Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Clin Res Hepatol Gastroenterol ; 48(6): 102343, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641250

RESUMEN

Various liver diseases pose great threats to humans. Although the etiologies of these liver diseases are quite diverse, they share similar pathologic phenotypes and molecular mechanisms such as oxidative stress, lipid and glucose metabolism disturbance, hepatic Kupffer cell (KC) proinflammatory polarization and inflammation, insulin resistance, and hepatic stellate cell (HSC) activation and proliferation. Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is expressed in various types of liver cells with relatively higher expression in KCs and HSCs. Accumulating evidence has revealed the versatile functions of PPARß/δ such as controlling lipid homeostasis, inhibiting inflammation, regulating glucose metabolism, and restoring insulin sensitivity, suggesting that PPARß/δ may serve as a potential molecular drug target for various liver diseases. This article aims to provide a concise review of the structure, expression pattern and biological functions of PPARß/δ in the liver and its roles in various liver diseases, and to discuss potential future research perspectives.


Asunto(s)
Hepatopatías , PPAR delta , PPAR-beta , Humanos , PPAR-beta/fisiología , PPAR-beta/metabolismo , PPAR delta/fisiología , PPAR delta/metabolismo , Hepatopatías/metabolismo , Hepatopatías/tratamiento farmacológico , Terapia Molecular Dirigida , Resistencia a la Insulina
2.
Trends Pharmacol Sci ; 45(1): 9-23, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38065777

RESUMEN

Peroxisome proliferator-activated receptors [PPARs; PPARα, PPARß/δ (also known as PPARδ), and PPARγ] widely recognized for their important role in glucose/lipid homeostasis, have recently received significant attention due to their additional anti-inflammatory and neuroprotective effects. Several newly developed PPAR agonists have shown high selectivity for specific PPAR isoforms in vitro and in vivo, offering the potential to achieve desired therapeutic outcomes while reducing the risk of adverse effects. In this review, we discuss the latest preclinical and clinical studies of the activation of PPARs by synthetic, natural, and isoform-specific (full, partial, and dual) agonists for the treatment of neuroinflammatory diseases, including HIV-associated neurocognitive disorders (HAND), Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and cerebral ischemia.


Asunto(s)
PPAR delta , PPAR-beta , Humanos , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/fisiología , Enfermedades Neuroinflamatorias , PPAR delta/agonistas , PPAR delta/fisiología , PPAR-beta/fisiología , PPAR alfa/agonistas , PPAR alfa/fisiología , PPAR gamma/agonistas , PPAR gamma/fisiología , Hipoglucemiantes
3.
Neuromolecular Med ; 23(1): 86-98, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33210212

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) ß/δ belongs to the family of hormone and lipid-activated nuclear receptors, which are involved in metabolism of long-chain fatty acids, cholesterol, and sphingolipids. Similar to PPAR-α and PPAR-γ, PPAR-ß/δ also acts as a transcription factor activated by dietary lipids and endogenous ligands, such as long-chain saturated and polyunsaturated fatty acids, and selected lipid metabolic products, such as eicosanoids, leukotrienes, lipoxins, and hydroxyeicosatetraenoic acids. Together with other PPARs, PPAR-ß/δ displays transcriptional activity through interaction with retinoid X receptor (RXR). In general, PPARs have been shown to regulate cell differentiation, proliferation, and development and significantly modulate glucose, lipid metabolism, mitochondrial function, and biogenesis. PPAR-ß/δ appears to play a special role in inflammatory processes and due to its proangiogenic and anti-/pro-carcinogenic properties, this receptor has been considered as a therapeutic target for treating metabolic syndrome, dyslipidemia, carcinogenesis, and diabetes. Until now, most studies were carried out in the peripheral organs, and despite of its presence in brain cells and in different brain regions, its role in neurodegeneration and neuroinflammation remains poorly understood. This review is intended to describe recent insights on the impact of PPAR-ß/δ and its novel agonists on neuroinflammation and neurodegenerative disorders, including Alzheimer's and Parkinson's, Huntington's diseases, multiple sclerosis, stroke, and traumatic injury. An important goal is to obtain new insights to better understand the dietary and pharmacological regulations of PPAR-ß/δ and to find promising therapeutic strategies that could mitigate these neurological disorders.


Asunto(s)
Enfermedades Neurodegenerativas/fisiopatología , PPAR delta/fisiología , PPAR-beta/fisiología , Antineoplásicos/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Sistemas de Liberación de Medicamentos , Células Endoteliales/metabolismo , Glioma/tratamiento farmacológico , Glioma/metabolismo , Inflamación , Metabolismo de los Lípidos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuroglía/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo , PPAR delta/agonistas , PPAR-beta/agonistas , Receptores X Retinoide/fisiología , Transducción de Señal , Transcripción Genética
4.
Exp Cell Res ; 382(1): 111455, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31163124

RESUMEN

Diabetic nephropathy (DN) is one of the most severe complications of diabetes mellitus. The pathomolecular events behind DN remain uncertain. Peroxisome proliferator-activated receptors (PPARs) play essential functions in the development of DN. Meanwhile, 20-hydroxyeicosatetraenoic acid (20-HETE) also plays central roles in the regulation of renal function. However, the relationship between PPARs and 20-HETE is rarely studied in DN. It was revealed in our study that both PPARs expression and CYP4A-20-HETE level were decreased under DN conditions in vivo and in vitro. Supplementation with bezafibrate, a PPAR pan-agonist, improved the damage of kidney in DN mice and in high glucose-induced NRK-52E cells, following the up-regulation of PPARs and the increase of CYP4A-20-HETE. PPARα antagonist (MK886), PPARß antagonist (GSK0660), and PPARγ antagonist (GW9662) reversed the protection of bezafibrate in NRK-52E, and abrogated the up-regulation of CYP4A-20-HETE produced by bezafibrate. Noteworthily, 20-HETE synthetase inhibitor, HET0016, also blocked the bezafibrate-mediated improvement of NRK-52E, and abolished the up-regulation of PPARs expression. Collectively, our data suggest that the concurrent down-regulation and interaction of PPARs and 20-HETE play crucial roles in the pathogenesis process of DN, and we provide a novel evidence that PPARs/20-HETE signaling may be served as a therapeutic target for DN patients.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Ácidos Hidroxieicosatetraenoicos/fisiología , PPAR alfa/fisiología , PPAR gamma/fisiología , PPAR-beta/fisiología , Amidinas/farmacología , Anilidas/farmacología , Animales , Línea Celular , Citocromo P-450 CYP4A/metabolismo , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/toxicidad , Ácidos Hidroxieicosatetraenoicos/biosíntesis , Indoles/farmacología , Túbulos Renales/citología , Masculino , Ratones , PPAR alfa/biosíntesis , PPAR alfa/genética , PPAR gamma/biosíntesis , PPAR gamma/genética , PPAR-beta/biosíntesis , PPAR-beta/genética , Ratas , Sulfonas/farmacología , Tiofenos/farmacología
5.
Int J Mol Sci ; 19(10)2018 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241392

RESUMEN

BACKGROUND: Peroxisome proliferator⁻activated receptor (PPAR) ß/δ, a ligand-activated transcription factor, is involved in diverse biological processes including cell proliferation, cell differentiation, inflammation and energy homeostasis. Besides its well-established roles in metabolic disorders, PPARß/δ has been linked to carcinogenesis and was reported to inhibit melanoma cell proliferation, anchorage-dependent clonogenicity and ectopic xenograft tumorigenicity. However, PPARß/δ's role in tumour progression and metastasis remains controversial. METHODS: In the present studies, the consequence of PPARß/δ inhibition either by global genetic deletion or by a specific PPARß/δ antagonist, 10h, on malignant transformation of melanoma cells and melanoma metastasis was examined using both in vitro and in vivo models. RESULTS: Our study showed that 10h promotes epithelial-mesenchymal transition (EMT), migration, adhesion, invasion and trans-endothelial migration of mouse melanoma B16/F10 cells. We further demonstrated an increased tumour cell extravasation in the lungs of wild-type mice subjected to 10h treatment and in Pparß/δ-/- mice in an experimental mouse model of blood-borne pulmonary metastasis by tail vein injection. This observation was further supported by an increased tumour burden in the lungs of Pparß/δ-/- mice as demonstrated in the same animal model. CONCLUSION: These results indicated a protective role of PPARß/δ in melanoma progression and metastasis.


Asunto(s)
Melanoma/genética , Metástasis de la Neoplasia/genética , PPAR delta/fisiología , PPAR-beta/fisiología , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Melanoma/patología , Ratones , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/patología , PPAR delta/genética , PPAR delta/metabolismo , PPAR-beta/genética , PPAR-beta/metabolismo
6.
Neuropharmacology ; 140: 150-161, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30086290

RESUMEN

Activation of peroxisome proliferator-activated receptor beta/delta (PPAR-ß/δ), a nuclear receptor acting as a transcription factor, was shown to be protective in various models of neurological diseases. However, there is no information about the role of PPAR-ß/δ as well as its molecular mechanisms in neonatal hypoxia-ischemia (HI). In the present study, we hypothesized that PPAR-ß/δ agonist GW0742 can activate miR-17-5p, consequently inhibiting TXNIP and ASK1/p38 pathway leading to attenuation of apoptosis. Ten-day-old rat pups were subjected to right common carotid artery ligation followed by 2.5 h hypoxia. GW0742 was administered intranasally 1 and 24 h post HI. PPAR-ß/δ receptor antagonist GSK3787 was administered intranasally 1 h before and 24 h after HI, antimir-17-5p and TXNIP CRISPR activation plasmid were administered intracerebroventricularly 24 and 48 h before HI, respectively. Brain infarct area measurement, neurological function tests, western blot, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), Fluoro-Jade C and immunofluorescence staining were conducted. GW0742 reduced brain infarct area, brain atrophy, apoptosis, and improved neurological function at 72 h and 4 weeks post HI. Furthermore, GW0742 treatment increased PPAR-ß/δ nuclear expression and miR-17-5p level and reduced TXNIP in ipsilateral hemisphere after HI, resulting in inhibition of ASK1/p38 pathway and attenuation of apoptosis. Inhibition of PPAR-ß/δ receptor and miR-17-5p and activation of TXNIP reversed the protective effects. For the first time, we provide evidence that intranasal administration of PPAR-ß/δ agonist GW0742 attenuated neuronal apoptosis at least in part via PPAR-ß/δ/miR-17/TXNIP pathway. GW0742 could represent a therapeutic target for treatment of neonatal hypoxic ischemic encephalopathy (HIE).


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/fisiología , Hipoxia-Isquemia Encefálica/fisiopatología , MicroARNs/fisiología , PPAR delta/fisiología , PPAR-beta/fisiología , Animales , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/patología , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Neuronas/patología , PPAR delta/agonistas , PPAR delta/antagonistas & inhibidores , PPAR delta/biosíntesis , PPAR-beta/agonistas , PPAR-beta/antagonistas & inhibidores , PPAR-beta/biosíntesis , Ratas , Transducción de Señal/fisiología , Sulfonas/farmacología , Tiazoles/farmacología , Tiazoles/uso terapéutico
7.
J Dairy Res ; 85(3): 295-302, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29941059

RESUMEN

The hypothesis of the study was that inhibition of PPARß/δ increases glucose uptake and lactose synthesis in bovine mammary epithelial cells by reducing the expression of the glucose transporter mRNA destabiliser calreticulin. Three experiments were conducted to test the hypothesis using immortalised bovine mammary alveolar (MACT) and primary bovine mammary (PBMC) cells. In Experiment 1, the most effective dose to inhibit PPARß/δ activity among two synthetic antagonists (GSK-3787 and PT-s58) was assessed using a gene reporter assay. In Experiment 2, the effect on glucose uptake and lactose synthesis was evaluated by measuring glucose and lactose in the media and expression of related key genes upon modulation of PPARß/δ using GSK-3787, the synthetic PPARß/δ agonist GW-501516, or a combination of the two in cells cultivated in plastic. In Experiment 3, the same treatments were applied to cells cultivated in Matrigel and glucose and lactose in media were measured. In Experiment 1 it was determined that a significant inhibition of PPARß/δ in the presence or absence of fetal bovine serum was achieved with ≥ 1000 nm GSK-3787 but no significant inhibition was observed with PT-s58. In Experiment 2, inhibition of PPARß/δ had no effect on glucose uptake and lactose synthesis but they were both increased by GW-501516 in PBMC. The mRNA abundance of PPARß/δ target gene pyruvate dehydrogenase kinase 4 was increased but transcription of calreticulin was decreased (only in MACT cells) by GW-501516. Treatment with GSK-3787 did not affect the transcription of measured genes. No effects on glucose uptake or lactose synthesis were detected by modulation of PPARß/δ activity on cells cultivated in Matrigel. The above data do not provide support for the original hypothesis and suggest that PPARß/δ does not play a major role in glucose uptake and lactose synthesis in bovine mammary epithelial cells.


Asunto(s)
Bovinos , Glucosa/metabolismo , Lactosa/biosíntesis , Glándulas Mamarias Animales/metabolismo , PPAR delta/fisiología , PPAR-beta/fisiología , Animales , Benzamidas/farmacología , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , PPAR delta/antagonistas & inhibidores , PPAR-beta/antagonistas & inhibidores , Proteínas Quinasas/genética , ARN Mensajero/análisis , Sulfonas/farmacología
8.
Future Cardiol ; 13(3): 279-296, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28581362

RESUMEN

The PPARs are a subfamily of three ligand-inducible transcription factors, which belong to the superfamily of nuclear hormone receptors. In mammals, the PPAR subfamily consists of three members: PPAR-α, PPAR-ß/δ and PPAR-γ. PPARs control the expression of a large number of genes involved in metabolic homeostasis, lipid, glucose and energy metabolism, adipogenesis and inflammation. PPARs regulate a large number of metabolic pathways that are implicated in the pathogenesis of metabolic diseases such as metabolic syndrome, Type 2 diabetes mellitus, nonalcoholic fatty liver disease and cardiovascular disease. The aim of this review is to provide up-to-date information about the biochemical and metabolic actions of PPAR-ß/δ and PPAR-γ, the therapeutic potential of their agonists currently under clinical development and the cardiovascular disease outcome of clinical trials of PPAR-γ agonists, pioglitazone and rosiglitazone.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , PPAR delta/agonistas , PPAR delta/fisiología , PPAR gamma/agonistas , PPAR gamma/fisiología , PPAR-beta/agonistas , PPAR-beta/fisiología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Síndrome Metabólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Pioglitazona , Rosiglitazona , Tiazolidinedionas/uso terapéutico
9.
Biochimie ; 136: 33-41, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27939528

RESUMEN

Peroxisome Proliferator-Activated Receptor Beta (PPARß) is a transcription factor playing an important role in both muscle myogenesis and remodeling, and in inflammation. However, its role in the coordination of the transient muscle inflammation and reparation process following muscle injury has not yet been fully determined. We postulated that activation of the PPARß pathway alters the early phase of the muscle regeneration process, i.e. when immune cells infiltrate in injured muscle. Tibialis anteriors of C57BL6/J mice treated or not with the PPARß agonist GW0742 were injected with cardiotoxin (or with physiological serum for the contralateral muscle). Muscle regeneration was monitored on days 4, 7, and 14 post-injury. We found that treatment of mice with GW0742 increased, at day 4 post-damage, the recruitment of immune cells (M1 and M2 macrophages) and upregulated the expression of the anti-inflammatory cytokine IL-10 and TGF-ß mRNA. Those effects were accompanied by a significant increase at day 4 of myogenic regulatory factors (Pax7, MyoD, Myf5, Myogenin) mRNA in GW0742-treated mice. However, we showed an earlier return (7 days vs 14 days) of Myf5 and Myogenin to basal levels in GW0742- compared to DMSO-treated mice. Differential effects of GW0742 observed during the regeneration were associated with variations of PPARß pathway activity. Collectively, our findings indicate that PPARß pathway activity shortens the early phases of skeletal muscle regeneration by increasing the immune response.


Asunto(s)
Músculo Esquelético/fisiología , PPAR-beta/fisiología , Regeneración/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Inmunofenotipificación , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inmunología , PPAR-beta/genética , Linfocitos T/citología , Linfocitos T/inmunología , Tiazoles/farmacología , Transcripción Genética
10.
Am J Physiol Heart Circ Physiol ; 312(2): H189-H200, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881385

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors, which is composed of three members encoded by distinct genes: PPARα, PPARß/δ, and PPARγ. The biological actions of PPARα and PPARγ and their potential as a cardiovascular therapeutic target have been extensively reviewed, whereas the biological actions of PPARß/δ and its effectiveness as a therapeutic target in the treatment of hypertension remain less investigated. Preclinical studies suggest that pharmacological PPARß/δ activation induces antihypertensive effects in direct [spontaneously hypertensive rat (SHR), ANG II, and DOCA-salt] and indirect (dyslipemic and gestational) models of hypertension, associated with end-organ damage protection. This review summarizes mechanistic insights into the antihypertensive effects of PPARß/δ activators, including molecular and functional mechanisms. Pharmacological PPARß/δ activation induces genomic actions including the increase of regulators of G protein-coupled signaling (RGS), acute nongenomic vasodilator effects, as well as the ability to improve the endothelial dysfunction, reduce vascular inflammation, vasoconstrictor responses, and sympathetic outflow from central nervous system. Evidence from clinical trials is also examined. These preclinical and clinical outcomes of PPARß/δ ligands may provide a basis for the development of therapies in combating hypertension.


Asunto(s)
Hipertensión/fisiopatología , PPAR delta/fisiología , PPAR-beta/fisiología , Vasodilatación/fisiología , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Endotelio Vascular/fisiopatología , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Humanos , Hipertensión/tratamiento farmacológico , Inflamación , PPAR delta/agonistas , PPAR delta/metabolismo , PPAR-beta/agonistas , PPAR-beta/metabolismo , Fenoxiacetatos/farmacología , Fenoxiacetatos/uso terapéutico , Proteínas RGS/efectos de los fármacos , Proteínas RGS/genética , Ratas , Ratas Endogámicas SHR , Sistema Nervioso Simpático/fisiopatología , Tiazoles/farmacología , Tiazoles/uso terapéutico , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos
11.
Biochimie ; 136: 55-58, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27914902

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) have emerged as key regulators of physiological and immunological processes. Recently, one of their members PPARß/δ has been identified as major player in the maintenance of bone homeostasis, by promoting Wnt signalling activity in osteoblast and mesenchymal stem cells (MSC). PPARß/δ not only controls the fate of MSC but also regulates their immunosuppressive properties by directly modulating their NF-κB activity. In this review, we discuss how the regulation of PPARß/δ provides an innovative strategy for an optimisation of MSC-based therapy.


Asunto(s)
Células Madre Mesenquimatosas/citología , PPAR gamma/fisiología , PPAR-beta/fisiología , Animales , Humanos , Tolerancia Inmunológica , Células Madre Mesenquimatosas/inmunología , Osteogénesis/fisiología
12.
Biochimie ; 136: 90-99, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27916645

RESUMEN

The nuclear receptor factor peroxisome proliferator-activated receptor (PPARß/δ) can regulate its target genes by transcriptional activation or repression through both ligand-dependent and independent mechanism as well as by interactions with other transcription factors. PPARß/δ exerts essential regulatory functions in intermediary metabolism that have been elucidated in detail, but clearly also plays a role in inflammation, differentiation, apoptosis and other cancer-associated processes, which is, however, mechanistically only partly understood. Consistent with these functions clinical associations link the expression of PPARß/δ and its target genes to an unfavorable outcome of several human cancers. However, the available data do not yield a clear picture of PPARß/δ's role in cancer-associated processes and are in fact partly controversial. This article provides an overview of this research area and discusses the role of PPARß/δ in cancer in light of the complex mechanisms of its transcriptional regulation and its potential as a druggable anti-cancer target.


Asunto(s)
Neoplasias/fisiopatología , PPAR delta/fisiología , PPAR-beta/fisiología , Angiopoyetinas/genética , Regulación de la Expresión Génica/fisiología , Humanos , Ligandos , Neoplasias/genética , PPAR delta/genética , PPAR-beta/genética , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , ARN Interferente Pequeño/genética , Transcripción Genética/fisiología
13.
Biochimie ; 136: 42-48, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27916646

RESUMEN

More than two decades of studying Peroxisome Proliferator-Activated Receptors (PPARs) has led to an understanding of their implications in various physiological processes that are key for health and disease. All three PPAR isotypes, PPARα, PPARß/δ, and PPARγ, are activated by a variety of molecules, including fatty acids, eicosanoids and phospholipids, and regulate a spectrum of genes involved in development, lipid and carbohydrate metabolism, inflammation, and proliferation and differentiation of many cell types in different tissues. The hypolipidemic and antidiabetic functions of PPARα and PPARγ in response to fibrate and thiazolidinedione treatment, respectively, are well documented. However, until more recently the functions of PPARß/δ were less well defined, but are now becoming more recognized in fatty acid metabolism, energy expenditure, and tissue repair. Skeletal muscle is an active metabolic organ with high plasticity for adaptive responses to varying conditions such as fasting or physical exercise. It is the major site of energy expenditure resulting from lipid and glucose catabolism. Here, we review the multifaceted roles of PPARß/δ in skeletal muscle physiology.


Asunto(s)
Músculo Esquelético/fisiología , PPAR gamma/fisiología , PPAR-beta/fisiología , Animales , Humanos , Músculo Esquelético/patología , Enfermedades Musculares/fisiopatología
14.
Subcell Biochem ; 81: 179-199, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27830505

RESUMEN

It has long been established that the transcriptional activity of retinoic acid (RA) is mediated by members of the nuclear receptor family of ligand-activated transcription factors termed RA receptors (RARs). More recent observations have established that RA also activates an additional nuclear receptor, PPARß/δ. Partitioning RA between RARs and PPARß/δ is governed by different intracellular lipid-binding proteins: cellular RA binding protein 2 (CRABP2) delivers RA to nuclear RARs and a fatty acid binding protein (FABP5) delivers the hormone from the cytosol to nuclear PPARß/δ. Consequently, RA signals through RARs in CRABP2-expressing cells, but activates PPARß/δ in cells that express a high level of FABP5. RA elicits different and sometimes opposing responses in cells that express different FABP5/CRABP2 ratios because PPARß/δ and RARs regulate the expression of distinct sets of genes. An overview of the observations that led to the discovery of this non-classical activity of RA are presented here, along with a discussion of evidence demonstrating the involvement of the dual transcriptional activities of RA in regulating energy homeostasis, insulin responses, and adipocyte and neuron differentiation.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , PPAR delta/fisiología , PPAR-beta/fisiología , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo/metabolismo , Animales , Transporte Biológico , Proteínas de Unión a Ácidos Grasos/fisiología , Predicción , Regulación de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Modelos Moleculares , Proteínas de Neoplasias/fisiología , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Obesidad/metabolismo , PPAR delta/efectos de los fármacos , PPAR-beta/efectos de los fármacos , Conformación Proteica , Receptores de Ácido Retinoico/fisiología
15.
Toxicology ; 363-364: 1-9, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27427494

RESUMEN

Peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) inhibits steatosis and inflammation, known risk factors for liver cancer. In this study, the effect of ligand activation of PPARß/δ in modulating liver tumorigenesis in transgenic hepatitis B virus (HBV) mice was examined. Activation of PPARß/δ in HBV mice reduced steatosis, the average number of liver foci, and tumor multiplicity. Reduced expression of hepatic CYCLIN D1 and c-MYC, tumor necrosis factor alpha (Tnfa) mRNA, serum levels of alanine aminotransaminase, and an increase in apoptotic signaling was also observed following ligand activation of PPARß/δ in HBV mice compared to controls. Inhibition of Tnfa mRNA expression was not observed in wild-type hepatocytes. Ligand activation of PPARß/δ inhibited lipopolysaccharide (LPS)-induced mRNA expression of Tnfa in wild-type, but not in Pparß/δ-null Kupffer cells. Interestingly, LPS-induced expression of Tnfa mRNA was also inhibited in Kupffer cells from a transgenic mouse line that expressed a DNA binding mutant form of PPARß/δ compared to controls. Combined, these results suggest that ligand activation of PPARß/δ attenuates hepatic tumorigenesis in HBV transgenic mice by inhibiting steatosis and cell proliferation, enhancing hepatocyte apoptosis, and modulating anti-inflammatory activity in Kupffer cells.


Asunto(s)
Hepatitis B/complicaciones , Neoplasias Hepáticas/prevención & control , PPAR delta/efectos de los fármacos , PPAR-beta/efectos de los fármacos , Alanina Transaminasa/sangre , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Ligandos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , PPAR delta/fisiología , PPAR-beta/fisiología , Reacción en Cadena de la Polimerasa , Tiazoles/farmacología
16.
Eur J Dermatol ; 25 Suppl 1: 4-11, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26287030

RESUMEN

We review the functions of peroxisome proliferator activated receptor (PPAR) ß/δ in skin wound healing and cancer. In particular, we highlight the roles of PPARß/δ in inhibiting keratinocyte apoptosis at wound edges via activation of the PI3K/PKBα/Akt1 pathway and its role during re-epithelialization in regulating keratinocyte adhesion and migration. In fibroblasts, PPARß/δ controls IL-1 signalling and thereby contributes to the homeostatic control of keratinocyte proliferation. We discuss its therapeutic potential for treating diabetic wounds and inflammatory skin diseases such as psoriasis and acne vulgaris. PPARß/δ is classified as a tumour growth modifier; it is activated by chronic low-grade inflammation, which promotes the production of lipids that, in turn, enhance PPARß/δ transcription activity. Our earlier work unveiled a cascade of events triggered by PPARß/δ that involve the oncogene Src, which promotes ultraviolet-induced skin cancer in mice via enhanced EGFR/Erk1/2 signalling and the expression of epithelial-to-mesenchymal transition (EMT) markers. Interestingly, PPARß/δ expression is correlated with the expression of SRC and EMT markers in human skin squamous cell carcinoma. Furthermore, there is a positive interaction between PPARß/δ, SRC, and TGFß1 at the transcriptional level in various human epithelial cancers. Taken together, these observations suggest the need for evaluating PPARß/δ modulators that attenuate or increase its activity, depending on the therapeutic target.


Asunto(s)
PPAR delta/fisiología , Neoplasias Cutáneas/fisiopatología , Cicatrización de Heridas/fisiología , Animales , Comunicación Celular , Uniones Célula-Matriz/fisiología , Humanos , Queratinocitos/fisiología , PPAR-beta/fisiología
17.
Physiol Rev ; 94(3): 795-858, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24987006

RESUMEN

The peroxisome proliferator-activated receptors, PPARα, PPARß, and PPARγ, are a family of transcription factors activated by a diversity of molecules including fatty acids and fatty acid metabolites. PPARs regulate the transcription of a large variety of genes implicated in metabolism, inflammation, proliferation, and differentiation in different cell types. These transcriptional regulations involve both direct transactivation and interaction with other transcriptional regulatory pathways. The functions of PPARα and PPARγ have been extensively documented mainly because these isoforms are activated by molecules clinically used as hypolipidemic and antidiabetic compounds. The physiological functions of PPARß remained for a while less investigated, but the finding that specific synthetic agonists exert beneficial actions in obese subjects uplifted the studies aimed to elucidate the roles of this PPAR isoform. Intensive work based on pharmacological and genetic approaches and on the use of both in vitro and in vivo models has considerably improved our knowledge on the physiological roles of PPARß in various cell types. This review will summarize the accumulated evidence for the implication of PPARß in the regulation of development, metabolism, and inflammation in several tissues, including skeletal muscle, heart, skin, and intestine. Some of these findings indicate that pharmacological activation of PPARß could be envisioned as a therapeutic option for the correction of metabolic disorders and a variety of inflammatory conditions. However, other experimental data suggesting that activation of PPARß could result in serious adverse effects, such as carcinogenesis and psoriasis, raise concerns about the clinical use of potent PPARß agonists.


Asunto(s)
PPAR-beta/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Inflamación/metabolismo , Músculos/fisiología
18.
Diabetologia ; 57(10): 2126-35, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25063273

RESUMEN

AIM/HYPOTHESIS: Endoplasmic reticulum (ER) stress, which is involved in the link between inflammation and insulin resistance, contributes to the development of type 2 diabetes mellitus. In this study, we assessed whether peroxisome proliferator-activated receptor (PPAR)ß/δ prevented ER stress-associated inflammation and insulin resistance in skeletal muscle cells. METHODS: Studies were conducted in mouse C2C12 myotubes, in the human myogenic cell line LHCN-M2 and in skeletal muscle from wild-type and PPARß/δ-deficient mice and mice exposed to a high-fat diet. RESULTS: The PPARß/δ agonist GW501516 prevented lipid-induced ER stress in mouse and human myotubes and in skeletal muscle of mice fed a high-fat diet. PPARß/δ activation also prevented thapsigargin- and tunicamycin-induced ER stress in human and murine skeletal muscle cells. In agreement with this, PPARß/δ activation prevented ER stress-associated inflammation and insulin resistance, and glucose-intolerant PPARß/δ-deficient mice showed increased phosphorylated levels of inositol-requiring 1 transmembrane kinase/endonuclease-1α in skeletal muscle. Our findings demonstrate that PPARß/δ activation prevents ER stress through the activation of AMP-activated protein kinase (AMPK), and the subsequent inhibition of extracellular-signal-regulated kinase (ERK)1/2 due to the inhibitory crosstalk between AMPK and ERK1/2, since overexpression of a dominant negative AMPK construct (K45R) reversed the effects attained by PPARß/δ activation. CONCLUSIONS/INTERPRETATION: Overall, these findings indicate that PPARß/δ prevents ER stress, inflammation and insulin resistance in skeletal muscle cells by activating AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Músculo Esquelético/metabolismo , PPAR delta/fisiología , PPAR-beta/fisiología , Animales , Línea Celular , Dieta Alta en Grasa/efectos adversos , Estrés del Retículo Endoplásmico/genética , Humanos , Técnicas In Vitro , Inflamación/etiología , Inflamación/genética , Resistencia a la Insulina/genética , Ratones , Fibras Musculares Esqueléticas/metabolismo , PPAR delta/deficiencia , PPAR delta/genética , PPAR-beta/deficiencia , PPAR-beta/genética
19.
Carcinogenesis ; 35(7): 1602-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24639079

RESUMEN

Whether peroxisome proliferator-activated receptor ß/δ (PPARß/δ) reduces skin tumorigenesis by altering aryl hydrocarbon receptor (AHR)-dependent activities was examined. Polycyclic aromatic hydrocarbons (PAH) increased expression of cytochrome P4501A1 (CYP1A1), CYP1B1 and phase II xenobiotic metabolizing enzymes in wild-type skin and keratinocytes. Surprisingly, this effect was not found in Pparß/δ-null skin and keratinocytes. Pparß/δ-null keratinocytes exhibited decreased AHR occupancy and histone acetylation on the Cyp1a1 promoter in response to a PAH compared with wild-type keratinocytes. Bisulfite sequencing of the Cyp1a1 promoter and studies using a DNA methylation inhibitor suggest that PPARß/δ promotes demethylation of the Cyp1a1 promoter. Experiments with human HaCaT keratinocytes stably expressing shRNA against PPARß/δ also support this conclusion. Consistent with the lower AHR-dependent activities in Pparß/δ-null mice compared with wild-type mice, 7,12-dimethylbenz[a]anthracene (DMBA)-induced skin tumorigenesis was inhibited in Pparß/δ-null mice compared with wild-type. Results from these studies demonstrate that PPARß/δ is required to mediate complete carcinogenesis by DMBA. The mechanisms underlying this PPARß/δ-dependent reduction of AHR signaling by PAH are not due to alterations in the expression of AHR auxiliary proteins, ligand binding or AHR nuclear translocation between genotypes, but are likely influenced by PPARß/δ-dependent demethylation of AHR target gene promoters including Cyp1a1 that reduces AHR accessibility as shown by reduced promoter occupancy. This PPARß/δ/AHR crosstalk is unique to keratinocytes and conserved between mice and humans.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Queratinocitos/metabolismo , PPAR delta/fisiología , PPAR-beta/fisiología , Receptores de Hidrocarburo de Aril/fisiología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Western Blotting , Carcinógenos/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Inmunoprecipitación de Cromatina , Dermis/citología , Dermis/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Queratinocitos/citología , Ratones , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
20.
Nutr J ; 13: 17, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24524207

RESUMEN

Peroxisome proliferator-activated receptors are expressed in many tissues, including adipocytes, hepatocytes, muscles and endothelial cells; however, the affinity depends on the isoform of PPAR, and different distribution and expression profiles, which ultimately lead to different clinical outcomes. Because they play an important role in lipid and glucose homeostasis, they are called lipid and insulin sensors. Their actions are limited to specific tissue types and thus, reveal a characteristic influence on target cells. PPARα mainly influences fatty acid metabolism and its activation lowers lipid levels, while PPARγ is mostly involved in the regulation of the adipogenesis, energy balance, and lipid biosynthesis. PPARß/δ participates in fatty acid oxidation, mostly in skeletal and cardiac muscles, but it also regulates blood glucose and cholesterol levels. Many natural and synthetic ligands influence the expression of these receptors. Synthetic ligands are widely used in the treatment of dyslipidemia (e.g. fibrates--PPARα activators) or in diabetes mellitus (e.g. thiazolidinediones--PPARγ agonists). New generation drugs--PPARα/γ dual agonists--reveal hypolipemic, hypotensive, antiatherogenic, anti-inflammatory and anticoagulant action while the overexpression of PPARß/δ prevents the development of obesity and reduces lipid accumulation in cardiac cells, even during a high-fat diet. Precise data on the expression and function of natural PPAR agonists on glucose and lipid metabolism are still missing, mostly because the same ligand influences several receptors and a number of reports have provided conflicting results. To date, we know that PPARs have the capability to accommodate and bind a variety of natural and synthetic lipophilic acids, such as essential fatty acids, eicosanoids, phytanic acid and palmitoylethanolamide. A current understanding of the effects of PPARs, their molecular mechanisms and the role of these receptors in nutrition and therapeutic treatment are delineated in this paper.


Asunto(s)
Receptores Activados del Proliferador del Peroxisoma/fisiología , Animales , Humanos , Ligandos , PPAR alfa/fisiología , PPAR delta/fisiología , PPAR gamma/fisiología , PPAR-beta/fisiología , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/efectos de los fármacos , Receptores X Retinoide/fisiología , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA