Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gastroenterology ; 160(4): 1330-1344.e11, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33212097

RESUMEN

BACKGROUND & AIMS: Molecular evidence of cellular heterogeneity in the human exocrine pancreas has not been yet established because of the local concentration and cascade of hydrolytic enzymes that can rapidly degrade cells and RNA upon pancreatic resection. We sought to better understand the heterogeneity and cellular composition of the pancreas in neonates and adults in healthy and diseased conditions using single-cell sequencing approaches. METHODS: We innovated single-nucleus RNA-sequencing protocols and profiled more than 120,000 cells from pancreata of adult and neonatal human donors. We validated the single-nucleus findings using RNA fluorescence in situ hybridization, in situ sequencing, and computational approaches. RESULTS: We created the first comprehensive atlas of human pancreas cells including epithelial and nonepithelial constituents, and uncovered 3 distinct acinar cell types, with possible implications for homeostatic and inflammatory processes of the pancreas. The comparison with neonatal single-nucleus sequencing data showed a different cellular composition of the endocrine tissue, highlighting the tissue dynamics occurring during development. By applying spatial cartography, involving cell proximity mapping through in situ sequencing, we found evidence of specific cell type neighborhoods, dynamic topographies in the endocrine and exocrine pancreas, and principles of morphologic organization of the organ. Furthermore, similar analyses in chronic pancreatitis biopsy samples showed the presence of acinar-REG+ cells, a reciprocal association between macrophages and activated stellate cells, and a new potential role of tuft cells in this disease. CONCLUSIONS: Our human pancreas cell atlas can be interrogated to understand pancreatic cell biology and provides a crucial reference set for comparisons with diseased tissue samples to map the cellular foundations of pancreatic diseases.


Asunto(s)
Núcleo Celular/metabolismo , Páncreas Exocrino/citología , Adolescente , Adulto , Factores de Edad , Anciano , Animales , Fraccionamiento Celular , Niño , Preescolar , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Masculino , Persona de Mediana Edad , Modelos Animales , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/metabolismo , RNA-Seq , Análisis de la Célula Individual/métodos , Porcinos , Adulto Joven
2.
Cell Mol Gastroenterol Hepatol ; 7(4): 841-856, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30831323

RESUMEN

BACKGROUND & AIMS: The Onecut 1 transcription factor (Oc1, a.k.a. HNF6) promotes differentiation of endocrine and duct cells of the pancreas; however, it has no known role in acinar cell differentiation. We sought to better understand the role of Oc1 in exocrine pancreas development and to identify its direct transcriptional targets. METHODS: Pancreata from Oc1Δpanc (Oc1fl/fl;Pdx1-Cre) mouse embryos and neonates were analyzed morphologically. High-throughput RNA-sequencing was performed on control and Oc1-deficient pancreas; chromatin immunoprecipitation sequencing was performed on wild-type embryonic mouse pancreata to identify direct Oc1 transcriptional targets. Immunofluorescence labeling was used to confirm the RNA-sequencing /chromatin immunoprecipitation sequencing results and to further investigate the effects of Oc1 loss on acinar cells. RESULTS: Loss of Oc1 from the developing pancreatic epithelium resulted in disrupted duct and acinar cell development. RNA-sequencing revealed decreased expression of acinar cell regulatory factors (Nr5a2, Ptf1a, Gata4, Mist1) and functional genes (Amylase, Cpa1, Prss1, Spink1) at embryonic day (e) 18.5 in Oc1Δpanc samples. Approximately 1000 of the altered genes were also identified as direct Oc1 targets by chromatin immunoprecipitation sequencing, including most of the previously noted genes. By immunolabeling, we confirmed that Amylase, Mist1, and GATA4 protein levels are significantly decreased by P2, and Spink1 protein levels were significantly reduced and mislocalized. The pancreatic duct regulatory factors Hnf1ß and FoxA2 were also identified as direct Oc1 targets. CONCLUSIONS: These findings confirm that Oc1 is an important regulator of both duct and acinar cell development in the embryonic pancreas. Novel transcriptional targets of Oc1 have now been identified and provide clarity into the mechanisms of Oc1 transcriptional regulation in the developing exocrine pancreas. Oc1 can now be included in the gene-regulatory network of acinar cell regulatory genes. Oc1 regulates other acinar cell regulatory factors and acinar cell functional genes directly, and it can also regulate some acinar cell regulatory factors (eg, Mist1) indirectly. Oc1 therefore plays an important role in acinar cell development.


Asunto(s)
Diferenciación Celular , Factor Nuclear 6 del Hepatocito/metabolismo , Morfogénesis , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/patología , Células Acinares/patología , Animales , Animales Recién Nacidos , Secuencia de Bases , Proliferación Celular , Embrión de Mamíferos/patología , Epitelio/crecimiento & desarrollo , Epitelio/patología , Regulación del Desarrollo de la Expresión Génica , Ratones , Páncreas Exocrino/metabolismo
3.
Sci Rep ; 8(1): 13451, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30194315

RESUMEN

Cadherin-mediated cell-cell adhesion plays an important role in organ development and changes in cadherin expression are often linked to morphogenetic and pathogenic events. Cadherins interact with other intracellular components to form adherens junctions (AJs) and provide mechanical attachments between adjacent cells. E-cadherin (Cdh1) represents an integral component of these intercellular junctions. To elucidate the function of E-cadherin in the developing pancreas, we generated and studied pancreas-specific Cdh1-knockout (Cdh1ΔPan/ΔPan) mice. Cdh1ΔPan/ΔPan mice exhibit normal body size at birth, but fail to gain weight and become hypoglycemic soon afterward. We found that E-cadherin is not required for the establishment of apical-basal polarity or pancreatic exocrine cell identity at birth. However, four days after birth, the pancreata of Cdh1ΔPan/ΔPan mutants display progressive deterioration of exocrine architecture and dysregulation of Wnt and YAP signaling. At this time point, the acinar cells of Cdh1ΔPan/ΔPan mutants begin to exhibit ductal phenotypes, suggesting acinar-to-ductal metaplasia (ADM) in the E-cadherin-deficient pancreas. Our findings demonstrate that E-cadherin plays an integral role in the maintenance of exocrine architecture and regulation of homeostatic signaling. The present study provides insights into the involvement of cadherin-mediated cell-cell adhesion in pathogenic conditions such as pancreatitis or pancreatic cancer.


Asunto(s)
Cadherinas/metabolismo , Páncreas Exocrino/crecimiento & desarrollo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Cadherinas/genética , Adhesión Celular/genética , Proteínas de Ciclo Celular , Ratones , Ratones Noqueados , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pancreatitis/genética , Pancreatitis/metabolismo , Pancreatitis/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Señalizadoras YAP
4.
J Biol Chem ; 293(31): 11971-11983, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-29871925

RESUMEN

Degenerative diseases of organs lead to their impaired function. The cellular and molecular mechanisms underlying organ degeneration are therefore of great research and clinical interest but are currently incompletely characterized. Here, using a forward-genetic screen for genes regulating liver development and function in zebrafish, we identified a cq5 mutant that exhibited a liver-degeneration phenotype at 5 days postfertilization, the developmental stage at which a functional liver develops. Positional cloning revealed that the liver degeneration was caused by a single point mutation in the gene zc3h8 (zinc finger CCCH-type containing 8), changing a highly conserved histidine to glutamine at position 353 of the Zc3h8 protein. The zc3h8 mutation-induced liver degeneration in the mutant was accompanied by reduced proliferation, increased apoptosis, and macrophage phagocytosis of hepatocytes. Transcriptional profile analyses revealed up-regulation and activation of both proinflammatory cytokines and the NF-κB signaling pathway in the zc3h8 mutant. Suppression of NF-κB signaling activity efficiently rescued the proinflammatory cytokine response, as well as the inflammation-mediated liver degeneration phenotype of the mutant. Of note, the zc3h8 mutation-induced degeneration of several other organs, including the gut and exocrine pancreas, indicating that Zc3h8 is a general repressor of inflammation in zebrafish. Collectively, our findings demonstrate that Zc3h8 maintains organ homeostasis by inhibiting the NF-κB-mediated inflammatory response in zebrafish and that Zc3h8 dysfunction causes degeneration of multiple organs, including the liver, gut, and pancreas.


Asunto(s)
Hepatocitos/metabolismo , Hígado/metabolismo , FN-kappa B/genética , Páncreas Exocrino/metabolismo , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Apoptosis , Proliferación Celular , Citocinas/genética , Citocinas/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Glutamina/metabolismo , Hepatocitos/patología , Histidina/metabolismo , Inflamación , Intestinos/anomalías , Intestinos/crecimiento & desarrollo , Hígado/anomalías , Hígado/crecimiento & desarrollo , Macrófagos/metabolismo , Macrófagos/patología , Mutación , FN-kappa B/metabolismo , Páncreas Exocrino/anomalías , Páncreas Exocrino/crecimiento & desarrollo , Fagocitosis , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Factores de Transcripción/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo , Dedos de Zinc
5.
PLoS Genet ; 13(4): e1006740, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28410364

RESUMEN

Arf4 is proposed to be a critical regulator of membrane protein trafficking in early secretory pathway. More recently, Arf4 was also implicated in regulating ciliary trafficking, however, this has not been comprehensively tested in vivo. To directly address Arf4's role in ciliary transport, we deleted Arf4 specifically in either rod photoreceptor cells, kidney, or globally during the early postnatal period. Arf4 deletion in photoreceptors did not cause protein mislocalization or retinal degeneration, as expected if Arf4 played a role in protein transport to the ciliary outer segment. Likewise, Arf4 deletion in kidney did not cause cystic disease, as expected if Arf4 were involved in general ciliary trafficking. In contrast, global Arf4 deletion in the early postnatal period resulted in growth restriction, severe pancreatic degeneration and early death. These findings are consistent with Arf4 playing a critical role in endomembrane trafficking, particularly in the pancreas, but not in ciliary function.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Enfermedades Renales Quísticas/genética , Páncreas Exocrino/patología , Degeneración Retiniana/genética , Animales , Cilios/genética , Cilios/patología , Modelos Animales de Enfermedad , Humanos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales Quísticas/patología , Ratones , Ratones Noqueados , Motivos de Nucleótidos/genética , Páncreas Exocrino/crecimiento & desarrollo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Degeneración Retiniana/patología , Eliminación de Secuencia
6.
J Exp Zool B Mol Dev Evol ; 324(8): 699-706, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26477613

RESUMEN

Despite the growing importance of zebrafish (Danio rerio) as an experimental model in biomedical research, some aspect of physiological and related morphological age dependent changes in digestive system during larval development are still unknown. In this paper, a biochemical and morphological study of the digestive tract of zebrafish was undertaken to record the functional changes occurring in this species during its ontogenetic development, particularly from 24 hr to 47 days post fertilization (dpf). Endo- and exo-proteases, as well as α-amylase enzymes, were quantified in zebrafish larvae before first feeding (7 dpf). The most morphologically significant events during the ontogenesis of the gut occurred between 3 dpf (mouth opening) and 7 dpf (end of exocrine pancreas differentiation). The presence of a wide range of digestive enzymes, already active at earlier zebrafish larval stages, closely related with the omnivorous diet of this species. Increasing enzyme activities were found with increasing age, probably in relation with intestinal mucosa folding and consequent absorption surface increase. J. Exp. Zool. (Mol. Dev. Evol.) 324B: 699-706, 2015. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Sistema Digestivo/enzimología , Pez Cebra/metabolismo , Animales , Sistema Digestivo/anatomía & histología , Sistema Digestivo/crecimiento & desarrollo , Larva/enzimología , Larva/crecimiento & desarrollo , Páncreas Exocrino/enzimología , Páncreas Exocrino/crecimiento & desarrollo , Pez Cebra/crecimiento & desarrollo
7.
Dev Dyn ; 244(6): 724-35, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25773748

RESUMEN

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.


Asunto(s)
Páncreas/embriología , Factores de Transcripción/fisiología , Pez Cebra/embriología , Células Acinares/citología , Animales , Animales Modificados Genéticamente , Linaje de la Célula , Cromosomas Artificiales Bacterianos , Vesícula Biliar/citología , Dosificación de Gen , Genotipo , Islotes Pancreáticos/citología , Islotes Pancreáticos/embriología , Islotes Pancreáticos/crecimiento & desarrollo , Especificidad de Órganos , Páncreas/citología , Páncreas/crecimiento & desarrollo , Páncreas/fisiología , Páncreas Exocrino/citología , Páncreas Exocrino/embriología , Páncreas Exocrino/crecimiento & desarrollo , Receptores Notch/fisiología , Recombinación Genética , Regeneración , Células Madre/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
8.
Dev Biol ; 381(2): 471-81, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23876428

RESUMEN

Development of the pancreas and cerebellum require Pancreas-specific transcription factor-1a (Ptf1a), which encodes a subunit of the transcription factor complex PTF1. Ptf1a is required in succession for specification of the pancreas, proper allocation of pancreatic progenitors to endocrine and exocrine fates, and the production of digestive enzymes from the exocrine acini. In several neuronal structures, including the cerebellum, hindbrain, retina and spinal cord, Ptf1a is transiently expressed and promotes inhibitory neuron fates at the expense of excitatory fates. Transcription of Ptf1a in mouse is maintained in part by PTF1 acting on an upstream autoregulatory enhancer. However, the transcription factors and enhancers that initially activate Ptf1a expression in the pancreas and in certain structures of the nervous system have not yet been identified. Here we describe a zebrafish autoregulatory element, conserved among teleosts, with activity similar to that described in mouse. In addition, we performed a comprehensive survey of all non-coding sequences in a 67kb interval encompassing zebrafish ptf1a, and identified several neuronal enhancers, and an enhancer active in the ventral pancreas prior to activation of the autoregulatory enhancer. To test the requirement for autoregulatory control during pancreatic development, we restored ptf1a function through BAC transgenesis in ptf1a morphants, either with an intact BAC or one lacking the autoregulatory enhancer. We find that ptf1a autoregulation is required for development of the exocrine pancreas and full rescue of the ptf1a morphant phenotype. Similarly, we demonstrate that a ptf1a locus lacking the early enhancer region is also capable of rescue, but only supports formation of a hypoplastic exocrine pancreas. Through our dissection of the complex regulatory control of ptf1a, we identified separate cis-regulatory elements that underlie different aspects of its expression and function, and further demonstrated the requirement of maintained ptf1a expression for normal pancreatic morphogenesis. We also identified a novel enhancer that mediates initiation of ptf1a expression in the pancreas, through which the signals that specify the ventral pancreas are expected to exert their action.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Páncreas Exocrino/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Cromosomas Artificiales Bacterianos/metabolismo , Secuencia Conservada , Embrión no Mamífero/metabolismo , Técnicas de Transferencia de Gen , Sitios Genéticos , Homeostasis , Datos de Secuencia Molecular , Páncreas Exocrino/metabolismo , Fenotipo , Secuencias Reguladoras de Ácidos Nucleicos , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética
9.
Zebrafish ; 10(2): 161-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23697888

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Páncreas Exocrino/embriología , Proteínas Ribosómicas/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Hibridación in Situ , Larva/genética , Larva/metabolismo , Microscopía Confocal , Especificidad de Órganos , Organogénesis , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Ribosómicas/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
10.
Zebrafish ; 10(2): 132-46, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23682805

RESUMEN

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.


Asunto(s)
Adenocarcinoma/etiología , Adenocarcinoma/terapia , Modelos Animales de Enfermedad , Páncreas/embriología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/terapia , Pez Cebra/embriología , Adenocarcinoma/embriología , Adenocarcinoma/genética , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Biomarcadores/metabolismo , Descubrimiento de Drogas , Genes ras , Humanos , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo , Páncreas Exocrino/embriología , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/metabolismo , Neoplasias Pancreáticas/embriología , Neoplasias Pancreáticas/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
11.
PLoS One ; 8(4): e58858, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23593122

RESUMEN

Digestive organs originate from the endoderm. Morphogenesis of the digestive system is precisely controlled by multiple factors that dictate the cell fate and behavior so that the specific digestive organs are timely formed in the right place and develop into right size and structure. We showed previously that digestive organ expansion factor (def) is a gene whose expression is enriched in the liver, pancreas and intestine. Loss-of-function of def in the def(hi429) mutant confers hypoplastic digestive organs partly due to alteration of expression of genes related to the p53 pathway. However, the molecular mechanism for the involvement of Def in the organogenesis of digestive organs is still largely unknown. For example, it is not known whether Def regulates specific pathways in a specific organ. To address this question, we generated four independent Tg(fabp10a:def) transgenic fish lines which over-expressed Def specifically in the liver. We characterized Tg-I, one of the transgenic lines, in detail with genetic, molecular and histological approaches. We found that Tg-I restored the liver but not exocrine pancreas and intestine development in the def(hi429) mutant. However, Tg-I adult fish in the wild type (WT) background exhibits reduced liver-to-body ratio and all four transgenic lines conferred abnormal intrahepatic structure. Microarray data analysis showed that certain specific functional pathways were affected in the liver of Tg-I. These results demonstrate that Def functions in a cell autonomous manner during early liver development and aberrant Def protein expression might lead to disruption of the structural integrity of a normal adult liver.


Asunto(s)
Sistema Digestivo/citología , Sistema Digestivo/crecimiento & desarrollo , Organogénesis , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Envejecimiento/metabolismo , Animales , Animales Modificados Genéticamente , Nucléolo Celular/metabolismo , Regulación hacia Abajo/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Intestinos/crecimiento & desarrollo , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Especificidad de Órganos , Páncreas Exocrino/crecimiento & desarrollo , Transporte de Proteínas , ARN Ribosómico 18S/metabolismo , Transgenes , Regulación hacia Arriba/genética , Proteínas de Pez Cebra/genética
13.
Gastroenterology ; 144(7): 1543-53, 1553.e1, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454691

RESUMEN

BACKGROUND & AIMS: The Hippo signaling pathway is a context-dependent regulator of cell proliferation, differentiation, and apoptosis in species ranging from Drosophila to humans. In this study, we investigated the role of the core Hippo kinases-Mst1 and Mst2-in pancreatic development and homeostasis. METHODS: We used a Cre/LoxP system to create mice with pancreas-specific disruptions in Mst1 and Mst2 (Pdx1-Cre;Mst1(-/-);Mst2(fl/fl) mice), the mammalian orthologs of Drosophila Hippo. We used a transgenic approach to overexpress Yap, the downstream mediator of Hippo signaling, in the developing pancreas of mice. RESULTS: Contrary to expectations, the pancreatic mass of Pdx1-Cre;Mst1(-/-);Mst2(fl/fl) mice was reduced compared with wild-type mice, largely because of postnatal de-differentiation of acinar cells into duct-like cells. Development of this phenotype coincided with postnatal reactivation of YAP expression. Ectopic expression of YAP during the secondary transition (a stage at which YAP is normally absent) blocked differentiation of the endocrine and exocrine compartments, whereas loss of a single Yap allele reduced acinar de-differentiation. The phenotype of Pdx1-Cre;Mst1(-/-);Mst2(fl/fl) mice recapitulated cellular and molecular changes observed during chemical-induced pancreatitis in mice. CONCLUSIONS: The mammalian Hippo kinases, and YAP, maintain postnatal pancreatic acinar differentiation in mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Páncreas Exocrino/crecimiento & desarrollo , Fosfoproteínas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Proliferación Celular , Ratones , Ratones Transgénicos , Páncreas Exocrino/fisiología , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Serina-Treonina Quinasa 3 , Transducción de Señal , Proteínas Señalizadoras YAP
14.
Gut ; 62(10): 1481-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23002247

RESUMEN

OBJECTIVES: Previous studies have suggested an important role of the transcription factor Gata6 in endocrine pancreas, while GATA6 haploinsufficient inactivating mutations cause pancreatic agenesis in humans. We aimed to analyse the effects of Gata6 inactivation on pancreas development and function. DESIGN: We deleted Gata6 in all epithelial cells in the murine pancreas at the onset of its development. Acinar proliferation, apoptosis, differentiation and exocrine functions were assessed using reverse transcriptase quantitative PCR (RT-qPCR), chromatin immunoprecipitation, immunohistochemistry and enzyme assays. Adipocyte transdifferentiation was assessed using electron microscopy and genetic lineage tracing. RESULTS: Gata6 is expressed in all epithelial cells in the adult mouse pancreas but it is only essential for exocrine pancreas homeostasis: while dispensable for pancreatic development after e10.5, it is required for complete acinar differentiation, for establishment of polarity and for the maintenance of acinar cells in the adult. Gata6 regulates directly the promoter of genes coding for digestive enzymes and the transcription factors Rbpjl and Mist1. Upon pancreas-selective Gata6 inactivation, massive loss of acinar cells and fat replacement take place. This is accompanied by increased acinar apoptosis and proliferation, acinar-to-ductal metaplasia and adipocyte transdifferentiation. By contrast, the endocrine pancreas is spared. CONCLUSIONS: Our data show that Gata6 is required for the complete differentiation of acinar cells through multiple transcriptional regulatory mechanisms. In addition, it is required for the maintenance of the adult acinar cell compartment. Our studies suggest that GATA6 alterations may contribute to diseases of the human adult exocrine pancreas.


Asunto(s)
Células Acinares/citología , Factor de Transcripción GATA6/fisiología , Páncreas Exocrino/citología , Células Acinares/patología , Células Acinares/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Femenino , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Eliminación de Gen , Masculino , Metaplasia/genética , Metaplasia/patología , Ratones , Ratones Noqueados , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/patología , Páncreas Exocrino/fisiología
15.
Eur J Cell Biol ; 90(12): 1052-60, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21906833

RESUMEN

The development of in vitro models able to support the long-term viability and function of acinar cells is critical for exploring pancreatic pathophysiology. Despite considerable efforts, no long-term culture models for non-transformed pancreatic acini exist. Our aim was to develop and validate culture conditions for this purpose. An explant outgrowth culture design was established in which mouse pancreatic explants were cultured at the gas-liquid interphase. An enriched culture medium, pH 7.8, was employed to promote the selective outgrowth of acinar cells and to support their differentiated phenotype. After 7 days, the outgrown primary acinar cells were subcultured and maintained up to an additional 7 days as secondary monolayers on tissue culture plastic. Measurements of basal and caerulein-induced amylase secretion, phase-contrast microscopy and immunohistochemical analyses were used to characterize the cultures. Explants retained their pancreatic cytoarchitecture for 2 days in vitro. A triphasic dose response to caerulein was detected in 7-day primary cultures. The maximal rate of secretion was 1.2-fold versus basal (p=0.009) and 1.7-fold versus 1 pM caerulein (p=0.014). In secondary cultures the response was biphasic with maximal rates of secretion being 1.9-fold in 3- to 4-day cultures at 0.01 nM (p=0.049) and 2-fold in 6- to 7-day cultures at 0.1 nM (p=0.003). The present culture model provides a means to obtain functionally competent normal mouse acinar cells for long-term in vitro experimentation.


Asunto(s)
Células Acinares/citología , Páncreas Exocrino/citología , Páncreas Exocrino/crecimiento & desarrollo , Células Acinares/química , Animales , Supervivencia Celular/fisiología , Técnicas Citológicas , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas Exocrino/química , Fenotipo
16.
Am J Physiol Gastrointest Liver Physiol ; 301(5): G791-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21852360

RESUMEN

Adaptive exocrine pancreatic growth is mediated primarily by dietary protein and the gastrointestinal hormone cholecystokinin (CCK). Feeding trypsin inhibitors such as camostat (FOY-305) is known to induce CCK release and stimulate pancreatic growth. However, camostat has also been reported to stimulate secretin release and, because secretin often potentiates the action of CCK, it could participate in the growth response. Our aim was to test the role of secretin in pancreatic development and adaptive growth through the use of C57BL/6 mice with genetic deletion of secretin or secretin receptor. The lack of secretin in the intestine or the secretin receptor in the pancreas was confirmed by RT-PCR. Other related components, such as vasoactive intestinal polypeptide (VIP) receptors (VPAC(1) and VPAC(2)), were not affected. Secretin increased cAMP levels in acini from wild-type (WT) mice but had no effect on acini from secretin receptor-deleted mice, whereas VIP and forskolin still induced a normal response. Secretin in vivo failed to induce fluid secretion in receptor-deficient mice. The pancreas of secretin or secretin receptor-deficient mice was of normal size and histology, indicating that secretin is not necessary for normal pancreatic differentiation or maintenance. When WT mice were fed 0.1% camostat in powdered chow, the pancreas doubled in size in 1 wk, accompanied by parallel increases in protein and DNA. Camostat-fed littermate secretin and secretin receptor-deficient mice had similar pancreatic mass to WT mice. These results indicate that secretin is not required for normal pancreatic development or adaptive growth mediated by CCK.


Asunto(s)
Páncreas Exocrino/crecimiento & desarrollo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de la Hormona Gastrointestinal/metabolismo , Secretina/metabolismo , Células Acinares/metabolismo , Animales , Colecistoquinina/metabolismo , AMP Cíclico/metabolismo , Masculino , Ratones , Ratones Noqueados , Páncreas Exocrino/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de la Hormona Gastrointestinal/genética , Secretina/genética , Péptido Intestinal Vasoactivo/metabolismo
17.
Development ; 138(4): 609-17, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21208992

RESUMEN

The Notch-signaling pathway is known to be fundamental in controlling pancreas differentiation. We now report on using Cre-based fate mapping to indelibly label pancreatic Notch-responsive cells (PNCs) at larval stages and follow their fate in the adult pancreas. We show that the PNCs represent a population of progenitors that can differentiate to multiple lineages, including adult ductal cells, centroacinar cells (CACs) and endocrine cells. These endocrine cells include the insulin-producing ß-cells. CACs are a functional component of the exocrine pancreas; however, our fate-mapping results indicate that CACs are more closely related to endocrine cells by lineage as they share a common progenitor. The majority of the exocrine pancreas consists of the secretory acinar cells; however, we only detect a very limited contribution of PNCs to acinar cells. To explain this observation we re-examined early events in pancreas formation. The pancreatic anlage that gives rise to the exocrine pancreas is located in the ventral gut endoderm (called the ventral bud). Ptf1a is a gene required for exocrine pancreas development and is first expressed as the ventral bud forms. We used transgenic marker lines to observe both the domain of cells expressing ptf1a and cells responding to Notch signaling. We do not detect any overlap in expression and demonstrate that the ventral bud consists of two cell populations: a ptf1-expressing domain and a Notch-responsive progenitor core. As pancreas organogenesis continues, the ventral bud derived PNCs align along the duct, remain multipotent and later in development differentiate to form secondary islets, ducts and CACs.


Asunto(s)
Envejecimiento , Linaje de la Célula , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Pez Cebra/crecimiento & desarrollo , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Larva/genética , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/metabolismo , Receptores Notch/metabolismo , Pez Cebra/metabolismo
18.
Dev Biol ; 351(2): 311-7, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21194527

RESUMEN

The tadpole pancreas has differentiated acinar cells but an underdeveloped ductal system. At the climax of metamorphosis thyroid hormone (TH) induces the tadpole acinar cells to dedifferentiate to a progenitor state. After metamorphosis is complete the exocrine pancreas redifferentiates in the growing frog forming a typical vertebrate pancreas including a complex ductal system. A micro array analysis found that TH up regulates stromelysin 3 (ST3, matrix metalloproteinase 11) in the exocrine pancreas at metamorphic climax. Transgenic tadpoles were prepared with an elastase promoter driving either the ST3 gene or the constitutively active form of Notch (IC). Expression of the transgenes was controlled by the tetracycline system. A few days after either of these transgenes is activated by doxycycline the pancreatic acinar cells turn into duct-like cells. This transdetermination occurs without cell division since both acinar and ductal markers can be visualized transiently in the same cell. We propose that remodeling of the tadpole acinar cells is initiated when ST3 is up regulated by TH. Stromelysin-3 then cleaves and activates Notch.


Asunto(s)
Diferenciación Celular , Metaloproteinasa 11 de la Matriz/fisiología , Metamorfosis Biológica , Páncreas Exocrino/crecimiento & desarrollo , Conductos Pancreáticos/crecimiento & desarrollo , Receptores Notch/fisiología , Xenopus laevis/crecimiento & desarrollo , Animales , Transdiferenciación Celular , Larva , Metamorfosis Biológica/efectos de los fármacos , Metaplasia , Páncreas Exocrino/citología , Hormonas Tiroideas/farmacología
19.
Am J Physiol Gastrointest Liver Physiol ; 299(5): G1154-63, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20798356

RESUMEN

Dietary protein can stimulate pancreatic growth in the absence of CCK release, but there is little data on the regulation of CCK-independent growth. To identify mechanisms whereby protein stimulates pancreatic growth in the absence of CCK release, C57BL/6 control and CCK-null male mice were fed normal-protein (14% casein) or high-protein (75% casein) chow for 7 days. The weight of the pancreas increased by 32% in C57BL/6 mice and 26% in CCK-null mice fed high-protein chow. Changes in pancreatic weight in control mice were due to both cell hypertrophy and hyperplasia since there was an increase in protein-to-DNA ratio, total DNA content, and DNA synthesis. In CCK-null mice pancreatic growth was almost entirely due to hypertrophy with both protein-to-DNA ratio and cell size increasing without significant increases in DNA content or DNA synthesis. ERK, calcineurin, and mammalian target of rapamycin complex 1 (mTORC1) are activated in models of CCK-induced growth, but there were no differences in ERK or calcineurin activation between fasted and fed CCK-null mice. In contrast, mTORC1 activation was increased after feeding and the duration of activation was prolonged in mice fed high-protein chow compared with normal-protein chow. Changes in pancreatic weight and RNA content were completely inhibited, and changes in protein content were partially abated, when the mTORC1 inhibitor rapamycin was administered during high-protein chow feeding. Prolonged mTORC1 activation is thus required for dietary protein-induced pancreatic growth in the absence of CCK.


Asunto(s)
Colecistoquinina/metabolismo , Proteínas en la Dieta/metabolismo , Páncreas Exocrino/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Análisis de Varianza , Animales , Western Blotting , Calcineurina/metabolismo , Colecistoquinina/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos , Tamaño de los Órganos , Páncreas Exocrino/metabolismo , Fosforilación , Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
20.
Fish Physiol Biochem ; 36(4): 993-1000, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20077135

RESUMEN

The ontogeny and differentiation stages of digestive systems related with trypsin expression in larvae of sharpsnout sea bream, Diplodus puntazzo, were investigated from hatching to 40 DAH (days after hatching), and total lengths and weights of larvae were determined. Histologic and enzymatic techniques were used to explain the functional development of the pancreas including trypsin activity. The pancreas was identified as a compact structure located in the region slightly posterior to the liver. At 3 DAH, first anus and then mouth opened. Incipient pancreas secretion polyhedral cells could be first observed as zymogen granules. During larval metamorphosis, the pancreas became diffuse, spreading throughout the mesentery in proximity to the stomach, the anterior intestine and the pyloric caeca. The specific activity of trypsin (42.54±6.8 mU/mg protein(-1)) was found as early as after hatching at larvae size of 2.87±0.34 mm at 0 DAH. Activity further increased until 10 DAH, especially after exogenous feeding. The highest trypsin activity was detected at 25 DAH as 119.26±11.6 mU/mg protein(-1). It is concluded that exocrine pancreas organogenesis is the main critical step in the development of digestive system that results in zymogen granules accumulation and increased trypsin activity.


Asunto(s)
Organogénesis/fisiología , Páncreas Exocrino/crecimiento & desarrollo , Dorada/fisiología , Tripsina/metabolismo , Análisis de Varianza , Animales , Técnicas Histológicas , Larva/crecimiento & desarrollo , Larva/metabolismo , Dorada/metabolismo , Espectrofotometría
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...