Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Metab ; 42: 101060, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32763423

RESUMO

OBJECTIVES: The main endocrine cell types in pancreatic islets are alpha, beta, and delta cells. Although these cell types have distinct roles in the regulation of glucose homeostasis, inadequate purification methods preclude the study of cell type-specific effects. We developed a reliable approach that enables simultaneous sorting of live alpha, beta, and delta cells from mouse islets for downstream analyses. METHODS: We developed an antibody panel against cell surface antigens to enable isolation of highly purified endocrine subsets from mouse islets based on the specific differential expression of CD71 on beta cells and CD24 on delta cells. We rigorously demonstrated the reliability and validity of our approach using bulk and single cell qPCR, immunocytochemistry, reporter mice, and transcriptomics. RESULTS: Pancreatic alpha, beta, and delta cells can be separated based on beta cell-specific CD71 surface expression and high expression of CD24 on delta cells. We applied our new sorting strategy to demonstrate that CD71, which is the transferrin receptor mediating the uptake of transferrin-bound iron, is upregulated in beta cells during early postnatal weeks. We found that beta cells express higher levels of several other genes implicated in iron metabolism and iron deprivation significantly impaired beta cell function. In human beta cells, CD71 is similarly required for iron uptake and CD71 surface expression is regulated in a glucose-dependent manner. CONCLUSIONS: This study provides a novel and efficient purification method for murine alpha, beta, and delta cells, identifies for the first time CD71 as a postnatal beta cell-specific marker, and demonstrates a central role of iron metabolism in beta cell function.


Assuntos
Antígenos de Superfície/imunologia , Células Secretoras de Insulina/metabolismo , Ferro/metabolismo , Animais , Antígenos CD/imunologia , Antígenos de Superfície/isolamento & purificação , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Antígeno CD24/imunologia , Linhagem Celular , Feminino , Células Secretoras de Glucagon/imunologia , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/fisiologia , Humanos , Imuno-Histoquímica/métodos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/fisiologia , Ferro/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/metabolismo , Pâncreas/fisiologia , Receptores da Transferrina/imunologia , Reprodutibilidade dos Testes , Células Secretoras de Somatostatina/imunologia , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/fisiologia
2.
Diabetes Obes Metab ; 18 Suppl 1: 128-36, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27615142

RESUMO

Diabetes (either type 1 or type 2) is due to insufficient functional ß-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either ß-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model ß-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human ß-cells share many similarities with rodent ß-cells, they also differ on a number of important parameters. In this context, developing ways to study human ß-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human ß-cells that should be useful tools to discover new ways to modulate functional ß-cell mass in humans.


Assuntos
Técnicas de Cultura de Células , Diabetes Mellitus/terapia , Células-Tronco Embrionárias Humanas , Células Secretoras de Insulina/citologia , Animais , Pesquisa Biomédica , Diferenciação Celular , Linhagem Celular , Humanos , Camundongos , Modelos Animais , Ratos
3.
Cell Death Dis ; 5: e1487, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25341041

RESUMO

During embryogenesis, the intrauterine milieu affects cell proliferation, differentiation, and function by modifying gene expression in susceptible cells, such as the pancreatic ß-cells. In this limited energy environment, mitochondrial dysfunction can lead to overproduction of reactive oxygen species (ROS) and to a decline in ß-cell function. In opposition to this toxicity, ROS are also required for insulin secretion. Here we investigated the role of ROS in ß-cell development. Surprisingly, decreasing ROS production in vivo reduced ß-cell differentiation. Moreover, in cultures of pancreatic explants, progenitors were highly sensitive to ROS stimulation and responded by generating ß-cells. ROS enhanced ß-cell differentiation through modulation of ERK1/2 signaling. Gene transfer and pharmacological manipulations, which diminish cellular ROS levels, also interfered with normal ß-cell differentiation. This study highlights the role of the redox balance on ß-cell development and provides information that will be useful for improving ß-cell production from embryonic stem cells, a step in cell therapy for diabetes.


Assuntos
Antioxidantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Oxidantes/toxicidade , Acetilcisteína/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Catalase/metabolismo , Embrião de Mamíferos/citologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Feminino , Peróxido de Hidrogênio/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Fosforilação/efeitos dos fármacos , Ratos Wistar
4.
Diabetes Metab ; 39(3): 276-80, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23562494

RESUMO

AIM: Permanent neonatal diabetes mellitus (PNDM) is a rare monogenic form of non-autoimmune diabetes. Genetic defects have been identified in∼60% of cases, with mutations in ABCC8, KCNJ11 and INS being the most frequent causes of PNDM. Recognition of genetic subtypes strongly impacts on both patients' care and family counseling. This study aimed to identify the genetic aetiology of PNDM in a diabetic girl born of consanguineous parents. METHODS: DNA samples from both the proband and her non-diabetic parents were analyzed for homozygosity mapping, using Illumina Infinium 660K SNP microarrays, focusing on the runs of homozygosity (ROHs) detected only in the patient. Standard Sanger sequencing of candidate genes (MNX1 and GATA6) present in the ROHs was subsequently performed, as well as expression analyses on human embryonic and adult pancreatic islet samples. RESULTS: A putative causal homozygous mutation in the transcription factor gene MNX1 (c.816C>A/p.Phe272Leu) was identified in the PNDM patient, who was clinically diagnosed as a typical case of PNDM with no developmental pancreatic defects or other clinical features. The probable deleterious mutation was located within the MNX1 homeodomain helix 2 that is highly conserved between species. In human embryonic pancreatic islet samples, it has been shown that MNX1 expression is significantly enriched in pancreatic epithelium compared with mesenchyme, suggesting a role for MNX1 in human pancreatic beta-cell development. CONCLUSION: This study found a new putative cause of PNDM in a consanguineous family. Replication in other cohorts would help to clarify the clinical spectrum of MNX1 mutations in PNDM patients.


Assuntos
Consanguinidade , Diabetes Mellitus/genética , Proteínas de Homeodomínio/genética , Doenças do Recém-Nascido/genética , Fatores de Transcrição/genética , Diabetes Mellitus/diagnóstico , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/diagnóstico , Análise de Sequência de DNA
5.
Diabetologia ; 56(6): 1327-38, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23462796

RESUMO

AIMS/HYPOTHESIS: It is not clear how small tyrosine kinase inhibitors, such as imatinib mesilate, protect against diabetes and beta cell death. The aim of this study was to determine whether imatinib, as compared with the non-cAbl-inhibitor sunitinib, affects pro-survival signalling events in the phosphatidylinositol 3-kinase (PI3K) pathway. METHODS: Human EndoC-ßH1 cells, murine beta TC-6 cells and human pancreatic islets were used for immunoblot analysis of insulin receptor substrate (IRS)-1, Akt and extracellular signal-regulated kinase (ERK) phosphorylation. Phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] plasma membrane concentrations were assessed in EndoC-ßH1 and MIN6 cells using evanescent wave microscopy. Src homology 2-containing inositol 5'-phosphatase 2 (SHIP2) tyrosine phosphorylation and phosphatase and tensin homologue deleted on chromosome 10 (PTEN) serine phosphorylation, as well as c-Abl co-localisation with SHIP2, were studied in HEK293 and EndoC-ßH1 cells by immunoprecipitation and immunoblot analysis. Gene expression was assessed using RT-PCR. Cell viability was measured using vital staining. RESULTS: Imatinib stimulated ERK(thr202/tyr204) phosphorylation in a c-Abl-dependent manner. Imatinib, but not sunitinib, also stimulated IRS-1(tyr612), Akt(ser473) and Akt(thr308) phosphorylation. This effect was paralleled by oscillatory bursts in plasma membrane PI(3,4,5)P3 levels. Wortmannin induced a decrease in PI(3,4,5)P3 levels, which was slower in imatinib-treated cells than in control cells, indicating an effect on PI(3,4,5)P3-degrading enzymes. In line with this, imatinib decreased the phosphorylation of SHIP2 but not of PTEN. c-Abl co-immunoprecipitated with SHIP2 and its binding to SHIP2 was largely reduced by imatinib but not by sunitinib. Imatinib increased total ß-catenin levels and cell viability, whereas sunitinib exerted negative effects on cell viability. CONCLUSIONS/INTERPRETATION: Imatinib inhibition of c-Abl in beta cells decreases SHIP2 activity, which results in enhanced signalling downstream of PI3 kinase.


Assuntos
Benzamidas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Insulina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Mesilato de Imatinib , Indóis/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosforilação , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Sunitinibe , Fatores de Tempo
6.
Diabetologia ; 56(2): 330-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23178930

RESUMO

AIMS/HYPOTHESIS: The main objective of this work was to discover new drugs that can activate the differentiation of multipotent pancreatic progenitors into endocrine cells. METHODS: In vitro experiments were performed using fetal pancreatic explants from rats and mice. In this assay, we examined the actions on pancreatic cell development of glibenclamide, a sulfonylurea derivative, and glycine hydrazide (GlyH-101), a small-molecule inhibitor of cystic fibrosis transmembrane conductance regulator (CFTR). We next tested the actions of GlyH-101 on in vivo pancreatic cell development. RESULTS: Glibenclamide (10 nmol/l-100 µmol/l) did not alter the morphology or growth of the developing pancreas and exerted no deleterious effects on exocrine cell development in the pancreas. Unexpectedly, glibenclamide at its highest concentration promoted endocrine differentiation. This glibenclamide-induced promotion of the endocrine pathway could not be reproduced when other sulfonylureas were used, suggesting that glibenclamide had an off-target action. This high concentration of glibenclamide had previously been reported to inhibit CFTR. We found that the effects of glibenclamide on the developing pancreas could be mimicked both in vitro and in vivo by GlyH-101. CONCLUSIONS/INTERPRETATION: Collectively, we demonstrate that two small-molecule inhibitors of the CFTR, glibenclamide and GlyH-101, increase the number of pancreatic endocrine cells by increasing the size of the pool of neurogenin 3-positive endocrine progenitors in the developing pancreas.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Glibureto/farmacologia , Glicina/análogos & derivados , Hidrazinas/farmacologia , Pâncreas/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Feminino , Glicina/farmacologia , Imunoquímica , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Cultura de Órgãos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Gravidez , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
7.
Diabetologia ; 55(10): 2713-2722, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22760788

RESUMO

AIMS/HYPOTHESIS: Pancreatic cell development is a tightly controlled process. Although information is available regarding the mesodermal signals that control pancreatic development, little is known about the role of environmental factors such as nutrients, including glucose, on pancreatic development. We previously showed that glucose and its metabolism through the hexosamine biosynthesis pathway (HBP) promote pancreatic endocrine cell differentiation. Here, we analysed the role of the transcription factor carbohydrate-responsive element-binding protein (ChREBP) in this process. This transcription factor is activated by glucose, and has been recently described as a target of the HBP. METHODS: We used an in vitro bioassay in which pancreatic endocrine and exocrine cells develop from rat embryonic pancreas in a way that mimics in vivo pancreatic development. Using this model, gain-of-function and loss-of-function experiments were undertaken. RESULTS: ChREBP was produced in the endocrine lineage during pancreatic development, its abundance increasing with differentiation. When rat embryonic pancreases were cultured in the presence of glucose or xylitol, the production of ChREBP targets was induced. Concomitantly, beta cell differentiation was enhanced. On the other hand, when embryonic pancreases were cultured with inhibitors decreasing ChREBP activity or an adenovirus producing a dominant-negative ChREBP, beta cell differentiation was reduced, indicating that ChREBP activity was necessary for proper beta cell differentiation. Interestingly, adenovirus producing a dominant-negative ChREBP also reduced the positive effect of N-acetylglucosamine, a substrate of the HBP acting on beta cell differentiation. CONCLUSIONS/INTERPRETATION: Our work supports the idea that glucose, through the transcription factor ChREBP, controls beta cell differentiation from pancreatic progenitors.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Glucose/farmacologia , Células Secretoras de Insulina/citologia , Acetilglucosamina/farmacologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Técnicas In Vitro , Modelos Animais , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/fisiologia , Gravidez , Ratos , Ratos Wistar , Xilitol/farmacologia
9.
Diabetologia ; 55(1): 154-65, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21947380

RESUMO

AIMS/HYPOTHESIS: The aim of the study was to identify surface bio-markers and corresponding antibody tools that can be used for the imaging and immunoisolation of the pancreatic beta cell and its progenitors. This may prove essential to obtain therapeutic grade human beta cells via stem cell differentiation. METHODS: Using bioinformatics-driven data mining, we generated a gene list encoding putative plasma membrane proteins specifically expressed at distinct stages of the developing pancreas and islet beta cells. In situ hybridisation and immunohistochemistry were used to further prioritise and identify candidates. RESULTS: In the developing pancreas seizure related 6 homologue like (SEZ6L2), low density lipoprotein receptor-related protein 11 (LRP11), dispatched homologue 2 (Drosophila) (DISP2) and solute carrier family 30 (zinc transporter), member 8 (SLC30A8) were found to be expressed in early islet cells, whereas discoidin domain receptor tyrosine kinase 1 (DDR1) and delta/notch-like EGF repeat containing (DNER) were expressed in early pancreatic progenitors. The expression pattern of DDR1 overlaps with the early pancreatic and duodenal homeobox 1 (PDX1)⁺/NK6 homeobox 1 (NKX6-1)⁺ multipotent progenitor cells from embryonic day 11, whereas DNER expression in part overlaps with neurogenin 3 (NEUROG3)⁺ cells. In the adult pancreas SEZ6L2, LRP11, DISP2 and SLC30A8, but also FXYD domain containing ion transport regulator 2 (FXYD2), tetraspanin 7 (TSPAN7) and transmembrane protein 27 (TMEM27), retain an islet-specific expression, whereas DDR1 is undetectable. In contrast, DNER is expressed at low levels in peripheral mouse and human islet cells. Re-expression of DDR1 and upregulation of DNER is observed in duct-ligated pancreas. Antibodies to DNER and DISP2 have been successfully used in cell sorting. CONCLUSIONS/INTERPRETATION: Extracellular epitopes of SEZ6L2, LRP11, DISP2, DDR1 and DNER have been identified as useful tags by applying specific antibodies to visualise pancreatic cell types at specific stages of development. Furthermore, antibodies recognising DISP2 and DNER are suitable for FACS-mediated cell purification.


Assuntos
Antígenos de Superfície/metabolismo , Separação Celular/métodos , Ilhotas Pancreáticas/metabolismo , Células-Tronco/metabolismo , Adulto , Animais , Biomarcadores/metabolismo , Linhagem Celular , Biologia Computacional/métodos , Mineração de Dados , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Camundongos , Camundongos Endogâmicos BALB C , Técnicas de Cultura de Órgãos , Células-Tronco/citologia
10.
Diabetologia ; 54(4): 819-27, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21181396

RESUMO

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


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Ilhotas Pancreáticas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/metabolismo , Glucose/farmacologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Modelos Biológicos , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/genética , Ratos
11.
Endocr Rev ; 32(2): 159-224, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20971919

RESUMO

Plasticity in developmental programming has evolved in order to provide the best chances of survival and reproductive success to the organism under changing environments. Environmental conditions that are experienced in early life can profoundly influence human biology and long-term health. Developmental origins of health and disease and life-history transitions are purported to use placental, nutritional, and endocrine cues for setting long-term biological, mental, and behavioral strategies in response to local ecological and/or social conditions. The window of developmental plasticity extends from preconception to early childhood and involves epigenetic responses to environmental changes, which exert their effects during life-history phase transitions. These epigenetic responses influence development, cell- and tissue-specific gene expression, and sexual dimorphism, and, in exceptional cases, could be transmitted transgenerationally. Translational epigenetic research in child health is a reiterative process that ranges from research in the basic sciences, preclinical research, and pediatric clinical research. Identifying the epigenetic consequences of fetal programming creates potential applications in clinical practice: the development of epigenetic biomarkers for early diagnosis of disease, the ability to identify susceptible individuals at risk for adult diseases, and the development of novel preventive and curative measures that are based on diet and/or novel epigenetic drugs.


Assuntos
Desenvolvimento Infantil/fisiologia , Proteção da Criança , Epigênese Genética/fisiologia , Adolescente , Envelhecimento/fisiologia , Criança , Pré-Escolar , Meio Ambiente , Feminino , Impressão Genômica/fisiologia , Humanos , Lactente , Recém-Nascido , Masculino , Diferenciação Sexual/fisiologia
12.
Diabetes Obes Metab ; 10 Suppl 4: 195-200, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18834447

RESUMO

Understanding in detail how pancreatic endocrine cells develop is important for many reasons. From a scientific point of view, elucidation of such a complex process is a major challenge. From a more applied point of view, this may help us to better understand and treat specific forms of diabetes. Although a variety of therapeutic approaches are well validated, no cure for diabetes is available. Many arguments indicate that the development of new strategies to cure diabetic patients will require precise understanding of the way beta-cells form during development. This is obvious for a future cell therapy using beta-cells produced from embryonic stem cells. This also holds true for therapeutic approaches based on regenerative medicine. In this review, we summarize our current knowledge concerning pancreatic development and focus on the role of extracellular signals implicated in beta-cell development from pancreatic progenitors.


Assuntos
Células-Tronco Adultas/citologia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células-Tronco Embrionárias/citologia , Células Secretoras de Insulina/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco Adultas/fisiologia , Animais , Diferenciação Celular , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Células-Tronco Embrionárias/fisiologia , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Camundongos , Células Precursoras de Linfócitos B/fisiologia , Regeneração/fisiologia
14.
Dev Dyn ; 236(6): 1517-25, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17450605

RESUMO

Calsenilin/DREAM/Kchip3 is a neuronal calcium-binding protein. It is a multifunctional protein, mainly expressed in neural tissues and implicated in regulation of presenilin processing, repression of transcription, and modulation of A-type potassium channels. Here, we performed a search for new genes expressed during pancreatic development and have studied the spatiotemporal expression pattern and possible role of calsenilin in pancreatic development in zebrafish. We detected calsenilin transcripts in the pancreas from 21 somites to 39 hours postfertilization stages. Using double in situ hybridization, we found that the calsenilin gene was expressed in pancreatic endocrine cells. Loss-of-function experiments with anti-calsenilin morpholinos demonstrated that injected morphants have a significant decrease in the number of pancreatic endocrine cells. Furthermore, the knockdown of calsenilin leads to perturbation in islet morphogenesis, suggesting that calsenilin is required for early islet cell migration. Taken together, our results show that zebrafish calsenilin is involved in endocrine cell differentiation and morphogenesis within the pancreas.


Assuntos
Sistema Endócrino/embriologia , Sistema Endócrino/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Pâncreas/embriologia , Pâncreas/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Interatuantes com Canais de Kv/genética , Mutação/genética , Hormônios Pancreáticos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Peixe-Zebra/genética
15.
Diabetologia ; 50(3): 585-95, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17221210

RESUMO

AIMS/HYPOTHESIS: Epithelium-mesenchyme interactions play a major role in pancreas development. Recently, we demonstrated that embryonic pancreatic mesenchyme enhanced progenitor cell proliferation but inhibited endocrine cell differentiation. Here, we investigated the role played by sulphated proteoglycans, which are known to be essential to embryonic development, in this inhibitory effect. MATERIALS AND METHODS: We first determined the expression of the genes encoding glypicans, syndecans and the main glycosaminoglycan chain-modifying enzymes in immature embryonic day (E) 13.5 and more differentiated E17.5 rat pancreases. Next, using an in vitro model of pancreas development, we blocked the action of endogenous sulphated proteoglycans by treating embryonic pancreases in culture with chlorate, an inhibitor of proteoglycan sulphation, and examined the effects on pancreatic endocrine cell differentiation. RESULTS: We first showed that expression of the genes encoding glypicans 1, 2, 3 and 5 and heparan sulphate 2-sulfotransferase decreased between E13.5 and E17.5. We next found that alteration of proteoglycan action by chlorate blocked the inhibitory effect of the mesenchyme on endocrine differentiation. Chlorate-treated pancreases exhibited a dramatic increase in beta cell number in a dose-dependent manner (169-and 375-fold increase with 30 mmol/l and 40 mmol/l chlorate, respectively) and in alpha cell development. Insulin-positive cells that developed in the presence of chlorate exhibited a phenotype of mature cells with regard to the expression of the following genes: pancreatic and duodenal homeobox gene 1 (Pdx1), proprotein convertase subtilisin/kexin type 1 (Pcsk1; previously known as pro-hormone convertase 1/3), proprotein convertase subtilisin/kexin type 2 (Pcsk2; previously known as pro-hormone convertase 2) and solute carrier family 2 (facilitated glucose transporter), member 2 (Slc2a1; previously known as glucose transporter 2). Finally, we showed that chlorate activated endocrine cell development by inducing neurogenin 3 (Neurog3) expression in early endocrine progenitor cells. CONCLUSIONS/INTERPRETATION: We demonstrated that sulphated proteoglycans control pancreatic endocrine cell differentiation. Understanding the mechanism by which sulphated proteoglycans affect beta cell development could be useful in the generation of beta cells from embryonic stem cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Glicosaminoglicanos/genética , Proteoglicanas de Heparan Sulfato/farmacologia , Ilhotas Pancreáticas/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Desenvolvimento Embrionário , Feminino , Glucagon/genética , Insulina/genética , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/embriologia , Proteínas do Tecido Nervoso/genética , Gravidez , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sindecanas/genética , Transcrição Gênica
17.
Diabetologia ; 49(12): 2939-47, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17001468

RESUMO

AIMS/HYPOTHESIS: Beta cell development is sensitive to glucocorticoid levels. Although direct effects of glucocorticoids on pancreatic precursors have been shown to control beta cell mass expansion, indirect effects of these hormones on pancreatic development remain unexplored. This issue was addressed in mice lacking the glucocorticoid receptor (GR) in the whole organism. MATERIALS AND METHODS: The pancreatic phenotype of GR(null/null) mice was studied at fetal ages (embryonic day [E]) E15.5 and E18 by immunohistochemistry and beta cell fraction measurements. To distinguish between direct and indirect effects, mutant E15.5 fetal pancreata were grafted under the kidney capsule of immunodeficient mice and analysed after 1 week. RESULTS: E18 GR(null/null) fetuses had smaller digestive tracts and tiny pancreata. Massive pancreatic disorganisation and apoptosis were observed despite the presence of all cell types. E15.5 GR(null/null) mutants were indistinguishable from wild-type regarding pancreatic size, tissue structure and organisation, beta cell fraction and production of exocrine transcription factor Ptf1a, neurogenin 3 and Pdx-1. Grafting E15.5 GR(null/null) pancreata into a GR-expressing environment rescued the increased apoptosis and mature islets were observed, suggesting that GR(null/null) pancreatic cell death can be attributed to indirect effects of glucocorticoids on this tissue. Heterozygous GR(+/null) mutants with reduced GR numbers showed no apoptosis but increased beta cell fraction at E18 and the adult age, strengthening the importance of an accurate GR dosage on beta cell mass expansion. CONCLUSIONS/INTERPRETATION: Our results provide evidence for GR involvement in pancreatic tissue organisation and survival through indirect effects. GR does not appear necessary for early phases, but its accurate dosage is critical to modulate beta cell mass expansion at later fetal stages, presumably through direct effects.


Assuntos
Glucocorticoides/farmacologia , Pâncreas/crescimento & desenvolvimento , Receptores de Glucocorticoides/genética , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Divisão Celular , Desenvolvimento Embrionário , Genótipo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Pâncreas/efeitos dos fármacos , Pâncreas/embriologia , Transplante de Pâncreas , Reação em Cadeia da Polimerase , Receptores de Glucocorticoides/deficiência , Transdução de Sinais/fisiologia , Ensaio de Cápsula Sub-Renal
18.
Diabetologia ; 48(4): 709-19, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15759109

RESUMO

AIMS/HYPOTHESIS: Gene transfer into pancreatic beta cells, which produce and secrete insulin, is a promising strategy to protect such cells against autoimmune destruction and also to generate beta cells in mass, thereby providing a novel therapeutic approach to treat diabetic patients. Until recently, exogenous DNA has been directly transferred into mature beta cells with various levels of success. We investigated whether exogenous DNA could be stably transferred into pancreatic stem/progenitor cells, which would subsequently differentiate into mature beta cells expressing the transgene. METHODS: We designed transplantation and tissue culture procedures to obtain ex vivo models of pancreatic development. We next constructed recombinant lentiviruses expressing enhanced green fluorescent protein (eGFP) under the control of either the rat insulin promoter or a ubiquitous promoter, and performed viral infection of rat embryonic pancreatic tissue. RESULTS: Embryonic pancreas infected with recombinant lentiviruses resulted in endocrine cell differentiation and restricted cell type expression of the transgene according to the specificity of the promoter used in the viral construct. We next demonstrated that the efficiency of infection could be further improved upon infection of embryonic pancreatic epithelia, followed by their in vitro culture, using conditions that favour endocrine cell differentiation. Under these conditions, endocrine stem/progenitor cells expressing neurogenin 3 are efficiently transduced by recombinant lentiviral vectors. Moreover, when eGFP was placed under the control of the insulin promoter, 70.4% of the developed beta cells were eGFP-expressing cells. All of the eGFP-positive cells were insulin-producing cells. CONCLUSIONS/INTERPRETATION: We have demonstrated that mature rat pancreatic beta cells can be stably modified by infecting pancreatic stem/progenitor cells that undergo endocrine differentiation.


Assuntos
Expressão Gênica/genética , Ilhotas Pancreáticas/metabolismo , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Carboxipeptidases A/metabolismo , Diferenciação Celular/fisiologia , Citomegalovirus/genética , Vetores Genéticos/genética , Glucagon/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hibridização in Situ Fluorescente , Insulina/genética , Insulina/metabolismo , Lentivirus/genética , Masculino , Camundongos , Camundongos SCID , Proteínas do Tecido Nervoso/genética , Pâncreas/citologia , Pâncreas/embriologia , Regiões Promotoras Genéticas/genética , Ratos , Ratos Wistar , Células-Tronco/citologia , Transfecção/métodos , Transplante Heterólogo , Transplantes
19.
Diabetologia ; 48(2): 277-81, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15690149

RESUMO

AIMS/HYPOTHESIS: The fibroblast growth factor (FGF) family consists of 22 members. In rodents, several FGFs are expressed in the pancreas, where they participate in epithelial-mesenchymal interactions. Our objective was to describe the pattern of expression of FGFs in the human embryonic pancreas and to analyse their effect on pancreas development. METHODS: The expression of FGFs was analysed by RT-PCR. To investigate the cell types expressing FGF7 and FGF10, we separated epithelial from mesenchymal cells using immunomagnetic beads linked to E-cadherin antibodies and performed real-time PCR. The effect of FGF7 and FGF10 on proliferation of human embryonic pancreatic epithelial cells was evaluated in vitro by measuring BrdU incorporation. RESULTS: We found that different FGFs are expressed in the human embryonic pancreas, and we focused on FGF7 and FGF10. We defined a new approach to separating epithelial cells (containing the pancreatic progenitor cells) from mesenchymal cells. This allowed us to demonstrate that human embryonic pancreatic mesenchymal cells express both FGF7 and FGF10. We next demonstrated that FGF7 and FGF10 were able to induce the proliferation of the epithelial cells in vitro. CONCLUSION/INTERPRETATION: These findings indicate that it is now possible to efficiently separate human embryonic pancreatic epithelial from mesenchymal cells, an important step to characterize and expand progenitor cells. This method allowed us to demonstrate that human embryonic pancreatic mesenchyme expresses FGF7 and FGF10 that act on epithelial cells to activate their proliferation. Such growth factors could thus be used to expand human embryonic pancreatic epithelial cells.


Assuntos
Células Epiteliais/citologia , Fatores de Crescimento de Fibroblastos/genética , Pâncreas/citologia , Pâncreas/embriologia , Sequência de Bases , Caderinas/genética , Divisão Celular , Ciclofilinas/genética , Primers do DNA , Fator 10 de Crescimento de Fibroblastos , Fator 7 de Crescimento de Fibroblastos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vimentina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...