Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Front Mol Neurosci ; 14: 757646, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35002618

RESUMEN

TALPID3/KIAA0586 is an evolutionary conserved protein, which plays an essential role in protein trafficking. Its role during gastrointestinal (GI) and enteric nervous system (ENS) development has not been studied previously. Here, we analyzed chicken, mouse and human embryonic GI tissues with TALPID3 mutations. The GI tract of TALPID3 chicken embryos was shortened and malformed. Histologically, the gut smooth muscle was mispatterned and enteric neural crest cells were scattered throughout the gut wall. Analysis of the Hedgehog pathway and gut extracellular matrix provided causative reasons for these defects. Interestingly, chicken intra-species grafting experiments and a conditional knockout mouse model showed that ENS formation did not require TALPID3, but was dependent on correct environmental cues. Surprisingly, the lack of TALPID3 in enteric neural crest cells (ENCC) affected smooth muscle and epithelial development in a non-cell-autonomous manner. Analysis of human gut fetal tissues with a KIAA0586 mutation showed strikingly similar findings compared to the animal models demonstrating conservation of TALPID3 and its necessary role in human GI tract development and patterning.

3.
J Physiol ; 596(17): 4237-4251, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29917237

RESUMEN

KEY POINTS: Tenascin-X (TNX) is an extracellular matrix glycoprotein with anti-adhesive properties in skin and joints. Here we report the novel finding that TNX is expressed in human and mouse gut tissue where it is exclusive to specific subpopulations of neurones. Our studies with TNX-deficient mice show impaired defecation and neural control of distal colonic motility that can be rescued with a 5-HT4 receptor agonist. However, colonic secretion is unchanged. They are also susceptible to internal rectal intussusception. Colonic afferent sensitivity is increased in TNX-deficient mice. Correspondingly, there is increased density of and sensitivity of putative nociceptive fibres in TNX-deficient mucosa. A group of TNX-deficient patients report symptoms highly consistent with those in the mouse model. These findings suggest TNX plays entirely different roles in gut to non-visceral tissues - firstly a role in enteric motor neurones and secondly a role influencing nociceptive sensory neurones Studying further the mechanisms by which TNX influences neuronal function will lead to new targets for future treatment. ABSTRACT: The extracellular matrix (ECM) is not only an integral structural molecule, but is also critical for a wide range of cellular functions. The glycoprotein tenascin-X (TNX) predominates in the ECM of tissues like skin and regulates tissue structure through anti-adhesive interactions with collagen. Monogenic TNX deficiency causes painful joint hypermobility and skin hyperelasticity, symptoms characteristic of hypermobility Ehlers Danlos syndrome (hEDS). hEDS patients also report consistently increased visceral pain and gastrointestinal (GI) dysfunction. We investigated whether there is a direct link between TNX deficiency and GI pain or motor dysfunction. We set out first to learn where TNX is expressed in human and mouse, then determine how GI function, specifically in the colon, is disordered in TNX-deficient mice and humans of either sex. In human and mouse tissue, TNX was predominantly associated with cholinergic colonic enteric neurones, which are involved in motor control. TNX was absent from extrinsic nociceptive peptidergic neurones. TNX-deficient mice had internal rectal prolapse and a loss of distal colonic contractility which could be rescued by prokinetic drug treatment. TNX-deficient patients reported increased sensory and motor GI symptoms including abdominal pain and constipation compared to controls. Despite absence of TNX from nociceptive colonic neurones, neuronal sprouting and hyper-responsiveness to colonic distension was observed in the TNX-deficient mice. We conclude that ECM molecules are not merely support structures but an integral part of the microenvironment particularly for specific populations of colonic motor neurones where TNX exerts functional influences.


Asunto(s)
Colon/patología , Matriz Extracelular/metabolismo , Enfermedades Gastrointestinales/patología , Neuronas Motoras/patología , Células Receptoras Sensoriales/patología , Tenascina/metabolismo , Animales , Movimiento Celular , Colon/metabolismo , Femenino , Enfermedades Gastrointestinales/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Células Receptoras Sensoriales/metabolismo , Tenascina/genética
4.
BMC Cancer ; 18(1): 622, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859059

RESUMEN

BACKGROUND: Metastasis is a complex process which is difficult to study and model. Experimental ingenuity is therefore essential when seeking to elucidate the biological mechanisms involved. Typically, in vitro models of metastasis have been overly simplistic, lacking the characteristic elements of the tumour microenvironment, whereas in vivo models are expensive, requiring specialist resources. Here we propose a pipeline approach for the study of cell migration and colonization, two critical steps in the metastatic cascade. METHODS: We used a range of extracellular matrix derived contexts to facilitate a progressive approach to the observation and quantification of cell behaviour in 2D, 3D and at border zones between dimensions. At the simplest level, cells were set onto collagen-coated plastic or encapsulated within a collagen matrix. To enhance this, a collagen compression technique provided a stiffened, denser substrate which could be used as a 2D surface or to encapsulate cells. Decellularized tissue from the chorioallantoic membrane of the developing chicken embryo was used to provide a more structured, biologically relevant extracellular matrix-based context in which cell behaviour could then be compared with its in vivo counterpart. RESULTS: Cell behaviour could be observed and quantified within each context using standard laboratory techniques of microscopy and immunostaining, affording the opportunity for comparison and contrast of behaviour across the whole range of contexts. In particular, the temporal constraints of the in vivo CAM were removed when cells were cultured on the decellularized CAM, allowing for much longer-term cell colonization and cell-cell interaction. CONCLUSIONS: Together the assays within this pipeline provide the opportunity for the study of cell behaviour in a replicable way across multiple environments. The assays can be set up and analysed using easily available resources and standard laboratory equipment. We believe this offers the potential for the detailed study of cell migration and colonization of tissue, essential steps in the metastatic cascade. Also, we propose that the pipeline could be used in the wider arena of cell culture in general with the increasingly more complex contexts allowing cell behaviours and interactions to be explored in a stepwise fashion in an integrated way.


Asunto(s)
Técnicas de Cultivo de Célula , Ensayos de Migración Celular/métodos , Invasividad Neoplásica/patología , Neoplasias/patología , Animales , Línea Celular Tumoral , Embrión de Pollo , Pollos , Membrana Corioalantoides , Colágeno , Matriz Extracelular , Humanos
5.
PLoS One ; 11(1): e0147989, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26824433

RESUMEN

OBJECTIVES: Enteric neuropathies are severe gastrointestinal disorders with unsatisfactory outcomes. We aimed to investigate the potential of enteric neural stem cell therapy approaches for such disorders by transplanting mouse enteric neural crest cells (ENCCs) into ganglionic and aganglionic mouse gut in vivo and analysing functional integration and long-term safety. DESIGN: Neurospheres generated from yellow fluorescent protein (YFP) expressing ENCCs selected from postnatal Wnt1-cre;R26R-YFP/YFP murine gut were transplanted into ganglionic hindgut of wild-type littermates or aganglionic hindgut of Ednrbtm1Ywa mice (lacking functional endothelin receptor type-B). Intestines were then assessed for ENCC integration and differentiation using immunohistochemistry, cell function using calcium imaging, and long-term safety using PCR to detect off-target YFP expression. RESULTS: YFP+ ENCCs engrafted, proliferated and differentiated into enteric neurons and glia within recipient ganglionic gut. Transplanted cells and their projections spread along the endogenous myenteric plexus to form branching networks. Electrical point stimulation of endogenous nerve fibres resulted in calcium transients (F/F0 = 1.16 ± 0.01;43 cells, n = 6) in YFP+ transplanted ENCCs (abolished with TTX). Long-term follow-up (24 months) showed transplanted ENCCs did not give rise to tumours or spread to other organs (PCR negative in extraintestinal sites). In aganglionic gut ENCCs similarly spread and differentiated to form neuronal and glial networks with projections closely associated with endogenous neural networks of the transition zone. CONCLUSIONS: Transplanted ENCCs successfully engrafted into recipient ganglionic and aganglionic gut showing appropriate spread, localisation and, importantly, functional integration without any long-term safety issues. This study provides key support for the development and use of enteric neural stem cell therapies.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Intestinos/citología , Cresta Neural/citología , Células-Madre Neurales/trasplante , Neuroglía/citología , Neuronas/citología , Animales , Animales Recién Nacidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biomarcadores/metabolismo , Calcio/metabolismo , Diferenciación Celular , Ingeniería Celular , Estimulación Eléctrica , Expresión Génica , Supervivencia de Injerto , Mucosa Intestinal/metabolismo , Intestinos/inervación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Fibras Nerviosas/metabolismo , Cresta Neural/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Receptor de Endotelina B/genética , Receptor de Endotelina B/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Transfección , Transgenes
6.
J Vis Exp ; (99): e52514, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-26065540

RESUMEN

All developing organs need to be connected to both the nervous system (for sensory and motor control) as well as the vascular system (for gas exchange, fluid and nutrient supply). Consequently both the nervous and vascular systems develop alongside each other and share striking similarities in their branching architecture. Here we report embryonic manipulations that allow us to study the simultaneous development of neural crest-derived nervous tissue (in this case the enteric nervous system), and the vascular system. This is achieved by generating chicken chimeras via transplantation of discrete segments of the neural tube, and associated neural crest, combined with vascular DiI injection in the same embryo. Our method uses transgenic chick(GFP) embryos for intraspecies grafting, making the transplant technique more powerful than the classical quail-chick interspecies grafting protocol used with great effect since the 1970s. Chick(GFP)-chick intraspecies grafting facilitates imaging of transplanted cells and their projections in intact tissues, and eliminates any potential bias in cell development linked to species differences. This method takes full advantage of the ease of access of the avian embryo (compared with other vertebrate embryos) to study the co-development of the enteric nervous system and the vascular system.


Asunto(s)
Vasos Sanguíneos/química , Carbocianinas/química , Colorantes Fluorescentes/química , Cresta Neural/citología , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/embriología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Embrión de Pollo , Pollos , Quimera , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Sistema Nervioso/química , Sistema Nervioso/embriología , Cresta Neural/química , Cresta Neural/embriología , Codorniz
7.
PLoS One ; 10(3): e0119467, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25799576

RESUMEN

OBJECTIVES: Enteric neural stem cells provide hope of curative treatment for enteric neuropathies. Current protocols for their harvesting from humans focus on the generation of 'neurospheres' from cultures of dissociated gut tissue. The study aims to better understand the derivation, generation and composition of enteric neurospheres. DESIGN: Gut tissue was obtained from Wnt1-Cre;Rosa26Yfp/Yfp transgenic mice (constitutively labeled neural crest cells) and paediatric patients. Gut cells were cultured either unsorted (mixed neural crest/non-neural crest), or following FACS selection into neural crest (murine-YFP+ve/human-p75+ve) or non-neural crest (YFP-ve/p75-ve) populations. Cultures and resultant neurospheres were characterized using immunolabelling in vitro and following transplantation in vivo. RESULTS: Cultures of (i) unsorted, (ii) neural crest, and (iii) non-neural crest cell populations generated neurospheres similar in numbers, size and morphology. Unsorted neurospheres were highly heterogeneous for neural crest content. Neural crest-derived (YFP+ve/p75+ve) neurospheres contained only neural derivatives (neurons and glia) and were devoid of non-neural cells (i.e. negative for SMA, c-Kit), with the converse true for non-neural crest-derived (YFP-ve/p75-ve) 'neurospheres'. Under differentiation conditions only YFP+ve cells gave rise to neural derivatives. Both YFP+ve and YFP-ve cells displayed proliferation and spread upon transplantation in vivo, but YFP-ve cells did not locate or integrate within the host ENS. CONCLUSIONS: Spherical accumulations of cells, so-called 'neurospheres' forming in cultures of dissociated gut contain variable proportions of neural crest-derived cells. If they are to be used for ENS cell replacement therapy then improved protocols for their generation, including cell selection, should be sought in order to avoid inadvertent transplantation of non-therapeutic, non-ENS cells.


Asunto(s)
Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Sistema Nervioso Entérico/citología , Tracto Gastrointestinal/citología , Cresta Neural/citología , Células-Madre Neurales/citología , Animales , Proteínas Bacterianas/metabolismo , Células Cultivadas , Sistema Nervioso Entérico/metabolismo , Femenino , Tracto Gastrointestinal/metabolismo , Humanos , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Proteína Wnt1/fisiología
8.
Arterioscler Thromb Vasc Biol ; 35(4): 845-54, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25767277

RESUMEN

OBJECTIVE: The antitumor effects of FK506-binding protein like (FKBPL) and its extracellular role in angiogenesis are well characterized; however, its role in physiological/developmental angiogenesis and the effect of FKBPL ablation has not been evaluated. This is important as effects of some angiogenic proteins are dosage dependent. Here we evaluate the regulation of FKBPL secretion under angiogenic stimuli, as well as the effect of FKBPL ablation in angiogenesis using mouse and zebrafish models. APPROACH AND RESULTS: FKBPL is secreted maximally by human microvascular endothelial cells and fibroblasts, and this was specifically downregulated by proangiogenic hypoxic signals, but not by the angiogenic cytokines, VEGF or IL8. FKBPL's critical role in angiogenesis was supported by our inability to generate an Fkbpl knockout mouse, with embryonic lethality occurring before E8.5. However, whilst Fkbpl heterozygotic embryos showed some vasculature irregularities, the mice developed normally. In murine angiogenesis models, including the ex vivo aortic ring assay, in vivo sponge assay, and tumor growth assay, Fkbpl(+/-) mice exhibited increased sprouting, enhanced vessel recruitment, and faster tumor growth, respectively, supporting the antiangiogenic function of FKBPL. In zebrafish, knockdown of zFkbpl using morpholinos disrupted the vasculature, and the phenotype was rescued with hFKBPL. Interestingly, this vessel disruption was ineffective when zcd44 was knocked-down, supporting the dependency of zFkbpl on zCd44 in zebrafish. CONCLUSIONS: FKBPL is an important regulator of angiogenesis, having an essential role in murine and zebrafish blood vessel development. Mouse models of angiogenesis demonstrated a proangiogenic phenotype in Fkbpl heterozygotes.


Asunto(s)
Aorta/metabolismo , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Inmunofilinas/metabolismo , Neovascularización Patológica , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Carcinoma Pulmonar de Lewis/patología , Hipoxia de la Célula , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Inmunofilinas/genética , Células MCF-7 , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica , Fenotipo , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Factores de Tiempo , Carga Tumoral , Pez Cebra , Proteínas de Pez Cebra/genética
9.
Dev Biol ; 385(2): 220-9, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24262984

RESUMEN

The vasculature and nervous system share striking similarities in their networked, tree-like architecture and in the way they are super-imposed in mature organs. It has previously been suggested that the intestinal microvasculature network directs the migration of enteric neural crest cells (ENCC) along the gut to promote the formation of the enteric nervous system (ENS). To investigate the inter-relationship of migrating ENCC, ENS formation and gut vascular development we combined fate-mapping of ENCC with immunolabelling and intravascular dye injection to visualise nascent blood vessel networks. We found that the enteric and vascular networks initially had very distinct patterns of development. In the foregut, ENCC migrated through areas devoid of established vascular networks. In vessel-rich areas, such as the midgut and hindgut, the distribution of migrating ENCC did not support the idea that these cells followed a pre-established vascular network. Moreover, when gut vascular development was impaired, either genetically in Vegfa(120/120) or Tie2-Cre;Nrp1(fl/-) mice or using an in vitro Wnt1-Cre;Rosa26(Yfp/+) mouse model of ENS development, ENCC still colonised the entire length of the gut, including the terminal hindgut. These results demonstrate that blood vessel networks are not necessary to guide migrating ENCC during ENS development. Conversely, in miRet(51) mice, which lack ENS in the hindgut, the vascular network in this region appeared to be normal suggesting that in early development both networks form independently of each other.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Intestinos/citología , Neovascularización Fisiológica , Cresta Neural/citología , Animales , Intestinos/irrigación sanguínea , Ratones
10.
Biomaterials ; 34(28): 6638-48, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23727263

RESUMEN

Tissue engineering of autologous lung tissue aims to become a therapeutic alternative to transplantation. Efforts published so far in creating scaffolds have used harsh decellularization techniques that damage the extracellular matrix (ECM), deplete its components and take up to 5 weeks to perform. The aim of this study was to create a lung natural acellular scaffold using a method that will reduce the time of production and better preserve scaffold architecture and ECM components. Decellularization of rat lungs via the intratracheal route removed most of the nuclear material when compared to the other entry points. An intermittent inflation approach that mimics lung respiration yielded an acellular scaffold in a shorter time with an improved preservation of pulmonary micro-architecture. Electron microscopy demonstrated the maintenance of an intact alveolar network, with no evidence of collapse or tearing. Pulsatile dye injection via the vasculature indicated an intact capillary network in the scaffold. Morphometry analysis demonstrated a significant increase in alveolar fractional volume, with alveolar size analysis confirming that alveolar dimensions were maintained. Biomechanical testing of the scaffolds indicated an increase in resistance and elastance when compared to fresh lungs. Staining and quantification for ECM components showed a presence of collagen, elastin, GAG and laminin. The intratracheal intermittent decellularization methodology could be translated to sheep lungs, demonstrating a preservation of ECM components, alveolar and vascular architecture. Decellularization treatment and methodology preserves lung architecture and ECM whilst reducing the production time to 3 h. Cell seeding and in vivo experiments are necessary to proceed towards clinical translation.


Asunto(s)
Matriz Extracelular/química , Pulmón/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Embrión de Pollo , Membrana Corioalantoides/química , Membrana Corioalantoides/citología , Pulmón/citología , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Neovascularización Fisiológica/fisiología , Ratas
11.
Int J Dev Biol ; 56(4): 245-54, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22562200

RESUMEN

Neural crest cells (NCC) are multipotent progenitors that migrate extensively throughout the developing embryo and generate a diverse range of cell types. Vagal NCC migrate from the hindbrain into the foregut and from there along the gastrointestinal tract to form the enteric nervous system (ENS), the intrinsic innervation of the gut, and into the developing lung buds to form the intrinsic innervation of the lungs. The aim of this study was to determine the developmental potential of vagal NCC that had already colonised the gut or the lungs. We used transgenic chicken embryos that ubiquitously express green fluorescent protein (GFP) to permanently mark and fate-map vagal NCC using intraspecies grafting. This was combined with back-transplantation of gut and lung segments, containing GFP-positive NCC, into the vagal region of a second recipient embryo to determine, using immunohistochemical staining, whether gut or lung NCC are competent of re-colonising both these organs, or whether their fate is restricted. Chick(GFP)-chick intraspecies grafting efficiently labelled NCC within the gut and lung of chick embryos. When segments of embryonic day (E)5.5 pre-umbilical midgut containing GFP-positive NCC were back-transplanted into the vagal region of E1.5 host embryos, the GFP-positive NCC remigrated to colonise both the gut and lungs and differentiated into neurons in stereotypical locations. However, GFP-positive lung NCC did not remigrate when back-transplanted. Our studies suggest that gut NCC are not restricted to colonising only this organ, since upon back-transplantation GFP-positive gut NCC colonised both the gut and the lung.


Asunto(s)
Movimiento Celular , Trasplante de Células/métodos , Proteínas Fluorescentes Verdes/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Animales , Embrión de Pollo , Pollos , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/metabolismo , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/inervación , Proteínas Fluorescentes Verdes/genética , Pulmón/citología , Pulmón/embriología , Pulmón/metabolismo , Microscopía Confocal , Cresta Neural/embriología , Factores de Tiempo , Nervio Vago/citología , Nervio Vago/embriología , Nervio Vago/metabolismo
12.
Mol Biol Cell ; 22(21): 3962-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21900494

RESUMEN

Despite high rates of cell death, epithelia maintain intact barriers by squeezing dying cells out using a process termed cell extrusion. Cells can extrude apically into the lumen or basally into the tissue the epithelium encases, depending on whether actin and myosin contract at the cell base or apex, respectively. We previously found that microtubules in cells surrounding a dying cell target p115 RhoGEF to the actin cortex to control where contraction occurs. However, what controls microtubule targeting to the cortex and whether the dying cell also controls the extrusion direction were unclear. Here we find that the tumor suppressor adenomatous polyposis coli (APC) controls microtubule targeting to the cell base to drive apical extrusion. Whereas wild-type cells preferentially extrude apically, cells lacking APC or expressing an oncogenic APC mutation extrude predominantly basally in cultured monolayers and zebrafish epidermis. Thus APC is essential for driving extrusion apically. Surprisingly, although APC controls microtubule reorientation and attachment to the actin cortex in cells surrounding the dying cell, it does so by controlling actin and microtubules within the dying cell. APC disruptions that are common in colon and breast cancer may promote basal extrusion of tumor cells, which could enable their exit and subsequent migration.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Células Epiteliales/fisiología , Mucosa Respiratoria/citología , Actinas/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Muerte Celular , Línea Celular , Células Epidérmicas , Epidermis/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Microscopía Confocal , Microtúbulos/metabolismo , Proteínas Oncogénicas/metabolismo , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/fisiología , Análisis de la Célula Individual , Tubulina (Proteína)/metabolismo , Pez Cebra
13.
Hum Mol Genet ; 19(18): 3642-51, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20621975

RESUMEN

Goldberg-Shprintzen syndrome (GOSHS) is a rare clinical disorder characterized by central and enteric nervous system defects. This syndrome is caused by inactivating mutations in the Kinesin Binding Protein (KBP) gene, which encodes a protein of which the precise function is largely unclear. We show that KBP expression is up-regulated during neuronal development in mouse cortical neurons. Moreover, KBP-depleted PC12 cells were defective in nerve growth factor-induced differentiation and neurite outgrowth, suggesting that KBP is required for cell differentiation and neurite development. To identify KBP interacting proteins, we performed a yeast two-hybrid screen and found that KBP binds almost exclusively to microtubule associated or related proteins, specifically SCG10 and several kinesins. We confirmed these results by validating KBP interaction with one of these proteins: SCG10, a microtubule destabilizing protein. Zebrafish studies further demonstrated an epistatic interaction between KBP and SCG10 in vivo. To investigate the possibility of direct interaction between KBP and microtubules, we undertook co-localization and in vitro binding assays, but found no evidence of direct binding. Thus, our data indicate that KBP is involved in neuronal differentiation and that the central and enteric nervous system defects seen in GOSHS are likely caused by microtubule-related defects.


Asunto(s)
Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Microtúbulos/metabolismo , Neurogénesis , Serpinas/metabolismo , Estatmina/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/genética , Línea Celular , Células Cultivadas , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Modelos Animales de Enfermedad , Células HeLa , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Microtúbulos , Células 3T3 NIH , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Unión Proteica , Ratas , Serpinas/genética , Estatmina/genética , Proteínas de Pez Cebra/genética
14.
Gene Expr Patterns ; 9(4): 231-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19272335

RESUMEN

SCG10 (Superior Cervical Ganglia 10, STMN2) is a member of the stathmin family of proteins. Stathmins regulate microtubule dynamics by inhibiting polymerization and promoting their depolymerization. SCG10 is believed to be a neuronal-specific stathmin that is enriched in the growth cones of developing neurons and plays a role in regulating neurite outgrowth. In all species examined so far, SCG10 is expressed in both the CNS and PNS. We have cloned two zebrafish SCG10 homologues and have determined the temporal and spatial expression pattern of both of these genes by RT-PCR and in situ hybridization. RT-PCR shows that both transcripts are expressed maternally and zygotically through at least 5 days. In situ hybridization analysis reveals that both SCG10 orthologues have dynamic, spatial expression patterns that are nearly identical to each other. Initially, these orthologues are expressed in discrete areas of the forebrain, midbrain, and hindbrain, as well as in the anterior and posterior lateral line ganglia and transiently in the spinal cord Rohon-Beard neurons. From 48hpf onwards, the level of expression of both genes increases and becomes mainly restricted to the anterior CNS (the forebrain region, retina, optic tectum, and hindbrain), and to the cranial ganglia. From 72 to 96hpf, SCG10 genes are also expressed in the developing neurons in the gut and in the surrounding intestinal mesenchyme. Our results provide a starting point for future studies that will investigate the in vivo function of SCG10 orthologues in zebrafish neural development.


Asunto(s)
Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Estatmina/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Femenino , Hibridación in Situ , Mucosa Intestinal/metabolismo , Intestinos/embriología , Masculino , Mesodermo/metabolismo , Datos de Secuencia Molecular , Filogenia , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Tiempo , Pez Cebra/embriología , Proteínas de Pez Cebra/clasificación
15.
Development ; 135(23): 3891-901, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18948415

RESUMEN

In zebrafish, BMP signaling establishes cell identity along the dorsoventral (DV) axis during gastrulation. Owing to the early requirements of BMP activity in DV patterning, it has been difficult to assign later roles in cell fate specification to specific BMP ligands. In this study, we have taken advantage of two follistatin-like genes (fstl1 and fstl2), as well as a transgenic zebrafish line carrying an inducible truncated form of the BMP-type 1 receptor to study the role of Bmp4 outside of the context of DV specification. Characterization of fstl1/2 suggests that they exert a redundant role as BMP antagonists during late gastrulation, regulating BMP activity in axial mesoderm. Maintenance of appropriate levels of BMP signaling is crucial for the proper development of chordamesoderm, a subset of axial mesoderm that gives rise to the notochord, but not prechordal mesoderm, which gives rise to the prechordal plate. Bmp4 activity in particular is required during a crucial window beginning at late gastrulation and lasting through early somitogenesis to promote chordamesoderm proliferation. In the absence of Bmp4, the notochord precursor pool is depleted, and the notochord differentiates prematurely. Our results illustrate a role for Bmp4 in the proliferation and timely differentiation of axial tissue after DV axis specification.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Notocorda/citología , Notocorda/embriología , Cola (estructura animal)/embriología , Pez Cebra/embriología , Animales , Tipificación del Cuerpo/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Gastrulación/efectos de los fármacos , Ligandos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Notocorda/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
16.
Dev Biol ; 318(1): 52-64, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18436202

RESUMEN

The zebrafish enteric nervous system (ENS), like those of all other vertebrate species, is principally derived from the vagal neural crest cells (NCC). The developmental controls that govern the migration, proliferation and patterning of the ENS precursors are not well understood. We have investigated the roles of endoderm and Sonic hedgehog (SHH) in the development of the ENS. We show that endoderm is required for the migration of ENS NCC from the vagal region to the anterior end of the intestine. We show that the expression of shh and its receptor ptc-1 correlate with the development of the ENS and demonstrate that hedgehog (HH) signaling is required in two phases, a pre-enteric and an enteric phase, for normal ENS development. We show that HH signaling regulates the proliferation of vagal NCC and ENS precursors in vivo. We also show the zebrafish hand2 is required for the normal development of the intestinal smooth muscle and the ENS. Furthermore we show that endoderm and HH signaling, but not hand2, regulate gdnf expression in the intestine, highlighting a central role of endoderm and SHH in patterning the intestine and the ENS.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Endodermo/metabolismo , Sistema Nervioso Entérico/embriología , Proteínas Hedgehog/metabolismo , Mesodermo/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Tipificación del Cuerpo , Movimiento Celular/fisiología , Endodermo/citología , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Hedgehog/genética , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Proteínas de la Membrana , Mesodermo/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores Patched , Receptor Patched-1 , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción SOX , Transducción de Señal/fisiología , Células Madre/citología , Células Madre/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Alcaloides de Veratrum/metabolismo , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
17.
Dev Dyn ; 237(4): 1060-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18351671

RESUMEN

Smad-interacting protein-1 (SIP1) has been implicated in the development of Mowat-Wilson syndrome whose patients exhibit Hirschsprung disease, an aganglionosis of the large intestine, as well as other phenotypes. We have identified and cloned two sip1 orthologues in zebrafish. Both sip1 orthologues are expressed maternally and have dynamic zygotic expression patterns that are temporally and spatially distinct. We have investigated the function of both orthologues using translation and splice-blocking morpholino antisense oligonucleotides. Knockdown of the orthologues causes axial and neural patterning defects consistent with the previously described function of SIP1 as an inhibitor of BMP signaling. In addition, knockdown of both genes leads to a significant reduction/loss of the post-otic cranial neural crest. This results in a subsequent absence of neural crest precursors in the posterior pharyngeal arches and a loss of enteric precursors in the intestine.


Asunto(s)
Tipificación del Cuerpo , Proteínas Portadoras/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Morfogénesis , Cresta Neural/citología , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , Isoformas de Proteínas/genética , Células Madre/fisiología , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
18.
Dev Biol ; 313(1): 279-92, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18031721

RESUMEN

The enteric nervous system (ENS) is formed from vagal and sacral neural crest cells (NCC). Vagal NCC give rise to most of the ENS along the entire gut, whereas the contribution of sacral NCC is mainly limited to the hindgut. This, and data from heterotopic quail-chick grafting studies, suggests that vagal and sacral NCC have intrinsic differences in their ability to colonize the gut, and/or to respond to signalling cues within the gut environment. To better understand the molecular basis of these differences, we studied the expression of genes known to be essential for ENS formation, in sacral NCC within the chick hindgut. Our results demonstrate that, as in vagal NCC, Sox10, EdnrB, and Ret are expressed in sacral NCC within the gut. Since we did not detect a qualitative difference in expression of these ENS genes we performed DNA microarray analysis of vagal and sacral NCC. Of 11 key ENS genes examined from the total data set, Ret was the only gene identified as being highly differentially expressed, with a fourfold increase in expression in vagal versus sacral NCC. We also found that over-expression of RET in sacral NCC increased their ENS developmental potential such that larger numbers of cells entered the gut earlier in development, thus promoting the fate of sacral NCC towards that of vagal NCC.


Asunto(s)
Movimiento Celular , Sistema Nervioso Entérico/embriología , Cresta Neural/citología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Embrión de Pollo , Proteínas de Unión al ADN/metabolismo , Sistema Digestivo/embriología , Sistema Digestivo/inervación , Sistema Digestivo/metabolismo , Embrión no Mamífero/metabolismo , Sistema Nervioso Entérico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/metabolismo , Cresta Neural/trasplante , Análisis de Secuencia por Matrices de Oligonucleótidos , Codorniz , Factores de Transcripción SOXE , Sacro/citología , Factores de Transcripción/metabolismo , Trasplante Heterólogo
19.
Development ; 133(3): 395-406, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16396911

RESUMEN

The zebrafish enteric nervous system (ENS), like those of all other vertebrate species, is principally derived from the vagal neural crest. The developmental controls that govern the specification and patterning of the ENS are not well understood. To identify genes required for the formation of the vertebrate ENS, we preformed a genetic screen in zebrafish. We isolated the lessen (lsn) mutation that has a significant reduction in the number of ENS neurons as well as defects in other cranial neural crest derived structures. We show that the lsn gene encodes a zebrafish orthologue of Trap100, one of the subunits of the TRAP/mediator transcriptional regulation complex. A point mutation in trap100 causes a premature stop codon that truncates the protein, causing a loss of function. Antisense-mediated knockdown of trap100 causes an identical phenotype to lsn. During development trap100 is expressed in a dynamic tissue-specific expression pattern consistent with its function in ENS and jaw cartilage development. Analysis of neural crest markers revealed that the initial specification and migration of the neural crest is unaffected in lsn mutants. Phosphohistone H3 immunocytochemistry revealed that there is a significant reduction in proliferation of ENS precursors in lsn mutants. Using cell transplantation studies, we demonstrate that lsn/trap100 acts cell autonomously in the pharyngeal mesendoderm and influences the development of neural crest derived cartilages secondarily. Furthermore, we show that endoderm is essential for ENS development. These studies demonstrate that lsn/trap100 is not required for initial steps of cranial neural crest development and migration, but is essential for later proliferation of ENS precursors in the intestine.


Asunto(s)
Tipificación del Cuerpo , Sistema Nervioso Entérico/embriología , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Animales , Movimiento Celular , Clonación Molecular , Endodermo/fisiología , Sistema Nervioso Entérico/fisiología , Huesos Faciales/anatomía & histología , Huesos Faciales/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Masculino , Mutación , Cresta Neural/citología , Cresta Neural/fisiología , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , Cráneo/anatomía & histología , Cráneo/embriología , Timo/anatomía & histología , Timo/embriología , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Pez Cebra/genética
20.
Dev Biol ; 277(1): 63-79, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15572140

RESUMEN

The development of intrinsic ganglia, comprised of neurons and glia cells that innervate airway smooth muscle, is a recognized component of the growing lung. However, the embryological origin of these neurons and glia is unclear. The lung buds develop as an outgrowth of the foregut, which contains migrating neural crest cells (NCC) that ultimately give rise to the enteric nervous system (ENS) along the entire length of the gut. It has therefore been proposed that the intrinsic ganglia of the lung arise from a subset of NCC that leave the gut and migrate into the lung buds during early development. We have tested this hypothesis using quail-chick interspecies grafting to selectively label the hindbrain-derived neural crest cell population that colonizes the gut. In conjunction with antibody labeling and in situ hybridization, we demonstrate that: (i) lung ganglia arise from vagal NCC that migrate from the foregut into the lung buds; (ii) like ENS precursors, these NCC express the transcription factor Sox10, and the receptors EDNRB and RET; (iii) the co-receptor for RET, GFRalpha1, is expressed in the lung mesenchyme and in ganglia; (iv) ganglia persist within the lung throughout development and contain cells immunopositive for the pan-neuronal markers ANNA-1 and PGP9.5, the inhibitory neurotransmitter NO, as shown by NADPH-diaphorase staining, and the glial marker GFAP.


Asunto(s)
Ganglios/embriología , Pulmón/embriología , Pulmón/inervación , Cresta Neural/citología , Animales , Diferenciación Celular , Movimiento Celular , Embrión de Pollo , Proteínas de Unión al ADN/análisis , Proteínas del Grupo de Alta Movilidad/análisis , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras/análisis , Factores de Transcripción SOXE , Transducción de Señal , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...