Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Diabetologia ; 67(6): 1066-1078, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38630142

RESUMO

AIMS/HYPOTHESIS: Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS: We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS: We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION: This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.


Assuntos
Diferenciação Celular , Proliferação de Células , Imageamento Tridimensional , Pâncreas , Humanos , Pâncreas/embriologia , Pâncreas/citologia , Pâncreas/metabolismo , Diferenciação Celular/fisiologia , Feminino , Células-Tronco/citologia , Células-Tronco/metabolismo , Gravidez , Insulina/metabolismo
2.
Cells ; 12(1)2022 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-36611907

RESUMO

Type 1 diabetes (T1D) is an auto-immune disease characterized by the progressive destruction of insulin-producing pancreatic beta cells. While beta cells are the target of the immune attack, the other islet endocrine cells, namely the alpha and delta cells, can also be affected by the inflammatory milieu. Here, using a flow cytometry-based strategy, we compared the impact of IFNγ, one of the main cytokines involved in T1D, on the three endocrine cell subsets isolated from C57BL/6 mouse islets. RNA-seq analyses revealed that alpha and delta cells exposed in vitro to IFNγ display a transcriptomic profile very similar to that of beta cells, with an increased expression of inflammation key genes such as MHC class I molecules, the CXCL10 chemokine and the programmed death-ligand 1 (PD-L1), three hallmarks of IFNγ signaling. Interestingly, at low IFNγ concentration, we observed two beta cell populations (responders and non-responders) based on PD-L1 protein expression. Our data indicate that this differential sensitivity relies on the location of the cells within the islet rather than on the existence of two different beta cells subsets. The same findings were corroborated by the in vivo analysis of pancreatic islets from the non-obese diabetic mouse model of T1D, showing more intense PD-L1 staining on endocrine cells close to immune infiltrate. Collectively, our work demonstrates that alpha and delta cells are as sensitive as beta cells to IFNγ, and suggests a gradual diffusion of the cytokine into an islet. These observations provide novel insights into the in situ inflammatory processes occurring in T1D progression.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Camundongos , Animais , Diabetes Mellitus Tipo 1/genética , Antígeno B7-H1/metabolismo , Camundongos Endogâmicos C57BL , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Insulina/metabolismo , Interferon gama/metabolismo , Citocinas/metabolismo
3.
Islets ; 13(1-2): 10-23, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33641620

RESUMO

During the secondary transition of rodent pancreatic development, mainly between E12.5 and E15.5 in mice, exocrine and endocrine populations differentiate from pancreatic progenitors. Here we describe an experimental system for its study in vitro. First, we show that spheres derived from dissociated E12.5 mouse pancreases differentiate within 7 days into most pancreatic exocrine and endocrine cell types, including beta cells. The proportion and spatial repartition of the different endocrine populations mirror those observed during normal development. Thus, dissociation and culture do not impair the developmental events affecting pancreatic progenitors during the secondary transition. Moreover, dissociated cells from mouse E12.5 pancreas were transduced with ecotropic MLV-based retroviral vectors or, though less efficiently, with a mixture of ALV(A)-based retroviral vectors and gesicles containing the TVA (Tumor Virus A) receptor. As an additional improvement, we also created a transgenic mouse line expressing TVA under the control of the 4.5 kB pdx1 promoter (pdx1-TVA). We demonstrate that pancreatic progenitors from dissociated pdx1-TVA pancreas can be specifically transduced by ALV(A)-based retroviral vectors. Using this model, we expressed an activated mutant of the YAP transcriptional co-activator in pancreatic progenitors. These experiments indicate that deregulated YAP activity reduces endocrine and exocrine differentiation in the resulting spheres, confirming and extending previously published data. Thus, our experimental model recapitulates in vitro the crucial developmental decisions arising at the secondary transition and provides a convenient tool to study their genetic control.


Assuntos
Proteínas de Homeodomínio , Células Secretoras de Insulina , Animais , Diferenciação Celular , Camundongos , Camundongos Transgênicos , Organogênese , Pâncreas
4.
Diabetes ; 68(4): 761-773, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30655386

RESUMO

Bromodomain and extraterminal (BET) proteins are epigenetic readers that interact with acetylated lysines of histone tails. Recent studies have demonstrated their role in cancer progression because they recruit key components of the transcriptional machinery to modulate gene expression. However, their role during embryonic development of the pancreas has never been studied. Using mouse embryonic pancreatic explants and human induced pluripotent stem cells (hiPSCs), we show that BET protein inhibition with I-BET151 or JQ1 enhances the number of neurogenin3 (NEUROG3) endocrine progenitors. In mouse explants, BET protein inhibition further led to increased expression of ß-cell markers but in the meantime, strongly downregulated Ins1 expression. Similarly, although acinar markers, such as Cpa1 and CelA, were upregulated, Amy expression was repressed. In hiPSCs, BET inhibitors strongly repressed C-peptide and glucagon during endocrine differentiation. Explants and hiPSCs were then pulsed with BET inhibitors to increase NEUROG3 expression and further chased without inhibitors. Endocrine development was enhanced in explants with higher expression of insulin and maturation markers, such as UCN3 and MAFA. In hiPSCs, the outcome was different because C-peptide expression remained lower than in controls, but ghrelin expression was increased. Altogether, by using two independent models of pancreatic development, we show that BET proteins regulate multiple aspects of pancreatic development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteínas/antagonistas & inibidores , Animais , Azepinas/farmacologia , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Triazóis/farmacologia
5.
Cell Cycle ; 13(14): 2221-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24870561

RESUMO

Type 2 diabetes is caused by a limited capacity of insulin-producing pancreatic ß cells to increase their mass and function in response to insulin resistance. The signaling pathways that positively regulate functional ß cell mass have not been fully elucidated. DYRK1A (also called minibrain/MNB) is a member of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) family. A significant amount of data implicates DYRK1A in brain growth and Down syndrome, and recent data indicate that Dyrk1A haploinsufficient mice have a low functional ß cell mass. Here we ask whether Dyrk1A upregulation could be a way to increase functional ß cell mass. We used mice overexpressing Dyrk1A under the control of its own regulatory sequences (mBACTgDyrk1A). These mice exhibit decreased glucose levels and hyperinsulinemia in the fasting state. Improved glucose tolerance is observed in these mice as early as 4 weeks of age. Upregulation of Dyrk1A in ß cells induces expansion of ß cell mass through increased proliferation and cell size. Importantly, mBACTgDyrk1A mice are protected against high-fat-diet-induced ß cell failure through increase in ß cell mass and insulin sensitivity. These studies show the crucial role of the DYRK1A pathway in the regulation of ß cell mass and carbohydrate metabolism in vivo. Activating the DYRK1A pathway could thus represent an innovative way to increase functional ß cell mass.


Assuntos
Glicemia/metabolismo , Proliferação de Células , Células Secretoras de Insulina/enzimologia , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Tirosina Quinases/biossíntese , Animais , Biomarcadores/sangue , Tamanho Celular , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/prevenção & controle , Dieta Hiperlipídica , Genótipo , Hiperinsulinismo/sangue , Hiperinsulinismo/enzimologia , Hiperinsulinismo/genética , Insulina/sangue , Células Secretoras de Insulina/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Transdução de Sinais , Fatores de Tempo , Regulação para Cima , Quinases Dyrk
6.
Exp Cell Res ; 322(2): 236-48, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24503054

RESUMO

Diabetes is caused by loss or dysfunction of pancreatic beta cells. Generation of beta cells in vitro is a promising strategy to develop a full-scale cell therapy against diabetes, and the development of methods without gene transfer may provide safer protocols for human therapy. Here we show that thyroid hormone receptors are expressed in embryonic murine pancreas. Addition of the thyroid hormone T3 in an ex vivo culture model of embryonic (E12.5) dorsal pancreas, mimicking embryonic pancreatic development, promoted an increase of ductal cell number at expenses of the acinar compartment. Double labeled cells expressing specific markers for ductal and acinar cells were observed, suggesting cell reprogramming. Increased mRNA levels of the pro-endocrine gene Ngn3 and an increased number of beta cells were detected in cultures treated previously with T3 suggesting that ductal cells promoted by T3 can subsequently differentiate into endocrine cells. So, indirectly, T3 induced endocrine differentiation. Moreover, T3 induced the expression of the pro-endocrine gene Ngn3 in the acinar 266-6 cell line. The pro-endocrine effect of T3 in the pancreatic explants and in the acinar cell line, was abrogated by the Akt inhibitor Ly294002 indicating the involvement of Akt signaling in this process. Altogether we show numerous evidences that define T3 as a promising candidate to generate endocrine cells from exocrine tissue, using ectopically gene expression free protocols, for cell therapy against diabetes.


Assuntos
Células Acinares/citologia , Células Secretoras de Insulina/citologia , Pâncreas/embriologia , Ductos Pancreáticos/citologia , Tri-Iodotironina/farmacologia , Células Acinares/efeitos dos fármacos , Células Acinares/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Morfolinas/farmacologia , Técnicas de Cultura de Órgãos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Ductos Pancreáticos/efeitos dos fármacos , Ductos Pancreáticos/metabolismo , Gravidez , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores dos Hormônios Tireóideos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Diabetologia ; 57(5): 960-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24477974

RESUMO

AIMS/HYPOTHESIS: Growth factors and nutrients are important regulators of pancreatic beta cell mass and function. However, the signalling pathways by which these factors modulate these processes have not yet been fully elucidated. DYRK1A (also named minibrain/MNB) is a member of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) family that has been conserved across evolution. A significant amount of data implicates DYRK1A in brain growth and function, as well as in neurodegenerative processes in Alzheimer's disease and Down's syndrome. We investigated here whether DYRK1A would be an attractive candidate for beta cell growth modulation. METHODS: To study the role of DYRK1A in beta cell growth, we used Dyrk1a-deficient mice. RESULTS: We show that DYRK1A is expressed in pancreatic islets and provide evidence that changes in Dyrk1a gene dosage in mice strongly modulate glycaemia and circulating insulin levels. Specifically, Dyrk1a-haploinsufficient mice show severe glucose intolerance, reduced beta cell mass and decreased beta cell proliferation. CONCLUSIONS/INTERPRETATION: Taken together, our data indicate that DYRK1A is a critical kinase for beta cell growth as Dyrk1a-haploinsufficient mice show a diabetic profile.


Assuntos
Diabetes Mellitus Experimental/genética , Células Secretoras de Insulina/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Proliferação de Células , Haploinsuficiência , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Transdução de Sinais , Quinases Dyrk
8.
Diabetes ; 62(10): 3479-87, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23835344

RESUMO

While sporadic human genetic studies have permitted some comparisons between rodent and human pancreatic development, the lack of a robust experimental system has not permitted detailed examination of human pancreatic development. We previously developed a xenograft model of immature human fetal pancreas grafted under the kidney capsule of immune-incompetent mice, which allowed the development of human pancreatic ß-cells. Here, we compared the development of human and murine fetal pancreatic grafts either under skeletal muscle epimysium or under the renal capsule. We demonstrated that human pancreatic ß-cell development occurs more slowly (weeks) than murine pancreas (days) both by differentiation of pancreatic progenitors and by proliferation of developing ß-cells. The superficial location of the skeletal muscle graft and its easier access permitted in vivo lentivirus-mediated gene transfer with a green fluorescent protein-labeled construct under control of the insulin or elastase gene promoter, which targeted ß-cells and nonendocrine cells, respectively. This model of engraftment under the skeletal muscle epimysium is a new approach for longitudinal studies, which allows localized manipulation to determine the regulation of human pancreatic development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diabetes Mellitus Experimental/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/citologia , Transativadores/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Diabetes Mellitus Experimental/genética , Feminino , Feto/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Transferência de Genes , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/fisiologia , Lentivirus/genética , Camundongos , Microscopia Eletrônica , Pâncreas/embriologia , Pâncreas/patologia , Gravidez , Fatores de Tempo
9.
Diabetes ; 61(2): 409-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22210321

RESUMO

Leucine (Leu) is an essential branched-chain amino acid, which activates the mammalian target of rapamycin (mTOR) signaling pathway. The effect of Leu on cell differentiation during embryonic development is unknown. Here, we show that Leu supplementation during pregnancy significantly increased fetal body weight, caused fetal hyperglycemia and hypoinsulinemia, and decreased the relative islet area. We also used rat embryonic pancreatic explant culture for elucidating the mechanism of Leu action on ß-cell development. We found that in the presence of Leu, differentiation of pancreatic duodenal homeobox-1-positive progenitor cells into neurogenin3-positive endocrine progenitor cells was inefficient and resulted in decreased ß-cell formation. Mechanistically, Leu increases the intracellular levels of hypoxia-inducible factor 1-α, a repressor of endocrine fate in the pancreas, by activating the mTOR complex 1 signaling pathway. Collectively, our findings indicate that Leu supplementation during pregnancy could potentially increase the risk of type 2 diabetes mellitus by inhibiting the differentiation of pancreatic endocrine progenitor cells during a susceptible period of fetal life.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Leucina/administração & dosagem , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/análise , Suplementos Nutricionais , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Proteínas do Tecido Nervoso/análise , Gravidez , Ratos , Ratos Wistar , Sirolimo/farmacologia , Células-Tronco/efeitos dos fármacos , Transativadores , Fatores de Transcrição/fisiologia
10.
Diabetes ; 56(5): 1248-58, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17322477

RESUMO

The importance of mesenchymal-epithelial interactions for normal development of the pancreas was recognized in the early 1960s, and mesenchymal signals have been shown to control the proliferation of early pancreatic progenitor cells. The mechanisms by which the mesenchyme coordinates cell proliferation and differentiation to produce the normal number of differentiated pancreatic cells are not fully understood. Here, we demonstrate that the mesenchyme positively controls the final number of beta-cells that develop from early pancreatic progenitor cells. In vitro, the number of beta-cells that developed from rat embryonic pancreatic epithelia was larger in cultures with mesenchyme than without mesenchyme. The effect of mesenchyme was not due to an increase in beta-cell proliferation but was due to increased proliferation of early pancreatic duodenal homeobox-1 (PDX1)-positive progenitor cells, as confirmed by bromodeoxyuridine incorporation. Consequently, the window during which early PDX1(+) pancreatic progenitor cells differentiated into endocrine progenitor cells expressing Ngn3 was extended. Fibroblast growth factor 10 mimicked mesenchyme effects on proliferation of early PDX1(+) progenitor cells and induction of Ngn3 expression. Taken together, our results indicate that expansion of early PDX1(+) pancreatic progenitor cells represents a way to increase the final number of beta-cells developing from early embryonic pancreas.


Assuntos
Células Epiteliais/fisiologia , Células Secretoras de Insulina/fisiologia , Pâncreas/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/efeitos dos fármacos , Embrião de Mamíferos , Células Epiteliais/citologia , Fator 10 de Crescimento de Fibroblastos/farmacologia , Homeostase , Hibridização In Situ , Insulina/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Técnicas de Cultura de Órgãos , Pâncreas/enzimologia , Pâncreas/fisiologia , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar
11.
Diabetes ; 52(8): 2035-42, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12882920

RESUMO

Islets of Langerhans are micro-organs scattered throughout the pancreas that contain insulin-producing cells, called beta-cells. Although new light has been recently shed on beta-cell development, information on the phenotype and location of beta-stem cells remains scarce. Here, we provide evidence that beta-stem cells are slow-cycling cells located within and around the islets of Langerhans. First, using a bromodeoxyuridine (BrdU) pulse/chase approach, we detected BrdU-retaining cells in vivo in the islet area of rat pancreata. These cells were negative for endocrine markers but expressed Pdx1, a marker for pancreatic stem cells. Next, using an in vitro model that mimicked endocrine cell development, we found that BrdU-retaining cells were capable of differentiating into beta-cells. Taken together, these observations demonstrate that BrdU retention is a property of beta-stem cells.


Assuntos
Ilhotas Pancreáticas/citologia , Células-Tronco/citologia , Animais , Antimetabólitos/farmacocinética , Bromodesoxiuridina/farmacocinética , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Feminino , Técnicas In Vitro , Ilhotas Pancreáticas/fisiologia , Gravidez , Ratos , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...