Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Endocrinol ; 496: 110524, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31362031

RESUMEN

Decreased insulin secretory capacity in Type 2 diabetes mellitus is associated with beta-cell dedifferentiation and inflammation. We hypothesize that prolonged exposure of beta-cells to low concentrations of IL-1ß induce beta-cell dedifferentiation characterized by impaired glucose-stimulated insulin secretion, reduced expression of key beta-cell genes and changes in histone modifications at gene loci known to affect beta-cell function. Ten days exposure to IL-1ß at non-cytotoxic concentrations reduced insulin secretion and beta-cell proliferation and decreased expression of key beta-cell identity genes, including MafA and Ucn3 and decreased H3K27ac at the gene loci, suggesting that inflammatory cytokines directly affects the epigenome. Following removal of IL-1ß, beta-cell function was normalized and mRNA expression of beta-cell identity genes, such as insulin and Ucn3 returned to pre-stimulation levels. Our findings indicate that prolonged exposure to low concentrations of IL-1ß induces epigenetic changes associated with loss of beta-cell identity as observed in Type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Diabetes Mellitus Tipo 2/patología , Epigénesis Genética , Células Secretoras de Insulina/patología , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Masculino , Ratones , Urocortinas/biosíntesis
2.
Obes Surg ; 26(4): 904-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26781599

RESUMEN

PURPOSE: Sleeve gastrectomy is an effective technique for the treatment of severe obesity, and its effects on the improved ß-cell function have not yet been fully understood. MATERIALS AND METHODS: From February 2014 to July 2015, sleeve gastrectomy was performed in 5 patients with T2D, who were assessed before and after sleeve gastrectomy (SG). Moreover, a high-fat-diet (HFD) mouse model was also used to study the molecular mechanisms of ß-cell functional improvement after SG. RESULTS: The glucose-stimulated acute insulin response was restored in the T2D patients after SG. The expression of GLP-1 in colonic tissue as well as ß-cell specific transcription factors (TFs), Pdx1, and MafA in islets was significantly increased after SG. CONCLUSION: ß-cell dysfunction can be ameliorated by SG. The re-activation of key TFs contributes to the improvement of ß cell function.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Gastrectomía , Proteínas de Homeodominio/biosíntesis , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Obesidad Mórbida/cirugía , Transactivadores/biosíntesis , Anciano , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/cirugía , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/biosíntesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad Mórbida/complicaciones , Obesidad Mórbida/metabolismo , Factores de Transcripción/biosíntesis , Regulación hacia Arriba
3.
Biochem Biophys Res Commun ; 468(4): 629-35, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26546820

RESUMEN

Lack or dysfunction of insulin producing ß cells results in the development of type 1 and type 2 diabetes mellitus, respectively. Insulin secretion is controlled by metabolic stimuli (glucose, fatty acids), but also by monoamine neurotransmitters, like dopamine, serotonin, and norepinephrine. Intracellular monoamine levels are controlled by monoamine oxidases (Mao) A and B. Here we show that MaoA and MaoB are expressed in mouse islet ß cells and that inhibition of Mao activity reduces insulin secretion in response to metabolic stimuli. Moreover, analysis of MaoA and MaoB protein expression in mouse and human type 2 diabetic islets shows a significant reduction of MaoB in type 2 diabetic ß cells suggesting that loss of Mao contributes to ß cell dysfunction. MaoB expression was also reduced in ß cells of MafA-deficient mice, a mouse model for ß cell dysfunction, and biochemical studies showed that MafA directly binds to and activates MaoA and MaoB transcriptional control sequences. Taken together, our results show that MaoA and MaoB expression in pancreatic islets is required for physiological insulin secretion and lost in type 2 diabetic mouse and human ß cells. These findings demonstrate that regulation of monoamine levels by Mao activity in ß cells is pivotal for physiological insulin secretion and that loss of MaoB expression may contribute to the ß cell dysfunction in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Monoaminooxidasa/metabolismo , Animales , Células Cultivadas , Humanos , Secreción de Insulina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Activación Transcripcional
4.
Peptides ; 52: 134-42, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24406898

RESUMEN

Glucagon-like peptide-1 (GLP-1) has been proved to have effects of anti-hyperglycemia and ß-cell preservation. However, it is still unclear whether there are differences between early and late GLP-1 intervention in type 2 diabetes mellitus (T2DM). We divided the mice into 5 groups: early treated group (n=7, 8-week old, fasting glucose>10mmol/l), late treated group (n=7, 10-week old, fasting glucose>20mmol/l), early control group (n=7), late control group (n=7) and wild type group (n=7). Treated group was injected with liraglutide (a GLP-1 analog) 300µg/kg bid for 4 weeks, while control group was given saline at the same time. The results showed that compared with control group, food intake and body weight gain were reduced in both early and late treated group (p<0.05), and there was no significance between the two treated groups. Early liraglutide intervention showed better improvements in glucose control, acute insulin response to glucose (AIRg) and disposition index (before vs. after treatment, AIRg 1.01±0.53 vs. 2.98±0.63, disposition index 10.81±0.89 vs. 27.4±2.15) than late intervention (AIRg 0.99±0.02 vs. 1.41±0.32, disposition index 3.47±0.38 vs. 6.43±1.62, p=0.001). The histopathology of the pancreas showed the estimated ß-cell mass (BCM) was increased more in early treated group than that in late one (0.03 vs. 0.01g). Expressions of the proliferation related genes PDX-1, MafA and GLP-1 receptor (GLP-1R) in early treated group were 1.81, 2.57 and 1.59 times as much as that in late treated group. In conclusion, early liraglutide intervention was better in glucose control, ß-cell function improvement and ß-cell mass preservation.


Asunto(s)
Índice de Masa Corporal , Ingestión de Alimentos/efectos de los fármacos , Péptido 1 Similar al Glucagón/análogos & derivados , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/metabolismo , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica/efectos de los fármacos , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Proteínas de Homeodominio/biosíntesis , Células Secretoras de Insulina/patología , Liraglutida , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Masculino , Ratones , Receptores de Glucagón/biosíntesis , Transactivadores/biosíntesis
5.
Dev Biol ; 385(1): 2-12, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24183936

RESUMEN

Specification and maturation of insulin(+) cells accompanies a transition in expression of Maf family of transcription factors. In development, MafA is expressed after specification of insulin(+) cells that are expressing another Maf factor, MafB; after birth, these insulin(+) MafA(+) cells stop MafB expression and gain glucose responsiveness. Current differentiation protocols for deriving insulin-producing ß-cells from stem cells result in ß-cells lacking both MafA expression and glucose-stimulated insulin secretion. So driving expression of MafA, a ß-cell maturation factor in endocrine precursors could potentially generate glucose-responsive MafA(+) ß cells. Using inducible transgenic mice, we characterized the final stages of ß-cell differentiation and maturation with MafA pause/release experiments. We found that forcing MafA transgene expression, out of its normal developmental context, in Ngn3(+) endocrine progenitors blocked endocrine differentiation and prevented the formation of hormone(+) cells. However, this arrest was reversible such that with stopping the transgene expression, the cells resumed their differentiation to hormone(+) cells, including α-cells, indicating that the block likely occurred after progenitors had committed to a specific hormonal fate. Interestingly, this delayed resumption of endocrine differentiation resulted in a greater proportion of immature insulin(+)MafB(+) cells at P5, demonstrating that during maturation the inhibition of MafB in ß-cell transitioning from insulin(+)MafB(+) to insulin(+)MafB(-) stage is regulated by cell-autonomous mechanisms. These results demonstrate the importance of proper context of initiating MafA expression on the endocrine differentiation and suggest that generating mature Insulin(+)MafA(+) ß-cells will require the induction of MafA in a narrow temporal window to achieve normal endocrine differentiation.


Asunto(s)
Células Endocrinas/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factor de Transcripción MafB/metabolismo , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Células Endocrinas/citología , Regulación del Desarrollo de la Expresión Génica , Insulina/biosíntesis , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Factores de Transcripción Maf de Gran Tamaño/genética , Factor de Transcripción MafB/biosíntesis , Factor de Transcripción MafB/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Páncreas/citología , Páncreas/metabolismo
6.
PLoS One ; 8(11): e79076, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260156

RESUMEN

Surrogate ß-cells derived from stem cells are needed to cure type 1 diabetes, and neonatal liver cells may be an attractive alternative to stem cells for the generation of ß-cells. In this study, we attempted to generate insulin-producing cells from neonatal porcine liver-derived cells using adenoviruses carrying three genes: pancreatic and duodenal homeobox factor1 (PDX1)/VP16, BETA2/NeuroD and v-maf musculo aponeurotic fibrosarcoma oncogene homolog A (MafA), which are all known to play critical roles in pancreatic development. Isolated neonatal porcine liver-derived cells were sequentially transduced with triple adenoviruses and grown in induction medium containing a high concentration of glucose, epidermal growth factors, nicotinamide and a low concentration of serum following the induction of aggregation for further maturation. We noted that the cells displayed a number of molecular characteristics of pancreatic ß-cells, including expressing several transcription factors necessary for ß-cell development and function. In addition, these cells synthesized and physiologically secreted insulin. Transplanting these differentiated cells into streptozotocin-induced immunodeficient diabetic mice led to the reversal of hyperglycemia, and more than 18% of the cells in the grafts expressed insulin at 6 weeks after transplantation. These data suggested that neonatal porcine liver-derived cells can be differentiated into functional insulin-producing cells under the culture conditions presented in this report and indicated that neonatal porcine liver-derived cells (NPLCs) might be useful as a potential source of cells for ß-cell replacement therapy in efforts to cure type I diabetes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Transactivadores/biosíntesis , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Glucosa/genética , Glucosa/metabolismo , Xenoinjertos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/terapia , Insulina/genética , Secreción de Insulina , Hígado , Factores de Transcripción Maf de Gran Tamaño/genética , Ratones , Porcinos , Transactivadores/genética
7.
J Biol Chem ; 288(22): 16155-66, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23595988

RESUMEN

One goal of diabetic regenerative medicine is to instructively convert mature pancreatic exocrine cells into insulin-producing cells. We recently reported that ligand-bound thyroid hormone receptor α (TRα) plays a critical role in expansion of the ß-cell mass during postnatal development. Here, we used an adenovirus vector that expresses TRα driven by the amylase 2 promoter (AdAmy2TRα) to induce the reprogramming of pancreatic acinar cells into insulin-producing cells. Treatment with l-3,5,3-triiodothyronine increases the association of TRα with the p85α subunit of phosphatidylinositol 3-kinase (PI3K), leading to the phosphorylation and activation of Akt and the expression of Pdx1, Ngn3, and MafA in purified acinar cells. Analyses performed with the lectin-associated cell lineage tracing system and the Cre/loxP-based direct cell lineage tracing system indicate that newly synthesized insulin-producing cells originate from elastase-expressing pancreatic acinar cells. Insulin-containing secretory granules were identified in these cells by electron microscopy. The inhibition of p85α expression by siRNA or the inhibition of PI3K by LY294002 prevents the expression of Pdx1, Ngn3, and MafA and the reprogramming to insulin-producing cells. In immunodeficient mice with streptozotocin-induced hyperglycemia, treatment with AdAmy2TRα leads to the reprogramming of pancreatic acinar cells to insulin-producing cells in vivo. Our findings suggest that ligand-bound TRα plays a critical role in ß-cell regeneration during postnatal development via activation of PI3K signaling.


Asunto(s)
Células Acinares/metabolismo , Desdiferenciación Celular , Células Secretoras de Insulina/metabolismo , Receptores de Hormona Tiroidea/biosíntesis , Triyodotironina/farmacología , Células Acinares/citología , Adenoviridae , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/citología , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Factores de Transcripción Maf de Gran Tamaño/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Morfolinas/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , alfa-Amilasas Pancreáticas/genética , alfa-Amilasas Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Hormona Tiroidea/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transactivadores/biosíntesis , Transactivadores/genética , Transducción Genética
8.
Cells Tissues Organs ; 197(4): 269-82, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23306211

RESUMEN

Insulin therapy for type 1 diabetes does not prevent serious long-term complications including vascular disease, neuropathy, retinopathy and renal failure. Stem cells, including amniotic fluid-derived stem (AFS) cells - highly expansive, multipotent and nontumorigenic cells - could serve as an appropriate stem cell source for ß-cell differentiation. In the current study we tested whether nonhuman primate (nhp)AFS cells ectopically expressing key pancreatic transcription factors were capable of differentiating into a ß-cell-like cell phenotype in vitro. nhpAFS cells were obtained from Cynomolgus monkey amniotic fluid by immunomagnetic selection for a CD117 (c-kit)-positive population. RT-PCR for endodermal and pancreatic lineage-specific markers was performed on AFS cells after adenovirally transduced expression of PDX1, NGN3 and MAFA. Expression of MAFA was sufficient to induce insulin mRNA expression in nhpAFS cell lines, whereas a combination of MAFA, PDX1 and NGN3 further induced insulin expression, and also induced the expression of other important endocrine cell genes such as glucagon, NEUROD1, NKX2.2, ISL1 and PCSK2. Higher induction of these and other important pancreatic genes was achieved by growing the triply infected AFS cells in media supplemented with a combination of B27, betacellulin and nicotinamide, as well as culturing the cells on extracellular matrix-coated plates. The expression of pancreatic genes such as NEUROD1, glucagon and insulin progressively decreased with the decline of adenovirally expressed PDX1, NGN3 and MAFA. Together, these experiments suggest that forced expression of pancreatic transcription factors in primate AFS cells induces them towards the pancreatic lineage.


Asunto(s)
Líquido Amniótico/fisiología , Páncreas/fisiología , Células Madre/fisiología , Líquido Amniótico/citología , Líquido Amniótico/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Expresión Génica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Macaca fascicularis , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Factores de Transcripción Maf de Gran Tamaño/genética , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares , Páncreas/citología , Páncreas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Transactivadores/biosíntesis , Transactivadores/genética , Factores de Transcripción
9.
Cell Transplant ; 22(1): 147-58, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22776709

RESUMEN

The use of pancreatic ß-cells differentiated from embryonic stem (ES) cells or induced pluripotent stem (iPS) cells is a promising strategy in cell therapy. Pancreatic ß-cell development is regulated by the sequential expression of a molecular network of transcription factors. In this experiment, we adopted a three-step differentiation protocol to differentiate mES (mouse ES) cells into insulin-secreting cells and overexpressed transcription factors by adenoviral vectors at various combinations at different time of differentiation. We found that the coexpression of Pdx1 and MafA with either Ngn3 or NeuroD, especially at the final stage of the three-step differentiation, significantly increased the differentiation efficiency. It also increased the glucose-stimulated insulin and C-peptide secretion in insulin-secreting cells derived from mES cells compared to the control green fluorescent protein (GFP) vector-transduced group. For the first time, we have demonstrated that the coexpression of Pdx1 and MafA during a specific time window of development can act synergistically with either Ngn3 or NeuroD to promote the differentiation of mES cells into insulin-secreting cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/biosíntesis , Células Secretoras de Insulina/fisiología , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Transactivadores/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/genética , Insulina/biosíntesis , Insulina/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Ratones , Proteínas del Tejido Nervioso/genética , Transactivadores/genética , Transcripción Genética
10.
Dev Neurobiol ; 70(7): 485-97, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20213756

RESUMEN

Dorsal root ganglia proceed from the coalescence of cell bodies of sensory neurons, which have migrated dorsoventrally from the delaminating neural crest. They are composed of different neuronal subtypes with specific sensory functions, including nociception, thermal sensation, proprioception, and mechanosensation. In contrast to proprioceptors and thermonociceptors, little is known about the molecular mechanisms governing the early commitment and later differentiation into mechanosensitive neurons. This is mainly due to the absence of specific molecular markers for this particular cell type. Using knockout mice, we identified the bZIP transcription factor MafA as the first specific marker of a subpopulation of "early c-ret" positive neurons characterized by medium-to-large diameters. This marker will allow further functional characterization of these neurons.


Asunto(s)
Ganglios Espinales/embriología , Factores de Transcripción Maf de Gran Tamaño/genética , Mecanorreceptores/metabolismo , Cresta Neural/embriología , Proteínas Proto-Oncogénicas c-ret/biosíntesis , Células Receptoras Sensoriales/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Tamaño de la Célula , Ganglios Espinales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Marcadores Genéticos/genética , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Mecanorreceptores/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutagénesis Insercional , Cresta Neural/citología , Proteínas Proto-Oncogénicas c-ret/genética , Células Receptoras Sensoriales/citología
11.
Dev Biol ; 333(1): 108-20, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19576197

RESUMEN

The transcription factor MafA regulates glucose-responsive expression of insulin. MafA-deficient mice have a normal proportion of insulin+ cells at birth but develop diabetes gradually with age, suggesting that MafA is required for maturation and not specification of pancreatic beta-cells. However, several studies show that ectopic expression of MafA may have a role in specification as it induces insulin+ cells in chicken gut epithelium, reprograms adult murine acinar cells into insulin+ cells in combination with Ngn3 and Pdx1, and triggers the lens differentiation. Hence, we examined whether MafA can induce specification of beta-cells during pancreatic development. When the MafA transgene is expressed in Pdx1+ pancreatic progenitors, both pancreatic mass and proliferation of progenitors are reduced, at least partially due to induction of cyclin kinase inhibitors p27 and p57. Expression of MafA in Pdx1+ cells until E12.5 was sufficient to cause these effects and to disproportionately inhibit the formation of endocrine cells in the remnant pancreas. Thus, in mice, MafA expression in Pdx1+ pancreatic progenitors is not sufficient to specify insulin+ cells but in fact deters pancreatic development and the differentiation of endocrine cells. These findings imply that MafA should be used to enhance maturation, rather than specification, of beta-cells from stem/progenitor cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Endocrinas/citología , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Páncreas/metabolismo , Células Madre/citología , Animales , Proliferación Celular , Células Endocrinas/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Ratones , Ratones Transgénicos , Páncreas/embriología , Células Madre/metabolismo
12.
Arch Biochem Biophys ; 480(2): 138-42, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18948074

RESUMEN

MafA is a basic leucine zipper transcription factor expressed within the beta cells of the pancreas and is required to maintain normal glucose homeostasis as it is involved in various aspects of beta cell biology. MafA protein levels are known to increase in response to high glucose through mechanisms that have yet to be fully characterized. We investigated whether discrete intracellular signaling events control mafA expression. We found that the general kinase inhibitor staurosporine induces mafA expression without altering the stability of the protein. Inhibition of the MAP-kinase JNK mimics the effects of staurosporine on the expression of mafA. Calmodulin kinase and calcium signaling are also important in stimulating mafA expression by high glucose. However, staurosporine, JNK, and calmodulin kinase have different effects on the induction of insulin expression. These data reveal that MafA levels are tightly controlled by the coordinated action of multiple kinase pathways.


Asunto(s)
Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Factores de Transcripción Maf de Gran Tamaño/fisiología , Calcio/metabolismo , Línea Celular , Diabetes Mellitus/metabolismo , Expresión Génica , Glucosa/metabolismo , Humanos , Insulina/metabolismo , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Modelos Biológicos , Transducción de Señal , Estaurosporina/farmacología , Transcripción Genética
13.
Biochem Biophys Res Commun ; 354(2): 334-9, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17239820

RESUMEN

Transcription factors, such as PDX-1, that normally mediate pancreatic development are capable of inducing hepatic progenitor cells to differentiate into cells with pancreatic islet characteristics. We hypothesized that simultaneous expression of multiple transcription factors involved in islet development might enhance the differentiation of hepatic progenitor cells. Bi- or tri-cistronic constructs were generated in hybrid adenovirus/adeno-associated virus (Ad/AAV) vectors containing neurogenin 3 (NGN3), BETA2 (NeuroD), and RIPE3b1 (MafA), each of which plays a role in islet cell differentiation. These vectors efficiently express multiple transcription factors and stimulate insulin promoter activity in a combinatorial manner. When these multi-cistronic constructs were administered in vivo, they induce hepatic expression of islet-specific markers, including PDX-1, insulin, glucagon, somatostatin, and islet-amyloid peptide. Administration of the Ad/AAV hybrid vectors to streptozotocin-induced diabetic mice reversed hyperglycemia, consistent the differentiation of functional hepatic insulin-secreting cells. These results indicate that Ad/AAV hybrid vectors can be used to administer combinations of factors that induce islet cell differentiation in hepatic progenitor cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular/genética , Islotes Pancreáticos/citología , Hígado/citología , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Línea Celular , Regulación de la Expresión Génica/fisiología , Insulina/biosíntesis , Insulina/genética , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/fisiología , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología
14.
J Biol Chem ; 282(3): 1577-84, 2007 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-17142462

RESUMEN

MafA is a basic leucine zipper transcription factor that regulates gene expression in both the neuroretina and pancreas. Within the pancreas, MafA is exclusively expressed in the beta cells and is involved in insulin gene transcription, insulin secretion, and beta cell survival. The expression of the mafA gene within beta cells is known to increase in response to high glucose levels by an unknown mechanism. In this study, we demonstrate that pyruvate, which is produced by glycolysis from glucose, is not sufficient to induce mafA gene expression compared with high glucose. This suggests that the signal for MafA induction is independent of ATP levels and that a metabolic event occurring upstream of pyruvate production leads to the induction of MafA. Furthermore, insulin secretion mediated by high glucose is not important for MafA expression. However, the addition of glucosamine to beta cell lines stimulates MafA expression in the absence of high glucose, and inhibition of the hexosamine biosynthetic pathway in the presence of high glucose abolishes MafA induction. Moreover, we demonstrate that the expression of UDP-N-acetylglucosaminyl transferase, the enzyme mediating O-linked glycosylation of cytosolic and nuclear proteins, is essential for glucose-dependent MafA expression. Consistent with this observation, inhibition of N-acetylglucosaminidase, the enzyme involved in the removal of the O-GlcNAc modification from proteins, with O-(2-acetamido-2-deoxy-d-glucopyranosylidene)amino-N-phenylcarbamate stimulates MafA expression under low glucose conditions. The presented data suggest that MafA expression mediated by high glucose requires flux through the hexosamine biosynthetic pathway and the O-linked glycosylation of an unknown protein(s) by UDP-N-acetylglucosaminyl transferase.


Asunto(s)
Regulación de la Expresión Génica , Glucosa/metabolismo , Hexosaminas/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/biosíntesis , Animales , Núcleo Celular/metabolismo , Supervivencia Celular , Citosol/metabolismo , Glicosilación , Insulina/metabolismo , Ratones , Modelos Biológicos , N-Acetilglucosaminiltransferasas/metabolismo , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...