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










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119235, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35151663

RESUMEN

Glucose homeostasis is maintained by hormones secreted from different types of pancreatic islets and its dysregulation can result in diseases including diabetes mellitus. The secretion of hormones from pancreatic islets is highly complex and tightly controlled by G protein-coupled receptors (GPCRs). Moreover, GPCR signaling may play a role in enhancing islet cell replication and proliferation. Thus, targeting GPCRs offers a promising strategy for regulating the functionality of pancreatic islets. Here, available RNAseq datasets from human and mouse islets were used to identify the GPCR expression profile and the impact of GPCR signaling for normal islet functionality is discussed.


Asunto(s)
Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/crecimiento & desarrollo , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Transcriptoma
2.
Nat Commun ; 12(1): 4458, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294685

RESUMEN

The cellular identity of pancreatic polypeptide (Ppy)-expressing γ-cells, one of the rarest pancreatic islet cell-type, remains elusive. Within islets, glucagon and somatostatin, released respectively from α- and δ-cells, modulate the secretion of insulin by ß-cells. Dysregulation of insulin production raises blood glucose levels, leading to diabetes onset. Here, we present the genetic signature of human and mouse γ-cells. Using different approaches, we identified a set of genes and pathways defining their functional identity. We found that the γ-cell population is heterogeneous, with subsets of cells producing another hormone in addition to Ppy. These bihormonal cells share identity markers typical of the other islet cell-types. In mice, Ppy gene inactivation or conditional γ-cell ablation did not alter glycemia nor body weight. Interestingly, upon ß-cell injury induction, γ-cells exhibited gene expression changes and some of them engaged insulin production, like α- and δ-cells. In conclusion, we provide a comprehensive characterization of γ-cells and highlight their plasticity and therapeutic potential.


Asunto(s)
Insulina/biosíntesis , Células Secretoras de Polipéptido Pancreático/metabolismo , Polipéptido Pancreático/metabolismo , Precursores de Proteínas/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal , Linaje de la Célula/genética , Femenino , Técnicas de Sustitución del Gen , Humanos , Células Secretoras de Insulina/clasificación , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Páncreas/citología , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Polipéptido Pancreático/deficiencia , Polipéptido Pancreático/genética , Células Secretoras de Polipéptido Pancreático/clasificación , Células Secretoras de Polipéptido Pancreático/citología , Embarazo , RNA-Seq
3.
PLoS One ; 15(3): e0230627, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32208453

RESUMEN

The gene encoding eukaryotic initiation factor 5A (EIF5A) is found in diabetes-susceptibility loci in mouse and human. eIF5A is the only protein known to contain hypusine (hydroxyputrescine lysine), a polyamine-derived amino acid formed post-translationally in a reaction catalyzed by deoxyhypusine synthase (DHPS). Previous studies showed pharmacologic blockade of DHPS in type 1 diabetic NOD mice and type 2 diabetic db/db mice improved glucose tolerance and preserved beta cell mass, which suggests that hypusinated eIF5A (eIF5AHyp) may play a role in diabetes pathogenesis by direct action on the beta cells and/or altering the adaptive or innate immune responses. To translate these findings to human, we examined tissue from individuals with and without type 1 and type 2 diabetes to determine the expression of eIF5AHyp. We detected eIF5AHyp in beta cells, exocrine cells and immune cells; however, there was also unexpected enrichment of eIF5AHyp in pancreatic polypeptide-expressing PP cells. Interestingly, the presence of eIF5AHyp co-expressing PP cells was not enhanced with disease. These data identify new aspects of eIF5A biology and highlight the need to examine human tissue to understand disease.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/patología , Lisina/análogos & derivados , Páncreas/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Bazo/metabolismo , Adulto , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/genética , Adulto Joven , Factor 5A Eucariótico de Iniciación de Traducción
4.
Nature ; 567(7746): 43-48, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30760930

RESUMEN

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Asunto(s)
Diabetes Mellitus/patología , Diabetes Mellitus/terapia , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/patología , Animales , Biomarcadores/análisis , Linaje de la Célula/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad , Femenino , Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/trasplante , Glucosa/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Ratones , Especificidad de Órganos/efectos de los fármacos , Polipéptido Pancreático/metabolismo , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/efectos de los fármacos , Células Secretoras de Polipéptido Pancreático/metabolismo , Proteómica , Análisis de Secuencia de ARN , Transactivadores/genética , Transactivadores/metabolismo , Transcriptoma , Transducción Genética
5.
Anat Histol Embryol ; 47(2): 159-166, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29315753

RESUMEN

Immunohistochemical techniques were employed to investigate the distribution of amylin-like immunoreactive cells in the pancreas of gecko Homopholis fasciata. Four types of endocrine cells were distinguished: insulin immunoreactive (B cells), pancreatic polypeptide immunoreactive (PP cells), glucagon and pancreatic polypeptide immunoreactive (A/PP cells) and somatostatin immunoreactive cells (D cells). Pancreatic islets contained B, A/PP and D cells, whereas extrainsular regions contained B, D and PP cells. In the pancreatic islets, amylin-like immunoreactive cells corresponded to B cells, but not to A/PP or D cells. In the extrainsular regions, amylin-like immunoreactive cells corresponded to either B or PP cells. Amylin secreted from intrainsular B cells may regulate pancreatic hormone secretion in an autocrine and/or a paracrine fashion. On the other hand, amylin secreted from extrainsular PP and B cells, and/or intrainsular B cells may participate in the modulation of calcium homoeostasis in an endocrine fashion.


Asunto(s)
Linfocitos B/citología , Células Endocrinas/clasificación , Células Secretoras de Glucagón/citología , Inmunohistoquímica/veterinaria , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Células Secretoras de Polipéptido Pancreático/citología , Animales , Células Endocrinas/metabolismo , Glucagón/inmunología , Glucagón/metabolismo , Insulina/inmunología , Insulina/metabolismo , Secreción de Insulina , Polipéptido Amiloide de los Islotes Pancreáticos/inmunología , Lagartos , Somatostatina/inmunología , Somatostatina/metabolismo
6.
Diabetes Obes Metab ; 19 Suppl 1: 124-136, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28880471

RESUMEN

The progressive loss of pancreatic ß-cell mass that occurs in both type 1 and type 2 diabetes is a primary factor driving efforts to identify strategies for effectively increasing, enhancing or restoring ß-cell mass. While factors that seem to influence ß-cell proliferation in specific contexts have been described, reliable stimulation of human ß-cell proliferation has remained a challenge. Importantly, ß-cells exist in the context of a complex, integrated pancreatic islet microenvironment where they interact with other endocrine cells, vascular endothelial cells, extracellular matrix, neuronal projections and islet macrophages. This review highlights different components of the pancreatic microenvironment, and reviews what is known about how signaling that occurs between ß-cells and these other components influences ß-cell proliferation. Future efforts to further define the role of the pancreatic islet microenvironment on ß-cell proliferation may lead to the development of successful approaches to increase or restore ß-cell mass in diabetes.


Asunto(s)
Comunicación Celular , Proliferación Celular , Microambiente Celular , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Modelos Biológicos , Animales , Apoptosis , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/inmunología , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/inervación , Islotes Pancreáticos/patología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/inmunología , Células Secretoras de Polipéptido Pancreático/metabolismo , Células Secretoras de Polipéptido Pancreático/patología , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/inmunología , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/patología , Especificidad de la Especie
7.
Sci Rep ; 7(1): 2505, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28566744

RESUMEN

Insulin-producing cells (IPCs) derived from a patient's own stem cells offer great potential for autologous transplantation in diabetic patients. However, the limited survival of engrafted cells remains a bottleneck in the application of this strategy. The present study aimed to investigate whether nanoparticle-based magnetic resonance (MR) tracking can be used to detect the loss of grafted stem cell-derived IPCs in a sensitive and timely manner in a diabetic monkey model. Pancreatic progenitor cells (PPCs) were isolated from diabetic monkeys and labeled with superparamagnetic iron oxide nanoparticles (SPIONs). The SPION-labeled cells presented as hypointense signals on MR imaging (MRI). The labeling procedure did not affect the viability or IPC differentiation of PPCs. Importantly, the total area of the hypointense signal caused by SPION-labeled IPCs on liver MRI decreased before the decline in C-peptide levels after autotransplantation. Histological analysis revealed no detectable immune response to the grafts and many surviving insulin- and Prussian blue-positive cell clusters on liver sections at one year post-transplantation. Collectively, this study demonstrates that SPIO nanoparticles can be used to label stem cells for noninvasive, sensitive, longitudinal monitoring of stem cell-derived IPCs in large animal models using a conventional MR imager.


Asunto(s)
Rastreo Celular/métodos , Diabetes Mellitus Tipo 1/diagnóstico por imagen , Diabetes Mellitus Tipo 1/terapia , Imagen por Resonancia Magnética , Células Secretoras de Polipéptido Pancreático/citología , Animales , Péptido C/sangre , Diferenciación Celular , Medios de Contraste/administración & dosificación , Medios de Contraste/química , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Compuestos Férricos/administración & dosificación , Compuestos Férricos/química , Humanos , Macaca fascicularis , Nanopartículas de Magnetita/administración & dosificación , Nanopartículas de Magnetita/química , Trasplante de Células Madre Mesenquimatosas/métodos , Células Secretoras de Polipéptido Pancreático/efectos de los fármacos , Trasplante Autólogo
8.
Diabetes Obes Metab ; 18 Suppl 1: 10-22, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27615127

RESUMEN

During embryonic development, endocrine cells of the pancreas are specified from multipotent progenitors. The transcription factor Neurogenin 3 (NEUROG3) is critical for this development and it has been shown that all endocrine cells of the pancreas arise from endocrine progenitors expressing NEUROG3. A thorough understanding of the role of NEUROG3 during development, directed differentiation of pluripotent stem cells and in models of cellular reprogramming, will guide future efforts directed at finding novel sources of ß-cells for cell replacement therapies. In this article, we review the expression and function of NEUROG3 in both mouse and human and present the further characterization of a monoclonal antibody directed against NEUROG3. This antibody has been previously been used for detection of both mouse and human NEUROG3. However, our results suggest that the epitope recognized by this antibody is specific to mouse NEUROG3. Thus, we have also generated a monoclonal antibody specifically recognizing human NEUROG3 and present the characterization of this antibody here. Together, these antibodies will provide useful tools for future studies of NEUROG3 expression, and the data presented in this article suggest that recently described expression patterns of NEUROG3 in human foetal and adult pancreas should be re-examined.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso/genética , Animales , Anticuerpos Monoclonales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Reprogramación Celular , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo
9.
J Endocrinol ; 229(2): 123-32, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26931137

RESUMEN

The aim of this study was to evaluate the location of PP and δ cells in relation to the vascularization within human pancreatic islets. To this end, pancreas sections were analysed by immunofluorescence using antibodies against endocrine islet and endothelial cells. Staining in different islet areas corresponding to islet cells adjacent or not to peripheral or central vascular channels was quantified by computerized morphometry. As results, α, PP and δ cells were preferentially found adjacent to vessels. In contrast to α cells, which were evenly distributed between islet periphery and intraislet vascular channels, PP and δ cells had asymmetric and opposite distributions: PP staining was higher and somatostatin staining was lower in the islet periphery than in the area around intraislet vascular channels. Additionally, frequencies of PP and δ cells were negatively correlated in the islets. No difference was observed between islets from the head and the tail of the pancreas, and from type 2 diabetic and non-diabetic donors. In conclusion, the distribution of δ cells differs from that of PP cells in human islets, suggesting that vessels at the periphery and at the centre of islets drain different hormonal cocktails.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo , Adolescente , Adulto , Anciano , Técnica del Anticuerpo Fluorescente , Humanos , Persona de Mediana Edad , Polipéptido Pancreático/metabolismo , Somatostatina/metabolismo , Distribución Tisular , Adulto Joven
10.
PLoS One ; 10(12): e0144597, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26658466

RESUMEN

The transcription factor Pax6 is an important regulator of development and cell differentiation in various organs. Thus, Pax6 was shown to promote neural development in the cerebral cortex and spinal cord, and to control pancreatic endocrine cell genesis. However, the role of Pax6 in distinct endocrine cells of the adult pancreas has not been addressed. We report the conditional inactivation of Pax6 in insulin and glucagon producing cells of the adult mouse pancreas. In the absence of Pax6, beta- and alpha-cells lose their molecular maturation characteristics. Our findings provide strong evidence that Pax6 is responsible for the maturation of beta-, and alpha-cells, but not of delta-, and PP-cells. Moreover, lineage-tracing experiments demonstrate that Pax6-deficient beta- and alpha-cells are shunted towards ghrelin marked cells, sustaining the idea that ghrelin may represent a marker for endocrine cell maturation.


Asunto(s)
Proteínas del Ojo/genética , Ghrelina/genética , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Paired Box/genética , Células Secretoras de Polipéptido Pancreático/metabolismo , Proteínas Represoras/genética , Células Secretoras de Somatostatina/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Diferenciación Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Cruzamientos Genéticos , Proteínas del Ojo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Ghrelina/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Integrasas/genética , Integrasas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/efectos de los fármacos , Proteínas Represoras/metabolismo , Transducción de Señal , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/efectos de los fármacos , Tamoxifeno/farmacología
11.
Islets ; 5(5): 226-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24045229

RESUMEN

The large size of the human pancreas challenges unbiased quantitative analyses that require a practical stereological approach. While many histological studies of the pancreas in the past lacked regional information, we have shown marked heterogeneity within an individual, where islet distribution/density is relatively low in the head and gradually increases through the body toward the tail region by>2-fold. Studies focusing on the tail region may be prone to overestimation of ß-cell/islet mass when normalizing measured values per person by using pancreas weight or volume. In this article, beyond technical issues, we discuss the pathophysiological importance of studying the head region of the human pancreas regarding its unique characteristics in early development, and the anatomical disposition that may lead to a preferential loss of ß-cells in patients with type 2 diabetes and the development of pancreatic cancer.


Asunto(s)
Páncreas/citología , Células Secretoras de Polipéptido Pancreático/citología , Humanos
12.
PLoS One ; 8(1): e55501, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383206

RESUMEN

The pancreatic islet is mainly composed of beta-, alpha- and delta-cells with small numbers of pancreatic polypeptide (PP) and epsilon cells. It is known that there is a region in the head of the pancreas that is rich in PP-cells. In the present study, we examined the distribution of PP-cells, and assessed the influence of the PP-cell rich region to quantify the total islet mass. Pancreatic tissues were collected from donors with no history of diabetes or pancreatic diseases (n = 12). A stereological approach with a computer-assisted large-scale analysis of whole pancreatic sections was applied to quantify the entire distribution of endocrine cells within a given section. The initial whole pancreas analysis showed that a PP-cell rich region was largely restricted to the uncinate process with a clear boundary. The distinct distribution of PP-cells includes irregularly shaped clusters composed solely of PP-cells. Furthermore, in the PP-cell rich region, beta- and alpha-cell mass is significantly reduced compared to surrounding PP-cell poor regions. The results suggest that the analysis of the head region should distinguish the PP-cell rich region, which is best examined separately. This study presents an important implication for the regional selection and interpretation of the results.


Asunto(s)
Páncreas/citología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Glucagón/citología , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/citología , Islotes Pancreáticos , Páncreas/anatomía & histología , Páncreas/metabolismo , Células Secretoras de Polipéptido Pancreático/metabolismo
13.
Development ; 139(1): 33-45, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22096075

RESUMEN

Neurog3-induced Dll1 expression in pancreatic endocrine progenitors ostensibly activates Hes1 expression via Notch and thereby represses Neurog3 and endocrine differentiation in neighboring cells by lateral inhibition. Here we show in mouse that Dll1 and Hes1 expression deviate during regionalization of early endoderm, and later during early pancreas morphogenesis. At that time, Ptf1a activates Dll1 in multipotent pancreatic progenitor cells (MPCs), and Hes1 expression becomes Dll1 dependent over a brief time window. Moreover, Dll1, Hes1 and Dll1/Hes1 mutant phenotypes diverge during organ regionalization, become congruent at early bud stages, and then diverge again at late bud stages. Persistent pancreatic hypoplasia in Dll1 mutants after eliminating Neurog3 expression and endocrine development, together with reduced proliferation of MPCs in both Dll1 and Hes1 mutants, reveals that the hypoplasia is caused by a growth defect rather than by progenitor depletion. Unexpectedly, we find that Hes1 is required to sustain Ptf1a expression, and in turn Dll1 expression in early MPCs. Our results show that Ptf1a-induced Dll1 expression stimulates MPC proliferation and pancreatic growth by maintaining Hes1 expression and Ptf1a protein levels.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Páncreas/embriología , Células Secretoras de Polipéptido Pancreático/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina , Proteínas de Unión al Calcio , Inmunoprecipitación de Cromatina , Galactósidos , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Indoles , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Células Madre/citología , Factor de Transcripción HES-1
14.
Mol Endocrinol ; 24(8): 1605-14, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20592160

RESUMEN

The major role of glucagon is to promote hepatic gluconeogenesis and glycogenolysis to raise blood glucose levels during hypoglycemic conditions. Several animal models have been established to examine the in vivo function of glucagon in the liver through attenuation of glucagon via glucagon receptor knockout animals and pharmacological interventions. To investigate the consequences of glucagon loss to hepatic glucose production and glucose homeostasis, we derived mice with a pancreas specific ablation of the alpha-cell transcription factor, Arx, resulting in a complete loss of the glucagon-producing pancreatic alpha-cell. Using this model, we found that glucagon is not required for the general health of mice but is essential for total hepatic glucose production. Our data clarifies the importance of glucagon during the regulation of fasting and postprandial glucose homeostasis.


Asunto(s)
Glucemia/metabolismo , Células Secretoras de Glucagón/citología , Glucagón/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Western Blotting , Glucagón/deficiencia , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Mutantes , Células Secretoras de Polipéptido Pancreático/citología , Reacción en Cadena de la Polimerasa , Células Secretoras de Somatostatina/citología , Factores de Transcripción/genética
15.
Mol Cell Biol ; 28(20): 6373-83, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18710955

RESUMEN

During pancreas development, transcription factors play critical roles in exocrine and endocrine differentiation. Transcriptional regulation in eukaryotes occurs within chromatin and is influenced by posttranslational histone modifications (e.g., acetylation) involving histone deacetylases (HDACs). Here, we show that HDAC expression and activity are developmentally regulated in the embryonic rat pancreas. We discovered that pancreatic treatment with different HDAC inhibitors (HDACi) modified the timing and determination of pancreatic cell fate. HDACi modified the exocrine lineage via abolition and enhancement of acinar and ductal differentiation, respectively. Importantly, HDACi treatment promoted the NGN3 proendocrine lineage, leading to an increased pool of endocrine progenitors and modified endocrine subtype lineage choices. Interestingly, treatments with trichostatin A and sodium butyrate, two inhibitors of both class I and class II HDACs, enhanced the pool of beta cells. These results highlight the roles of HDACs at key points in exocrine and endocrine differentiation. They show the powerful use of HDACi to switch pancreatic cell determination and amplify specific cellular subtypes, with potential applications in cell replacement therapies in diabetes.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Islotes Pancreáticos/citología , Páncreas/efectos de los fármacos , Páncreas/enzimología , Células Madre/citología , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Ácidos Hidroxámicos/farmacología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/enzimología , Proteínas del Tejido Nervioso/metabolismo , Páncreas/citología , Páncreas/embriología , Páncreas Exocrino/citología , Páncreas Exocrino/efectos de los fármacos , Páncreas Exocrino/enzimología , Conductos Pancreáticos/citología , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/enzimología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/efectos de los fármacos , Células Secretoras de Polipéptido Pancreático/enzimología , Ratas , Ratas Wistar , Células Madre/efectos de los fármacos , Ácido Valproico/farmacología
16.
Lab Invest ; 88(7): 761-72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18490899

RESUMEN

Cultured human islets can be dedifferentiated to duct-like structures composed mainly of cytokeratin+ and nestin+ cells. Given that these structures possess the potential to redifferentiate into islet-like structures, we sought to elucidate their specific cellular origins. Adenoviral vectors were engineered for beta-, alpha-, delta- or PP-cell-specific GFP expression. A double-stranded system was designed whereby cultures were infected with two vectors: one expressed GFP behind the cumate-inducible promoter sequence, and the other expressed the requisite transactivator behind the human insulin, glucagon, somatostatin or pancreatic polypeptide promoter. This system labels hormone+ cells in the islet in a cell-specific manner, allowing these cells to be tracked during the course of transformation from islet to duct-like structure. Post-infection, islets were cultured to induce dedifferentiation. Fluorescence microscopy demonstrated that alpha-, delta- and PP-cells contributed equally to the cytokeratin+ population, with minimal beta-cell contribution, whereas the converse was true for nestin+ cells. Complementary targeted cell ablation studies, using streptozotocin or similar adenoviral expression of the Bax (Bcl2-associated X protein) toxigene, validated these findings and suggested a redundancy between alpha-, delta- and PP-cells with respect to cytokeratin+ cell derivation. These results call into question the traditional understanding of islet cells as being terminally differentiated and provide support for the concept of adult islet morphogenetic plasticity.


Asunto(s)
Islotes Pancreáticos/citología , Adenoviridae/genética , Adulto , Diferenciación Celular , Células Cultivadas , Vectores Genéticos , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Islotes Pancreáticos/metabolismo , Queratinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nestina , Conductos Pancreáticos/citología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Regiones Promotoras Genéticas , Somatostatina/fisiología , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo , Células Madre/citología , Estreptozocina/farmacología , Proteína X Asociada a bcl-2/biosíntesis , Proteína X Asociada a bcl-2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...