Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594183

RESUMEN

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


Asunto(s)
Huesos/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Intestino Delgado/citología , Células Madre Mesenquimatosas/citología , Animales , Cartílago/metabolismo , Intestino Delgado/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Nature ; 573(7774): 421-425, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31511693

RESUMEN

Early human embryonic development involves extensive lineage diversification, cell-fate specification and tissue patterning1. Despite its basic and clinical importance, early human embryonic development remains relatively unexplained owing to interspecies divergence2,3 and limited accessibility to human embryo samples. Here we report that human pluripotent stem cells (hPSCs) in a microfluidic device recapitulate, in a highly controllable and scalable fashion, landmarks of the development of the epiblast and amniotic ectoderm parts of the conceptus, including lumenogenesis of the epiblast and the resultant pro-amniotic cavity, formation of a bipolar embryonic sac, and specification of primordial germ cells and primitive streak cells. We further show that amniotic ectoderm-like cells function as a signalling centre to trigger the onset of gastrulation-like events in hPSCs. Given its controllability and scalability, the microfluidic model provides a powerful experimental system to advance knowledge of human embryology and reproduction. This model could assist in the rational design of differentiation protocols of hPSCs for disease modelling and cell therapy, and in high-throughput drug and toxicity screens to prevent pregnancy failure and birth defects.


Asunto(s)
Amnios/embriología , Estratos Germinativos/embriología , Modelos Biológicos , Células Madre Pluripotentes/citología , Amnios/citología , Diferenciación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Femenino , Estratos Germinativos/citología , Humanos , Embarazo , Línea Primitiva/citología
3.
Annu Rev Physiol ; 83: 359-380, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33035430

RESUMEN

The hedgehog (Hh) signaling pathway plays several diverse regulatory and patterning roles during organogenesis of the intestine and in the regulation of adult intestinal homeostasis. In the embryo, fetus, and adult, intestinal Hh signaling is paracrine: Hh ligands are expressed in the endodermally derived epithelium, while signal transduction is confined to the mesenchymal compartment, where at least a dozen distinct cell types are capable of responding to Hh signals. Epithelial Hh ligands not only regulate a variety of mesenchymal cell behaviors, but they also direct these mesenchymal cells to secrete additional soluble factors (e.g., Wnts, Bmps, inflammatory mediators) that feed back to regulate the epithelial cells themselves. Evolutionary conservation of the core Hh signaling pathway, as well as conservation of epithelial/mesenchymal cross talk in the intestine, has meant that work in many diverse model systems has contributed to our current understanding of the role of this pathway in intestinal organogenesis, which is reviewed here.


Asunto(s)
Proteínas Hedgehog/metabolismo , Homeostasis/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiología , Intestinos/fisiología , Transducción de Señal/fisiología , Animales , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Humanos
4.
Development ; 147(20)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32994164

RESUMEN

Between embryonic days 10.5 and 14.5, active proliferation drives rapid elongation of the murine midgut epithelial tube. Within this pseudostratified epithelium, nuclei synthesize DNA near the basal surface and move apically to divide. After mitosis, the majority of daughter cells extend a long, basally oriented filopodial protrusion, building a de novo path along which their nuclei can return to the basal side. WNT5A, which is secreted by surrounding mesenchymal cells, acts as a guidance cue to orchestrate this epithelial pathfinding behavior, but how this signal is received by epithelial cells is unknown. Here, we have investigated two known WNT5A receptors: ROR2 and RYK. We found that epithelial ROR2 is dispensable for midgut elongation. However, loss of Ryk phenocopies the Wnt5a-/- phenotype, perturbing post-mitotic pathfinding and leading to apoptosis. These studies reveal that the ligand-receptor pair WNT5A-RYK acts as a navigation system to instruct filopodial pathfinding, a process that is crucial for continuous cell cycling to fuel rapid midgut elongation.


Asunto(s)
Sistema Digestivo/crecimiento & desarrollo , Sistema Digestivo/metabolismo , Seudópodos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Apoptosis , Núcleo Celular/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Femenino , Masculino , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo
5.
Development ; 143(13): 2261-72, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27381224

RESUMEN

The vertebrate small intestine requires an enormous surface area to effectively absorb nutrients from food. Morphological adaptations required to establish this extensive surface include generation of an extremely long tube and convolution of the absorptive surface of the tube into villi and microvilli. In this Review, we discuss recent findings regarding the morphogenetic and molecular processes required for intestinal tube elongation and surface convolution, examine shared and unique aspects of these processes in different species, relate these processes to known human maladies that compromise absorptive function and highlight important questions for future research.


Asunto(s)
Absorción Intestinal , Intestinos/crecimiento & desarrollo , Animales , Humanos , Microvellosidades/metabolismo , Modelos Biológicos , Morfogénesis , Transducción de Señal
6.
Development ; 143(3): 427-36, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26721501

RESUMEN

In the intestine, finger-like villi provide abundant surface area for nutrient absorption. During murine villus development, epithelial Hedgehog (Hh) signals promote aggregation of subepithelial mesenchymal clusters that drive villus emergence. Clusters arise first dorsally and proximally and spread over the entire intestine within 24 h, but the mechanism driving this pattern in the murine intestine is unknown. In chick, the driver of cluster pattern is tensile force from developing smooth muscle, which generates deep longitudinal epithelial folds that locally concentrate the Hh signal, promoting localized expression of cluster genes. By contrast, we show that in mouse, muscle-induced epithelial folding does not occur and artificial deformation of the epithelium does not determine the pattern of clusters or villi. In intestinal explants, modulation of Bmp signaling alters the spatial distribution of clusters and changes the pattern of emerging villi. Increasing Bmp signaling abolishes cluster formation, whereas inhibiting Bmp signaling leads to merged clusters. These dynamic changes in cluster pattern are faithfully simulated by a mathematical model of a Turing field in which an inhibitor of Bmp signaling acts as the Turing activator. In vivo, genetic interruption of Bmp signal reception in either epithelium or mesenchyme reveals that Bmp signaling in Hh-responsive mesenchymal cells controls cluster pattern. Thus, unlike in chick, the murine villus patterning system is independent of muscle-induced epithelial deformation. Rather, a complex cocktail of Bmps and Bmp signal modulators secreted from mesenchymal clusters determines the pattern of villi in a manner that mimics the spread of a self-organizing Turing field.


Asunto(s)
Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/metabolismo , Intestinos/embriología , Microvellosidades/metabolismo , Transducción de Señal , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Epitelio/embriología , Proteínas Hedgehog/metabolismo , Hibridación in Situ , Ligandos , Mesodermo/embriología , Ratones Endogámicos C57BL , Modelos Biológicos , Músculo Liso/embriología , Tamaño de los Órganos , Resistencia a la Tracción
7.
Development ; 143(20): 3711-3722, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27802136

RESUMEN

During late gestation, villi extend into the intestinal lumen to dramatically increase the surface area of the intestinal epithelium, preparing the gut for the neonatal diet. Incomplete development of the intestine is the most common gastrointestinal complication in neonates, but the causes are unclear. We provide evidence in mice that Yin Yang 1 (Yy1) is crucial for intestinal villus development. YY1 loss in the developing endoderm had no apparent consequences until late gestation, after which the intestine differentiated poorly and exhibited severely stunted villi. Transcriptome analysis revealed that YY1 is required for mitochondrial gene expression, and ultrastructural analysis confirmed compromised mitochondrial integrity in the mutant intestine. We found increased oxidative phosphorylation gene expression at the onset of villus elongation, suggesting that aerobic respiration might function as a regulator of villus growth. Mitochondrial inhibitors blocked villus growth in a fashion similar to Yy1 loss, thus further linking oxidative phosphorylation with late-gestation intestinal development. Interestingly, we find that necrotizing enterocolitis patients also exhibit decreased expression of oxidative phosphorylation genes. Our study highlights the still unappreciated role of metabolic regulation during organogenesis, and suggests that it might contribute to neonatal gastrointestinal disorders.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/citología , Organogénesis/fisiología , Factor de Transcripción YY1/metabolismo , Aerobiosis/genética , Aerobiosis/fisiología , Animales , Western Blotting , Genotipo , Inmunohistoquímica , Masculino , Ratones , Organogénesis/genética , Fosforilación Oxidativa , Transcriptoma/genética , Factor de Transcripción YY1/genética
8.
Nat Mater ; 16(4): 419-425, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27941807

RESUMEN

Amniogenesis-the development of amnion-is a critical developmental milestone for early human embryogenesis and successful pregnancy. However, human amniogenesis is poorly understood due to limited accessibility to peri-implantation embryos and a lack of in vitro models. Here we report an efficient biomaterial system to generate human amnion-like tissue in vitro through self-organized development of human pluripotent stem cells (hPSCs) in a bioengineered niche mimicking the in vivo implantation environment. We show that biophysical niche factors act as a switch to toggle hPSC self-renewal versus amniogenesis under self-renewal-permissive biochemical conditions. We identify a unique molecular signature of hPSC-derived amnion-like cells and show that endogenously activated BMP-SMAD signalling is required for the amnion-like tissue development by hPSCs. This study unveils the self-organizing and mechanosensitive nature of human amniogenesis and establishes the first hPSC-based model for investigating peri-implantation human amnion development, thereby helping advance human embryology and reproductive medicine.


Asunto(s)
Amnios/metabolismo , Materiales Biomiméticos , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Nicho de Células Madre , Ingeniería de Tejidos/métodos , Amnios/citología , Línea Celular , Humanos , Células Madre Pluripotentes/citología , Medicina Reproductiva/métodos
9.
Dev Dyn ; 245(5): 614-26, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26930384

RESUMEN

BACKGROUND: Digestion is facilitated by coordinated contractions of the intestinal muscularis externa, a bilayered smooth muscle structure that is composed of inner circular muscles (ICM) and outer longitudinal muscles (OLM). We performed transcriptome analysis of intestinal mesenchyme tissue at E14.5, when the ICM, but not the OLM, is present, to investigate the transcriptional program of the ICM. RESULTS: We identified 3967 genes enriched in E14.5 intestinal mesenchyme. The gene expression profiles were clustered and annotated to known muscle genes, identifying a muscle-enriched subcluster. Using publically available in situ data, 127 genes were verified as expressed in ICM. Examination of the promoter and regulatory regions for these co-expressed genes revealed enrichment for cJUN transcription factor binding sites, and cJUN protein was enriched in ICM. cJUN ChIP-seq, performed at E14.5, revealed that cJUN regulatory regions contain characteristics of muscle enhancers. Finally, we show that cJun is a target of Hedgehog (Hh), a signaling pathway known to be important in smooth muscle development, and identify a cJun genomic enhancer that is responsive to Hh. CONCLUSIONS: This work provides the first transcriptional catalog for the developing ICM and suggests that cJun regulates gene expression in the ICM downstream of Hh signaling. Developmental Dynamics 245:614-626, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Intestinos/embriología , Músculo Liso/embriología , Transcriptoma , Animales , Genes jun/fisiología , Proteínas Hedgehog , Ratones
10.
BMC Dev Biol ; 16: 4, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26912062

RESUMEN

BACKGROUND: The Hedgehog (Hh) signaling pathway, acting through three homologous transcription factors (GLI1, GLI2, GLI3) in vertebrates, plays multiple roles in embryonic organ development and adult tissue homeostasis. At the level of the genome, GLI factors bind to specific motifs in enhancers, some of which are hundreds of kilobases removed from the gene promoter. These enhancers integrate the Hh signal in a context-specific manner to control the spatiotemporal pattern of target gene expression. Importantly, a number of genes that encode Hh pathway molecules are themselves targets of Hh signaling, allowing pathway regulation by an intricate balance of feed-back activation and inhibition. However, surprisingly few of the critical enhancer elements that control these pathway target genes have been identified despite the fact that such elements are central determinants of Hh signaling activity. Recently, ChIP studies have been carried out in multiple tissue contexts using mouse models carrying FLAG-tagged GLI proteins (GLI(FLAG)). Using these datasets, we tested whether a meta-analysis of GLI binding sites, coupled with a machine learning approach, could reveal genomic features that could be used to empirically identify Hh-regulated enhancers linked to loci of the Hh signaling pathway. RESULTS: A meta-analysis of four existing GLI(FLAG) datasets revealed a library of GLI binding motifs that was substantially more restricted than the potential sites predicted by previous in vitro binding studies. A machine learning method (kmer-SVM) was then applied to these datasets and enriched k-mers were identified that, when applied to the mouse genome, predicted as many as 37,000 potential Hh enhancers. For functional analysis, we selected nine regions which were annotated to putative Hh pathway molecules and found that seven exhibited GLI-dependent activity, indicating that they are directly regulated by Hh signaling (78% success rate). CONCLUSIONS: The results suggest that Hh enhancer regions share common sequence features. The kmer-SVM machine learning approach identifies those features and can successfully predict functional Hh regulatory regions in genomic DNA surrounding Hh pathway molecules and likely, other Hh targets. Additionally, the library of enriched GLI binding motifs that we have identified may allow improved identification of functional GLI binding sites.


Asunto(s)
Biología Computacional/métodos , Elementos de Facilitación Genéticos/genética , Proteínas Hedgehog/genética , Transducción de Señal/genética , Animales , Secuencia de Bases , Línea Celular , Proteínas Hedgehog/metabolismo , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Motivos de Nucleótidos/genética , Proteínas Oncogénicas/metabolismo , Unión Proteica , Reproducibilidad de los Resultados , Máquina de Vectores de Soporte , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1
11.
Gastroenterology ; 146(1): 157-165.e10, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24120474

RESUMEN

BACKGROUND & AIMS: Infantile hypertrophic pyloric stenosis is a common birth anomaly characterized by obstruction of the pyloric lumen. A genome-wide association study implicated NKX2-5, which encodes a transcription factor that is expressed in embryonic heart and pylorus, in the pathogenesis of infantile hypertrophic pyloric stenosis. However, the function of the NKX2-5 in pyloric smooth muscle development has not been examined directly. We investigated the pattern of Nkx2-5 during the course of murine pyloric sphincter development and examined coexpression of Nkx2-5 with Gata3 and Sox9-other transcription factors with pyloric-specific mesenchymal expression. We also assessed pyloric sphincter development in mice with disruption of Nkx2-5 or Gata3. METHODS: We used immunofluorescence analysis to compare levels of NKX2-5, GATA3, and SOX9 in different regions of smooth muscle cells. Pyloric development was assessed in mice with conditional or germline deletion of Nkx2-5 or Gata3, respectively. RESULTS: Gata3, Nkx2-5, and Sox9 are coexpressed in differentiating smooth muscle cells of a distinct fascicle of the pyloric outer longitudinal muscle. Expansion of this fascicle coincides with development of the pyloric sphincter. Disruption of Nkx2-5 or Gata3 causes severe hypoplasia of this fascicle and alters pyloric muscle shape. Although expression of Sox9 requires Nkx2-5 and Gata3, there is no apparent hierarchical relationship between Nkx2-5 and Gata3 during pyloric outer longitudinal muscle development. CONCLUSIONS: Nkx2-5 and Gata3 are independently required for the development of a pyloric outer longitudinal muscle fascicle, which is required for pyloric sphincter morphogenesis in mice. These data indicate that regulatory changes that alter Nkx2-5 or Gata3 expression could contribute to pathogenesis of infantile hypertrophic pyloric stenosis.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Proteínas de Homeodominio/metabolismo , Desarrollo de Músculos/fisiología , Músculo Liso/embriología , Miocitos del Músculo Liso/metabolismo , Píloro/embriología , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Proteína Homeótica Nkx-2.5 , Ratones , Músculo Liso/metabolismo , Píloro/metabolismo
12.
Development ; 139(3): 488-97, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22190634

RESUMEN

Notch signaling is known to regulate the proliferation and differentiation of intestinal stem and progenitor cells; however, direct cellular targets and specific functions of Notch signals had not been identified. We show here in mice that Notch directly targets the crypt base columnar (CBC) cell to maintain stem cell activity. Notch inhibition induced rapid CBC cell loss, with reduced proliferation, apoptotic cell death and reduced efficiency of organoid initiation. Furthermore, expression of the CBC stem cell-specific marker Olfm4 was directly dependent on Notch signaling, with transcription activated through RBP-Jκ binding sites in the promoter. Notch inhibition also led to precocious differentiation of epithelial progenitors into secretory cell types, including large numbers of cells that expressed both Paneth and goblet cell markers. Analysis of Notch function in Atoh1-deficient intestine demonstrated that the cellular changes were dependent on Atoh1, whereas Notch regulation of Olfm4 gene expression was Atoh1 independent. Our findings suggest that Notch targets distinct progenitor cell populations to maintain adult intestinal stem cells and to regulate cell fate choice to control epithelial cell homeostasis.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Regulación de la Expresión Génica , Intestino Delgado/citología , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Caliciformes/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Técnicas de Cultivo de Órganos , Células de Paneth/metabolismo , Regiones Promotoras Genéticas , Receptor Notch1/antagonistas & inhibidores , Receptor Notch2/antagonistas & inhibidores , Transducción de Señal , Células Madre/citología , Células Madre/fisiología
13.
Proc Natl Acad Sci U S A ; 109(39): 15817-22, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23019366

RESUMEN

In the adult intestine, an organized array of finger-like projections, called villi, provide an enormous epithelial surface area for absorptive function. Villi first emerge at embryonic day (E) 14.5 from a previously flat luminal surface. Here, we analyze the cell biology of villus formation and examine the role of paracrine epithelial Hedgehog (Hh) signals in this process. We find that, before villus emergence, tight clusters of Hh-responsive mesenchymal cells form just beneath the epithelium. Cluster formation is dynamic; clusters first form dorsally and anteriorly and spread circumferentially and posteriorly. Statistical analysis of cluster distribution reveals a patterned array; with time, new clusters form in spaces between existing clusters, promoting approximately four rounds of villus emergence by E18.5. Cells within mesenchymal clusters express Patched1 and Gli1, as well as Pdgfrα, a receptor previously shown to participate in villus development. BrdU-labeling experiments show that clusters form by migration and aggregation of Hh-responsive cells. Inhibition of Hh signaling prevents cluster formation and villus development, but does not prevent emergence of villi in areas where clusters have already formed. Conversely, increasing Hh signaling increases the size of villus clusters and results in exceptionally wide villi. We conclude that Hh signals dictate the initial aspects of the formation of each villus by controlling mesenchymal cluster aggregation and regulating cluster size.


Asunto(s)
Proteínas Hedgehog/metabolismo , Mucosa Intestinal/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Hedgehog/genética , Humanos , Mucosa Intestinal/citología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Transgénicos , Receptores Patched , Receptor Patched-1 , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteína con Dedos de Zinc GLI1
14.
Dev Biol ; 382(1): 82-97, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23916850

RESUMEN

The adult fungiform taste papilla is a complex of specialized cell types residing in the stratified squamous tongue epithelium. This unique sensory organ includes taste buds, papilla epithelium and lateral walls that extend into underlying connective tissue to surround a core of lamina propria cells. Fungiform papillae must contain long-lived, sustaining or stem cells and short-lived, maintaining or transit amplifying cells that support the papilla and specialized taste buds. Shh signaling has established roles in supporting fungiform induction, development and patterning. However, for a full understanding of how Shh transduced signals act in tongue, papilla and taste bud formation and maintenance, it is necessary to know where and when the Shh ligand and pathway components are positioned. We used immunostaining, in situ hybridization and mouse reporter strains for Shh, Ptch1, Gli1 and Gli2-expression and proliferation markers to identify cells that participate in hedgehog signaling. Whereas there is a progressive restriction in location of Shh ligand-expressing cells, from placode and apical papilla cells to taste bud cells only, a surrounding population of Ptch1 and Gli1 responding cells is maintained in signaling centers throughout papilla and taste bud development and differentiation. The Shh signaling targets are in regions of active cell proliferation. Using genetic-inducible lineage tracing for Gli1-expression, we found that Shh-responding cells contribute not only to maintenance of filiform and fungiform papillae, but also to taste buds. A requirement for normal Shh signaling in fungiform papilla, taste bud and filiform papilla maintenance was shown by Gli2 constitutive activation. We identified proliferation niches where Shh signaling is active and suggest that epithelial and mesenchymal compartments harbor potential stem and/or progenitor cell zones. In all, we report a set of hedgehog signaling centers that regulate development and maintenance of taste organs, the fungiform papilla and taste bud, and surrounding lingual cells. Shh signaling has roles in forming and maintaining fungiform papillae and taste buds, most likely via stage-specific autocrine and/or paracrine mechanisms, and by engaging epithelial/mesenchymal interactions.


Asunto(s)
Epitelio/embriología , Epitelio/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Papilas Gustativas/embriología , Papilas Gustativas/metabolismo , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Compartimento Celular , Linaje de la Célula , Proliferación Celular , Microambiente Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Factores de Transcripción de Tipo Kruppel/metabolismo , Ligandos , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo , Papilas Gustativas/citología , Papilas Gustativas/ultraestructura , Factores de Tiempo , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
15.
Am J Physiol Gastrointest Liver Physiol ; 307(4): G430-6, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24970776

RESUMEN

The gastric ligaments are superficial cord-like structures, located on the lesser curvature of the stomach, that extend from the pylorus to the esophagus. These ligaments have been documented in a wide variety of mammalian species, including humans, but their composition and ontogeny is unexplored. Here, we demonstrate that, during ontogeny, the gastric ligaments are first visible as extensions from a C-shaped domain of Gata3-expressing cells that surround the future pylorus; this domain will later give rise to the pyloric outer longitudinal muscle (OLM). The open ends of the C are located ventrally, and, beginning at embryonic day (E) 13.5, the ligaments grow anteriorly from these points. Whereas most other ligaments of the stomach are neurovascular in nature, the gastric ligaments are composed of smooth muscle cells that mature between E14.5 and E16.5. The gastric ligaments coexpress the transcription factors Gata3, Nkx2-5, and Sox9, and germline loss of Gata3 or conditional deletion of Nkx2-5 abrogates Sox9 expression and impairs gastric ligament smooth muscle development; similar phenotypes were previously seen in the OLM. In accord with this molecular contiguity between the OLM and gastric ligaments, three-dimensional image reconstruction highlights physical contiguity between these smooth muscle structures, suggesting that they may work together as a unit to control flexure of the pyloric region, a function similar to the ligament of Treitz at the duodenojejunal junction. These findings may have implications for our understanding of normal pyloric sphincter function, as well as the common human congenital pathology idiopathic hypertrophic pyloric stenosis.


Asunto(s)
Factor de Transcripción GATA3/fisiología , Proteínas de Homeodominio/fisiología , Ligamentos/embriología , Músculo Liso/embriología , Factores de Transcripción/fisiología , Animales , Proteína Homeótica Nkx-2.5 , Ligamentos/metabolismo , Ratones , Miocitos del Músculo Liso/metabolismo , Factor de Transcripción SOX9/biosíntesis
16.
Gastroenterology ; 144(7): 1478-87, 1487.e1-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23470617

RESUMEN

BACKGROUND & AIMS: Stress alters brain-gut interactions and could exacerbate intestinal disorders, including irritable bowel syndrome. Alterations in the intestinal microbiota have been associated with irritable bowel syndrome. Maintenance of healthy microbiota requires nucleotide-binding oligomerization domain protein-like receptors, pyrin-domain containing (NLRP)-6 inflammasomes. We investigated the involvement of NLRP6 in water-avoidance stress (WAS)-induced intestinal disorders in mice. METHODS: B57BL6 mice were subjected to WAS for 1 hour each day for 10 days; body weights and intestinal inflammation and permeability were analyzed. We investigated signaling via the NLRP3 and NLRP6 inflammasomes, and the role of corticotropin-releasing hormone (CRH) in WAS-associated inflammation and NLRP6 inhibition. Mice that were not exposed to stress were co-housed with mice subjected to WAS to determine the effects of WAS-induced dysbiosis, measured by sequencing bacterial 16S ribosomal RNA. We also assessed the effects of a peroxisome proliferator-activated receptor-γ agonist and probiotics. RESULTS: WAS-induced small-bowel inflammation (enteritis) was associated with inhibition of NLRP6, but not NLRP3, and was prevented by a peroxisome proliferator-activated receptor-γ agonist, which induced epithelial expression of NLRP6. CRH was released during WAS and inhibited NLRP6 expression. WAS induced alterations in the gut microbiota of mice; co-housed nonstressed mice developed enteritis associated with increased CRH and decreased levels of NLRP6. Probiotic therapy reduced intestinal inflammation in mice with WAS-induced enteritis. CONCLUSIONS: Exposure of mice to stress inhibits NLRP6 and alters the composition of the gut microbiota, leading to intestinal inflammation. These findings might explain the benefits of probiotics for patients with stress-associated gastrointestinal disorders.


Asunto(s)
Hormona Liberadora de Corticotropina/fisiología , Enteritis/etiología , Receptores de Superficie Celular/fisiología , Estrés Psicológico/fisiopatología , Animales , Modelos Animales de Enfermedad , Enteritis/terapia , Femenino , Inflamasomas/metabolismo , Síndrome del Colon Irritable/fisiopatología , Síndrome del Colon Irritable/psicología , Metagenoma/fisiología , Ratones , Ratones Endogámicos C57BL , PPAR gamma/agonistas , Probióticos/uso terapéutico , Estrés Psicológico/complicaciones
17.
Development ; 138(20): 4423-32, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21880782

RESUMEN

The cellular mechanisms that drive growth and remodeling of the early intestinal epithelium are poorly understood. Current dogma suggests that the murine fetal intestinal epithelium is stratified, that villi are formed by an epithelial remodeling process involving the de novo formation of apical surface at secondary lumina, and that radial intercalation of the stratified cells constitutes a major intestinal lengthening mechanism. Here, we investigate cell polarity, cell cycle dynamics and cell shape in the fetal murine intestine between E12.5 and E14.5. We show that, contrary to previous assumptions, this epithelium is pseudostratified. Furthermore, epithelial nuclei exhibit interkinetic nuclear migration, a process wherein nuclei move in concert with the cell cycle, from the basal side (where DNA is synthesized) to the apical surface (where mitosis takes place); such nuclear movements were previously misinterpreted as the radial intercalation of cells. We further demonstrate that growth of epithelial girth between E12.5 and E14.5 is driven by microtubule- and actinomyosin-dependent apicobasal elongation, rather than by progressive epithelial stratification as was previously thought. Finally, we show that the actin-binding protein Shroom3 is crucial for the maintenance of the single-layered pseudostratified epithelium. In mice lacking Shroom3, the epithelium is disorganized and temporarily stratified during villus emergence. These results favor an alternative model of intestinal morphogenesis in which the epithelium remains single layered and apicobasally polarized throughout early intestinal development.


Asunto(s)
Mucosa Intestinal/embriología , Animales , Ciclo Celular , Polaridad Celular , Forma de la Célula , Femenino , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Morfogénesis , Embarazo
18.
Dev Biol ; 361(1): 39-56, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22024319

RESUMEN

Although canonical Wnt signaling is known to regulate taste papilla induction and numbers, roles for noncanonical Wnt pathways in tongue and taste papilla development have not been explored. With mutant mice and whole tongue organ cultures we demonstrate that Wnt5a protein and message are within anterior tongue mesenchyme across embryo stages from the initiation of tongue formation, through papilla placode appearance and taste papilla development. The Wnt5a mutant tongue is severely shortened, with an ankyloglossia, and lingual mesenchyme is disorganized. However, fungiform papilla morphology, number and innervation are preserved, as is expression of the papilla marker, Shh. These data demonstrate that the genetic regulation for tongue size and shape can be separated from that directing lingual papilla development. Preserved number of papillae in a shortened tongue results in an increased density of fungiform papillae in the mutant tongues. In tongue organ cultures, exogenous Wnt5a profoundly suppresses papilla formation and simultaneously decreases canonical Wnt signaling as measured by the TOPGAL reporter. These findings suggest that Wnt5a antagonizes canonical Wnt signaling to dictate papilla number and spacing. In all, distinctive roles for Wnt5a in tongue size, fungiform papilla patterning and development are shown and a necessary balance between non-canonical and canonical Wnt paths in regulating tongue growth and fungiform papillae is proposed in a model, through the Ror2 receptor.


Asunto(s)
Mesodermo/metabolismo , Transducción de Señal/fisiología , Papilas Gustativas/embriología , Lengua/embriología , Proteínas Wnt/metabolismo , Animales , Western Blotting , Bromodesoxiuridina , Femenino , Galactósidos , Proteínas Hedgehog/metabolismo , Inmunohistoquímica , Hibridación in Situ , Indoles , Mesodermo/embriología , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Ratas , Ratas Sprague-Dawley , Papilas Gustativas/metabolismo , Proteína Wnt-5a
19.
Am J Pathol ; 181(6): 2114-25, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23036899

RESUMEN

Gastric adenocarcinoma is one of the leading causes of cancer mortality worldwide. It arises through a stepwise process that includes prominent inflammation with expression of interferon-γ (IFN-γ) and multiple other pro-inflammatory cytokines. We engineered mice expressing IFN-γ under the control of the stomach-specific H(+)/K(+) ATPase ß promoter to test the potential role of this cytokine in gastric tumorigenesis. Stomachs of H/K-IFN-γ transgenic mice exhibited inflammation, expansion of myofibroblasts, loss of parietal and chief cells, spasmolytic polypeptide expressing metaplasia, and dysplasia. Proliferation was elevated in undifferentiated and metaplastic epithelial cells in H/K-IFN-γ transgenic mice, and there was increased apoptosis. H/K-IFN-γ mice had elevated levels of mRNA for IFN-γ target genes and the pro-inflammatory cytokines IL-6, IL-1ß, and tumor necrosis factor-α. Intracellular mediators of IFN-γ and IL-6 signaling, pSTAT1 and pSTAT3, respectively, were detected in multiple cell types within stomach. H/K-IFN-γ mice developed dysplasia as early as 3 months of age, and 4 of 39 mice over 1 year of age developed antral polyps or tumors, including one adenoma and one adenocarcinoma, which expressed high levels of nuclear ß-catenin. Our data identified IFN-γ as a pivotal secreted factor that orchestrates complex changes in inflammatory, epithelial, and mesenchymal cell populations to drive pre-neoplastic progression in stomach; however, additional alterations appear to be required for malignant conversion.


Asunto(s)
Mucosa Gástrica/metabolismo , Inflamación/patología , Interferón gamma/genética , Estómago/patología , Animales , Apoptosis/genética , Atrofia , Linaje de la Célula/genética , Proliferación Celular , Progresión de la Enfermedad , Femenino , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Proteínas Hedgehog/metabolismo , Inflamación/genética , Péptidos y Proteínas de Señalización Intercelular , Interferón gamma/metabolismo , Masculino , Metaplasia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamaño de los Órganos , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/patología , Péptidos/metabolismo , Lesiones Precancerosas/patología , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Regulación hacia Arriba/genética
20.
Nature ; 446(7135): 557-61, 2007 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-17361131

RESUMEN

Deregulation of intestinal immune responses seems to have a principal function in the pathogenesis of inflammatory bowel disease. The gut epithelium is critically involved in the maintenance of intestinal immune homeostasis-acting as a physical barrier separating luminal bacteria and immune cells, and also expressing antimicrobial peptides. However, the molecular mechanisms that control this function of gut epithelial cells are poorly understood. Here we show that the transcription factor NF-kappaB, a master regulator of pro-inflammatory responses, functions in gut epithelial cells to control epithelial integrity and the interaction between the mucosal immune system and gut microflora. Intestinal epithelial-cell-specific inhibition of NF-kappaB through conditional ablation of NEMO (also called IkappaB kinase-gamma (IKKgamma)) or both IKK1 (IKKalpha) and IKK2 (IKKbeta)-IKK subunits essential for NF-kappaB activation-spontaneously caused severe chronic intestinal inflammation in mice. NF-kappaB deficiency led to apoptosis of colonic epithelial cells, impaired expression of antimicrobial peptides and translocation of bacteria into the mucosa. Concurrently, this epithelial defect triggered a chronic inflammatory response in the colon, initially dominated by innate immune cells but later also involving T lymphocytes. Deficiency of the gene encoding the adaptor protein MyD88 prevented the development of intestinal inflammation, demonstrating that Toll-like receptor activation by intestinal bacteria is essential for disease pathogenesis in this mouse model. Furthermore, NEMO deficiency sensitized epithelial cells to tumour-necrosis factor (TNF)-induced apoptosis, whereas TNF receptor-1 inactivation inhibited intestinal inflammation, demonstrating that TNF receptor-1 signalling is crucial for disease induction. These findings demonstrate that a primary NF-kappaB signalling defect in intestinal epithelial cells disrupts immune homeostasis in the gastrointestinal tract, causing an inflammatory-bowel-disease-like phenotype. Our results identify NF-kappaB signalling in the gut epithelium as a critical regulator of epithelial integrity and intestinal immune homeostasis, and have important implications for understanding the mechanisms controlling the pathogenesis of human inflammatory bowel disease.


Asunto(s)
Colitis/inmunología , Colitis/patología , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Quinasa I-kappa B/metabolismo , Inmunidad Innata/inmunología , Animales , Apoptosis/efectos de los fármacos , Enfermedad Crónica , Colitis/enzimología , Colon/inmunología , Colon/patología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Homeostasis , Quinasa I-kappa B/deficiencia , Intestinos/enzimología , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Factores de Necrosis Tumoral/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA