Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Dis Model Mech ; 12(1)2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30679186

RESUMO

Islet inflammation and cytokine production are implicated in pancreatic ß-cell dysfunction and diabetes pathogenesis. However, we lack therapeutics to protect the insulin-producing ß-cells from inflammatory damage. Closing this clinical gap requires the establishment of new disease models of islet inflammation to facilitate screening efforts aimed at identifying new protective agents. Here, we have developed a genetic model of Interleukin-1ß (Il-1ß)-driven islet inflammation in zebrafish, a vertebrate that allows for non-invasive imaging of ß-cells and in vivo drug discovery. Live imaging of immune cells and ß-cells in our model revealed dynamic migration, increased visitation and prolonged macrophage retention in the islet, together with robust activation of NF-κB signalling in ß-cells. We find that Il-1ß-mediated inflammation does not cause ß-cell destruction but, rather, it impairs ß-cell function and identity. In vivo, ß-cells exhibit impaired glucose-stimulated calcium influx and reduced expression of genes involved in function and maturity. These defects are accompanied by α-cell expansion, glucose intolerance and hyperglycemia following a glucose challenge. Notably, we show that a medicinal plant derivative (wedelolactone) is capable of reducing the immune-cell infiltration while also ameliorating the hyperglycemic phenotype of our model. Importantly, these anti-diabetic properties in zebrafish are predictive of wedelolactone's efficacy in protecting rodent and human islets from cytokine-induced apoptosis. In summary, this new zebrafish model of diabetes opens a window to study the interactions between immune and ß-cells in vivo, while also allowing the identification of therapeutic agents for protecting ß-cells from inflammation.


Assuntos
Produtos Biológicos/farmacologia , Cumarínicos/farmacologia , Inflamação/patologia , Células Secretoras de Insulina/patologia , Animais , Animais Geneticamente Modificados , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Citocinas/farmacologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Glucose/farmacologia , Humanos , Hiperglicemia/genética , Hiperglicemia/patologia , Inflamação/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Larva/efeitos dos fármacos , Larva/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Modelos Genéticos , Imagem com Lapso de Tempo , Transcrição Gênica/efeitos dos fármacos , Peixe-Zebra
2.
Elife ; 72018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29624168

RESUMO

The pancreatic islet, a cellular community harboring the insulin-producing beta-cells, is known to undergo age-related alterations. However, only a handful of signals associated with aging have been identified. By comparing beta-cells from younger and older zebrafish, here we show that the aging islets exhibit signs of chronic inflammation. These include recruitment of tnfα-expressing macrophages and the activation of NF-kB signaling in beta-cells. Using a transgenic reporter, we show that NF-kB activity is undetectable in juvenile beta-cells, whereas cells from older fish exhibit heterogeneous NF-kB activity. We link this heterogeneity to differences in gene expression and proliferation. Beta-cells with high NF-kB signaling proliferate significantly less compared to their neighbors with low activity. The NF-kB signalinghi cells also exhibit premature upregulation of socs2, an age-related gene that inhibits beta-cell proliferation. Together, our results show that NF-kB activity marks the asynchronous decline in beta-cell proliferation with advancing age.


Assuntos
Envelhecimento , Proliferação de Células , Mediadores da Inflamação/metabolismo , Inflamação/patologia , Células Secretoras de Insulina/patologia , NF-kappa B/metabolismo , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Células Cultivadas , Perfilação da Expressão Gênica , Inflamação/imunologia , Inflamação/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , NF-kappa B/genética , Transdução de Sinais , Análise de Célula Única , Ativação Transcricional , Peixe-Zebra/imunologia
3.
Sci Rep ; 7(1): 3994, 2017 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-28652605

RESUMO

The pancreatic beta-cells control glucose homeostasis by secreting insulin in response to nutrient intake. The number of beta-cells is under tight metabolic control, as this number increases with higher nutrient intake. However, the signaling pathways matching nutrition with beta-cell mass plasticity remain poorly defined. By applying pharmacological and genetic manipulations, we show that reactive oxygen species (ROS) regulate dose-dependently beta-cell proliferation in vivo and in vitro. In particular, reducing ROS levels in beta-cells blocks their proliferation in response to nutrients. Using a non-invasive genetic sensor of intracellular hydrogen peroxide (H2O2), we reveal that glucose can directly increase the levels of H2O2. Furthermore, a moderate increase in H2O2 levels can stimulate beta-cell proliferation. Interestingly, while high H2O2 levels are inhibitory to beta-cell proliferation, they expand beta-cell mass in vivo by inducing rapid beta-cell neogenesis. Our study thus reveals a ROS-level-dependent mechanism linking nutrients with beta-cell mass plasticity. Hence, given the requirement of ROS for beta-cell mass expansion, antioxidant therapies should be applied with caution in diabetes.


Assuntos
Plasticidade Celular , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Espécies Reativas de Oxigênio , Animais , Animais Geneticamente Modificados , Linhagem Celular , Proliferação de Células , Peróxido de Hidrogênio/metabolismo , Peixe-Zebra
4.
Nat Commun ; 8(1): 664, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28939870

RESUMO

The proliferative and functional heterogeneity among seemingly uniform cells is a universal phenomenon. Identifying the underlying factors requires single-cell analysis of function and proliferation. Here we show that the pancreatic beta-cells in zebrafish exhibit different growth-promoting and functional properties, which in part reflect differences in the time elapsed since birth of the cells. Calcium imaging shows that the beta-cells in the embryonic islet become functional during early zebrafish development. At later stages, younger beta-cells join the islet following differentiation from post-embryonic progenitors. Notably, the older and younger beta-cells occupy different regions within the islet, which generates topological asymmetries in glucose responsiveness and proliferation. Specifically, the older beta-cells exhibit robust glucose responsiveness, whereas younger beta-cells are more proliferative but less functional. As the islet approaches its mature state, heterogeneity diminishes and beta-cells synchronize function and proliferation. Our work illustrates a dynamic model of heterogeneity based on evolving proliferative and functional beta-cell states.Βeta-cells have recently been shown to be heterogeneous with regard to morphology and function. Here, the authors show that ß-cells in zebrafish switch from proliferative to functional states with increasing time since ß-cell birth, leading to functional and proliferative heterogeneity.


Assuntos
Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Linhagem da Célula , Proliferação de Células , Técnicas Citológicas/métodos , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Glucose/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/embriologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Urocortinas/metabolismo , Peixe-Zebra/genética
5.
J Vis Exp ; (88)2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24961304

RESUMO

The zebrafish is an important model to understand the cell and molecular biology of organ and appendage regeneration. However, molecular strategies to employ reverse genetics have not yet been adequately developed to assess gene function in regeneration or tissue homeostasis during larval stages after zebrafish embryogenesis, and several tissues within the zebrafish larva are difficult to target. Intraventricular injections of gene-specific morpholinos offer an alternative method for the current inability to genomically target zebrafish genes in a temporally controlled manner at these stages. This method allows for complete dispersion and subsequent incorporation of the morpholino into various tissues throughout the body, including structures that were formerly impossible to reach such as those in the larval caudal fin, a structure often used to noninvasively research tissue regeneration. Several genes activated during larval finfold regeneration are also present in regenerating adult vertebrate tissues, so the larva is a useful model to understand regeneration in adults. This morpholino dispersion method allows for the quick and easy identification of genes required for the regeneration of larval tissues as well as other physiological phenomena regulating tissue homeostasis after embryogenesis. Therefore, this delivery method provides a currently needed strategy for temporal control to the evaluation of gene function after embryogenesis. 


Assuntos
Morfolinos/administração & dosagem , Morfolinos/genética , Regeneração/genética , Transfecção/métodos , Peixe-Zebra/fisiologia , Animais , Vias de Administração de Medicamentos , Fluoresceína/química , Ventrículos do Coração , Larva , Peixe-Zebra/embriologia , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA