Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Birth Defects Res ; 116(1): e2277, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38158794

RESUMEN

PURPOSE: The purpose of this study is to explore the regulatory function of COL1A1 against the apoptosis of embryonic stem cells (ESCs) and the potential function in congenital talipes equinovarus (CTEV). METHODS: Muscle tissues were collected from 20 children with CTEV and 20 children without CTEV, followed by detecting the expression of COL1A1 using the RT-PCR method. COL1A1 was knocked down in H1 and H9 human ESCs using the RNA interference technology, followed by determining the level of COL1A1, PITX1, TBX4, HOXD10, Fas, FasL, and Bax using the Western blotting assay. RESULTS: COL1A1 was found markedly upregulated in muscle tissues of CTEV children. In H1 and H9 human ESCs, compared to the empty vector, COL1A1, PITX1, TBX4, HOXD10, Fas, FasL, and Bax were found notably downregulated after transfected with the siRNA targeting COL1A1. CONCLUSION: COL1A1 induced the apoptosis of ESCs by mediating the PITX1/TBX4 signaling and might be a potential target for treating CTEV.


Asunto(s)
Pie Equinovaro , Niño , Humanos , Apoptosis/genética , Proteína X Asociada a bcl-2/genética , Pie Equinovaro/genética , Células Madre Embrionarias , Proteínas de Dominio T Box/genética
2.
Comput Math Methods Med ; 2022: 6840716, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832129

RESUMEN

Background: A radius neck fracture in children is a common fracture that not only affects the growth and development of children but also has a certain impact on the function of children's elbow joints. Objective: To probe into the application value of ultrasonography- (US-) guided combination with elbow arthrography in the minimally invasive treatment of radial neck fractures in young children, summarize its clinical effect and provide a minimally invasive, safe, effective, and reliable method for treating radial neck fractures in young children. Methods: Seventy-three patients with type III or IV radial neck fractures were treated from June 2013 to December 2020 and were divided into the Métaizeau group (n = 31, treatment group) and Kirschner wire (k-wire) k-wire group (n = 42, control group). The Métaizeau group was given US-guided combination with elbow arthrography-assisted modified Métaizeau technique, the k-wire group received open reduction and internal fixation with k-wire and compared the surgical effect of the two groups. Results: In comparison with the k-wire group, time of operation, intraoperative bleeding volume, and hospital stay were signally junior to those in the Métaizeau group (P < 0.05). After surgery, in comparison with the k-wire group, the number of degrees to contralateral flexion or forearm rotation was visually lower in the Métaizeau group (P < 0.05), and postoperative complication incidence in the Métaizeau group was visually lower than that in k-wire group (P < 0.05). Conclusion: In the minimally invasive treatment of radial neck fractures, US-guided combination with elbow arthrography in young children has better efficacy and high safety. It can be widely promoted and applied clinically.


Asunto(s)
Fracturas del Radio , Radio (Anatomía) , Artrografía , Niño , Preescolar , Codo/diagnóstico por imagen , Fijación Interna de Fracturas/métodos , Humanos , Radio (Anatomía)/diagnóstico por imagen , Radio (Anatomía)/cirugía , Fracturas del Radio/diagnóstico por imagen , Fracturas del Radio/cirugía , Estudios Retrospectivos , Resultado del Tratamiento , Ultrasonografía
4.
Front Physiol ; 9: 1712, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30542297

RESUMEN

Growth/differentiation factor-15 (GDF-15) is a distant member of the transforming growth factor ß (TGF-ß) superfamily and is widely expressed in multiple mammalian tissues. Its expression is highly regulated and is often induced in response to conditions associated with cellular stress. GDF15 serum levels have a strong association with many diseases, including inflammation, cancer, cardiovascular diseases, and obesity, and potentially serve as reliable predictor of disease progression. A functional role for GDF15 has been suggested in cancer, cardiovascular disease, kidney disease and metabolic disease. However, the knowledge of its pathophysiological function at the molecular level is still limited and requires more investigation. Recent identification of the endogenous receptor for GDF15 may provide additional insight in to its' molecular mechanisms and relationship to disease states.

5.
Nat Med ; 23(10): 1215-1219, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28846098

RESUMEN

Growth/differentiation factor 15 (GDF15), also known as MIC-1, is a distant member of the transforming growth factor-ß (TGF-ß) superfamily and has been implicated in various biological functions, including cancer cachexia, renal and heart failure, atherosclerosis and metabolism. A connection between GDF15 and body-weight regulation was initially suggested on the basis of an observation that increasing GDF15 levels in serum correlated with weight loss in individuals with advanced prostate cancer. In animal models, overexpression of GDF15 leads to a lean phenotype, hypophagia and other improvements in metabolic parameters, suggesting that recombinant GDF15 protein could potentially be used in the treatment of obesity and type 2 diabetes. However, the signaling and mechanism of action of GDF15 are poorly understood owing to the absence of a clearly identified cognate receptor. Here we report that GDNF-family receptor α-like (GFRAL), an orphan member of the GFR-α family, is a high-affinity receptor for GDF15. GFRAL binds to GDF15 in vitro and is required for the metabolic actions of GDF15 with respect to body weight and food intake in vivo in mice. Gfral-/- mice were refractory to the effects of recombinant human GDF15 on body-weight, food-intake and glucose parameters. Blocking the interaction between GDF15 and GFRAL with a monoclonal antibody prevented the metabolic effects of GDF15 in rats. Gfral mRNA is highly expressed in the area postrema of mouse, rat and monkey, in accordance with previous reports implicating this region of the brain in the metabolic actions of GDF15 (refs. 4,5,6). Together, our data demonstrate that GFRAL is a receptor for GDF15 that mediates the metabolic effects of GDF15.


Asunto(s)
Área Postrema/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor 15 de Diferenciación de Crecimiento/farmacología , Obesidad/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Encéfalo/metabolismo , Ingestión de Alimentos/genética , Citometría de Flujo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células HEK293 , Humanos , Immunoblotting , Macaca fascicularis , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Resonancia por Plasmón de Superficie , Pérdida de Peso/genética
6.
EBioMedicine ; 2(3): 214-24, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26151067

RESUMEN

The polarization of tissue resident macrophages toward the alternatively activated, anti-inflammatory M2 phenotype is believed to positively impact obesity and insulin resistance. Here we show that the soluble form of the extracellular domain (ECD) of C-type lectin-like receptor 2, CLEC2, regulates Kupffer cell polarization in the liver and improves glucose and lipid parameters in diabetic animal models. Over-expression of Fc-CLEC2(ECD) in mice via in vivo gene delivery, or injection of recombinant Fc-CLEC2(ECD) protein, results in a reduction of blood glucose and liver triglyceride levels and improves glucose tolerance. Furthermore, Fc-CLEC2(ECD) treatment improves cytokine profiles and increases both the M2 macrophage population and the genes involved in the oxidation of lipid metabolism in the liver. These data reveal a previously unidentified role for CLEC2 as a regulator of macrophage polarity, and establish CLEC2 as a promising therapeutic target for treatment of diabetes and liver disease.


Asunto(s)
Glucosa/metabolismo , Macrófagos del Hígado/metabolismo , Lectinas Tipo C/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Polaridad Celular , Hígado Graso/genética , Hígado Graso/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Macrófagos del Hígado/citología , Macrófagos del Hígado/efectos de los fármacos , Lectinas Tipo C/genética , Metabolismo de los Lípidos/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos , Estructura Terciaria de Proteína , Receptores Fc/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Solubilidad
7.
PLoS One ; 10(6): e0126924, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26083576

RESUMEN

Trefoil factor 3 (TFF3), also called intestinal trefoil factor or Itf, is a 59 amino acid peptide found as a homodimer predominantly along the gastrointestinal tract and in serum. TFF3 expression is elevated during gastrointestinal adenoma progression and has been shown to promote mucosal wound healing. Here we show that in contrast to other trefoil factor family members, TFF1 and TFF2, TFF3 is highly expressed in mouse duodenum, jejunum and ileum and that its expression is regulated by food intake. Overexpression of TFF3 using a recombinant adeno-associated virus (AAV) vector, or daily administration of recombinant TFF3 protein in vivo improved glucose tolerance in a diet-induced obesity mouse model. Body weight, fasting insulin, triglyceride, cholesterol and leptin levels were not affected by TFF3 treatment. Induction of mucinous metaplasia was observed in mice with AAV-mediated TFF3 overexpression, however, no such adverse histological effect was seen after the administration of recombinant TFF3 protein. Altogether these results suggest that the therapeutic potential of targeting TFF3 to treat T2D may be limited.


Asunto(s)
Glucemia/metabolismo , Ingestión de Alimentos/genética , Vectores Genéticos/efectos adversos , Metaplasia/genética , Mucinas/genética , Obesidad/genética , Animales , Colesterol/sangre , Dependovirus/genética , Dieta Alta en Grasa , Duodeno/metabolismo , Duodeno/patología , Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Íleon/metabolismo , Íleon/patología , Insulina/sangre , Yeyuno/metabolismo , Yeyuno/patología , Leptina/sangre , Masculino , Metaplasia/etiología , Metaplasia/metabolismo , Metaplasia/patología , Ratones , Mucinas/administración & dosificación , Mucinas/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factor Trefoil-2 , Factor Trefoil-3 , Triglicéridos/sangre
8.
Expert Opin Ther Targets ; 18(11): 1253-64, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25287216

RESUMEN

OBJECTIVE: Although the human genome encodes ∼ 20,000 protein-coding genes, only a very small fraction of these have been explored as potential targets for therapeutic development. The challenge of identifying and validating new protein targets has contributed to the significant reduction in the productivity of the pharmaceutical industry in the recent decade, highlighting the continued need to find new therapeutic targets. RESEARCH DESIGN AND METHODS: The traditional methods to discover new targets are expensive, low throughput and time consuming, usually taking years to validate or invalidate a target. To address these limitations, as a proof of concept, we explored the hydrodynamic tail vein (HTV) injection as a gene delivery method for direct in vivo phenotypic screening of novel secreted factor targets for Type II diabetes therapeutics. RESULTS: High levels and sustained expression of target proteins were observed in diabetic mouse models tested, allowing us to identify multiple novel hormones that may regulate glucose metabolism. CONCLUSIONS: These results suggest that HTV is a low-cost, high-throughput method for direct in vivo phenotypic drug screening in metabolic disorders and could be applicable to many other disease areas as well. This method if combined with other approaches such as human genetic studies could provide a significant value to future drug discovery.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Proteoma , Animales , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Descubrimiento de Drogas/métodos , Técnicas de Transferencia de Gen , Glucosa/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Hidrodinámica , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Cola (estructura animal)/irrigación sanguínea
9.
PLoS One ; 8(4): e61432, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23630589

RESUMEN

The endocrine hormone FGF21 has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to the native cytokine, we generated bispecific Avimer polypeptides that bind with high affinity and specificity to one of the receptor and coreceptor pairs used by FGF21, FGFR1c and ß-Klotho. These Avimers exhibit FGF21-like activity in in vitro assays with potency greater than FGF21. In a study conducted in obese male cynomolgus monkeys, animals treated with an FGFR1c/ß-Klotho bispecific Avimer showed improved metabolic parameters and reduced body weight comparable to the effects seen with FGF21. These results not only demonstrate the essential roles of FGFR1c and ß-Klotho in mediating the metabolic effects of FGF21, they also describe a first bispecific activator of this unique receptor complex and provide validation for a novel therapeutic approach to target this potentially important pathway for treating diabetes and obesity.


Asunto(s)
Fármacos Antiobesidad/farmacología , Factores de Crecimiento de Fibroblastos/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Péptidos/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Fármacos Antiobesidad/farmacocinética , Sitios de Unión , Unión Competitiva , Peso Corporal/efectos de los fármacos , Línea Celular , Evaluación Preclínica de Medicamentos , Factores de Crecimiento de Fibroblastos/química , Insulina/sangre , Proteínas Klotho , Macaca fascicularis , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Imitación Molecular , Datos de Secuencia Molecular , Obesidad/sangre , Péptidos/farmacocinética , Unión Proteica , Ratas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/biosíntesis , Albúmina Sérica/farmacocinética , Albúmina Sérica/farmacología , Transducción de Señal , Triglicéridos/sangre
10.
BioDrugs ; 27(2): 159-66, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23456652

RESUMEN

BACKGROUND AND OBJECTIVE: Fibroblast growth factor 21 (FGF21) has potent effects on normalizing glucose, lipid, and energy homeostasis, and represents an attractive novel therapy for type 2 diabetes mellitus and obesity. Approaches to improve the pharmacokinetic properties of FGF21, such as conjugation with polyethylene glycol, have been explored for therapeutic development. However, not only is there room for further pharmacokinetic improvements, additional re-engineering approaches to improve the potency and stability of FGF21 have not been reported. Here, we describe a novel approach to modify and improve the function of FGF21 by altering its C-terminal ßKlotho interaction domain. METHODS: We first identified Avimer proteins that are capable of binding ßKlotho. Then we explored replacing the C-terminal ßKlotho interaction domain of FGF21 with a ßKlotho-binding Avimer protein. RESULTS: Such a ßKlotho-binding Avimer protein was able to fully complement the C-terminal domain function of FGF21. The resulting FGF21-Avimer fusion is functionally indistinguishable from wild type FGF21, and more tolerant of C-terminal modification. CONCLUSION: These results demonstrate a viable strategy to modulate the affinity, potency, and engineering of FGF21, paving the way for further improvements of FGF21 as a therapeutic.


Asunto(s)
Fármacos Antiobesidad/farmacología , Factores de Crecimiento de Fibroblastos/farmacología , Hipoglucemiantes/farmacología , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/farmacología , Secuencia de Aminoácidos , Animales , Fármacos Antiobesidad/química , Fármacos Antiobesidad/uso terapéutico , Glucemia/análisis , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/normas , Factores de Crecimiento de Fibroblastos/uso terapéutico , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/uso terapéutico , Masculino , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Obesidad/sangre , Obesidad/tratamiento farmacológico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/normas , Proteínas Recombinantes de Fusión/uso terapéutico
11.
J Lipid Res ; 54(2): 325-32, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23204296

RESUMEN

Elevated triglyceride (TG) and cholesterol levels are risk factors for cardiovascular disease and are often associated with diabetes and metabolic syndrome. Recent reports suggest that fibroblast growth factor (FGF)19 and FGF21 can dramatically improve metabolic dysfunction, including hyperglycemia, hypertriglyceridemia, and hypercholesterolemia. Due to their similar receptor specificities and co-receptor requirements, FGF19 and FGF21 share many common properties and have been thought to be interchangeable in metabolic regulation. Here we directly compared how pharmacological administration of recombinant FGF19 or FGF21 proteins affect metabolism in B6.V-Lep(ob)/J leptin-deficient mice. FGF19 and FGF21 equally improved glucose parameters; however, we observed increased serum TG and cholesterol levels after treatment with FGF19 but not with FGF21. Increases in serum TGs were also observed after a 4-day treatment with FGF19 in C57BL6/J mice on a high-fat diet. This is in contrast to many literature reports that showed significant improvements in hyperlipidemia after chronic treatment with FGF19 or FGF21 in high-fat diet models. We propose that FGF19 has lipid-raising and lipid-lowering actions mediated through different FGF receptors and target tissues, and the results described here provide a potential mechanism that may explain the inconsistency in the reported effects of FGF19 on lipid metabolism.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Línea Celular , Colesterol/sangre , Dieta/efectos adversos , Factores de Crecimiento de Fibroblastos/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Obesidad/sangre , Obesidad/etiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Triglicéridos/sangre
12.
Sci Transl Med ; 4(162): 162ra153, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197570

RESUMEN

Fibroblast growth factor 21 (FGF21) is a distinctive member of the FGF family with potent beneficial effects on lipid, body weight, and glucose metabolism and has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to native FGF21, we have developed a monoclonal antibody, mimAb1, that binds to ßKlotho with high affinity and specifically activates signaling from the ßKlotho/FGFR1c (FGF receptor 1c) receptor complex. In obese cynomolgus monkeys, injection of mimAb1 led to FGF21-like metabolic effects, including decreases in body weight, plasma insulin, triglycerides, and glucose during tolerance testing. Mice with adipose-selective FGFR1 knockout were refractory to FGF21-induced improvements in glucose metabolism and body weight. These results in obese monkeys (with mimAb1) and in FGFR1 knockout mice (with FGF21) demonstrated the essential role of FGFR1c in FGF21 function and suggest fat as a critical target tissue for the cytokine and antibody. Because mimAb1 depends on ßKlotho to activate FGFR1c, it is not expected to induce side effects caused by activating FGFR1c alone. The unexpected finding of an antibody that can activate FGF21-like signaling through cell surface receptors provided preclinical validation for an innovative therapeutic approach to diabetes and obesity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/inmunología , Glucuronidasa/metabolismo , Obesidad/tratamiento farmacológico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Anticuerpos Monoclonales/farmacología , Peso Corporal/genética , Diabetes Mellitus/sangre , Epítopos/química , Glucosa/metabolismo , Glucuronidasa/inmunología , Humanos , Proteínas Klotho , Macaca fascicularis , Ratones , Obesidad/sangre , Obesidad/complicaciones , Fosfatos/sangre , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/agonistas , Transducción de Señal/efectos de los fármacos , Triglicéridos/sangre
13.
FEBS Lett ; 586(8): 1214-9, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22575658

RESUMEN

Adhesion G-protein-coupled receptors (GPCR) are special members of GPCRs with long N-termini containing multiple domains. We overexpressed our collection of receptors together with G-proteins in mammalian cell lines and measured the concentrations of intracellular signaling molecules, such as inositol phosphate and cAMP. Our results show that a subset of tested adhesion GPCRs has constitutive activities and is capable of coupling to a variety of G-proteins. In addition, we have identified a small molecule compound that specifically activates one of the subfamily members, GPR97, and the activation was confirmed by an independent GTPγS assay. These findings suggest classical GPCR screening assays could be applied to de-orphanize these receptors, and provide pharmacological tools to improve understanding of the physiological functions of these receptors.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Células CHO , Línea Celular , Cricetinae , AMP Cíclico/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Transfección
14.
PLoS One ; 7(3): e33603, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457778

RESUMEN

Diabetes and associated metabolic conditions have reached pandemic proportions worldwide, and there is a clear unmet medical need for new therapies that are both effective and safe. FGF19 and FGF21 are distinctive members of the FGF family that function as endocrine hormones. Both have potent effects on normalizing glucose, lipid, and energy homeostasis, and therefore, represent attractive potential next generation therapies for combating the growing epidemics of type 2 diabetes and obesity. The mechanism responsible for these impressive metabolic effects remains unknown. While both FGF19 and FGF21 can activate FGFRs 1c, 2c, and 3c in the presence of co-receptor ßKlotho in vitro, which receptor is responsible for the metabolic activities observed in vivo remains unknown. Here we have generated a variant of FGF19, FGF19-7, that has altered receptor specificity with a strong bias toward FGFR1c. We show that FGF19-7 is equally efficacious as wild type FGF19 in regulating glucose, lipid, and energy metabolism in both diet-induced obesity and leptin-deficient mouse models. These results are the first direct demonstration of the central role of the ßKlotho/FGFR1c receptor complex in glucose and lipid regulation, and also strongly suggest that activation of this receptor complex alone might be sufficient to achieve all the metabolic functions of endocrine FGF molecules.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucosa/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Animales , Western Blotting , Línea Celular , Dependovirus/genética , Factores de Crecimiento de Fibroblastos/genética , Vectores Genéticos , Prueba de Tolerancia a la Glucosa , Hígado/metabolismo , Masculino , Ratones , Unión Proteica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
15.
Adv Exp Med Biol ; 728: 195-213, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22396171

RESUMEN

FGF19 differs from the classical FGFs in that it has a much-reduced heparan sulfate proteoglycan binding affinity that allows it to act as endocrine hormone. Although FGF19 regulates several different metabolic activities, it still activates downstream signaling pathways through FGF receptors, in a similar manner to that seen in classical FGFs. Aberrant FGF signaling has been implicated in tumor development, and mouse models have confirmed that FGF19 has the potential to induce hepatocellular carcinoma. Treatment with anti-FGF19 antibody suppressed tumor progression in both FGF19 transgenic mice and colon cancer cell xenograft models. FGFR4, the predominant FGF receptor expressed in the liver, may play an important role in FGF19-mediated tumorigenesis. This review reports the current advances in understanding the structure-function relationship between FGF19 and its interactions with FGFRs, its physiological activities, and its differences from FGF21. The review also discusses strategies to separate the mitogenic and metabolic activities for the development of potential therapeutic molecules based on FGF19.


Asunto(s)
Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/metabolismo , Sustancias de Crecimiento/química , Sustancias de Crecimiento/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Relación Estructura-Actividad
16.
J Mol Biol ; 418(1-2): 82-9, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22370560

RESUMEN

Three fibroblast growth factor (FGF) molecules, FGF19, FGF21, and FGF23, form a unique subfamily that functions as endocrine hormones. FGF19 and FGF21 can regulate glucose, lipid, and energy metabolism, while FGF23 regulates phosphate homeostasis. The FGF receptors and co-receptors for these three FGF molecules have been identified, and domains important for receptor interaction and specificity determination are beginning to be elucidated. However, a number of questions remain unanswered, such as the identification of fibroblast growth factor receptor responsible for glucose regulation. Here, we have generated a variant of FGF23: FGF23-21c, where the C-terminal domain of FGF23 was replaced with the corresponding regions from FGF21. FGF23-21c showed a number of interesting and unexpected properties in vitro. In contrast to wild-type FGF23, FGF23-21c gained the ability to activate FGFR1c and FGFR2c in the presence of ßKlotho and was able to stimulate glucose uptake into adipocytes in vitro and lower glucose levels in ob/ob diabetic mice model to similar extent as FGF21 in vivo. These results suggest that ßKlotho/FGFR1c or FGFR2c receptor complexes are sufficient for glucose regulation. Interestingly, without the FGF23 C-terminal domain, FGF23-21c was still able to activate fibroblast growth factor receptors in the presence of αKlotho. This suggests not only that sequences outside of the C-terminal region may also contribute to the interaction with co-receptors but also that FGF23-21c may be able to regulate both glucose and phosphate metabolisms. This raises an interesting concept of designing an FGF molecule that may be able to address multiple diseases simultaneously. Further understanding of FGF/receptor interactions may allow the development of exciting opportunities for novel therapeutic discovery.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Proteínas de la Membrana/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Adipocitos/metabolismo , Animales , Glucemia/efectos de los fármacos , Línea Celular , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/farmacología , Glucosa/metabolismo , Proteínas Klotho , Masculino , Ratones , Ratones Obesos
17.
Expert Opin Ther Targets ; 15(11): 1307-16, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21961833

RESUMEN

INTRODUCTION: Diabetes and associated metabolic conditions have reached pandemic proportions worldwide and there is a clear unmet medical need for new therapies that are both effective and safe. FGF19 is a distinctive member of the FGF family that functions as an endocrine hormone. AREAS COVERED: An up-to-date report on the exciting findings related to the involvement of FGF19 in the regulation of glucose, bile acid metabolism and energy expenditure. The role of FGF receptors in these different activities. The therapeutic potential of FGF19 and the engineering opportunities for removing undesirable mitogenic activity. EXPERT OPINION: The ability of FGF19 to regulate bile acid homeostasis, gallbladder filling and tumor development and its potent ability to normalize glucose, lipid and energy homeostasis have made it a potential therapeutic target for the treatment of patients with gallstones, cancer and metabolic diseases, among others. Its potential utility as a novel therapeutic for both type 1 and type 2 diabetes is of particular interest. The ability to separate the undesired mitogenic activity from its potent metabolic activities has opened new opportunities for the development of potential therapeutic molecules based on FGF19 in treating various conditions associated with metabolic syndrome.


Asunto(s)
Sistemas de Liberación de Medicamentos , Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/fisiopatología , Diseño de Fármacos , Factores de Crecimiento de Fibroblastos/uso terapéutico , Glucosa/metabolismo , Humanos , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/fisiopatología , Ingeniería de Proteínas
18.
J Mol Biol ; 408(3): 491-502, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21392510

RESUMEN

FGF21 is a member of a unique subfamily of fibroblast growth factors that function as endocrine hormones and regulate a variety of metabolic activities. Unlike paracrine FGFs, FGF21 does not bind heparin and requires ßKlotho as a co-receptor to activate FGFR signaling. In the presence of ßKlotho, FGF21 is able to activate FGFRs 1c, 2c and 3c but not FGFR4. Chimeric FGFR1c/FGFR4 receptors were constructed to identify domains that confer this specificity and to understand regions important for FGF21-induced receptor activation. With these chimeras, we showed that domain 3 of the FGFR1c extracellular domain plays a critical role in specificity determination and receptor activation by FGF21. Furthermore, we were able to narrow down the sequences important for this function to a six amino acid region within domain 3 of FGFR1c. It is interesting to note that this region falls into the ßC'-ßE loop, which has been shown to be important for receptor specificity determination in paracrine FGFs, suggesting a common principle in both endocrine and paracrine FGF receptor interaction and activation.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Línea Celular , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Ratas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Proc Natl Acad Sci U S A ; 107(32): 14158-63, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660733

RESUMEN

FGF19 and FGF21 are distinctive members of the FGF family that function as endocrine hormones. Their potent effects on normalizing glucose, lipid, and energy homeostasis in disease models have made them an interesting focus of research for combating the growing epidemics of diabetes and obesity. Despite overlapping functions, FGF19 and FGF21 have many discrete effects, the most important being that FGF19 has both metabolic and proliferative effects, whereas FGF21 has only metabolic effects. Here we identify the structural determinants dictating differential receptor interactions that explain and distinguish these two physiological functions. We also have generated FGF19 variants that have lost the ability to induce hepatocyte proliferation but that still are effective in lowering plasma glucose levels and improving insulin sensitivity in mice. Our results add valuable insight into the structure-function relationship of FGF19/FGF21 and identify the structural basis underpinning the distinct proliferative feature of FGF19 compared with FGF21. In addition, these studies provide a road map for engineering FGF19 as a potential therapeutic candidate for treating diabetes and obesity.


Asunto(s)
Glucemia , Proliferación Celular , Factores de Crecimiento de Fibroblastos/fisiología , Hepatocitos/citología , Secuencia de Aminoácidos , Animales , Diabetes Mellitus/tratamiento farmacológico , Variación Genética , Resistencia a la Insulina , Ratones , Obesidad/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes/farmacología
20.
J Biol Chem ; 285(8): 5165-70, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20018895

RESUMEN

FGF19 and FGF21, unique members of the fibroblast growth factor (FGF) family, are hormones that regulate glucose, lipid, and energy homeostasis. Increased hepatocyte proliferation and liver tumor formation have also been observed in FGF19 transgenic mice. Here, we report that, in contrast to FGF19, FGF21 does not induce hepatocyte proliferation in vivo. To identify the mechanism for FGF19-induced hepatocyte proliferation, we explored similarities and differences in receptor specificity between FGF19 and FGF21. We find that although both are able to activate FGF receptors (FGFRs) 1c, 2c, and 3c, only FGF19 activates FGFR4, the predominant receptor in the liver. Using a C-terminal truncation mutant of FGF19 and a series of FGF19/FGF21 chimeric molecules, we determined that amino acids residues 38-42 of FGF19 are sufficient to confer both FGFR4 activation and increased hepatocyte proliferation in vivo to FGF21. These data suggest that activation of FGFR4 is the mechanism whereby FGF19 can increase hepatocyte proliferation and induce hepatocellular carcinoma formation.


Asunto(s)
Carcinoma Hepatocelular/mortalidad , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Células 3T3-L1 , Secuencia de Aminoácidos , Animales , Carcinoma Hepatocelular/genética , Transformación Celular Neoplásica/genética , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ratones Transgénicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...