Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Horm Metab Res ; 47(1): 56-63, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25350519

RESUMEN

Early postnatal life is a critical period for development of the endocrine pancreas, involving remodelling of islet cells and maturation of secretory responses. Factors that regulate these processes are undefined. Somatostatin-secreting delta-cells are abundant in the developing pancreas and, because somatostatin inhibits growth, the hormone may regulate islet expansion in early life. The aim of this study was to investigate effects of somatostatin-deficiency on proliferation, apoptosis and pancreas expansion in the first 3 weeks of life in mice. Pancreases from control or somatostatin-knockout mice were analysed for beta cell, alpha cell and pancreatic volumes by morphometry, proliferation by BrdU incorporation and apoptosis by TUNEL labelling. Signalling pathways associated with proliferation and apoptosis were studied by immunohistochemistry and Western blotting. Knockout mice grew normally in the first 3 weeks of life, but had high circulating insulin that normalised by day 21. Beta cell, alpha cell and pancreatic volumes were decreased in knockout mice, accompanied by reduced proliferation and increased apoptosis in the pancreas. Decreased growth was not due to impaired Akt signalling, as Akt phosphorylation and nuclear cyclin-D2 increased in the knockout pancreas. Levels of TGF-ß1, a factor implicated in tissue remodelling, together with SMAD phosphorylation through which TGF-ß mediates its effects, were increased in the knockout pancreas. Beta cell expansion was impaired in knockout mice, potentially compensating for increased insulin secretion from islets lacking inhibitory effects of somatostatin, and was associated with increased TGF-ß1 levels. TGF-ß1 may represent an important regulator of beta cell mass in early life.


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Transducción de Señal , Somatostatina/deficiencia , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Peso Corporal , Bromodesoxiuridina/metabolismo , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Fosforilación , Fosfoserina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Smad/metabolismo , Somatostatina/metabolismo
2.
Diabetes Obes Metab ; 16(10): 947-56, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24720683

RESUMEN

AIMS: Islets are innervated by parasympathetic nerves which release acetylcholine (ACh) to amplify glucose-induced insulin secretion, primarily via muscarinic M3 receptors (M3R). Here we investigate the consequence of chronic hyperglycaemia on islet M3R expression and secretory sensitivity of mouse islets to cholinergic receptor activation. METHODS: The impact of hyperglycaemia was studied in (i) islets isolated from ob/ob mice, (ii) alginate-encapsulated mouse islets transplanted intraperitoneally into streptozotocin-induced diabetic mice and (iii) mouse and human islets maintained in vitro at 5.5 or 16 mmol/l glucose. Blood glucose levels were assessed by a commercial glucose meter, insulin content by RIA and M3R expression by qPCR and immunohistochemistry. RESULTS: M3R mRNA expression was reduced in both ob/ob islets and islets maintained at 16 mmol/l glucose for 3 days (68 and 50% control, respectively). In all three models of hyperglycaemia the secretory sensitivity to the cholinergic receptor agonist, carbachol, was reduced by 60-70% compared to control islets. Treatment for 72 h with the irreversible PKC activator, PMA, or the PKC inhibitor, Gö6983, did not alter islet M3R mRNA expression nor did incubation with the PI3K-inhibitor, LY294002, although enhancement of glucose-induced insulin secretion by LY294002 was reduced in islets maintained at 16 mmol/l glucose, as was mRNA expression of the PI3K regulatory subunit, p85α. CONCLUSIONS: Cholinergic regulation of insulin release is impaired in three experimental islet models of hyperglycaemia consistent with reduced expression of M3 receptors. Our data suggest that the receptor downregulation is a PKC- and PI3K-independent consequence of the hyperglycaemic environment, and they imply that M3 receptors could be potential targets in the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Hiperglucemia/metabolismo , Islotes Pancreáticos/metabolismo , Agonistas Muscarínicos/farmacología , Receptor Muscarínico M3/agonistas , Animales , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Obesos , Fosfatidilinositol 3-Quinasas/metabolismo
3.
Diabetologia ; 55(7): 1995-2004, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22526610

RESUMEN

AIMS/HYPOTHESIS: Somatostatin from islet delta cells inhibits insulin and glucagon secretion, but knowledge of the regulation of pancreatic somatostatin release is limited. Some insulin secretagogues stimulate somatostatin secretion, and here we investigated whether delta cell secretory responses are indirectly regulated in a paracrine manner by insulin released from beta cells. METHODS: Hormone release from static incubations of primary mouse islets or somatostatin-secreting TGP52 cells was measured by RIA. mRNA expression was assessed by RT-PCR. RESULTS: Glucose and a range of other physiological and pharmacological agents stimulated insulin and somatostatin release, and insulin receptor mRNA was expressed in islets, MIN6 beta cells and TGP52 cells. However, exogenous insulin did not modulate basal or glucose-induced somatostatin secretion from islets, nor did pre-incubation with an antibody against the insulin receptor or with the insulin receptor tyrosine kinase inhibitor, HNMPA(AM)(3). Glucose and tolbutamide stimulated somatostatin release from TGP52 cells, whereas a range of receptor-operating agents had no effect, the latter being consistent with a lack of corresponding receptor mRNA expression in these cells. Parasympathetic activation stimulated insulin, but inhibited somatostatin release from mouse islets in accordance with differences in muscarinic receptor mRNA expression in islets, MIN6 and TGP52 cells. The inhibitory effect on somatostatin secretion was reversed by pertussis toxin or the muscarinic receptor 2 antagonist, methoctramine. CONCLUSIONS/INTERPRETATIONS: A number of insulin secretagogues have analogous effects on insulin and somatostatin release, but this similarity of response is not mediated by an indirect, paracrine action of insulin on delta cells.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptor de Insulina/metabolismo , Células Secretoras de Somatostatina/metabolismo , Animales , Apoptosis , Línea Celular , Insulina/farmacología , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos ICR , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Diabetologia ; 54(5): 1127-35, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21267536

RESUMEN

AIMS/HYPOTHESIS: Recent studies have shown that mesenchymal stem cells (MSCs) secrete several factors that improve survival and function of transplanted islets. Implantation of islets beneath the kidney capsule results in morphological changes, due to interactions of the graft with the host, thus impairing islet function. We co-transplanted MSCs with islets to determine their effects on the remodelling process and studied graft function in a mouse model of minimal islet mass. METHODS: Islets were syngeneically transplanted, either alone or with kidney-derived MSCs, underneath the kidney capsule of streptozotocin-induced diabetic C57Bl/6 mice. Blood glucose levels were monitored and intraperitoneal glucose tolerance tests carried out. Hormone contents of grafts and pancreas were assessed by radioimmunoassay. Graft morphology and vascularisation were evaluated by immunohistochemistry. RESULTS: MSCs improved the capacity of islet grafts to reverse hyperglycaemia, with 92% of mice co-transplanted with MSCs reverting to normoglycaemia, compared with 42% of those transplanted with islets alone. Average blood glucose concentrations were lower throughout the 1 month monitoring period in MSC co-transplanted mice. MSCs did not alter graft hormone content. Islets co-transplanted with MSCs maintained a morphology that more closely resembled that of islets in the endogenous pancreas, both in terms of size, and of endocrine and endothelial cell distribution. Vascular engraftment was superior in MSC co-transplanted mice, as shown by increased endothelial cell numbers within the endocrine tissue. CONCLUSIONS/INTERPRETATION: Co-transplantation of islets with MSCs had a profound impact on the remodelling process, maintaining islet organisation and improving islet revascularisation. MSCs also improved the capacity of islets to reverse hyperglycaemia.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Diabetes Mellitus Experimental/terapia , Supervivencia de Injerto , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Diabetologia ; 49(9): 2131-5, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16826407

RESUMEN

AIMS/HYPOTHESIS: We investigated the production of kisspeptin (KISS1) and the KISS1 receptor, GPR54, in pancreatic islets and determined the effects of exogenous kisspeptin on insulin secretion. METHODS: RT-PCR and immunohistochemistry were used to detect expression of KISS1 and GPR54 mRNAs and the production of KISS1 and GPR54 in human and mouse islets and in beta (MIN6) and alpha- (alphaTC1) cell lines. The effects of KISS1 on basal and glucose-induced insulin secretion from mouse and human islets were measured in a perifusion system. RESULTS: KISS1 and GPR54 mRNAs were both detected in human and mouse islets, and GPR54 mRNA expression was also found in the MIN6 and alphaTC1 endocrine cell lines. In sections of mouse pancreas, KISS1 and GPR54 immunoreactivities were co-localised in both beta and alpha cells within islets, but were not detected in the exocrine pancreas. Exposure of mouse and human islets to KISS1 caused a stimulation of glucose-induced (20 mmol/l) insulin secretion, but had no effect on the basal rate of secretion at a sub-stimulatory concentration of glucose (2 mmol/l). In contrast, KISS1 inhibited insulin secretion from MIN6 cells at both 2 and 20 mmol/l glucose. KISS1 had no significant effect on glucagon secretion from mouse islets. CONCLUSIONS/INTERPRETATION: This is the first report to show that the GPR54/KISS1 system is expressed in the endocrine pancreas, where it influences beta cell secretory function. These observations suggest an important role for this system in the normal regulation of islet function.


Asunto(s)
Islotes Pancreáticos/metabolismo , Proteínas/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Línea Celular Tumoral , Expresión Génica , Glucagón/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Kisspeptinas , Ratones , Proteínas/genética , Proteínas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Receptores de Kisspeptina-1 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/farmacología
6.
Cell Calcium ; 31(5): 209-19, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12098223

RESUMEN

Isolated beta-cells are heterogeneous in sensory, biosynthetic and secretory capabilities, however, to enable efficient and appropriate secretion, cellular activity within the intact islet is synchronised. Historically, the entrainment of activity to a common pattern has been attributed to gap-junction mediated cell-to-cell communication. Although clearly influential, the possibility remains for other local synchronising mechanisms. In this study, we have used small clusters of insulin-secreting MIN6 cells to assess how contact-dependent, homotypic interactions between cells influences nutrient- and non-nutrient- evoked Ca(2+)-handling and insulin secretion, and to determine whether a secreted product plays a role in the synchronisation of oscillatory activity. Tolbutamide evoked a concentration-dependent recruitment of active cells within cell clusters, both in terms of numbers of cells and amplitude of the evoked Ca(2+)-response. The change in [Ca(2+)](i) was characteristically oscillatory above a mean elevated plateau, and was in phase between member cells of an individual cluster. Even at maximal concentrations (100 microM) some cells within a cluster responded before their immediate neighbours. Subsequent oscillatory behaviour then became entrained between member cells within that cluster. Inhibiting exocytosis using the microtubule inhibitors vincristine and nocodazole, or the adrenergic agent noradrenaline, did not prevent tolbutamide-evoked oscillatory changes in [Ca(2+)](i) but did reduce the probability of obtaining synchronous activity within an individual cluster. Above a threshold glucose concentration, the number of cells secreting insulin increased, without a commensurate change in secretory efficiency. This recruitment of cells secreting insulin mirrored Ca(2+) data that showed a glucose-dependent increase in cell number, without a change in the mean basal-to-peak change in [Ca(2+)](i). Together these data suggest that synchronised behaviour in MIN6 cells is dependent, in part, on a secreted factor that acts in a local paracrine fashion to recruit heterogeneous individual cellular activity into an organised group response.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Comunicación Celular/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/fisiología , Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Hipoglucemiantes/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Nocodazol/farmacología , Tolbutamida/farmacología , Vincristina/farmacología
7.
Mol Cell Endocrinol ; 191(2): 167-76, 2002 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-12062900

RESUMEN

Insulin secretion from MIN6 cells configured as cell aggregates by culture on a gelatin substrate (pseudoislets) is enhanced compared to that of MIN6 cells grown as monolayers on tissue culture plastic, indicating the importance of beta-cell-to-beta-cell proximity for insulin release. In this study we have shown that glucose induced a biphasic release of insulin from pseudoislets, whereas the amplitude and duration of the responses of equivalent monolayer cells were much reduced. Purinergic aqonists have been implicated in intercellular communication between beta-cells, so we investigated whether adenine nucleotides co-released with insulin are responsible for the enhanced secretory responses of pseudoislets. We have demonstrated that MIN6 cells express purinergic A(1) and P2Y receptors, and that adenine nucleotides increased [Ca(2+)](i) with an efficacy of agonists being ATP > ADP > AMP. However, neither suramin nor the more selective A(1) antagonist 1,3-dipropyl-8-cyclopentylxanthine reduced glucose-induced insulin secretion from pseudoislets, and stimulation of monolayer cells with a range of adenine nucleotides did not enhance glucose-induced secretion. These results suggest that enhanced secretion from MIN6 pseudoislets is not due to increased paracrine/autocrine action of adenine nucleotides.


Asunto(s)
Nucleótidos de Adenina/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Nucleótidos de Adenina/metabolismo , Nucleótidos de Adenina/farmacología , Animales , Comunicación Autocrina , Calcio/metabolismo , Línea Celular , Glucoquinasa/análisis , Glucosa/farmacología , Transportador de Glucosa de Tipo 2 , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Cinética , Ratones , Proteínas de Transporte de Monosacáridos/análisis , Comunicación Paracrina
8.
Cell Calcium ; 27(5): 287-96, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10859595

RESUMEN

The secretory response of the intact islet is greater than the response of individual beta-cells in isolation, and functional coupling between cells is critical in insulin release. The changes in intracellular Ca(2+)([Ca(2+)](i)) which initiate insulin secretory responses are synchronized between groups of cells within the islet, and gap-junctions are thought to play a central role in coordinating signalling events. We have used the MIN6 insulin-secreting cell line, to examine whether uncoupling gap-junctions alters the synchronicity of nutrient- and non-nutrient-evoked Ca(2+)oscillations, or affects insulin secretion. MIN6 cells express mRNA species that can be amplified using PCR primers for connexin 36. A commonly used gap-junctional inhibitor, heptanol, inhibited glucose- and tolbutamide-induced Ca(2+)-oscillations to basal levels in MIN6 cell clusters at concentrations of 0.5 mM and greater, and it had similar effects in pseudoislets when used at 2.5 mM. Lower heptanol concentrations altered the frequency of Ca(2+)transients without affecting their synchronicity, in both monolayers and pseudoislets. Heptanol also had effects on insulin secretion from MIN6 pseudoislets such that 1 mM enhanced secretion while 2.5 mM was inhibitory. These data suggest that heptanol has multiple effects in pancreatic beta-cells, none of which appears to be related to uncoupling of synchronicity of Ca(2+)signalling between cells. A second gap-junction uncoupler, 18 alpha-glycyrrhetinic acid, also failed to uncouple synchronized Ca(2+)-oscillations, and it had no effect on insulin secretion. These data provide evidence that Ca(2+)signalling events occur simultaneously across the bulk mass of the pseudoislet, and suggest that gap-junctions are not required to coordinate the synchronicity of these events, nor is communication via gap junctions essential for integrated insulin secretory responses.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Administración Tópica , Animales , Antiinflamatorios/farmacología , Señalización del Calcio/fisiología , Línea Celular , Uniones Comunicantes/metabolismo , Glucosa/farmacología , Ácido Glicirretínico/farmacología , Heptanol/farmacología , Humanos , Hipoglucemiantes/farmacología , Islotes Pancreáticos/metabolismo , Ratones , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Tolbutamida/farmacología
9.
Diabetes ; 48(7): 1402-8, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10389845

RESUMEN

The effect of cell-to-cell contact on Ca2+ influx and secretory responses in the beta-cell line MIN6 was studied using MIN6 pseudoislets, which are three-dimensional islet-like cell aggregates that develop when MIN6 cells are cultured for 6-8 days on gelatin. The formation of pseudoislets is dependent on the Ca2+-dependent adhesion molecule E-cadherin (E-CAD), since the process can be inhibited by incubation in the absence of Ca2+ or in the presence of an anti-E-CAD antibody. Glucose and alpha-ketoisocaproic acid (KIC) evoked a Ca2+ influx in only a small fraction of the MIN6 monolayer cells, whereas >80% of cell groups within the pseudoislets responded to both nutrients. In contrast, changes in the intracellular free Ca2+ concentration ([Ca2+]i) were observed in all or most monolayer cells or pseudoislet cell groups in response to physical or pharmacological depolarizing stimuli. No significant increase in insulin release was observed from MIN6 monolayer cells in response to nutrient or nonnutrient insulin secretagogues. Conversely, pseudoislets were found to respond significantly to both nutrients and nonnutrients. These results suggest that close cell-to-cell contact improves the functional responsiveness of MIN6 cells and that pseudoislets may therefore serve as a useful research model in the study of beta-cell function.


Asunto(s)
Calcio/metabolismo , Agregación Celular/fisiología , Comunicación Celular/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Estado Nutricional , Línea Celular Transformada , Humanos , Secreción de Insulina , Tasa de Secreción , Estimulación Química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...