Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 473(1): 107-113, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-27012212

RESUMEN

Plasma cholesterol levels are associated with an increased risk of developing atherosclerosis. An elevated low-density lipoprotein cholesterol (LDL-C) level is a hallmark of hypercholesterolemia in metabolic syndrome. Our previous study suggested that when acetylated LDL (AC-LDL) was co-applied with a PPARγ agonist, rosiglitazone (ROSI), many oil red O-positive macrophages could be observed. However, addition of cyclic phosphatidic acid (cPA) to ROSI-stimulated macrophages completely abolished oil red O-stained cells, indicating that cPA inhibits PPARγ-regulated AC-LDL uptake. This study aimed to determine whether metabolically stabilized cPA, in the form of a carba-derivative of cPA (2ccPA), could reduce plasma cholesterol levels and affect the expression of genes related to atherosclerosis in apolipoprotein E-knockout (apoE(-/-)) mice. 2ccPA reduced LDL-C levels in these mice (n = 3) from 460 to 330 mg/ml, from 420 to 350 mg/ml, and 420 to 281 mg/ml under a western-type diet. 2ccPA also reduced expression of lipid metabolism-related genes, cytokines, and chemokines in ApoE-deficient mice on a high-fat diet. Taken together, these results suggest that 2ccPA governs anti-atherogenic activities in the carotid arteries of apoE-deficient mice.


Asunto(s)
LDL-Colesterol/sangre , Ácidos Fosfatidicos/administración & dosificación , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Arterias Carótidas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inflamación , Metabolismo de los Lípidos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , PPAR gamma/metabolismo , Ácidos Fosfatidicos/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Rosiglitazona , Tiazolidinedionas/agonistas , Triglicéridos/metabolismo
2.
Urology ; 81(5): 1109.e1-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23522297

RESUMEN

OBJECTIVE: To investigate the role of peroxisome proliferator-activated receptor γ (PPARγ) in bladder cancer (BCa) progression. MATERIALS AND METHODS: The gene copy number of PPARγ in human BCa tissue samples was analyzed by fluorescence in situ hybridization. The migration and invasive ability of human BCa cell lines with different PPARγ expression levels or treated with thiazolidinedione-rosiglitazone, a PPARγ agonist and an antidiabetic drug, were investigated. RESULTS: PPARγ amplification was increased dramatically in BCa tissue compared with normal urothelium (38.1% vs 4.3%, P = .0082) and in tumors with lymph node metastasis compared with those without metastasis (75.0% vs 15.4%, P = .0176). The human BCa cell line 5637 with strong PPARγ expression demonstrated a greater ability for cell migration and invasion than the UMUC-3 cell line with weak PPARγ expression. Knocking down PPARγ in BCa 5637 cells led to decreased cell migration, and activation of PPARγ with thiazolidinedione-rosiglitazone promoted their migration and invasive ability. CONCLUSION: PPARγ amplification in BCa could play an important role in BCa cell migration and invasion. Alteration of PPARγ expression by PPARγ-small interfering ribonucleic acid or activation by its agonist rosiglitazone, a diabetic thiazolidinedione drug, could lead to alternation of BCa cell migration and invasion.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , PPAR gamma/genética , ARN Neoplásico/genética , Tiazolidinedionas/agonistas , Neoplasias de la Vejiga Urinaria/genética , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Humanos , Hipoglucemiantes/farmacología , Hibridación Fluorescente in Situ , PPAR gamma/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Rosiglitazona , Tiazolidinedionas/farmacología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
3.
Gene ; 512(2): 438-43, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22967709

RESUMEN

Both vitamin D receptor (VDR) and peroxisome proliferator-activated receptor γ (PPAR-γ) are ligand-activated nuclear transcription factors that are instrumental for bone health. While 1α,25-dihydroxyvitamin D3 (1,25D3), the ligand for VDR, is essential for the development and maintenance of healthy bone, PPAR-γ agonists cause detrimental skeletal effects. Recent studies have revealed evidence for a cross-talk between 1,25D3- and PPAR-α/-δ ligand-mediated signaling but there is a current lack of knowledge regarding cross-talk between signaling of 1,25D3 and the PPAR-γ ligand mediated signaling. In this study, we investigated the cross-talk between 1,25D3- and PPAR-γ agonist rosiglitazone-mediated signaling in human osteoblasts. 1,25D3 slightly but significantly induced expression of the primary PPAR-γ target gene ANGPTL4 but did not influence FABP4. 1,25D3 did not change rosiglitazone regulation of ANGPTL4 and FABP4. The other way around, rosiglitazone reduced CYP24A1 gene expression but this did not change CYP24A1 induction by 1,25D3. The findings regarding CYP24A1 gene expression are in line with the observation that 1,25D3 levels in medium were not affected by rosiglitazone. Furthermore, rosiglitazone significantly inhibited 1,25D3-induction of BGLAP while rosiglitazone alone did not change BGLAP. Additionally, 1,25D3 and rosiglitazone increase osteoblast alkaline phosphatase activity and synergistically stimulated extracellular matrix mineralization. In conclusion, these data provide evidence for a cross-talk between rosiglitazone- and 1,25D3-mediated signaling leading to an acceleration of extracellular matrix mineralization. The data suggest that the reduction of the mineralization inhibitor BGLAP and the increased differentiation status underlie the increased mineralization.


Asunto(s)
Conservadores de la Densidad Ósea/agonistas , Calcificación Fisiológica/efectos de los fármacos , Calcitriol/agonistas , Hipoglucemiantes/agonistas , Osteoblastos/metabolismo , Tiazolidinedionas/agonistas , Fosfatasa Alcalina/biosíntesis , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/metabolismo , Conservadores de la Densidad Ósea/farmacología , Calcitriol/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Matriz Extracelular/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Osteoblastos/citología , PPAR alfa/metabolismo , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Receptores de Calcitriol/metabolismo , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Esteroide Hidroxilasas/biosíntesis , Tiazolidinedionas/farmacología , Vitamina D3 24-Hidroxilasa
4.
J Nutr Biochem ; 24(3): 531-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22749137

RESUMEN

To test the hypothesis that Lactobacillus casei Shirota (Lcs) protects against the onset of non-alcoholic fatty liver disease (NAFLD) in a mouse model of fructose-induced steatosis, C57BL/6J mice were either fed tap water or 30% fructose solution +/- Lcs for 8 weeks. Chronic consumption of 30% fructose solution led to a significant increase in hepatic steatosis as well as plasma alanine-aminotransferase (ALT) levels, which was attenuated by treatment with Lcs. Protein levels of the tight junction protein occludin were found to be markedly lower in both fructose treated groups in the duodenum, whereas microbiota composition in this part of the intestine was not affected. Lcs treatment markedly attenuated the activation of the Toll-like receptor (TLR) 4 signalling cascade found in the livers of mice only treated with fructose. Moreover, in livers of fructose fed mice treated with Lcs peroxisome proliferator-activated receptor (PPAR)-γ activity was markedly higher than in mice only fed fructose. Taken together, the results of the present study suggest that the dietary intake of Lcs protects against the onset of fructose-induced NAFLD through mechanisms involving an attenuation of the TLR-4-signalling cascade in the liver.


Asunto(s)
ADN Bacteriano/aislamiento & purificación , Hígado Graso/patología , Hígado Graso/prevención & control , Fructosa/efectos adversos , Lacticaseibacillus casei/metabolismo , Alanina Transaminasa/análisis , Alanina Transaminasa/metabolismo , Animales , Butiratos/sangre , Línea Celular , Proliferación Celular , Cromanos/agonistas , Cromanos/farmacología , Dermatoglifia del ADN , ADN Bacteriano/genética , Modelos Animales de Enfermedad , Hígado Graso/inducido químicamente , Hipoglucemiantes/agonistas , Hipoglucemiantes/farmacología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Lacticaseibacillus casei/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , PPAR gamma/genética , PPAR gamma/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal , Tiazolidinedionas/agonistas , Tiazolidinedionas/farmacología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Troglitazona , Regulación hacia Arriba
5.
J Biol Chem ; 286(22): 20087-99, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21467033

RESUMEN

Here, we demonstrate that troglitazone (Rezulin), a peroxisome proliferator-activated receptor agonist, acted in synergy with heregulin to induce massive cell death in breast cancer cells. Although the combination of heregulin and troglitazone (HRG/TGZ) induced both apoptosis and necrosis, the main mode of cell death was caspase-independent and occurred via necrosis. This combination increased generation of superoxide in mitochondria, which in turn destabilized mitochondria potential. Pretreatment with N-acetyl-l-cysteine and catalase expression ameliorated cell death induced by the combination treatment, indicating a role of oxidative stress in mediating HRG/TGZ-induced cell death. Notably, pretreatment with pyruvate significantly prevented the cell death, suggesting a potential mechanistic link between metabolic stress and HRG/TGZ-induced cell death. The activation of the HRG signaling axis has been considered as a poor prognostic factor in breast cancer and confers resistance to gefitinib (Iressa) and tamoxifen. However, our data presented here paradoxically suggest that HRG expression can actually be beneficial when it comes to treating breast cancer with peroxisome proliferator-activated receptor-γ ligands. Taken together, the combination of HRG and TGZ may provide a basis for the development of a novel strategy in the treatment of apoptosis-resistant and/or hormone-refractory breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Cromanos/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neurregulina-1/farmacología , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Antineoplásicos/agonistas , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cromanos/agonistas , Sinergismo Farmacológico , Femenino , Humanos , Necrosis , Neurregulina-1/agonistas , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/genética , PPAR gamma/metabolismo , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/agonistas , Troglitazona
6.
Neurotox Res ; 19(3): 462-71, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20524105

RESUMEN

The neuroprotective effect of rosiglitazone, a peroxisome proliferator-activated receptor γ (PPARγ) agonist, has been investigated using both in vivo and in vitro models of global ischemia in CD1 mice. Behavioral tests were carried out prior to and at various times (up to 14 days) subsequent to bilateral common carotid artery occlusion followed by reperfusion. Mice at each time point were euthanized under anesthesia and the brain was removed, serially sliced and stained with 1% triphenyltetrazolium (TTC) to quantify infarct size. Administration of rosiglitazone (5 or 10 mg/kg, i.p.) 10 min prior to occlusion significantly reduced the postsurgical mortality rate (10-11 vs. 36%, P < 0.05). The higher dose of rosiglitazone (10 mg/kg) also significantly reduced the mean area of brain infarct at 1, 3, 7 and 14 days post-ischemia, reduced post-occlusion deficits in limb grasping and forelimb placing at various time points, and reduced total nitrite concentration in serum and brain homogenate at day 7 post-occlusion. To model global ischemia in vitro, coronal brain slices were incubated in oxygenated artificial cerebrospinal fluid (ACSF) in the presence of either glutamate (1 mM) or hydrogen peroxide (H(2)O(2)) (5 µM) for 30 min. Both H(2)O(2) and glutamate caused significant tissue damage, and co-incubation with rosiglitazone (5 µM) significantly reduced H(2)O(2)-induced damage but did not significantly reduce glutamate-induced brain damage in this model. Our observations provide further evidence for a neuroprotective effect of rosiglitazone in rodent models of ischemia.


Asunto(s)
Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/prevención & control , Fármacos Neuroprotectores/uso terapéutico , PPAR gamma/metabolismo , Tiazolidinedionas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/metabolismo , Masculino , Ratones , Fármacos Neuroprotectores/agonistas , Estrés Oxidativo/fisiología , PPAR gamma/agonistas , Rosiglitazona , Tiazolidinedionas/agonistas
7.
Biochem Pharmacol ; 80(1): 80-5, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20298676

RESUMEN

As a ligand for peroxisome proliferators-activated receptor gamma (PPAR gamma), troglitazone inhibits cell growth by mechanisms besides activating PPAR gamma. In this study, we found that troglitazone interfered with the interactions between estrogen-related receptor alpha and gamma (ERR alpha and ERR gamma) and their coactivator PPAR gamma coactivator-1 alpha (PGC-1 alpha) functioning as an inverse agonist. Additionally, troglitazone suppressed the expressions of PGC-1 alpha and its related member PGC-1 beta which are key regulators of mitochondrial function. Consequently, troglitazone reduced mitochondrial mass and suppressed the expressions of superoxide dismutases to elevate reactive oxygen species (ROS) production. The increase in ROS in turn induced the expression of cell cycle inhibitor p21(WAF1). We therefore propose that ERR alpha and ERR gamma are alternative targets of troglitazone important for mediating its growth suppressive effect.


Asunto(s)
Cromanos/agonistas , Cromanos/metabolismo , PPAR gamma/agonistas , Receptores de Estrógenos/agonistas , Tiazolidinedionas/agonistas , Tiazolidinedionas/metabolismo , Humanos , Ligandos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Troglitazona , Receptor Relacionado con Estrógeno ERRalfa
8.
Br J Pharmacol ; 159(3): 678-88, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20105179

RESUMEN

BACKGROUND AND PURPOSE: Peroxisome proliferator-activated receptor gamma (PPARgamma) agonists, such as rosiglitazone and pioglitazone, sensitize cells to insulin, and are therefore used to treat type 2 diabetes. However, in some patients, these drugs induce oedema, and the present study tests the hypothesis that this side effect reflects serum and glucocorticoid-inducible kinase 1 (SGK1)-dependent enhancement of epithelia Na(+) absorption. EXPERIMENTAL APPROACH: Na(+) absorbing epithelial cells (H441 cells, mpkCCD cells) on permeable membranes were mounted in Ussing chambers, and the effects of rosiglitazone (2 microM) and pioglitazone (10 microM) on transepithelial Na(+) absorption were quantified electrometrically. Changes in SGK1 activity were assessed by monitoring phosphorylation of residues within an endogenous protein. KEY RESULTS: Both cell types absorbed Na(+) via an electrogenic process that was enhanced by insulin. In mpkCCD cells, this stimulation of Na(+) transport was associated with increased activity of SGK1, whereas insulin regulated Na(+) transport in H441 cells through a mechanism that did not involve activation of this kinase. Rosiglitazone and pioglitazone had no discernible effect on transepithelial Na(+) absorption in unstimulated or insulin-stimulated cells and failed to alter cellular SGK1 activity. CONCLUSIONS AND IMPLICATIONS: Our results do not support the view that PPARgamma agonists stimulate epithelial Na(+) absorption or alter the control of cellular SGK1 activity. It is therefore likely that other mechanisms are involved in PPARgamma-mediated fluid retention, and a better understanding of these mechanisms may help with the identification of patients likely to develop oedema or heart failure when treated with these drugs.


Asunto(s)
Células Epiteliales/metabolismo , Epitelio/metabolismo , PPAR gamma/agonistas , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Transporte Biológico/efectos de los fármacos , Recuento de Células , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Humanos , Insulina/metabolismo , Insulina/farmacología , Riñón/metabolismo , Pulmón/metabolismo , Fosforilación , Fosfotransferasas/metabolismo , Pioglitazona , Rosiglitazona , Tiazolidinedionas/agonistas , Tiazolidinedionas/metabolismo
9.
Bioorg Med Chem Lett ; 20(3): 1298-301, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20064714

RESUMEN

Systematic structure-activity relationship (SAR) studies of a screening lead led to the discovery of a series of thiazolidinediones (TZDs) as potent GPR40 agonists. Among them, compound C demonstrated an acute mechanism-based glucose-lowering in an intraperitoneal glucose tolerance test (IPGTT) in lean mice, while no effects were observed in GPR40 knock-out mice.


Asunto(s)
Descubrimiento de Drogas/métodos , Receptores Acoplados a Proteínas G/agonistas , Tiazolidinedionas/química , Animales , Ratones , Ratones Noqueados , Unión Proteica/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Relación Estructura-Actividad , Tiazolidinedionas/agonistas , Tiazolidinedionas/farmacología
10.
Rio de Janeiro; s.n; 2010. 138 p. ilus, tab.
Tesis en Portugués | LILACS | ID: lil-601469

RESUMEN

Este trabalho teve o objetivo de estudar o efeito de medicamentos com diferentes ações agonista PPAR (rosiglitazona, fenofibrato e bezafibrato) sobre o perfil lipídico, glicídico e alterações na massa corporal e morfologia do tecido adiposo e pancreático em modelo de diabetes e sobrepeso induzido por dieta. Camundongos C57BL/6 (2 meses de idade) foram alimentados com dieta padrão (SC, n=10) ou dieta hiperlipídica rica em sacarose (HFHS, n=40) por 6 semanas. Logo após, os animais HFHS foram subdividos em: HFHS não tratado e HFHS tratado com rosiglitazona (HFHS-Ro), fenofibrato (HFHS-Fe) ou bezafibrato (HFHS-BZ) (5 semanas). Os camundongos alimentados com dieta HFHS apresentaram maior glicemia e insulina de jejum (+33% e +138%, respectivamente), intolerância à glicose, resistência à insulina, aumento da massa corporal (MC) (+20%) e adiposidade, hipertrofia de adipócitos e redução da imunocoloração para adiponectina no tecido adiposo. No pâncreas houve aumento da massa (+28%), acúmulo de gordura (+700%), hipertrofia da ilhota (+38%) e redução da imunocoloração para GLUT-2 (-60%). A rosiglitazona diminuiu a glicemia e insulina de jejum, porém induziu o ganho de MC e hipertrofia cardíaca. O fenofibrato estabilizou a MC, enquanto o bezafibrato levou a perda de MC. Apenas o bezafibrato impediu a hipertrofia da ilhota. A imunocoloração para GLUT-2 foi aumentada por todos os medicamentos, e não houve alterações na imunocoloração para o PPARalfa. Sinais morfológicos de pancreatite foram vistos no grupo HFHS-Fe, apesar dos níveis normais de amilase e lipase séricos. A rosiglitazona exacerbou a infiltração intrapancreática de gordura (+75% vs. HFHS), e o bezafibrato aumentou a imunocoloração para o PPARbeta/delta nas ilhotas pancreáticas. Em conclusão, o bezafibrato apresentou um efeito mais amplo sobre as alterações metabólicas, morfológicas e biométricas decorrentes da dieta HFHS, sugerindo que a inibição das três isoformas do PPAR seria melhor do que a inibição...


This work aimed to evaluate the effect of peroxisome proliferator-activated receptor (PPAR) agonists (rosiglitazone, fenofibrate and bezafibrate) on lipid and glucose metabolism, body mass, and adipose and pancreatic tissue morphology in a model of diet-induced type 2 diabetes and overweight in mice. Two-month-old male C57BL/6 mice were fed a standard chow (SC, n=10) or a high-fat high-sucrose chow (HFHS, n=40) for 6 weeks, and then HFHS-fed mice were subdivided by treatment: untreated HFHS and HFHS treated with rosiglitazone (HFHS-Ro), fenofibrate (HFHS-Fe), or bezafibrate (HFHS-Bz) (5 weeks on medication). HFHS-fed mice have altered fasting glucose (+33%) and insulin (+138%), GI, IR, increased body mass (+20%) and fat pad weight, adipocyte hypertrophy, and decreased adiponectin immunostain. They also presented increased pancreatic (+28%) mass, intrapancreatic fat (+700%), islet hypertrophy (+38%), and decreased GLUT-2 immunostain (-60%). Rosiglitazone reduced fasting glucose and insulin but induced weight gain and heart hypertrophy. Fenofibrate impaired body mass gain, while bezafibrate induced weight loss. Only bezafibrate impaired islet hypertrophy. GLUT-2 immunostain was improved by all treatments, and there were no alterations in PPAR-alfa stain. There were morphological signs of pancreatitis in fenofibrate-treated mice, although there was no alteration in serum amylase and lipase. Rosiglitazone exacerbated pancreatic fat infiltration (+75% vs. HFHS group), and bezafibrate increased PPAR-beta expression in pancreatic islets. In conclusion, bezafibrate showed a wider range of action on metabolic, morphologic, and biometric alterations due to HFHS intake, suggesting that inhibiting the three PPAR isoforms is better than inhititing each isoform alone. Rosiglitazone exacerbated body mass gain, pancreatic fat infiltration and induced heart hyperthophy as well, thus, precaution has to be taken in prescribing rosiglitazone to obese patients.


Asunto(s)
Animales , Ratones , Adiponectina , Bezafibrato/agonistas , Grasas de la Dieta , Fenofibrato/agonistas , Metabolismo de los Lípidos , PPAR alfa/metabolismo , PPAR-beta/metabolismo , PPAR gamma/metabolismo , Sacarosa , Tiazolidinedionas/agonistas , /inducido químicamente , Enfermedades Cardiovasculares/metabolismo , Modelos Animales , Páncreas/metabolismo
11.
Bioorg Med Chem Lett ; 16(22): 5913-6, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17010604

RESUMEN

MCSS and LeapFrog, two de novo drug design programs, were used for the novel indole-based PPARgamma ligands' study. The designed compounds were synthesized and tested for the PPARgamma protein binding activities in vitro. Out of the compounds that were synthesized, two molecules (compounds 14d and 7d) possessed potent PPARgamma protein binding activity close to rosiglitazone in vitro.


Asunto(s)
Indoles/síntesis química , Indoles/farmacología , PPAR gamma/agonistas , Tiazolidinedionas/agonistas , Algoritmos , Simulación por Computador , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Enlace de Hidrógeno , Ligandos , PPAR gamma/química , Unión Proteica , Rosiglitazona , Relación Estructura-Actividad , Tiazolidinedionas/química
12.
Bone ; 38(1): 74-84, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16137931

RESUMEN

Thiazolidinediones are effective anti-diabetic drugs that improve insulin sensitivity through the activation of the nuclear receptor and adipocyte-specific transcription factor, peroxisome proliferator-activated receptor gamma (PPAR-gamma). Recent evidence suggests that PPAR-gamma also controls bone cell development and bone homeostasis. In mice, PPAR-gamma insufficiency results in increased bone mass, whereas administration of the specific PPAR-gamma agonist rosiglitazone leads to bone loss and increased bone marrow adiposity. Although the pro-adipocytic and anti-osteoblastic activities of PPAR-gamma can be separated in vitro using ligands with distinct chemical structures, little evidence exists supporting this functional separation in vivo. Netoglitazone (MCC-555, RWJ-241947) is a thiazolidinedione, which acts as either a full or partial PPAR-gamma agonist, or antagonist, in a cell type specific manner. In this study, the pro-adipocytic and anti-osteoblastic activities of netoglitazone were evaluated in vitro, using both U-33/gamma2 cells as a model of marrow mesenchymal cell differentiation under the control of PPAR-gamma2 and primary bone marrow cultures, and in vivo in C57BL/6 mice. In vitro, netoglitazone induced adipocyte and inhibited osteoblast formation in a PPAR-gamma2-dependent manner; however, it was 100-fold less effective than rosiglitazone. In vivo, the administration of netoglitazone at an effective hyperglycemic dose (10 microg/g body weight/day) did not result in trabecular bone loss. Bone quality parameters such as bone mineral density and bone microarchitecture were not affected in netoglitazone-treated animals. The observed lack of an in vivo effect of netoglitazone on bone was entirely consistent with its low anti-osteoblastic activity in vitro. In contrast to the observed in vitro effects, netoglitazone in vivo effectively induced marrow adipocyte formation and induced changes in the weights of extramedullary fat depots. Consistent with these cell type-specific effects, expression of the adipocyte-specific gene marker FABP4/aP2 was increased, whereas the expression of osteoblast-specific gene markers, Runx2, Dlx5, osteocalcin, and collagen were not affected by netoglitazone. In conclusion, netoglitazone is a member of a new class of PPAR-gamma ligands with distinct anti-diabetic, anti-osteoblastic, and pro-adipocytic activities in vivo.


Asunto(s)
Adipocitos/fisiología , Diferenciación Celular/fisiología , Osteoblastos/fisiología , Tiazoles/farmacología , Adipocitos/efectos de los fármacos , Animales , Densidad Ósea/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Línea Celular , Medios de Cultivo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos/efectos de los fármacos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , PPAR gamma , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Tiazoles/agonistas , Tiazolidinedionas/agonistas , Tiazolidinedionas/farmacología , Tomografía Computarizada por Rayos X
13.
Clin Cardiol ; 27(7 Suppl 4): IV11-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15470906

RESUMEN

Patients with type 2 diabetes are at high risk of cardiovascular disease. In addition to treating hyperglycemia, the thiazolidinedione (TZD) class of antidiabetic agents may also benefit the cardiovascular complications associated with the disease. The two available TZDs, pioglitazone and rosiglitazone, are peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists that influence gene expression of key proteins involved in regulating glucose and lipid metabolism. Tumor necrosis factor-alpha (TNF-alpha) and adiponectin are believed to be important in the development of insulin resistance and atherosclerosis. Understanding the role of these cytokines in the inflammatory processes that trigger plaque development might lead to identification of other potential mechanisms that could be exploited to enhance future treatments for patients with diabetes and atherosclerosis.


Asunto(s)
Resistencia a la Insulina/inmunología , Péptidos y Proteínas de Señalización Intercelular , Proteínas/efectos de los fármacos , Tiazolidinedionas/farmacología , Factores de Transcripción/farmacología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Adiponectina , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/fisiología , Arteriosclerosis/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Citocinas/efectos de los fármacos , Citocinas/inmunología , Diabetes Mellitus Tipo 2/fisiopatología , Glucosa/metabolismo , Humanos , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos , Proteínas/inmunología , Receptores Citoplasmáticos y Nucleares/agonistas , Tiazolidinedionas/agonistas , Factores de Transcripción/agonistas , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...