Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 17: 1192464, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37377777

RESUMEN

Introduction: Clinical trials demonstrated that co-targeting angiopoietin-2 (Ang-2) and vascular endothelial growth factor (VEGF-A) with faricimab controls anatomic outcomes and maintains vision improvements, with strong durability, through 2 years in patients with neovascular age-related macular degeneration and diabetic macular edema. The mechanism(s) underlying these findings is incompletely understood and the specific role that Ang-2 inhibition plays requires further investigation. Methods: We examined the effects of single and dual Ang-2/VEGF-A inhibition in diseased vasculatures of JR5558 mice with spontaneous choroidal neovascularization (CNV) and in mice with retinal ischemia/reperfusion (I/R) injuries. Results: In JR5558 mice, Ang-2, VEGF-A, and dual Ang-2/VEGF-A inhibition reduced CNV area after 1 week; only dual Ang-2/VEGF-A inhibition decreased neovascular leakage. Only Ang-2 and dual Ang-2/VEGF-A inhibition maintained reductions after 5 weeks. Dual Ang-2/VEGF-A inhibition reduced macrophage/microglia accumulation around lesions after 1 week. Both Ang-2 and dual Ang-2/VEGF-A inhibition reduced macrophage/microglia accumulation around lesions after 5 weeks. In the retinal I/R injury model, dual Ang-2/VEGF-A inhibition was statistically significantly more effective than Ang-2 or VEGF-A inhibition alone in preventing retinal vascular leakage and neurodegeneration. Discussion: These data highlight the role of Ang-2 in dual Ang-2/VEGF-A inhibition and indicate that dual inhibition has complementary anti-inflammatory and neuroprotective effects, suggesting a mechanism for the durability and efficacy of faricimab in clinical trials.

2.
Proc Natl Acad Sci U S A ; 117(33): 19854-19865, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32759214

RESUMEN

The blood-retina barrier and blood-brain barrier (BRB/BBB) are selective and semipermeable and are critical for supporting and protecting central nervous system (CNS)-resident cells. Endothelial cells (ECs) within the BRB/BBB are tightly coupled, express high levels of Claudin-5 (CLDN5), a junctional protein that stabilizes ECs, and are important for proper neuronal function. To identify novel CLDN5 regulators (and ultimately EC stabilizers), we generated a CLDN5-P2A-GFP stable cell line from human pluripotent stem cells (hPSCs), directed their differentiation to ECs (CLDN5-GFP hPSC-ECs), and performed flow cytometry-based chemogenomic library screening to measure GFP expression as a surrogate reporter of barrier integrity. Using this approach, we identified 62 unique compounds that activated CLDN5-GFP. Among them were TGF-ß pathway inhibitors, including RepSox. When applied to hPSC-ECs, primary brain ECs, and retinal ECs, RepSox strongly elevated barrier resistance (transendothelial electrical resistance), reduced paracellular permeability (fluorescein isothiocyanate-dextran), and prevented vascular endothelial growth factor A (VEGFA)-induced barrier breakdown in vitro. RepSox also altered vascular patterning in the mouse retina during development when delivered exogenously. To determine the mechanism of action of RepSox, we performed kinome-, transcriptome-, and proteome-profiling and discovered that RepSox inhibited TGF-ß, VEGFA, and inflammatory gene networks. In addition, RepSox not only activated vascular-stabilizing and barrier-establishing Notch and Wnt pathways, but also induced expression of important tight junctions and transporters. Taken together, our data suggest that inhibiting multiple pathways by selected individual small molecules, such as RepSox, may be an effective strategy for the development of better BRB/BBB models and novel EC barrier-inducing therapeutics.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematorretinal/efectos de los fármacos , Barrera Hematorretinal/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular/efectos de los fármacos , Claudina-5/genética , Claudina-5/metabolismo , Evaluación Preclínica de Medicamentos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Edición Génica , Genoma , Humanos , Ratones , Ratones Noqueados , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Uniones Estrechas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Biochim Biophys Acta Mol Cell Res ; 1866(11): 118474, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30954571

RESUMEN

Discoidin domain receptor1 (DDR1) is a collagen activated receptor tyrosine kinase and an attractive anti-fibrotic target. Its expression is mainly limited to epithelial cells located in several organs including skin, kidney, liver and lung. DDR1's biology is elusive, with unknown downstream activation pathways; however, it may act as a mediator of the stromal-epithelial interaction, potentially controlling the activation state of the resident quiescent fibroblasts. Increased expression of DDR1 has been documented in several types of cancer and fibrotic conditions including skin hypertrophic scars, idiopathic pulmonary fibrosis, cirrhotic liver and renal fibrosis. The present review article focuses on: a) detailing the evidence for a role of DDR1 as an anti-fibrotic target in different organs, b) clarifying DDR1 tissue distribution in healthy and diseased tissues as well as c) exploring DDR1 protective mode of action based on literature evidence and co-authors experience; d) detailing pharmacological efforts attempted to drug this subtle anti-fibrotic target to date.


Asunto(s)
Receptor con Dominio Discoidina 1/efectos de los fármacos , Receptor con Dominio Discoidina 1/metabolismo , Fibrosis/metabolismo , Animales , Aterosclerosis/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/tratamiento farmacológico , Humanos , Riñón/metabolismo , Riñón/patología , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Ratones , Neoplasias/metabolismo , Nefritis Intersticial/patología , Células Plasmáticas , Proteínas Tirosina Quinasas Receptoras , Piel/metabolismo , Piel/patología , Enfermedades Vasculares/metabolismo , Cicatrización de Heridas
5.
ACS Chem Biol ; 14(1): 37-49, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30452219

RESUMEN

The importance of Discoidin Domain Receptor 1 (DDR1) in renal fibrosis has been shown via gene knockout and use of antisense oligonucleotides; however, these techniques act via a reduction of DDR1 protein, while we prove the therapeutic potential of inhibiting DDR1 phosphorylation with a small molecule. To date, efforts to generate a selective small-molecule to specifically modulate the activity of DDR1 in an in vivo model have been unsuccessful. We performed parallel DNA encoded library screens against DDR1 and DDR2, and discovered a chemical series that is highly selective for DDR1 over DDR2. Structure-guided optimization efforts yielded the potent DDR1 inhibitor 2.45, which possesses excellent kinome selectivity (including 64-fold selectivity over DDR2 in a biochemical assay), a clean in vitro safety profile, and favorable pharmacokinetic and physicochemical properties. As desired, compound 2.45 modulates DDR1 phosphorylation in vitro as well as prevents collagen-induced activation of renal epithelial cells expressing DDR1. Compound 2.45 preserves renal function and reduces tissue damage in Col4a3-/- mice (the preclinical mouse model of Alport syndrome) when employing a therapeutic dosing regime, indicating the real therapeutic value of selectively inhibiting DDR1 phosphorylation in vivo. Our results may have wider significance as Col4a3-/- mice also represent a model for chronic kidney disease, a disease which affects 10% of the global population.


Asunto(s)
ADN/genética , Receptor con Dominio Discoidina 1/antagonistas & inhibidores , Riñón/fisiopatología , Nefritis Hereditaria/genética , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Pruebas de Función Renal , Ratones , Ratones Noqueados , Nefritis Hereditaria/fisiopatología , Fosforilación , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo
6.
J Transl Med ; 16(1): 148, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859097

RESUMEN

BACKGROUND: Discoidin domain receptor 1 (DDR1) is a collagen-activated receptor tyrosine kinase extensively implicated in diseases such as cancer, atherosclerosis and fibrosis. Multiple preclinical studies, performed using either a gene deletion or a gene silencing approaches, have shown this receptor being a major driver target of fibrosis and glomerulosclerosis. METHODS: The present study investigated the role and relevance of DDR1 in human crescentic glomerulonephritis (GN). Detailed DDR1 expression was first characterized in detail in human GN biopsies using a novel selective anti-DDR1 antibody using immunohistochemistry. Subsequently the protective role of DDR1 was investigated using a highly selective, novel, small molecule inhibitor in a nephrotoxic serum (NTS) GN model in a prophylactic regime and in the NEP25 GN mouse model using a therapeutic intervention regime. RESULTS: DDR1 expression was shown to be mainly limited to renal epithelium. In humans, DDR1 is highly induced in injured podocytes, in bridging cells expressing both parietal epithelial cell (PEC) and podocyte markers and in a subset of PECs forming the cellular crescents in human GN. Pharmacological inhibition of DDR1 in NTS improved both renal function and histological parameters. These results, obtained using a prophylactic regime, were confirmed in the NEP25 GN mouse model using a therapeutic intervention regime. Gene expression analysis of NTS showed that pharmacological blockade of DDR1 specifically reverted fibrotic and inflammatory gene networks and modulated expression of the glomerular cell gene signature, further validating DDR1 as a major mediator of cell fate in podocytes and PECs. CONCLUSIONS: Together, these results suggest that DDR1 inhibition might be an attractive and promising pharmacological intervention for the treatment of GN, predominantly by targeting the renal epithelium.


Asunto(s)
Receptor con Dominio Discoidina 1/antagonistas & inhibidores , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Animales , Receptor con Dominio Discoidina 1/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/patología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Glomerulonefritis/genética , Glomerulonefritis/patología , Humanos , Inflamación/patología , Riñón/patología , Masculino , Ratones , Persona de Mediana Edad , Fenotipo
7.
Mol Metab ; 5(1): 47-56, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26844206

RESUMEN

OBJECTIVE: Type 2 diabetes and obesity are emerging pandemics in the 21st century creating worldwide urgency for the development of novel and safe therapies. We investigated trace amine-associated receptor 1 (TAAR1) as a novel target contributing to the control of glucose homeostasis and body weight. METHODS: We investigated the peripheral human tissue distribution of TAAR1 by immunohistochemistry and tested the effect of a small molecule TAAR1 agonist on insulin secretion in vitro using INS1E cells and human islets and on glucose tolerance in C57Bl6, and db/db mice. Body weight effects were investigated in obese DIO mice. RESULTS: TAAR1 activation by a selective small molecule agonist increased glucose-dependent insulin secretion in INS1E cells and human islets and elevated plasma PYY and GLP-1 levels in mice. In diabetic db/db mice, the TAAR1 agonist normalized glucose excursion during an oral glucose tolerance test. Sub-chronic treatment of diet-induced obese (DIO) mice with the TAAR1 agonist resulted in reduced food intake and body weight. Furthermore insulin sensitivity was improved and plasma triglyceride levels and liver triglyceride content were lower than in controls. CONCLUSIONS: We have identified TAAR1 as a novel integrator of metabolic control, which acts on gastrointestinal and pancreatic islet hormone secretion. Thus TAAR1 qualifies as a novel and promising target for the treatment of type 2 diabetes and obesity.

8.
Nat Med ; 21(1): 27-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25485909

RESUMEN

We report the discovery of a new monomeric peptide that reduces body weight and diabetic complications in rodent models of obesity by acting as an agonist at three key metabolically-related peptide hormone receptors: glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and glucagon receptors. This triple agonist demonstrates supraphysiological potency and equally aligned constituent activities at each receptor, all without cross-reactivity at other related receptors. Such balanced unimolecular triple agonism proved superior to any existing dual coagonists and best-in-class monoagonists to reduce body weight, enhance glycemic control and reverse hepatic steatosis in relevant rodent models. Various loss-of-function models, including genetic knockout, pharmacological blockade and selective chemical knockout, confirmed contributions of each constituent activity in vivo. We demonstrate that these individual constituent activities harmonize to govern the overall metabolic efficacy, which predominantly results from synergistic glucagon action to increase energy expenditure, GLP-1 action to reduce caloric intake and improve glucose control, and GIP action to potentiate the incretin effect and buffer against the diabetogenic effect of inherent glucagon activity. These preclinical studies suggest that, so far, this unimolecular, polypharmaceutical strategy has potential to be the most effective pharmacological approach to reversing obesity and related metabolic disorders.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Obesidad/metabolismo , Péptidos/administración & dosificación , Animales , Glucemia/efectos de los fármacos , Peso Corporal/genética , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/genética , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/metabolismo , Células HEK293 , Humanos , Insulina/biosíntesis , Insulina/metabolismo , Ratones , Obesidad/tratamiento farmacológico , Obesidad/genética , Péptidos/síntesis química , Péptidos/metabolismo , Ratas , Receptores de la Hormona Gastrointestinal/agonistas , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Roedores
9.
Sci Transl Med ; 5(209): 209ra151, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174327

RESUMEN

We report the discovery and translational therapeutic efficacy of a peptide with potent, balanced co-agonism at both of the receptors for the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). This unimolecular dual incretin is derived from an intermixed sequence of GLP-1 and GIP, and demonstrated enhanced antihyperglycemic and insulinotropic efficacy relative to selective GLP-1 agonists. Notably, this superior efficacy translated across rodent models of obesity and diabetes, including db/db mice and ZDF rats, to primates (cynomolgus monkeys and humans). Furthermore, this co-agonist exhibited synergism in reducing fat mass in obese rodents, whereas a selective GIP agonist demonstrated negligible weight-lowering efficacy. The unimolecular dual incretins corrected two causal mechanisms of diabesity, adiposity-induced insulin resistance and pancreatic insulin deficiency, more effectively than did selective mono-agonists. The duration of action of the unimolecular dual incretins was refined through site-specific lipidation or PEGylation to support less frequent administration. These peptides provide comparable pharmacology to the native peptides and enhanced efficacy relative to similarly modified selective GLP-1 agonists. The pharmacokinetic enhancement lessened peak drug exposure and, in combination with less dependence on GLP-1-mediated pharmacology, avoided the adverse gastrointestinal effects that typify selective GLP-1-based agonists. This discovery and validation of a balanced and high-potency dual incretin agonist enables a more physiological approach to management of diseases associated with impaired glucose tolerance.


Asunto(s)
Haplorrinos/metabolismo , Incretinas/farmacología , Roedores/metabolismo , Acilación/efectos de los fármacos , Adolescente , Adulto , Anciano , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Exenatida , Femenino , Polipéptido Inhibidor Gástrico/administración & dosificación , Polipéptido Inhibidor Gástrico/farmacología , Péptido 1 Similar al Glucagón/administración & dosificación , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/tratamiento farmacológico , Incretinas/administración & dosificación , Incretinas/uso terapéutico , Insulina/metabolismo , Liraglutida , Masculino , Ratones , Persona de Mediana Edad , Péptidos/farmacología , Ratas , Receptores de la Hormona Gastrointestinal , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Resultado del Tratamiento , Ponzoñas/farmacología , Pérdida de Peso/efectos de los fármacos , Adulto Joven
10.
FASEB J ; 24(6): 1824-37, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20061534

RESUMEN

Phosphatidylinositide 3-kinases (PI3Ks) play central roles in insulin signal transduction. While the contribution of class Ia PI3K members has been extensively studied, the role of class II members remains poorly understood. The diverse actions of class II PI3K-C2alpha have been attributed to its lipid product PI(3)P. By applying pharmacological inhibitors, transient overexpression and small-interfering RNA-based knockdown of PI3K and PKB/Akt isoforms, together with PI-lipid profiling and live-cell confocal and total internal reflection fluorescence microscopy, we now demonstrate that in response to insulin, PI3K-C2alpha generates PI(3,4)P(2), which allows the selective activation of PKBalpha/Akt1. Knockdown of PI3K-C2alpha expression and subsequent reduction of PKBalpha/Akt1 activity in the pancreatic beta-cell impaired glucose-stimulated insulin release, at least in part, due to reduced glucokinase expression and increased AS160 activity. Hence, our results identify signal transduction via PI3K-C2alpha as a novel pathway whereby insulin activates PKB/Akt and thus discloses PI3K-C2alpha as a potential drugable target in type 2 diabetes. The high degree of codistribution of PI3K-C2alpha and PKBalpha/Akt1 with insulin receptor B type, but not A type, in the same plasma membrane microdomains lends further support to the concept that selectivity in insulin signaling is achieved by the spatial segregation of signaling events.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Edulcorantes/farmacología , Androstadienos/farmacología , Animales , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Fosfatidilinositol 3-Quinasas Clase II , Técnica del Anticuerpo Fluorescente , Glucoquinasa/metabolismo , Inmunoprecipitación , Antagonistas de Insulina/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Lípidos , Ratones , Ratones Obesos , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Receptor de Insulina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Wortmanina
11.
FASEB J ; 21(7): 1609-21, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17264162

RESUMEN

Insulin exerts pleiotropic effects at the cellular level. Signaling via the two isoforms of the insulin receptor (IR) may explain the activation of different signaling cascades, while it remains to be explored how selectivity is achieved when utilizing the same IR isoform. We now demonstrate that insulin-stimulated transcription of c-fos and glucokinase genes is activated simultaneously in the insulin-producing beta-cell via IR-B localized in different cellular compartments. Insulin activates the glucokinase gene from plasma membrane-standing IR-B, while c-fos gene activation is dependent on clathrin-mediated IR-B-endocytosis and signaling from early endosomes. Moreover, glucokinase gene up-regulation requires the integrity of the juxtamembrane IR-B NPEY-motif and signaling via PI3K-C2alpha-like/PDK1/PKB, while c-fos gene activation requires the intact C-terminal YTHM-motif and signaling via PI3K Ia/Shc/MEK1/ERK. By using IR-B as an example it is thus possible to demonstrate how spatial segregation allows simultaneous and selective signaling via the same receptor isoform in the same cell.


Asunto(s)
Regulación de la Expresión Génica , Receptor de Insulina/metabolismo , Transducción de Señal , Secuencia de Bases , Línea Celular , Cartilla de ADN , Genes fos , Humanos , Islotes Pancreáticos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional
12.
J Biol Chem ; 280(16): 16272-7, 2005 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-15722354

RESUMEN

Clathrin-dependent endocytosis of Na(+),K(+)-ATPase molecules in response to G protein-coupled receptor signals is triggered by phosphorylation of the alpha-subunit and the binding of phosphoinositide 3-kinase. In this study, we describe a molecular mechanism linking phosphorylation of Na(+),K(+)-ATPase alpha-subunit to binding and activation of phosphoinositide 3-kinase. Co-immunoprecipitation studies, as well as experiments using confocal microscopy, revealed that dopamine favored the association of 14-3-3 protein with the basolateral plasma membrane and its co-localization with the Na(+),K(+)-ATPase alpha-subunit. The functional relevance of this interaction was established in opossum kidney cells expressing a 14-3-3 dominant negative mutant, where dopamine failed to decrease Na(+),K(+)-ATPase activity and to promote its endocytosis. The phosphorylated Ser-18 residue within the alpha-subunit N terminus is critical for 14-3-3 binding. Activation of phosphoinositide 3-kinase by dopamine during Na(+),K(+)-ATPase endocytosis requires the binding of the kinase to a proline-rich domain within the alpha-subunit, and this effect was blocked by the presence of a 14-3-3 dominant negative mutant. Thus, the 14-3-3 protein represents a critical linking mechanism for recruiting phosphoinositide 3-kinase to the site of Na(+),K(+)-ATPase endocytosis.


Asunto(s)
Proteínas 14-3-3/metabolismo , Endocitosis/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Proteínas 14-3-3/genética , Secuencias de Aminoácidos , Animales , Sitios de Unión , Técnicas de Transferencia de Gen , Fosforilación , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
13.
Cell ; 119(2): 273-84, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15479643

RESUMEN

An oscillatory increase in pancreatic beta cell cytoplasmic free Ca2+ concentration, [Ca2+]i, is a key feature in glucose-induced insulin release. The role of the voltage-gated Ca2+ channel beta3 subunit in the molecular regulation of these [Ca2+]i oscillations has now been clarified by using beta3 subunit-deficient beta cells. beta3 knockout mice showed a more efficient glucose homeostasis compared to wild-type mice due to increased glucose-stimulated insulin secretion. This resulted from an increased glucose-induced [Ca2+]i oscillation frequency in beta cells lacking the beta3 subunit, an effect accounted for by enhanced formation of inositol 1,4,5-trisphosphate (InsP3) and increased Ca2+ mobilization from intracellular stores. Hence, the beta3 subunit negatively modulated InsP3-induced Ca2+ release, which is not paralleled by any effect on the voltage-gated L type Ca2+ channel. Since the increase in insulin release was manifested only at high glucose concentrations, blocking the beta3 subunit in the beta cell may constitute the basis for a novel diabetes therapy.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Exocitosis/fisiología , Insulina/metabolismo , Subunidades de Proteína/metabolismo , Animales , Células COS , Canales de Calcio/genética , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Homeostasis , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Subunidades de Proteína/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Tapsigargina/farmacología
14.
Proc Natl Acad Sci U S A ; 101(27): 10090-4, 2004 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-15210953

RESUMEN

In type 1 diabetes (T1D), there is a specific destruction of the insulin secreting pancreatic beta cell. Although the exact molecular mechanisms underlying beta cell destruction are not known, sera from T1D patients have been shown to promote Ca(2+)-induced apoptosis. We now demonstrate that apolipoprotein CIII (apoCIII) is increased in serum from T1D patients and that this serum factor both induces increased cytoplasmic free intracellular Ca(2+) concentration ([Ca(2+)](i)) and beta cell death. The apoCIII-induced increase in [Ca(2+)](i) reflects an activation of the voltage-gated L-type Ca(2+) channel. Both the effects of T1D sera and apoCIII on the beta cell are abolished in the presence of antibody against apoCIII. Increased serum levels of apoCIII can thus account for the increase in beta cell [Ca(2+)](i) and thereby beta cell apoptosis associated with T1D.


Asunto(s)
Apolipoproteínas C/fisiología , Apoptosis , Calcio/fisiología , Diabetes Mellitus Tipo 1/patología , Islotes Pancreáticos/patología , Adulto , Animales , Apolipoproteína C-III , Canales de Calcio Tipo L/fisiología , Diabetes Mellitus Tipo 1/sangre , Femenino , Humanos , Masculino , Ratones
15.
J Biol Chem ; 279(23): 24152-62, 2004 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-15056668

RESUMEN

Interferon (IFN) alpha induces a caspase-dependent apoptosis that is associated with activation of the proapoptotic Bak and Bax, loss of mitochondrial membrane potential, and release of cytochrome c. In addition to the onset of the classical Jak-STAT pathway, IFNalpha also induced phosphoinositide 3-kinase (PI3K) activity. Pharmacological inhibition of PI3K activity by Ly294002 disrupted IFN-induced apoptosis upstream of mitochondria. Inhibition of mTOR by rapamycin or by overexpression of a kinase dead mutant of mTOR, efficiently blocked IFNalpha-induced apoptosis. A PI3K and mTOR-dependent phosphorylation of p70S6 kinase and 4E-BP1 repressor was induced by IFNalpha treatment of cells and was strongly inhibited by Ly294002 or rapamycin. The activation of Jak-STAT signaling upon IFNalpha stimulation was not affected by abrogating PI3K/mTOR pathway. Neither was the expression of several IFNalpha target genes affected, nor the ability of IFNalpha to protect against virus-induced cell death affected by inhibition of the PI3K/mTOR pathway. These data demonstrate that an intact PI3K/mTOR pathway is necessary for the ability of IFNalpha to induce apoptosis, whereas activation of the Jak-STAT pathway alone appears to be insufficient for this specific IFNalpha-induced effect.


Asunto(s)
Apoptosis , Interferón-alfa/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Anexina A5/farmacología , Antibióticos Antineoplásicos/farmacología , Western Blotting , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Proteínas de Ciclo Celular , Muerte Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cromonas/farmacología , Colorantes/farmacología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Interferón-alfa/metabolismo , Queratinocitos/metabolismo , Proteínas Luminiscentes/metabolismo , Potenciales de la Membrana , Mitocondrias/metabolismo , Morfolinas/farmacología , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Transfección
16.
J Cell Biol ; 163(6): 1327-37, 2003 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-14691140

RESUMEN

In pancreatic beta-cells, insulin selectively up-regulates the transcription of its own gene and that of the glucokinase gene by signaling through the two isoforms of the insulin receptor, i.e., A-type (Ex11-) and B-type (Ex11+), using different signaling pathways. However, the molecular mechanism(s) that allows the discrete activation of signaling cascades via the two receptor isoforms remains unclear. Here we show that activation of the insulin promoter via A-type and of the glucokinase promoter via B-type insulin receptor is not dependent on receptor isoform-specific differences in internalization but on the different localization of the receptor types in the plasma membrane. Our data demonstrate that localization and function of the two receptor types depend on the 12-amino acid string encoded by exon 11, which acts as a sorting signal rather than as a physical spacer. Moreover, our data suggest that selective activation of the insulin and glucokinase promoters occurs by signaling from noncaveolae lipid rafts that are differently sensitive toward cholesterol depletion.


Asunto(s)
Membrana Celular/metabolismo , Insulina/biosíntesis , Islotes Pancreáticos/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos/genética , Animales , Caveolinas/metabolismo , Línea Celular Tumoral , Membrana Celular/ultraestructura , Colesterol/deficiencia , Cricetinae , Exones/genética , Glucoquinasa/biosíntesis , Glucoquinasa/genética , Insulina/genética , Islotes Pancreáticos/ultraestructura , Microdominios de Membrana/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína/genética , Ratas , Ratas Wistar , Receptor de Insulina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...