Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(16): e2200476119, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35412887

RESUMEN

Augmentor α and ß (Augα and Augß) are newly discovered ligands of the receptor tyrosine kinases Alk and Ltk. Augα functions as a dimeric ligand that binds with high affinity and specificity to Alk and Ltk. However, a monomeric Augα fragment and monomeric Augß also bind to Alk and potently stimulate cellular responses. While previous studies demonstrated that oncogenic Alk mutants function as important drivers of a variety of human cancers, the physiological roles of Augα and Augß are poorly understood. Here, we investigate the physiological roles of Augα and Augß by exploring mice deficient in each or both Aug ligands. Analysis of mutant mice showed that both Augα single knockout and double knockout of Augα and Augß exhibit a similar thinness phenotype and resistance to diet-induced obesity. In the Augα-knockout mice, the leanness phenotype is coupled to increased physical activity. By contrast, Augß-knockout mice showed similar weight curves as the littermate controls. Experiments are presented demonstrating that Augα is robustly expressed and metabolically regulated in agouti-related peptide (AgRP) neurons, cells that control whole-body energy homeostasis in part via their projections to the paraventricular nucleus (PVN). Moreover, both Alk and melanocortin receptor-4 are expressed in discrete neuronal populations in the PVN and are regulated by projections containing Augα and AgRP, respectively, demonstrating that two distinct mechanisms that regulate pigmentation operate in the hypothalamus to control body weight. These experiments show that Alk-driven cancers were co-opted from a neuronal pathway in control of body weight, offering therapeutic opportunities for metabolic diseases and cancer.


Asunto(s)
Quinasa de Linfoma Anaplásico , Peso Corporal , Citocinas , Hipotálamo , Animales , Ratones , Quinasa de Linfoma Anaplásico/genética , Quinasa de Linfoma Anaplásico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Hipotálamo/metabolismo , Ligandos , Redes y Vías Metabólicas , Ratones Noqueados , Neoplasias/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Delgadez/genética
2.
Environ Sci Technol ; 57(39): 14737-14746, 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37738479

RESUMEN

Mn-based catalysts preferred in low-temperature selective catalytic reduction (SCR) are susceptible to SO2 poisoning. The stubborn sulfates make insufficient O2 activation and result in deficient reactive oxygen species (ROS) for activating reaction molecules. H2O has long been regarded as an accomplice to SO2, hastening catalyst deactivation. However, such a negative impression of the SCR reaction was reversed by our recent research. Here, we reported a H2O contribution over Mn-based SCR catalysts to counteract SO2 poisoning through accessible O2 activation, in which O2 was synergistically activated with H2O to generate ROS for less deactivation and more expected regeneration. The resulting ROS benefited from the energetically favorable route supported by water-induced Ea reduction and was actively involved in the NH3 activation and NO oxidation process. Besides, ROS maintained high stability over the SO2 + H2O-deactivated γ-MnO2 catalyst throughout the mild thermal treatment, achieving complete regeneration of its own NO disposal ability. This strategy was proven to be universally applicable to other Mn-based catalysts.

3.
Proc Natl Acad Sci U S A ; 114(45): 12027-12032, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29078341

RESUMEN

Anaplastic lymphoma kinase (Alk) and leucocyte tyrosine kinase (Ltk) were identified as "orphan" receptor tyrosine kinases (RTKs) with oncogenic potential. Recently ALKAL1 and ALKAL2 (also named "augmentor-ß" and "augmentor-α" or "FAM150A" and "FAM150B," respectively) were discovered as physiological ligands of Alk and Ltk. Here, we employ zebrafish as a model system to explore the physiological function and to characterize in vivo links between Alk and Ltk with their ligands. Unlike the two ligands encoded by mammalian genomes, the zebrafish genome contains three genes: aug-α1, aug-α2, and aug-ß Our experiments demonstrate that these ligands play an important role in zebrafish pigment development. Deficiency in aug-α1, aug-α2, and aug-ß results in strong impairment in iridophore patterning of embryonic and adult zebrafish that is phenocopied in zebrafish deficient in Ltk. We show that aug-α1 and aug-α2 are essential for embryonic iridophore development and adult body coloration. In contrast, aug-α2 and aug-ß are essential for iridophore formation in the adult eye. Importantly, these processes are entirely mediated by Ltk and not by Alk. These experiments establish a physiological link between augmentor ligands and Ltk and demonstrate that particular augmentors activate Ltk in a tissue-specific context to induce iridophore differentiation from neural crest-derived cells and pigment progenitor cells.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Quinasa de Linfoma Anaplásico , Animales , Diferenciación Celular/genética , Ojo/metabolismo , Genoma/genética , Ligandos , Cresta Neural/fisiología , Pigmentos Retinianos/genética , Células Madre/fisiología , Pez Cebra/fisiología
4.
Clin Sci (Lond) ; 131(3): 247-260, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27980130

RESUMEN

G-protein-coupled receptor 120 (GPR120) is a putative target for obesity and diabetes therapies. However, it remains controversial whether resident GPR120 plays a direct regulatory role in islet ß-cell insulin secretion. The present study examined this issue in isolated rodent islets and rat ß-cell line INS-1E, and assessed the role of GPR120 in islet insulin secretion in obese non-diabetic (OND) and diabetic states. GPR120 expression was detected in rodent islet ß-cells. Docosahexaenoic acid (DHA) and synthetic GPR120 agonist GSK137647 (GSK) augmented insulin release from rat/mouse islets and INS-1E; DHA effects were partially mediated by GPR40. GPR120 knockdown and overexpression attenuated and enhanced DHA effects in INS-1E respectively. DHA and GSK improved postprandial hyperglycaemia of diabetic mice. Inhibition of calcium signalling in INS-1E reduced GPR120 activation-induced insulinotropic effects. The insulinotropic effects of DHA/GSK were amplified in OND rat islets, but diminished in diabetic rat islets. GPR120 and peroxisome proliferator-activated receptor γ (PPARγ) expression were elevated in OND islets and palmitic acid (PA)-treated INS-1E, but reduced in diabetic islets and high glucose-treated INS-1E. PPARγ activation increased GPR120 expression in rat islets and INS-1E. DHA and GSK induced protein kinase B (Akt)/extracellular signal-regulated kinase (ERK) phosphorylation in rodent islets and INS-1E, and these effects were altered in OND and diabetic states. Taken together, the present study indicates that (i) GPR120 activation has an insulinotropic influence on ß-cells with the involvement of calcium signalling; (ii) GPR120 expression in ß-cells and GPR120-mediated insulinotropic effects are altered in OND and diabetic states in distinct ways, and these alterations may be mediated by PPARγ.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Compuestos de Anilina/farmacología , Animales , Apoptosis , Señalización del Calcio , Línea Celular , Ácidos Docosahexaenoicos , Técnicas de Silenciamiento del Gen , Prueba de Tolerancia a la Glucosa , Secreción de Insulina , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Wistar , Ratas Zucker , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Sulfonamidas/farmacología
5.
Anal Chim Acta ; 1188: 339179, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34794567

RESUMEN

The electrochemical collision-blocking technique, equipped with the nanoelectrode of Pt was proposed for determination of the critical micelle concentration (CMC) of non-ionic surfactant TX-100. The approach was found on detection of individual collided nanomicelles in amperometric measurements of the oxidation of K4Fe(CN)6 varying the titrated concentration of TX-100 whereas the formed micelles above the CMC stick on the electrode surface during collision to locally block the flux of electroactive species and further to change the faradaic current. The step-like current transients observed in i-t curves have been demonstrated corresponding to electrochemical collision events of individual TX-100 micelles and micelle aggregates by 3D COMSOL simulations. The logarithm relations between the collision frequency of micelle(s) and the concentration of TX-100 were derived by regression analysis to give the corresponding values of CMC in salt solutions. Further, an 'ideal' CMC of TX-100 without influence of additional salts was estimated to be 0.194 mM using the McDevit-Long theory. The more accurate CMC determined in this work has shown less than the previously reported, mainly due to the detection limit for micelle as low as 0.41 fM. Also, we determined the second CMC of 1.21 mM as the first observation of the collision response of micelle aggregates during TX-100 titration. Owing to its analytical characteristics in single-particle tracking and material insensitivity, the approach we proposed is potentially to be a universal tool for accurate determination of CMC of surfactants, and also for studying the formation of polymer particles at a single-particle level, which is not easily accessible using conventional ensemble measurements.


Asunto(s)
Micelas , Tensoactivos , Polietilenglicoles
6.
Am J Physiol Cell Physiol ; 298(2): C313-23, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19889960

RESUMEN

Hyperglycemia-associated glucotoxicity induces beta-cell apoptosis but the underlying mechanisms are unknown. Interestingly, prolonged exposure to high glucose upregulates the expression and function of the renin-angiotensin system (RAS). We hypothesize that the voltage-gated outward potassium (K(v)) current, which governs beta-cell membrane potential and insulin secretion, has a role in glucotoxicity. In this study, we investigated the effects of prolonged exposure to high glucose on mouse pancreatic beta-cells and concurrent effects on the RAS by examining changes in expression of angiotensin II (ANG II) receptors and changes in the expression and activity of K(v) channels. beta-Cells were incubated in high glucose medium for 1-7 days and then were examined with electrophysiological and molecular biology techniques. Prolonged exposure to high glucose produced a marked increase in beta-cell primary K(v) channel subunit, K(v)2.1, expression and K(v) current amplitude. Enhanced expression of ANG II type 1 receptor (AT(1)R) was also observed under high glucose conditions, whereas blockade of AT(1)R by losartan did not alter K(v) channel expression. External application of ANG II reduced K(v) current amplitude under normal, but not high, glucose conditions. The effect of ANG II on K(v) channel gating was abolished by ANG II type 2 receptor (AT(2)R) antagonism. These data suggest that hyperglycemia alters beta-cell function through modification of the K(v) channel which may be associated with the RAS.


Asunto(s)
Angiotensina II/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Potasio/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Sistema Renina-Angiotensina , Canales de Potasio Shab/metabolismo , Transducción de Señal , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II , Animales , Células Cultivadas , Imidazoles/farmacología , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Activación del Canal Iónico , Losartán/farmacología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Piridinas/farmacología , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Sistema Renina-Angiotensina/efectos de los fármacos , Canales de Potasio Shab/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
7.
Adv Exp Med Biol ; 654: 339-61, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20217505

RESUMEN

Pancreatic islets secrete multiple factors that act as endocrine, paracrine, and/or autocrine pathways in regulating pancreatic endocrine function. As such, the islets perform critical biological activities in synthesizing metabolic peptide hormones, notably insulin and regulating body glucose homeostasis. These functions are controlled by various conditions and signaling molecules, particularly nutrients like glucose levels. However, more and more clinically relevant regulators, including molecules which stimulate islet beta-cell metabolism, regulate beta-cell [Ca(2+)] homeostasis and related channels or adjust beta-cell membrane, and nuclear receptors activity continue to be discovered and characterized. Of great interest in this context, glucagon-like peptide-1 can improve glycemic control by regulating insulin secretion and islet cell mass; vitamin D can regulate islet physiology directly by binding its receptors; in addition, the peptide hormone angiotensin II has been implicated in islet function and exhibits effects on islet cell secretion as well as cell mass. In this chapter, these three novel regulators in islet function and thus its clinical relevance to type 2 diabetes mellitus will undergo critical appraisal. Since all of these molecules have biological interactions with pancreatic islets, potential relationships may exist among them and they will also be discussed.


Asunto(s)
Angiotensina II/fisiología , Péptido 1 Similar al Glucagón/fisiología , Islotes Pancreáticos/metabolismo , Vitamina D/fisiología , Angiotensinas/metabolismo , Animales , Presión Sanguínea , Núcleo Celular/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Ratones , Modelos Biológicos , Péptidos/química , Vitamina D/metabolismo
8.
J Pharmacol Exp Ther ; 327(3): 683-91, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18787107

RESUMEN

LAF237 [(S)-1-[(3-hydroxy-1-adamantyl)ammo]acetyl-2-cyanopyrrolidine] is an inhibitor of dipeptidyl peptidase IV that delays the degradation of glucagon-like peptide-1 (GLP-1). Valsartan [N-(1-oxopentyl)-N-[[2'-(1H-tetrazol-5-yl)[1,1'-biphenyl]-4-yl]methyl]-l-valine] is an antagonist of the angiotensin II type 1 receptor (AT1R) that reduces the incidence of type 2 diabetes mellitus. LAF237 and valsartan act on a common target through separate pathways to improve pancreatic islet cell function. We hypothesize that the combination of these two drugs acts in a synergistic or additive manner on islet function and structure. To test this hypothesis, we performed in vitro and in vivo studies. To measure the acute effect of the treatment, pancreatic islets of db/db mice were isolated and stimulated in vitro with glucose in the presence of valsartan (1 microM) and exendin-4 (100 nM), a GLP-1 receptor agonist. Combination treatment with valsartan and exendin-4 significantly enhanced glucose-stimulated insulin secretion from isolated islets. For studies of chronic effect, db/db mice received LAF237 (1 mg/kg/day) and/or valsartan (10 mg/kg/day). Islet cell reactive oxygen species (ROS), proliferation, apoptosis, fibrosis, beta-cell area, and glucose homeostasis were evaluated after 8 weeks of treatment, which showed that combination treatment resulted in a significant increase in pancreatic islet beta-cell area compared with monotherapy. This beneficial effect correlated with an increase in beta-cell proliferation and a decrease in ROS-induced islet apoptosis and fibrosis. These in vitro and in vivo data indicate that combination treatment with LAF237 and valsartan has significant beneficial additive effects on pancreatic beta-cell structure and function compared with their respective monotherapeutic effects.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Islotes Pancreáticos/efectos de los fármacos , Adamantano/análogos & derivados , Adamantano/farmacología , Adamantano/uso terapéutico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Apoptosis , Proliferación Celular , Diabetes Mellitus Tipo 2/patología , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Sinergismo Farmacológico , Quimioterapia Combinada , Fibrosis , Glucosa/farmacología , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/patología , Ratones , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , Especies Reactivas de Oxígeno , Tetrazoles/farmacología , Tetrazoles/uso terapéutico , Valina/análogos & derivados , Valina/farmacología , Valina/uso terapéutico , Valsartán
9.
Cell Death Differ ; 24(2): 225-237, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27740628

RESUMEN

Bone plays a role in energy metabolism, but the interplay between bone and other organs in this process is not completely understood. Here, we show that upregulated Hh signaling in bones results in increased whole-body energy expenditure, white adipose tissue (WAT) browning, hypoglycemia and skeletal muscle atrophy. We found that Hh signaling induces PTHrP secretion from bones and causes WAT browning. Injection of PTHrP-neutralizing antibody attenuates WAT browning and improves the circulating blood glucose level while high-fat diet treatment only rescues hypoglycemia. Furthermore, bone-derived PTHrP stimulates adiponectin secretion in WAT and results in systemic increase of fatty acid oxidation and glucose uptake. Mechanistically, PTHrP activates both PKA/cAMP and Akt/Foxo pathways for Ucp1 expression in WAT. PTHrP couples adiponectin actions to activate the AMPK pathway in the skeletal muscles and liver, respectively, for fatty acid oxidation. Our findings establish a new bone-adipose hormonal relay that regulates whole-body energy metabolism.


Asunto(s)
Adiponectina/metabolismo , Huesos/metabolismo , Metabolismo Energético/fisiología , Proteínas Hedgehog/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Transducción de Señal , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Células Cultivadas , Humanos , Hipoglucemia/metabolismo , Hipoglucemia/patología , Insulina/metabolismo , Peroxidación de Lípido , Ratones , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Consumo de Oxígeno , Proteína Relacionada con la Hormona Paratiroidea/inmunología , Receptor Patched-1/genética , Receptor Patched-1/metabolismo
10.
Mol Cell Endocrinol ; 404: 56-66, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25622782

RESUMEN

The widely used lipid-lowering drug niacin is reported to induce hyperglycemia during chronic and high-dose treatments, but the mechanism is poorly understood. Recently, the niacin receptor [G-protein-coupled receptor, (GPR) 109a], has been localized to islet cells while its potential role therein remains unclear. We, therefore, aimed at investigating how GPR109a regulates islet beta-cell function and its downstream signaling using high-fat diet-induced obese mice and INS-1E beta cells. Eight-week niacin treatment elevated blood glucose concentration in obese mice with increased areas under the curve at oral glucose and intraperitoneal insulin tolerance tests. Additionally, niacin treatment significantly decreased glucose-stimulated insulin secretion (GSIS) but induced peroxisome proliferator-activated receptor gamma (Pparg) and GPR109a expression in isolated pancreatic islets; concomitantly, reactive oxygen species (ROS) were transiently increased, with decreases in GSIS, intracellular cyclic adenosine monophosphate (cAMP) accumulation and mitochondrial membrane potential (ΔΨm), but with increased expression of uncoupling protein 2 (Ucp2), Pparg and Gpr109a in INS-1E cells. Corroborating these findings, the decreases in GSIS, ΔΨm and cAMP production and increases in ROS, Pparg and GPR109a expression were abolished in INS-1E cells by GPR109a knockdown. Our data indicate that niacin-induced pancreatic islet dysfunction is probably modulated through activation of the islet beta-cell GPR109a-induced ROS-PPARγ-UCP2 pathways.


Asunto(s)
Hiperglucemia/inducido químicamente , Hipolipemiantes/administración & dosificación , Células Secretoras de Insulina/patología , Niacina/efectos adversos , Obesidad/tratamiento farmacológico , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Glucosa/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Hipolipemiantes/efectos adversos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Canales Iónicos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Niacina/administración & dosificación , Obesidad/fisiopatología , PPAR gamma/metabolismo , Ratas , Proteína Desacopladora 2
11.
Stem Cell Rev Rep ; 10(2): 280-94, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24395006

RESUMEN

Recent advance in directed differentiation of pancreatic stem cells offers potential to the development of replacement therapy for diabetic patients. However, the existing differentiation protocols are complex, time-consuming, and costly; thus there is a need for alternative protocols. Given the common developmental origins of liver and pancreas, we sought to develop a novel protocol, devoid of growth factors, by using liver stromal cells (LSCs) derived from human fetal liver. We examined the effects of the LSCs on the differentiation of pancreatic progenitor cells (PPCs) into islet-like cell clusters (ICCs). PPCs and LSCs isolated from 1st to 2nd trimester human fetal tissues underwent co-cultures; differentiation and functionality of ICCs were determined by examining expression of critical markers and secretion of insulin. Co-culture with 2nd but not 1st trimester LSCs enhanced ICC differentiation and functionality without the use of exogenous differentiation 'cocktails'. Differential expression profiles of growth factors from 1st versus 2nd trimester fetal liver were compared. Many morphogenic factors were expressed by LSCs, while insulin-like growth factor 1 (IGF1) was identified as one of the key molecules responsible for the ICC differentiation. This is the first report showing that an LSC-induced microenvironment can enhance ICC differentiation and functionality. Further modifications of the stroma microenvironment may offer an alternative, efficient and cost-effective approach to providing islets for transplantation.


Asunto(s)
Diferenciación Celular , Islotes Pancreáticos/citología , Hígado/citología , Células Madre/fisiología , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/terapia , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Ratones Desnudos , Células del Estroma/fisiología , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Diabetes ; 62(11): 3751-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23897951

RESUMEN

Circulating fibroblast growth factor 21 (FGF21) levels are elevated in diabetic subjects and correlate directly with abnormal glucose metabolism, while pharmacologically administered FGF21 can ameliorate hyperglycemia. The pancreatic islet is an FGF21 target, yet the actions of FGF21 in the islet under normal and diabetic conditions are not fully understood. This study investigated the effects of high glucose on islet FGF21 actions in a diabetic mouse model by investigating db/db mouse islet responses to exogenous FGF21, the direct effects of glucose on FGF21 signaling, and the involvement of peroxisome proliferator-activated receptor γ (PPARγ) in FGF21 pathway activation. Results showed that both adult db/db mouse islets and normal islets treated with high glucose ex vivo displayed reduced ß-klotho expression, resistance to FGF21, and decreased PPARγ expression. Rosiglitazone, an antidiabetic PPARγ ligand, ameliorated these effects. Our data indicate that hyperglycemia in type 2 diabetes mellitus may lead to FGF21 resistance in pancreatic islets, probably through reduction of PPARγ expression, which provides a novel mechanism for glucose-mediated islet dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Glucosa/farmacología , Islotes Pancreáticos/metabolismo , Proteínas de la Membrana/biosíntesis , PPAR gamma/fisiología , Envejecimiento/fisiología , Animales , Modelos Animales de Enfermedad , Glucosa/administración & dosificación , Islotes Pancreáticos/efectos de los fármacos , Proteínas Klotho , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Fosforilación , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas
13.
Peptides ; 32(5): 1087-95, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21396973

RESUMEN

The traditional renin-angiotensin system (RAS) components have been studied extensively since the rate-limiting component of RAS, renin, was first characterized. The ongoing identification of various novel RAS components and signaling pathways continues to elaborate the complexity of this system. Regulation of RAS according to the conventional and contemporary views of its functions in various tissues under pathophysiological conditions is a main treatment strategy for many metabolic diseases. The local pancreatic RAS, first proposed to exist in pancreatic islets two decades ago, could regulate islet function and glycemic control via influences on islet cell mass, inflammation, and ion channels. Insulin secretion, the major function of pancreatic islets, is controlled by numerous factors. Among these factors and of particular interest are glucagon-like peptide-1 (GLP-1) and vitamin D, which may regulate islet function by directly binding receptors on islet beta cells. These factors may work with local RAS signaling in islets to protect and maintain islet function under diabetic and hyperglycemic conditions. In this concise review, the local islet RAS will be discussed with particular attention being paid to recent notable findings.


Asunto(s)
Islotes Pancreáticos/metabolismo , Sistema Renina-Angiotensina/fisiología , Animales , Diabetes Mellitus/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Humanos
14.
Diabetes ; 60(1): 320-30, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20870970

RESUMEN

OBJECTIVE: Suppression of Kinesin-1 by antisense oligonucleotides, or overexpression of dominant-negative acting kinesin heavy chain, has been reported to affect the sustained phase of glucose-stimulated insulin secretion in ß-cells in vitro. In this study, we examined the in vivo physiological role of Kinesin-1 in ß-cell development and function. RESEARCH DESIGN AND METHODS: A Cre-LoxP strategy was used to generate conditional knockout mice in which the Kif5b gene is specifically inactivated in pancreatic ß-cells. Physiological and histological analyses were carried out in Kif5b knockout mice as well as littermate controls. RESULTS: Mice with ß-cell specific deletion of Kif5b (Kif5b(fl/)⁻:RIP2-Cre) displayed significantly retarded growth as well as slight hyperglycemia in both nonfasting and 16-h fasting conditions compared with control littermates. In addition, Kif5b(fl/)⁻:RIP2-Cre mice displayed significant glucose intolerance, which was not due to insulin resistance but was related to an insulin secretory defect in response to glucose challenge. These defects of ß-cell function in mutant mice were not coupled with observable changes in islet morphology, islet cell composition, or ß-cell size. However, compared with controls, pancreas of Kif5b(fl/)⁻:RIP2-Cre mice exhibited both reduced islet size and increased islet number, concomitant with an increased insulin vesicle density in ß-cells. CONCLUSIONS: In addition to being essential for maintaining glucose homeostasis and regulating ß-cell function, Kif5b may be involved in ß-cell development by regulating ß-cell proliferation and insulin vesicle synthesis.


Asunto(s)
Células Secretoras de Insulina/fisiología , Insulina/metabolismo , Cinesinas/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Genotipo , Humanos , Insulina/deficiencia , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Cinesinas/química , Cinesinas/deficiencia , Cinesinas/efectos de los fármacos , Cinesinas/genética , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/farmacología , Fragmentos de Péptidos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA