Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 503: 43-52, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37597605

RESUMO

Transmembrane p24 trafficking protein 10 (TMED10) is a conserved vesicle trafficking protein. It is dysregulated in Alzheimer disease and plays a pivotal role in the pathogenesis of Alzheimer disease. In addition to the brain, TMED10 is highly expressed in the exocrine pancreas; however, its biological functions and underlying mechanisms remain largely unknown. We studied reduced Tmed10 in zebrafish embryos by morpholino oligonucleotide knockdown and CRISPR-Cas9 mutagenesis. Tmed10-deficient embryos showed extensive loss of acinar mass and impaired acinar differentiation. TMED10 has been reported to have an inhibitory effect on γ-secretase. As one of the substrates of γ-secretase, membrane-bound ß-catenin was significantly reduced in Tmed10-deficient embryos. Increased γ-secretase activity in wild-type embryos resulted in a phenotype similar to that of tmed10 mutants. And the mutant phenotype could be rescued by treatment with the γ-secretase inhibitor, N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester (DAPT). In addition, the reduced membrane-bound ß-catenin was accompanied with up-regulated ß-catenin target genes in Tmed10-deficient embryos. Overexpression of ß-catenin signaling inhibitor Dickkopf-1 (DKK-1) could rescue the exocrine pancreas defects. Taken together, our study reveals that Tmed10 regulates exocrine pancreatic differentiation through γ-secretase. Reduced membrane-bound ß-catenin, accompanied with hyperactivation of ß-catenin signaling, is an important cause of exocrine pancreas defects in Tmed10-deficient embryos. Our study reaffirms the importance of appropriate ß-catenin signaling in exocrine pancreas development. These findings may provide a theoretical basis for the development of treatment strategies for TMED10-related diseases.


Assuntos
Doença de Alzheimer , Proteínas de Transporte Nucleocitoplasmático , Pâncreas Exócrino , Animais , Secretases da Proteína Precursora do Amiloide/genética , beta Catenina/genética , Larva , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/metabolismo , Peixe-Zebra/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo
2.
Adv Exp Med Biol ; 1236: 65-85, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32304069

RESUMO

The pancreas is a glandular organ responsible for diverse homeostatic functions, including hormone production from the endocrine islet cells to regulate blood sugar levels and enzyme secretion from the exocrine acinar cells to facilitate food digestion. These pancreatic functions are essential for life; therefore, preserving pancreatic function is of utmost importance. Pancreas dysfunction can arise either from developmental disorders or adult onset disease, both of which are caused by defects in shared molecular pathways. In this chapter, we discuss what is known about the molecular mechanisms controlling pancreas development, how disruption of these mechanisms can lead to developmental defects and disease, and how essential pancreas functions can be modeled using human pluripotent stem cells. At the core of understanding of these molecular processes are animal model studies that continue to be essential for elucidating the mechanisms underlying human pancreatic functions and diseases.


Assuntos
Modelos Animais , Organogênese , Pâncreas/embriologia , Pâncreas/patologia , Células Acinares/metabolismo , Células Acinares/patologia , Animais , Humanos , Pâncreas/citologia , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/patologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-32200130

RESUMO

MicroRNAs (miRNAs), as important regulators of post-transcriptional gene expression, play important roles in the occurrence and function of organs. In this study, morpholino (MO) knockdown of miR-462/miR-731 was used to investigate the potential mechanisms of the miR-462-731 cluster during zebrafish liver development. The results showed significant reduction of digestive organs, especially liver and exocrine pancreas after the miR-462/miR-731 knockdown, and those phenotypes could be partially rescued by corresponding miRNA duplex. Acinar cells of the exocrine pancreas were also severely affected with pancreatic insufficiency. In particular, knockdown of miR-462 caused pancreas morphogenesis abnormity with specific bilateral exocrine pancreas. Additionally, it was found that miR-731 played a role in liver and exocrine pancreas development by directly targeting dkk3b, instead of the down-regulation of Wnt/ß-catenin signaling. These findings contribute significantly to our understanding of molecular mechanisms of miR-462-731 cluster in development of digestive organs.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Pâncreas Exócrino/embriologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Pâncreas Exócrino/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
4.
Development ; 147(7)2020 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165492

RESUMO

Although the developing pancreas is exquisitely sensitive to nutrient supply in utero, it is not entirely clear how nutrient-driven post-translational modification of proteins impacts the pancreas during development. We hypothesized that the nutrient-sensing enzyme O-GlcNAc transferase (Ogt), which catalyzes an O-GlcNAc-modification onto key target proteins, integrates nutrient-signaling networks to regulate cell survival and development. In this study, we investigated the heretofore unknown role of Ogt in exocrine and endocrine islet development. By genetic manipulation in vivo and by using morphometric and molecular analyses, such as immunofluorescence imaging and single cell RNA sequencing, we show the first evidence that Ogt regulates pancreas development. Genetic deletion of Ogt in the pancreatic epithelium (OgtKOPanc) causes pancreatic hypoplasia, in part by increased apoptosis and reduced levels of of Pdx1 protein. Transcriptomic analysis of single cell and bulk RNA sequencing uncovered cell-type heterogeneity and predicted upstream regulator proteins that mediate cell survival, including Pdx1, Ptf1a and p53, which are putative Ogt targets. In conclusion, these findings underscore the requirement of O-GlcNAcylation during pancreas development and show that Ogt is essential for pancreatic progenitor survival, providing a novel mechanistic link between nutrients and pancreas development.


Assuntos
Acetilglucosamina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Nutrientes/farmacologia , Pâncreas Exócrino/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Animais , Embrião de Mamíferos , Feminino , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilglucosaminiltransferases/efeitos dos fármacos , N-Acetilglucosaminiltransferases/metabolismo , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Artigo em Inglês | MEDLINE | ID: mdl-32061751

RESUMO

Carboxyl ester lipase (Cel), is a lipolytic enzyme secreted by the pancreas, which hydrolyzes various species of lipids in the gut. Cel is also secreted by mammary gland during lactation and exists in breast milk. It facilitates dietary fat digestion and absorption, thus contributing to normal infant development. This study aimed to examine whether the Cel in zebrafish embryos has a similar role of maternal lipid utilization as in human infants, and how Cel contributes to the utilization of yolk lipids in zebrafish. The cel1 and cel2 genes were expressed ubiquitously in the blastodisc and yolk syncytial layer before 24 hpf, and in the exocrine pancreas after 72 hpf. The cel1 and cel2 morphants exhibited developmental retardation and yolk sac retention. The total cholesterol, cholesterol ester, free cholesterol, and triglyceride were reduced in the morphants' body while accumulated in the yolk (except triglyceride). The FFA content of whole embryos was much lower in morphants than in standard controls. Moreover, the delayed development in cel (cel1/cel2) double morphants was partially rescued by FFA and cholesterol supplementation. Delayed and weakened cholesterol ester transport to the brain and eyes was observed in cel morphants. Correspondingly, shrunken midbrain tectum, microphthalmia, pigmentation-delayed eyes as well as down-regulated Shh target genes were observed in the CNS of double morphants. Interestingly, cholesterol injections reversed these CNS alterations. Our findings suggested that cel genes participate in the lipid releasing from yolk sac to developing body, thereby contributing to the normal growth rate and CNS development in zebrafish.


Assuntos
Carboxilesterase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transtornos do Crescimento/genética , Saco Vitelino/enzimologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Carboxilesterase/genética , Sistema Nervoso Central/embriologia , Colesterol/metabolismo , Ésteres do Colesterol/metabolismo , Modelos Animais de Doenças , Embrião não Mamífero , Desenvolvimento Embrionário , Técnicas de Silenciamento de Genes , Transtornos do Crescimento/embriologia , Transtornos do Crescimento/enzimologia , Proteínas Hedgehog/metabolismo , Humanos , Metabolismo dos Lipídeos , Morfolinos/administração & dosagem , Morfolinos/genética , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/enzimologia , Triglicerídeos/metabolismo , Saco Vitelino/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
6.
J Morphol ; 281(1): 110-121, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31714625

RESUMO

This study report about the differentiation of pancreatic acinar tissue in grass snake, Natrix natrix, embryos using light microscopy, transmission electron microscopy, and immuno-gold labeling. Differentiation of acinar cells in the embryonic pancreas of the grass snake is similar to that of other amniotes. Pancreatic acini occurred for the first time at Stage VIII, which is the midpoint of embryonic development. Two pattern of acinar cell differentiation were observed. The first involved formation of zymogen granules followed by cell migration from ducts. In the second, one zymogen granule was formed at the end of acinar cell differentiation. During embryonic development in the pancreatic acini of N. natrix, five types of zymogen granules were established, which correlated with the degree of their maturation and condensation. Within differentiating acini of the studied species, three types of cells were present: acinar, centroacinar, and endocrine cells. The origin of acinar cells as well as centroacinar cells in the pancreas of the studied species was the pancreatic ducts, which is similar as in other vertebrates. In the differentiating pancreatic acini of N. natrix, intermediate cells were not present. It may be related to the lack of transdifferentiation activity of acinar cells in the studied species. Amylase activity of exocrine pancreas was detected only at the end of embryonic development, which may be related to animal feeding after hatching from external sources that are rich in carbohydrates and presence of digestive enzymes in the egg yolk. Mitotic division of acinar cells was the main mechanism of expansion of acinar tissue during pancreas differentiation in the grass snake embryos.


Assuntos
Células Acinares/citologia , Colubridae/embriologia , Embrião não Mamífero/anatomia & histologia , Pâncreas Exócrino/embriologia , Células Acinares/ultraestrutura , Animais , Diferenciação Celular , Embrião não Mamífero/ultraestrutura , Desenvolvimento Embrionário , Feminino
7.
Genesis ; 58(2): e23345, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31705616

RESUMO

Myosin phosphatase targeting subunit 1 (Mypt1) is the regulatory subunit of myosin phosphatase which dephosphorylates the light chain of myosin II to inhibit its contraction. Although biochemical properties of Mypt1 have been characterized in detail, its biological functions in organisms are not well understood. The zebrafish mypt1 sq181 allele was found defective in the ventral pancreatic bud and extrapancreatic duct development, resulting in dysplasia of exocrine pancreas. In mypt1 sq181 mutant, the early growth of the ventral pancreatic bud was initiated but failed to expand due to impaired cell proliferation and increased cell apoptosis. As Mypt1 is essential for cell migration, the loss-of-function of Mypt1 in the mutant disrupted the lateral plate mesoderm migration during gut looping, therefore, altering the Bmp2a expression pattern within it, and eventually leading to impaired Bmp signaling in the adjacent exocrine pancreas. Overexpression of bmp2a could rescue the development of exocrine pancreas, suggesting that the impaired Bmp2a signaling is responsible for the pancreatic development defects. Bmp2a has been reported to promote the early specification of the ventral pancreatic bud, and our study reveals that it continues to serve as a cell proliferation/survival signal to ensure pancreatic bud growth properly in zebrafish.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Pâncreas Exócrino/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Apoptose , Proteína Morfogenética Óssea 2/genética , Regulação da Expressão Gênica no Desenvolvimento , Mutação com Perda de Função , Fosfatase de Miosina-de-Cadeia-Leve/genética , Pâncreas Exócrino/embriologia , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
8.
J Morphol ; 279(6): 724-746, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29464754

RESUMO

We analyzed the development of the pancreatic ducts in grass snake Natrix natrix L. embryos with special focus on the three-dimensional (3D)-structure of the duct network, ultrastructural differentiation of ducts with attention to cell types and lumen formation. Our results indicated that the system of ducts in the embryonic pancreas of the grass snake can be divided into extralobular, intralobular, and intercalated ducts, similarly as in other vertebrate species. However, the pattern of branching was different from that in other vertebrates, which was related to the specific topography of the snake's internal organs. The process of duct remodeling in Natrix embryos began when the duct walls started to change from multilayered to single-layered and ended together with tube formation. It began in the dorsal pancreatic bud and proceeded toward the caudal direction. The lumen of pancreatic ducts differentiated by cavitation because a population of centrally located cells was cleared through cell death resembling anoikis. During embryonic development in the pancreatic duct walls of the grass snake four types of cells were present, that is, principal, endocrine, goblet, and basal cells, which is different from other vertebrate species. The principal cells were electron-dense, contained indented nuclei with abundant heterochromatin, microvilli and cilia, and were connected by interdigitations of lateral membranes and junctional complexes. The endocrine cells were electron-translucent and some of them included endocrine granules. The goblet cells were filled with large granules with nonhomogeneous, moderately electron-dense material. The basal cells were small, electron-dense, and did not reach the duct lumen.


Assuntos
Colubridae/embriologia , Desenvolvimento Embrionário , Pâncreas Exócrino/embriologia , Animais , Pâncreas Exócrino/ultraestrutura
9.
Eur J Histochem ; 61(1): 2761, 2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-28348424

RESUMO

At present, data on the endocannabinoid system expression and distribution in the pancreatic gland appear scarce and controversial as descriptions are limited to humans and laboratory animals. Since the bovine pancreas is very similar to the human in endocrine portion development and control, studies on the fetal gland could prove to be very interesting, as an abnormal maternal condition during late pregnancy may be a predisposing trigger for adult metabolic disorders. The present investigation studied cannabinoid receptor type 2 presence and distribution in the bovine fetal pancreas towards the end of gestation. Histological analyses revealed numerous endocrinal cell clusters or islets which were distributed among exocrine adenomeri in connectival tissue. Immunohistochemistry showed that endocrine-islets contained some CB2-positive cells with a very peculiar localization that is a few primarily localized at the edges of islets and some of them also scattered in the center of the cluster. Characteristically, also the epithelium of the excretory ducts and the smooth muscle layers of the smaller arteries, in the interlobular glandular septa, tested positive for the CB2 endocannabinoid receptor. Conse - quently, the endocannabinoid system, via the cannabinoid receptor type 2, was hypothesized to play a major role in controlling pancreas function from normal fetal development to correct metabolic functioning in adulthood.


Assuntos
Feto/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ilhotas Pancreáticas/embriologia , Pâncreas Exócrino/embriologia , Receptor CB2 de Canabinoide/biossíntese , Animais , Bovinos , Feminino , Feto/citologia , Humanos , Ilhotas Pancreáticas/citologia , Pâncreas Exócrino/citologia , Gravidez
10.
Dis Model Mech ; 10(3): 307-321, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28138096

RESUMO

The exocrine pancreas displays a significant capacity for regeneration and renewal. In humans and mammalian model systems, the partial loss of exocrine tissue, such as after acute pancreatitis or partial pancreatectomy induces rapid recovery via expansion of surviving acinar cells. In mouse it was further found that an almost complete removal of acinar cells initiates regeneration from a currently not well-defined progenitor pool. Here, we used the zebrafish as an alternative model to study cellular mechanisms of exocrine regeneration following an almost complete removal of acinar cells. We introduced and validated two novel transgenic approaches for genetically encoded conditional cell ablation in the zebrafish, either by caspase-8-induced apoptosis or by rendering cells sensitive to diphtheria toxin. By using the ela3l promoter for exocrine-specific expression, we show that both approaches allowed cell-type-specific removal of >95% of acinar tissue in larval and adult zebrafish without causing any signs of unspecific side effects. We find that zebrafish larvae are able to recover from a virtually complete acinar tissue ablation within 2 weeks. Using short-term lineage-tracing experiments and EdU incorporation assays, we exclude duct-associated Notch-responsive cells as the source of regeneration. Rather, a rare population of slowly dividing ela3l-negative cells expressing ptf1a and CPA was identified as the origin of the newly forming exocrine cells. Cells are actively maintained, as revealed by a constant number of these cells at different larval stages and after repeated cell ablation. These cells establish ela3l expression about 4-6 days after ablation without signs of increased proliferation in between. With onset of ela3l expression, cells initiate rapid proliferation, leading to fast expansion of the ela3l-positive population. Finally, we show that this proliferation is blocked by overexpression of the Wnt-signaling antagonist dkk1b In conclusion, we show a conserved requirement for Wnt signaling in exocrine tissue expansion and reveal a potential novel progenitor or stem cell population as a source for exocrine neogenesis after complete loss of acinar cells.


Assuntos
Células Acinares/metabolismo , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Elastase Pancreática/metabolismo , Regeneração , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Células Acinares/citologia , Animais , Animais Geneticamente Modificados , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Larva/metabolismo , Especificidade de Órgãos , Pâncreas Exócrino/metabolismo , Ductos Pancreáticos/metabolismo , Células-Tronco , Via de Sinalização Wnt/genética
11.
Dev Biol ; 418(1): 108-123, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27474396

RESUMO

The stepwise progression of common endoderm progenitors into differentiated liver and pancreas organs is regulated by a dynamic array of signals that are not well understood. The nuclear receptor subfamily 5, group A, member 2 gene nr5a2, also known as Liver receptor homolog-1 (Lrh-1) is expressed in several tissues including the developing liver and pancreas. Here, we interrogate the role of Nr5a2 at multiple developmental stages using genetic and chemical approaches and uncover novel pleiotropic requirements during zebrafish liver and pancreas development. Zygotic loss of nr5a2 in a targeted genetic null mutant disrupted the development of the exocrine pancreas and liver, while leaving the endocrine pancreas intact. Loss of nr5a2 abrogated exocrine pancreas markers such as trypsin, while pancreas progenitors marked by ptf1a or pdx1 remained unaffected, suggesting a role for Nr5a2 in regulating pancreatic acinar cell differentiation. In the developing liver, Nr5a2 regulates hepatic progenitor outgrowth and differentiation, as nr5a2 mutants exhibited reduced hepatoblast markers hnf4α and prox1 as well as differentiated hepatocyte marker fabp10a. Through the first in vivo use of Nr5a2 chemical antagonist Cpd3, the iterative requirement for Nr5a2 for exocrine pancreas and liver differentiation was temporally elucidated: chemical inhibition of Nr5a2 function during hepatopancreas progenitor specification was sufficient to disrupt exocrine pancreas formation and enhance the size of the embryonic liver, suggesting that Nr5a2 regulates hepatic vs. pancreatic progenitor fate choice. Chemical inhibition of Nr5a2 at a later time during pancreas and liver differentiation was sufficient to block the formation of mature acinar cells and hepatocytes. These findings define critical iterative and pleiotropic roles for Nr5a2 at distinct stages of pancreas and liver organogenesis, and provide novel perspectives for interpreting the role of Nr5a2 in disease.


Assuntos
Células Acinares/citologia , Hepatócitos/citologia , Hepatopâncreas/embriologia , Fígado/embriologia , Pâncreas Exócrino/embriologia , Receptores Citoplasmáticos e Nucleares/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Animais , Diferenciação Celular/genética , Endoderma/citologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Técnicas de Silenciamento de Genes , Fator 4 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Morfolinos/genética , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Transativadores/genética , Fatores de Transcrição/genética , Tripsina/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/metabolismo
12.
Sci Rep ; 6: 21211, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26887806

RESUMO

Endocrine and exocrine pancreas tissues are both derived from the posterior foregut endoderm, however, the interdependence of these two cell types during their formation is not well understood. In this study, we generated mutant mice, in which the exocrine tissue is hypoplastic, in order to reveal a possible requirement for exocrine pancreas tissue in endocrine development and/or function. Since previous studies showed an indispensable role for Pdx1 in pancreas organogenesis, we used Elastase-Cre-mediated recombination to inactivate Pdx1 in the pancreatic exocrine lineage during embryonic stages. Along with exocrine defects, including impaired acinar cell maturation, the mutant mice exhibited substantial endocrine defects, including disturbed tip/trunk patterning of the developing ductal structure, a reduced number of Ngn3-expressing endocrine precursors, and ultimately fewer ß cells. Notably, postnatal expansion of the endocrine cell content was extremely poor, and the mutant mice exhibited impaired glucose homeostasis. These findings suggest the existence of an unknown but essential factor(s) in the adjacent exocrine tissue that regulates proper formation of endocrine precursors and the expansion and function of endocrine tissues during embryonic and postnatal stages.


Assuntos
Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Integrases , Pâncreas Exócrino/metabolismo , Elastase Pancreática , Transativadores/deficiência , Animais , Diabetes Mellitus/embriologia , Diabetes Mellitus/genética , Proteínas de Homeodomínio , Camundongos , Camundongos Knockout , Pâncreas Exócrino/embriologia
13.
Proc Natl Acad Sci U S A ; 112(40): 12426-31, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26392552

RESUMO

The gene encoding the E3 ubiquitin ligase Ligand of Numb protein-X (Lnx)2a is expressed in the ventral-anterior pancreatic bud of zebrafish embryos in addition to its expression in the brain. Knockdown of Lnx2a by using an exon 2/intron 2 splice morpholino resulted in specific inhibition of the differentiation of ventral bud derived exocrine cell types, with little effect on endocrine cell types. A frame shifting null mutation in lnx2a did not mimic this phenotype, but a mutation that removed the exon 2 splice donor site did. We found that Lnx2b functions in a redundant manner with its paralog Lnx2a. Inhibition of lnx2a exon 2/3 splicing causes exon 2 skipping and leads to the production of an N-truncated protein that acts as an interfering molecule. Thus, the phenotype characterized by inhibition of exocrine cell differentiation requires inactivation of both Lnx2a and Lnx2b. Human LNX1 is known to destabilize Numb, and we show that inhibition of Numb expression rescues the Lnx2a/b-deficient phenotype. Further, Lnx2a/b inhibition leads to a reduction in the number of Notch active cells in the pancreas. We suggest that Lnx2a/b function to fine tune the regulation of Notch through Numb in the differentiation of cell types in the early zebrafish pancreas. Further, the complex relationships among genotype, phenotype, and morpholino effect in this case may be instructive in the ongoing consideration of morpholino use.


Assuntos
Proteínas de Transporte/genética , Diferenciação Celular/genética , Pâncreas Exócrino/metabolismo , Pâncreas/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteínas de Peixe-Zebra/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Immunoblotting , Hibridização In Situ , Microscopia Confocal , Morfolinos/genética , Mutação , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
14.
Dev Dyn ; 244(6): 724-35, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25773748

RESUMO

BACKGROUND: Pancreas development in zebrafish shares many features with mammals, including the participation of epithelial progenitor cells expressing pancreas transcription factor 1a (ptf1a). However, to date it has remained unclear whether, as in mammals, ptf1a-expressing zebrafish pancreatic progenitors are able to contribute to multiple exocrine and endocrine lineages. To delineate the lineage potential of ptf1a-expressing cells, we generated ptf1a:creER(T2) transgenic fish and performed genetic-inducible lineage tracing in developmental, regenerating, and ptf1a-deficient zebrafish pancreas. RESULTS: In addition to their contribution to the acinar cell lineage, ptf1a-expressing cells give rise to both pancreatic Notch-responsive-cells (PNCs) as well as small numbers of endocrine cells during pancreatic development. In fish with ptf1a haploinsufficiency, a higher proportion of ptf1a lineage-labeled cells are traced into the PNC and endocrine compartments. Further reduction of ptf1a gene dosage converts pancreatic progenitor cells to gall bladder and other non-pancreatic cell fates. CONCLUSIONS: Our results confirm the presence of multipotent ptf1a-expressing progenitor cells in developing zebrafish pancreas, with reduced ptf1a dosage promoting greater contributions towards non-acinar lineages. As in mammals, loss of ptf1a results in conversion of nascent pancreatic progenitor cells to non-pancreatic cell fates, underscoring the central role of ptf1a in foregut tissue specification.


Assuntos
Pâncreas/embriologia , Fatores de Transcrição/fisiologia , Peixe-Zebra/embriologia , Células Acinares/citologia , Animais , Animais Geneticamente Modificados , Linhagem da Célula , Cromossomos Artificiais Bacterianos , Vesícula Biliar/citologia , Dosagem de Genes , Genótipo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Especificidade de Órgãos , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/fisiologia , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/crescimento & desenvolvimento , Receptores Notch/fisiologia , Recombinação Genética , Regeneração , Células-Tronco/citologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
15.
PLoS One ; 9(7): e101980, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000307

RESUMO

The liver plays a vital role in metabolism, detoxification, digestion, and the maintenance of homeostasis. During development, the vertebrate embryonic liver undergoes a series of morphogenic processes known as hepatogenesis. Hepatogenesis can be separated into three interrelated processes: endoderm specification, hepatoblast differentiation, and hepatic outgrowth. Throughout this process, signaling molecules and transcription factors initiate and regulate the coordination of cell proliferation, apoptosis, differentiation, intercellular adhesion, and cell migration. Hifs are already recognized to be essential in embryonic development, but their role in hepatogenesis remains unknown. Using the zebrafish embryo as a model organism, we report that the lack of Hif2-alpha but not Hif1-alpha blocks hepatic outgrowth. While Hif2-alpha is not involved in hepatoblast specification, this transcription factor regulates hepatocyte cell proliferation during hepatic outgrowth. Furthermore, we demonstrated that the lack of Hif2-alpha can reduce the expression of liver-enriched gene 1 (leg1), which encodes a secretory protein essential for hepatic outgrowth. Additionally, exogenous mRNA expression of leg1 can rescue the small liver phenotype of hif2-alpha morphants. We also showed that Hif2-alpha directly binds to the promoter region of leg1 to control leg1 expression. Interestingly, we discovered overrepresented, high-density Hif-binding sites in the potential upstream regulatory sequences of leg1 in teleosts but not in terrestrial mammals. We concluded that hif2-alpha is a key factor required for hepatic outgrowth and regulates leg1 expression in zebrafish embryos. We also proposed that the hif2-alpha-leg1 axis in liver development may have resulted from the adaptation of teleosts to their environment.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fígado/embriologia , Transcrição Gênica , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células/efeitos dos fármacos , Cobalto/farmacologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Fator de Crescimento de Hepatócito/metabolismo , Intestinos/embriologia , Fígado/citologia , Tamanho do Órgão/efeitos dos fármacos , Pâncreas Exócrino/embriologia , Fenótipo , Regiões Promotoras Genéticas/genética , Elementos de Resposta/genética , Via de Sinalização Wnt/efeitos dos fármacos
16.
Dev Dyn ; 243(3): 415-27, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24166718

RESUMO

BACKGROUND: We previously identified a local renin-angiotensin system (RAS) regulating the differentiation of an isolated population of human pancreatic progenitor cells. Major RAS components that regulate organogenesis have been also described in embryos; however, it is not known whether a local RAS is present in the fetal pancreas. We now hypothesize that angiotensin II type 1 (AT1 ) and type 2 (AT2 ) receptors are expressed in mouse embryonic pancreas and involved in regulating endocrine cell development. RESULTS: Differential expression of AT1 and AT2 receptors was observed in the mouse pancreata in late embryogenesis. Systemic AT2 , but not AT1 , receptor blockade during the second transition in pancreatic development (from embryonic day 12.0 onward) reduced the ß-cell to α-cell ratio of the neonate islets, impaired their insulin secretory function and the glucose tolerance of the pups. Studies with pancreas explants ex vivo revealed regulation by AT2 receptors of the differentiation of pancreatic progenitors into insulin-producing cells and of the proliferation of the differentiated cell, actions that did not result from reduced angiogenesis as a secondary effect of AT2 receptor antagonism. CONCLUSIONS: These data revealed an AT2 receptor-mediated mechanism regulating pancreatic endocrine cell development in vivo.


Assuntos
Diferenciação Celular/fisiologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Pâncreas Exócrino , Receptor Tipo 2 de Angiotensina/biossíntese , Células-Tronco , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Teste de Tolerância a Glucose , Humanos , Camundongos , Camundongos Endogâmicos ICR , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Receptor Tipo 1 de Angiotensina/biossíntese , Células-Tronco/citologia , Células-Tronco/metabolismo
17.
Zebrafish ; 10(2): 161-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23697888

RESUMO

Ribosomal protein L (rpl) genes are essential for assembly of the 60S subunit of the eukaryotic ribosome and may also carry out additional extra-ribosomal functions. We have identified a common expression pattern for rpl genes in developing zebrafish larvae. After initially widespread expression in early embryos, the expression of multiple rpl genes becomes increasingly restricted to the endoderm. With respect to the pancreas, rpl genes are highly expressed in ptf1a-expressing pancreatic progenitors at 48 hpf, suggesting possible functional roles in pancreatic morphogenesis and/or differentiation. Utilizing two available mutant lines, rpl23a(hi2582) and rpl6(hi3655b), we found that ptf1a-expressing pancreatic progenitors fail to properly expand in embryos homozygous for either of these genes. In addition to these durable homozygous phenotypes, we also demonstrated recoverable delays in ptf1a-expressing pancreatic progenitor expansion in rpl23a(hi2582) and rpl6(hi3655b) heterozygotes. Disruptions in ribosome assembly are generally understood to initiate a p53-dependent cellular stress response. However, concomitant p53 knockdown was unable to rescue normal pancreatic progenitor expansion in either rpl23a(hi2582) or rpl6(hi3655b) mutant embryos, suggesting required and p53-independent roles for rpl23a and rpl6 in pancreas development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Pâncreas Exócrino/embriologia , Proteínas Ribossômicas/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Hibridização In Situ , Larva/genética , Larva/metabolismo , Microscopia Confocal , Especificidade de Órgãos , Organogênese , Pâncreas Exócrino/crescimento & desenvolvimento , Pâncreas Exócrino/metabolismo , Reação em Cadeia da Polimerase , Proteínas Ribossômicas/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
18.
Zebrafish ; 10(2): 132-46, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23682805

RESUMO

Abstract Experimental studies in the zebrafish have greatly facilitated understanding of genetic regulation of the early developmental events in the pancreas. Various approaches using forward and reverse genetics, chemical genetics, and transgenesis in zebrafish have demonstrated generally conserved regulatory roles of mammalian genes and discovered novel genetic pathways in exocrine pancreatic development. Accumulating evidence has supported the use of zebrafish as a model of human malignant diseases, including pancreatic cancer. Studies have shown that the genetic regulators of exocrine pancreatic development in zebrafish can be translated into potential clinical biomarkers and therapeutic targets in human pancreatic adenocarcinoma. Transgenic zebrafish expressing oncogenic K-ras and zebrafish tumor xenograft model have emerged as valuable tools for dissecting the pathogenetic mechanisms of pancreatic cancer and for drug discovery and toxicology. Future analysis of the pancreas in zebrafish will continue to advance understanding of the genetic regulation and biological mechanisms during organogenesis. Results of those studies are expected to provide new insights into how aberrant developmental pathways contribute to formation and growth of pancreatic neoplasia, and hopefully generate valid biomarkers and targets as well as effective and safe therapeutics in pancreatic cancer.


Assuntos
Adenocarcinoma/etiologia , Adenocarcinoma/terapia , Modelos Animais de Doenças , Pâncreas/embriologia , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/terapia , Peixe-Zebra/embriologia , Adenocarcinoma/embriologia , Adenocarcinoma/genética , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/crescimento & desenvolvimento , Biomarcadores/metabolismo , Descoberta de Drogas , Genes ras , Humanos , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/crescimento & desenvolvimento , Pâncreas Exócrino/metabolismo , Neoplasias Pancreáticas/embriologia , Neoplasias Pancreáticas/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
19.
Semin Cell Dev Biol ; 23(6): 711-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22743232

RESUMO

This review summarizes our current understanding of exocrine pancreas development, including the formation of acinar, ductal and centroacinar cells. We discuss the transcription factors associated with various stages of exocrine differentiation, from multipotent progenitor cells to fully differentiated acinar and ductal cells. Within the branching epithelial tree of the embryonic pancreas, this involves the progressive restriction of multipotent pancreatic progenitor cells to either a central "trunk" domain giving rise to the islet and ductal lineages, or a peripheral "tip" domain giving rise to acinar cells. This review also discusses the soluble morphogens and other signaling pathways that influence these events. Finally, we examine centroacinar cells as an enigmatic pancreatic cell type whose lineage remains uncertain, and whose possible progenitor capacities continue to be explored.


Assuntos
Células Acinares/citologia , Morfogênese , Pâncreas Exócrino/embriologia , Ductos Pancreáticos/embriologia , Células Acinares/metabolismo , Animais , Humanos , Organogênese , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas Exócrino/citologia , Pâncreas Exócrino/metabolismo , Ductos Pancreáticos/citologia , Ductos Pancreáticos/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo
20.
Gastroenterology ; 142(4): 999-1009.e6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22178591

RESUMO

BACKGROUND & AIMS: The exocrine portion of the pancreas functions in digestion and preserves pancreatic homeostasis. Learning how this tissue forms during embryogenesis could improve our understanding of human pancreatic diseases. Expression of the homeobox gene Prox1 in the exocrine pancreas changes throughout development in mice. We investigated the role of Prox1 in development of the exocrine pancreas in mice. METHODS: Mice with pancreas-specific deletion of Prox1 (Prox1(ΔPanc)) were generated and their pancreatic tissues were analyzed using immunohistochemistry, transmission electron microscopy, histologic techniques, quantitative real-time polymerase chain reaction, immunoblotting, and morphometric analysis. RESULTS: Loss of Prox1 from the pancreas led to multiple exocrine alterations, most notably premature acinar cell differentiation, increased ductal cell proliferation, altered duct morphogenesis, and imbalanced expression of claudin proteins. Prox1(ΔPanc) mice also had some minor alterations in islet cells, but beta-cell development was not affected. The exocrine congenital defects of Prox1(ΔPanc) pancreata appeared to initiate a gradual process of deterioration that resulted in extensive loss of acinar cells, lipomatosis, and damage to ductal tissue in adult mice. CONCLUSIONS: Pancreas-specific deletion of Prox1 causes premature differentiation of acinar cells and poor elongation of epithelial branches; these defects indicate that Prox1 controls the expansion of tip progenitors in the early developing pancreas. During later stages of embryogenesis, Prox1 appears to regulate duct cell proliferation and morphogenesis. These findings identify Prox1 as an important regulator of pancreatic exocrine development.


Assuntos
Células-Tronco Embrionárias/metabolismo , Pâncreas Exócrino/metabolismo , Proteínas Supressoras de Tumor/deficiência , Fatores Etários , Envelhecimento , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Claudinas/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Proteínas de Homeodomínio/genética , Homeostase , Imuno-Histoquímica , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Morfogênese , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/ultraestrutura , Ductos Pancreáticos/embriologia , Ductos Pancreáticos/metabolismo , Fenótipo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...