Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(50): e2201097119, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36469766

RESUMEN

Despite the robust healing capacity of the liver, regenerative failure underlies numerous hepatic diseases, including the JAG1 haploinsufficient disorder, Alagille syndrome (ALGS). Cholestasis due to intrahepatic duct (IHD) paucity resolves in certain ALGS cases but fails in most with no clear mechanisms or therapeutic interventions. We find that modulating jag1b and jag2b allele dosage is sufficient to stratify these distinct outcomes, which can be either exacerbated or rescued with genetic manipulation of Notch signaling, demonstrating that perturbations of Jag/Notch signaling may be causal for the spectrum of ALGS liver severities. Although regenerating IHD cells proliferate, they remain clustered in mutants that fail to recover due to a blunted elevation of Notch signaling in the distal-most IHD cells. Increased Notch signaling is required for regenerating IHD cells to branch and segregate into the peripheral region of the growing liver, where biliary paucity is commonly observed in ALGS. Mosaic loss- and-gain-of-function analysis reveals Sox9b to be a key Notch transcriptional effector required cell autonomously to regulate these cellular dynamics during IHD regeneration. Treatment with a small-molecule putative Notch agonist stimulates Sox9 expression in ALGS patient fibroblasts and enhances hepatic sox9b expression, rescues IHD paucity and cholestasis, and increases survival in zebrafish mutants, thereby providing a proof-of-concept therapeutic avenue for this disorder.


Asunto(s)
Síndrome de Alagille , Conductos Biliares Intrahepáticos , Transducción de Señal , Animales , Humanos , Síndrome de Alagille/genética , Síndrome de Alagille/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Mosaicismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Regeneración , Conductos Biliares Intrahepáticos/citología , Conductos Biliares Intrahepáticos/patología , Fibroblastos
2.
Hepatology ; 75(3): 567-583, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34569629

RESUMEN

BACKGROUND AND AIMS: Alagille Syndrome (ALGS) is a congenital disorder caused by mutations in the Notch ligand gene JAGGED1, leading to neonatal loss of intrahepatic duct (IHD) cells and cholestasis. Cholestasis can resolve in certain patients with ALGS, suggesting regeneration of IHD cells. However, the mechanisms driving IHD cell regeneration following Jagged loss remains unclear. Here, we show that cholestasis due to developmental loss of IHD cells can be consistently phenocopied in zebrafish with compound jagged1b and jagged2b mutations or knockdown. APPROACH AND RESULTS: Leveraging the transience of jagged knockdown in juvenile zebrafish, we find that resumption of Jagged expression leads to robust regeneration of IHD cells through a Notch-dependent mechanism. Combining multiple lineage tracing strategies with whole-liver three-dimensional imaging, we demonstrate that the extrahepatic duct (EHD) is the primary source of multipotent progenitors that contribute to the regeneration, but not to the development, of IHD cells. Hepatocyte-to-IHD cell transdifferentiation is possible but rarely detected. Progenitors in the EHD proliferate and migrate into the liver with Notch signaling loss and differentiate into IHD cells if Notch signaling increases. Tissue-specific mosaic analysis with an inducible dominant-negative Fgf receptor suggests that Fgf signaling from the surrounding mesenchymal cells maintains this extrahepatic niche by directly preventing premature differentiation and allocation of EHD progenitors to the liver. Indeed, transcriptional profiling and functional analysis of adult mouse EHD organoids uncover their distinct differentiation and proliferative potential relative to IHD organoids. CONCLUSIONS: Our data show that IHD cells regenerate upon resumption of Jagged/Notch signaling, from multipotent progenitors originating from an Fgf-dependent extrahepatic stem cell niche. We posit that if Jagged/Notch signaling is augmented, through normal stochastic variation, gene therapy, or a Notch agonist, regeneration of IHD cells in patients with ALGS may be enhanced.


Asunto(s)
Síndrome de Alagille , Conductos Biliares Extrahepáticos , Conductos Biliares Intrahepáticos , Proteínas de Unión al Calcio , Proteína Jagged-1 , Regeneración Hepática/fisiología , Receptores Notch/metabolismo , Proteínas de Pez Cebra , Síndrome de Alagille/genética , Síndrome de Alagille/metabolismo , Animales , Conductos Biliares Extrahepáticos/crecimiento & desarrollo , Conductos Biliares Extrahepáticos/fisiología , Conductos Biliares Intrahepáticos/crecimiento & desarrollo , Conductos Biliares Intrahepáticos/fisiología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Transdiferenciación Celular , Modelos Animales de Enfermedad , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Elife ; 102021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33544077

RESUMEN

Genetic variants associated with type 2 diabetes (T2D) risk affect gene regulation in metabolically relevant tissues, such as pancreatic islets. Here, we investigated contributions of regulatory programs active during pancreatic development to T2D risk. Generation of chromatin maps from developmental precursors throughout pancreatic differentiation of human embryonic stem cells (hESCs) identifies enrichment of T2D variants in pancreatic progenitor-specific stretch enhancers that are not active in islets. Genes associated with progenitor-specific stretch enhancers are predicted to regulate developmental processes, most notably tissue morphogenesis. Through gene editing in hESCs, we demonstrate that progenitor-specific enhancers harboring T2D-associated variants regulate cell polarity genes LAMA1 and CRB2. Knockdown of lama1 or crb2 in zebrafish embryos causes a defect in pancreas morphogenesis and impairs islet cell development. Together, our findings reveal that a subset of T2D risk variants specifically affects pancreatic developmental programs, suggesting that dysregulation of developmental processes can predispose to T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Epigenoma , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
4.
Science ; 370(6515): 463-467, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33093109

RESUMEN

Vertebrate sensory organs arise from epithelial thickenings called placodes. Along with neural crest cells, cranial placodes are considered ectodermal novelties that drove evolution of the vertebrate head. The anterior-most placode generates the endocrine lobe [adenohypophysis (ADH)] of the pituitary, a master gland controlling growth, metabolism, and reproduction. In addition to known ectodermal contributions, we use lineage tracing and time-lapse imaging in zebrafish to identify an endodermal contribution to the ADH. Single-cell RNA sequencing of the adult pituitary reveals similar competency of endodermal and ectodermal epithelia to generate all endocrine cell types. Further, endoderm can generate a rudimentary ADH-like structure in the near absence of ectodermal contributions. The fish condition supports the vertebrate pituitary arising through interactions of an ancestral endoderm-derived proto-pituitary with newly evolved placodal ectoderm.


Asunto(s)
Endodermo/embriología , Adenohipófisis/embriología , Animales , Linaje de la Célula , Endodermo/citología , Adenohipófisis/citología , RNA-Seq , Análisis de la Célula Individual , Pez Cebra
5.
Development ; 146(14)2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31142539

RESUMEN

An early step in pancreas development is marked by the expression of the transcription factor Pdx1 within the pancreatic endoderm, where it is required for the specification of all endocrine cell types. Subsequently, Pdx1 expression becomes restricted to the ß-cell lineage, where it plays a central role in ß-cell function. This pivotal role of Pdx1 at various stages of pancreas development makes it an attractive target to enhance pancreatic ß-cell differentiation and increase ß-cell function. In this study, we used a newly generated zebrafish reporter to screen over 8000 small molecules for modulators of pdx1 expression. We found four hit compounds and validated their efficacy at different stages of pancreas development. Notably, valproic acid treatment increased pancreatic endoderm formation, while inhibition of TGFß signaling led to α-cell to ß-cell transdifferentiation. HC toxin, another HDAC inhibitor, enhances ß-cell function in primary mouse and human islets. Thus, using a whole organism screening strategy, this study identified new pdx1 expression modulators that can be used to influence different steps in pancreas and ß-cell development.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Islotes Pancreáticos/embriología , Modelos Animales , Organogénesis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/análisis , Pez Cebra , Animales , Animales Modificados Genéticamente , Células COS , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Chlorocebus aethiops , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/aislamiento & purificación , Inhibidores de Histona Desacetilasas/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Organogénesis/genética , Bibliotecas de Moléculas Pequeñas/aislamiento & purificación , Transactivadores/genética , Transactivadores/metabolismo , Ácido Valproico/aislamiento & purificación , Ácido Valproico/farmacología , Pez Cebra/embriología , Pez Cebra/genética
6.
Dev Biol ; 441(1): 127-131, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29964026

RESUMEN

In mouse, retinoic acid (RA) is required for the early phase of body axis extension controlled by a population of neuromesodermal progenitors (NMPs) in the trunk called expanding-NMPs, but not for the later phase of body axis extension controlled by a population of NMPs in the tail called depleting-NMPs. Recent observations suggest that zebrafish utilize depleting-NMPs but not expanding-NMPs for body axis extension. In zebrafish, a role for RA in body axis extension was not supported by previous studies on aldh1a2 (raldh2) mutants lacking RA synthesis. Here, by treating zebrafish embryos with an RA synthesis inhibitor, we also found that body axis extension and somitogenesis was not perturbed, although loss of pectoral fin and cardiac edema were observed consistent with previous studies. The conclusion that zebrafish diverges from mouse in not requiring RA for body axis extension is consistent with zebrafish lacking early expanding-NMPs to generate the trunk. We suggest that RA control of body axis extension was added to higher vertebrates during evolution of expanding-NMPs.


Asunto(s)
Embrión de Mamíferos/embriología , Embrión no Mamífero/embriología , Mesodermo/embriología , Células-Madre Neurales/metabolismo , Tretinoina/metabolismo , Pez Cebra/embriología , Animales , Embrión de Mamíferos/citología , Embrión no Mamífero/citología , Mesodermo/citología , Ratones , Células-Madre Neurales/citología , Especificidad de la Especie
7.
Cell Rep ; 23(11): 3146-3151, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29898387

RESUMEN

A standard approach in the identification of transcriptional enhancers is the use of transgenic animals carrying DNA elements joined to reporter genes inserted randomly in the genome. We examined elements near Tbx5, a gene required for forelimb development in humans and other vertebrates. Previous transgenic studies reported a mammalian Tbx5 forelimb enhancer located in intron 2 containing a putative retinoic acid response element and a zebrafish tbx5a forelimb (pectoral fin) enhancer located downstream that is conserved from fish to mammals. We used CRISPR/Cas9 gene editing to knockout the endogenous elements and unexpectedly found that deletion of the intron 2 and downstream elements, either singly or together in double knockouts, resulted in no effect on forelimb development. Our findings show that reporter transgenes may not identify endogenous enhancers and that in vivo genetic loss-of-function studies are required, such as CRISPR/Cas9, which is similar in effort to production of animals carrying reporter transgenes.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Miembro Anterior/crecimiento & desarrollo , Edición Génica , Proteínas de Dominio T Box/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente/metabolismo , Sistemas CRISPR-Cas/genética , Miembro Anterior/metabolismo , Intrones , Ratones , Pez Cebra/metabolismo
8.
Nat Commun ; 8(1): 769, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974684

RESUMEN

Liver duct paucity is characteristic of children born with Alagille Syndrome (ALGS), a disease associated with JAGGED1 mutations. Here, we report that zebrafish embryos with compound homozygous mutations in two Notch ligand genes, jagged1b (jag1b) and jagged2b (jag2b) exhibit a complete loss of canonical Notch activity and duct cells within the liver and exocrine pancreas, whereas hepatocyte and acinar pancreas development is not affected. Further, animal chimera studies demonstrate that wild-type endoderm cells within the liver and pancreas can rescue Notch activity and duct lineage specification in adjacent cells lacking jag1b and jag2b expression. We conclude that these two Notch ligands are directly and solely responsible for all duct lineage specification in these organs in zebrafish. Our study uncovers genes required for lineage specification of the intrahepatopancreatic duct cells, challenges the role of duct cells as progenitors, and suggests a genetic mechanism for ALGS ductal paucity.The hepatopancreatic duct cells connect liver hepatocytes and pancreatic acinar cells to the intestine, but the mechanism for their lineage specification is unclear. Here, the authors reveal that Notch ligands Jagged1b and Jagged2b induce duct cell lineage in the liver and pancreas of the zebrafish.


Asunto(s)
Conductos Biliares Intrahepáticos/embriología , Proteínas de Unión al Calcio/genética , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Jagged-2/genética , Conductos Pancreáticos/embriología , Proteínas de Pez Cebra/genética , Síndrome de Alagille/genética , Animales , Linaje de la Célula , Endodermo/citología , Pez Cebra
9.
Dev Biol ; 395(1): 96-110, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25176043

RESUMEN

To gain insight into liver and pancreas development, we investigated the target of 2F11, a monoclonal antibody of unknown antigen, widely used in zebrafish studies for labeling hepatopancreatic ducts. Utilizing mass spectrometry and in vivo assays, we determined the molecular target of 2F11 to be Annexin A4 (Anxa4), a calcium binding protein. We further found that in both zebrafish and mouse endoderm, Anxa4 is broadly expressed in the developing liver and pancreas, and later becomes more restricted to the hepatopancreatic ducts and pancreatic islets, including the insulin producing ß-cells. Although Anxa4 is a known target of several monogenic diabetes genes and its elevated expression is associated with chemoresistance in malignancy, its in vivo role is largely unexplored. Knockdown of Anxa4 in zebrafish leads to elevated expression of caspase 8 and Δ113p53, and liver bud specific activation of Caspase 3 and apoptosis. Mosaic knockdown reveal that Anxa4 is required cell-autonomously in the liver bud for cell survival. This finding is further corroborated with mosaic anxa4 knockout studies using the CRISPR/Cas9 system. Collectively, we identify Anxa4 as a new, evolutionarily conserved hepatopancreatic factor that is required in zebrafish for liver progenitor viability, through inhibition of the extrinsic apoptotic pathway. A role for Anxa4 in cell survival may have implications for the mechanism of diabetic ß-cell apoptosis and cancer cell chemoresistance.


Asunto(s)
Anexina A4/metabolismo , Hígado/metabolismo , Páncreas/metabolismo , Proteínas de Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Anexina A4/genética , Apoptosis/genética , Secuencia de Bases , Caspasa 3/metabolismo , Supervivencia Celular , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Hibridación in Situ , Hígado/citología , Hígado/embriología , Microscopía Confocal , Datos de Secuencia Molecular , Páncreas/citología , Páncreas/embriología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
10.
Mol Cell ; 53(6): 1005-19, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24530304

RESUMEN

Here, we generated a genome-scale shRNA library targeting long intergenic noncoding RNAs (lincRNAs) in the mouse. We performed an unbiased loss-of-function study in mouse embryonic stem cells (mESCs) and identified 20 lincRNAs involved in the maintenance of pluripotency. Among these, TUNA (Tcl1 Upstream Neuron-Associated lincRNA, or megamind) was required for pluripotency and formed a complex with three RNA-binding proteins (RBPs). The TUNA-RBP complex was detected at the promoters of Nanog, Sox2, and Fgf4, and knockdown of TUNA or the individual RBPs inhibited neural differentiation of mESCs. TUNA showed striking evolutionary conservation of both sequence- and CNS-restricted expression in vertebrates. Accordingly, knockdown of tuna in zebrafish caused impaired locomotor function, and TUNA expression in the brains of Huntington's disease patients was significantly associated with disease grade. Our results suggest that the lincRNA TUNA plays a vital role in pluripotency and neural differentiation of ESCs and is associated with neurological function of adult vertebrates.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Enfermedad de Huntington/genética , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Evolución Biológica , Diferenciación Celular , Secuencia Conservada , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Datos de Secuencia Molecular , Actividad Motora , Proteína Homeótica Nanog , Neuronas/citología , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Homología de Secuencia de Aminoácido , Índice de Severidad de la Enfermedad , Transducción de Señal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
11.
Development ; 140(13): 2669-79, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23720049

RESUMEN

Although the liver and ventral pancreas are thought to arise from a common multipotent progenitor pool, it is unclear whether these progenitors of the hepatopancreas system are specified by a common genetic mechanism. Efforts to determine the role of Hnf1b and Wnt signaling in this crucial process have been confounded by a combination of factors, including a narrow time frame for hepatopancreas specification, functional redundancy among Wnt ligands, and pleiotropic defects caused by either severe loss of Wnt signaling or Hnf1b function. Using a novel hypomorphic hnf1ba zebrafish mutant that exhibits pancreas hypoplasia, as observed in HNF1B monogenic diabetes, we show that hnf1ba plays essential roles in regulating ß-cell number and pancreas specification, distinct from its function in regulating pancreas size and liver specification, respectively. By combining Hnf1ba partial loss of function with conditional loss of Wnt signaling, we uncover a crucial developmental window when these pathways synergize to specify the entire ventrally derived hepatopancreas progenitor population. Furthermore, our in vivo genetic studies demonstrate that hnf1ba generates a permissive domain for Wnt signaling activity in the foregut endoderm. Collectively, our findings provide a new model for HNF1B function, yield insight into pancreas and ß-cell development, and suggest a new mechanism for hepatopancreatic specification.


Asunto(s)
Factor Nuclear 1-beta del Hepatocito/metabolismo , Hepatopáncreas/citología , Hepatopáncreas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Factor Nuclear 1-beta del Hepatocito/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Wnt/genética , Pez Cebra , Proteínas de Pez Cebra/genética
12.
Dev Biol ; 334(1): 213-23, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19631206

RESUMEN

Developmental mechanisms regulating gene expression and the stable acquisition of cell fate direct cytodifferentiation during organogenesis. Moreover, it is likely that such mechanisms could be exploited to repair or regenerate damaged organs. DNA methyltransferases (Dnmts) are enzymes critical for epigenetic regulation, and are used in concert with histone methylation and acetylation to regulate gene expression and maintain genomic integrity and chromosome structure. We carried out two forward genetic screens for regulators of endodermal organ development. In the first, we screened for altered morphology of developing digestive organs, while in the second we screed for the lack of terminally differentiated cell types in the pancreas and liver. From these screens, we identified two mutant alleles of zebrafish dnmt1. Both lesions are predicted to eliminate dnmt1 function; one is a missense mutation in the catalytic domain and the other is a nonsense mutation that eliminates the catalytic domain. In zebrafish dnmt1 mutants, the pancreas and liver form normally, but begin to degenerate after 84 h post fertilization (hpf). Acinar cells are nearly abolished through apoptosis by 100 hpf, though neither DNA replication, nor entry into mitosis is halted in the absence of detectable Dnmt1. However, endocrine cells and ducts are largely spared. Surprisingly, dnmt1 mutants and dnmt1 morpholino-injected larvae show increased capacity for pancreatic beta cell regeneration in an inducible model of pancreatic beta cell ablation. Thus, our data suggest that Dnmt1 is dispensable for pancreatic duct or endocrine cell formation, but not for acinar cell survival. In addition, Dnmt1 may influence the differentiation of pancreatic beta cell progenitors or the reprogramming of cells toward the pancreatic beta cell fate.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Páncreas/citología , Regeneración/fisiología , Proteínas de Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Supervivencia Celular , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Células Endocrinas/metabolismo , Técnica del Anticuerpo Fluorescente , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Datos de Secuencia Molecular , Páncreas/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
13.
Dev Biol ; 322(2): 237-50, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18687323

RESUMEN

Liver, pancreas and lung originate from the presumptive foregut in temporal and spatial proximity. This requires precisely orchestrated transcriptional activation and repression of organ-specific gene expression within the same cell. Here, we show distinct roles for the chromatin remodelling factor and transcriptional repressor Histone deacetylase 1 (Hdac1) in endodermal organogenesis in zebrafish. Loss of Hdac1 causes defects in timely liver specification and in subsequent differentiation. Mosaic analyses reveal a cell-autonomous requirement for hdac1 within the hepatic endoderm. Our studies further reveal specific functions for Hdac1 in pancreas development. Loss of hdac1 causes the formation of ectopic endocrine clusters anteriorly to the main islet, as well as defects in exocrine pancreas specification and differentiation. In addition, we observe defects in extrahepatopancreatic duct formation and morphogenesis. Finally, loss of hdac1 results in an expansion of the foregut endoderm in the domain from which the liver and pancreas originate. Our genetic studies demonstrate that Hdac1 is crucial for regulating distinct steps in endodermal organogenesis. This suggests a model in which Hdac1 may directly or indirectly restrict foregut fates while promoting hepatic and exocrine pancreatic specification and differentiation, as well as pancreatic endocrine islet morphogenesis. These findings establish zebrafish as a tractable system to investigate chromatin remodelling factor functions in controlling gene expression programmes in vertebrate endodermal organogenesis.


Asunto(s)
Histona Desacetilasas/metabolismo , Hígado/embriología , Páncreas/embriología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Endodermo/embriología , Hepatocitos/citología , Hepatocitos/fisiología , Histona Desacetilasa 1 , Histona Desacetilasas/genética , Hígado/enzimología , Pulmón/embriología , Pulmón/enzimología , Datos de Secuencia Molecular , Mutación , Especificidad de Órganos , Páncreas/enzimología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
14.
Genes Dev ; 22(11): 1445-50, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519637

RESUMEN

The mechanisms regulating pancreatic endocrine versus exocrine fate are not well defined. By analyzing the effects of Ptf1a partial loss of function, we uncovered novel roles for this transcription factor in determining pancreatic fates. In a newly identified hypomorphic ptf1a mutant, pancreatic cells that would normally express ptf1a and become exocrine cells, express the endocrine marker Isl1, indicating a cell fate switch. Surprisingly, a milder reduction of Ptf1a leads to an even greater increase of ectopic endocrine cells, suggesting that Ptf1a also plays a role in promoting endocrine development. We propose that low levels of Ptf1a promote endocrine fate, whereas high levels repress endocrine fate and promote exocrine fate.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Islotes Pancreáticos/embriología , Páncreas Exocrino/embriología , Páncreas/embriología , Proteínas de Pez Cebra/genética , Animales , Páncreas/metabolismo , Factores de Transcripción/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
Nat Genet ; 39(3): 397-402, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17259985

RESUMEN

During organogenesis, the foregut endoderm gives rise to the many different cell types that comprise the hepatopancreatic system, including hepatic, pancreatic and gallbladder cells, as well as the epithelial cells of the hepatopancreatic ductal system that connects these organs together and with the intestine. However, the mechanisms responsible for demarcating ducts versus organs are poorly understood. Here, we show that Fgf10 signaling from the adjacent mesenchyme is responsible for refining the boundaries between the hepatopancreatic duct and organs. In zebrafish fgf10 mutants, the hepatopancreatic ductal epithelium is severely dysmorphic, and cells of the hepatopancreatic ductal system and adjacent intestine misdifferentiate toward hepatic and pancreatic fates. Furthermore, Fgf10 also functions to prevent the differentiation of the proximal pancreas and liver into hepatic and pancreatic cells, respectively. These data shed light onto how the multipotent cells of the foregut endoderm, and subsequently those of the hepatopancreatic duct, are directed toward different organ fates.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatopáncreas/embriología , Mesodermo/citología , Organogénesis , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Tipificación del Cuerpo , Diferenciación Celular , Embrión no Mamífero , Factor 10 de Crecimiento de Fibroblastos/genética , Técnica del Anticuerpo Fluorescente , Hepatopáncreas/anatomía & histología , Hepatopáncreas/metabolismo , Mesodermo/metabolismo , Transducción de Señal , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
16.
Dev Biol ; 286(1): 114-35, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16125164

RESUMEN

Recent analysis of a novel strain of transgenic zebrafish (gutGFP) has provided a detailed description of the early morphological events that occur during the development of the liver and pancreas. In this paper, we aim to complement these studies by providing an analysis of the morphological events that shape the zebrafish intestinal epithelium. One of our goals is to provide a framework for the future characterization of zebrafish mutant phenotypes in which intestinal epithelial morphogenesis has been disrupted. Our analysis encompasses the period between 26 and 126 h post-fertilization (hpf) and follows the growth, lumen formation and differentiation of a continuous layer of endoderm into a functional intestinal epithelium with three morphologically distinct segments: the intestinal bulb, mid-intestine and posterior intestine. Between 26 hpf and 76 hpf, the entire intestinal endoderm is a highly proliferative organ. To make a lumen, the zebrafish endoderm cells undergo apical membrane biogenesis, adopt a bilayer configuration and form small cavities that coalesce without cell death. Thereafter, the endoderm cells polarize and differentiate into distinct cell lineages. Enteroendocrine cells are distinguished first at 52 hpf in the caudal region of the intestine in a new stable transgenic line, Tg[nkx2.2a:mEGFP]. The differentiation of mucin-containing goblet cells is first evident at 100 hpf and is tightly restricted to a middle segment of the intestine, designated the mid-intestine, that is also demarcated by the presence of enterocytes with large supranuclear vacuoles. Meanwhile, striking expansion of the lumen in the rostral intestine forms the intestinal bulb. Here the epithelium elaborates folds and proliferating cells become progressively restricted to a basal compartment analogous to the crypts of Lieberkühn in mammals. At 126 hpf, the posterior intestine remains an unfolded monolayer of simple columnar epithelium.


Asunto(s)
Intestinos/embriología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Diferenciación Celular , Polaridad Celular , Proliferación Celular , ADN Recombinante/genética , Endodermo/citología , Endodermo/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Mucosa Intestinal/metabolismo , Intestinos/citología , Morfogénesis , Proteínas Recombinantes/genética , Pez Cebra/genética , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...