Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(11): 113326, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37897727

RESUMEN

Glucagon-like peptide 1 (GLP-1R) and glucose-dependent insulinotropic polypeptide (GIPR) receptors are G-protein-coupled receptors involved in glucose homeostasis. Diabetogenic conditions decrease ß-arrestin 2 (ARRB2) levels in human islets. In mouse ß cells, ARRB2 dampens insulin secretion by partially uncoupling cyclic AMP (cAMP)/protein kinase A (PKA) signaling at physiological doses of GLP-1, whereas at pharmacological doses, the activation of extracellular signal-related kinase (ERK)/cAMP-responsive element-binding protein (CREB) requires ARRB2. In contrast, GIP-potentiated insulin secretion needs ARRB2 in mouse and human islets. The GIPR-ARRB2 axis is not involved in cAMP/PKA or ERK signaling but does mediate GIP-induced F-actin depolymerization. Finally, the dual GLP-1/GIP agonist tirzepatide does not require ARRB2 for the potentiation of insulin secretion. Thus, ARRB2 plays distinct roles in regulating GLP-1R and GIPR signaling, and we highlight (1) its role in the physiological context and the possible functional consequences of its decreased expression in pathological situations such as diabetes and (2) the importance of assessing the signaling pathways engaged by the agonists (biased/dual) for therapeutic purposes.


Asunto(s)
Células Secretoras de Insulina , Ratones , Humanos , Animales , Células Secretoras de Insulina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Arrestina beta 2/metabolismo , beta-Arrestina 1/metabolismo , Glucosa/metabolismo
2.
Front Endocrinol (Lausanne) ; 13: 918733, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35813647

RESUMEN

Pancreatic islets are highly vascularized micro-organs ensuring whole body glucose homeostasis. Islet vascular cells play an integral part in sustaining adequate insulin release by beta cells. In particular, recent studies have demonstrated that islet pericytes regulate local blood flow velocity and are required for maintenance of beta cell maturity and function. In addition, increased metabolic demand accompanying obesity alters islet pericyte morphology. Here, we sought to explore the effects of metabolic stress on islet pericyte functional response to stimulation in a mouse model of type 2 diabetes, directly in the pancreas in vivo . We found that high fat diet induced islet pericyte hypertrophy without alterations in basal local blood flow. However, optogenetic stimulation of pericyte activity revealed impaired islet vascular responses, despite increased expression of genes encoding proteins directly or indirectly involved in cell contraction. These findings suggest that metabolic stress impinges upon islet pericyte function, which may contribute to beta cell failure during T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Optogenética , Pericitos , Estrés Fisiológico
3.
Cell Death Dis ; 13(4): 353, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35428762

RESUMEN

Pancreatic ß-cell failure in type 2 diabetes mellitus (T2DM) is associated with impaired regulation of autophagy which controls ß-cell development, function, and survival through clearance of misfolded proteins and damaged organelles. However, the mechanisms responsible for defective autophagy in T2DM ß-cells remain unknown. Since recent studies identified circadian clock transcriptional repressor REV-ERBα as a novel regulator of autophagy in cancer, in this study we set out to test whether REV-ERBα-mediated inhibition of autophagy contributes to the ß-cell failure in T2DM. Our study provides evidence that common diabetogenic stressors (e.g., glucotoxicity and cytokine-mediated inflammation) augment ß-cell REV-ERBα expression and impair ß-cell autophagy and survival. Notably, pharmacological activation of REV-ERBα was shown to phenocopy effects of diabetogenic stressors on the ß-cell through inhibition of autophagic flux, survival, and insulin secretion. In contrast, negative modulation of REV-ERBα was shown to provide partial protection from inflammation and glucotoxicity-induced ß-cell failure. Finally, using bioinformatic approaches, we provide further supporting evidence for augmented REV-ERBα activity in T2DM human islets associated with impaired transcriptional regulation of autophagy and protein degradation pathways. In conclusion, our study reveals a previously unexplored causative relationship between REV-ERBα expression, inhibition of autophagy, and ß-cell failure in T2DM.


Asunto(s)
Relojes Circadianos , Diabetes Mellitus Tipo 2 , Autofagia/genética , Ritmo Circadiano/fisiología , Diabetes Mellitus Tipo 2/genética , Humanos , Inflamación , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo
4.
Int J Mol Sci ; 22(10)2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-34069914

RESUMEN

Type 2 diabetes (T2D) is characterized by chronic hyperglycemia secondary to the decline of functional beta-cells and is usually accompanied by a reduced sensitivity to insulin. Whereas altered beta-cell function plays a key role in T2D onset, a decreased beta-cell mass was also reported to contribute to the pathophysiology of this metabolic disease. The decreased beta-cell mass in T2D is, at least in part, attributed to beta-cell apoptosis that is triggered by diabetogenic situations such as amyloid deposits, lipotoxicity and glucotoxicity. In this review, we discussed the molecular mechanisms involved in pancreatic beta-cell apoptosis under such diabetes-prone situations. Finally, we considered the molecular signaling pathways recruited by glucagon-like peptide-1-based therapies to potentially protect beta-cells from death under diabetogenic situations.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/fisiología , Animales , Apoptosis/fisiología , Células Cultivadas , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/farmacología , Péptido 1 Similar al Glucagón/fisiología , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Transducción de Señal
5.
Methods Mol Biol ; 1957: 345-364, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30919365

RESUMEN

Novel findings reveal important functional roles for ß-arrestin 1 and ß-arrestin 2 in the regulation of insulin secretion, ß-cell survival, and ß-cell mass plasticity not only by glucose but also by G-protein-coupled receptors, such as the glucagon-like peptide-1 (GLP-1) and the pituitary adenylate cyclase-activating polypeptide (PACAP) receptors or GPR40, or tyrosine kinase receptors, such as the insulin receptor. Here, we describe experimental protocols to knock down ß-arrestins by small interference RNA, to follow subcellular localization of ß-arrestins in the cytosol and nucleus of the insulinoma INS-1E rat pancreatic ß-cell line, and to analyze ß-arrestin protein expression by Western blot using INS-1E cells and isolated mouse or human pancreatic islets. We also provide details on how to genotype ß-arrestin 2 knockout (Arrb2-/-) mice and to evaluate ß-arrestin-mediated roles in ß-cell mass plasticity and ß-cell signaling using immunocytochemistry on pancreatic sections or on primary dispersed ß-cells from wild-type mice and Arrb2-/- mice.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Biología Molecular/métodos , beta-Arrestinas/metabolismo , Animales , Citoplasma/metabolismo , Técnicas de Silenciamiento del Gen , Genotipo , Ratones Noqueados , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/metabolismo
6.
Cell Death Dis ; 9(6): 600, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789539

RESUMEN

In type 2 diabetes, amyloid oligomers, chronic hyperglycemia, lipotoxicity, and pro-inflammatory cytokines are detrimental to beta-cells, causing apoptosis and impaired insulin secretion. The histone acetyl transferase p300, involved in remodeling of chromatin structure by epigenetic mechanisms, is a key ubiquitous activator of the transcriptional machinery. In this study, we report that loss of p300 acetyl transferase activity and expression leads to beta-cell apoptosis, and most importantly, that stress situations known to be associated with diabetes alter p300 levels and functional integrity. We found that proteasomal degradation is the mechanism subserving p300 loss in beta-cells exposed to hyperglycemia or pro-inflammatory cytokines. We also report that melatonin, a hormone produced in the pineal gland and known to play key roles in beta-cell health, preserves p300 levels altered by these toxic conditions. Collectively, these data imply an important role for p300 in the pathophysiology of diabetes.


Asunto(s)
Diabetes Mellitus/enzimología , Diabetes Mellitus/patología , Proteína p300 Asociada a E1A/metabolismo , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Acetilación , Animales , Apoptosis/efectos de los fármacos , Citocinas/metabolismo , Proteína p300 Asociada a E1A/genética , Glucosa/toxicidad , Histonas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Melatonina/metabolismo , Ratones Endogámicos C57BL , Proteolisis/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Melatonina/metabolismo , Transducción de Señal
7.
Diabetologia ; 60(10): 1999-2010, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28721437

RESUMEN

AIMS/HYPOTHESIS: Insufficient insulin secretion from pancreatic beta cells, which is associated with a decrease in beta cell mass, is a characteristic of type 2 diabetes. Extracellular signal-related kinase 1 and 2 (ERK1/2) inhibition in beta cells has been reported to affect insulin secretion, gene transcription and survival, although whether ERK1 and ERK2 play distinct roles is unknown. The aim of this study was to assess the individual roles of ERK1 and ERK2 in beta cells using ERK1 (also known as Mapk3)-knockout mice (Erk1 -/- mice) and pharmacological approaches. METHODS: NAD(P)H, free cytosolic Ca2+ concentration and insulin secretion were determined in islets. ERK1 and ERK2 subplasmalemmal translocation and activity was monitored using total internal reflection fluorescence microscopy. ERK1/2, mitogen and stress-activated kinase1 (MSK1) and cAMP-responsive element-binding protein (CREB) activation were evaluated by western blot and/or immunocytochemistry. The islet mass was determined from pancreatic sections. RESULTS: Glucose induced rapid subplasmalemmal recruitment of ERK1 and ERK2. When both ERK1 and ERK2 were inhibited simultaneously, the rapid transient peak of the first phase of glucose-induced insulin secretion was reduced by 40% (p < 0.01), although ERK1 did not appear to be involved in this process. By contrast, ERK1 was required for glucose-induced full activation of several targets involved in beta cell survival; MSK1 and CREB were less active in Erk1 -/- mouse beta cells (p < 0.01) compared with Erk1 +/+ mouse beta cells, and their phosphorylation could only be restored when ERK1 was re-expressed and not when ERK2 was overexpressed. Finally, the islet mass of Erk1 -/- mice was slightly increased in young animals (4-month-old mice) vs Erk1 +/+ mice (section occupied by islets [mean ± SEM]: 0.74% ± 0.03% vs 0.62% ± 0.04%; p < 0.05), while older mice (10 months old) were less prone to age-associated pancreatic peri-insulitis (infiltrated islets [mean ± SEM]: 7.51% ± 1.34% vs 2.03% ± 0.51%; p < 0.001). CONCLUSIONS/INTERPRETATION: ERK1 and ERK2 play specific roles in beta cells. ERK2 cannot always compensate for the lack of ERK1 but the absence of a clear-cut phenotype in Erk1 -/- mice shows that ERK1 is dispensable in normal conditions.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética
8.
J Biol Chem ; 290(34): 21131-21140, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26157145

RESUMEN

FFAR1/GPR40 is a seven-transmembrane domain receptor (7TMR) expressed in pancreatic ß cells and activated by FFAs. Pharmacological activation of GPR40 is a strategy under consideration to increase insulin secretion in type 2 diabetes. GPR40 is known to signal predominantly via the heterotrimeric G proteins Gq/11. However, 7TMRs can also activate functionally distinct G protein-independent signaling via ß-arrestins. Further, G protein- and ß-arrestin-based signaling can be differentially modulated by different ligands, thus eliciting ligand-specific responses ("biased agonism"). Whether GPR40 engages ß-arrestin-dependent mechanisms and is subject to biased agonism is unknown. Using bioluminescence resonance energy transfer-based biosensors for real-time monitoring of cell signaling in living cells, we detected a ligand-induced GPR40-ß-arrestin interaction, with the synthetic GPR40 agonist TAK-875 being more effective than palmitate or oleate in recruiting ß-arrestins 1 and 2. Conversely, TAK-875 acted as a partial agonist of Gq/11-dependent GPR40 signaling relative to both FFAs. Pharmacological blockade of Gq activity decreased FFA-induced insulin secretion. In contrast, knockdown or genetic ablation of ß-arrestin 2 in an insulin-secreting cell line and mouse pancreatic islets, respectively, uniquely attenuated the insulinotropic activity of TAK-875, thus providing functional validation of the biosensor data. Collectively, these data reveal that in addition to coupling to Gq/11, GPR40 is functionally linked to a ß-arrestin 2-mediated insulinotropic signaling axis. These observations expose previously unrecognized complexity for GPR40 signal transduction and may guide the development of biased agonists showing improved clinical profile in type 2 diabetes.


Asunto(s)
Arrestinas/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Animales , Arrestinas/antagonistas & inhibidores , Arrestinas/metabolismo , Benzofuranos/farmacología , Técnicas Biosensibles , Línea Celular Tumoral , Espectroscopía de Resonancia por Spin del Electrón , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Insulina/agonistas , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Cinética , Ratones , Ácido Oléico/farmacología , Ácido Palmítico/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Sulfonas/farmacología , Técnicas de Cultivo de Tejidos , Arrestina beta 2 , beta-Arrestinas
9.
EMBO Mol Med ; 7(6): 802-18, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25828351

RESUMEN

The metabolic syndrome covers metabolic abnormalities including obesity and type 2 diabetes (T2D). T2D is characterized by insulin resistance resulting from both environmental and genetic factors. A genome-wide association study (GWAS) published in 2010 identified TP53INP1 as a new T2D susceptibility locus, but a pathological mechanism was not identified. In this work, we show that mice lacking TP53INP1 are prone to redox-driven obesity and insulin resistance. Furthermore, we demonstrate that the reactive oxygen species increase in TP53INP1-deficient cells results from accumulation of defective mitochondria associated with impaired PINK/PARKIN mitophagy. This chronic oxidative stress also favors accumulation of lipid droplets. Taken together, our data provide evidence that the GWAS-identified TP53INP1 gene prevents metabolic syndrome, through a mechanism involving prevention of oxidative stress by mitochondrial homeostasis regulation. In conclusion, this study highlights TP53INP1 as a molecular regulator of redox-driven metabolic syndrome and provides a new preclinical mouse model for metabolic syndrome clinical research.


Asunto(s)
Síndrome Metabólico/fisiopatología , Mitofagia , Proteínas Nucleares/metabolismo , Animales , Modelos Animales de Enfermedad , Resistencia a la Insulina , Ratones , Proteínas Nucleares/deficiencia , Obesidad , Oxidación-Reducción , Estrés Oxidativo , Especies Reactivas de Oxígeno/análisis
10.
Diabetologia ; 57(3): 532-41, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24317793

RESUMEN

AIMS/HYPOTHESIS: Beta cell failure due to progressive secretory dysfunction and limited expansion of beta cell mass is a key feature of type 2 diabetes. Beta cell function and mass are controlled by glucose and hormones/neurotransmitters that activate G protein-coupled receptors or receptor tyrosine kinases. We have investigated the role of ß-arrestin (ARRB)2, a scaffold protein known to modulate such receptor signalling, in the modulation of beta cell function and mass, with a specific interest in glucagon-like peptide-1 (GLP-1), muscarinic and insulin receptors. METHODS: ß-arrestin2-knockout mice and their wild-type littermates were fed a normal or a high-fat diet (HFD). Glucose tolerance, insulin sensitivity and insulin secretion were assessed in vivo. Beta cell mass was evaluated in pancreatic sections. Free cytosolic [Ca(2+)] and insulin secretion were determined using perifused islets. The insulin signalling pathway was evaluated by western blotting. RESULTS: Arrb2-knockout mice exhibited impaired glucose tolerance and insulin secretion in vivo, but normal insulin sensitivity compared with wild type. Surprisingly, the absence of ARRB2 did not affect glucose-stimulated insulin secretion or GLP-1- and acetylcholine-mediated amplifications from perifused islets, but it decreased the islet insulin content and beta cell mass. Additionally, there was no compensatory beta cell mass expansion through proliferation in response to the HFD. Furthermore, Arrb2 deletion altered the islet insulin signalling pathway. CONCLUSIONS/INTERPRETATION: ARRB2 is unlikely to be involved in the regulation of insulin secretion, but it is required for beta cell mass plasticity. Additionally, we provide new insights into the mechanisms involved in insulin signalling in beta cells.


Asunto(s)
Arrestinas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Páncreas/metabolismo , Animales , Western Blotting , Dieta Alta en Grasa , Secreción de Insulina , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Insulina , Transducción de Señal , Arrestina beta 2 , beta-Arrestinas
11.
Cell Signal ; 23(3): 522-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20849951

RESUMEN

Defective insulin secretion is a feature of type 2 diabetes that results from inadequate compensatory increase in ß-cell mass, decreased ß-cell survival and impaired glucose-dependent insulin release. Pancreatic ß-cell proliferation, survival and secretion are thought to be regulated by signalling pathways linked to G-protein coupled receptors (GPCRs), such as the glucagon-like peptide-1 (GLP-1) and the pituitary adenylate cyclase-activating polypeptide (PACAP) receptors. ß-arrestin-1 serves as a multifunctional adaptor protein that mediates receptor desensitization, receptor internalization, and links GPCRs to downstream pathways such as tyrosine kinase Src, ERK1/2 or Akt/PKB. Importantly, recent studies found that ß-arrestin-1 mediates GLP-1 signalling to insulin secretion, GLP-1 antiapoptotic effect by phosphorylating the proapoptotic protein Bad through ERK1/2 activation, and PACAP potentiation of glucose-induced long-lasting ERK1/2 activation controlling IRS-2 expression. Together, these novel findings reveal an important functional role for ß-arrestin-1 in the regulation of insulin secretion and ß-cell survival by GPCRs.


Asunto(s)
Arrestinas/fisiología , Diabetes Mellitus/patología , Células Secretoras de Insulina/fisiología , Animales , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Evaluación Preclínica de Medicamentos , Receptor del Péptido 1 Similar al Glucagón , Glucosa/fisiología , Humanos , Células Secretoras de Insulina/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glucagón/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Transducción de Señal , beta-Arrestina 1 , beta-Arrestinas
12.
J Biol Chem ; 285(3): 1989-2002, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19915011

RESUMEN

Strategies based on activating GLP-1 receptor (GLP-1R) are intensively developed for the treatment of type 2 diabetes. The exhaustive knowledge of the signaling pathways linked to activated GLP-1R within the beta-cells is of major importance. In beta-cells, GLP-1 activates the ERK1/2 cascade by diverse pathways dependent on either Galpha(s)/cAMP/cAMP-dependent protein kinase (PKA) or beta-arrestin 1, a scaffold protein. Using pharmacological inhibitors, beta-arrestin 1 small interfering RNA, and islets isolated from beta-arrestin 1 knock-out mice, we demonstrate that GLP-1 stimulates ERK1/2 by two temporally distinct pathways. The PKA-dependent pathway mediates rapid and transient ERK1/2 phosphorylation that leads to nuclear translocation of the activated kinases. In contrast, the beta-arrestin 1-dependent pathway produces a late ERK1/2 activity that is restricted to the beta-cell cytoplasm. We further observe that GLP-1 phosphorylates the cytoplasmic proapoptotic protein Bad at Ser-112 but not at Ser-155. We find that the beta-arrestin 1-dependent ERK1/2 activation engaged by GLP-1 mediates the Ser-112 phosphorylation of Bad, through p90RSK activation, allowing the association of Bad with the scaffold protein 14-3-3, leading to its inactivation. beta-Arrestin 1 is further found to mediate the antiapoptotic effect of GLP-1 in beta-cells through the ERK1/2-p90RSK-phosphorylation of Bad. This new regulatory mechanism engaged by activated GLP-1R involving a beta-arrestin 1-dependent spatiotemporal regulation of the ERK1/2-p90RSK activity is now suspected to participate in the protection of beta-cells against apoptosis. Such signaling mechanism may serve as a prototype to generate new therapeutic GLP-1R ligands.


Asunto(s)
Apoptosis/efectos de los fármacos , Arrestinas/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Proteínas 14-3-3/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteína Letal Asociada a bcl/química , beta-Arrestina 1 , beta-Arrestinas
13.
EMBO Rep ; 10(8): 873-80, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19575010

RESUMEN

A previously uncharacterized putative ion channel, NALCN (sodium leak channel, non-selective), has been recently shown to be responsible for the tetrodotoxin (TTX)-resistant sodium leak current implicated in the regulation of neuronal excitability. Here, we show that NALCN encodes a current that is activated by M3 muscarinic receptors (M3R) in a pancreatic beta-cell line. This current is primarily permeant to sodium ions, independent of intracellular calcium stores and G proteins but dependent on Src activation, and resistant to TTX. The current is recapitulated by co-expression of NALCN and M3R in human embryonic kidney-293 cells and in Xenopus oocytes. We also show that NALCN and M3R belong to the same protein complex, involving the intracellular I-II loop of NALCN and the intracellular i3 loop of M3R. Taken together, our data show the molecular basis of a muscarinic-activated inward sodium current that is independent of G-protein activation, and provide new insights into the properties of NALCN channels.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Canales Iónicos/metabolismo , Receptor Muscarínico M3/metabolismo , Western Blotting , Línea Celular , Electrofisiología , Inhibidores Enzimáticos/farmacología , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Canales Iónicos/genética , Técnicas de Placa-Clamp , Pirazoles/farmacología , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Receptor Muscarínico M3/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Diabetes ; 58(5): 1105-15, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19223597

RESUMEN

OBJECTIVE: In type 2 diabetes, chronic hyperglycemia is detrimental to beta-cells, causing apoptosis and impaired insulin secretion. The transcription factor cAMP-responsive element-binding protein (CREB) is crucial for beta-cell survival and function. We investigated whether prolonged exposure of beta-cells to high glucose affects the functional integrity of CREB. RESEARCH DESIGN AND METHODS: INS-1E cells and rat and human islets were used. Gene expression was analyzed by RT-PCR and Western blotting. Apoptosis was detected by cleaved caspase-3 emergence, DNA fragmentation, and electron microscopy. RESULTS: Chronic exposure of INS-1E cells and rat and human islets to high glucose resulted in decreased CREB protein expression, phosphorylation, and transcriptional activity associated with apoptosis and impaired beta-cell function. High-glucose treatment increased CREB polyubiquitination, while treatment of INS-1E cells with the proteasome inhibitor MG-132 prevented the decrease in CREB content. The emergence of apoptosis in INS-1E cells with decreased CREB protein expression knocked down by small interfering RNA suggested that loss of CREB protein content induced by high glucose contributes to beta-cell apoptosis. Loading INS-1E cells or human islets with a cell-permeable peptide mimicking the proteasomal targeting sequence of CREB blocked CREB degradation and protected INS-1E cells and human islets from apoptosis induced by high glucose. The insulin secretion in response to glucose and the insulin content were preserved in human islets exposed to high glucose and loaded with the peptide. CONCLUSIONS: These studies demonstrate that the CREB degradation by the ubiquitin-proteasome pathway contributes to beta-cell dysfunction and death upon glucotoxicity and provide new insight into the cellular mechanisms of glucotoxicity.


Asunto(s)
Modulador del Elemento de Respuesta al AMP Cíclico/metabolismo , Glucosa/toxicidad , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Muerte Encefálica , Proteína de Unión a CREB/efectos de los fármacos , Proteína de Unión a CREB/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Modulador del Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Fragmentación del ADN , Diabetes Mellitus Experimental/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Biol Chem ; 284(7): 4332-42, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19074139

RESUMEN

In pancreatic beta-cells, the pituitary adenylate cyclase-activating polypeptide (PACAP) exerts a potent insulin secretory effect via PAC(1) and VPAC receptors (Rs) through the Galpha(s)/cAMP/protein kinase A pathway. Here, we investigated the mechanisms linking PAC(1)R to ERK1/2 activation in INS-1E beta-cells and pancreatic islets. PACAP caused a transient (5 min) increase in ERK1/2 phosphorylation via PAC(1)Rs and promoted nuclear translocation of a fraction of cytosolic p-ERK1/2. Both protein kinase A- and Src-dependent pathways mediated this transient ERK1/2 activation. Moreover, PACAP potentiated glucose-induced long-lasting ERK1/2 activation. Blocking Ca(2+) influx abolished glucose-induced ERK1/2 activation and PACAP potentiating effect. Glucose stimulation during KCl depolarization showed that, in addition to the triggering signal (rise in cytosolic [Ca(2+)]), the amplifying pathway was also involved in glucose-induced sustained ERK1/2 activation and was required for PACAP potentiation. The finding that at 30 min glucose-induced p-ERK1/2 was detected in both cytosol and nucleus while the potentiating effect of PACAP was only observed in the cytosol, suggested the involvement of the scaffold protein beta-arrestin. Indeed, beta-arrestin 1 (beta-arr1) depletion (in beta-arr1 knockout mouse islets or in INS-1E cells by siRNA) completely abolished PACAP potentiation of long-lasting ERK1/2 activation by glucose. Finally, PACAP potentiated glucose-induced CREB transcriptional activity and IRS-2 mRNA expression mainly via the ERK1/2 signaling pathway, and likewise, beta-arr1 depletion reduced the PACAP potentiating effect on IRS-2 expression. These results establish for the first time that PACAP potentiates glucose-induced long-lasting ERK1/2 activation via a beta-arr1-dependent pathway and thus provide new insights concerning the mechanisms of PACAP and glucose actions in pancreatic beta-cells.


Asunto(s)
Arrestinas/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Edulcorantes/farmacología , Animales , Arrestinas/genética , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glucosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Células Secretoras de Insulina/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Edulcorantes/metabolismo , Factores de Tiempo , beta-Arrestina 1 , beta-Arrestinas
16.
Endocrinology ; 148(9): 4136-46, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17495006

RESUMEN

Recently, we synthesized and characterized the first selective V(1b) vasopressin (VP)/oxytocin receptor agonist, d[Cha(4)]arginine vasopressin. However, this agonist was only selective for the human receptors. We thus decided to design a selective V(1b) agonist for the rodent species. We started from previous observations showing that modifying [deamino(1),Arg(8)]VP in positions 4 and 8 altered the rat VP/oxytocin receptor selectivity. We synthesized a series of 13 [deamino(1),Arg(8)]VP analogs modified in positions 4 and 8. Among them, one seemed very promising, d[Leu(4), Lys(8)]VP. In this paper, we describe its pharmacological and physiological properties. This analog exhibited a nanomolar affinity for the rat, human, and mouse V(1b) VP receptors and a strong V(1b) selectivity for the rat species. On AtT20 cells stably transfected with the rat V(1b) receptor, d[Leu(4), Lys(8)]VP behaved as a full agonist on both phospholipase C and MAPK assays. Additional experiments revealed its ability to induce the internalization of enhanced green fluorescent protein-tagged human and mouse V(1b) receptors as expected for a full agonist. Additional physiological experiments were performed to further confirm the selectivity of this peptide. Its antidiuretic, vasopressor, and in vitro oxytocic activities were weak compared with those of VP. In contrast, used at low doses, its efficiency to stimulate adrenocorticotropin or insulin release from mouse pituitary or perfused rat pancreas, respectively, was similar to that obtained with VP. In conclusion, d[Leu(4), Lys(8)]VP is the first selective agonist available for the rat V(1b) VP receptor. It will allow a better understanding of V(1b) receptor-mediated effects in rodents.


Asunto(s)
Lipresina/análogos & derivados , Receptores de Oxitocina/agonistas , Receptores de Vasopresinas/agonistas , Adenilil Ciclasas/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Humanos , Riñón/efectos de los fármacos , Riñón/fisiología , Lactancia , Hígado/efectos de los fármacos , Hígado/fisiología , Lipresina/síntesis química , Lipresina/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/fisiología , Ratones , Adenohipófisis/efectos de los fármacos , Adenohipófisis/fisiología , Ratas , Ratas Wistar , Receptores de Oxitocina/efectos de los fármacos , Receptores de Oxitocina/genética , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/efectos de los fármacos , Transfección
17.
Diabetes ; 55(8): 2220-30, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16873684

RESUMEN

cAMP-responsive element-binding protein (CREB) is required for beta-cell survival by regulating expression of crucial genes such as bcl-2 and IRS-2. Using MIN6 cells and isolated rat pancreatic islets, we investigated the signaling pathway that controls phosphorylation and protein level of CREB. We observed that 10 mmol/l glucose-induced CREB phosphorylation was totally inhibited by the protein kinase A (PKA) inhibitor H89 (2 micromol/l) and reduced by 50% with the extracellular signal-regulated kinase (ERK)1/2 inhibitor PD98059 (20 micromol/l). This indicates that ERK1/2, reported to be located downstream of PKA, participates in the PKA-mediated CREB phosphorylation elicited by glucose. In ERK1/2-downregulated MIN6 cells by siRNA, glucose-stimulated CREB phosphorylation was highly reduced and CREB protein content was decreased by 60%. In MIN6 cells and islets cultured for 24-48 h in optimal glucose concentration (10 mmol/l), which promotes survival, blockade of ERK1/2 activity with PD98059 caused a significant decrease in CREB protein level, whereas CREB mRNA remained unaffected (measured by real-time quantitative PCR). This was associated with loss of bcl-2 mRNA and protein contents, caspase-3 activation, and emergence of ultrastructural apoptotic features detected by electron microscopy. Our results indicate that ERK1 and -2 control the phosphorylation and protein level of CREB and play a key role in glucose-mediated pancreatic beta-cell survival.


Asunto(s)
Proteína de Unión a CREB/análisis , Proteína de Unión a CREB/metabolismo , Supervivencia Celular , Islotes Pancreáticos/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Animales , Apoptosis , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Flavonoides/farmacología , Expresión Génica , Genes bcl-2/genética , Glucosa/farmacología , Islotes Pancreáticos/ultraestructura , Isoquinolinas/farmacología , Masculino , Microscopía Electrónica de Transmisión , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/análisis , ARN Mensajero/análisis , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Sulfonamidas/farmacología , Transfección
18.
J Biol Chem ; 277(36): 32883-91, 2002 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-12087106

RESUMEN

Glucose increases insulin secretion by raising cytoplasmic Ca(2+) ([Ca(2+)](i)) in beta-cells (triggering pathway) and augmenting the efficacy of Ca(2+) on exocytosis (amplifying pathway). It has been suggested that glutamate formed from alpha-ketoglutarate is a messenger of the amplifying pathway (Maechler, P., and Wollheim, C. B. (1999) Nature 402, 685-689). This hypothesis was tested with mouse islets depolarized with 30 mm KCl (+ diazoxide) or with a saturating concentration of sulfonylurea. Because [Ca(2+)](i) was elevated under these conditions, insulin secretion was stimulated already in 0 mm glucose. The amplification of secretion produced by glucose was accompanied by an increase in islet glutamate. However, glutamine (0.5-2 mm) markedly augmented islet glutamate without affecting insulin secretion, whereas glucose augmented secretion without influencing glutamate levels when these were elevated by glutamine. Allosteric activation of glutamate dehydrogenase by BCH (2-amino 2-norbornane carboxylic acid) lowered islet glutamate but increased insulin secretion. Similar insulin secretion thus occurred at very different cellular glutamate levels. Glutamine did not affect islet [Ca(2+)](i) and pH(i), whereas glucose and BCH slightly raised pH(i) and either slightly decreased (30 mm KCl) or increased (tolbutamide) [Ca(2+)](i). The general dissociation between changes in islet glutamate and insulin secretion refutes a role of beta-cell glutamate in the amplification of insulin secretion by glucose.


Asunto(s)
Calcio/metabolismo , Glucosa/farmacología , Ácido Glutámico/metabolismo , Insulina/metabolismo , Sitio Alostérico , Aminoácidos Cíclicos/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Exocitosis , Femenino , Glucosa/metabolismo , Concentración de Iones de Hidrógeno , Secreción de Insulina , Masculino , Ratones , Páncreas , Potasio/metabolismo , Cloruro de Potasio/farmacología , Unión Proteica , Ratas , Ratas Wistar , Factores de Tiempo , Tolbutamida/farmacología
19.
Br J Clin Pharmacol ; 53(6): 641-3, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12047489

RESUMEN

AIMS: To investigate the effects of glutamate on insulin secretion and glucose tolerance in humans. METHODS: Monosodium (L)-glutamate (10 g) was given orally in a double-blind placebo-controlled cross-over study to 18 healthy volunteers, aged 19-28 years, with an oral (75 g) glucose load. RESULTS: The 75 min insulin response (AUC(0,75 min)), up to tmax of glutamate kinetics, was significantly correlated with the AUC(0,75 min) of glutamate concentrations (r=0.485, P=0.049). Glucose tolerance was not affected. CONCLUSIONS: Oral (L)-glutamate enhances glucose-induced insulin secretion in healthy volunteers in a concentration-dependent manner.


Asunto(s)
Hipoglucemiantes/farmacología , Insulina/metabolismo , Glutamato de Sodio/farmacología , Administración Oral , Adulto , Área Bajo la Curva , Glucemia/análisis , Estudios Cruzados , Método Doble Ciego , Prueba de Tolerancia a la Glucosa , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/sangre , Secreción de Insulina , Glutamato de Sodio/efectos adversos , Glutamato de Sodio/sangre
20.
Br J Pharmacol ; 135(7): 1810-8, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11934823

RESUMEN

1. alpha-Endosulphine, isolated as an endogenous equivalent for sulphonylureas, is a 121-amino acids protein of 19 kDa apparent molecular mass, member of a cyclic AMP-regulated phosphoprotein family. We have previously shown that alpha-endosulphine inhibits sulphonylurea binding and K(ATP) channel activity, thereby stimulating basal insulin secretion. 2. We now describe that in the perfused rat pancreas, no stimulation was detected and that alpha-endosulphine inhibited glucose stimulated insulin release. This inhibition was dose-dependent and affected both phases of insulin secretion. 3. This inhibitory effect of alpha-endosulphine also occurred on MIN6 beta-cells when insulin release was stimulated either by glucose, sulphonylureas or a high K(+) depolarization. Inhibition was concentration-dependent with a half-maximal inhibition at 0.5 microM and was mirrored by inhibition of calcium influx. 4. Electrophysiological experiments demonstrated, in comparison to the effects of the sulphonylurea tolbutamide, that these inhibitory effects were linked to a direct inhibition of L-type Ca(2+)-channels and were independent from a regulation of K(ATP) channels. 5. Although alpha-endosulphine is able to stimulate insulin release under specific conditions acting via modulation of K(ATP) channel activity, the present study suggests that, under physiological conditions, the peptide mainly acts to block voltage-gated Ca(2+)-channels. This block leads to the inhibition of calcium influx and triggers inhibition of insulin release. 6. We conclude that alpha-endosulphine is not exclusively an endogenous equivalent for sulphonylureas and not solely a K(ATP) channel regulator.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Proteínas de Drosophila , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Páncreas/efectos de los fármacos , Péptidos/farmacología , Animales , Calcio/metabolismo , Canales de Calcio/efectos de los fármacos , Canales de Calcio/fisiología , Electrofisiología , Péptidos y Proteínas de Señalización Intercelular , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Modelos Animales , Páncreas/metabolismo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...