Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Endocr Regul ; 54(4): 275-283, 2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33885253

RESUMEN

Objectives. Bisphenol A (BPA), as an indispensable plastic additive, has also been proven as an endocrine disruptor associated with adverse health effects including impaired ovarian function and cancer. Due to the restrictions of its usage, several analogs have been employed to replace BPA. Although many studies revealed a harmfulness in the biological effects of BPA analogs, their specific targets remain largely unknown. Nuclear receptors (NRs) may be one of the most important targets of bisphenols. Therefore, in this study, our attention was directed to explore the effect of BPA and its analogs, AF and S, on the mRNA expression of selected NRs involved in the steroidogenic and carcinogenic pathways in the human granulosa cell line COV434. The NRs investigated included: thyroid hormone receptor α (THRA), peroxisome proliferator activating receptor ß/δ (PPARD), retinoid X receptor α (RXRA), chicken ovalbumin upstream promoter-transcription factor II (COUPTFII), nuclear receptor-related protein 1 (NURR1), and liver receptor homolog-1 (LRH1).Methods. COV434 cells were treated with the bisphenols at the concentrations of 10-9 M, 10-7 M, and 10-5 M, and after 24 and 48 h, cell viability was monitored by the MTS assay and gene expressions were analyzed using RT-qPCR.Results. Bisphenol treatment did not alter the COV434 cell viability. After 24 h, the expression of neither of the NRs was changed. Likewise, after 48 h, the expression of the selected genes was not altered. However, both BPAF and BPS increased, at the highest concentration (10-5 M) used, the mRNA levels of both PPARD and NURR1 NRs after 48 h of the treatment. In the BPA-treated groups, no significant upregulation was observed.Conclusions. In the present study, the effect of bisphenols on COUP-TFII, Nurr1, and LRH-1 NRs was investigated for the first time. Although generally we did not observe that BPs provoked any alterations in the expression of the selected NRs in COV434 cells, at specific concentrations and time points they might alter mRNA expression of certain NRs (NURR1, PPARD).


Asunto(s)
Compuestos de Bencidrilo/farmacología , Disruptores Endocrinos/farmacología , Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Ovario/efectos de los fármacos , Fenoles/farmacología , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Compuestos de Bencidrilo/análisis , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Disruptores Endocrinos/análisis , Femenino , Humanos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/efectos de los fármacos , Ovario/citología , PPAR delta/efectos de los fármacos , Fenoles/análisis
2.
Am J Physiol Endocrinol Metab ; 316(5): E908-E921, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30807216

RESUMEN

The prevalence of cardiometabolic syndrome (CMS) is increased in women after menopause. While hormone replacement therapy has been prescribed to relieve several components of CMS in postmenopausal women, some aspects of cardiometabolic dysfunction cannot be completely restored. The present study examined the effectiveness of estrogen replacement alone and in combination with exercise by voluntary wheel running (VWR) for alleviating the risks of CMS, insulin-mediated skeletal muscle glucose transport, and hepatic fat accumulation in ovariectomized Sprague-Dawley rats fed a high-fat high-fructose diet (OHFFD). We compared a sham-operated group with OHFFD rats that were subdivided into a sedentary, estradiol replacement (E2), and E2 plus VWR for 12 wk. E2 prevented the development of insulin resistance in skeletal muscle glucose transport and decreased hepatic fat accumulation in OHFFD rats. Furthermore, E2 treatment decreased visceral fat mass and low-density lipoprotein (LDL)-cholesterol in OHFFD rats, while VWR further decreased LDL-cholesterol and increased the ratio of high-density lipoprotein-cholesterol to total cholesterol to a greater extent. Although E2 treatment alone did not reduce serum triglyceride levels in OHFFD rats, the combined intervention of E2 and VWR lowered serum triglycerides in E2-treated OHFFD rats. The addition of VWR to E2-treated OHFFD rats led to AMPK activation and upregulation of peroxisome proliferator-activated receptor-γ (PPARγ) coactivator-1α and PPARδ in skeletal muscle along with increased fatty acid oxidation and suppressed fatty acid synthesis in the liver. Collectively, our findings indicate that, to achieve greater health benefits, physical exercise is required for E2-treated individuals under ovarian hormone deprivation with high-energy consumption.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Hígado Graso/metabolismo , Hígado/efectos de los fármacos , Síndrome Metabólico/metabolismo , Actividad Motora , Músculo Esquelético/efectos de los fármacos , Animales , HDL-Colesterol/efectos de los fármacos , HDL-Colesterol/metabolismo , LDL-Colesterol/efectos de los fármacos , LDL-Colesterol/metabolismo , Dieta Alta en Grasa , Azúcares de la Dieta , Terapia de Reemplazo de Estrógeno , Femenino , Fructosa , Glucosa/metabolismo , Resistencia a la Insulina , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Menopausia , Músculo Esquelético/metabolismo , Ovariectomía , PPAR delta/efectos de los fármacos , PPAR delta/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Ratas , Ratas Sprague-Dawley
3.
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
4.
J Neurochem ; 130(4): 563-74, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24806616

RESUMEN

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a potential regulator of neuroinflammation. Toll-like receptors (TLR) are innate immunity-related receptors of inflammatory stimuli. In the present report, we evaluate the molecular mechanisms of regulation of mRNA, protein, and transcriptional activity levels of PPARß/δ by agonists of TLR4, TLR1/2, and TLR5, using lipopolysaccharide (LPS), peptidoglycan, and flagellin, respectively. We found that these stimuli increase the PPARß/δ levels in astrocytes. Expression and activity of PPARß/δ are separately regulated by inhibitors of p38, MEK1/2, extracellular signal-regulated kinases 1/2, and c-Jun N-terminal Kinase mitogen-activated protein kinases. The LPS-induced kinetics of PPARß/δ expression is similar to that of the proinflammatory gene cyclooxygenase 2. Moreover, for both genes the expression depends on nuclear factor kappa-light-chain-enhancer of activated B cells and p38, and is induced after inhibition of protein synthesis. The up-regulation of the expression after inhibition of protein synthesis signifies the participation of a labile protein in regulation of PPARß/δ expression. In contrast to cyclooxygenase 2, the cycloheximide-sensitive PPARß/δ expression was not responsive to nuclear factor kappa-light-chain-enhancer of activated B cells inhibition. Measurements of PPARß/δ mRNA stability showed that the PPARß/δ mRNA levels are regulated post-transcriptionally. We found that in LPS-stimulated astrocytes, the half-life of PPARß/δ mRNA was 50 min. Thus, we demonstrate that PPARß/δ expression and activity are regulated in TLR agonist-stimulated astrocytes by mechanisms that are widely used for regulation of proinflammatory genes. Protein expression level of nuclear receptor PPARß/δ is important for functions of this transcription factor. We investigate the regulatory mechanisms of PPARß/δ in rat primary astrocytes stimulated by agonists of toll-like receptors (TLR): TLR4, TLR1/2, and TLR5. Expression, activity, mRNA stability, and superinduction of PPARß/δ were up-regulated after TLR stimulation. These processes are sensitive to MAPKs and NF-kB inhibitors. Superinduction is up-regulation of mRNA expression after inhibition of protein synthesis.


Asunto(s)
Astrocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , PPAR delta/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Receptores Toll-Like/agonistas , Animales , Astrocitos/efectos de los fármacos , Western Blotting , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Inducción Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Lipopolisacáridos/farmacología , PPAR delta/efectos de los fármacos , PPAR delta/genética , Cultivo Primario de Células , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN/biosíntesis , ARN/genética , ARN/aislamiento & purificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
5.
Nutr Rev ; 78(3): 207-214, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31584650

RESUMEN

The peroxisome proliferator-activated receptor (PPAR) ß/δ has an important role in multiple inflammatory conditions, including obesity, hypertension, cancer, cardiovascular disease, diabetes mellitus, and autoimmune diseases. PPARß/δ forms a heterodimer with the retinoic acid receptor and binds to peroxisome proliferator response elements to initiate transcription of its target genes. PPARß/δ is also able to suppress the activities of several transcription factors, including nuclear factor κB, and activator protein 1, thus regulating anti-inflammatory cellular responses and playing a protective role in several diseases. Recent studies have shown that nutritional compounds, including nutrients and bioactive compounds, can regulate PPARß/δ expression. This review discusses key nutritional compounds that are known to modulate PPARß/δ and are likely to affect human health.


Asunto(s)
Dieta , Inflamación/metabolismo , PPAR delta/metabolismo , PPAR-beta/metabolismo , Animales , Curcumina/farmacología , Flavonoides/farmacología , Humanos , Inflamación/dietoterapia , FN-kappa B/metabolismo , PPAR delta/efectos de los fármacos , PPAR-beta/efectos de los fármacos , Fitoquímicos/farmacología , Polifenoles/farmacología , Receptores de Ácido Retinoico/metabolismo , Vitamina A/farmacología
6.
J Cell Physiol ; 214(2): 434-41, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17620284

RESUMEN

Prostacyclin (PGI2) has been shown to inhibit proliferation in vascular smooth muscle cells. To clarify the underlying molecular mechanism, we investigated the vasoprotection of beraprost (a PGI2 agonist) both in vivo and in vitro. Beraprost eliminated increases in proliferation of rat aortic smooth muscle cells (RASMCs) by 12-O-tetradecanoylphorbol 13-acetate, and enhanced the peroxisome proliferator-activated receptor-delta (PPARdelta) and inducible nitric oxide synthetase (iNOS) expressions, which were associated with the antiproliferative action of beraprost according to inhibition experiments by [(3)H]thymidine incorporation. Additionally, elimination of iNOS activity by PPARdelta antagonists suggested that iNOS is the downstream target of PPARdelta. Furthermore, beraprost increased both consensus PPARdelta-responsive element (PPRE)-driven luciferase activity and the binding activity of the PPARdelta to the putative PPRE in the iNOS promoter; nevertheless, it was abolished by PPARdelta antagonists. Deletion of PPRE (-1,349/-1,330) in the iNOS promoter region (-1,359/+2) strongly reduced promoter-driven activity, representing a novel mechanism of iNOS induction by beraprost. Consistent with this, PPARdelta and the concomitant iNOS induction by beraprost were also evident in vivo. Beraprost-mediated protection in a murine model of balloon angioplasty was significantly attenuated by 13S-HODE, a PPARdelta antagonist. Taken together, the results suggest that the causal relationship between PPARdelta and iNOS contributes to the vasoprotective action of beraprost in RASMCs.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Epoprostenol/análogos & derivados , Epoprostenol/agonistas , Músculo Liso Vascular/efectos de los fármacos , Animales , Aorta Torácica/citología , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inducción Enzimática/efectos de los fármacos , Epoprostenol/farmacología , Formazáns/análisis , Formazáns/metabolismo , Eliminación de Gen , Genes Reporteros , Luciferasas/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Técnicas de Cultivo de Órganos , PPAR delta/efectos de los fármacos , PPAR delta/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Elementos de Respuesta/efectos de los fármacos , Elementos de Respuesta/genética , Acetato de Tetradecanoilforbol/farmacología , Sales de Tetrazolio/análisis , Sales de Tetrazolio/metabolismo , Timidina/antagonistas & inhibidores , Factores de Tiempo , Transfección
7.
FEBS Lett ; 582(1): 26-31, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18036566

RESUMEN

PPAR delta is the only member in the PPAR subfamily of nuclear receptors that is not a target of current drugs. Animal studies demonstrate PPAR delta activation exerts many favorable effects, including reducing weight gain, increasing skeletal muscle metabolic rate and endurance, improving insulin sensitivity and cardiovascular function and suppressing atherogenic inflammation. These activities stem largely from the ability of PPAR delta to control energy balance, reduce fat burden and protect against lipotoxicity caused by ectopic lipid deposition. Therefore, PPAR delta represents a novel therapeutic target and the development of PPAR delta gonists/modulators may be useful for treating the whole spectrum of metabolic syndrome.


Asunto(s)
Síndrome Metabólico/tratamiento farmacológico , PPAR delta/efectos de los fármacos , Humanos , PPAR delta/metabolismo
8.
Curr Opin Investig Drugs ; 9(3): 247-55, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18311660

RESUMEN

Nuclear hormone receptors, including peroxisome proliferator-activated receptors (PPARs), liver X receptors (LXRs), and the farnesoid X receptor (FXR), are transcription factors involved in the regulation of essential metabolic functions, including glucose and lipid metabolism, reverse cholesterol transport, and the regulation of bile acids. This review summarizes new developments in the use of PPAR, LXR and FXR agonists for the treatment of obesity and cardiovascular diseases, including dyslipidemia and atherosclerosis. Currently available drugs and future areas of research for new therapies are also discussed.


Asunto(s)
Fármacos Antiobesidad/farmacología , Aterosclerosis/tratamiento farmacológico , Dislipidemias/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Animales , Proteínas de Unión al ADN/efectos de los fármacos , Humanos , Receptores X del Hígado , Receptores Nucleares Huérfanos , PPAR alfa/efectos de los fármacos , PPAR delta/efectos de los fármacos , PPAR gamma/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos
9.
Eur J Pharmacol ; 584(1): 192-201, 2008 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-18346728

RESUMEN

The use of the thiazolidinedione insulin sensitizers rosiglitazone and pioglitazone for the treatment of type 2 diabetes mellitus in recent years has proven to be effective in helping patients resume normal glycemic control. However, their use is often associated with undesirable side effects including peripheral edema, congestive heart failure and weight gain. Here, we report the identification and characterization of a novel selective PPARgamma modulator, SPPARgammaM5 ((2S)-2-(2-chloro-5-{[3-(4-chlorophenoxy)-2-methyl-6-(trifluoromethoxy)-1H-indol-1-yl]methyl} phenoxy)propionic acid), which has notable insulin sensitizing properties and a superior tolerability profile to that of rosiglitazone. SPPARgammaM5 is a potent ligand of human PPARgamma with high selectivity versus PPARalpha or PPARdelta in receptor competitive binding assays. In cell-based transcriptional activation assays, SPPARgammaM5 was a potent partial agonist of human PPARgamma in comparison to the PPARgamma full agonist rosiglitazone. Compared to rosiglitazone or the PPARgamma full agonist COOH (2-(2-(4-phenoxy-2-propylphenoxy)ethyl)indole-5-acetic acid), SPPARgammaM5 induced an attenuated PPARgamma-regulated gene expression profile in fully differentiated 3T3-L1 adipocytes and white adipose tissue of chronically treated db/db mice. SPPARgammaM5 treatment also reduced the insulin resistance index by homeostasis model assessment (HOMA), suggesting an improvement in insulin resistance in these db/db mice. Treatment of obese Zucker rats with either rosiglitazone or SPPARgammaM5 resulted in an improvement in selected parameters that serve as surrogate indicators of insulin resistance and hyperlipidemia. However, unlike rosiglitazone, SPPARgammaM5 did not cause significant fluid retention or cardiac hypertrophy in these rats. Thus, compounds such as SPPARgammaM5 may offer beneficial effects on glycemic control with significantly attenuated adverse effects.


Asunto(s)
Acetatos/farmacología , Enfermedades Cardiovasculares/inducido químicamente , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Indoles/farmacología , Resistencia a la Insulina , PPAR gamma/efectos de los fármacos , Propionatos/farmacología , Tiazolidinedionas/farmacología , Células 3T3-L1 , Acetatos/efectos adversos , Acetatos/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Células COS , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Chlorocebus aethiops , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Agonismo Parcial de Drogas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hemodilución , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/metabolismo , Indoles/efectos adversos , Indoles/metabolismo , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos , PPAR alfa/efectos de los fármacos , PPAR alfa/metabolismo , PPAR delta/efectos de los fármacos , PPAR delta/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Propionatos/efectos adversos , Propionatos/metabolismo , Unión Proteica , Ratas , Ratas Zucker , Rosiglitazona , Tiazolidinedionas/efectos adversos , Tiazolidinedionas/metabolismo , Activación Transcripcional/efectos de los fármacos , Transfección , Equilibrio Hidroelectrolítico/efectos de los fármacos
10.
J Anim Sci ; 96(7): 2763-2776, 2018 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-29688535

RESUMEN

All-trans retinoic acid (ATRA) has been associated with various physiological phenomenon in mammalian adipose tissue and skeletal muscle. We hypothesized that ATRA may affect skeletal muscle fiber type in bovine satellite cell culture through various transcriptional processes. Bovine primary satellite cell (BSC) culture experiments were conducted to determine dose effects of ATRA on expression of genes and protein levels related to skeletal muscle fiber type and metabolism. The semimembranosus from crossbred steers (n = 2 steers), aged approximately 24 mo, were used to isolate BSC for 3 separate assays. Myogenic differentiation was induced using 3% horse serum upon cultured BSC with increasing doses (0, 1, 10, 100, and 1,000 nM) of ATRA. After 96 h of incubation, cells were harvested and used to measure the gene expression of protein kinase B (Akt), AMP-activated protein kinase alpha (AMPK), glucose transporter 4 (GLUT4), myogenin, lipoprotein lipase (LPL), myosin heavy chain (MHC) I, MHC IIA, MHC IIX, insulin like growth factor-1 (IGF-1), Peroxisome proliferator activated receptor gamma (PPARγ), PPARδ, and Smad transcription factor 3 (SMAD3) mRNA relative to ribosomal protein subunit 9 (RPS9). The mRNA expression of LPL was increased (P < 0.05) with 100 and 1,000 nM of ATRA. Expression of GLUT4 was altered (P < 0.05) by ATRA. The treatment of ATRA (1,000 nM) also increased (P < 0.05) mRNA gene expression of SMAD3. The gene expression of both PPARδ and PPARγ were increased (P < 0.05) with 1,000 nM of ATRA. Protein level of PPARδ was also affected (P < 0.05) by 1,000 nM of ATRA and resulted in a greater (P < 0.05) protein level of PPARδ compared to CON. All-trans retinoic acid (10 nM) increased gene expression of MHC I (P < 0.05) compared to CON. Expression of MHC IIA was also influenced (P < 0.05) by ATRA. The mRNA expression of MHC IIX was decreased (P < 0.05) with 100 and 1,000 nM of ATRA. In muscle cells, ATRA may cause muscle fibers to transition towards the MHC isoform that prefers oxidative metabolism, as evidenced by increased expression of genes associated with the MHC I isoform. These changes in MHC isoforms appeared to be brought about by changing PPARδ gene expression and protein levels.


Asunto(s)
Bovinos/fisiología , Cadenas Pesadas de Miosina/efectos de los fármacos , PPAR delta/efectos de los fármacos , Tretinoina/farmacología , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Animales , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Miogenina/efectos de los fármacos , Miogenina/genética , Cadenas Pesadas de Miosina/genética , Oxidación-Reducción , PPAR delta/genética , Células Satélite del Músculo Esquelético
11.
Sci Rep ; 7(1): 14021, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-29070884

RESUMEN

Major depression is a common mental disorder that has been established to be associated with a decrease in serotonin and/or serotonin transporters in the brain. Peroxisome proliferator-activated receptor δ (PPARδ) has been introduced as a potential target for depression treatment. Telmisartan was recently shown to activate PPARδ expression; therefore, the effectiveness of telmisartan in treating depression was investigated. In unpredictable chronic mild stress (UCMS) model, treatment with telmisartan for five weeks notably decrease in the time spent in the central and the reduced frequency of grooming and rearing in open filed test (OFT) and the decreased sucrose consumption in sucrose preference test (SPT) compared with the paradigms. Telmisartan also reversed the decrease in PPARδ and 5-HTT levels in the hippocampus of depression-like mice. Administration of PPARδ antagonist GSK0660 and direct infusion of sh-PPARδ into the brain blocked the effects of telmisartan on the improvement of depression-like behavior in these mice. Moreover, telmisartan enhanced the expression of PPARδ and 5HTT in H19-7 cells. In conclusion, the obtained results suggest that telmisartan improves symptoms of stress-induced depression in animals under chronic stress through activation of PPARδ. Therefore, telmisartan may be developed as a potential anti-depressant in the future.


Asunto(s)
Depresión/tratamiento farmacológico , PPAR delta/efectos de los fármacos , Estrés Fisiológico , Telmisartán/uso terapéutico , Animales , Línea Celular , Depresión/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Hipocampo/metabolismo , Ratones , Ratones Endogámicos BALB C , PPAR delta/genética , PPAR delta/metabolismo , Telmisartán/farmacología
12.
Hypertension ; 69(4): 641-650, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28242713

RESUMEN

Women with systemic lupus erythematosus exhibit a high prevalence of hypertension, endothelial dysfunction, and renal injury. We tested whether GW0742, a peroxisome proliferator activator receptor ß/δ (PPARß/δ) agonist, ameliorates disease activity and cardiovascular complications in a female mouse model of lupus. Thirty-week-old NZBWF1 (lupus) and NZW/LacJ (control) mice were treated with GW0742 or with the PPARß/δ antagonist GSK0660 plus GW0742 for 5 weeks. Blood pressure, plasma double-stranded DNA autoantibodies and cytokines, nephritis, hepatic opsonins, spleen lymphocyte populations, endothelial function, and vascular oxidative stress were compared in treated and untreated mice. GW0742 treatment reduced lupus disease activity, blood pressure, cardiac and renal hypertrophy, splenomegaly, albuminuria, and renal injury in lupus mice, but not in control. GW0742 increased hepatic opsonins mRNA levels in lupus mice and reduced the elevated T, B, Treg, and Th1 cells in spleens from lupus mice. GW0742 lowered the higher plasma concentration of proinflammatory cytokines observed in lupus mice. Aortae from lupus mice showed reduced endothelium-dependent vasodilator responses to acetylcholine and increased nicotinamide adenine dinucleotide phosphate oxidase-driven vascular reactive oxygen species production, which were normalized by GW0742 treatment. All these effects of GW0742 were inhibited by PPARß/δ blockade with GSK0660. Pharmacological activation of PPARß/δ reduced hypertension, endothelial dysfunction, and organ damage in severe lupus mice, which was associated with reduced plasma antidouble-stranded DNA autoantibodies and anti-inflammatory and antioxidant effects in target tissues. Our findings identify PPARß/δ as a promising target for an alternative approach in the treatment of systemic lupus erythematosus and its associated vascular damage.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Activación Enzimática , Hipertensión/tratamiento farmacológico , Lupus Eritematoso Sistémico/tratamiento farmacológico , PPAR delta/metabolismo , Tiazoles/farmacología , Animales , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Femenino , Hipertensión/enzimología , Hipertensión/etiología , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/enzimología , Ratones , Ratones Endogámicos NZB , PPAR delta/efectos de los fármacos
13.
Minerva Med ; 108(1): 1-12, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27706112

RESUMEN

BACKGROUND: TRPV4, a non-selective cation channel, is involved in lipometabolism and atherosclerosis. However, whether TRPV4 participates in oxygenized low density lipoprotein (oxLDL)-induced foam cell formation remains unknown. The present study investigates the effect of oxLDL on the expression of TRPV4 in macrophages and its underlying mechanisms. METHODS: The expression of TRPV4 in RAW264.7 and phorbol-12-myristate-13-acetate (PMA) induced U937, THP-1 cells was detected by immunofluorescence, and western blot was used to detect the TRPV4 expression before and after PMA induction. Each cell line was divided into three groups, including control group, native low-density lipoprotein (nLDL) (100 µg/mL) group and oxLDL (100µg/mL) group; the expression of TRPV4 in each group was measured using immunohistochemistry and western blot. TRPV4 protein expression was detected by western blot after RAW 264.7 cells were treated with 0, 0.01 µM, 0.1 µM and 1 µM T0070907 or preincubated with 0.1 µM T0070709 for 1 h before incubation with oxLDL for 24 h. RESULTS: In all macrophage cell lines, TRPV4 was widely expressed. PMA increased TRPV4 expression in U937 and THP-1 cells. There was no significant difference in TRPV4 expression in the nLDL group compared to that in the control group; however a significant reduction in TRPV4 expression was detected in the oxLDL group compared to that in the control and nLDL groups using measurements obtained from both immunohistochemistry and western blot. The PPARγ inhibitor T0070907 enhanced the basal expression of TRPV4 and protected RAW264.7 cells from oxLDL-induced TRPV4 down-regulation. CONCLUSIONS: This study revealed that TRPV4 was widely expressed in macrophages and that oxLDL could induce the down-regulation of TRPV4 expression through its actions on PPARγ. This study may serve as an important first step for further investigation into the roles of TRPV4 in macrophage-derived foam cell formation in atherosclerosis.


Asunto(s)
Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , PPAR gamma/fisiología , Animales , Benzamidas/farmacología , Antígenos CD36/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Macrófagos/metabolismo , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , PPAR delta/efectos de los fármacos , PPAR gamma/antagonistas & inhibidores , Piridinas/farmacología , Células RAW 264.7 , Acetato de Tetradecanoilforbol/farmacología , Células U937
14.
Curr Opin Investig Drugs ; 7(4): 360-70, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16625823

RESUMEN

GlaxoSmithKline and Ligand are developing GW-501516, a peroxisome proliferator-activator receptor-delta agonist for the potential treatment of dyslipidemia. Phase II clinical trials of this compound are ongoing.


Asunto(s)
Hipolipemiantes/farmacología , Tiazoles/farmacología , Animales , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Humanos , Macaca mulatta , Ratones , Ratones Obesos , PPAR delta/efectos de los fármacos , PPAR delta/fisiología , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/uso terapéutico
15.
J Dairy Sci ; 89(10): 3826-32, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16960057

RESUMEN

Recent interest in conjugated linoleic acid (CLA) research stems from the well-documented anticarcinogenic, antiatherogenic, antidiabetic, and antiobesity properties of CLA in animal models. The objective of this study was to examine the effects of 2 CLA isomers (cis-9,trans-11 and trans-10,cis-12) on phorbol 12,13-dibutyrate (PDBu)-induced PGF2alpha production in cultured bovine endometrial (BEND) cells. Confluent BEND cells were incubated in the absence (control) or presence of 100 microM each of linoleic acid, cis-9,trans-11 CLA, or trans-10,cis-12 CLA for 24 h. After incubation, cells were rinsed and then stimulated with PDBu (100 ng/mL) for 6 h. Compared with untreated cells, PDBu stimulated PGF2alpha secretion (+25-fold) within 6 h. The increases in PGF(2alpha) secretion were paralleled by signifi-cant induction of prostaglandin endoperoxide synthase-2 (PGHS-2) mRNA (+63-fold) and protein (+1.6-fold) expression. In spite of stimulatory effects on PGHS-2 and peroxisome proliferator-activated receptor delta (PPARdelta) mRNA responses, CLA greatly decreased PGF2alpha production by PDBu-stimulated BEND cells. There was no evidence for PDBu or CLA modulation of PPARdelta protein synthesis in cultured BEND cells. Results indicated that CLA modulation of PGF2alpha production by BEND cells was not mediated through PGHS-2 or PPARdelta gene repression.


Asunto(s)
Bovinos/metabolismo , Dinoprost/biosíntesis , Endometrio/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Forbol 12,13-Dibutirato/farmacología , Animales , Células Cultivadas , Ciclooxigenasa 2/efectos de los fármacos , Cartilla de ADN/química , Endometrio/citología , Endometrio/metabolismo , Femenino , PPAR delta/análisis , PPAR delta/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/análisis , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos
16.
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
17.
Curr Pharm Des ; 21(21): 2956-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26004416

RESUMEN

Peroxisome proliferator-activated receptorδ (PPARδ) belongs to the PPARs receptor family including PPARα, PPARδ, and PPARγ. PPARδ is a ligand-activated transcription factor that plays a critical role in regulating cancer progression. PPARδ-linked tumorigenesis was first identified in colorectal cancer, which is demonstrated by the following evidences, so PPARε is a potential drug target for colorectal cancer. In contrast, some observations show that PPARδ negatively regulates colorectal cancer event. In the present review, the recent progress of PPARδ signaling-mediated colorectal cancer is covered.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/fisiopatología , PPAR delta/genética , Animales , Antineoplásicos/uso terapéutico , Humanos , PPAR delta/efectos de los fármacos , PPAR gamma , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , beta Catenina/genética
18.
J Dermatol Sci ; 80(3): 186-95, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26481780

RESUMEN

BACKGROUND: The phenotypic changes that accompany differentiation of resident fibroblasts into myofibroblasts are important aspects of the wound healing process. Recent studies showed that peroxisome proliferator-activated receptor (PPAR) δ plays a critical role in wound healing. OBJECTIVE: To determine whether the nuclear receptor PPARδ can modulate the differentiation of human dermal fibroblasts (HDFs) into myofibroblasts. METHODS: These studies were undertaken in primary HDFs using Western blot analyses, small interfering (si)RNA-mediated gene silencing, reporter gene assays, chromatin immunoprecipitation (ChIP), migration assays, collagen gel contraction assays, and real-time PCR. RESULTS: Activation of PPARδ by GW501516, a specific ligand of PPARδ, specifically upregulated the myofibroblast marker α-smooth muscle actin (α-SMA) in a time- and concentration-dependent manner. This induction was significantly inhibited by the presence of siRNA against PPARδ, indicating that PPARδ is involved in myofibroblast transdifferentiation of HDFs. Ligand-activated PPARδ increased α-SMA promoter activity in a dual mode by directly binding a direct repeat-1 (DR1) site in the α-SMA promoter, and by inducing expression of transforming growth factor (TGF)-ß, whose downstream effector Smad3 interacts with a Smad-binding element (SBE) in another region of the promoter. Mutations in these cis-elements totally abrogated transcriptional activation of the α-SMA gene by the PPARδ ligand; thus both sites represent novel types of PPARδ response elements. GW501516-activated PPARδ also increased the migration and contractile properties of HDFs, as demonstrated by Transwell and collagen lattice contraction assays, respectively. In addition, PPARδ-mediated upregulation of α-SMA was correlated with elevated expression of myofibroblast markers such as collagen I and fibronectin, with a concomitant reduction in expression of the epithelial marker E-cadherin. CONCLUSION: PPARδ plays pivotal roles in wound healing by promoting fibroblast-to-myofibroblast differentiation via TGF-ß/Smad3 signaling.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular , PPAR delta/efectos de los fármacos , PPAR delta/metabolismo , Tiazoles/farmacología , Cicatrización de Heridas , Actinas/genética , Cadherinas/metabolismo , Movimiento Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Fibroblastos , Fibronectinas/metabolismo , Humanos , Ligandos , PPAR delta/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño/farmacología , Elementos de Respuesta , Transducción de Señal , Piel/citología , Proteína smad3/metabolismo , Activación Transcripcional , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
J Nutr Biochem ; 26(10): 1058-67, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26101135

RESUMEN

The initiation of obesity entails an imbalance wherein energy intake exceeds expenditure. Obesity is increasing in prevalence and is now a worldwide health problem. Food-derived peroxisome proliferator-activated receptor δ (PPARδ) stimulators represent potential treatment options for obesity. Ginger (Zingiber officinale Roscoe) was previously shown to regulate the PPARγ signaling pathway in adipocytes. In this study, we investigated the antiobesity effects of ginger in vivo and the mechanism of action in vitro. Energy expenditure was increased, and diet-induced obesity was attenuated in C57BL/6J mice treated with dietary ginger extract (GE). GE also increased the number of Type I muscle fibers, improved running endurance capacity and upregulated PPARδ-targeted gene expression in skeletal muscle and the liver. 6-Shogaol and 6-gingerol acted as specific PPARδ ligands and stimulated PPARδ-dependent gene expression in cultured human skeletal muscle myotubes. An analysis of cellular respiration revealed that pretreating cultured skeletal muscle myotubes with GE increased palmitate-induced oxygen consumption rate, which suggested an increase in cellular fatty acid catabolism. These results demonstrated that sustained activation of the PPARδ pathway with GE attenuated diet-induced obesity and improved exercise endurance capacity by increasing skeletal muscle fat catabolism. 6-Shogaol and 6-gingerol may be responsible for the regulatory effects of dietary ginger on PPARδ signaling.


Asunto(s)
Fármacos Antiobesidad , Dieta Alta en Grasa , Obesidad/prevención & control , PPAR delta/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Zingiber officinale/química , Tejido Adiposo/citología , Animales , Línea Celular , Dieta , Metabolismo Energético/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Obesidad/etiología , PPAR delta/fisiología , Resistencia Física/efectos de los fármacos
20.
J Mol Endocrinol ; 33(2): 533-44, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15525607

RESUMEN

Fatty acids are an important ligand for peroxisome proliferator-activated receptor (PPAR) activation and transcriptional regulation of metabolic genes. To examine whether reduced plasma free fatty acid (FFA) availability affects the mRNA content of proteins involved in fuel metabolism in vivo, the skeletal muscle mRNA content of various transcription factors, transcriptional coactivators and genes encoding for lipid regulatory proteins were examined before and after 3 h of cycle exercise with (NA) and without (CON) pre-exercise ingestion of nicotinic acid (NA). NA resulted in a marked (3- to 6-fold) increase (P<0.05) in PPARalpha, PPARdelta and PPAR coactivator 1alpha (PGC1alpha) mRNA, but was without effect on nuclear respiratory factor-1 and Forkhead transcription factor, fatty acid transcolase/CD36, carnitine palmitoyl transferase 1, hormone sensitive lipase (HSL) and pyruvate dehydrogenase kinase 4. Exercise in CON was associated with increased (P<0.05) PPARalpha, PPARdelta and PGC1alpha mRNA, which was similar in magnitude to levels observed with NA at rest. Exercise was generally without effect on the mRNA content of lipid regulatory proteins in CON and did not affect the mRNA content of the measured subset of transcription factors, transcriptional co-activators and lipid regulatory proteins during NA. To determine the possible mechanisms by which NA might affect PGC1alpha expression, we measured p38 MAP kinase (MAPK) and plasma epinephrine. Phosphorylation of p38 MAPK was increased (P<0.05) by NA treatment at rest, and this correlated (r2=0.84, P<0.01) with increased PGC1alpha. Despite this close relationship, increasing p38 MAPK in human primary myotubes was without effect on PGC1alpha mRNA content. Plasma epinephrine was elevated (P<0.05) by NA at rest (CON: 0.27+/-0.06, NA: 0.72+/-0.11 nM) and throughout exercise. Incubating human primary myotubes with epinephrine increased PGC1alpha independently of changes in p38 MAPK phosphorylation. Hence, despite the fact that NA ingestion decreased FFA availability, it promoted the induction of PPARalpha/delta and PGC1alpha gene expression to a similar degree as prolonged exercise. We suggest that the increase in PGC1alpha may be due to the elevated plasma epinephrine levels. Despite these changes in transcription factors/coactivators, the mRNA content of lipid regulatory proteins was generally unaffected by plasma FFA availability.


Asunto(s)
Ácidos Grasos/sangre , Proteínas de Choque Térmico/metabolismo , Hipolipemiantes/farmacología , Músculo Esquelético/fisiología , Niacina/farmacología , PPAR alfa/metabolismo , PPAR delta/metabolismo , Factores de Transcripción/metabolismo , Antígenos CD36/efectos de los fármacos , Antígenos CD36/genética , Calcimicina/farmacología , Carnitina O-Palmitoiltransferasa/efectos de los fármacos , Carnitina O-Palmitoiltransferasa/genética , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Epinefrina/sangre , Prueba de Esfuerzo , Factores de Transcripción Forkhead , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/genética , Humanos , Imidazoles/farmacología , Lípidos/genética , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , PPAR alfa/efectos de los fármacos , PPAR alfa/genética , PPAR delta/efectos de los fármacos , PPAR delta/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosforilación , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/genética , Piridinas/farmacología , Esterol Esterasa/efectos de los fármacos , Esterol Esterasa/genética , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA