Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119235, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35151663

RESUMO

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.


Assuntos
Ilhotas Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Transcriptoma
2.
Nat Commun ; 12(1): 4458, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294685

RESUMO

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.


Assuntos
Insulina/biossíntese , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Polipeptídeo Pancreático/metabolismo , Precursores de Proteínas/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Linhagem da Célula/genética , Feminino , Técnicas de Introdução de Genes , Humanos , Células Secretoras de Insulina/classificação , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Polipeptídeo Pancreático/deficiência , Polipeptídeo Pancreático/genética , Células Secretoras de Polipeptídeo Pancreático/classificação , Células Secretoras de Polipeptídeo Pancreático/citologia , Gravidez , RNA-Seq
3.
Nat Genet ; 53(4): 455-466, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33795864

RESUMO

Single-nucleus assay for transposase-accessible chromatin using sequencing (snATAC-seq) creates new opportunities to dissect cell type-specific mechanisms of complex diseases. Since pancreatic islets are central to type 2 diabetes (T2D), we profiled 15,298 islet cells by using combinatorial barcoding snATAC-seq and identified 12 clusters, including multiple alpha, beta and delta cell states. We cataloged 228,873 accessible chromatin sites and identified transcription factors underlying lineage- and state-specific regulation. We observed state-specific enrichment of fasting glucose and T2D genome-wide association studies for beta cells and enrichment for other endocrine cell types. At T2D signals localized to islet-accessible chromatin, we prioritized variants with predicted regulatory function and co-accessibility with target genes. A causal T2D variant rs231361 at the KCNQ1 locus had predicted effects on a beta cell enhancer co-accessible with INS and genome editing in embryonic stem cell-derived beta cells affected INS levels. Together our findings demonstrate the power of single-cell epigenomics for interpreting complex disease genetics.


Assuntos
Cromatina/química , Diabetes Mellitus Tipo 2/genética , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Canal de Potássio KCNQ1/genética , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Células Secretoras de Somatostatina/metabolismo , Glicemia/metabolismo , Diferenciação Celular , Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Epigenômica , Jejum , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Células Secretoras de Glucagon/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células Secretoras de Insulina/patologia , Canal de Potássio KCNQ1/metabolismo , Família Multigênica , Células Secretoras de Polipeptídeo Pancreático/patologia , Polimorfismo Genético , Análise de Célula Única , Células Secretoras de Somatostatina/patologia , Fatores de Transcrição/classificação , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
PLoS One ; 15(3): e0230627, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32208453

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/patologia , Lisina/análogos & derivados , Pâncreas/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Baço/metabolismo , Adulto , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Lisina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Fatores de Iniciação de Peptídeos/genética , Proteínas de Ligação a RNA/genética , Adulto Jovem , Fator de Iniciação de Tradução Eucariótico 5A
5.
Nature ; 567(7746): 43-48, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30760930

RESUMO

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.


Assuntos
Diabetes Mellitus/patologia , Diabetes Mellitus/terapia , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/patologia , Animais , Biomarcadores/análise , Linhagem da Célula/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Diabetes Mellitus/imunologia , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Feminino , Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/transplante , Glucose/farmacologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Masculino , Camundongos , Especificidade de Órgãos/efeitos dos fármacos , Polipeptídeo Pancreático/metabolismo , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/efeitos dos fármacos , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Proteômica , Análise de Sequência de RNA , Transativadores/genética , Transativadores/metabolismo , Transcriptoma , Transdução Genética
6.
Cell Metab ; 28(4): 557-572.e6, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30017352

RESUMO

The gut microbiota is essential for the normal function of the gut immune system, and microbiota alterations are associated with autoimmune disorders. However, how the gut microbiota prevents autoimmunity in distant organs remains poorly defined. Here we reveal that gut microbiota conditioned innate lymphoid cells (ILCs) induce the expression of mouse ß-defensin 14 (mBD14) by pancreatic endocrine cells, preventing autoimmune diabetes in the non-obese diabetic (NOD) mice. MBD14 stimulates, via Toll-like receptor 2, interleukin-4 (IL-4)-secreting B cells that induce regulatory macrophages, which in turn induce protective regulatory T cells. The gut microbiota-derived molecules, aryl hydrocarbon receptor (AHR) ligands and butyrate, promote IL-22 secretion by pancreatic ILCs, which induce expression of mBD14 by endocrine cells. Dysbiotic microbiota and low-affinity AHR allele explain the defective pancreatic expression of mBD14 observed in NOD mice. Our study reveals a yet unidentified crosstalk between ILCs and endocrine cells in the pancreas that is essential for the prevention of autoimmune diabetes development.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Microbioma Gastrointestinal/imunologia , Células Secretoras de Insulina/metabolismo , Linfócitos/metabolismo , Células Secretoras de Polipeptídeo Pancreático/metabolismo , beta-Defensinas/metabolismo , Animais , Linfócitos B Reguladores/metabolismo , Feminino , Humanos , Imunidade Inata , Interleucinas/metabolismo , Ilhotas Pancreáticas/metabolismo , Estimativa de Kaplan-Meier , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Estatísticas não Paramétricas , Linfócitos T Reguladores/metabolismo , Receptor 2 Toll-Like/metabolismo , Interleucina 22
7.
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
8.
Pancreas ; 46(6): 820-824, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28609372

RESUMO

A partial pancreaticogastrodudenectomy was performed on a 66-year old man with type 2 diabetes mellitus because of an invasive, moderately differentiated adenocarcinoma in the head of the pancreas. In the adjacent grossly normal tissue of the uncinate process, there was a massive proliferation of pancreatic polypeptide (PP) cells confined to this region and showed invasive pattern. Strikingly, in the heaped area of his duodenum, there was a strikingly large number of PP, glucagon, a few insulin cells in a mini-islet-like patterns composed of glucagon and insulin cells. Among the etiological factors, the possible long-lasting effects of the GLP-1 analog, with which the patient was treated, are discussed. This is the first report in the literature of both the coexistence of a pancreatic adenocarcinoma and invasive PPoma and the occurrence of PP and insulin cells in human duodenal mucosa.


Assuntos
Adenocarcinoma/complicações , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Duodeno/efeitos dos fármacos , Hipoglicemiantes/efeitos adversos , Liraglutida/efeitos adversos , Pâncreas/efeitos dos fármacos , Neoplasias Pancreáticas/complicações , Células Secretoras de Polipeptídeo Pancreático/efeitos dos fármacos , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Idoso , Quimioterapia Adjuvante , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/diagnóstico , Duodeno/metabolismo , Duodeno/patologia , Duodeno/cirurgia , Evolução Fatal , Humanos , Hiperplasia , Imuno-Histoquímica , Masculino , Pâncreas/metabolismo , Pâncreas/patologia , Pâncreas/cirurgia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Pancreaticoduodenectomia , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
9.
Diabetes Obes Metab ; 19(9): 1267-1275, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28345790

RESUMO

AIMS: Ghrelin is a gastric-derived hormone that stimulates growth hormone (GH) secretion and has a multi-faceted role in the regulation of energy homeostasis, including glucose metabolism. Circulating ghrelin concentrations are modulated in response to nutritional status, but responses to ghrelin in altered metabolic states are poorly understood. We investigated the metabolic effects of ghrelin in obesity and early after Roux-en-Y gastric bypass (RYGB). MATERIALS AND METHODS: We assessed central and peripheral metabolic responses to acyl ghrelin infusion (1 pmol kg-1 min-1 ) in healthy, lean subjects (n = 9) and non-diabetic, obese subjects (n = 9) before and 2 weeks after RYGB. Central responses were assessed by GH and pancreatic polypeptide (surrogate for vagal activity) secretion. Peripheral responses were assessed by hepatic and skeletal muscle insulin sensitivity during a hyperinsulinaemic-euglycaemic clamp. RESULTS: Ghrelin-stimulated GH secretion was attenuated in obese subjects, but was restored by RYGB to a response similar to that of lean subjects. The heightened pancreatic polypeptide response to ghrelin infusion in the obese was attenuated after RYGB. Hepatic glucose production and hepatic insulin sensitivity were not altered by ghrelin infusion in RYGB subjects. Skeletal muscle insulin sensitivity was impaired to a similar degree in lean, obese and post-RYGB individuals in response to ghrelin infusion. CONCLUSIONS: These data suggest that obesity is characterized by abnormal central, but not peripheral, responsiveness to ghrelin that can be restored early after RYGB before significant weight loss. Further work is necessary to fully elucidate the role of ghrelin in the metabolic changes that occur in obesity and following RYGB.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Derivação Gástrica , Grelina/uso terapêutico , Hormônio do Crescimento Humano/agonistas , Resistência à Insulina , Obesidade Mórbida/tratamento farmacológico , Obesidade Mórbida/cirurgia , Acilação , Fármacos Antiobesidade/administração & dosagem , Fármacos Antiobesidade/efeitos adversos , Fármacos Antiobesidade/química , Estudos de Coortes , Terapia Combinada/efeitos adversos , Estudos Cross-Over , Metabolismo Energético/efeitos dos fármacos , Grelina/administração & dosagem , Grelina/efeitos adversos , Grelina/química , Gluconeogênese/efeitos dos fármacos , Técnica Clamp de Glucose , Hormônio do Crescimento Humano/sangue , Hormônio do Crescimento Humano/metabolismo , Humanos , Infusões Intravenosas , Fígado/efeitos dos fármacos , Fígado/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Obesidade Mórbida/sangue , Obesidade Mórbida/metabolismo , Polipeptídeo Pancreático/agonistas , Polipeptídeo Pancreático/sangue , Polipeptídeo Pancreático/metabolismo , Células Secretoras de Polipeptídeo Pancreático/efeitos dos fármacos , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Cuidados Pós-Operatórios , Cuidados Pré-Operatórios , Precursores de Proteínas/agonistas , Precursores de Proteínas/sangue , Precursores de Proteínas/metabolismo , Método Simples-Cego
10.
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
11.
PLoS One ; 11(6): e0157138, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27270601

RESUMO

The transcription factor Glis-similar 3 (Glis3) has been implicated in the development of neonatal, type 1 and type 2 diabetes. In this study, we examined the spatiotemporal expression of Glis3 protein during embryonic and neonatal pancreas development as well as its function in PP cells. To obtain greater insights into the functions of Glis3 in pancreas development, we examined the spatiotemporal expression of Glis3 protein in a knockin mouse strain expressing a Glis3-EGFP fusion protein. Immunohistochemistry showed that Glis3-EGFP was not detectable during early pancreatic development (E11.5 and E12.5) and at E13.5 and 15.5 was not expressed in Ptf1a+ cells in the tip domains indicating that Glis3 is not expressed in multipotent pancreatic progenitors. Glis3 was first detectable at E13.5 in the nucleus of bipotent progenitors in the trunk domains, where it co-localized with Sox9, Hnf6, and Pdx1. It remained expressed in preductal and Ngn3+ endocrine progenitors and at later stages becomes restricted to the nucleus of pancreatic beta and PP cells as well as ductal cells. Glis3-deficiency greatly reduced, whereas exogenous Glis3, induced Ppy expression, as reported for insulin. Collectively, our study demonstrates that Glis3 protein exhibits a temporal and cell type-specific pattern of expression during embryonic and neonatal pancreas development that is consistent with a regulatory role for Glis3 in promoting endocrine progenitor generation, regulating insulin and Ppy expression in beta and PP cells, respectively, and duct morphogenesis.


Assuntos
Células Secretoras de Insulina/metabolismo , Pâncreas/crescimento & desenvolvimento , Ductos Pancreáticos/metabolismo , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/genética , Transativadores/metabolismo , Animais , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas de Ligação a DNA , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Camundongos , Pâncreas/citologia , Polipeptídeo Pancreático/metabolismo
12.
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
13.
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
14.
Tissue Cell ; 46(6): 535-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25458814

RESUMO

tIn this study, we investigated the presence of ovoid or ellipsoidal amylin-immunoreactive cells of the pancreatic islets of the black-spotted frog Rana (Pelophylax) nigromaculata. Using double immunofluorescent staining, all amylin-immunoreactive cells were shown to be immuno-negative for insulin, glucagon, and somatostatin, and they were often observed in peripheral regions of clusters of insulin-immunoreactive cells. Under immunoelectron microscopy, amylin-immunoreactive signals were detected on the secretory granules in a specific type of endocrine cells. From our results, we conclude that the amylin-immunoreactive cells correspond to X cells among the 4 distinct types of endocrine cells (B, A/PP, D, and X) previously identified in the frog. Amylin secreted from X cells may regulate the hormone secretion from A/PP cells and/or B cells through a paracrine mechanism.


Assuntos
Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Pâncreas/metabolismo , Animais , Tecido Conjuntivo/metabolismo , Glucagon , Células Secretoras de Glucagon/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/imunologia , Pâncreas/citologia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Ranidae
15.
Pancreas ; 43(4): 648-50, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24713672

RESUMO

A 74-year-old man with recurrent duodenal ulcers underwent somatostatin receptor scintigraphy (SRS) in suspicion of gastrinoma. A 2-cm area of focal uptake was visualized within the pancreatic head. Serum chromogranin A levels were elevated, but serum gastrin levels and the secretin test were normal. Computed tomography and endoscopic ultrasonography were not conclusive. After partial duodenopancreatectomy, pathological examination failed to reveal any neuroendocrine tumor. Instead, the dorsal portion of the pancreatic head was found to be densely populated by pancreatic polypeptide cell-rich islets. This area correlated with the site of tracer uptake seen on SRS. Thus, pancreatic polypeptide cell-rich islets in elderly patients should be kept in mind when interpreting SRS results to avoid unnecessary major pancreatic resections.


Assuntos
Erros de Diagnóstico , Tumores Neuroendócrinos/diagnóstico por imagem , Octreotida/análogos & derivados , Neoplasias Pancreáticas/diagnóstico por imagem , Células Secretoras de Polipeptídeo Pancreático/diagnóstico por imagem , Compostos Radiofarmacêuticos , Receptores de Somatostatina/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Idoso , Biópsia , Humanos , Masculino , Imagem Multimodal/métodos , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/cirurgia , Pancreatectomia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/cirurgia , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Valor Preditivo dos Testes , Tomografia Computadorizada por Raios X , Procedimentos Desnecessários
16.
Regul Pept ; 187: 42-50, 2013 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-24183983

RESUMO

Xenin-25 (Xen) is a 25-amino acid neurotensin-related peptide that activates neurotensin receptor-1 (NTSR1). We previously showed that Xen increases the effect of glucose-dependent insulinotropic polypeptide (GIP) on insulin release 1) in hyperglycemic mice via a cholinergic relay in the periphery independent from the central nervous system and 2) in humans with normal or impaired glucose tolerance, but not type 2 diabetes mellitus (T2DM). Since this blunted response to Xen defines a novel defect in T2DM, it is important to understand how Xen regulates islet physiology. On separate visits, subjects received intravenous graded glucose infusions with vehicle, GIP, Xen, or GIP plus Xen. The pancreatic polypeptide response was used as an indirect measure of cholinergic input to islets. The graded glucose infusion itself had little effect on the pancreatic polypeptide response whereas administration of Xen equally increased the pancreatic polypeptide response in humans with normal glucose tolerance, impaired glucose tolerance, and T2DM. The pancreatic polypeptide response to Xen was similarly amplified by GIP in all 3 groups. Antibody staining of human pancreas showed that NTSR1 is not detectable on islet endocrine cells, sympathetic neurons, blood vessels, or endothelial cells but is expressed at high levels on PGP9.5-positive axons in the exocrine tissue and at low levels on ductal epithelial cells. PGP9.5 positive nerve fibers contacting beta cells in the islet periphery were also observed. Thus, a neural relay, potentially involving muscarinic acetylcholine receptors, indirectly increases the effects of Xen on pancreatic polypeptide release in humans.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Polipeptídeo Inibidor Gástrico/farmacologia , Neurotensina/farmacologia , Pâncreas/inervação , Polipeptídeo Pancreático/metabolismo , Adulto , Glicemia , Estudos de Casos e Controles , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Polipeptídeo Pancreático/sangue , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Receptores de Neurotensina/metabolismo
17.
PLoS One ; 8(8): e72213, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977255

RESUMO

Mutations in the human homolog of the Vhlh gene [encoding the von-Hippel Lindau (VHL) protein] lead to tumor development. In mice, depletion of Vhlh in pancreatic ß-cells causes perturbed glucose homeostasis, but the role of this gene in other pancreatic cells is poorly understood. To investigate the function of VHL/HIF pathway in pancreatic cells, we inactivated Vhlh in the pancreatic epithelium as well as in the endocrine and exocrine lineages. Our results show that embryonic depletion of Vhlh within the pancreatic epithelium causes postnatal lethality due to severe hypoglycemia. The hypoglycemia is recapitulated in mice with endocrine-specific removal of Vhlh, while animals with loss of Vhlh predominantly in the exocrine compartment survive to adulthood with no overt defects in glucose metabolism. Mice with hypoglycemia display diminished insulin release in response to elevated glucose. Significantly, the glucagon response is impaired both in vivo (circulating glucagon levels) as well as in an in vitro secretion assay in isolated islets. Hypoxia also impairs glucagon secretion in a glucagon-expressing cell line in culture. Our results reveal a novel role for the hypoxia/HIF pathway in islet hormone secretion and maintenance of the fine balance that allows for the establishment of normoglycemia.


Assuntos
Sistema Endócrino/metabolismo , Células Secretoras de Glucagon/metabolismo , Fator 1 Induzível por Hipóxia/genética , Hipóxia/genética , Células Secretoras de Somatostatina/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Animais , Células Cultivadas , Embrião de Mamíferos , Sistema Endócrino/patologia , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Glucose/metabolismo , Homeostase/genética , Hipóxia/metabolismo , Hipóxia/patologia , Fator 1 Induzível por Hipóxia/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Knockout , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Células Secretoras de Polipeptídeo Pancreático/patologia , Transdução de Sinais , Células Secretoras de Somatostatina/patologia , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
18.
PLoS One ; 8(1): e55501, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383206

RESUMO

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.


Assuntos
Pâncreas/citologia , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Glucagon/citologia , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas , Pâncreas/anatomia & histologia , Pâncreas/metabolismo , Células Secretoras de Polipeptídeo Pancreático/metabolismo
19.
Pancreas ; 39(6): 836-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20182388

RESUMO

OBJECTIVES: Somatostatin inhibits hormone release through 5 G protein-coupled somatostatin receptors (sst1-sst5). However, the role of somatostatin in islet physiology is not fully known. The immunoreactivity to sst1 to sst5 in normal human endocrine pancreas has been described. The present study reports the expression of sst1 to sst5 in human pancreatic islets with type 2 diabetes mellitus. METHODS: Pancreatic autopsy specimens from individuals with type 2 diabetes mellitus and matched controls were double immunostained to demonstrate sst1 to sst5 in the major islet cell types. RESULTS: Most apparent differences in type 2 diabetic islets were the lack of sst1 and sst4 in glucagon cells and sst1-3 and 4 in somatostatin cells, whereas minor changes were demonstrated in insulin cells. The pancreatic polypeptide cells showed a reversed staining pattern in diabetic islets compared with the controls. CONCLUSIONS: In type 2 diabetes mellitus, the sst pattern differed from that of the controls in somatostatin, pancreatic polypeptide, and glucagon cells, to a minor extent in insulin cells. It is unclear whether the changes in sst patterns are primarily due to the diabetes or secondary to metabolic disturbances. However, this study may be the basis for further functional studies to evaluate the role of sst1 to sst5 in the diabetic state.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Autopsia , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Isoformas de Proteínas/metabolismo , Células Secretoras de Somatostatina/metabolismo
20.
Eur J Histochem ; 53(2): 81-5, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19683981

RESUMO

We used immunofluorescence double staining method to investigate the cellular localization of glucagon and pancreatic polypeptide (PP) in rat pancreatic islets. The results showed that both A-cells (glucagon-secreting cells) and PP-cells (PP-secreting cells) were located in the periphery of the islets. However, A-cells and PP-cells had a different regional distribution. Most of A-cells were located in the splenic lobe but a few of them were in the duodenal lobe of the pancreas. In contrast, the majority of PP-cells were found in the duodenal lobe and a few of them were in the splenic lobe of the pancreas. Furthermore, we found that 67.74% A-cells had PP immunoreactivity, 70.92% PP-cells contained glucagon immunoreactivity with immunofluorescence double staining. Our data support the concept of a common precursor stem cell for pancreatic hormone-producing cells.


Assuntos
Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Polipeptídeo Pancreático/metabolismo , Animais , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA