Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Bone Miner Res ; 32(4): 834-845, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27505721

RESUMEN

Results of prior studies suggest that fibroblast growth factor 21 (FGF21) may be involved in bone turnover and in the actions of peroxisome proliferator-activated receptor (PPAR) α and γ in mice. We have conducted independent studies to examine the effects of FGF21 on bone homeostasis and the role of FGF21 in PPARα and γ actions. High-fat-diet-induced obesity (DIO) mice were administered vehicle or recombinant human FGF21 (rhFGF21) intraperitoneally at 0 (vehicle), 0.1, 1, and 3 mg/kg daily for 2 weeks. Additional groups of DIO mice received water or 10 mg/kg rosiglitazone daily. Mice treated with rhFGF21 or rosiglitazone showed expected metabolic improvements in glucose, insulin, and lipid levels. However, bone loss was not detected in rhFGF21-treated mice by dual-energy X-ray absorptiometry (DXA), micro-CT, and histomorphometric analyses. Mineral apposition rate, a key bone formation parameter, was unchanged by rhFGF21, while significantly decreased by rosiglitazone in DIO mice. Bone resorption markers, OPG/RANKL mRNA expression, and histological bone resorption indices were unchanged by rhFGF21 or rosiglitazone. Bone marrow fat was unchanged by rhFGF21, while increased by rosiglitazone. Furthermore, FGF21 knockout mice did not show high bone mass phenotype. Treatment with PPARα or PPARγ agonists caused similar metabolic effects in FGF21 knockout and wild-type mice. These results contrast with previous findings and suggest that FGF21 is not critical for bone homeostasis or actions of PPARα and PPARγ. © 2016 American Society for Bone and Mineral Research.


Asunto(s)
Densidad Ósea , Factores de Crecimiento de Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , PPAR alfa , PPAR gamma , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/genética , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Insulina/genética , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/metabolismo , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , PPAR alfa/agonistas , PPAR alfa/biosíntesis , PPAR alfa/genética , PPAR gamma/agonistas , PPAR gamma/biosíntesis , PPAR gamma/genética , Ligando RANK/biosíntesis , Ligando RANK/genética , Rosiglitazona , Tiazolidinedionas/farmacología
2.
PLoS One ; 7(11): e49345, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209571

RESUMEN

Fibroblast growth factor 21 (FGF21) is a promising drug candidate for the treatment of type 2 diabetes. However, the use of wild type native FGF21 is challenging due to several limitations. Among these are its short half-life, its susceptibility to in vivo proteolytic degradation and its propensity to in vitro aggregation. We here describe a rationale-based protein engineering approach to generate a potent long-acting FGF21 analog with improved resistance to proteolysis and aggregation. A recombinant Fc-FGF21 fusion protein was constructed by fusing the Fc domain of human IgG1 to the N-terminus of human mature FGF21 via a linker peptide. The Fc positioned at the N-terminus was determined to be superior to the C-terminus as the N-terminal Fc fusion retained the ßKlotho binding affinity and the in vitro and in vivo potency similar to native FGF21. Two specific point mutations were introduced into FGF21. The leucine to arginine substitution at position 98 (L98R) suppressed FGF21 aggregation at high concentrations and elevated temperatures. The proline to glycine replacement at position 171 (P171G) eliminated a site-specific proteolytic cleavage of FGF21 identified in mice and cynomolgus monkeys. The derived Fc-FGF21(RG) molecule demonstrated a significantly improved circulating half-life while maintaining the in vitro activity similar to that of wild type protein. The half-life of Fc-FGF21(RG) was 11 h in mice and 30 h in monkeys as compared to 1-2 h for native FGF21 or Fc-FGF21 wild type. A single administration of Fc-FGF21(RG) in diabetic mice resulted in a sustained reduction in blood glucose levels and body weight gains up to 5-7 days, whereas the efficacy of FGF21 or Fc-FGF21 lasted only for 1 day. In summary, we engineered a potent and efficacious long-acting FGF21 analog with a favorable pharmaceutical property for potential clinical development.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Hipoglucemiantes/farmacología , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/farmacología , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Macaca fascicularis , Masculino , Ratones , Mutación , Proteolisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
3.
Endocrinology ; 153(1): 69-80, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22067317

RESUMEN

Circulating levels of fibroblast growth factor 21 (FGF21), a metabolic regulator of glucose, lipid, and energy homeostasis, are elevated in obese diabetic subjects, raising questions about potential FGF21 resistance. Here we report tissue expression changes in FGF21 and its receptor components, and we describe the target-organ and whole-body responses to FGF21 in ob/ob and diet-induced obese (DIO) mice. Plasma FGF21 concentrations were elevated 8- and 16-fold in DIO and ob/ob mice, respectively, paralleling a dramatic increase in hepatic FGF21 mRNA expression. Concurrently, expression levels of ßKlotho, FGF receptor (FGFR)-1c, and FGFR2c were markedly down-regulated in the white adipose tissues (WAT) of ob/ob and DIO mice. However, dose-response curves of recombinant human FGF21 (rhFGF21) stimulation of ERK phosphorylation in the liver and WAT were not right shifted in disease models, although the magnitude of induction in ERK phosphorylation was partially attenuated in DIO mice. Whole-body metabolic responses were preserved in ob/ob and DIO mice, with disease models being more sensitive and responsive than lean mice to the glucose-lowering and weight-loss effects of rhFGF21. Endogenous FGF21 levels, although elevated in diseased mice, were below the half-maximal effective concentrations of rhFGF21, suggesting a state of relative deficiency. Hepatic and WAT FGF21 mRNA expression levels declined after rhFGF21 treatment in the absence of the increased expression levels of ßKlotho and FGFR. We conclude that overt FGF21 resistance was not evident in the disease models, and increased hepatic FGF21 expression as a result of local metabolic changes is likely a major cause of elevated circulating FGF21 levels.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/genética , Expresión Génica , Humanos , Resistencia a la Insulina/genética , Proteínas Klotho , Hígado/efectos de los fármacos , Hígado/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/etiología , Obesidad/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes/administración & dosificación
4.
Am J Physiol Endocrinol Metab ; 297(5): E1105-14, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19706786

RESUMEN

Recombinant fibroblast growth factor (FGF)21 has antihyperglycemic, antihyperlipidemic, and antiobesity effects in diabetic rodent and monkey models. Previous studies were confined to measuring steady-state effects of FGF21 following subchronic or chronic administration. The present study focuses on the kinetics of biological actions of FGF21 following a single injection and on the associated physiological and cellular mechanisms underlying FGF21 actions. We show that FGF21 resulted in rapid decline of blood glucose levels and immediate improvement of glucose tolerance and insulin sensitivity in two animal models of insulin resistance (ob/ob and DIO mice). In ob/ob mice, FGF21 led to a 40-60% decrease in blood glucose, insulin, and amylin levels within 1 h after injection, and the maximal effects were sustained for more than 6 h despite the 1- to 2-h half-life of FGF21. In DIO mice, FGF21 reduced fasting blood glucose and insulin levels and improved glucose tolerance and insulin sensitivity within 3 h of treatment. The acute improvement of glucose metabolism was associated with a 30% reduction of hepatic glucose production and an increase in peripheral glucose turnover. FGF21 appeared to have no direct effect on ex vivo pancreatic islet insulin or glucagon secretion. However, it rapidly induced typical FGF signaling in liver and adipose tissues and in several hepatoma-derived cell lines and differentiated adipocytes. FGF21 was able to inhibit glucose release from H4IIE hepatoma cells and stimulate glucose uptake in 3T3-L1 adipocytes. We conclude that the acute glucose-lowering and insulin-sensitizing effects of FGF21 are potentially associated with its metabolic actions in liver and adipose tissues.


Asunto(s)
Tejido Adiposo/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Hipoglucemiantes , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Animales , Área Bajo la Curva , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Dieta , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Factores de Crecimiento de Fibroblastos/farmacocinética , Técnica de Clampeo de la Glucosa , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Hígado/efectos de los fármacos , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Proteínas Recombinantes/farmacología
5.
Mol Pharmacol ; 75(4): 901-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19129426

RESUMEN

Ghrelin is a 28-amino acid peptide secreted mainly by the stomach. Acyl-ghrelin, which binds to and activates the growth hormone secretagogue receptor type 1a (GHS-R1a), is considered to be the active form for its orexigenic effects. It has been demonstrated that peripheral administration of ghrelin stimulates food intake and adiposity in rodents and humans. Accordingly, different approaches to antagonize ghrelin/GHS-R1a signaling have been pursued for the treatment of obesity. In the present study, we generated and characterized high-affinity anti-acyl ghrelin-specific monoclonal antibodies (mAbs). In vitro, the lead mAb (33A) displayed specific binding to acyl-ghrelin, with an estimated K(d) value < 100 pM. In recombinant receptor cell-based assays, 33A dose-dependently inhibited the ghrelin-mediated calcium signal, with an IC(50) of approximately 3.5 nM. In vivo, ghrelin dose-dependently stimulated food intake in mice, and this effect was fully blocked by a single injection of 33A. In a 4-week chronic study, 33A was shown to effectively bind to endogenous acyl-ghrelin; however, long-term administration of 33A did not affect food intake or body weight gain in a mouse model of diet-induced obesity. Our results indicate that peripheral neutralization of ghrelin can suppress appetite stimulated by a transient surge in ghrelin levels. The lack of long-term effects on body weight control by 33A suggests that compensatory mechanisms may contribute to the regulation of energy balance.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Ingestión de Alimentos/fisiología , Ghrelina/antagonistas & inhibidores , Ghrelina/fisiología , Obesidad/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Depresores del Apetito/administración & dosificación , Depresores del Apetito/metabolismo , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Ghrelina/inmunología , Ghrelina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/prevención & control , Aumento de Peso/efectos de los fármacos , Aumento de Peso/fisiología
6.
Endocrinology ; 143(9): 3490-7, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12193562

RESUMEN

We have conducted three studies to examine the role of TNFalpha in islet destruction in female nonobese diabetic mouse (NOD) mice, a model of human autoimmune diabetes, using polyethylene glycolated (PEGylated) soluble TNF receptor type I (PEG sTNF-RI) as TNFalpha antagonist. PEG sTNF-RI (3 mg/kg, sc) was given every other day to NOD mice from age wk 8 for 12 wk (study 1), from age wk 12 for 8 wk (study 2), or from age wk 8 for 3 wk, with cyclophosphamide (6 mg/mouse) injected at wk 9 to accelerate the onset of diabetes (study 3). Diabetic incidence was reduced (control vs. PEG sTNF-RI) from 68.7% (11 of 16) to 18.3% (3 of 16) in study 1, from 84.6% (11 of 13) to 28.5% (4 of 14) in study 2, and from 66.6% (8 of 12) to 23.1% (3 of 13) in study 3, respectively. The incidence of insulitis was also reduced from 91.6% (11 of 12) to 12.5% (2 of 16) in study 1 and from 100% (7 of 7) to 16.6% (2 of 12) in study 2 by PEG sTNF-RI. PEG sTNF-RI also largely preserved islet insulin content, reduced mRNA of inducible nitric oxide synthase and IL-6 in pancreases, and lowered plasma corticosterone, glycerol, and free fatty acid levels. These results confirm a pathogenic role of TNFalpha in mediating insulitis in NOD mice and suggest the prophylactic and therapeutic potential of PEG sTNF-RI for human autoimmune diabetes.


Asunto(s)
Ciclofosfamida/administración & dosificación , Diabetes Mellitus Tipo 1/prevención & control , Animales , Enfermedades Autoinmunes/prevención & control , Corticosterona/sangre , Diabetes Mellitus Tipo 1/inmunología , Ácidos Grasos no Esterificados/sangre , Femenino , Expresión Génica/efectos de los fármacos , Glicerol/sangre , Insulina/análisis , Interleucina-6/genética , Islotes Pancreáticos/química , Islotes Pancreáticos/patología , Linfocitos/patología , Ratones , Ratones Endogámicos NOD , Necrosis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Pancreatitis/patología , Pancreatitis/prevención & control , ARN Mensajero/análisis , Receptores del Factor de Necrosis Tumoral/fisiología , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA