Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 33(2): 2553-2562, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30285581

RESUMEN

The implication of αß and γδ T cells in obesity-associated inflammation and insulin resistance (IR) remains uncertain. Mice lacking γδ T cells show either no difference or a decrease in high-fat diet (HFD)-induced IR, whereas partial depletion in γδ T cells does not protect from HFD-induced IR. αß T-cell deficiency leads to a decrease in white adipose tissue (WAT) inflammation and IR without weight change, but partial depletion of these cells has not been studied. We previously described a mouse model overexpressing peroxisome proliferator-activated receptor ß (PPAR-ß) specifically in T cells [transgenic (Tg) T-PPAR-ß] that exhibits a partial depletion in αß T cells and no change in γδ T-cell number. This results in a decreased αß/γδ T-cell ratio in lymphoid organs. We now show that Tg T-PPAR-ß mice are partially protected against HFD-induced weight gain and exhibit decreased IR and liver steatosis independently of animal weight. These mice display an alteration of WAT-depots distribution with an increased epididymal-WAT mass and a decreased subcutaneous WAT mass. Immune cell number is decreased in both WAT-depots, except for γδ T cells, which are increased in epididymal-WAT. Overall, we show that decreasing αß/γδ T-cell ratio in WAT-depots alters their inflammatory state and mass repartition, which might be involved in improvement of insulin sensitivity.-Le Menn, G., Sibille, B., Murdaca, J., Rousseau, A.-S., Squillace, R., Vergoni, B., Cormont, M., Niot, I., Grimaldi, P. A., Mothe-Satney, I., Neels, J. G. Decrease in αß/γδ T-cell ratio is accompanied by a reduction in high-fat diet-induced weight gain, insulin resistance, and inflammation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Inflamación/prevención & control , Resistencia a la Insulina , Obesidad/prevención & control , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Aumento de Peso , Animales , Peso Corporal , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Intolerancia a la Glucosa/prevención & control , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Linfocitos T/inmunología
2.
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
3.
Sci Rep ; 6: 34317, 2016 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-27680392

RESUMEN

Metabolism plays an important role in T cell biology and changes in metabolism drive T cell differentiation and fate. Most research on the role of metabolism in T lymphocytes focuses on mature T cells while only few studies have investigated the role of metabolism in T cell development. In this study, we report that activation or overexpression of the transcription factor Peroxisome Proliferator-Activated Receptor ß (PPARß) increases fatty acid oxidation in T cells. Furthermore, using both in vivo and in vitro models, we demonstrate that PPARß activation/overexpression inhibits thymic T cell development by decreasing proliferation of CD4-CD8- double-negative stage 4 (DN4) thymocytes. These results support a model where PPARß activation/overexpression favours fatty acid- instead of glucose-oxidation in developing T cells, thereby hampering the proliferative burst normally occurring at the DN4 stage of T cell development. As a consequence, the αß T cells that are derived from DN4 thymocytes are dramatically decreased in peripheral lymphoid tissues, while the γδ T cell population remains untouched. This is the first report of a direct role for a member of the PPAR family of nuclear receptors in the development of T cells.

4.
FASEB J ; 30(3): 1287-99, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26655383

RESUMEN

We hypothesized that α-lipoic acid (α-LA) might interact with the transcriptional control of peroxisome proliferator-activated receptor (PPAR)ß in skeletal muscle. Molecular mechanisms were investigated using differentiated C2C12 myotubes treated with α-LA and/or PPARß agonist GW0742. In vivo studies with 3-mo-old C57Bl6 mice were realized: voluntary wheel running (VWR) training (7 wk), and a 6 wk diet containing (or not) α-LA (0.25% wt/wt). This last condition was combined with (or not) 1 bout of treadmill exercise (18 m/min for 1 h). Using a reporter assay, we demonstrate that α-LA is not an agonist of PPARß but regulates PPARß target gene expression through an active PPARß pathway. GW0742-induced pyruvate dehydrogenase kinase 4 mRNA is potentiated by α-LA. In C2C12, α-LA lowers the activation of the JNK signaling pathway and increases PPARß mRNA and protein levels (2-fold) to the same extent as with the JNK inhibitor SP600125. Similarly to VWR training effect, PPARß expression increases (2-fold) in vastus lateralis of animals fed an α-LA-enriched diet. However, α-LA treatment does not further stimulate the adaptive up-regulation of PPARß observed in response to 1 bout of exercise. We have identified a novel mechanism of regulation of PPARß expression/action in skeletal muscle with potential physiologic application through the action of α-LA, involving the JNK pathway.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , PPAR-beta/metabolismo , Transducción de Señal/efectos de los fármacos , Ácido Tióctico/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/métodos , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
5.
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
6.
Diabetes ; 61(9): 2311-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22688342

RESUMEN

Leukotrienes (LTs) are potent proinflammatory mediators, and many important aspects of innate and adaptive immune responses are regulated by LTs. Key members of the LT synthesis pathway are overexpressed in adipose tissue (AT) during obesity, resulting in increased LT levels in this tissue. We observed that several mouse adipocyte cell lines and primary adipocytes from mice and humans both can secrete large amounts of LTs. Furthermore, this production increases with a high-fat diet (HFD) and positively correlates with adipocyte size. LTs produced by adipocytes play an important role in attracting macrophages and T cells in in vitro chemotaxis assays. Mice that are deficient for the enzyme 5-lipoxygenase (5-LO), and therefore lack LTs, exhibit a decrease in HFD-induced AT macrophage and T-cell infiltration and are partially protected from HFD-induced insulin resistance. Similarly, treatment of HFD-fed wild-type mice with the 5-LO inhibitor Zileuton also results in a reduction of AT macrophages and T cells, accompanied by a decrease in insulin resistance. Together, these findings suggest that LTs represent a novel target in the prevention or treatment of obesity-associated inflammation and insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Inflamación/etiología , Resistencia a la Insulina/fisiología , Leucotrienos/metabolismo , Obesidad/complicaciones , Tejido Adiposo/metabolismo , Animales , Araquidonato 5-Lipooxigenasa/deficiencia , Línea Celular , Quimiocinas/sangre , Citocinas/sangre , Dieta Alta en Grasa , Femenino , Humanos , Hidroxiurea/análogos & derivados , Hidroxiurea/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Grasa Subcutánea/metabolismo
7.
Curr Opin Lipidol ; 21(3): 186-91, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20480546

RESUMEN

PURPOSE OF REVIEW: This review focuses on the emerging knowledge about peroxisome proliferator-activated receptor beta regulatory functions on metabolism, inflammation, and cellular stress. RECENT FINDINGS: Recent publications have confirmed the important roles of peroxisome proliferator-activated receptor beta in adaptive metabolic responses of skeletal muscle and have also implicated the nuclear receptor in the regulation of inflammation and oxidative stress in various tissues. The mechanisms implicated in these effects have been partially elucidated. SUMMARY: Peroxisome proliferator-activated receptors mediate the transcriptional effects of fatty acids and fatty acid derivatives and regulate many physiological functions, including metabolism and development. Use of potent and specific agonists revealed that activation of peroxisome proliferator-activated receptor beta efficiently reverses some metabolic syndrome-associated abnormalities by affecting metabolic, inflammatory, and oxidative stress responses in several tissues through both genomic and nongenomic modes of action.


Asunto(s)
Inflamación/metabolismo , Síndrome Metabólico/metabolismo , Estrés Oxidativo , PPAR-beta/metabolismo , Animales , Humanos , Inflamación/fisiopatología , Síndrome Metabólico/tratamiento farmacológico , Fibras Musculares Esqueléticas/metabolismo , Neovascularización Fisiológica
8.
Int J Mol Med ; 24(1): 63-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19513536

RESUMEN

C2C12 cells exposed to hyperthermia (41 degrees C) experienced an increase in both protein synthesis and degradation. The addition of IL15 under hyperthermic conditions resulted in an important increase in protein synthesis with no changes in protein degradation, except when cells overexpressed PPARdelta. The PPARdelta agonist GW501516 exerted similar effects on protein synthesis to IL15. Expression of a mutant dominant negative form of PPARdelta prevented the effect of the cytokine on protein synthesis, suggesting that this transcription factor is involved in the anabolic action of IL15. The present study also suggests that the effects of IL15 on lipid oxidation could be mediated by PPARdelta.


Asunto(s)
Calor , Interleucina-15/fisiología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , PPAR delta/metabolismo , Animales , Línea Celular , Interleucina-15/farmacología , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Mutación , PPAR delta/antagonistas & inhibidores , PPAR delta/genética , Biosíntesis de Proteínas , Tiazoles/farmacología
9.
Am J Physiol Endocrinol Metab ; 295(2): E297-304, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18492772

RESUMEN

Recent studies have shown that administration of peroxisome proliferator-activated receptor-beta (PPARbeta) agonists enhances fatty acid oxidation in rodent and human skeletal muscle and that muscle-restricted PPARbeta overexpression affects muscle metabolic profile by increasing oxidative myofiber number, which raises the possibility that PPARbeta agonists alter muscle morphology in adult animals. This possibility was examined in this study in which adult mice were treated with a PPARbeta agonist, and the resulting changes in myofiber metabolic phenotype and angiogenesis were quantified in tibialis anterior muscles. The findings indicate a muscle remodeling that is completed within 2 days and is characterized by a 1.63-fold increase in oxidative fiber number and by a 1.55-fold increase in capillary number. These changes were associated with a quick and transient upregulation of myogenic and angiogenic markers. Both myogenic and angiogenic responses were dependent on the calcineurin pathway, as they were blunted by cyclosporine A administration. In conclusion, the data indicate that PPARbeta activation is associated with a calcineurin-dependent effect on muscle morphology that enhances the oxidative phenotype.


Asunto(s)
Calcineurina/fisiología , Músculo Esquelético/fisiología , PPAR-beta/agonistas , Condicionamiento Físico Animal/fisiología , Tiazoles/farmacología , Animales , Inhibidores de la Calcineurina , Ciclosporina/farmacología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Proteína MioD/fisiología , Factor 5 Regulador Miogénico/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Succinato Deshidrogenasa/metabolismo
10.
J Biol Chem ; 283(21): 14317-26, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18308721

RESUMEN

The transcription factor FoxO1 contributes to the metabolic adaptation to fasting by suppressing muscle oxidation of glucose, sparing it for glucose-dependent tissues. Previously, we reported that FoxO1 activation in C(2)C(12) muscle cells recruits the fatty acid translocase CD36 to the plasma membrane and increases fatty acid uptake and oxidation. This, together with FoxO1 induction of lipoprotein lipase, would promote the reliance on fatty acid utilization characteristic of the fasted muscle. Here, we show that CD36-mediated fatty acid uptake, in turn, up-regulates protein levels and activity of FoxO1 as well as its target PDK4, the negative regulator of glucose oxidation. Increased fatty acid flux or enforced CD36 expression in C(2)C(12) cells is sufficient to induce FoxO1 and PDK4, whereas CD36 knockdown has opposite effects. In vivo, CD36 loss blunts fasting induction of FoxO1 and PDK4 and the associated suppression of glucose oxidation. Importantly, CD36-dependent regulation of FoxO1 is mediated by the nuclear receptor PPARdelta/beta. Loss of PPARdelta/beta phenocopies CD36 deficiency in blunting fasting induction of muscle FoxO1 and PDK4 in vivo. Expression of PPARdelta/beta in C(2)C(12) cells, like that of CD36, robustly induces FoxO1 and suppresses glucose oxidation, whereas co-expression of a dominant negative PPARdelta/beta compromises FoxO1 induction. Finally, several PPRE sites were identified in the FoxO1 promoter, which was responsive to PPARdelta/beta. Agonists of PPARdelta/beta were sufficient to confer responsiveness and transactivate the heterologous FoxO1 promoter but not in the presence of dominant negative PPARdelta/beta. Taken together, our findings suggest that CD36-dependent FA activation of PPARdelta/beta results in the transcriptional regulation of FoxO1 as well as PDK4, recently shown to be a direct PPARdelta/beta target. FoxO1 in turn can regulate CD36, lipoprotein lipase, and PDK4, reinforcing the action of PPARdelta/beta to increase muscle reliance on FA. The findings could have implications in the chronic abnormalities of fatty acid metabolism associated with obesity and diabetes.


Asunto(s)
Adaptación Fisiológica , Antígenos CD36/metabolismo , Factores de Transcripción Forkhead/metabolismo , Músculos/metabolismo , PPAR delta/metabolismo , PPAR-beta/metabolismo , Proteínas Quinasas/metabolismo , Animales , Secuencia de Bases , Antígenos CD36/genética , Línea Celular , Ácidos Grasos/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Glucosa/metabolismo , Cinética , Ratones , Ratones Noqueados , Oxidación-Reducción , Transcripción Genética/genética , Regulación hacia Arriba
11.
PPAR Res ; 2007: 86394, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17389772

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are transcription factors that act as lipid sensors and adapt the metabolic rates of various tissues to the concentration of dietary lipids. PPARs are pharmacological targets for the treatment of metabolic disorders. PPARalpha and PPARgamma are activated by hypolipidemic and insulin-sensitizer compounds, such as fibrates and thiazolidinediones. The roles of PPARbeta/delta in metabolic regulations remained unclear until recently. Treatment of obese monkeys and rodents by specific PPARbeta/delta agonists promoted normalization of metabolic parameters and reduction of adiposity. Recent evidences strongly suggested that some of these beneficial actions are related to activation of fatty acid catabolism in skeletal muscle and also that PPARbeta/delta is involved in the adaptive responses of skeletal muscle to environmental changes, such as long-term fasting or physical exercise, by controlling the number of oxidative myofibers. These observations indicated that PPARbeta/delta agonists might have therapeutic usefulness in metabolic syndrome by increasing fatty acid consumption in skeletal muscle and reducing obesity.

12.
Biochim Biophys Acta ; 1771(8): 983-90, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17392020

RESUMEN

The prevalence of metabolic disturbances, collectively known as metabolic syndrome, has reached an epidemic proportion in industrialized countries. Lifestyle interventions and pharmacological treatments of such pathologies are only partially efficient and new therapeutic approaches are urgently needed. This review focuses on the recent findings describing the regulatory functions of peroxisome proliferator-activated receptor beta (PPARbeta) on lipid metabolism in several tissues and on the implications of such findings on the therapeutic usefulness of PPARbeta agonists in the treatment of particular features of the metabolic syndrome, such as insulin resistance, obesity, dyslipidemia and cardiac dysfunctions.


Asunto(s)
Síndrome Metabólico/tratamiento farmacológico , PPAR-beta/fisiología , Corazón/fisiopatología , Cardiopatías/patología , Cardiopatías/fisiopatología , Homeostasis , Humanos , Músculo Esquelético/fisiopatología , Miocardio/patología , PPAR-beta/agonistas
13.
Pharmacol Rev ; 58(4): 726-41, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17132851

RESUMEN

The three peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear hormone receptor superfamily. They share a high degree of structural homology with all members of the superfamily, particularly in the DNA-binding domain and ligand- and cofactor-binding domain. Many cellular and systemic roles have been attributed to these receptors, reaching far beyond the stimulation of peroxisome proliferation in rodents after which they were initially named. PPARs exhibit broad, isotype-specific tissue expression patterns. PPARalpha is expressed at high levels in organs with significant catabolism of fatty acids. PPARbeta/delta has the broadest expression pattern, and the levels of expression in certain tissues depend on the extent of cell proliferation and differentiation. PPARgamma is expressed as two isoforms, of which PPARgamma2 is found at high levels in the adipose tissues, whereas PPARgamma1 has a broader expression pattern. Transcriptional regulation by PPARs requires heterodimerization with the retinoid X receptor (RXR). When activated by a ligand, the dimer modulates transcription via binding to a specific DNA sequence element called a peroxisome proliferator response element (PPRE) in the promoter region of target genes. A wide variety of natural or synthetic compounds was identified as PPAR ligands. Among the synthetic ligands, the lipid-lowering drugs, fibrates, and the insulin sensitizers, thiazolidinediones, are PPARalpha and PPARgamma agonists, respectively, which underscores the important role of PPARs as therapeutic targets. Transcriptional control by PPAR/RXR heterodimers also requires interaction with coregulator complexes. Thus, selective action of PPARs in vivo results from the interplay at a given time point between expression levels of each of the three PPAR and RXR isotypes, affinity for a specific promoter PPRE, and ligand and cofactor availabilities.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Animales , Sitios de Unión , Humanos , Hipoglucemiantes/farmacología , Ligandos , Mutación , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/genética , Pioglitazona , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tiazolidinedionas/farmacología
14.
Endocr Rev ; 27(3): 318-29, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16556851

RESUMEN

Metabolic disturbances associated with alterations in lipid metabolism, such as obesity, type 2 diabetes, and syndrome X, are becoming more and more prominent in Western societies. Despite extensive research in such pathologies and their molecular basis, we are still far from completely understanding how these metabolic perturbations are produced and interrelate and, consequently, how to treat them efficiently. The discovery that adipose tissue is, in fact, an endocrine tissue able to secrete active molecules related to lipid homeostasis--the adipokines--has dramatically changed our understanding of the molecular events that take place in such diseases. This knowledge has been further improved by the discovery of peroxisome proliferator-activated receptors and their ligands, at present commonly used for the clinical treatment of lipid disturbances. However, a key point remains to be solved, and that is the role of muscle lipid metabolism, notably because of the main role played by this tissue in the development of such pathologies. In addition, a reciprocal regulation between adipose tissue and skeletal muscle has been proposed. New discoveries on the role of peroxisome proliferator-activated receptor-delta in skeletal muscle functions as well as the secretory capabilities of muscle, now considered as an endocrine tissue, have changed the general point of view on lipid homeostasis, opening new and promising doors for the treatment of lipid disorders.


Asunto(s)
Músculo Esquelético/metabolismo , Obesidad/metabolismo , Obesidad/terapia , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Tejido Adiposo/metabolismo , Animales , Ejercicio Físico , Ácidos Grasos/metabolismo , Humanos , Resistencia a la Insulina , Metabolismo de los Lípidos , Ratones , Receptores Activados del Proliferador del Peroxisoma/agonistas
15.
Cardiovasc Res ; 69(2): 440-9, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16337160

RESUMEN

OBJECTIVE: Activation of peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARgamma plays beneficial roles in cardiovascular disorders such as atherosclerosis and heart reperfusion. Although PPARalpha and gamma have been documented to reduce oxidative stress in the vasculature and the heart, the role of PPARdelta remains poorly studied. METHODS AND RESULTS: We focused on PPARdelta function in the regulation of oxidative stress-induced apoptosis in the rat cardiomyoblast cell line H9c2. Using semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), we showed that PPARdelta is the predominantly expressed isotype whereas PPARalpha was weakly detected. By performing cell viability assays, we also showed that the selective PPARdelta agonist GW501516 protected cells from H(2)O(2)-induced cell death. The protective effect of GW501516 was due to an inhibition of H(2)O(2)-triggered apoptosis as shown by annexin-V labeling, DNA fragmentation analysis, and caspase-3 activity measurement. We demonstrated by transient transfection of a dominant negative mutant of PPARdelta that the protection induced by GW501516 was totally dependent on PPARdelta. Semi-quantitative RT-PCR and Western blotting analysis demonstrated that GW501516 treatment upregulated catalase. Moreover, forced overexpression of catalase inhibited H(2)O(2)-triggered apoptosis, as evidenced by annexin-V labeling. CONCLUSION: Taken together, our results account for an important role of PPARdelta in inhibiting the onset of oxidative stress-induced apoptosis in H9c2 cells. PPARdelta appears to be a new therapeutic target for the regulation of heart reperfusion-associated oxidative stress and stimulation of enzymatic antioxidative defences.


Asunto(s)
Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patología , PPAR delta/metabolismo , Tiazoles/farmacología , Animales , Apoptosis , Western Blotting/métodos , Caspasa 3 , Caspasas/metabolismo , Catalasa/metabolismo , Línea Celular , Fragmentación del ADN , Peróxido de Hidrógeno/farmacología , Etiquetado Corte-Fin in Situ , Mioblastos Cardíacos/efectos de los fármacos , Estrés Oxidativo , PPAR delta/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
16.
Biochim Biophys Acta ; 1740(2): 313-7, 2005 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-15949697

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are lipid-activated transcription factors exerting several functions in development and metabolism. PPARalpha, activated by polyunsaturated fatty acids and fibrates, is implicated in regulation of lipid metabolism, lipoprotein synthesis and metabolism and inflammatory response in liver and other tissues. PPARgamma plays important roles in regulation of proliferation and differentiation of several cell types, including adipose cells. Its activation by thiazolidinediones results in insulin sensibilization and antidiabetic action. Until recently, the physiological functions of PPARdelta remain elusive. The utilization of specific agonists and of appropriate cellular and animal models revealed that PPARdelta has an important role in metabolic adaptation of several tissues to environmental changes. Treatment of obese animals by specific PPARdelta agonists results in normalization of metabolic parameters and reduction of adiposity. The nuclear receptor appeared to be implicated in the regulation of fatty acid burning capacities of skeletal muscle and adipose tissue by controlling the expression of genes involved in fatty acid uptake, beta-oxidation and energy uncoupling. PPARdelta is also implicated in the adaptive metabolic response of skeletal muscle to endurance exercise by controlling the number of oxidative myofibers. Given the results obtained with animal models, PPARdelta agonists may have therapeutic usefulness in metabolic syndrome by increasing fatty acid consumption in skeletal muscle and adipose tissue.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Metabolismo de los Lípidos , PPAR delta/metabolismo , Tejido Adiposo/metabolismo , Animales , Proteínas Portadoras/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Proteínas de Unión a Ácidos Grasos , Ácidos Grasos/metabolismo , Humanos , Síndrome Metabólico/tratamiento farmacológico , Proteína Trifuncional Mitocondrial , Complejos Multienzimáticos/metabolismo , Músculo Esquelético/metabolismo , PPAR delta/agonistas
17.
Mol Pharmacol ; 68(1): 193-203, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15829700

RESUMEN

It has been suggested that the teratogenic effects of the antiepileptic drug valproic acid (VPA) is reflected in vitro by the differentiation of F9 cells, activation of peroxisome proliferator-activated receptor delta (PPARdelta), and inhibition of histone deacetylases (HDACs). The aim of this study was to identify genes involved in the differentiation of F9 cells induced by VPA, teratogenic VPA derivatives, or the HDAC inhibitor trichostatin A (TSA) and to characterize the role of PPARdelta. Treatment of the cells with teratogenic VPA derivatives or TSA induced differentiation of F9 cells, mRNA, and protein expression of the neural cell adhesion molecule (NCAM) as well as activated the 5'-flanking region of the NCAM promoter, whereas nonteratogenic VPA derivatives had no effect at all. The polysialyltransferases [ST8SiaIV (PST1) and ST8SiaII] are responsible for the addition of polysialic acid (PSA) to NCAM. The mRNA expression of PST1 was highly induced by only teratogenic VPA derivatives and TSA. As shown by fluorescence-activated cell sorting analysis the level of PSA was higher after treatment of F9 cells with teratogenic VPA derivatives. It is interesting that overexpression of the PPARdelta but not PPARalpha or PPARgamma in F9 cells resulted in higher induction of NCAM mRNA and protein expression and of PST1 mRNA expression (and a higher PSA level) than in mock-transfected F9 cells. Furthermore, repression of PPARdelta activity in F9 cells inhibited these effects. We conclude that NCAM and PST1 are molecular markers in F9 cell differentiation caused by treatment with teratogenic VPA compounds or TSA and suggest that in addition to HDAC inhibition PPARdelta is involved in the signaling pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , PPAR delta/metabolismo , Sialiltransferasas/biosíntesis , Teratógenos/farmacología , Ácido Valproico/análogos & derivados , Animales , Diferenciación Celular/fisiología , Línea Celular , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Ratones , Moléculas de Adhesión de Célula Nerviosa/genética , PPAR delta/genética , PPAR delta/fisiología , Sialiltransferasas/genética , Ácido Valproico/farmacología
19.
Diabetes ; 53(9): 2209-16, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15331529

RESUMEN

High levels of intramyocellular triglycerides are linked to insulin resistance and reflect conditions in which fatty acid uptake exceeds the myocyte oxidative capacity. CD36 facilitates fatty acid uptake by myocytes, and its level is increased in diabetic muscle. We examined whether high CD36 levels would increase lipid content and susceptibility of myocytes to fatty acid-induced insulin resistance. C2C12 myoblasts with stable fivefold overexpression of CD36 (+CD36) were generated and differentiated into myotubes. CD36 expression increased palmitate uptake, oxidation, and lipid incorporation but had no effect on cell triglyceride content. Importantly, glycerol release increased fourfold, indicating enhanced triglyceride turnover and suggesting that CD36 promotes futile cycling of fatty acids into triglyceride. When +CD36 myotubes were incubated with excess palmitate, CD36 enhancement of glycerol release was blunted, triglyceride content increased above wild-type cells, and insulin resistance of glucose metabolism was observed. In contrast to palmitate, oleate-treated +CD36 cells exhibited enhanced glycerol release and no alteration in triglyceride content or insulin responsiveness. Furthermore, increased expression of hormone-sensitive lipase was measured with CD36 expression and with oleate treatment. In conclusion, high futile cycling of fatty acids is important for maintaining low triglyceride content and insulin responsiveness of myocytes. The findings provide a new perspective related to the etiology of lipid accumulation and insulin resistance in myocytes.


Asunto(s)
Antígenos CD36/metabolismo , Lipasa/metabolismo , Células Musculares/metabolismo , Ácido Oléico/farmacocinética , Palmitatos/farmacocinética , Triglicéridos/metabolismo , Animales , Antígenos CD36/genética , Línea Celular , Expresión Génica , Glucógeno/biosíntesis , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Insulina/metabolismo , Insulina/farmacología , Ratones , Células Musculares/citología , Oxidación-Reducción , Ratas
20.
Curr Opin Clin Nutr Metab Care ; 7(4): 377-81, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15192438

RESUMEN

PURPOSE OF THE REVIEW: Peroxisome proliferator-activated receptors mediate the transcriptional effects of fatty acids and fatty acid metabolites and regulate many physiological functions including development and metabolism. The roles of peroxisome proliferator-activated receptor alpha and gamma isotypes have been well established, while the functions of the third member of the family, peroxisome proliferator-activated receptor delta, remained unclear until very recently. This review focuses on the physiological functions of the nuclear receptor and especially on its roles in the control of fatty acid metabolism. RECENT FINDINGS: We review very recent data demonstrating that peroxisome proliferator-activated receptor delta plays a central role in the regulation of fatty acid oxidation in several tissues, such as skeletal muscle and adipose tissue, and also that the nuclear receptor is involved in cholesterol metabolism. SUMMARY: Use of potent and specific peroxisome proliferator-activated receptor delta agonists and appropriate transgenic animal models revealed the importance of this nuclear receptor in regulation of fatty acid catabolism in skeletal muscle and other tissues. It also indicated the potential of peroxisome proliferator-activated receptor delta as a therapeutic target in pathologies such as metabolic syndrome. However, the effects of peroxisome proliferator-activated receptor delta activation in atherosclerosis and the control of cell proliferation remain to be established.


Asunto(s)
Ácidos Grasos/metabolismo , Músculo Esquelético/fisiología , PPAR delta/fisiología , Animales , Animales Modificados Genéticamente , Arteriosclerosis/metabolismo , Proliferación Celular , Colesterol/metabolismo , Humanos , Músculo Esquelético/metabolismo , Oxidación-Reducción , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...