Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 136(2): 235-48, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-19135240

RESUMEN

Dysfunction and loss of insulin-producing pancreatic beta cells represent hallmarks of diabetes mellitus. Here, we show that mice lacking the mitogen-activated protein kinase (MAPK) p38delta display improved glucose tolerance due to enhanced insulin secretion from pancreatic beta cells. Deletion of p38delta results in pronounced activation of protein kinase D (PKD), the latter of which we have identified as a pivotal regulator of stimulated insulin exocytosis. p38delta catalyzes an inhibitory phosphorylation of PKD1, thereby attenuating stimulated insulin secretion. In addition, p38delta null mice are protected against high-fat-feeding-induced insulin resistance and oxidative stress-mediated beta cell failure. Inhibition of PKD1 reverses enhanced insulin secretion from p38delta-deficient islets and glucose tolerance in p38delta null mice as well as their susceptibility to oxidative stress. In conclusion, the p38delta-PKD pathway integrates regulation of the insulin secretory capacity and survival of pancreatic beta cells, pointing to a pivotal role for this pathway in the development of overt diabetes mellitus.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Animales , Exocitosis , Femenino , Glucosa/metabolismo , Aparato de Golgi/metabolismo , Secreción de Insulina , Masculino , Ratones , Proteína Quinasa 13 Activada por Mitógenos/genética , Fosfolipasas de Tipo C/metabolismo
2.
Nat Immunol ; 12(8): 761-9, 2011 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-21706006

RESUMEN

The migration of neutrophils into inflamed tissues is a fundamental component of innate immunity. A decisive step in this process is the polarized migration of blood neutrophils through endothelial cells (ECs) lining the venular lumen (transendothelial migration (TEM)) in a luminal-to-abluminal direction. By real-time confocal imaging, we found that neutrophils had disrupted polarized TEM ('hesitant' and 'reverse') in vivo. We noted these events in inflammation after ischemia-reperfusion injury, characterized by lower expression of junctional adhesion molecule C (JAM-C) at EC junctions, and they were enhanced by blockade or genetic deletion of JAM-C in ECs. Our results identify JAM-C as a key regulator of polarized neutrophil TEM in vivo and suggest that reverse TEM of neutrophils can contribute to the dissemination of systemic inflammation.


Asunto(s)
Moléculas de Adhesión Celular/inmunología , Endotelio Vascular/inmunología , Inmunoglobulinas/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Migración Transendotelial y Transepitelial/inmunología , Animales , Endotelio Vascular/citología , Endotelio Vascular/patología , Procesamiento de Imagen Asistido por Computador , Inflamación/patología , Ratones , Microscopía Confocal , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología
3.
Diabetologia ; 58(8): 1827-35, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26037202

RESUMEN

AIMS/HYPOTHESIS: Exposure of pancreatic beta cells to cytokines released by islet-infiltrating immune cells induces alterations in gene expression, leading to impaired insulin secretion and apoptosis in the initial phases of type 1 diabetes. Long non-coding RNAs (lncRNAs) are a new class of transcripts participating in the development of many diseases. As little is known about their role in insulin-secreting cells, this study aimed to evaluate their contribution to beta cell dysfunction. METHODS: The expression of lncRNAs was determined by microarray in the MIN6 beta cell line exposed to proinflammatory cytokines. The changes induced by cytokines were further assessed by real-time PCR in islets of control and NOD mice. The involvement of selected lncRNAs modified by cytokines was assessed after their overexpression in MIN6 cells and primary islet cells. RESULTS: MIN6 cells were found to express a large number of lncRNAs, many of which were modified by cytokine treatment. The changes in the level of selected lncRNAs were confirmed in mouse islets and an increase in these lncRNAs was also seen in prediabetic NOD mice. Overexpression of these lncRNAs in MIN6 and mouse islet cells, either alone or in combination with cytokines, favoured beta cell apoptosis without affecting insulin production or secretion. Furthermore, overexpression of lncRNA-1 promoted nuclear translocation of nuclear factor of κ light polypeptide gene enhancer in B cells 1 (NF-κB). CONCLUSIONS/INTERPRETATION: Our study shows that lncRNAs are modulated during the development of type 1 diabetes in NOD mice, and that their overexpression sensitises beta cells to apoptosis, probably contributing to their failure during the initial phases of the disease.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Estado Prediabético/metabolismo , ARN Largo no Codificante , Animales , Línea Celular , Diabetes Mellitus Tipo 1/patología , Progresión de la Enfermedad , Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos NOD , Estado Prediabético/patología
4.
Hum Mol Genet ; 19(2): 262-75, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19864490

RESUMEN

Targeting of numerous transmembrane proteins to the cell surface is thought to depend on their recognition by cargo receptors that interact with the adaptor machinery for anterograde traffic at the distal end of the Golgi complex. We report here on consortin, a novel integral membrane protein that is predicted to be intrinsically disordered, i.e. that contains large segments whose native state is unstructured. We identified consortin as a binding partner of connexins, the building blocks of gap junctions. Consortin is located at the trans-Golgi network (TGN), in tubulovesicular transport organelles, and at the plasma membrane. It directly interacts with the TGN clathrin adaptors GGA1 and GGA2, and disruption of this interaction by expression of a consortin mutant lacking the acidic cluster-dileucine (DXXLL) GGA interaction motif causes an intracellular accumulation of several connexins. RNA interference-mediated silencing of consortin expression in HeLa cells blocks the cell surface targeting of these connexins, which accumulate intracellularly, whereas partial depletion and redistribution of the consortin pool slows down the intracellular degradation of gap junction plaques. Altogether, our results show that, by studying connexin trafficking, we have identified the first TGN cargo receptor for the targeting of transmembrane proteins to the plasma membrane. The identification of consortin provides in addition a potential target for therapies aimed at diseases in which connexin traffic is altered, including cardiac ischemia, peripheral neuropathies, cataracts and hearing impairment. Sequence accession numbers. GenBank: Human CNST cDNA, NM_152609; mouse Cnst cDNA, NM_146105.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Conexinas/metabolismo , Proteínas de la Membrana/metabolismo , Red trans-Golgi/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas Portadoras/genética , Membrana Celular/genética , Conexinas/genética , Células HeLa , Humanos , Proteínas de la Membrana/genética , Ratones , Unión Proteica , Transporte de Proteínas , Red trans-Golgi/genética
5.
Hum Mol Genet ; 18(3): 428-39, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19000992

RESUMEN

Previous studies have documented that the insulin-producing beta-cells of laboratory rodents are coupled by gap junction channels made solely of the connexin36 (Cx36) protein, and have shown that loss of this protein desynchronizes beta-cells, leading to secretory defects reminiscent of those observed in type 2 diabetes. Since human islets differ in several respects from those of laboratory rodents, we have now screened human pancreas, and islets isolated thereof, for expression of a variety of connexin genes, tested whether the cognate proteins form functional channels for islet cell exchanges, and assessed whether this expression changes with beta-cell function in islets of control and type 2 diabetics. Here, we show that (i) different connexin isoforms are differentially distributed in the exocrine and endocrine parts of the human pancreas; (ii) human islets express at the transcript level different connexin isoforms; (iii) the membrane of beta-cells harbors detectable levels of gap junctions made of Cx36; (iv) this protein is concentrated in lipid raft domains of the beta-cell membrane where it forms gap junctions; (v) Cx36 channels allow for the preferential exchange of cationic molecules between human beta-cells; (vi) the levels of Cx36 mRNA correlated with the expression of the insulin gene in the islets of both control and type 2 diabetics. The data show that Cx36 is a native protein of human pancreatic islets, which mediates the coupling of the insulin-producing beta-cells, and contributes to control beta-cell function by modulating gene expression.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Conexinas/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Uniones Comunicantes/genética , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína delta-6 de Union Comunicante
6.
Pediatr Res ; 70(2): 142-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21527868

RESUMEN

Diabetes develops when the insulin needs of peripheral cells exceed the availability or action of the hormone. This situation results from the death of most beta-cells in type 1 diabetes, and from an inability of the beta-cell mass to adapt to increasing insulin needs in type 2 and gestational diabetes. We analyzed several lines of transgenic mice and showed that connexins (Cxs), the transmembrane proteins that form gap junctions, are implicated in the modulation of the beta-cell mass. Specifically, we found that the native Cx36 does not alter islet size or insulin content, whereas the Cx43 isoform increases both parameters, and Cx32 has a similar effect only when combined with GH. These findings open interesting perspectives for the in vitro and in vivo regulation of the beta-cell mass.


Asunto(s)
Tamaño de la Célula , Conexina 43/metabolismo , Conexinas/metabolismo , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Conexina 43/genética , Conexinas/genética , Cruzamientos Genéticos , Técnica del Anticuerpo Fluorescente , Hormona del Crecimiento/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Radioinmunoensayo , Estadísticas no Paramétricas , Proteína beta1 de Unión Comunicante , Proteína delta-6 de Union Comunicante
7.
J Biol Chem ; 284(37): 25004-14, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19584051

RESUMEN

The SLC25 carrier family mediates solute transport across the inner mitochondrial membrane, a process that is still poorly characterized regarding both the mechanisms and proteins implicated. This study investigated mitochondrial glutamate carrier GC1 in insulin-secreting beta-cells. GC1 was cloned from insulin-secreting cells, and sequence analysis revealed hydropathy profile of a six-transmembrane protein, characteristic of mitochondrial solute carriers. GC1 was found to be expressed at the mRNA and protein levels in INS-1E beta-cells and pancreatic rat islets. Immunohistochemistry showed that GC1 was present in mitochondria, and ultrastructural analysis by electron microscopy revealed inner mitochondrial membrane localization of the transporter. Silencing of GC1 in INS-1E beta-cells, mediated by adenoviral delivery of short hairpin RNA, reduced mitochondrial glutamate transport by 48% (p < 0.001). Insulin secretion at basal 2.5 mM glucose and stimulated either by intermediate 7.5 mM glucose or non-nutrient 30 mM KCl was not modified by GC1 silencing. Conversely, insulin secretion stimulated with optimal 15 mM glucose was reduced by 23% (p < 0.005) in GC1 knocked down cells compared with controls. Adjunct of cell-permeant glutamate (5 mM dimethyl glutamate) fully restored the secretory response at 15 mM glucose (p < 0.005). Kinetics of insulin secretion were investigated in perifused isolated rat islets. GC1 silencing in islets inhibited the secretory response induced by 16.7 mM glucose, both during first (-25%, p < 0.05) and second (-33%, p < 0.05) phases. This study demonstrates that insulin-secreting cells depend on GC1 for maximal glucose response, thereby assigning a physiological function to this newly identified mitochondrial glutamate carrier.


Asunto(s)
Ácido Glutámico/metabolismo , Insulina/metabolismo , Proteínas de Transporte de Membrana/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología , Animales , Línea Celular , Silenciador del Gen , Glucosa/metabolismo , Humanos , Inmunohistoquímica , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Cinética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , ARN Interferente Pequeño/metabolismo , Ratas
8.
Biochim Biophys Acta ; 1719(1-2): 82-101, 2005 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-16359942

RESUMEN

Glands were the first type of tissues in which the permissive role of gap junctions in the cell-to-cell transfer of membrane-impermeant molecules was shown. During the 40 years that have followed this seminal finding, gap junctions have been documented in all types of multicellular secretory systems, whether of the exocrine, endocrine or pheromonal nature. Also, compelling evidence now indicates that gap junction-mediated coupling, and/or the connexin proteins per se, play significant regulatory roles in various aspects of gland functions, ranging from the biosynthesis, storage and release of a variety of secretory products, to the control of the growth and differentiation of secretory cells, and to the regulation of gland morphogenesis. This review summarizes this evidence in the light of recent reports.


Asunto(s)
Comunicación Celular , Conexinas/fisiología , Glándulas Endocrinas/metabolismo , Glándulas Exocrinas/metabolismo , Uniones Comunicantes/fisiología , Animales , Calcio/metabolismo , Membrana Celular/metabolismo , Conexinas/metabolismo , Humanos , Modelos Biológicos , Isoformas de Proteínas , Transducción de Señal , Distribución Tisular , Transgenes
9.
Diabetes ; 54(6): 1798-807, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15919802

RESUMEN

Normal insulin secretion requires the coordinated functioning of beta-cells within pancreatic islets. This coordination depends on a communications network that involves the interaction of beta-cells with extracellular signals and neighboring cells. In particular, adjacent beta-cells are coupled via channels made of connexin36 (Cx36). To assess the function of this protein, we investigated islets of transgenic mice in which the Cx36 gene was disrupted by homologous recombination. We observed that compared with wild-type and heterozygous littermates that expressed Cx36 and behaved as nontransgenic controls, mice homozygous for the Cx36 deletion (Cx36(-/-)) featured beta-cells devoid of gap junctions and failing to exchange microinjected Lucifer yellow. During glucose stimulation, islets of Cx36(-/-) mice did not display the regular oscillations of intracellular calcium concentrations ([Ca(2+)](i)) seen in controls due to the loss of cell-to-cell synchronization of [Ca(2+)](i) changes. The same islets did not release insulin in a pulsatile fashion, even though the overall output of the hormone in response to glucose stimulation was normal. However, under nonstimulatory conditions, islets lacking Cx36 showed increased basal release of insulin. These data show that Cx36-dependent signaling is essential for the proper functioning of beta-cells, particularly for the pulsatility of [Ca(2+)](i) and insulin secretion during glucose stimulation.


Asunto(s)
Calcio/metabolismo , Conexinas/fisiología , Glucosa/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Conexinas/genética , Femenino , Heterocigoto , Homocigoto , Masculino , Ratones , Ratones Noqueados , Proteína delta-6 de Union Comunicante
10.
Arch Physiol Biochem ; 112(2): 74-81, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16931449

RESUMEN

Most cell types are functionally coupled by connexin (Cx) channels, i.e. exchange cytoplasmic ions and small metabolites through gap junction domains of their membrane. This form of direct cell-to-cell communication occurs in all existing animals, whatever their position in the phylogenetic scale, and up to humans. Pancreatic beta-cells are no exception, and normally cross-talk with their neighbors via channels made of Cx36. These exchanges importantly contribute to coordinate and synchronize the function of individual cells within pancreatic islets, particularly in the context of glucose-induced insulin secretion. Compelling evidence now indicates that Cx36-mediated coupling, and/or the Cx36 protein per se, play significant regulatory roles in various beta-cell functions, ranging from the biosynthesis, storage and release of insulin. Recent preliminary data further suggest that the protein may also be implicated in the balance of beta-cell growth versus necrosis and apoptosis, and in the regulated expression of specific genes. Here, we review this evidence, discuss the possible involvement of Cx36 in the pathophysiology of diabetes, and evaluate the relevance of this connexin in the therapeutic approaches to the disease.


Asunto(s)
Conexinas/fisiología , Células Secretoras de Insulina/metabolismo , Animales , Conexinas/genética , Humanos , Proteína delta-6 de Union Comunicante
11.
Hum Mutat ; 25(6): 543-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15880785

RESUMEN

Mutations in claudin 14 (CLDN14) cause nonsyndromic DFNB29 deafness in humans. The analysis of a murine model indicated that this phenotype is associated with degeneration of hair cells, possibly due to cation overload. However, the mechanism linking these alterations to CLDN14 mutations is unknown. To investigate this mechanism, we compared the ability of wild-type and missense mutant CLDN14 to form tight junctions. Ectopic expression in L mouse fibroblasts (LM cells) of wild-type CLDN14 protein induced the formation of tight junctions, while both the c.254T>A (p.V85D) mutant, previously identified in a Pakistani family, and the c.301 G>A (p.G101R) mutant, identified in this study through the screen of 183 Spanish and Greek patients affected with sporadic nonsyndromic deafness, failed to form such junctions. However, the two mutant proteins differed in their ability to localize at the plasma membrane. We further identified hitherto undescribed exons of CLDN14 that are utilized in alternative spliced transcripts. We demonstrated that different mutations of CLDN14 impaired by different mechanisms the ability of the protein to form tight junctions. Our results indicate that the ability of CLDN14 to be recruited to these junctions is crucial for the hearing process.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación/genética , Uniones Estrechas/metabolismo , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Claudinas , Perros , Exones/genética , Fibroblastos/citología , Humanos , Intrones/genética , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Uniones Estrechas/química
12.
J Mol Endocrinol ; 51(1): 49-58, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23613279

RESUMEN

Channels formed by the gap junction protein Connexin36 (CX36) contribute to the proper control of insulin secretion. We previously demonstrated that chronic exposure to glucose decreases Cx36 levels in insulin-secreting cells in vitro. Here, we investigated whether hyperglycemia also regulates Cx36 in vivo. Using a model of continuous glucose infusion in adult rats, we showed that prolonged (24-48 h) hyperglycemia reduced the Cx36 gene Gjd2 mRNA levels in pancreatic islets. Accordingly, prolonged exposure to high glucose concentrations also reduced the expression and function of Cx36 in the rat insulin-producing INS-1E cell line. The glucose effect was blocked after inhibition of the cAMP/PKA pathway and was associated with an overexpression of the inducible cAMP early repressor ICER-1/ICER-1γ, which binds to a functional cAMP-response element in the promoter of the Cx36 gene Gjd2. The involvement of this repressor was further demonstrated using an antisense strategy of ICER-1 inhibition, which prevented glucose-induced downregulation of Cx36. The data indicate that chronic exposure to glucose alters the in vivo expression of Cx36 by the insulin-producing ß-cells through ICER-1/ICER-1γ overexpression. This mechanism may contribute to the reduced glucose sensitivity and altered insulin secretion, which contribute to the pathophysiology of diabetes.


Asunto(s)
Conexinas/genética , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Regulación de la Expresión Génica , Hiperglucemia/genética , Hiperglucemia/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Glucemia , Línea Celular Tumoral , Conexinas/metabolismo , Modulador del Elemento de Respuesta al AMP Cíclico/metabolismo , Glucosa/metabolismo , Masculino , Ratas , Proteína delta-6 de Union Comunicante
13.
Diabetes ; 61(7): 1742-51, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22537941

RESUMEN

During the initial phases of type 1 diabetes, pancreatic islets are invaded by immune cells, exposing ß-cells to proinflammatory cytokines. This unfavorable environment results in gene expression modifications leading to loss of ß-cell functions. To study the contribution of microRNAs (miRNAs) in this process, we used microarray analysis to search for changes in miRNA expression in prediabetic NOD mice islets. We found that the levels of miR-29a/b/c increased in islets of NOD mice during the phases preceding diabetes manifestation and in isolated mouse and human islets exposed to proinflammatory cytokines. Overexpression of miR-29a/b/c in MIN6 and dissociated islet cells led to impairment in glucose-induced insulin secretion. Defective insulin release was associated with diminished expression of the transcription factor Onecut2, and a consequent rise of granuphilin, an inhibitor of ß-cell exocytosis. Overexpression of miR-29a/b/c also promoted apoptosis by decreasing the level of the antiapoptotic protein Mcl1. Indeed, a decoy molecule selectively masking the miR-29 binding site on Mcl1 mRNA protected insulin-secreting cells from apoptosis triggered by miR-29 or cytokines. Taken together, our findings suggest that changes in the level of miR-29 family members contribute to cytokine-mediated ß-cell dysfunction occurring during the initial phases of type 1 diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , MicroARNs/biosíntesis , Estado Prediabético/metabolismo , Animales , Apoptosis/efectos de los fármacos , Citocinas/farmacología , Diabetes Mellitus Tipo 1/metabolismo , Exocitosis/efectos de los fármacos , Femenino , Glucosa/administración & dosificación , Proteínas de Homeodominio/biosíntesis , Humanos , Insulina/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Factores de Transcripción/biosíntesis , Proteínas de Transporte Vesicular/análisis
14.
PLoS One ; 7(9): e45844, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029270

RESUMEN

The absence of the transcriptional repressor RE-1 Silencing Transcription Factor (REST) in insulin-secreting beta cells is a major cue for the specific expression of a large number of genes. These REST target genes were largely ascribed to a function of neurotransmission in a neuronal context, whereas their role in pancreatic beta cells has been poorly explored. To identify their functional significance, we have generated transgenic mice expressing REST in beta cells (RIP-REST mice), and previously discovered that REST target genes are essential to insulin exocytosis. Herein we characterized a novel line of RIP-REST mice featuring diabetes. In diabetic RIP-REST mice, high levels of REST were associated with postnatal beta cell apoptosis, which resulted in gradual beta cell loss and sustained hyperglycemia in adults. Moreover, adenoviral REST transduction in INS-1E cells led to increased cell death under control conditions, and sensitized cells to death induced by cytokines. Screening for REST target genes identified several anti-apoptotic genes bearing the binding motif RE-1 that were downregulated upon REST expression in INS-1E cells, including Gjd2, Mapk8ip1, Irs2, Ptprn, and Cdk5r2. Decreased levels of Cdk5r2 in beta cells of RIP-REST mice further confirmed that it is controlled by REST, in vivo. Using siRNA-mediated knock-down in INS-1E cells, we showed that Cdk5r2 protects beta cells against cytokines and palmitate-induced apoptosis. Together, these data document that a set of REST target genes, including Cdk5r2, is important for beta cell survival.


Asunto(s)
Proteínas Portadoras/genética , Supervivencia Celular , Regulación de la Expresión Génica , Células Secretoras de Insulina/fisiología , Proteínas Represoras/fisiología , Animales , Apoptosis , Secuencia de Bases , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Secuencia de Consenso , Proteínas del Citoesqueleto , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Homeostasis , Hiperglucemia/metabolismo , Hiperglucemia/patología , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Proteínas Ligadas a Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Páncreas/metabolismo , Páncreas/patología , Interferencia de ARN , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Elementos de Respuesta
15.
J Clin Invest ; 121(12): 4870-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22056383

RESUMEN

Type 1 diabetes develops when most insulin-producing ß cells of the pancreas are killed by an autoimmune attack. The in vivo conditions modulating the sensitivity and resistance of ß cells to this attack remain largely obscure. Here, we show that connexin 36 (Cx36), a trans-membrane protein that forms gap junctions between ß cells in the pancreatic islets, protects mouse ß cells against both cytotoxic drugs and cytokines that prevail in the islet environment at the onset of type 1 diabetes. We documented that this protection was at least partially dependent on intercellular communication, which Cx36 and other types of connexin channels establish within pancreatic islets. We further found that proinflammatory cytokines decreased expression of Cx36 and that experimental reduction or augmentation of Cx36 levels increased or decreased ß cell apoptosis, respectively. Thus, we conclude that Cx36 is central to ß cell protection from toxic insults.


Asunto(s)
Conexinas/fisiología , Diabetes Mellitus Experimental/prevención & control , Islotes Pancreáticos/patología , Aloxano/farmacología , Aloxano/toxicidad , Animales , Apoptosis/efectos de los fármacos , Comunicación Celular , Microambiente Celular , Conexinas/antagonistas & inhibidores , Conexinas/deficiencia , Conexinas/genética , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Uniones Comunicantes/fisiología , Dosificación de Gen , Insulina/genética , Interferón gamma/toxicidad , Interleucina-1beta/toxicidad , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico/biosíntesis , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Proteínas Recombinantes de Fusión/fisiología , Estreptozocina/farmacología , Estreptozocina/toxicidad , Factor de Necrosis Tumoral alfa/toxicidad , Proteína delta-6 de Union Comunicante
16.
PLoS One ; 6(9): e25178, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949881

RESUMEN

Imbalance of the excitatory neurotransmitter glutamate and of the inhibitory neurotransmitter GABA is one of several causes of seizures. ATP has also been implicated in epilepsy. However, little is known about the mechanisms involved in the release of ATP from cells and the consequences of the altered ATP signaling during seizures. Pannexin1 (Panx1) is found in astrocytes and in neurons at high levels in the embryonic and young postnatal brain, declining in adulthood. Panx1 forms large-conductance voltage sensitive plasma membrane channels permeable to ATP that are also activated by elevated extracellular K(+) and following P2 receptor stimulation. Based on these properties, we hypothesized that Panx1 channels may contribute to seizures by increasing the levels of extracellular ATP. Using pharmacological tools and two transgenic mice deficient for Panx1 we show here that interference with Panx1 ameliorates the outcome and shortens the duration of kainic acid-induced status epilepticus. These data thus indicate that the activation of Panx1 in juvenile mouse hippocampi contributes to neuronal hyperactivity in seizures.


Asunto(s)
Conducta Animal/efectos de los fármacos , Conexinas/fisiología , Epilepsia/prevención & control , Proteínas del Tejido Nervioso/fisiología , Convulsiones/prevención & control , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Western Blotting , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Epilepsia/inducido químicamente , Epilepsia/metabolismo , Técnica del Anticuerpo Fluorescente , Hipocampo/citología , Hipocampo/metabolismo , Ácido Kaínico/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Potasio/metabolismo , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Estado Epiléptico/metabolismo
17.
Stem Cell Res ; 2(1): 29-40, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19383407

RESUMEN

We have previously described the derivation of insulin-producing cell lines from mouse embryonic stem cells (mESCs) by differentiation of an intermediate lineage-restricted E-RoSH cell line through nutrient depletion in the presence of nicotinamide followed by limiting dilution. Here we investigated whether insulin-producing cell lines could be similarly derived directly from mouse embryo cells or tissues. Using a similar approach, we generated the RoSH2.K and MEPI-1 to -14 insulin-producing cell lines from the 5.5-dpc embryo-derived E-RoSH-analogous RoSH2 cell line and a 6.0-dpc mouse embryo culture, respectively. Insulin content was approximately 8 microg/10(6) MEPI-1 cells and 0.5 microg/10(6) RoSH2.K cells. Like insulin-producing mESC-derived ERoSHK cell lines, both MEPI and RoSH2.K lines were amenable to repeated cycles of freeze and thaw, replicated for months with a doubling time of 3-4 days, and exhibited genomic, structural, biochemical, and pharmacological properties of pancreatic beta-cells, including storage and release of insulin and C-peptide in an equimolar ratio. Transplantation of these cells also reversed hyperglycemia in streptozotocin-treated SCID mice and did not induce teratoma. Like ERoSHK cells, both RoSH2.K and MEPI-1 cells also induced hypoglycemia in the mice. Therefore, our protocol is robust and could reproducibly generate insulin-producing cell lines from different embryonic cell sources.


Asunto(s)
Trasplante de Células , Hiperglucemia/terapia , Células Secretoras de Insulina/citología , Animales , Péptido C/biosíntesis , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Embrión de Mamíferos , Hiperglucemia/inducido químicamente , Insulina/análisis , Insulina/biosíntesis , Células Secretoras de Insulina/trasplante , Ratones , Ratones SCID , Estreptozocina , Resultado del Tratamiento
18.
Stem Cell Res ; 2(1): 41-55, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19383408

RESUMEN

Generating surrogate insulin-producing cells from embryonic stem cells (ESCs) through in vitro replication of successive steps during pancreatic development has been challenging . Here we describe a novel reproducible protocol to establish homogeneous and scalable insulin-producing cell lines from mouse (m) ESCs via differentiation of the previously described lineage-restricted clonal mESC-derived E-RoSH cells. Unlike their parental mESCs, E-RoSH cells expressed high levels of mesodermal and endodermal genes. Nutrient depletion in the presence of nicotinamide inhibited proliferation of E-RoSH cells and induced differentiation into heterogeneous cultures comprising vascular-like structures that produced detectable levels of insulin and C-peptide in an equimolar ratio. Limiting dilution of these cultures resulted in the isolation of eight independent insulin-producing cell lines in five experiments. All these lines were cloned and shown to be amenable to repeated cycles of freeze and thaw and to replicate for months with a doubling time of 3-4 days. Under such conditions, the cultured cells exhibited genomic, structural, biochemical, and pharmacological properties of pancreatic beta cells, including storage of an equimolar ratio of insulin and C-peptide in granules and release of the contents of these organelles through a glucose-sensitive machinery. After transplantation, these cells reversed hyperglycemia in streptozotocin-treated SCID mice and did not form teratomas.


Asunto(s)
Células Madre Embrionarias/citología , Células Secretoras de Insulina/citología , Animales , Péptido C/análisis , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Trasplante de Células , Endodermo , Hiperglucemia/terapia , Insulina/análisis , Mesodermo , Ratones , Ratones SCID , Resultado del Tratamiento
19.
Exp Cell Res ; 314(5): 997-1012, 2008 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-18258229

RESUMEN

Connexin36 (Cx36) is the main connexin isoform expressed in neurons of the central nervous system (CNS) and in pancreatic beta-cells, i.e. two types of excitable cells that share - in spite of their different origins - a number of common features. Previous studies on Cx36 deficient mice have documented that loss of Cx36 resulted in phenotypic abnormalities in both the CNS and the pancreas which, however, could not be attributed to specific cell types due to the general deletion nature of the animal model used. Attempts to address this limitation using cell type specific deletions generated by the Cre/loxP strategy have so far been complicated by the lack of Cx36 expression from the floxed allele. We have now generated a conditional Cx36 deficient mouse mutant in which the coding region of Cx36 is flanked by loxP sites, followed by a cyan fluorescent protein (CFP) reporter gene. Here we show that Cx36 was still expressed from the floxed allele in neurons and pancreatic beta-cells. In these cells, a 30-60% decrease of this protein, relative to the expression level of the wildtype allele, did not significantly perturb cell coupling. The deletion of Cx36 by ubiquitously and cell type specifically expressed Cre recombinases revealed that CFP functions as a reliable reporter for Cx36 expression in brain neurons and to some extent in retina neurons, but not in pancreas. Loss of Cx36 by Cre-mediated recombination was documented at transcript and protein levels. Cell type specific deletion of Cx36 in the endocrine pancreas revealed major alterations in the basal as well as the glucose-induced insulin secretion, hence specifically attributing to pancreatic Cx36 an important regulatory role in the control of beta-cell function. Cell type specific deletion of Cx36 in the CNS by suitable Cre recombinases should also help to elucidate the functional role of Cx36 in different neuronal subtypes.


Asunto(s)
Conexinas/genética , Conexinas/fisiología , Células Secretoras de Insulina/química , Neuronas/química , Animales , Encéfalo/citología , Conexinas/deficiencia , Regulación de la Expresión Génica , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Integrasas , Ratones , Retina/citología , Distribución Tisular , Proteína delta-6 de Union Comunicante
20.
Diabetes ; 57(5): 1176-85, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18420492

RESUMEN

OBJECTIVE: Emerging evidence suggests that dietary phytoestrogens can have beneficial effects on obesity and diabetes, although their mode of action is not known. Here, we investigate the mechanisms mediating the action of dietary phytoestrogens on lipid and glucose metabolism in rodents. RESEARCH DESIGN AND METHODS: Male CD-1 mice were fed from conception to adulthood with either a high soy-containing diet or a soy-free diet. Serum levels of circulating isoflavones, ghrelin, leptin, free fatty acids, triglycerides, and cholesterol were quantified. Tissue samples were analyzed by quantitative RT-PCR and Western blotting to investigate changes of gene expression and phosphorylation state of key metabolic proteins. Glucose and insulin tolerance tests and euglycemic-hyperinsulinemic clamp were used to assess changes in insulin sensitivity and glucose uptake. In addition, insulin secretion was determined by in situ pancreas perfusion. RESULTS: In peripheral tissues of soy-fed mice, especially in white adipose tissue, phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase was increased, and expression of genes implicated in peroxisomal fatty acid oxidation and mitochondrial biogenesis was upregulated. Soy-fed mice also showed reduced serum insulin levels and pancreatic insulin content and improved insulin sensitivity due to increased glucose uptake into skeletal muscle. Thus, mice fed with a soy-rich diet have improved adipose and glucose metabolism. CONCLUSIONS: Dietary soy could prove useful to prevent obesity and associated disorders. Activation of the AMPK pathway by dietary soy is likely involved and may mediate the beneficial effects of dietary soy in peripheral tissues.


Asunto(s)
Glucemia/metabolismo , Dieta , Isoflavonas/sangre , Lípidos/sangre , Complejos Multienzimáticos/metabolismo , Fitoestrógenos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Alimentación Animal , Animales , Glucemia/efectos de los fármacos , Cruzamientos Genéticos , Activación Enzimática/efectos de los fármacos , Femenino , Insulina/sangre , Insulina/metabolismo , Masculino , Ratones , Páncreas/efectos de los fármacos , Páncreas/fisiología , Fitoestrógenos/administración & dosificación , Alimentos de Soja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA