Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 436
Filtrar
1.
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
2.
Acta Cir Bras ; 38: e387023, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38055403

RESUMEN

PURPOSE: Cerebral ischemia-reperfusion (I/R) is a neurovascular disorder that leads to brain injury. In mice, Fasudil improves nerve injury induced by I/R. However, it is unclear if this is mediated by increased peroxisome proliferator-activated receptor-α (PPARα) expression and reduced oxidative damage. This study aimed to investigate the neuroprotective mechanism of action of Fasudil. METHODS: MCAO (Middle cerebral artery occlusion) was performed in male C57BL/6J wild-type and PPARα KO mice between September 2021 to April 2023. Mice were treated with Fasudil and saline; 2,3,5-Triphenyltetrazolium chloride (TTC) staining was performed to analyze cerebral infarction. PPARα and Rho-associated protein kinase (ROCK) expression were detected using Western blot, and the expression of NADPH subunit Nox2 mRNA was detected using real-time polymerase chain reaction. The NADPH oxidase activity level and reactive oxygen species (ROS) content were also investigated. RESULTS: After cerebral ischemia, the volume of cerebral necrosis was reduced in wild-type mice treated with Fasudil. The expression of PPARα was increased, while ROCK was decreased. Nox2 mRNA expression, NADPH oxidase activity, and ROS content decreased. There were no significant changes in cerebral necrosis volumes, NADPH oxidase activity, and ROS content in the PPARα KO mice treated with Fasudil. CONCLUSIONS: In mice, the neuroprotective effect of Fasudil depends on the expression of PPARα induced by ROCK-PPARα-NOX axis-mediated reduction in ROS and associated oxidative damage.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Ratones , Masculino , Animales , PPAR alfa/fisiología , Especies Reactivas de Oxígeno/metabolismo , Neuroprotección , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Daño por Reperfusión/genética , Ratones Endogámicos C57BL , Isquemia , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/prevención & control , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Reperfusión , Necrosis , ARN Mensajero
3.
J Proteome Res ; 22(7): 2327-2338, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37232578

RESUMEN

Incidence and mortality rates of alcoholic liver disease (ALD) is one of the highest in the world. In the present study, we found that the genetic knockout nuclear receptor the peroxisome proliferator-activated receptor α (PPARα) exacerbated ALD. Lipidomics of the liver revealed changed levels of lipid species encompassing phospholipids, ceramides (CM), and long-chain fatty acids in Ppara-null mice induced by ethanol. Moreover, 4-hydroxyphenylacetic acid (4-HPA) was changed as induced by ethanol in the metabolome of urine. Moreover, the phylum level analysis showed a decrease in the level of Bacteroidetes and an increase in the level of Firmicutes after alcohol feeding in Ppara-null mice, while there was no change in wild-type mice. In Ppara-null mice, the level of Clostridium_sensu_stricto_1 and Romboutsia were upregulated after alcohol feeding. These data revealed that PPARα deficiency potentiated alcohol-induced liver injury through promotion of lipid accumulation, changing the metabolome of urine, and increasing the level of Clostridium_sensu_stricto_1 and Romboutsia. 4-HPA could improve ALD in mice by regulating inflammation and lipid metabolism. Therefore, our findings suggest a novel approach to the treatment of ALD: focusing on the gut microbiota and its metabolites. Data are available via ProteomeXchange (PXD 041465).


Asunto(s)
Microbioma Gastrointestinal , Hepatopatías Alcohólicas , Animales , Ratones , Etanol/efectos adversos , Etanol/metabolismo , Etanol/toxicidad , Hígado/metabolismo , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Metabolómica , Ratones Noqueados , Fosfolípidos/metabolismo , PPAR alfa/fisiología
4.
Yakugaku Zasshi ; 141(11): 1267-1274, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34719550

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor-type transcription factors that consist of three subtypes (α, γ, and ß/δ) with distinct physiological functions and ligand recognition. PPARs regulate energy metabolism and therefore become therapeutic targets for various metabolic diseases. While PPARα agonists are used as anti-dyslipidemia drugs and PPARγ agonists as anti-type 2 diabetes drugs, PPAR dual/pan agonists (that acts on two or three subtypes) are expected to treat non-alcoholic steatohepatitis (NASH), pulmonary fibrosis, etc. Structural analyses of PPAR-ligand-binding domain (LBD)-ligand co-crystals using X-ray crystallography have been done mainly on PPARγ, in which ligand-free apocrystals were prepared; however, the information on PPARα-LBD and PPARδ-LBD is limited. Recently, we succeeded to obtain 34 novel co-crystal structures of PPARα-LBD and various PPARα ligands (including fibrates) using various co-crystallization techniques. This procedure is applicable to preparation of PPARδ-LBD co-crystals, and contributes to molecular design of new PPAR targeted drugs based on all three PPAR-LBD structures.


Asunto(s)
Cristalografía por Rayos X/métodos , Ligandos , PPAR alfa/química , PPAR alfa/metabolismo , Metabolismo Energético , Hipoglucemiantes , Hipolipemiantes , Enfermedades Metabólicas/metabolismo , PPAR alfa/agonistas , PPAR alfa/fisiología , Unión Proteica , Dominios Proteicos
5.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445672

RESUMEN

In mammalian cells, two cellular organelles, mitochondria and peroxisomes, share the ability to degrade fatty acid chains. Although each organelle harbors its own fatty acid ß-oxidation pathway, a distinct mitochondrial system feeds the oxidative phosphorylation pathway for ATP synthesis. At the same time, the peroxisomal ß-oxidation pathway participates in cellular thermogenesis. A scientific milestone in 1965 helped discover the hepatomegaly effect in rat liver by clofibrate, subsequently identified as a peroxisome proliferator in rodents and an activator of the peroxisomal fatty acid ß-oxidation pathway. These peroxisome proliferators were later identified as activating ligands of Peroxisome Proliferator-Activated Receptor α (PPARα), cloned in 1990. The ligand-activated heterodimer PPARα/RXRα recognizes a DNA sequence, called PPRE (Peroxisome Proliferator Response Element), corresponding to two half-consensus hexanucleotide motifs, AGGTCA, separated by one nucleotide. Accordingly, the assembled complex containing PPRE/PPARα/RXRα/ligands/Coregulators controls the expression of the genes involved in liver peroxisomal fatty acid ß-oxidation. This review mobilizes a considerable number of findings that discuss miscellaneous axes, covering the detailed expression pattern of PPARα in species and tissues, the lessons from several PPARα KO mouse models and the modulation of PPARα function by dietary micronutrients.


Asunto(s)
Ácidos Grasos/metabolismo , PPAR alfa/metabolismo , Peroxisomas/metabolismo , Acil-CoA Oxidasa/metabolismo , Animales , Humanos , Hígado/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , PPAR alfa/fisiología , Proliferadores de Peroxisomas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Ácido Retinoico/metabolismo , Elementos de Respuesta/genética , Receptores X Retinoide/metabolismo , Activación Transcripcional/genética
6.
Molecules ; 26(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067839

RESUMEN

Recent work has shown that bilirubin has a hormonal function by binding to the peroxisome proliferator-activated receptor-α (PPARα), a nuclear receptor that drives the transcription of genes to control adiposity. Our previous in silico work predicted three potential amino acids that bilirubin may interact with by hydrogen bonding in the PPARα ligand-binding domain (LBD), which could be responsible for the ligand-induced function. To further reveal the amino acids that bilirubin interacts with in the PPARα LBD, we harnessed bilirubin's known fluorescent properties when bound to proteins such as albumin. Our work here revealed that bilirubin interacts with threonine 283 (T283) and alanine 333 (A333) for ligand binding. Mutational analysis of T283 and A333 showed significantly reduced bilirubin binding, reductions of 11.4% and 17.0%, respectively. Fenofibrate competitive binding studies for the PPARα LBD showed that bilirubin and fenofibrate possibly interact with different amino acid residues. Furthermore, bilirubin showed no interaction with PPARγ. This is the first study to reveal the amino acids responsible for bilirubin binding in the ligand-binding pocket of PPARα. Our work offers new insight into the mechanistic actions of a well-known molecule, bilirubin, and new fronts into its mechanisms.


Asunto(s)
Bilirrubina/metabolismo , PPAR alfa/metabolismo , Bilirrubina/fisiología , Unión Competitiva , Células HEK293 , Humanos , Ligandos , PPAR alfa/fisiología , Unión Proteica/fisiología
7.
Int J Mol Sci ; 22(10)2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34069420

RESUMEN

Selenium-binding protein 1 (Selenbp1) is a 2,3,7,8-tetrechlorodibenzo-p-dioxin inducible protein whose function is yet to be comprehensively elucidated. As the highly homologous isoform, Selenbp2, is expressed at low levels in the kidney, it is worthwhile comparing wild-type C57BL mice and Selenbp1-deficient mice under dioxin-free conditions. Accordingly, we conducted a mouse metabolomics analysis under non-dioxin-treated conditions. DNA microarray analysis was performed based on observed changes in lipid metabolism-related factors. The results showed fluctuations in the expression of numerous genes. Real-time RT-PCR confirmed the decreased expression levels of the cytochrome P450 4a (Cyp4a) subfamily, known to be involved in fatty acid ω- and ω-1 hydroxylation. Furthermore, peroxisome proliferator-activated receptor-α (Pparα) and retinoid-X-receptor-α (Rxrα), which form a heterodimer with Pparα to promote gene expression, were simultaneously reduced. This indicated that reduced Cyp4a expression was mediated via decreased Pparα and Rxrα. In line with this finding, increased levels of leukotrienes and prostaglandins were detected. Conversely, decreased hydrogen peroxide levels and reduced superoxide dismutase (SOD) activity supported the suppression of the renal expression of Sod1 and Sod2 in Selenbp1-deficient mice. Therefore, we infer that ablation of Selenbp1 elicits oxidative stress caused by increased levels of superoxide anions, which alters lipid metabolism via the Pparα pathway.


Asunto(s)
Metabolismo de los Lípidos/genética , Proteínas de Unión al Selenio/metabolismo , Animales , Citocromo P-450 CYP4A/metabolismo , Expresión Génica , Riñón/patología , Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/genética , PPAR alfa/metabolismo , PPAR alfa/fisiología , ARN Mensajero/genética , Receptor alfa X Retinoide/metabolismo , Receptor alfa X Retinoide/fisiología , Proteínas de Unión al Selenio/genética , Factores de Transcripción/metabolismo
8.
Metabolism ; 116: 154705, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33422545

RESUMEN

The preservation of body proteins is essential to guarantee their functions in organisms. Therefore, the utilization of amino acids as energy substrates is regulated by a precise fine-tuned mechanism. Recent evidence suggests that the transcription factors peroxisome proliferator-activated receptor alpha (PPARα) and hepatocyte nuclear factor 4 alpha (HNF4α) are involved in this regulatory mechanism. Thus, the aim of this study was to determine how these transcription factors interact to regulate the expression of amino acid catabolism genes. In vivo studies using PPARα-knockout mice (Pparα-null) fed different amounts of dietary protein showed that in the absence of PPARα, there was a significant increase in HNF4α abundance in the liver, which corresponded with an increase in amino acid catabolizing enzyme (AACE) expression and the generation of increased amounts of postprandial urea. Moreover, this effect was proportional to the increase in dietary protein consumed. Chromatin immunoprecipitation assays showed that HNF4α can bind to the promoter of AACE serine dehydratase (SDS), an effect that was potentiated by dietary protein in the Pparα-null mice. The mechanistic studies revealed that the presence of retinoid X receptor alpha (RXRα) is essential to repress HNF4α activity in the presence of PPARα, and this interaction accelerates HNF4α degradation via the proteasome pathway. These results showed that PPARα can downregulate liver amino acid catabolism in the presence of RXRα by inhibiting HNF4α activity.


Asunto(s)
Aminoácidos/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/metabolismo , PPAR alfa/fisiología , Receptor alfa X Retinoide/fisiología , Animales , Regulación hacia Abajo/genética , Células HEK293 , Células Hep G2 , Humanos , Masculino , Metabolismo/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR alfa/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , Receptor alfa X Retinoide/genética
9.
Am J Physiol Endocrinol Metab ; 320(2): E191-E207, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284088

RESUMEN

Recent research on bilirubin, a historically well-known waste product of heme catabolism, suggests an entirely new function as a metabolic hormone that drives gene transcription by nuclear receptors. Studies are now revealing that low plasma bilirubin levels, defined as "hypobilirubinemia," are a possible new pathology analogous to the other end of the spectrum of extreme hyperbilirubinemia seen in patients with jaundice and liver dysfunction. Hypobilirubinemia is most commonly seen in patients with metabolic dysfunction, which may lead to cardiovascular complications and possibly stroke. We address the clinical significance of low bilirubin levels. A better understanding of bilirubin's hormonal function may explain why hypobilirubinemia might be deleterious. We present mechanisms by which bilirubin may be protective at mildly elevated levels and research directions that could generate treatment possibilities for patients with hypobilirubinemia, such as targeting of pathways that regulate its production or turnover or the newly designed bilirubin nanoparticles. Our review here calls for a shift in the perspective of an old molecule that could benefit millions of patients with hypobilirubinemia.


Asunto(s)
Bilirrubina/sangre , Bilirrubina/fisiología , Metabolismo Energético , Hormonas/fisiología , Animales , Bilirrubina/deficiencia , Metabolismo Energético/genética , Regulación de la Expresión Génica , Enfermedad de Gilbert/sangre , Enfermedad de Gilbert/genética , Enfermedad de Gilbert/metabolismo , Hemo/metabolismo , Humanos , Hiperbilirrubinemia/complicaciones , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/metabolismo , Redes y Vías Metabólicas/genética , PPAR alfa/metabolismo , PPAR alfa/fisiología
10.
Cell Rep ; 33(3): 108278, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33086073

RESUMEN

Dendritic cells (DCs) orchestrate the initiation, programming, and regulation of anti-tumor immune responses. Emerging evidence indicates that the tumor microenvironment (TME) induces immune dysfunctional tumor-infiltrating DCs (TIDCs), characterized with both increased intracellular lipid content and mitochondrial respiration. The underlying mechanism, however, remains largely unclear. Here, we report that fatty acid-carrying tumor-derived exosomes (TDEs) induce immune dysfunctional DCs to promote immune evasion. Mechanistically, peroxisome proliferator activated receptor (PPAR) α responds to the fatty acids delivered by TDEs, resulting in excess lipid droplet biogenesis and enhanced fatty acid oxidation (FAO), culminating in a metabolic shift toward mitochondrial oxidative phosphorylation, which drives DC immune dysfunction. Genetic depletion or pharmacologic inhibition of PPARα effectively attenuates TDE-induced DC-based immune dysfunction and enhances the efficacy of immunotherapy. This work uncovers a role for TDE-mediated immune modulation in DCs and reveals that PPARα lies at the center of metabolic-immune regulation of DCs, suggesting a potential immunotherapeutic target.


Asunto(s)
Células Dendríticas/fisiología , PPAR alfa/metabolismo , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/inmunología , Ácidos Grasos/metabolismo , Femenino , Humanos , Metabolismo de los Lípidos , Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , PPAR alfa/fisiología
11.
Cell Death Dis ; 11(8): 691, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32826849

RESUMEN

Endoplasmic reticulum stress (ER stress) just like a double-edged sword depending on different conditions in the development of multiple hepatic diseases. But the molecular mechanisms of functional conversion during ER stress have not been fully elucidated. In this study, we aim to illustrate the role of PPARα and the subtle mechanism in the functional conversion of ER stress. Tunicamycin (TM) and thapsigargin (TG), as ER stress inducers, were used to induce ER stress in AML12 cells. During the ER stress, qRT-PCR and immunoblotting was used to measure the expression levels of GRP78 and CHOP which show a gradually increasing trend, while PPARα and autophagy was significantly activated in the early stage but was inhibited in the late stage. Moreover, PPARα inhibition by siRNA promoted cell injury in the mild-ER stress and PPARα activation by WY-14643 reduced cell apoptosis in the serious ER stress. In the mild-ER stress with PPARα knocked down, activation of autophagy by rapamycin significantly improved cell survival, in the serious ER stress with PPARα activation, inhibition of autophagy by 3-MA aggravate cell injury. In addition, in the mild-ER stress with PPARα knocked down, CHOP knocked down by siRNA reduced cell apoptosis, in the serious ER stress activated PPARα, CHOP over-expression mediated by lentiviral vector contributed to serious cell injury. Furthermore, C57BL/6 mice was used to induce ER stress with TM intraperitoneal injection, PPARα and autophagy was upregulated in the mild-ER stress while downregulated in the serious ER stress, measured by qRT-PCR and immunoblotting, further confirmed the finding in vitro. Our results firstly demonstrated that PPARα is a key molecule in the functional conversion of ER stress: protective effects in the mild ER stress was mediated by PPARα-autophagy pathway and destructive effects in the serious ER stress was mediated by PPARα-CHOP pathway.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , PPAR alfa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Retículo Endoplásmico/patología , Retículo Endoplásmico/fisiología , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR alfa/fisiología , Tapsigargina/farmacología , Factor de Transcripción CHOP/análisis , Factor de Transcripción CHOP/metabolismo , Tunicamicina/farmacología
12.
J Endocrinol ; 247(1): 11-24, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32698143

RESUMEN

Fructose dietary intake affects the composition of the intestinal microbiota and influences the development of hepatic steatosis. Endotoxins produced by gram-negative bacteria alter intestinal permeability and cause bacterial translocation. This study evaluated the effects of gut microbiota modulation by a purified PPAR-alpha agonist (WY14643), a DPP-4 inhibitor (linagliptin), or their association on intestinal barrier integrity, endotoxemia, and hepatic energy metabolism in high-fructose-fed C57BL/6 mice. Fifty mice were divided to receive the control diet (C group) or the high-fructose diet (HFRU) for 12 weeks. Subsequently, the HFRU group was divided to initiate the treatment with PPAR-alpha agonist (3.5 mg/kg/BM) and DPP-4 inhibitor (15 mg/kg/BM). The HFRU group had glucose intolerance, endotoxemia, and dysbiosis (with increased Proteobacteria) without changes in body mass in comparison with the C group. HFRU group showed damaged intestinal ultrastructure, which led to liver inflammation and marked hepatic steatosis in the HFRU group when compared to the C group. PPAR-alpha activation and DPP-4 inhibition countered glucose intolerance, endotoxemia, and dysbiosis, ameliorating the ultrastructure of the intestinal barrier and reducing Tlr4 expression in the liver of treated animals. These beneficial effects suppressed lipogenesis and mitigated hepatic steatosis. In conclusion, the results herein propose a role for PPAR-alpha activation, DPP-4 inhibition, and their association in attenuating hepatic steatosis by gut-liver axis modulation in high-fructose mice model. These observations suggest these treatments as potential targets to treat hepatic steatosis and avoid its progression.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Fructosa/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Linagliptina/farmacología , Hígado/efectos de los fármacos , PPAR alfa/fisiología , Animales , Glucemia/análisis , Dieta , Endotoxemia/prevención & control , Hígado Graso/prevención & control , Microbioma Gastrointestinal/fisiología , Intestinos/efectos de los fármacos , Intestinos/ultraestructura , Lipogénesis/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR alfa/efectos de los fármacos , Proliferadores de Peroxisomas , Pirimidinas/farmacología
13.
Cytokine ; 134: 155207, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32693363

RESUMEN

The nuclear transcriptional factor peroxisome proliferator activated receptor alpha (PPARα) plays a role in regulating genes involved in lipid metabolism, adipogenesis and inflammation. We aimed to assess the role of PPARα on exercise-mediated locally produced cytokines in adipose fat deposits and skeletal muscle. C57BL/6 (WT) and PPARα knockout (PPARα-/-) mice were examined. Each genotype was randomly subdivided into three groups: non-exercised, and euthanized 2 or 24 h after a moderate aerobic exercise session (run on a treadmill at 60% of maximum speed for 1 h). Fat content in gastrocnemius muscle and lipolytic activity in isolated adipose tissue from mesenteric (MEAT) and retroperitoneal (RPAT) adipose tissue were evaluated. In addition, Interleukin 6 (IL-6), interleukin 10 (IL-10), tumor necrosis factor α (TNF-α) and monocyte chemoattractant protein 1 (MCP-1) content were evaluated by ELISA. WT mice showed a maximum lipolysis rate, as well as higher IL-6, IL-10, and IL10/TNF-α ratio values 2 h post-exercise (RPAT only) compared with PPARα-/- mice. Taken together, our data suggests that PPARα knockout mice exhibited reduced lipolysis and anti-inflammatory response in adipose tissue following exercise, PPARα appears to play an important role in immunomodulatory and lipolysis signaling after acute moderate exercise.


Asunto(s)
Citocinas/metabolismo , PPAR alfa/fisiología , Condicionamiento Físico Animal , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Interleucina-6/metabolismo , Lipólisis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/inmunología , PPAR alfa/genética
14.
Bull Exp Biol Med ; 168(5): 658-661, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32246376

RESUMEN

Reduced expression of the key regulator of cardiac metabolism, transcription factor PPARα, in surgical samples of the auricles from patients with coronary heart disease and heart failure was detected by real-time quantitative PCR. These changes indicate reduced activity of this factor and a shift of energy metabolism from oxidative phosphorylation to glycolysis typical of dedifferentiated cells. Electron microscopy revealed dedifferentiated cardiomyocytes with disassembled contractile apparatus and disorganized sarcomeres. In the examined specimens from patients with heart failure, severe myocardial fibrosis was revealed.


Asunto(s)
Metabolismo Energético/fisiología , Corazón/fisiología , Miocitos Cardíacos/metabolismo , PPAR alfa/fisiología , Regeneración/fisiología , Biopsia , Desdiferenciación Celular/genética , Enfermedad Coronaria/genética , Enfermedad Coronaria/metabolismo , Enfermedad Coronaria/patología , Enfermedad Coronaria/fisiopatología , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/fisiopatología , Metabolismo Energético/genética , Regulación de la Expresión Génica , Glucólisis/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Fosforilación Oxidativa , PPAR alfa/genética , PPAR alfa/metabolismo
15.
J Invest Dermatol ; 140(10): 1909-1918.e8, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32142797

RESUMEN

We have shown previously that endocannabinoids promote sebaceous lipogenesis, and sebocytes are involved in the metabolism of the endocannabinoid-like substance oleoylethanolamide (OEA). OEA is an endogenous activator of GPR119, a recently deorphanized receptor, which currently is being investigated as a promising antidiabetic drug target. In this study, we investigated the effects of OEA as well as the expression and role of GPR119 in human sebocytes. We found that OEA promoted differentiation of human SZ95 sebocytes (elevated lipogenesis, enhanced granulation, and the induction of early apoptotic events), and it switched the cells to a proinflammatory phenotype (increased expression and release of several proinflammatory cytokines). Moreover, we could also demonstrate that GPR119 was expressed in human sebocytes, and its small interfering RNA-mediated gene silencing suppressed OEA-induced sebaceous lipogenesis, which was mediated via c-Jun N-terminal kinase, extracellular signal-regulated kinase 1/2, protein kinase B, and CRE-binding protein activation. Finally, our pilot data demonstrated that GPR119 was downregulated in the sebaceous glands of patients with acne, arguing that GPR119 signaling may indeed be disturbed in acne. Collectively, our findings introduce the OEA/GPR119 signaling as a positive regulator of sebocyte differentiation and highlight the possibility that dysregulation of this pathway may contribute to the development of seborrhea and acne.


Asunto(s)
Receptores Acoplados a Proteínas G/fisiología , Glándulas Sebáceas/citología , Glándulas Sebáceas/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Endocannabinoides/farmacología , Humanos , Ácidos Oléicos/farmacología , PPAR alfa/fisiología , Glándulas Sebáceas/inmunología , Transducción de Señal/fisiología
16.
Biochem Pharmacol ; 169: 113613, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31445019

RESUMEN

Previously, we reported that hepatic muscarinic receptors modulate both acute and chronic liver injury, however, the role of muscarinic receptors in fatty liver disease is unclear. We observed in patients who underwent weight loss surgery, a decrease in hepatic expression of M3 muscarinic receptors (M3R). We also observed that fat loading of hepatocytes, increased M3R expression. Based on these observations, we tested the hypothesis that M3R regulate hepatocyte lipid accumulation. Incubation of AML12 hepatocytes with 1 mM oleic acid resulted in lipid accumulation that was significantly reduced by co-treatment with a muscarinic agonist (pilocarpine or carbachol), an effect blocked by atropine (a muscarinic antagonist). Similar treatment of Hepa 1-6 cells, a mouse hepatoblastoma cell line, showed comparable results. In both, control and fat-loaded AML12 cells, pilocarpine induced time-dependent AMPKα phosphorylation and significantly up-regulated lipolytic genes (ACOX1, CPT1, and PPARα). Compound C, a selective and reversible AMPK inhibitor, significantly blunted pilocarpine-mediated reduction of lipid accumulation and pilocarpine-mediated up-regulation of lipolytic genes. BAPTA-AM, a calcium chelator, and STO-609, a calcium/calmodulin-dependent protein kinase kinase inhibitor, attenuated agonist-induced AMPKα phosphorylation. Finally, M3R siRNA attenuated agonist-induced AMPKα phosphorylation as well as agonist-mediated reduction of hepatocyte steatosis. In conclusion, this proof-of-concept study demonstrates that M3R has protective effects against hepatocyte lipid accumulation by activating AMPK pathway and is a potential therapeutic target for non-alcoholic fatty liver disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Receptor Muscarínico M3/fisiología , Animales , Células Cultivadas , Humanos , Ratones , PPAR alfa/fisiología , Fosforilación , Receptor Muscarínico M1/fisiología , Transducción de Señal/fisiología
17.
Neurochem Int ; 129: 104501, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31299417

RESUMEN

Brain is a site of diabetic end-organ damage. Diabetes-associated cognitive dysfunction, referred as "diabetic encephalopathy" (DE) has been coined for the patients with type 2 diabetes mellitus showing decline in their cognitive function, especially weak episodic memory, cognitive inflexibility and poor psychomotor performance leading towards Alzheimer's disease. Current evidence supported that aberrant synapses, energy metabolism imbalance, advanced glycation end products (AGEs) accumulation and Tau hyperphosphorylation are associated with cognition deficits induced by diabetes. Oleoylethanolamide (OEA), an endogenous peroxisome proliferator-activated receptor alpha (PPARα) agonist, has anti-hyperlipidemia, anti-inflammatory and neuroprotective activities. However, the effect of OEA on DE is unknown. Therefore, we tested its influence against cognitive dysfunction in high fat diet and streptozotocin (HFD + STZ)-induced diabetic C57BL/6J and PPARα--/- mice using Morris water maze (MWM) test. Neuron staining, dementia markers and neuroplasticity in the hippocampus were assessed to evaluate the neuropathological changes. The results showed that chronic OEA treatment significantly lowered hyperglycemia, recovered cognitive performance, reduced dementia markers, and inhibited hippocampal neuron loss and neuroplasticity impairments in diabetic mice. In contrast, the changes in MWM performance and neuron loss were not observed in PPARα knockout mice via OEA administration. These results indicated that OEA may provide a potential alternative therapeutic for DE by activating PPARα signaling.


Asunto(s)
Encefalopatías/prevención & control , Trastornos del Conocimiento/prevención & control , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Endocannabinoides/uso terapéutico , Ácidos Oléicos/uso terapéutico , PPAR alfa/agonistas , Animales , Glucemia/análisis , Encefalopatías/tratamiento farmacológico , Encefalopatías/etiología , Encefalopatías/patología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/psicología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/psicología , Dieta Alta en Grasa/efectos adversos , Productos Finales de Glicación Avanzada/sangre , Hipocampo/patología , Resistencia a la Insulina , Lípidos/sangre , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/patología , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , PPAR alfa/deficiencia , PPAR alfa/genética , PPAR alfa/fisiología , Organismos Libres de Patógenos Específicos , Estreptozocina , Proteínas tau/metabolismo
18.
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
19.
Biol Psychiatry ; 85(12): 1036-1045, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30955840

RESUMEN

BACKGROUND: The endocannabinoid and neurosteroid systems regulate emotions and stress responses. Activation of peroxisome proliferator-activated receptor (PPAR)-α by the endocannabinoid congener N-palmitoylethanolamine (PEA) regulates pathophysiological systems (e.g., inflammation, oxidative stress) and induces peripheral biosynthesis of allopregnanolone, a gamma-aminobutyric acidergic neurosteroid implicated in mood disorders. However, effects of PPAR-α on emotional behavior are poorly understood. METHODS: We studied the impact of PPAR-α activation on emotional behavior in a mouse model of posttraumatic stress disorder. Neurosteroid levels before and after PEA treatment were measured by gas chromatography-mass spectrometry in relevant brain regions of socially isolated versus group-housed mice exposed to the contextual fear conditioning test, elevated plus maze test, forced swim test, and tail suspension test. Neurosteroidogenic enzyme levels were quantified in hippocampus by Western blot. RESULTS: PEA administered in a model of conditioned contextual fear reconsolidation blockade facilitated fear extinction and fear extinction retention and induced marked antidepressive- and anxiolytic-like effects in socially isolated mice with reduced brain allopregnanolone levels. These effects were mimicked by the PPAR-α synthetic agonists, fenofibrate and GW7647, and were prevented by PPAR-α deletion, PPAR-α antagonists, and neurosteroid-enzyme inhibitors. Behavioral improvements correlated with PEA-induced upregulation of PPAR-α, neurosteroidogenic enzyme expression, and normalization of corticolimbic allopregnanolone levels. CONCLUSIONS: This evidence supports a previously unknown role for PPAR-α in behavior regulation and suggests new strategies for the treatment of neuropsychopathologies characterized by deficient neurosteroidogenesis, including posttraumatic stress disorder and major depressive disorder.


Asunto(s)
Emociones/fisiología , Etanolaminas/administración & dosificación , PPAR alfa/fisiología , Ácidos Palmíticos/administración & dosificación , Pregnanolona/biosíntesis , Trastornos por Estrés Postraumático/fisiopatología , Amidas , Animales , Ansiedad/fisiopatología , Modelos Animales de Enfermedad , Emociones/efectos de los fármacos , Miedo/efectos de los fármacos , Miedo/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , PPAR alfa/metabolismo , Aislamiento Social
20.
Inflammopharmacology ; 27(6): 1131-1142, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30945071

RESUMEN

Osteoarthritis (OA) is characterized by cartilage degeneration, subchondral sclerosis, and pain. Cannabinoids have well-established anti-nociceptive properties in animal models of chronic pain. The aim of this study is to evaluate the anti-nociceptive effects of synthetic cannabinoids (WIN-55,212 and HU210) and the cannabinoid-like compound palmitoylethanolamide (PEA) in rat models of OA and to assess the role of cannabinoid receptor 1 (CB1) and the peroxisome proliferator-activated receptor α (PPARα) in mediating these effects. Intra-articular injection of monosodium iodoacetate (MIA) in the knee joint was used as a model of osteoarthritis. The von Frey filament test and weight-bearing difference were used to assess the anti-nociceptive effects of WIN-55,212, HU210, and PEA on MIA-induced OA in rats. Open-field locomotor activity system was used confirm the analgesic effects of those compounds. HU210, WIN55, 212, and PEA in a dose-dependent manner restored the paw withdrawal threshold (PWT) and the weight-bearing difference induced by MIA injection. SR141716A (a CB1 antagonist) significantly reversed the anti-nociceptive effects of all the administered drugs in terms of PWT. However, in terms of weight-bearing difference, SR141716A significantly reduced the anti-nociceptive effect of HU210 but not PEA or WIN55, 212. GW6471 (a PPARα antagonist) significantly reversed the anti-nociceptive effects of PEA but not those of HU210 or WIN55, 212. HU210, WIN55, 212 and PEA significantly restored the MIA-induced reduction in locomotor activity. In conclusions, both CB1 and PPARα receptors are involved in mediating pain in osteoarthritis. Therefore, targeting these receptors may be of great clinical value.


Asunto(s)
Analgésicos/farmacología , Cannabinoides/farmacología , Osteoartritis/tratamiento farmacológico , PPAR alfa/fisiología , Receptor Cannabinoide CB1/fisiología , Amidas , Animales , Dronabinol/análogos & derivados , Dronabinol/farmacología , Etanolaminas/farmacología , Ácido Yodoacético/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Osteoartritis/fisiopatología , Oxazoles/farmacología , Ácidos Palmíticos/farmacología , Ratas , Ratas Sprague-Dawley , Rimonabant/farmacología , Tirosina/análogos & derivados , Tirosina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...