Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 253
Filtrar
1.
J Cell Biol ; 220(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34047771

RESUMO

Mesenchymal-to-epithelial transition (MET) converts cells from migratory mesenchymal to polarized epithelial states. Despite its importance for both normal and pathological processes, very little is known about the regulation of MET in vivo. Here we exploit midgut morphogenesis in Drosophila melanogaster to investigate the mechanisms underlying MET. We show that down-regulation of the EMT transcription factor Serpent is required for MET, but not sufficient, as interactions with the surrounding mesoderm are also essential. We find that midgut MET relies on the secretion of specific laminins via the CopII secretory pathway from both mesoderm and midgut cells. We show that secretion of the laminin trimer containing the Wingblister α-subunit from the mesoderm is an upstream cue for midgut MET, leading to basal polarization of αPS1 integrin in midgut cells. Polarized αPS1 is required for the formation of a monolayered columnar epithelium and for the apical polarization of αPS3, Baz, and E-Cad. Secretion of a distinct LamininA-containing trimer from midgut cells is required to reinforce the localization of αPS1 basally, and αPS3 apically, for robust repolarization. Our data suggest that targeting these MET pathways, in conjunction with therapies preventing EMT, may present a two-pronged strategy toward blocking metastasis in cancer.


Assuntos
Sistema Digestório/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Laminina/metabolismo , Animais , Animais Geneticamente Modificados , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Polaridade Celular , Sistema Digestório/embriologia , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Laminina/genética , Microscopia Confocal , Microscopia de Fluorescência , Transdução de Sinais , Fatores de Tempo , Imagem com Lapso de Tempo
2.
Dev Biol ; 474: 37-47, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33571486

RESUMO

Canonical Wnt signaling plays a key role during organ development, homeostasis and regeneration and these processes are conserved between invertebrates and vertebrates. Mutations in Wnt pathway components are commonly found in various types of cancer. Upon activation of canonical Wnt signaling, ß-catenin binds in the nucleus to members of the TCF-LEF family and activates the transcription of target genes. Multiple Wnt target genes, including Lgr5/LGR5 and Axin2/AXIN2, have been identified in mouse models and human cancer cell lines. Here we set out to identify the transcriptional targets of Wnt signaling in five human tissues using organoid technology. Organoids are derived from adult stem cells and recapitulate the functionality as well as the structure of the original tissue. Since the Wnt pathway is critical to maintain the organoids from the human intestine, colon, liver, pancreas and stomach, organoid technology allows us to assess Wnt target gene expression in a human wildtype situation. We performed bulk mRNA sequencing of organoids immediately after inhibition of Wnt pathway and identified 41 genes as commonly regulated genes in these tissues. We also identified large numbers of target genes specific to each tissue. One of the shared target genes is TEAD4, a transcription factor driving expression of YAP/TAZ signaling target genes. In addition to TEAD4, we identified a variety of genes which encode for proteins that are involved in Wnt-independent pathways, implicating the possibility of direct crosstalk between Wnt signaling and other pathways. Collectively, this study identified tissue-specific and common Wnt target gene signatures and provides evidence for a conserved role for these Wnt targets in different tissues.


Assuntos
Sistema Digestório/citologia , Regulação da Expressão Gênica no Desenvolvimento , Organoides/metabolismo , Via de Sinalização Wnt , Adulto , Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Endoderma , Perfilação da Expressão Gênica , Humanos , Especificidade de Órgãos
3.
Dev Biol ; 427(1): 72-83, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28501478

RESUMO

During early fetal development, paracrine Hedgehog (HH) ligands secreted from the foregut epithelium activate Gli transcription factors in the surrounding mesenchyme to coordinate formation of the respiratory system, digestive track and the cardiovascular network. Although disruptions to this process can lead to devastating congenital defects, the underlying mechanisms and downstream targets, are poorly understood. We show that the zinc finger transcription factor Osr1 is a novel HH target as Osr1 expression in the foregut mesenchyme depends on HH signaling and the effector of HH pathway Gli3 binds to a conserved genomic loci near Osr1 promoter region. Molecular analysis of mouse germline Osr1 mutants reveals multiple functions of Osr1 during foregut development. Osr1 mutants exhibit fewer lung progenitors in the ventral foregut. Osr is then required for the proper branching of the primary lung buds, with mutants exhibiting miss-located lung lobes. Finally, Osr1 is essential for proper mesenchymal differentiation including pulmonary arteries, esophageal and tracheal smooth muscle as well as tracheal cartilage rings. Tissue specific conditional knockouts in combination with lineage tracing indicate that Osr1 is required cell autonomously in the foregut mesenchyme. We conclude that Osr1 is a novel downstream target of HH pathway, required for lung specification, branching morphogenesis and foregut mesenchymal differentiation.


Assuntos
Sistema Digestório/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Organogênese/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Animais , Sistema Digestório/embriologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Proteínas Hedgehog/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição/metabolismo , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco
4.
Cell Signal ; 29: 12-22, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27693749

RESUMO

Wnt signaling plays essential roles in both embryonic pattern formation and postembryonic tissue homoestasis. High levels of Wnt activity repress foregut identity and facilitate hindgut fate through forming a gradient of Wnt signaling activity along the anterior-posterior axis. Here, we examined the mechanisms of Wnt signaling in hindgut development by differentiating human embryonic stem cells (hESCs) into the hindgut progenitors. We observed severe morphological changes when Wnt signaling was blocked by using Wnt antagonist Dkk1. We performed deep-transcriptome sequencing (RNA-seq) and identified 240 Wnt-activated genes and 2023 Wnt-repressed genes, respectively. Clusters of Wnt targets showed enrichment in specific biological functions, such as "gastrointestinal or skeletal development" in the Wnt-activated targets and "neural or immune system development" in the Wnt-repressed targets. Moreover, we adopted a high-throughput chromatin immunoprecipitation and deep sequencing (ChIP-seq) approach to identify the genomic regions through which Wnt-activated transcription factor TCF7L2 regulated transcription. We identified 83 Wnt direct target candidates, including the hindgut marker CDX2 and the genes relevant to morphogenesis (MSX1, MSX2, LEF1, T, PDGFRB etc.) through combinatorial analysis of the RNA-seq and ChIP-seq data. Together, our study identified a series of direct and indirect Wnt targets in hindgut differentiation, and uncovered the diverse mechanisms of Wnt signaling in regulating multi-lineage differentiation.


Assuntos
Diferenciação Celular , Linhagem da Célula , Sistema Digestório/diagnóstico por imagem , Sistema Digestório/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Humanas/citologia , Via de Sinalização Wnt , Receptores de Apelina/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Sistema Digestório/citologia , Endoderma/metabolismo , Ontologia Genética , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Mesoderma/metabolismo , Morfogênese/genética , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Ligação Proteica/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores CXCR4/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Transcrição Gênica , Via de Sinalização Wnt/genética
5.
Development ; 142(24): 4288-98, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26493402

RESUMO

In developing organisms, divergence from the canonical cell division cycle is often necessary to ensure the proper growth, differentiation, and physiological function of a variety of tissues. An important example is endoreplication, in which endocycling cells alternate between G and S phase without intervening mitosis or cytokinesis, resulting in polyploidy. Although significantly different from the canonical cell cycle, endocycles use regulatory pathways that also function in diploid cells, particularly those involved in S phase entry and progression. A key S phase regulator is the Cyclin E-Cdk2 kinase, which must alternate between periods of high (S phase) and low (G phase) activity in order for endocycling cells to achieve repeated rounds of S phase and polyploidy. The mechanisms that drive these oscillations of Cyclin E-Cdk2 activity are not fully understood. Here, we show that the Drosophila Cyclin E-Cdk2 inhibitor Dacapo (Dap) is targeted for destruction during S phase via a PIP degron, contributing to oscillations of Dap protein accumulation during both mitotic cycles and endocycles. Expression of a PIP degron mutant Dap attenuates endocycle progression but does not obviously affect proliferating diploid cells. A mathematical model of the endocycle predicts that the rate of destruction of Dap during S phase modulates the endocycle by regulating the length of G phase. We propose from this model and our in vivo data that endo S phase-coupled destruction of Dap reduces the threshold of Cyclin E-Cdk2 activity necessary to trigger the subsequent G-S transition, thereby influencing endocycle oscillation frequency and the extent of polyploidy.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Endorreduplicação , Proteínas Nucleares/metabolismo , Fase S , Sequência de Aminoácidos , Animais , Sistema Digestório/citologia , Sistema Digestório/embriologia , Proteínas de Drosophila/química , Drosophila melanogaster/embriologia , Células Epidérmicas , Feminino , Mitose , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Nucleares/química , Folículo Ovariano/citologia , Proteólise , Glândulas Salivares/citologia
6.
J Neurosci ; 33(11): 4901-12, 2013 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-23486961

RESUMO

Enteric neural crest-derived cells (ENCCs) migrate from the anterior foregut in a rostrocaudal direction to colonize the entire gastrointestinal tract and to form the enteric nervous system. Genetic approaches have identified many signaling molecules regulating the migration of ENCCs; however, it remains elusive how the activities of the signaling molecules are regulated spatiotemporally during migration. In this study, transgenic mice expressing biosensors based on Förster resonance energy transfer were generated to video the activity changes of the signaling molecules in migrating ENCCs. In an organ culture of embryonic day 11.25 (E11.25) to E13 guts, ENCCs at the rostral wavefront migrated as a cellular chain faster than the following ENCCs that formed a network. The faster-migrating cells at the wavefront exhibited lower protein kinase A (PKA) activity than did the slower-migrating trailing cells. The activities of Rac1 and Cdc42 exhibited an inverse correlation with the PKA activity, and PKA activation decreased the Rac1 activity and migration velocity. PKA activity in ENCCs was correlated positively with the distribution of GDNF and inversely with the distribution of endothelin 3 (ET-3). Accordingly, PKA was activated by GDNF and inhibited by ET-3 in cultured ENCCs. Finally, although the JNK and ERK pathways were previously reported to control the migration of ENCCs, we did not find any correlation of JNK or ERK activity with the migration velocities. These results suggest that external cues regulate the migration of ENCCs by controlling PKA activity, but not ERK or JNK activity, and argue for the importance of live imaging of signaling molecule activities in developing organs.


Assuntos
Movimento Celular/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endotelina-3/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Crista Neural/citologia , Neurônios/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Fatores Etários , Animais , Técnicas Biossensoriais , Proteína de Ligação a CREB/metabolismo , Movimento Celular/efeitos dos fármacos , Sistema Digestório/citologia , Sistema Digestório/embriologia , Embrião de Mamíferos , Endotelina-3/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Transferência Ressonante de Energia de Fluorescência , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Tionucleotídeos/farmacologia , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína Vermelha Fluorescente
7.
Res Vet Sci ; 94(1): 1-4, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22895155

RESUMO

RET receptor is a transmembrane protein which, together with the glial-cell-line derived neurotrophic factor family receptors alpha, forms a receptor complex upon activation by the glial-cell-line-derived neurotrophic ligands (GFLs). RET signaling is crucial for: (a) development of the enteric nervous system and kidney; (b) development of sympathetic, parasympathetic, motor, and sensory neurons; (c) postnatal maintenance of dopaminergic neurons; (d) spermatogenesis. In humans, RET mutations cause the Hirschsprung's disease, characterized by megacolon aganglionosis, and different types of cancer, the multiple endocrine neoplasia type 2A and type 2B and familial medullary thyroid. In the earliest aged cat embryos studied (stage 9 according to Knopse), RET immunoreactivity (IR) was observed in few cells detected in bilateral rows extending latero-ventrally to the neural tube and dorso-laterally to the foregut. In the successive aged group (stage 11), RET IR was observed in few single or grouped epithelial cells of the anterior gut and in small clustered cells scattered in the mesenchyme around the anterior gut. From stage 14-22 (the last stage 22 includes foetuses around the birth), RET IR was seen in neurons and fibers of the enteric nervous system. The appearance and intensification of RET-IR in the gut occurred with cranio/caudal and external/internal directions during the development. These results, thus, suggest the involvement of GFLs in the neuroblast migration, proliferation and differentiation. For a short period of development, these molecules might also act on some cells of the epithelium.


Assuntos
Gatos/embriologia , Sistema Digestório/embriologia , Receptores Proteína Tirosina Quinases/fisiologia , Animais , Sistema Nervoso Entérico/embriologia , Epitélio/fisiologia , Tubo Neural/fisiologia
8.
Int. j. morphol ; 30(4): 1285-1294, dic. 2012. ilus
Artigo em Espanhol | LILACS | ID: lil-670139

RESUMO

El aparato digestivo deriva del endodermo y el mesodermo, que forman su epitelio y la musculatura lisa respectivamente. Al igual que en el resto de los sistemas, existe un interacción epitelio-mesenquimática mediada por moléculas como Hedgehog, BMP y FoxF1 que determinan el crecimiento intestinal en sus ejes principales. Los genes Hox, junto con el resto de las moléculas, participan en la regionalización del sistema digestivo. En sus inicios lo denominaremos intestino primitivo, formado por un tubo endodérmico que deriva del saco vitelino; dividiéndose en intestino anterior, medio y posterior. En esta revisión veremos cómo estos 3 segmentos darán origen a las diferentes estructuras del sistema digestivo en los vertebrados.


The digestive system is derived from the endoderm and mesoderm, which form its epithelium and smooth muscle, respectively. As in the other systems, there is an epithelial-mesenchymal interactions mediated by molecules such as Hedgehog, BMP and FoxF1, determining intestinal growth in the main axes. The Hox genes, together the rest of the molecules, involved in the regionalization of the digestive system. In the beginning we call it primitive gut, consisting of a tube derived of endodermal yolk sac, divided into foregut, midgut and hindgut. In this review we will see how these 3 segments give rise to different structures of the digestive system in vertebrates.


Assuntos
Humanos , Animais , Sistema Digestório/embriologia , Vertebrados , Genes Homeobox , Proteínas Morfogenéticas Ósseas , Sistema Digestório/crescimento & desenvolvimento , Endoderma/embriologia , Proteínas Hedgehog , Mesoderma/embriologia
10.
Development ; 138(20): 4511-22, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21903671

RESUMO

Rhou encodes a Cdc42-related atypical Rho GTPase that influences actin organization in cultured cells. In mouse embryos at early-somite to early-organogenesis stages, Rhou is expressed in the columnar endoderm epithelium lining the lateral and ventral wall of the anterior intestinal portal. During foregut development, Rhou is downregulated in regions where the epithelium acquires a multilayered morphology heralding the budding of organ primordia. In embryos generated from Rhou knockdown embryonic stem (ES) cells, the embryonic foregut displays an abnormally flattened shape. The epithelial architecture of the endoderm is disrupted, the cells are depleted of microvilli and the phalloidin-stained F-actin content of their sub-apical cortical domain is reduced. Rhou-deficient cells in ES cell-derived embryos and embryoid bodies are less efficient in endoderm differentiation. Impaired endoderm differentiation of Rhou-deficient ES cells is accompanied by reduced expression of c-Jun/AP-1 target genes, consistent with a role for Rhou in regulating JNK activity. Downregulation of Rhou in individual endoderm cells results in a reduced ability of these cells to occupy the apical territory of the epithelium. Our findings highlight epithelial morphogenesis as a required intermediate step in the differentiation of endoderm progenitors. In vivo, Rhou activity maintains the epithelial architecture of the endoderm progenitors, and its downregulation accompanies the transition of the columnar epithelium in the embryonic foregut to a multilayered cell sheet during organ formation.


Assuntos
Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Endoderma/embriologia , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Camundongos , Camundongos Knockout , Células NIH 3T3 , RNA Interferente Pequeno/genética , Transdução de Sinais , Proteínas Wnt/metabolismo , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/genética
11.
Development ; 138(16): 3557-68, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21771809

RESUMO

The vertebrate liver, pancreas and lung arise in close proximity from the multipotent foregut endoderm. Tissue-explant experiments uncovered instructive signals emanating from the neighbouring lateral plate mesoderm, directing the endoderm towards specific organ fates. This suggested that an intricate network of signals is required to control the specification and differentiation of each organ. Here, we show that sequential functions of Wnt2bb and Wnt2 control liver specification and proliferation in zebrafish. Their combined specific activities are essential for liver specification, as their loss of function causes liver agenesis. Conversely, excess wnt2bb or wnt2 induces ectopic liver tissue at the expense of pancreatic and anterior intestinal tissues, revealing the competence of intestinal endoderm to respond to hepatogenic signals. Epistasis experiments revealed that the receptor frizzled homolog 5 (fzd5) mediates part of the broader hepatic competence of the alimentary canal. fzd5 is required for early liver formation and interacts genetically with wnt2 as well as wnt2bb. In addition, lack of both ligands causes agenesis of the swim bladder, the structural homolog of the mammalian lung. Thus, tightly regulated spatiotemporal expression of wnt2bb, wnt2 and fzd5 is central to coordinating early liver, pancreas and swim bladder development from a multipotent foregut endoderm.


Assuntos
Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt2/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Proliferação de Células , Sistema Digestório/citologia , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Wnt/genética , Proteína Wnt2/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
12.
Med Sci (Paris) ; 26(12): 1061-6, 2010 Dec.
Artigo em Francês | MEDLINE | ID: mdl-21187045

RESUMO

Hepatocyte transplantation is considered as an alternative to organ transplantation in particular for the treatment of liver metabolic diseases. However, due to the difficulties to obtain a large number of hepatocytes, new sources of cells are needed. These cells could be either of hepatic origin (hepatic stem cells) or extrahepatic such as mesenchymal stem cells or pluripotent stem cells (human embryonic stem cells [hESC] or iPS). We developed a new method to differentiate hESCs into fetal hepatocytes. These conditions recapitulate the main liver developmental stages, using fully defined medium devoid of animal products or unknown factors. The differentiated cells express many fetal hepatocytes markers (cytochrome P450 3A7, albumin, alpha-1-antitrypsin, etc.). The cells display specific hepatic functions (ammonia metabolism, excretion of indocyanin green) and are capable to engraft and express hepatic proteins two months after transplantation into newborn uPAxrag2gc-/- mouse liver. We have also showed that this approach is transposable to human iPS, and further studies on animal models will allow us to compare the in vivo potential of these two sources of pluripotent cells. Finally, only studies on large animals such as nonhuman primates will validate an eventual clinical application.


Assuntos
Células-Tronco Embrionárias/citologia , Hepatócitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Animais Recém-Nascidos , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Transdiferenciação Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/transplante , Meios de Cultura/farmacologia , Sistema Digestório/embriologia , Células-Tronco Embrionárias/efeitos dos fármacos , Endoderma/citologia , Regulação da Expressão Gênica no Desenvolvimento , Hepatócitos/metabolismo , Hepatócitos/transplante , Humanos , Fígado/citologia , Fígado/embriologia , Camundongos , Células-Tronco Pluripotentes/transplante , Transplante Heterólogo
13.
Artigo em Inglês | MEDLINE | ID: mdl-19788926

RESUMO

Changes in water temperature and salinity are responsible for a variety of physiological stress responses in aquatic organisms. Stress induced by these factors was recently associated with enhanced reactive oxygen species (ROS) generation, which caused oxidative damage. In the present study, we investigated the time-related effects of changes in water temperature and salinity on mRNA expression and the activities of antioxidant enzymes (SOD and CAT) and lipid peroxidation (LPO) in the gills and digestive glands of the ark shell, Scapharca broughtonii. To investigate physiological responses, hydrogen peroxide (H(2)O(2)), lysozyme activity, aspartate aminotransferase (AspAT), and alanine aminotransferase (AlaAT) were measured in the hemolymph. Water temperature and salinity changes significantly increased antioxidant enzyme mRNA expression and activity in the digestive glands and gills in a time-dependent manner. H(2)O(2) concentrations increased significantly in the high-temperature and hyposalinity treatments. LPO, AspAT and AlaAT levels also increased significantly in a time-dependent manner, while lysozyme activity decreased. These results suggest that antioxidant enzymes play important roles in reducing oxidative stress in ark shells exposed to changes in water temperature and salinity.


Assuntos
Catalase/metabolismo , Scapharca/enzimologia , Estresse Fisiológico , Superóxido Dismutase/metabolismo , Alanina Transaminase/metabolismo , Animais , Aspartato Aminotransferases/metabolismo , Catalase/genética , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Sistema Digestório/embriologia , Sistema Digestório/enzimologia , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Brânquias/enzimologia , Brânquias/metabolismo , Hemolinfa/enzimologia , Hemolinfa/metabolismo , Peróxido de Hidrogênio/metabolismo , Peroxidação de Lipídeos , Dados de Sequência Molecular , Muramidase/metabolismo , Concentração Osmolar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Scapharca/genética , Scapharca/metabolismo , Análise de Sequência de DNA , Superóxido Dismutase/genética , Temperatura
14.
Dev Cell ; 17(2): 290-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19686689

RESUMO

Patterning of the primitive foregut promotes appropriate organ specification along its anterior-posterior axis. However, the molecular pathways specifying foregut endoderm progenitors are poorly understood. We show here that Wnt2/2b signaling is required to specify lung endoderm progenitors within the anterior foregut. Embryos lacking Wnt2/2b expression exhibit complete lung agenesis and do not express Nkx2.1, the earliest marker of the lung endoderm. In contrast, other foregut endoderm-derived organs, including the thyroid, liver, and pancreas, are correctly specified. The phenotype observed is recapitulated by an endoderm-restricted deletion of beta-catenin, demonstrating that Wnt2/2b signaling through the canonical Wnt pathway is required to specify lung endoderm progenitors within the foregut. Moreover, activation of canonical Wnt/beta-catenin signaling results in the reprogramming of esophagus and stomach endoderm to a lung endoderm progenitor fate. Together, these data reveal that canonical Wnt2/2b signaling is required for the specification of lung endoderm progenitors in the developing foregut.


Assuntos
Sistema Digestório , Pulmão , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/metabolismo , Proteína Wnt2/metabolismo , beta Catenina/metabolismo , Animais , Padronização Corporal/fisiologia , Sistema Digestório/anatomia & histologia , Sistema Digestório/embriologia , Endoderma/citologia , Endoderma/fisiologia , Pulmão/anatomia & histologia , Pulmão/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco/citologia , Proteínas Wnt/genética , Proteína Wnt2/genética , beta Catenina/genética
15.
Biocell ; 33(1): 49-65, Apr. 2009. ilus
Artigo em Inglês | LILACS | ID: lil-595029

RESUMO

Development of Pomacea canaliculata from the gastrula stage until the first day after hatching is described. Trochophore embryos are developed after gastrulation, showing the prototroch as a crown of ciliated orange-brownish cells. However, no true veliger embryos are formed, since the prototroch does not fully develop into a velum. Afterward, the connection between the fore- and midgut is permeated and the midgut becomes full of the pink-reddish albumen, which is stored into a central archenteron's lake, from where it is accumulated into the large cells forming the midgut wall ("giant cells"). Electron microscopy of giant cells in late embryos showed that albumen is engulfed by large endocytic vesicles formed between the irregular microvilli at the top of these cells. By the end of intracapsular development, giant cells become gradually replaced by two new epithelial cell types which are similar to those found in the adult midgut gland: the pre-columnar and the pre-pyramidal cells. Pre-columnar cells have inconspicuous basal nuclei and are crowned by stereocilia, between which small endocytic vesicles are formed. Pre-pyramidal cells have large nuclei with 2-3 nucleoli and show a striking development of the rough endoplasmic reticulum. The genesis of the three cell lineages (giant, pre-columnar and pre-pyramidal cells) is hypothetically attributed to epithelial streaks that occur at both sides of the midgut since early stages of development.


Assuntos
Animais , Caramujos/citologia , Caramujos/embriologia , Caramujos/ultraestrutura , Gástrula/citologia , Organogênese/fisiologia , Sistema Digestório/embriologia
16.
Development ; 136(8): 1295-303, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19279135

RESUMO

Steel factor is an essential survival and proliferation factor for primordial germ cells (PGCs) during their migration in the early mouse embryo. PGCs arise during gastrulation, and migrate into the posterior endoderm that becomes the hindgut. Previous reports have suggested that PGCs become dependent on Steel factor when they colonize the hindgut. However, in the absence of a good marker for living PGCs, their behavior before hindgut colonization has not been previously studied. We report here the normal behavior of PGCs in live embryos before hindgut colonization, and the roles of Steel factor, using a reporter line in which GFP is driven by the promoter of the Stella gene, whose activation accompanies the initial specification of PGCs. We show first that PGCs are surrounded by Steel factor-expressing cells from their first appearance in the allantois to the time they enter the genital ridges. Second, fewer PGCs are found in the allantois in Steel-null embryos, but this is not due to a failure of PGC specification. Third, the analysis of cultured Steel-null early embryos shows that Steel factor is required for normal PGC motility, both in the allantois and in the hindgut. Germ cells migrate actively in the allantois, and move directionally from the allantois into the proximal epiblast. In the absence of Steel factor, caused by either null mutation or antibody blockade, PGC motility is dramatically decreased, but directionality is maintained, demonstrating a primary role for Steel factor in PGC motility. This was found both before and after colonization of the hindgut. These data, together with previously published data, show that PGCs are Steel factor dependent from their initial specification until they colonize the genital ridges, and suggest the existence of a ;spatio-temporal niche' that travels with this important pluripotential cell population in the embryo.


Assuntos
Alantoide/citologia , Alantoide/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco/citologia , Nicho de Células-Tronco/metabolismo , Alantoide/embriologia , Animais , Morte Celular , Movimento Celular , Sobrevivência Celular , Sistema Digestório/citologia , Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Fator de Células-Tronco/genética , Fatores de Tempo
17.
Dev Cell ; 15(3): 470-477, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18804441

RESUMO

Mechanical deformations associated with embryonic morphogenetic movements have been suggested to actively participate in the signaling cascades regulating developmental gene expression. Here we develop an appropriate experimental approach to ascertain the existence and the physiological relevance of this phenomenon. By combining the use of magnetic tweezers with in vivo laser ablation, we locally control physiologically relevant deformations in wild-type Drosophila embryonic tissues. We demonstrate that the deformations caused by germ band extension upregulate Twist expression in the stomodeal primordium. We find that stomodeal compression triggers Src42A-dependent nuclear translocation of Armadillo/beta-catenin, which is required for Twist mechanical induction in the stomodeum. Finally, stomodeal-specific RNAi-mediated silencing of Twist during compression impairs the differentiation of midgut cells, resulting in larval lethality. These experiments show that mechanically induced Twist upregulation in stomodeal cells is necessary for subsequent midgut differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese , Proteína 1 Relacionada a Twist/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Gastrulação , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Transdução de Sinais/fisiologia , Estresse Mecânico , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 1 Relacionada a Twist/genética
18.
Development ; 135(18): 3007-11, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18684739

RESUMO

Neural crest cells (NCCs) form at the dorsal margin of the neural tube and migrate along distinct pathways throughout the vertebrate embryo to generate multiple cell types. A subpopulation of vagal NCCs invades the foregut and colonises the entire gastrointestinal tract to form the enteric nervous system (ENS). The colonisation of embryonic gut by NCCs has been studied extensively in chick embryos, and genetic studies in mice have identified genes crucial for ENS development, including Ret. Here, we have combined mouse embryo and organotypic gut culture to monitor and experimentally manipulate the progenitors of the ENS. Using this system, we demonstrate that lineally marked intestinal ENS progenitors from E11.5 mouse embryos grafted into the early vagal NCC pathway of E8.5 embryos colonise the entire length of the gastrointestinal tract. By contrast, similar progenitors transplanted into Ret-deficient host embryos are restricted to the proximal foregut. Our findings establish an experimental system that can be used to explore the interactions of NCCs with their cellular environment and reveal a previously unrecognised non-cell-autonomous effect of Ret deletion on ENS development.


Assuntos
Sistema Digestório/embriologia , Sistema Nervoso Entérico/fisiologia , Proteínas Proto-Oncogênicas c-ret/genética , Deleção de Sequência , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Sistema Digestório/metabolismo , Embrião de Mamíferos , Sistema Nervoso Entérico/citologia , Camundongos , Camundongos Knockout , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/fisiologia , Técnicas de Cultura de Órgãos , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células-Tronco/metabolismo , Fatores de Tempo
19.
Dev Biol ; 322(1): 145-55, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18692041

RESUMO

Tracheal agenesis/atresia (TA) is a rare but fatal congenital disease in which the breathing tube fails to grow. The etiology of this serious condition remains largely unknown. We found that Bmp signaling is prominently present in the anterior foregut where the tracheal primordium originates and targeted ablation of Bmp4 (Bmp4(cko)) resulted in a loss-of-trachea phenotype that closely resembles the Floyd type II pathology, the most common form of TA in humans. In Bmp4(cko) embryos, tracheal specification was not affected; however, its outgrowth was severely impaired due to reduced epithelial and mesenchymal proliferation. In agreement, we also observed significant reduction in the expression of Cyclin D1, a key cell cycle regulator associated with cellular proliferation. However, the proliferative effect of Bmp signaling appears to be independent of Wnt signaling. Interestingly, we found significantly reduced expression of activated extracellular signal-regulated kinase (Erk) in the Bmp4(cko) ventral foregut, suggesting that Bmp signaling promotes Erk phosphorylation which has been associated with cellular proliferation. This study provides the first evidence linking Bmp signaling to tracheal formation by regulating the proliferative response of the anterior ventral foregut. Our finding sheds light on human tracheal malformations by providing a novel mouse model implicating Bmp signaling, non-canonical Erk activation and cellular proliferation.


Assuntos
Proteínas Morfogenéticas Ósseas/deficiência , Transdução de Sinais/genética , Traqueia/anormalidades , Doenças da Traqueia/congênito , Doenças da Traqueia/genética , Animais , Padronização Corporal/genética , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Proliferação de Células , Ciclina D1/metabolismo , Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Sistema Digestório/patologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Genes Reporter , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Traqueia/patologia , Doenças da Traqueia/embriologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
20.
Histochem Cell Biol ; 130(2): 387-97, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18386042

RESUMO

Recent evidence shows that the serine protease inhibitor Kazal type 3 (Spink3) has more diverse functions than expected. To gain insight into its function, we analyzed the spatiotemporal expression profile of Spink3, using in situ hybridization (ISH) and a Spink3+/lacZ knock-in mouse, in which lacZ was inserted into the Spink3 locus. Spink3lacZ expression was first observed in the foregut, midgut, hindgut and the forebrain/midbrain junction region at 9.5 days post coitus (dpc). In the pancreas, Spink3 mRNA was detected at 11.5 dpc, before formation of the typical shape of the exocrine structure of the pancreas. Acinar cell expression was clearly identified by 13.5 dpc. After differentiation of the intestinal tract, Spink3lacZ expression was observed in the large intestine at 11.5 dpc, followed by expression in the small intestine at 13.5 dpc, before appearance of intestinal digestive enzymes. Spink3 mRNA and Spink3lacZ activity were also detected in other tissues, including the mesonephric tubules and the urogenital ridge at 11.5 dpc, the genital swelling at 13.5 dpc, the ductus epididymis at 17.5 dpc, and the seminal vesicle at 8 weeks. These data suggest that Spink3 may play important roles in proliferation and/or differentiation of various cell types during development.


Assuntos
Sistema Digestório/embriologia , Embrião de Mamíferos/metabolismo , Glicoproteínas/biossíntese , Pâncreas/embriologia , Proteínas Secretadas pela Próstata/biossíntese , Sistema Urogenital/embriologia , Animais , Sistema Digestório/citologia , Sistema Digestório/metabolismo , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Glicoproteínas/genética , Camundongos , Camundongos Knockout , Pâncreas/citologia , Pâncreas/metabolismo , Proteínas Secretadas pela Próstata/genética , Inibidor da Tripsina Pancreática de Kazal , Sistema Urogenital/citologia , Sistema Urogenital/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA