidation and increased susceptibility to diet-induced liver injury. In cancer, overexpression of PPARβ/δ in colon tumors enhances tumorigenesis by promoting and survival pathways. Studies in and models show elevated PPARβ/δ levels in tissues compared to mucosa, correlating with increased tumor susceptibility and . Recent research highlights PPARβ/δ dysregulation in the , where it modulates immune evasion, though data remain limited and coverage of stromal interactions is outdated. Antagonizing PPARβ/δ has emerged as a potential anticancer approach in 2025 strategies, targeting immune modulation to reshape the suppressive tumor milieu. Dysregulation of PPARγ contributes to inflammatory diseases, with reduced expression observed in (IBD) and . In IBD, decreased colonic PPARγ levels correlate with heightened signaling and mucosal inflammation, exacerbating barrier dysfunction. In , PPARγ deficiency in endothelial and cells promotes formation and plaque progression by impairing anti-inflammatory responses. PPARδ plays a protective role in liver diseases, where its downregulation facilitates through enhanced SMAD3 signaling and activation, as noted in 2024 reviews. Beyond metabolic and inflammatory conditions, PPARγ dysregulation influences neurological and adipose pathologies. In , PPARγ activation offers by mitigating amyloid-β-induced and , with 2024 preclinical trials demonstrating cognitive benefits in mouse models. In , PPARγ deacetylation is essential for remodeling; hyperacetylation in macrophages promotes visceral fat infiltration and metabolic dysfunction, as shown in 2023 studies. Research gaps persist in understanding PPAR's role in , with limited linking PPAR modulation to outcomes in conditions like , where 2023 analyses noted potential effects but lacked robust clinical validation.

References

  1. [1]
    Peroxisome proliferator-activated receptors and their ligands
    Feb 14, 2014 · Peroxisome proliferator-activated receptors are ligand-activated transcription factors that regulate genes important in cell differentiation and various ...Ppar Structure And Function · Types Of Ppars And Their... · Ppar Ligands
  2. [2]
    The peroxisome proliferator-activated receptor: A family of nuclear ...
    The present review critically analyzes the protective and detrimental effect of PPAR agonists in dyslipidemia, diabetes, adipocyte differentiation ...
  3. [3]
    Recent Insights into the Role of PPARs in Disease - PMC
    Jun 7, 2023 · Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that play important roles in cell proliferation, differentiation, ...<|control11|><|separator|>
  4. [4]
    Chemistory of Fibrates - PMC - PubMed Central - NIH
    Since the description of the synthetic chemical clofibrate in 1962, various derivatives of fibrates with a diversity of chemical structures have been developed.Missing: 1960s | Show results with:1960s
  5. [5]
    The clofibrate saga: a retrospective commentary - PMC - NIH
    After many years of study clofibrate, the first of the fibrates, seemed in the 1960s, to be an effective means of lowering raised plasma cholesterol.
  6. [6]
    The PPAR Story | News & Announcements - Cayman Chemical
    In 1990, Isabelle Issemann and Stephen Green reported the cloning of a novel member of the steroid hormone receptor superfamily that was activated by ...Missing: history | Show results with:history
  7. [7]
    [DOC] Clofibrate molecule was discovered by Thorp and Waring in 1962 ...
    Fibrates and TZDs were developed decades ago for the treatment of dyslipidemia and type 2 diabetes. ... Mechanism of action of fibrates on lipid and lipoprotein ...Missing: 1960s | Show results with:1960s
  8. [8]
    Activation of a member of the steroid hormone receptor superfamily ...
    The receptor homologue is activated by a diverse class of rodent hepatocarcinogens that causes proliferation of peroxisomes.
  9. [9]
    cDNA Cloning and Characterization of the Transcriptional ... - PubMed
    cDNA Cloning and Characterization of the Transcriptional Activities of the Hamster Peroxisome Proliferator-Activated Receptor haPPAR Gamma. Gene. 1995 Sep ...
  10. [10]
    PPARs: Nuclear Receptors Controlled by, and Controlling, Nutrient ...
    Taken together, PPARα or NR1C, PPARδ (also known as PPARβ) or NR1C2, and PPARγ or NR1C3 constitute group C in subfamily 1 of the superfamily of nuclear ...
  11. [11]
    Peroxisome Proliferator-Activated Receptors: Nuclear Control of ...
    They were named PPARα (NR1C1), PPARβ (NR1C2), and PPARγ (NR1C3) when the group of three was originally found in Xenopus (15 ), shortly after the ...
  12. [12]
    Peroxisome Proliferator - an overview | ScienceDirect Topics
    There are three PPAR subtypes (α, γ, and δ). PPARs were originally identified by their ability to induce peroxisome proliferation, and this led to the current ...
  13. [13]
    tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat
    PPAR-alpha is highly expressed in hepatocytes, cardiomyocytes, enterocytes, and the proximal tubule cells of kidney. PPAR-beta is expressed ubiquitously and ...
  14. [14]
    PPARs: diverse regulators in energy metabolism and ... - Nature
    Jan 26, 2010 · Through their distinct yet overlapping functions and tissue distribution, the three PPARs act as fatty acid sensors to control many metabolic ...Pparγ Is A Master Regulator... · Pparα Regulates Hepatic... · Pparδ In The Skeletal...
  15. [15]
    Transcriptional and epigenetic regulation of PPARγ expression ...
    May 29, 2014 · It is expressed as two isoforms, PPARγ1 and PPARγ2, as a result of different promoter usage and alternative splicing [16, 17]. The PPARγ1 ...
  16. [16]
    170998 - PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR ...
    Gene Structure​​ Vohl et al. (2000) determined that the PPARA gene spans 83.7 kb and contains 8 exons.
  17. [17]
    The role of Peroxisome Proliferator-Activated Receptors (PPAR) in ...
    PPARs are expressed in immune cells and have an emerging critical role in immune cell differentiation and fate commitment.
  18. [18]
    [PDF] Peroxisome Proliferator-Activated Receptors - Endocrine Society
    In 1990, Issemann and Green reported the cloning and initial characterization ... in rodent models of hyperlipidemia prior to the discovery of the PPARs.
  19. [19]
    Review of the Structural and Dynamic Mechanisms of PPARγ Partial ...
    The PPAR ligand binding domain near the β-sheet, proximal loops, and small helices (H2 and H2′) contacts the RXR DNA binding domain (rather than the ligand ...
  20. [20]
    Heterodimer formation with retinoic acid receptor RXRα modulates ...
    Heterodimers formed between PPARγ and its obligatory dimer partner retinoid X receptor α (RXRα) can activate transcription of target genes in response to ...
  21. [21]
    Allosteric Pathways in the PPARγ-RXRα nuclear receptor complex
    Jan 29, 2016 · Asymmetry in the ppar gamma/rxr alpha crystal structure reveals the molecular basis of heterodimerization among nuclear receptors. Mol. Cell ...
  22. [22]
    Nuclear receptor corepressors and PPARγ - PMC - NIH
    On these genes, corepressors repress PPARγ activity in the absence of ligand. The presence of ligands such as TZDs causes release of corepressors and ...Missing: upon | Show results with:upon
  23. [23]
    Ligand binding and co-activator assembly of the peroxisome ...
    Here we present the X-ray crystal structure of the human apo-PPAR-gamma ligand-binding domain (LBD), at 2.2 A resolution; this structure reveals a large ...
  24. [24]
    Coactivators in PPAR-Regulated Gene Expression - PMC
    Most coactivators possess one or more LXXLL motifs (L: leucine and X: any amino acid) some of which may make contact with a coactivator-binding groove in the ...
  25. [25]
    An Ideal PPAR Response Element Bound to and Activated by PPARα
    Aug 4, 2015 · The consensus sequence of PPRE/DR1 is composed of 2 core hexad sequences directionally aligned and separated by a single nucleotide spacer ( ...
  26. [26]
    Partial Agonists Activate PPARγ Using a Helix 12 Independent ...
    Oct 16, 2007 · These transcriptional coactivators bind to the receptor complex, modify local chromatin structure, and recruit the transcription machinery to ...
  27. [27]
    Functional and Structural Insights into the Human PPARα/δ/γ ... - MDPI
    Jul 29, 2023 · We have previously obtained the PPARα-LBD–clofibric acid (a low affinity PPARα ligand with an EC50 value of 574 µM in the PGC1α recruitment ...
  28. [28]
    Gene Expression Changes Induced by PPAR Gamma Agonists in ...
    Marked tissue-differences are observed in the response to glitazones: relative to the liver and the skeletal muscle, PPARγ is 10- to 30-fold higher expressed in ...
  29. [29]
    Differential expression of peroxisome proliferator-activated receptors ...
    Aug 7, 2025 · PPAR-beta is expressed ubiquitously and often at higher levels than PPAR-alpha and -gamma. PPAR-gamma is expressed predominantly in adipose ...<|control11|><|separator|>
  30. [30]
    Unraveling the role of peroxisome proliferator-activated receptor-β/δ ...
    Oct 7, 2019 · In human tissues, relative expression of PPARβ/δ protein is very high to moderate in colon, skin, breast, thyroid gland, gall bladder, adrenal ...
  31. [31]
    Localization of PPAR isotypes in the adult mouse and human brain
    Jun 10, 2016 · Moreover, PPARα, PPARβ/δ and PPARγ show unique tissue distribution in the peripheral nervous system and select regions of the central nervous ...
  32. [32]
    Ligand-dependent nucleo-cytoplasmic shuttling of peroxisome ...
    We show here that PPARα and PPARγ dynamically shuttle between nucleus and cytoplasm, although they constitutively and predominantly appear in nucleus.
  33. [33]
    Effects of peroxisome proliferator-activated receptor δ on ... - PNAS
    Targeting of the nuclear prostaglandin receptor peroxisome proliferator-activated receptor δ (PPARδ) by homologous recombination results in placental defects.Missing: PPARβ/ | Show results with:PPARβ/
  34. [34]
    Deficiency of PPARβ/δ in the Epidermis Results in Defective ...
    The purpose of this study was to determine if there are any changes in skin physiology, ultrastructure, epidermal lipid synthesis, as well as cutaneous ...Missing: lethal | Show results with:lethal
  35. [35]
    PPAR gamma is required for placental, cardiac, and adipose tissue ...
    The nuclear hormone receptor PPAR gamma promotes adipogenesis and macrophage differentiation and is a primary pharmacological target in the treatment of type ...Missing: adipocyte | Show results with:adipocyte
  36. [36]
    alpha and PPAR-gamma in human atherosclerosis - PubMed
    Jul 15, 2010 · The mRNA levels of both PPARs increased significantly in atherosclerosis and tended to increase in proportion to the severity of the lesion, ...
  37. [37]
    PPARγ in coronary atherosclerosis: in vivo expression pattern and ...
    Result: PPARγ expression was higher in coronary plaques and peripheral blood monocytes of statin-treated patients, and it significantly increased in monocytes ...
  38. [38]
    Hypoxia exacerbates nonalcoholic fatty liver disease via the HIF-2α ...
    Oct 1, 2019 · HIF-2α overexpression under hypoxic conditions suppressed PPARα, leading to PGC-1α, NRF-1, ESRRα downregulation, and mitochondrial impairment.
  39. [39]
    TIR8 protects against nonalcoholic steatohepatitis by antagonizing ...
    Jun 6, 2024 · TIR8 protects against nonalcoholic steatohepatitis by antagonizing lipotoxicity-induced PPARα downregulation and reducing the sensitivity of ...
  40. [40]
    A paradigm for gene regulation: inflammation, NF-kappaB and PPAR
    First, NF-kappaB activity is regulated by cytoplasmic degradation of the IkappaB inhibitor and nuclear translocation.
  41. [41]
    Down-regulation of STAT5b transcriptional activity by ligand ...
    Previous studies have shown that growth hormone (GH)-activated STAT5b can inhibit PPAR-regulated transcription. Here, we show that this inhibitory cross-talk ...
  42. [42]
    STAT6 transcription factor is a facilitator of the nuclear receptor ...
    Nov 24, 2010 · STAT6 acts as a facilitating factor for PPARγ by promoting DNA binding and consequently increasing the number of regulated genes and the magnitude of responses.Missing: STAT | Show results with:STAT
  43. [43]
    Epigenetic regulation of macrophage polarization and inflammation ...
    We find that inhibiting DNA methylation pharmacologically using 5-aza-2'-deoxycytidine or genetically by DNA methyltransferase 1 (DNMT1) deletion promotes ...
  44. [44]
    Epigenetic Regulation of Peroxisome Proliferator-Activated ...
    May 31, 2021 · This study demonstrated that HFD-induction of pparγ DNA promoter demethylation increased the expression of PPARγ and its target genes, vldlr and ...
  45. [45]
    Age-related decrease in expression of peroxisome proliferator ...
    The decrease in the hepatic PPARalpha expression is probably directly related to the lipid metabolic disturbances observed in old animals.
  46. [46]
    PPARα: An emerging target of metabolic syndrome ... - Frontiers
    PPARα is highly expressed in hepatocytes, cardiomyocytes, proximal renal tubular cells, and brown adipocytes. PPARβ/δ is more ubiquitous but mainly found in ...
  47. [47]
    Integrated physiology and systems biology of PPARα - ScienceDirect
    The first identified target gene of PPARα was Acyl-CoA oxidase (Acox1), which encodes the first enzyme in peroxisomal long-chain fatty acid oxidation [2], [5].
  48. [48]
    PPAR-α–Null Mice Are Protected From High-Fat Diet–Induced ...
    Dec 1, 2001 · PPAR-α–null mice (3) exhibit a reduced capacity to metabolize long-chain fatty acids (4,5), which likely contributes to dyslipidemia (6) and ...RESEARCH DESIGN AND... · RESULTS · DISCUSSION
  49. [49]
    PPARγ signaling and metabolism: the good, the bad and the future
    In adipose tissue, PPARγ upregulates genes involved in glucose uptake and also controls the expression of adipocyte-secreted factors, such as adiponectin, that ...
  50. [50]
    Peroxisome Proliferator-Activated Receptor Delta - PubMed Central
    PPARδ stimulates the expression of genes involved in (a) increasing of lipid oxidation (fatty acid binding protein 3 (FABP3) and CPT1) and (b) reducing ...
  51. [51]
    AMPK and PPARδ Agonists Are Exercise Mimetics - ScienceDirect
    Aug 8, 2008 · We found that PPARβ/δ agonist and exercise training synergistically increase oxidative myofibers and running endurance in adult mice.
  52. [52]
    Cross-Talk between Peroxisome Proliferator-Activated Receptor ...
    These data suggest that PPARα activation can suppress LXR-SREBP-1c pathway through reduction of LXR/RXR formation, proposing a novel transcription factor cross- ...
  53. [53]
  54. [54]
    Parsing the Role of PPARs in Macrophage Processes - Frontiers
    One study demonstrated that PPARγ reduced the secretion of influenza-induced proinflammatory cytokines TNF-a, IL-8, and RANTES in humans (85). PPARγ activation ...
  55. [55]
    PPARγ and the Innate Immune System Mediate the Resolution of ...
    PPARγ alters macrophage trafficking, increases efferocytosis and phagocytosis, and promotes alternative M2 macrophage activation. There are also roles for this ...
  56. [56]
    Molecular mechanism of PPARα action and its impact on lipid ...
    PPARα negatively regulates pro-inflammatory and acute phase response (APR) ... PPARα in acute and chronic liver inflammation. PPARα and acute hepatic ...Review · Pparα In The Regulation Of... · Pparα In Acute And Chronic...
  57. [57]
    PPARs at the crossroads of T cell differentiation and type 1 diabetes
    Meanwhile, the activation of PPARβ/δ employs an anti-inflammatory role, leading to abridged production of proinflammatory cytokines, contributing important ...
  58. [58]
    Critical roles of PPARβ/δ in keratinocyte response to inflammation
    Activated PPARβ/δ regulates the expression of genes associated with apoptosis resulting in an increased resistance of cultured keratinocytes to cell death.
  59. [59]
    PPARs and molecular mechanisms of transrepression - PMC
    PPARγ also negatively modulates the AP-1 and NF-κB pro-inflammatory pathways. In human vascular endothelial cells PPARγ has been shown to alter c-Jun ...
  60. [60]
    Nuclear Receptors and Inflammation Control: Molecular ...
    Similar to PPARγ, PPARα activation also decreases NF-κB and AP-1 activities in liver and endothelial cells. Three major mechanisms have been described for the ...Ppars · Nuclear Receptor 4a Family · Perspectives
  61. [61]
    The role of peroxisome proliferator-activated receptors in the ...
    Feb 17, 2023 · ... PPARγ is inversely related to pulmonary fibrosis caused by chronic inflammation in COVID-19 patients (147, 171–173). It has been also ...
  62. [62]
    Molecular scanning of the human PPARα gene
    Jun 1, 2000 · The exact length in base pairs of the exons is indicated in parenthesis. ... Using information available from GenBank, we have determined the exon ...Missing: PPARA | Show results with:PPARA<|control11|><|separator|>
  63. [63]
    Entry - *601487 - PEROXISOME PROLIFERATOR ... - OMIM
    (1995) demonstrated that PPAR-gamma-2 regulates adipocyte expression of the phosphoenolpyruvate carboxykinase gene (PCK1, 261680; PCK2, 261650). Lowell ...
  64. [64]
    Evolution of the nuclear receptor gene superfamily. - EMBO Press
    Mar 1, 1992 · These data suggest a complex evolutionary history for nuclear receptor genes in which gene duplication events and swapping between domains of ...
  65. [65]
    Evolutionary Genomics of Nuclear Receptors: From Twenty-Five ...
    The nuclear receptor distribution was also shaped by periods of gene duplication, essentially in vertebrates, as well as a lineage-specific duplication burst ...
  66. [66]
    PPARs are a unique set of fatty acid regulated transcription factors ...
    A series of gene duplication events during early vertebrate evolution produced (among other subfamilies) the three members of the PPAR subfamily.
  67. [67]
    Paternal hyperglycemia induces transgenerational inheritance of ...
    Paternal hyperglycemia may induce epigenetic modification of Ppara in offspring's liver, probably through interaction with SP1 binding, causing impaired lipid ...<|separator|>
  68. [68]
    PPARγ and NF-κB regulate the gene promoter activity of their ...
    Our novel findings include PPAR and NF-κB regulation of the TNIP1 promoter via elements within the distal region of the defined promoter; additionally, we ...
  69. [69]
    Divergence between human and murine peroxisome proliferator ...
    Although there exists remarkable overlap in the activities of PPARα across species, human and mouse PPARα proteins promote transcription to a different extent ...Missing: genomics | Show results with:genomics
  70. [70]
    PPARα-Humanized Mouse: A Model to Investigate Species ...
    Abstract. To determine the impact of the species difference between rodents and humans in response to peroxisome proliferators (PPs) mediated by peroxisome.
  71. [71]
    PPARG (Pro12Ala) genetic variant and risk of T2DM - Nature
    Jul 29, 2020 · The present study was focused on assessing the association between Pro12Ala variation in the PPARG and T2DM risk through a comprehensive meta-analysis.
  72. [72]
    Pro12Ala Variant of the PPARG Gene Is a Risk Factor for ...
    The 12Ala variant of the PPARγ protein has a 30–50% lower biological activity (dependent on the assay), compared with the 12Pro protein (7), and therefore ...
  73. [73]
    Association Between the PPARA L162V Polymorphism and Plasma ...
    A L162V polymorphism at the PPARA locus has been associated with alterations in lipid and apolipoprotein concentrations. We studied the association among lipids ...Missing: Leu162Val | Show results with:Leu162Val
  74. [74]
    604367 - LIPODYSTROPHY, FAMILIAL PARTIAL, TYPE 3; FPLD3
    Familial partial lipodystrophy type 3 (FPLD3) is an autosomal dominant disorder characterized by marked loss of subcutaneous fat from the extremities.
  75. [75]
    A critical role for the peroxisome proliferator-activated receptor α ...
    These results indicate that fasting induces a significant derangement in hepatic lipid balance in the PPARα-null mice. Figure 1. Open in Viewer Accumulation of ...Missing: mutations | Show results with:mutations
  76. [76]
    The Effect of PPARγ rs1801282 Variant on Mortality Risk Among ...
    Although the frequency of the PPARG Pro12Ala allele is 4% in Asians, it is 11% in Caucasians. Besides the possible differences in ethnicity, the enrolled ...
  77. [77]
    Studies of the Pro12Ala Polymorphism of the PPAR-γ Gene in the ...
    The allelic frequency of the Pro12Ala polymorphism in the total study population of 2245 subjects was 14.0% (95% confidence interval, 13.0–15.0%). Five ...
  78. [78]
    PPARs and lipid ligands in inflammation and metabolism - PMC
    Natural ligands for PPARs consist of fatty acids and cyclooxygenase-derived eicosanoids and prostaglandins that bind to PPARs with relatively low affinity ...
  79. [79]
    Fatty acids and eicosanoids regulate gene expression through direct ...
    We show that certain mono- and polyunsaturated fatty acids bind directly to PPARα and PPARγ at physiological concentrations.Results · Fas Bind To Pparα And... · Eicosanoids Are Subtype...
  80. [80]
    Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids ...
    Thus, PPARα activators such as PGA1, PGA2, PGB2, PGD2, and 15d-J2 may be inactive precursors that are metabolized to PPARα ligands. PPARα and -δ Possess ...
  81. [81]
    “New” hepatic fat activates PPARα to maintain glucose, lipid, and ...
    New fat synthesized by FAS may be the preferred endogenous PPARα activator. ... PPARα with an affinity similar to that of some unsaturated fatty acids (Xu ...
  82. [82]
  83. [83]
    Natural product agonists of peroxisome proliferator-activated ...
    Agonists of the nuclear receptor PPARγ are therapeutically used to combat hyperglycaemia associated with the metabolic syndrome and type 2 diabetes.Part Of The Special Issue... · 2. Pparγ And The Metabolic... · 3. Pparγ Activation By...<|control11|><|separator|>
  84. [84]
    Novel PPAR-γ agonists as potential neuroprotective agents against ...
    Oct 21, 2024 · Glitazones possess three essential structural features in their binding portion: an acidic head, an aromatic trunk with two carbon linkers, and ...
  85. [85]
    Fenofibric Acid, an Active Form of Fenofibrate, Increases ...
    Several subtype-specific synthetic compounds have been developed for clinical use, including fibric acids (PPARα agonist) and glitazones (PPARγ agonist).Missing: paper | Show results with:paper
  86. [86]
    Molecular Mechanism of Peroxisome Proliferator-Activated Receptor ...
    Less clear are determinants that influence affinity of ligands for PPARα. Several fibrates—that are PPARα agonists—are used to treat dyslipidemia and ...
  87. [87]
    Lobeglitazone - Diabetes & Metabolism Journal
    Apr 19, 2021 · Other TZDs, rosiglitazone and pioglitazone ... The EC50 values of rosiglitazone and pioglitazone for PPARγ were 0.1076 and 0.5492 μM, respectively ...
  88. [88]
    Article Peroxisome-Proliferator-Activated Receptor δ Activates Fat ...
    Apr 18, 2003 · Consistent with our observations in adipocytes, the PPARδ agonist GW501516 significantly increases fatty acid oxidation in myotubes, with no ...
  89. [89]
    PPARδ, a Potential Therapeutic Target for Heart Disease - PMC
    Another study shows that PPARδ ligands (GW0742 or GW501516) treatment in mice leads to rapid development of cardiac hypertrophy and angiogenesis, presumably via ...
  90. [90]
    GW9662, a potent antagonist of PPARγ, inhibits growth of breast ...
    This study demonstrated that the potent, irreversible and selective PPARγ antagonist GW9662 prevented activation of PPARγ and inhibited growth of human mammary ...
  91. [91]
    A phase 3 randomized, placebo (Pbo)-controlled study of amezalpat ...
    May 28, 2025 · Amezalpat (TPST-1120) is an investigational PPARα antagonist that inhibits FAO, targeting the bioenergetic requirements of cancer cells and ...
  92. [92]
    Elafibranor, an Agonist of the Peroxisome Proliferator−Activated ...
    Elafibranor (GFT505) is a dual PPARα/δ agonist that has demonstrated efficacy in disease models of nonalcoholic fatty liver disease (NAFLD)/NASH and liver ...
  93. [93]
    Functional and Structural Insights into the Human PPARα/δ/γ ... - NIH
    Jul 29, 2023 · Elafibranor, the PPARα/δ dual agonist developed by Genfit (Loos, France), has been abandoned at the phase 3 clinical trial against NAFLD/NASH ...
  94. [94]
    Fibrates, glitazones, and peroxisome proliferator-activated receptors
    Several decades ago, fibrates were approved for the treatment of dyslipidemia, whereas thiazolidinediones were screened in animal models to improve glucose ...
  95. [95]
    PPAR Agonists and Metabolic Syndrome: An Established Role?
    Fibrates, activators of the PPAR system, mainly PPAR-α, have shown significant benefit in clinical trials of CV prevention, i.e., reducing the occurrence of ...
  96. [96]
    Peroxisome Proliferator–Activated Receptors as Transcriptional ...
    PPARα, the first PPAR cloned, plays an important role in regulating the β ... All PPAR isoforms regulate central metabolic pathways in human physiology ...Missing: timeline | Show results with:timeline
  97. [97]
    Peroxisome proliferator-activated receptor-γ agonists and diabetes
    PPARγ agonists, like thiazolidinediones (TZDs), are used for type 2 diabetes, improving insulin sensitivity and glucose uptake, and reducing fasting insulin.
  98. [98]
    Efficacy and Safety of Elafibranor in Primary Biliary Cholangitis
    Nov 13, 2023 · Treatment with elafibranor resulted in significantly greater improvements in relevant biochemical indicators of cholestasis than placebo.Missing: lanifibranor approval
  99. [99]
    Drugs targeting peroxisome proliferator-activated receptors
    Currently, selective PPAR agonists are used to manage hyperlipidemia, type 2 diabetes mellitus (T2DM), and PBC, and dual/pan-PPAR agonists have been developed ...
  100. [100]
    PPAR agonists provide new treatment options for inflammatory liver ...
    Sep 9, 2024 · The FDA has granted accelerated approval to Gilead's PPAR-δ selective agonist seladelpar (Livdelzi) for primary biliary cholangitis (PBC).
  101. [101]
    A dual PPAR α/γ agonist for managing metabolic NAFLD - PubMed
    Apr 11, 2024 · SARO, administered at a dose of 4 mg/day, has been consistently studied in clinical trials with different time points ranging from 4 to 24 weeks ...
  102. [102]
    Saroglitazar in patients with non-alcoholic fatty liver disease ... - Nature
    Dec 3, 2020 · We conclude that Saroglitazar leads to significant improvement in transaminases, LSM, and CAP in NAFLD patients with DD.
  103. [103]
    PPARs and Microbiota in Skeletal Muscle Health and Wasting - MDPI
    Herein, we review functions of PPARs in skeletal muscle and their interaction with the gut microbiota in the context of muscle wasting.Ppars And Microbiota In... · 3. Roles Of Ppars In Muscle · 4. Ppars In Muscle Wasting
  104. [104]
    A multicentre, double‐blind, placebo‐controlled, randomized ...
    Feb 29, 2024 · Pioglitazone administration resulted in a significant reduction in HbA1c levels (from 7.80% ± 0.72% to 7.27% ± 0.82%) compared with placebo ( ...
  105. [105]
    Efficacy and Safety of Pioglitazone Monotherapy in Type 2 Diabetes ...
    Mar 29, 2019 · Despite the favourable effect, pioglitazone treatment in patients with underlying heart disease may be harmful since pioglitazone-mediated ...
  106. [106]
    Rethinking pioglitazone as a cardioprotective agent
    May 18, 2021 · Although pioglitazone might worsen HF by inducing sodium-water retention and oedema, there are some counterarguments [10] : first, it does not ...
  107. [107]
    Tempest Receives Orphan Drug Designation from the U.S. Food ...
    Jan 6, 2025 · Amezalpat is an oral, small molecule, selective PPAR⍺ antagonist. Data suggest that amezalpat treats cancer by targeting tumor cells ...
  108. [108]
    Peroxisome proliferator-activated receptor antagonists as emerging ...
    PPARα predominantly modulates lipid metabolism and may either inhibit or promote tumorigenesis, depending on the cancer context. PPARβ/δ displays dual functions ...
  109. [109]
    Phase 1B trial of PPARG inhibitor launches in urothelial carcinoma
    Aug 5, 2025 · FX-909 is an investigational small molecule PPARG inhibitor for patients with locally advanced or metastatic urothelial carcinoma.
  110. [110]
    Peroxisome proliferator-activated receptor antagonists as emerging ...
    Jun 16, 2025 · These antagonists selectively modulate oncogenic pathways by disrupting metabolic and signaling networks that support tumor growth, survival, and metastasis.
  111. [111]
  112. [112]
    Genomic and Bioinformatics Analysis of Familial Partial ... - PubMed
    Nov 10, 2024 · FPLD type 3 is caused by mutations in the PPARγ gene, which encodes for the protein peroxisome proliferator-activated receptor gamma (PPARγ). We ...
  113. [113]
    The lack of PPARα exacerbated the progression of non-alcoholic ...
    Apr 2, 2024 · Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, affecting 12.5% of the global population (1). Non- ...
  114. [114]
    Hepatocyte-specific deletion of Pparα promotes NAFLD in ... - Nature
    Apr 16, 2020 · Non alcoholic fatty liver disease (NAFLD) has become a major public health concern worldwide. NAFLD ranges from benign steatosis to non ...
  115. [115]
    Regulatory role of PPAR in colorectal cancer | Cell Death Discovery
    Jan 28, 2025 · PPARβ/δ is widely distributed in various tissues, with the highest expression levels observed in colonic epithelial cells, skin, and adipocytes.
  116. [116]
    The role of peroxisome proliferator-activated receptors in the tumor ...
    PPARα inhibits cancer by inhibiting angiogenesis and promotes cancer through escape from immunity and growth of the carcinoma. PPARγ inhibits cancer by ...
  117. [117]
    PPARγ in Inflammatory Bowel Disease - PMC - PubMed Central
    PPARγ receptors are widely and highly expressed in the colon, being a key regulator factor of bacteria-induced mucosal inflammation.
  118. [118]
    Disruption of Endothelial Peroxisome Proliferator-Activated ...
    Recently, conditional disruption of PPARγ in macrophages demonstrated an increase in atherosclerosis under conditions of mild and severe hypercholesterolemia.
  119. [119]
    PPARβ/δ attenuates hepatic fibrosis by reducing SMAD3 ... - PubMed
    Aug 18, 2024 · The PPARβ/δ agonist GW501516 completely prevented glucose intolerance and peripheral insulin resistance, blocked the accumulation of collagen in the liver,
  120. [120]
    Novel PPAR-γ agonists as potential neuroprotective agents against ...
    Studies have shown that PPAR-γ activation has neuroprotective effects, can potentially reduce inflammation and oxidative stress, and stimulates ...
  121. [121]
    Acetylation of PPARγ in macrophages promotes visceral fat ...
    Here, we report that PPARγ acetylation in macrophages promotes their infiltration into adipose tissue, exacerbating metabolic dysregulation.
  122. [122]
    Fenofibrate for COVID-19 and related complications as an approach ...
    Aug 8, 2022 · These findings imply that PPAR-α agonists like fenofibrate suppress the immune system and may increase the risk of herpes virus infection. As a ...