Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 18(1): 6-15, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26228188

RESUMEN

Diabetes mellitus is a growing worldwide epidemic disease, currently affecting 1 in 12 adults. Treatment of disease complications typically consumes ∼10% of healthcare budgets in developed societies. Whilst immune-mediated destruction of insulin-secreting pancreatic ß cells is responsible for Type 1 diabetes, both the loss and dysfunction of these cells underly the more prevalent Type 2 diabetes. The establishment of robust drug development programmes aimed at ß-cell restoration is still hampered by the absence of means to measure ß-cell mass prospectively in vivo, an approach which would provide new opportunities for understanding disease mechanisms and ultimately assigning personalized treatments. In the present review, we describe the progress towards this goal achieved by the Innovative Medicines Initiative in Diabetes, a collaborative public-private consortium supported by the European Commission and by dedicated resources of pharmaceutical companies. We compare several of the available imaging methods and molecular targets and provide suggestions as to the likeliest to lead to tractable approaches. Furthermore, we discuss the simultaneous development of animal models that can be used to measure subtle changes in ß-cell mass, a prerequisite for validating the clinical potential of the different imaging tracers.


Asunto(s)
Diabetes Mellitus/patología , Células Secretoras de Insulina/patología , Imagen Molecular/métodos , Adulto , Animales , Adhesión Celular , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Mediciones Luminiscentes , Manganeso , Glicoproteínas de Membrana/metabolismo , Ratones , Ratas , Receptores de Sulfonilureas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Zinc
2.
Diabetologia ; 57(1): 122-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24149837

RESUMEN

AIMS/HYPOTHESIS: Type 2 diabetes is characterised by progressive beta cell dysfunction, with changes in gene expression playing a crucial role in its development. MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression and therefore alterations in miRNA levels may be involved in the deterioration of beta cell function. METHODS: Global TaqMan arrays and individual TaqMan assays were used to measure islet miRNA expression in discovery (n = 20) and replication (n = 20) cohorts from individuals with and without type 2 diabetes. The role of specific dysregulated miRNAs in regulating insulin secretion, content and apoptosis was subsequently investigated in primary rat islets and INS-1 cells. Identification of miRNA targets was assessed using luciferase assays and by measuring mRNA levels. RESULTS: In the discovery and replication cohorts miR-187 expression was found to be significantly increased in islets from individuals with type 2 diabetes compared with matched controls. An inverse correlation between miR-187 levels and glucose-stimulated insulin secretion (GSIS) was observed in islets from normoglycaemic donors. This correlation paralleled findings in primary rat islets and INS-1 cells where overexpression of miR-187 markedly decreased GSIS without affecting insulin content or apoptotic index. Finally, the gene encoding homeodomain-interacting protein kinase-3 (HIPK3), a known regulator of insulin secretion, was identified as a direct target of miR-187 and displayed reduced expression in islets from individuals with type 2 diabetes. CONCLUSIONS/INTERPRETATION: Our findings suggest a role for miR-187 in the blunting of insulin secretion, potentially involving regulation of HIPK3, which occurs during the pathogenesis of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , MicroARNs/metabolismo , Adulto , Anciano , Animales , Línea Celular , Células Cultivadas , Humanos , Secreción de Insulina , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MicroARNs/genética , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Diabet Med ; 31(12): 1480-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25186316

RESUMEN

Whilst the heritable nature of Type 2 diabetes has been recognized for many years, only in the past two decades have linkage analyses in families and genome-wide association studies in large populations begun to reveal the genetic landscape of the disease in detail. Whilst the former have provided a powerful means of identifying the genes responsible for monogenic forms of the disease, the latter highlight relatively large genomic regions. These often harbour multiple genes, whose relative contribution to exaggerated disease risk is uncertain. In the present study, the approaches that have been used to dissect the role of just a few (TCF7L2, SLC30A8, ADCY5, MTNR1B and CDKAL1) of the ~ 500 genes identified at dozens of implicated loci are described. These are usually selected based on the strength of their effect on disease risk, and predictions as to their likely biological role. Direct determination of the effects of identified polymorphisms on gene expression in disease-relevant tissues, notably the pancreatic islet, are then performed to identify genes whose expression is affected by a particular polymorphism. Subsequent functional analyses then involve perturbing gene expression in vitro in ß-cell lines or isolated islets and in vivo in animal models. Although the majority of polymorphisms affect insulin production rather than action, and mainly affect the ß cell, effects via other tissues may also contribute, requiring careful consideration in the design and interpretation of experiments in model systems. These considerations illustrate the scale of the task needed to exploit genome-wide association study data for the development of new therapeutic strategies.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Estudio de Asociación del Genoma Completo , Adenilil Ciclasas/genética , Proteínas de Transporte de Catión/genética , Quinasa 5 Dependiente de la Ciclina/genética , Humanos , Receptor de Melatonina MT2/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Transportador 8 de Zinc , ARNt Metiltransferasas
4.
Mol Metab ; 85: 101947, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38677509

RESUMEN

OBJECTIVE: Type 2 diabetes (T2D) is characterised by the loss of first-phase insulin secretion. We studied mice with ß-cell selective loss of the glucagon receptor (Gcgrfl/fl X Ins-1Cre), to investigate the role of intra-islet glucagon receptor (GCGR) signalling on pan-islet [Ca2+]I activity and insulin secretion. METHODS: Metabolic profiling was conducted on Gcgrß-cell-/- and littermate controls. Crossing with GCaMP6f (STOP flox) animals further allowed for ß-cell specific expression of a fluorescent calcium indicator. These islets were functionally imaged in vitro and in vivo. Wild-type mice were transplanted with islets expressing GCaMP6f in ß-cells into the anterior eye chamber and placed on a high fat diet. Part of the cohort received a glucagon analogue (GCG-analogue) for 40 days and the control group were fed to achieve weight matching. Calcium imaging was performed regularly during the development of hyperglycaemia and in response to GCG-analogue treatment. RESULTS: Gcgrß-cell-/- mice exhibited higher glucose levels following intraperitoneal glucose challenge (control 12.7 mmol/L ± 0.6 vs. Gcgrß-cell-/- 15.4 mmol/L ± 0.0 at 15 min, p = 0.002); fasting glycaemia was not different to controls. In vitro, Gcgrß-cell-/- islets showed profound loss of pan-islet [Ca2+]I waves in response to glucose which was only partially rescued in vivo. Diet induced obesity and hyperglycaemia also resulted in a loss of co-ordinated [Ca2+]I waves in transplanted islets. This was reversed with GCG-analogue treatment, independently of weight-loss (n = 8). CONCLUSION: These data provide novel evidence for the role of intra-islet GCGR signalling in sustaining synchronised [Ca2+]I waves and support a possible therapeutic role for glucagonergic agents to restore the insulin secretory capacity lost in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucagón , Glucosa , Homeostasis , Secreción de Insulina , Células Secretoras de Insulina , Receptores de Glucagón , Transducción de Señal , Animales , Glucagón/metabolismo , Ratones , Células Secretoras de Insulina/metabolismo , Glucosa/metabolismo , Receptores de Glucagón/metabolismo , Receptores de Glucagón/genética , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Masculino , Islotes Pancreáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Dieta Alta en Grasa , Glucemia/metabolismo , Femenino
5.
Diabetes Obes Metab ; 15(6): 503-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23121289

RESUMEN

Outside of the biological arena the term 'repression' often has a negative connotation. However, in the pancreatic ß-cell a small group of genes, which are abundantly expressed in most if not all other mammalian tissues, are highly selectively repressed, with likely functional consequences. The two 'founder' members of this group, lactate dehydrogenase A (Ldha) and monocarboxylate transporter-1 (MCT-1/Slc16a1), are inactivated by multiple mechanisms including histone modifications and microRNA-mediated silencing. Their inactivation ensures that pyruvate and lactate, derived from muscle during exercise, do not stimulate insulin release inappropriately. Correspondingly, activating mutations in the MCT-1 promoter underlie 'exercise-induced hyperinsulinism' (EIHI) in man, a condition mimicked by forced over-expression of MCT-1 in the ß-cell in mice. Furthermore, LDHA expression in the ß-cell is upregulated in both human type 2 diabetes and in rodent models of the disease. Recent work by us and by others has identified a further ∼60 genes which are selectively inactivated in the ß-cell, a list which we refine here up to seven by detailed comparison of the two studies. These genes include key regulators of cell proliferation and stimulus-secretion coupling. The present, and our earlier results, thus highlight the probable importance of shutting down a subset of 'disallowed' genes for the differentiated function of ß-cells, and implicate previously unsuspected signalling pathways in the control of ß-cell expansion and insulin secretion. Targeting of deregulated 'disallowed' genes in these cells may thus, in the future, provide new therapeutic avenues for type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Marcación de Gen/métodos , Hiperinsulinismo/genética , Células Secretoras de Insulina/metabolismo , L-Lactato Deshidrogenasa/genética , Transportadores de Ácidos Monocarboxílicos/genética , Simportadores/genética , Animales , Proliferación Celular , Diabetes Mellitus Tipo 2/fisiopatología , Epigenómica , Ejercicio Físico , Femenino , Silenciador del Gen , Humanos , Hiperinsulinismo/etiología , Hiperinsulinismo/fisiopatología , Isoenzimas/genética , Lactato Deshidrogenasa 5 , Masculino , Ratones , Mutación , Estrés Oxidativo
6.
Diabetologia ; 55(10): 2667-2676, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22717537

RESUMEN

AIMS/HYPOTHESIS: Individuals carrying type 2 diabetes risk alleles in TCF7L2 display decreased beta cell levels of T cell factor 7 like-2 (TCF7L2) immunoreactivity, and impaired insulin secretion and beta cell sensitivity to glucagon-like peptide 1 (GLP-1). Here, we sought to determine whether selective deletion of Tcf7l2 in mouse pancreas impairs insulin release and glucose homeostasis. METHODS: Pancreas-specific Tcf7l2-null (pTcf7l2) mice were generated by crossing mice carrying conditional knockout alleles of Tcf7l2 (Tcf7l2-flox) with mice expressing Cre recombinase under the control of the Pdx1 promoter (Pdx1.Cre). Gene expression was assessed by real-time quantitative PCR and beta cell mass by optical projection tomography. Glucose tolerance, insulin secretion from isolated islets, and plasma insulin, glucagon and GLP-1 content were assessed by standard protocols. RESULTS: From 12 weeks of age, pTcf7l2 mice displayed decreased oral glucose tolerance vs control littermates; from 20 weeks they had glucose intolerance upon administration of glucose by the intraperitoneal route. pTcf7l2 islets displayed impaired insulin secretion in response to 17 (vs 3.0) mmol/l glucose (54.6 ± 4.6%, p < 0.01) or to 17 mmol/l glucose plus 100 nmol/l GLP-1 (44.3 ± 4.9%, p < 0.01) compared with control islets. Glp1r (42 ± 0.08%, p < 0.01) and Ins2 (15.4 ± 4.6%, p < 0.01) expression was significantly lower in pTcf7l2 islets than in controls. Maintained on a high-fat (but not on a normal) diet, pTcf7l2 mice displayed decreased expansion of pancreatic beta cell volume vs control littermates. No differences were observed in plasma insulin, proinsulin, glucagon or GLP-1 concentrations. CONCLUSIONS/INTERPRETATION: Selective deletion of Tcf7l2 in the pancreas replicates key aspects of the altered glucose homeostasis in human carriers of TCF7L2 risk alleles, indicating the direct role of this factor in controlling beta cell function.


Asunto(s)
Glucosa/metabolismo , Homeostasis/fisiología , Insulina/metabolismo , Páncreas/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/deficiencia , Animales , Células Cultivadas , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/farmacología , Prueba de Tolerancia a la Glucosa , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Páncreas/citología , Páncreas/efectos de los fármacos , Proinsulina/sangre , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
7.
Proc Natl Acad Sci U S A ; 106(35): 14872-7, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19706465

RESUMEN

Zinc co-crystallizes with insulin in dense core secretory granules, but its role in insulin biosynthesis, storage and secretion is unknown. In this study we assessed the role of the zinc transporter ZnT8 using ZnT8-knockout (ZnT8(-/-)) mice. Absence of ZnT8 expression caused loss of zinc release upon stimulation of exocytosis, but normal rates of insulin biosynthesis, normal insulin content and preserved glucose-induced insulin release. Ultrastructurally, mature dense core insulin granules were rare in ZnT8(-/-) beta cells and were replaced by immature, pale insulin "progranules," which were larger than in ZnT8(+/+) islets. When mice were fed a control diet, glucose tolerance and insulin sensitivity were normal. However, after high-fat diet feeding, the ZnT8(-/-) mice became glucose intolerant or diabetic, and islets became less responsive to glucose. Our data show that the ZnT8 transporter is essential for the formation of insulin crystals in beta cells, contributing to the packaging efficiency of stored insulin. Interaction between the ZnT8(-/-) genotype and diet to induce diabetes is a model for further studies of the mechanism of disease of human ZNT8 gene mutations.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Insulina/química , Insulina/metabolismo , Animales , Calcio/metabolismo , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Cristalización , Glucosa/administración & dosificación , Glucosa/metabolismo , Intolerancia a la Glucosa/inducido químicamente , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Zinc/metabolismo , Transportador 8 de Zinc
8.
Diabetologia ; 54(1): 125-34, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20938634

RESUMEN

AIM/HYPOTHESIS: AMP-activated protein kinase (AMPK), encoded by Prkaa genes, is emerging as a key regulator of overall energy homeostasis and the control of insulin secretion and action. We sought here to investigate the role of AMPK in controlling glucagon secretion from pancreatic islet alpha cells. METHODS: AMPK activity was modulated in vitro in clonal alphaTC1-9 cells and isolated mouse pancreatic islets using pharmacological agents and adenoviruses encoding constitutively active or dominant negative forms of AMPK. Glucagon secretion was measured during static incubation by radioimmunoassay. AMPK activity was assessed by both direct phosphotransfer assay and by western (immuno-)blotting of the phosphorylated AMPK α subunits and the downstream target acetyl-CoA carboxylase 1. Intracellular free [Ca²(+)] was measured using Fura-Red. RESULTS: Increasing glucose concentrations strongly inhibited AMPK activity in clonal pancreatic alpha cells. Forced increases in AMPK activity in alphaTC1-9 cells, achieved through the use of pharmacological agents including metformin, phenformin and A-769662, or via adenoviral transduction, resulted in stimulation of glucagon secretion at both low and high glucose concentrations, whereas AMPK inactivation inhibited both [Ca²(+)](i) increases and glucagon secretion at low glucose. Transduction of isolated mouse islets with an adenovirus encoding AMPK-CA under the control of the preproglucagon promoter increased glucagon secretion selectively at elevated glucose concentrations. CONCLUSIONS/INTERPRETATION: AMPK is strongly regulated by glucose in pancreatic alpha cells, and increases in AMPK activity are sufficient and necessary for the stimulation of glucagon release in vitro. Modulation of AMPK activity in alpha cells may therefore provide a novel approach to controlling blood glucose concentrations.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Secretoras de Glucagón/enzimología , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Acetiltransferasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Compuestos de Bifenilo , Western Blotting , Calcio/metabolismo , Línea Celular , Activación Enzimática/efectos de los fármacos , Femenino , Células Secretoras de Glucagón/efectos de los fármacos , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Inmunohistoquímica , Metformina/farmacología , Ratones , Fenformina/farmacología , Fosforilación/efectos de los fármacos , Pironas/farmacología , Tiofenos/farmacología
9.
Diabetologia ; 54(12): 3078-82, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21913056

RESUMEN

AIMS/HYPOTHESIS: Intronic single nucleotide polymorphisms within the transcription factor 7-like 2 (TCF7L2) gene are associated with risk of type 2 diabetes. It is widely hypothesised that the predisposing variation is involved in cis-regulation of TCF7L2 activity. The aim of this study was to seek evidence for the existence of novel TCF7L2 isoforms encoded within the type 2 diabetes-associated genomic region. METHODS: We searched expressed sequence tag (EST) databases for novel TCF7L2 transcripts and sought to validate the function and integrity of any isoforms found using a combination of RT-PCR, western blotting and reporter gene techniques. RESULTS: Analysis of EST databases suggested the presence of an alternative polyadenylation site located in intron 4 of TCF7L2. We used 3' rapid amplification of cDNA ends and real-time PCR to validate the integrity of this polyadenylation signal and show its wide use across human tissues. Western blotting results are consistent with the use of this polyadenylation signal to generate novel protein isoforms. The alternative polyadenylation signal results in the production of isoforms that retain the ß-catenin binding domain but do not possess the high-mobility group box DNA-binding domain. Promoter-reporter gene assays suggest that these isoforms inhibit TCF7L2-dependent target genes by sequestering ß-catenin. CONCLUSIONS/INTERPRETATION: We have identified a novel polyadenylation signal within TCF7L2 that can result in the production of isoforms that act to repress TCF/LEF-dependent target genes. These findings may provide new insights into the association of TCF7L2 with susceptibility to type 2 diabetes.


Asunto(s)
Regulación de la Expresión Génica , Poliadenilación , Isoformas de Proteínas/metabolismo , Transducción de Señal , Factores de Transcripción TCF/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Empalme Alternativo , Secuencia de Bases , Línea Celular , Bases de Datos Genéticas , Diabetes Mellitus Tipo 2/genética , Exones , Etiquetas de Secuencia Expresada , Humanos , Intestino Delgado/metabolismo , Intrones , Datos de Secuencia Molecular , Páncreas/metabolismo , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Factores de Transcripción TCF/genética , beta Catenina/metabolismo
10.
Diabetologia ; 54(4): 819-27, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21181396

RESUMEN

AIMS/HYPOTHESIS: We assessed whether per-arnt-sim (PAS) domain-containing protein kinase (PASK) is involved in the regulation of glucagon secretion. METHODS: mRNA levels were measured in islets by quantitative PCR and in pancreatic beta cells obtained by laser capture microdissection. Glucose tolerance, plasma hormone levels and islet hormone secretion were analysed in C57BL/6 Pask homozygote knockout mice (Pask-/-) and control littermates. Alpha-TC1-9 cells, human islets or cultured E13.5 rat pancreatic epithelia were transduced with anti-Pask or control small interfering RNAs, or with adenoviruses encoding enhanced green fluorescent protein or PASK. RESULTS: PASK expression was significantly lower in islets from human type 2 diabetic than control participants. PASK mRNA was present in alpha and beta cells from mouse islets. In Pask-/- mice, fasted blood glucose and plasma glucagon levels were 25 ± 5% and 50 ± 8% (mean ± SE) higher, respectively, than in control mice. At inhibitory glucose concentrations (10 mmol/l), islets from Pask-/- mice secreted 2.04 ± 0.2-fold (p < 0.01) more glucagon and 2.63 ± 0.3-fold (p < 0.01) less insulin than wild-type islets. Glucose failed to inhibit glucagon secretion from PASK-depleted alpha-TC1-9 cells, whereas PASK overexpression inhibited glucagon secretion from these cells and human islets. Extracellular insulin (20 nmol/l) inhibited glucagon secretion from control and PASK-deficient alpha-TC1-9 cells. PASK-depleted alpha-TC1-9 cells and pancreatic embryonic explants displayed increased expression of the preproglucagon (Gcg) and AMP-activated protein kinase (AMPK)-alpha2 (Prkaa2) genes, implying a possible role for AMPK-alpha2 downstream of PASK in the control of glucagon gene expression and release. CONCLUSIONS/INTERPRETATION: PASK is involved in the regulation of glucagon secretion by glucose and may be a useful target for the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Islotes Pancreáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Glucosa/farmacología , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Ratones , Ratones Mutantes , Modelos Biológicos , Reacción en Cadena de la Polimerasa , Proteínas Serina-Treonina Quinasas/genética , Ratas
11.
Diabetologia ; 53(8): 1656-68, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20424817

RESUMEN

AIMS/HYPOTHESIS: Zinc is highly concentrated in pancreatic beta cells, is critical for normal insulin storage and may regulate glucagon secretion from alpha cells. Zinc transport family member 8 (ZnT8) is a zinc efflux transporter that is highly abundant in beta cells. Polymorphisms of ZnT8 (also known as SLC30A8) gene in man are associated with increased risk of type 2 diabetes. While global Znt8 knockout (Znt8KO) mice have been characterised, ZnT8 is also present in other islet cell types and extra-pancreatic tissues. Therefore, it is important to find ways of understanding the role of ZnT8 in beta and alpha cells without the difficulties caused by the confounding effects of ZnT8 in these other tissues. METHODS: We generated mice with beta cell-specific (Znt8BKO) and alpha cell-specific (Znt8AKO) knockout of Znt8, and performed in vivo and in vitro characterisation of the phenotypes to determine the functional and anatomical impact of ZnT8 in these cells. Thus we assessed zinc accumulation, insulin granule morphology, insulin biosynthesis and secretion, and glucose homeostasis. RESULTS: Znt8BKO mice are glucose-intolerant, have reduced beta cell zinc accumulation and atypical insulin granules. They also display reduced first-phase glucose-stimulated insulin secretion, reduced insulin processing enzyme transcripts and increased proinsulin levels. In contrast, Znt8AKO mice show no evident abnormalities in plasma glucagon and glucose homeostasis. CONCLUSIONS/INTERPRETATION: This is the first report of specific beta and alpha cell deletion of Znt8. Our data indicate that while, under the conditions studied, ZnT8 is absolutely essential for proper beta cell function, it is largely dispensable for alpha cell function.


Asunto(s)
Proteínas de Transporte de Catión/genética , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Zinc/metabolismo , Análisis de Varianza , Animales , Western Blotting , Proteínas de Transporte de Catión/metabolismo , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/metabolismo , Células Secretoras de Glucagón/metabolismo , Inmunohistoquímica , Insulina/genética , Secreción de Insulina , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Inmunoelectrónica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transportador 8 de Zinc
12.
Diabetologia ; 53(5): 924-36, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20221584

RESUMEN

AIMS/HYPOTHESIS: AMP-activated protein kinase (AMPK) is an evolutionarily conserved enzyme and a target of glucose-lowering agents, including metformin. However, the precise role or roles of the enzyme in controlling insulin secretion remain uncertain. METHODS: The catalytic alpha1 and alpha2 subunits of AMPK were ablated selectively in mouse pancreatic beta cells and hypothalamic neurons by breeding Ampkalpha1 [also known as Prkaa1]-knockout mice, bearing floxed Ampkalpha2 [also known as Prkaa2] alleles (Ampkalpha1 ( -/- ),alpha2( fl/fl ),), with mice expressing Cre recombinase under the rat insulin promoter (RIP2). RIP2 was used to express constitutively activated AMPK selectively in beta cells in transgenic mice. Food intake, body weight and urinary catecholamines were measured using metabolic cages. Glucose and insulin tolerance were determined after intraperitoneal injection. Beta cell mass and morphology were analysed by optical projection tomography and confocal immunofluorescence microscopy, respectively. Granule docking, insulin secretion, membrane potential and intracellular free Ca(2+) were measured with standard techniques. RESULTS: Trigenic Ampkalpha1 ( -/- ),alpha2( fl/fl ) expressing Cre recombinase and lacking both AMPKalpha subunits in the beta cell, displayed normal body weight and increased insulin sensitivity, but were profoundly insulin-deficient. Secreted catecholamine levels were unchanged. Total beta cell mass was unaltered, while mean islet and beta cell volume were reduced. AMPK-deficient beta cells displayed normal glucose-induced changes in membrane potential and intracellular free Ca(2+), while granule docking and insulin secretion were enhanced. Conversely, betaAMPK transgenic mice were glucose-intolerant and displayed defective insulin secretion. CONCLUSIONS/INTERPRETATION: Inhibition of AMPK activity within the beta cell is necessary, but not sufficient for stimulation of insulin secretion by glucose to occur. AMPK activation in extrapancreatic RIP2.Cre-expressing cells might also influence insulin secretion in vivo.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Hipotálamo/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Análisis de Varianza , Animales , Glucemia/metabolismo , Peso Corporal/genética , Grasas de la Dieta , Ingestión de Alimentos/genética , Electrofisiología , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/genética , Secreción de Insulina , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Ratas
13.
J Cell Biol ; 155(1): 41-51, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11571310

RESUMEN

The role of dense core secretory vesicles in the control of cytosolic-free Ca(2+) concentrations ([Ca(2+)](c)) in neuronal and neuroendocrine cells is enigmatic. By constructing a vesicle-associated membrane protein 2-synaptobrevin.aequorin chimera, we show that in clonal pancreatic islet beta-cells: (a) increases in [Ca(2+)](c) cause a prompt increase in intravesicular-free Ca(2+) concentration ([Ca(2+)]SV), which is mediated by a P-type Ca(2+)-ATPase distinct from the sarco(endo) plasmic reticulum Ca(2+)-ATPase, but which may be related to the PMR1/ATP2C1 family of Ca(2+) pumps; (b) steady state Ca(2+) concentrations are 3-5-fold lower in secretory vesicles than in the endoplasmic reticulum (ER) or Golgi apparatus, suggesting the existence of tightly bound and more rapidly exchanging pools of Ca(2+); (c) inositol (1,4,5) trisphosphate has no impact on [Ca(2+)](SV) in intact or permeabilized cells; and (d) ryanodine receptor (RyR) activation with caffeine or 4-chloro-3-ethylphenol in intact cells, or cyclic ADPribose in permeabilized cells, causes a dramatic fall in [Ca(2+)](SV). Thus, secretory vesicles represent a dynamic Ca(2+) store in neuroendocrine cells, whose characteristics are in part distinct from the ER/Golgi apparatus. The presence of RyRs on secretory vesicles suggests that local Ca(2+)-induced Ca(2+) release from vesicles docked at the plasma membrane could participate in triggering exocytosis.


Asunto(s)
Aequorina/metabolismo , Calcio/metabolismo , Imidazoles , Proteínas de la Membrana/metabolismo , Vesículas Secretoras/metabolismo , Adenosina Trifosfato/metabolismo , Adenoviridae/fisiología , Aequorina/genética , Animales , Cafeína/farmacología , Línea Celular , Estimulantes del Sistema Nervioso Central/farmacología , Quelantes/farmacología , Ácido Egtácico/farmacología , Retículo Endoplásmico/metabolismo , Genes Reporteros/genética , Inmunohistoquímica , Inositol 1,4,5-Trifosfato/farmacología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Pirazinas/farmacología , Proteínas R-SNARE , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Vesículas Secretoras/química
14.
Trends Biochem Sci ; 25(5): 215-21, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10782088

RESUMEN

New live-cell imaging techniques indicate that mitochondria exist in the living cell as a continuous interconnected mitochondrial reticulum, or 'MR', closely associated with the endoplasmic reticulum (ER). Ca2+ ions released from the ER in response to hormonal stimulation might thus be preferentially transferred into the mitochondrial matrix causing the local activation of ATP synthesis. Ca2+ uptake into the MR might also subtly modify the activity of ER Ca2+ release channels and thus the dynamics of cytosolic Ca2+ oscillations and waves.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Transporte Iónico
15.
Curr Biol ; 9(12): R443-5, 1999 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-10375519

RESUMEN

It has long been accepted wisdom that insulin secreted from islet beta cells has either no effect, or an inhibitory feedback effect, on insulin synthesis and secretion. Recent work suggests, instead, that secreted insulin acts directly on beta cells, via its own receptor, to enhance insulin production in an autocrine feed-forward loop.


Asunto(s)
Insulina/biosíntesis , Insulina/metabolismo , Animales , Retroalimentación , Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Humanos , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Proinsulina/genética , Precursores de Proteínas/genética , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
16.
Curr Biol ; 10(20): R736-8, 2000 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-11069096

RESUMEN

Insulin resistance, the hallmark of non-insulin dependent diabetes mellitus, is characterized by the failure of tissues to take up and store glucose in response to insulin. Two recent studies shed new light on the importance of insulin signalling in the liver and how this may become defective in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus/fisiopatología , Insulina/fisiología , Hígado/fisiopatología , Receptor de Insulina/fisiología , Animales , Diabetes Mellitus/metabolismo , Humanos , Resistencia a la Insulina , Hígado/fisiología , Ratones , Ratones Noqueados , Receptor de Insulina/deficiencia , Receptor de Insulina/genética
17.
Curr Biol ; 5(8): 890-9, 1995 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-7583147

RESUMEN

BACKGROUND: Studies of the mechanisms by which signals are transmitted from receptor tyrosine kinases would be facilitated by a way of monitoring events at the single-cell level. We have explored how luciferase imaging can be used to examine the role of specific signalling pathways in insulin-stimulated gene expression. The analysis of luciferase expression in single cells has previously been hampered by the insensitivity of existing methodologies and the lack of a way of monitoring quantitatively, and independently, more than one promoter within the same cell. We have developed a technique for examining the dynamics of insulin-stimulated AP-1-dependent transcription in single living cells, and have explored the signalling pathway involved. RESULTS: Luciferase and aequorin gene expression were examined in single living cells with a high-sensitivity photon-counting camera. The technique involved the comicroinjection of luciferase- and aequorin-based reporter plasmids directly into the cell nucleus, and the subsequent analysis of luminescence in the presence of luciferin and coelenterazine, respectively. The method is quantitative and allows insulin-stimulated gene expression to be monitored in real time. We found that insulin promoted a substantial increase in the expression of a luciferase gene under the control of the AP-1-binding site from the collagenase gene promoter. Aequorin expression, under the control of a cytomegalovirus promoter, was unaffected by insulin. The effect of insulin on luciferase expression was specifically blocked by overexpression of either the mitogen-activated protein (MAP) kinase phosphatase CL100, or the dominant-negative mutant MAP kinase kinase, MEKS217/221A. CONCLUSIONS: Microinjection coupled with luciferase imaging allows hormone-regulated gene expression from relatively weak promoters to be monitored in single living cells. We have used this method to demonstrate that MAP kinase plays a central role in the ability of insulin to stimulate AP-1-dependent gene transcription.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas de Ciclo Celular , Proteínas Inmediatas-Precoces/genética , Insulina/metabolismo , Luciferasas/genética , Proteínas Quinasas Activadas por Mitógenos , Fosfoproteínas Fosfatasas , Proteínas Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/genética , Transducción de Señal , Aequorina/genética , Animales , Transporte Biológico , Células CHO , Núcleo Celular/metabolismo , Colagenasas/genética , Cricetinae , Fosfatasa 1 de Especificidad Dual , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Procesamiento de Imagen Asistido por Computador , Insulina/farmacología , Microinyecciones , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Regiones Promotoras Genéticas , Proteína Fosfatasa 1 , Factor de Transcripción AP-1/metabolismo
18.
Curr Biol ; 10(20): 1307-10, 2000 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-11069115

RESUMEN

The classical model of secretory vesicle recycling after exocytosis involves the retrieval of membrane (the omega figure) at a different site. An alternative model involves secretory vesicles transiently fusing with the plasma membrane (the 'kiss and run' mechanism) [1,2]. No continuous observation of the fate of a single secretory vesicle after exocytosis has been made to date. To study the dynamics of fusion immediately following exocytosis of insulin-containing vesicles, enhanced green fluorescent protein (EGFP) fused to the vesicle membrane protein phogrin [3] was delivered to the secretory vesicle membrane of INS-1 beta-cells using an adenoviral vector. The behaviour of the vesicle membrane during single exocytotic events was then examined using evanescent wave microscopy [4-6]. In unstimulated cells, secretory vesicles showed only slow Brownian movement. After a depolarizing pulse, most vesicles showed a small decrease in phogrin-EGFP fluorescence, and some moved laterally over the plasma membrane for approximately 1 microm. In contrast, secretory vesicles loaded with acridine orange all showed a transient (33-100 ms) increase in fluorescence intensity followed by rapid disappearance. Simultaneous observations of phogrin-EGFP and acridine orange indicated that the decrease in EGFP fluorescence occurred at the time of the acridine orange release, and that the lateral movement of EGFP-expressing vesicles occurred after this. Post-exocytotic retrieval of the vesicle membrane in INS-1 cells is thus slow, and can involve the movement of empty vesicles under the plasma membrane ('kiss and glide').


Asunto(s)
Exocitosis/fisiología , Insulina/fisiología , Proteínas de la Membrana , Vesículas Secretoras/fisiología , Naranja de Acridina/análisis , Animales , Línea Celular , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Estimulación Eléctrica , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/análisis , Fusión de Membrana , Glicoproteínas de Membrana/análisis , Microscopía Fluorescente/métodos , Microscopía por Video/métodos , Proteínas Tirosina Fosfatasas/análisis , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores , Vesículas Secretoras/ultraestructura
19.
Curr Biol ; 9(16): 915-8, 1999 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-10469598

RESUMEN

Regulated exocytosis involves the Ca(2+)-triggered fusion of secretory vesicles with the plasma membrane, by activation of vesicle membrane Ca(2+)-binding proteins [1]. The Ca(2+)-binding sites of these proteins are likely to lie within 30 nm of the vesicle surface, a domain in which changes in Ca2+ concentration cannot be resolved by conventional fluorescence microscopy. A fluorescent indicator for Ca2+ called a yellow 'cameleon' (Ycam2) - comprising a fusion between a cyan-emitting mutant of the green fluorescent protein (GFP), calmodulin, the calmodulin-binding peptide M13 and an enhanced yellow-emitting GFP - which is targetable to specific intracellular locations, has been described [2]. Here, we generated a fusion between phogrin, a protein that is localised to secretory granule membranes [3], and Ycam2 (phogrin-Ycam2) to monitor changes in Ca2+ concentration ([Ca2+]) at the secretory vesicle surface ([Ca2+]gd) through alterations in fluorescence resonance energy transfer (FRET) between the linked cyan and yellow fluorescent proteins (CFP and YFP, respectively) in Ycam2. In both neuroendocrine PC12 and MIN6 pancreatic beta cells, apparent resting values of cytosolic [Ca2+] and [Ca2+](gd) were similar throughout the cell. In MIN6 cells following the activation of Ca2+ influx, the minority of vesicles that were within approximately 1 microm of the plasma membrane underwent increases in [Ca2+](gd) that were significantly greater than those experienced by deeper vesicles, and greater than the apparent cytosolic [Ca2+] change. The ability to image both global and compartmentalised [Ca2+] changes with recombinant targeted cameleons should extend the usefulness of these new Ca2+ probes.


Asunto(s)
Calcio/metabolismo , Gránulos Citoplasmáticos/metabolismo , Proteínas de la Membrana , Microscopía Fluorescente/métodos , Proteínas Recombinantes de Fusión/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Calcio/análisis , Línea Celular , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/metabolismo , Glicoproteínas de Membrana/metabolismo , Células PC12 , Proteínas Tirosina Fosfatasas/metabolismo , Ratas , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores
20.
Bioinformatics ; 22(20): 2547-53, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16877753

RESUMEN

MOTIVATION: Many standard statistical techniques are effective on data that are normally distributed with constant variance. Microarray data typically violate these assumptions since they come from non-Gaussian distributions with a non-trivial mean-variance relationship. Several methods have been proposed that transform microarray data to stabilize variance and draw its distribution towards the Gaussian. Some methods, such as log or generalized log, rely on an underlying model for the data. Others, such as the spread-versus-level plot, do not. We propose an alternative data-driven multiscale approach, called the Data-Driven Haar-Fisz for microarrays (DDHFm) with replicates. DDHFm has the advantage of being 'distribution-free' in the sense that no parametric model for the underlying microarray data is required to be specified or estimated; hence, DDHFm can be applied very generally, not just to microarray data. RESULTS: DDHFm achieves very good variance stabilization of microarray data with replicates and produces transformed intensities that are approximately normally distributed. Simulation studies show that it performs better than other existing methods. Application of DDHFm to real one-color cDNA data validates these results. AVAILABILITY: The R package of the Data-Driven Haar-Fisz transform (DDHFm) for microarrays is available in Bioconductor and CRAN.


Asunto(s)
Algoritmos , Interpretación Estadística de Datos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/normas , Colorimetría/métodos , Colorimetría/normas , Simulación por Computador , Bases de Datos Genéticas , Almacenamiento y Recuperación de la Información/métodos , Modelos Genéticos , Modelos Estadísticos , Valores de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA