Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 6344, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39068220

RESUMO

Dysfunction of pancreatic δ cells contributes to the etiology of diabetes. Despite their important role, human δ cells are scarce, limiting physiological studies and drug discovery targeting δ cells. To date, no directed δ-cell differentiation method has been established. Here, we demonstrate that fibroblast growth factor (FGF) 7 promotes pancreatic endoderm/progenitor differentiation, whereas FGF2 biases cells towards the pancreatic δ-cell lineage via FGF receptor 1. We develop a differentiation method to generate δ cells from human stem cells by combining FGF2 with FGF7, which synergistically directs pancreatic lineage differentiation and modulates the expression of transcription factors and SST activators during endoderm/endocrine precursor induction. These δ cells display mature RNA profiles and fine secretory granules, secrete somatostatin in response to various stimuli, and suppress insulin secretion from in vitro co-cultured ß cells and mouse ß cells upon transplantation. The generation of human pancreatic δ cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation studies in diabetes.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes , Humanos , Animais , Camundongos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/citologia , Endoderma/citologia , Endoderma/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Pâncreas/citologia , Pâncreas/metabolismo , Somatostatina/metabolismo , Linhagem da Célula , Insulina/metabolismo , Secreção de Insulina
2.
Nat Commun ; 10(1): 3700, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420552

RESUMO

Little is known about the role of islet delta cells in regulating blood glucose homeostasis in vivo. Delta cells are important paracrine regulators of beta cell and alpha cell secretory activity, however the structural basis underlying this regulation has yet to be determined. Most delta cells are elongated and have a well-defined cell soma and a filopodia-like structure. Using in vivo optogenetics and high-speed Ca2+ imaging, we show that these filopodia are dynamic structures that contain a secretory machinery, enabling the delta cell to reach a large number of beta cells within the islet. This provides for efficient regulation of beta cell activity and is modulated by endogenous IGF-1/VEGF-A signaling. In pre-diabetes, delta cells undergo morphological changes that may be a compensation to maintain paracrine regulation of the beta cell. Our data provides an integrated picture of how delta cells can modulate beta cell activity under physiological conditions.


Assuntos
Ilhotas Pancreáticas/ultraestrutura , Comunicação Parácrina , Estado Pré-Diabético/patologia , Pseudópodes/ultraestrutura , Células Secretoras de Somatostatina/ultraestrutura , Animais , Glicemia/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Microscopia Intravital , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Imagem Óptica , Optogenética , Estado Pré-Diabético/metabolismo , Pseudópodes/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Diabetes Obes Metab ; 19 Suppl 1: 124-136, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28880471

RESUMO

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.


Assuntos
Comunicação Celular , Proliferação de Células , Microambiente Celular , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Modelos Biológicos , Animais , Apoptose , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/imunologia , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/inervação , Ilhotas Pancreáticas/patologia , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/imunologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Células Secretoras de Polipeptídeo Pancreático/patologia , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/imunologia , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/patologia , Especificidade da Espécie
4.
J Clin Invest ; 127(7): 2631-2646, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28604389

RESUMO

Somatostatin secreted by pancreatic δ cells mediates important paracrine interactions in Langerhans islets, including maintenance of glucose metabolism through the control of reciprocal insulin and glucagon secretion. Disruption of this circuit contributes to the development of diabetes. However, the precise mechanisms that control somatostatin secretion from islets remain elusive. Here, we found that a super-complex comprising the cullin 4B-RING E3 ligase (CRL4B) and polycomb repressive complex 2 (PRC2) epigenetically regulates somatostatin secretion in islets. Constitutive ablation of CUL4B, the core component of the CRL4B-PRC2 complex, in δ cells impaired glucose tolerance and decreased insulin secretion through enhanced somatostatin release. Moreover, mechanistic studies showed that the CRL4B-PRC2 complex, under the control of the δ cell-specific transcription factor hematopoietically expressed homeobox (HHEX), determines the levels of intracellular calcium and cAMP through histone posttranslational modifications, thereby altering expression of the Cav1.2 calcium channel and adenylyl cyclase 6 (AC6) and modulating somatostatin secretion. In response to high glucose levels or urocortin 3 (UCN3) stimulation, increased expression of cullin 4B (CUL4B) and the PRC2 subunit histone-lysine N-methyltransferase EZH2 and reciprocal decreases in Cav1.2 and AC6 expression were found to regulate somatostatin secretion. Our results reveal an epigenetic regulatory mechanism of δ cell paracrine interactions in which CRL4B-PRC2 complexes, Cav1.2, and AC6 expression fine-tune somatostatin secretion and facilitate glucose homeostasis in pancreatic islets.


Assuntos
Proteínas Culina/metabolismo , Insulina/metabolismo , Complexos Multienzimáticos/metabolismo , Comunicação Parácrina , Células Secretoras de Somatostatina/metabolismo , Somatostatina/metabolismo , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Proteínas Culina/genética , AMP Cíclico/metabolismo , Epigênese Genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Insulina/genética , Secreção de Insulina , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/genética , Somatostatina/genética , Células Secretoras de Somatostatina/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Sci Rep ; 7(1): 90, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273890

RESUMO

Glucagon is the main counterregulatory hormone in the body. Still, the mechanism involved in the regulation of glucagon secretion from pancreatic alpha cells remains elusive. Dysregulated glucagon secretion is common in patients with Cystic Fibrosis (CF) that develop CF related diabetes (CFRD). CF is caused by a mutation in the Cl- channel Cystic fibrosis transmembrane conductance regulator (CFTR), but whether CFTR is present in human alpha cells and regulate glucagon secretion has not been investigated in detail. Here, both human and mouse alpha cells showed CFTR protein expression, whereas CFTR was absent in somatostatin secreting delta cells. CFTR-current activity induced by cAMP was measured in single alpha cells. Glucagon secretion at different glucose levels and in the presence of forskolin was increased by CFTR-inhibition in human islets, whereas depolarization-induced glucagon secretion was unaffected. CFTR is suggested to mainly regulate the membrane potential through an intrinsic alpha cell effect, as supported by a mathematical model of alpha cell electrophysiology. In conclusion, CFTR channels are present in alpha cells and act as important negative regulators of cAMP-enhanced glucagon secretion through effects on alpha cell membrane potential. Our data support that loss-of-function mutations in CFTR contributes to dysregulated glucagon secretion in CFRD.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Secretoras de Glucagon/citologia , Glucagon/metabolismo , Animais , Células Cultivadas , Colforsina/metabolismo , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/metabolismo , Glucose/farmacologia , Humanos , Masculino , Potenciais da Membrana , Camundongos , Pessoa de Meia-Idade , Mutação , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/efeitos dos fármacos , Células Secretoras de Somatostatina/metabolismo
6.
Diabetes Obes Metab ; 18 Suppl 1: 10-22, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27615127

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Ilhotas Pancreáticas/citologia , Proteínas do Tecido Nervoso/genética , Animais , Anticorpos Monoclonais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Reprogramação Celular , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo
7.
J Endocrinol ; 229(2): 123-32, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26931137

RESUMO

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.


Assuntos
Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Adolescente , Adulto , Idoso , Imunofluorescência , Humanos , Pessoa de Meia-Idade , Polipeptídeo Pancreático/metabolismo , Somatostatina/metabolismo , Distribuição Tecidual , Adulto Jovem
8.
PLoS One ; 10(12): e0144597, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26658466

RESUMO

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.


Assuntos
Proteínas do Olho/genética , Grelina/genética , Células Secretoras de Glucagon/metabolismo , Proteínas de Homeodomínio/genética , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Box Pareados/genética , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Proteínas Repressoras/genética , Células Secretoras de Somatostatina/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Diferenciação Celular , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Cruzamentos Genéticos , Proteínas do Olho/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Grelina/metabolismo , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Integrases/genética , Integrases/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Transdução de Sinais , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/efeitos dos fármacos , Tamoxifeno/farmacologia
9.
Diabetes ; 64(9): 3182-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25931474

RESUMO

Diffuse congenital hyperinsulinism in infancy (CHI-D) arises from mutations inactivating the KATP channel; however, the phenotype is difficult to explain from electrophysiology alone. Here we studied wider abnormalities in the ß-cell and other pancreatic lineages. Islets were disorganized in CHI-D compared with controls. PAX4 and ARX expression was decreased. A tendency toward increased NKX2.2 expression was consistent with its detection in two-thirds of CHI-D δ-cell nuclei, similar to the fetal pancreas, and implied immature δ-cell function. CHI-D δ-cells also comprised 10% of cells displaying nucleomegaly. In CHI-D, increased proliferation was most elevated in duct (5- to 11-fold) and acinar (7- to 47-fold) lineages. Increased ß-cell proliferation observed in some cases was offset by an increase in apoptosis; this is in keeping with no difference in INSULIN expression or surface area stained for insulin between CHI-D and control pancreas. However, nuclear localization of CDK6 and P27 was markedly enhanced in CHI-D ß-cells compared with cytoplasmic localization in control cells. These combined data support normal ß-cell mass in CHI-D, but with G1/S molecules positioned in favor of cell cycle progression. New molecular abnormalities in δ-cells and marked proliferative increases in other pancreatic lineages indicate CHI-D is not solely a ß-cell disorder.


Assuntos
Hiperinsulinismo Congênito/genética , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Somatostatina/metabolismo , Estudos de Casos e Controles , Linhagem da Célula , Proliferação de Células , Criança , Pré-Escolar , Hiperinsulinismo Congênito/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feto/citologia , Células Secretoras de Glucagon/citologia , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/metabolismo , Humanos , Lactente , Recém-Nascido , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Mutação , Proteínas Nucleares , Fatores de Transcrição Box Pareados/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Células Secretoras de Somatostatina/citologia , Receptores de Sulfonilureias/genética , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra
10.
Cell Stem Cell ; 15(2): 139-53, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25105579

RESUMO

The adult pancreas is capable of limited regeneration after injury but has no defined stem cell population. The cell types and molecular signals that govern the production of new pancreatic tissue are not well understood. Here, we show that inactivation of the SCF-type E3 ubiquitin ligase substrate recognition component Fbw7 induces pancreatic ductal cells to reprogram into α, δ, and ß cells. Loss of Fbw7 stabilized the transcription factor Ngn3, a key regulator of endocrine cell differentiation. The induced ß cells resemble islet ß cells in morphology and histology, express genes essential for ß cell function, and release insulin after glucose challenge. Thus, loss of Fbw7 appears to reawaken an endocrine developmental differentiation program in adult pancreatic ductal cells. Our study highlights the plasticity of seemingly differentiated adult cells, identifies Fbw7 as a master regulator of cell fate decisions in the pancreas, and reveals adult pancreatic duct cells as a latent multipotent cell type.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas F-Box/fisiologia , Células Secretoras de Glucagon/citologia , Células Secretoras de Insulina/citologia , Proteínas do Tecido Nervoso/fisiologia , Células Secretoras de Somatostatina/citologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Glucose/metabolismo , Células HEK293 , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Células-Tronco Multipotentes/citologia , Proteínas do Tecido Nervoso/genética , Ductos Pancreáticos/citologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Regeneração/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
11.
Nature ; 514(7523): 503-7, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25141178

RESUMO

Total or near-total loss of insulin-producing ß-cells occurs in type 1 diabetes. Restoration of insulin production in type 1 diabetes is thus a major medical challenge. We previously observed in mice in which ß-cells are completely ablated that the pancreas reconstitutes new insulin-producing cells in the absence of autoimmunity. The process involves the contribution of islet non-ß-cells; specifically, glucagon-producing α-cells begin producing insulin by a process of reprogramming (transdifferentiation) without proliferation. Here we show the influence of age on ß-cell reconstitution from heterologous islet cells after near-total ß-cell loss in mice. We found that senescence does not alter α-cell plasticity: α-cells can reprogram to produce insulin from puberty through to adulthood, and also in aged individuals, even a long time after ß-cell loss. In contrast, before puberty there is no detectable α-cell conversion, although ß-cell reconstitution after injury is more efficient, always leading to diabetes recovery. This process occurs through a newly discovered mechanism: the spontaneous en masse reprogramming of somatostatin-producing δ-cells. The juveniles display 'somatostatin-to-insulin' δ-cell conversion, involving dedifferentiation, proliferation and re-expression of islet developmental regulators. This juvenile adaptability relies, at least in part, upon the combined action of FoxO1 and downstream effectors. Restoration of insulin producing-cells from non-ß-cell origins is thus enabled throughout life via δ- or α-cell spontaneous reprogramming. A landscape with multiple intra-islet cell interconversion events is emerging, offering new perspectives for therapy.


Assuntos
Envelhecimento/fisiologia , Transdiferenciação Celular , Diabetes Mellitus Experimental/patologia , Células Secretoras de Insulina/citologia , Insulina/biossíntese , Regeneração , Células Secretoras de Somatostatina/citologia , Animais , Desdiferenciação Celular , Proliferação de Células , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/terapia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos , Maturidade Sexual , Somatostatina/biossíntese , Somatostatina/metabolismo , Células Secretoras de Somatostatina/metabolismo
12.
Coll Antropol ; 35(1): 5-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21661347

RESUMO

The aim of our paper was to investigate the influence of the different morphological changes on gastric mucosa on somatostatin D-cell number in antral mucosa and serum Somatostatin. We analyzed according to Sydney classification to what extent the severity of gastritis affect the observed hormonal values. somatostatin D-cell number in antral mucosa and serum Somatostatin values were compared between three groups of patients; mild, moderate and severe chronic gastritis. The average number of somatostatin cell in biopsy sample of antrum mucosa was 30.41 +/- 35.38 (N = 17) in the case of middle form, 18.69 +/- 26.65 (N = 56) in moderate and in severe case of chronic gastritis 5.23 +/- 5.93 (N = 7) cells in mm2 of mucosa. The level of somatostatin in the serum of middle form gastritis were 26.43 +/- 28.76, moderate 19.95 +/- 35.93 and severe 17.88 +/- 17.66 pg/mL. In order to determine the number of somatostatin cells in antrum mucosa and serum somatostatin with present morphological changes of mucosa, it might helpful to exclude the patients with non-ulcer dyspepsia, but with the higher risk of premalignant and malignant changes.


Assuntos
Mucosa Gástrica/citologia , Gastrite/patologia , Células Secretoras de Somatostatina/citologia , Somatostatina/sangue , Estudos de Coortes , Mucosa Gástrica/patologia , Gastrite/sangue , Humanos , Células Secretoras de Somatostatina/patologia
13.
Mol Endocrinol ; 24(8): 1605-14, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20592160

RESUMO

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.


Assuntos
Glicemia/metabolismo , Células Secretoras de Glucagon/citologia , Glucagon/fisiologia , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/fisiologia , Animais , Western Blotting , Glucagon/deficiência , Proteínas de Homeodomínio/genética , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Mutantes , Células Secretoras de Polipeptídeo Pancreático/citologia , Reação em Cadeia da Polimerase , Células Secretoras de Somatostatina/citologia , Fatores de Transcrição/genética
14.
Lab Invest ; 88(7): 761-72, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18490899

RESUMO

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.


Assuntos
Ilhotas Pancreáticas/citologia , Adenoviridae/genética , Adulto , Diferenciação Celular , Células Cultivadas , Vetores Genéticos , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Ilhotas Pancreáticas/metabolismo , Queratinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Ductos Pancreáticos/citologia , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Regiões Promotoras Genéticas , Somatostatina/fisiologia , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Células-Tronco/citologia , Estreptozocina/farmacologia , Proteína X Associada a bcl-2/biossíntese , Proteína X Associada a bcl-2/genética
15.
Diabetologia ; 50(6): 1239-42, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17440705

RESUMO

AIMS/HYPOTHESIS: Insulin secretion is a highly regulated mechanism involving a complex insulin-dependent network of communication between alpha, beta and delta cells. However, whereas the role of insulin in beta cells has been well documented, very little is known about its role in alpha and delta cells. Having recently demonstrated heterogeneity of insulin receptor (INSR) isoform expression in these three endocrine cell types, our current study aimed to characterise the expression pattern of the multiple isoforms involved in the insulin signal transduction cascade in human alpha, beta and delta cells in vitro. MATERIALS AND METHODS: cDNA samples prepared from single human islet cells were subjected to nested PCRs. RESULTS: Of 706 cells analysed, 15% were alpha cells, 28% beta cells, 8% delta cells and 46% non-endocrine cells. Profiling of expression of the insulin signalling cascade elements showed a heterogeneity between islet cell types, although at least one member of each protein family was expressed in the three populations of endocrine cells. Thus, the mRNAs coding for INSR-B, phosphoinositide-dependent protein kinase-1 and the human homologue of v-akt murine thymoma viral oncogene homologue 1 (AKT1) could not be detected in alpha cells, but were expressed by beta and delta cells. In addition, while the insulin receptor substrates IRS1 and IRS2, phosphoinositide-3-kinase, catalytic, beta polypeptide (PIK3CB) and AKT2 were expressed with relatively low frequencies in alpha and delta cells (<17% for IRS1, IRS2, PIK3CB; <25% for AKT2), their frequencies of expression in beta cells were 50, 33, 33 and 100%, respectively. CONCLUSIONS/INTERPRETATION: Our results suggest that insulin signalling cascade elements in human alpha, beta and delta cells have distinct expression patterns.


Assuntos
Regulação da Expressão Gênica , Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Células Cultivadas , Amplificação de Genes , Marcadores Genéticos , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/fisiologia , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/citologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA