Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37272619

RESUMEN

WDR62 is a spindle pole-associated scaffold protein with pleiotropic functions. Recessive mutations in WDR62 cause structural brain abnormalities and account for the second most common cause of autosomal recessive primary microcephaly (MCPH), indicating WDR62 as a critical hub for human brain development. Here, we investigated WDR62 function in corticogenesis through the analysis of a C-terminal truncating mutation (D955AfsX112). Using induced Pluripotent Stem Cells (iPSCs) obtained from a patient and his unaffected parent, as well as isogenic corrected lines, we generated 2D and 3D models of human neurodevelopment, including neuroepithelial stem cells, cerebro-cortical progenitors, terminally differentiated neurons, and cerebral organoids. We report that WDR62 localizes to the Golgi apparatus during interphase in cultured cells and human fetal brain tissue, and translocates to the mitotic spindle poles in a microtubule-dependent manner. Moreover, we demonstrate that WDR62 dysfunction impairs mitotic progression and results in alterations of the neurogenic trajectories of iPSC neuroderivatives. In summary, impairment of WDR62 localization and function results in severe neurodevelopmental abnormalities, thus delineating new mechanisms in the etiology of MCPH.


Asunto(s)
Proteínas de Ciclo Celular , Aparato de Golgi , Microcefalia , Proteínas del Tejido Nervioso , Polos del Huso , Humanos , Microcefalia/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Ciclo Celular/metabolismo , Masculino , Células Madre Pluripotentes Inducidas , Mitosis , Niño , Adolescente
2.
Cell Rep ; 22(4): 992-1002, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29386140

RESUMEN

Neurogenesis continues in the ventricular-subventricular zone (V-SVZ) of the adult forebrain from quiescent neural stem cells (NSCs). V-SVZ NSCs are a reservoir for new olfactory bulb (OB) neurons that migrate through the rostral migratory stream (RMS). To generate neurons, V-SVZ NSCs need to activate and enter the cell cycle. The mechanisms underlying NSC transition from quiescence to activity are poorly understood. We show that Notch2, but not Notch1, signaling conveys quiescence to V-SVZ NSCs by repressing cell-cycle-related genes and neurogenesis. Loss of Notch2 activates quiescent NSCs, which proliferate and generate new neurons of the OB lineage. Notch2 deficiency results in accelerated V-SVZ NSC exhaustion and an aging-like phenotype. Simultaneous loss of Notch1 and Notch2 resembled the total loss of Rbpj-mediated canonical Notch signaling; thus, Notch2 functions are not compensated in NSCs, and Notch2 is indispensable for the maintenance of NSC quiescence in the adult V-SVZ.


Asunto(s)
Ventrículos Laterales/crecimiento & desarrollo , Células-Madre Neurales/metabolismo , Receptor Notch2/genética , Animales , Diferenciación Celular , Ratones , Transducción de Señal
3.
Sci Rep ; 7: 43708, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272472

RESUMEN

Recessive mutations in WD repeat domain 62 (WDR62) cause microcephaly and a wide spectrum of severe brain malformations. Disruption of the mouse ortholog results in microcephaly underlain by reduced proliferation of neocortical progenitors during late neurogenesis, abnormalities in asymmetric centrosome inheritance leading to neuronal migration delays, and altered neuronal differentiation. Spindle pole localization of WDR62 and mitotic progression are defective in patient-derived fibroblasts, which, similar to mouse neocortical progenitors, transiently arrest at prometaphase. Expression of WDR62 is closely correlated with components of the chromosome passenger complex (CPC), a key regulator of mitosis. Wild type WDR62, but not disease-associated mutant forms, interacts with the CPC core enzyme Aurora kinase B and staining of CPC components at centromeres is altered in patient-derived fibroblasts. Our findings demonstrate critical and diverse functions of WDR62 in neocortical development and provide insight into the mechanisms by which its disruption leads to a plethora of structural abnormalities.


Asunto(s)
Aurora Quinasa B/genética , Centrosoma/metabolismo , Epistasis Genética , Patrón de Herencia , Microcefalia/genética , Proteínas del Tejido Nervioso/genética , Animales , Encéfalo/anomalías , Encéfalo/metabolismo , Encéfalo/patología , Ciclo Celular/genética , Proteínas de Ciclo Celular , Diferenciación Celular/genética , Proliferación Celular , Consanguinidad , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Microcefalia/diagnóstico por imagen , Microcefalia/patología , Mutación , Células-Madre Neurales/metabolismo , Linaje , Secuenciación Completa del Genoma
4.
Gut ; 66(6): 1001-1011, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-26933171

RESUMEN

OBJECTIVE: We tested the ability of Notch pathway receptors Notch1 and Notch2 to regulate stem and epithelial cell homoeostasis in mouse and human gastric antral tissue. DESIGN: Mice were treated with the pan-Notch inhibitor dibenzazepine (DBZ) or inhibitory antibodies targeting Notch1 and/or Notch2. Epithelial proliferation, apoptosis and cellular differentiation were measured by histological and molecular approaches. Organoids were established from mouse and human antral glands; growth and differentiation were measured after treatment with Notch inhibitors. RESULTS: Notch1 and Notch2 are the predominant Notch receptors expressed in mouse and human antral tissue and organoid cultures. Combined inhibition of Notch1 and Notch2 in adult mice led to decreased epithelial cell proliferation, including reduced proliferation of LGR5 stem cells, and increased apoptosis, similar to the response to global Notch inhibition with DBZ. Less pronounced effects were observed after inhibition of individual receptors. Notch pathway inhibition with DBZ or combined inhibition of Notch1 and Notch2 led to increased differentiation of all gastric antral lineages, with remodelling of cells to express secretory products normally associated with other regions of the GI tract, including intestine. Analysis of mouse and human organoids showed that Notch signalling through Notch1 and Notch2 is intrinsic to the epithelium and required for organoid growth. CONCLUSIONS: Notch signalling is required to maintain gastric antral stem cells. Notch1 and Notch2 are the primary Notch receptors regulating epithelial cell homoeostasis in mouse and human stomach.


Asunto(s)
Células Epiteliales/fisiología , Homeostasis , Organoides/crecimiento & desarrollo , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Células Madre/fisiología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Apoptosis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Dibenzazepinas/farmacología , Células Epiteliales/efectos de los fármacos , Femenino , Mucosa Gástrica/citología , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Organoides/efectos de los fármacos , Antro Pilórico , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Receptor Notch2/antagonistas & inhibidores , Receptor Notch2/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/efectos de los fármacos
5.
Science ; 343(6177): 1353-6, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24653033

RESUMEN

Wnt/ß-catenin signaling is critical for tissue regeneration. However, it is unclear how ß-catenin controls stem cell behaviors to coordinate organized growth. Using live imaging, we show that activation of ß-catenin specifically within mouse hair follicle stem cells generates new hair growth through oriented cell divisions and cellular displacement. ß-Catenin activation is sufficient to induce hair growth independently of mesenchymal dermal papilla niche signals normally required for hair regeneration. Wild-type cells are co-opted into new hair growths by ß-catenin mutant cells, which non-cell autonomously activate Wnt signaling within the neighboring wild-type cells via Wnt ligands. This study demonstrates a mechanism by which Wnt/ß-catenin signaling controls stem cell-dependent tissue growth non-cell autonomously and advances our understanding of the mechanisms that drive coordinated regeneration.


Asunto(s)
Folículo Piloso/citología , Folículo Piloso/metabolismo , Cabello/crecimiento & desarrollo , Células Madre/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Diferenciación Celular , División Celular , Ligandos , Ratones , Modelos Biológicos , Mutación , Nicho de Células Madre , Células Madre/citología , Tamoxifeno/farmacología , Regulación hacia Arriba , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
6.
Nat Commun ; 5: 3543, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24667544

RESUMEN

A fundamental goal in cancer biology is to identify the cells and signalling pathways that are keys to induce tumour regression. Here we use a spontaneously self-regressing tumour, cutaneous keratoacanthoma (KAs), to identify physiological mechanisms that drive tumour regression. By using a mouse model system that recapitulates the behaviour of human KAs, we show that self-regressing tumours shift their balance to a differentiation programme during regression. Furthermore, we demonstrate that developmental programs utilized for skin hair follicle regeneration, such as Wnt, are hijacked to sustain tumour growth and that the retinoic acid (RA) signalling pathway promotes tumour regression by inhibiting Wnt signalling. Finally, we find that RA signalling can induce regression of malignant tumours that do not normally spontaneously regress, such as squamous cell carcinomas. These findings provide new insights into the physiological mechanisms of tumour regression and suggest therapeutic strategies to induce tumour regression.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Folículo Piloso/metabolismo , Queratoacantoma/metabolismo , Neoplasias Cutáneas/metabolismo , Células Madre/metabolismo , Tretinoina/metabolismo , Vía de Señalización Wnt , Animales , Modelos Animales de Enfermedad , Folículo Piloso/citología , Ratones , Remisión Espontánea , Células Madre/citología
7.
J Immunol ; 191(8): 4048-58, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043890

RESUMEN

Ezrin is a member of the ezrin-radixin-moesin family of membrane-actin cytoskeleton cross-linkers that participate in a variety of cellular processes. In B cells, phosphorylation of ezrin at different sites regulates multiple processes, such as lipid raft coalescence, BCR diffusion, microclustering, and endosomal JNK activation. In this study, we generated mice with conditional deletion of ezrin in the B cell lineage to investigate the physiological significance of ezrin's function in Ag receptor-mediated B cell activation and humoral immunity. B cell development, as well as the proportion and numbers of major B cell subsets in peripheral lymphoid organs, was unaffected by the loss of ezrin. Using superresolution imaging methods, we show that, in the absence of ezrin, BCRs respond to Ag binding by accumulating into larger and more stable signaling microclusters. Loss of ezrin led to delayed BCR capping and accelerated lipid raft coalescence. Although proximal signaling proteins showed stronger activation in the absence of ezrin, components of the distal BCR signaling pathways displayed distinct effects. Ezrin deficiency resulted in increased B cell proliferation and differentiation into Ab-secreting cells ex vivo and stronger T cell-independent and -dependent responses to Ag in vivo. Overall, our data demonstrate that ezrin regulates amplification of BCR signals and tunes the strength of B cell activation and humoral immunity.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Proteínas del Citoesqueleto/metabolismo , Inmunidad Humoral , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Citoesqueleto de Actina/inmunología , Citoesqueleto de Actina/metabolismo , Animales , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Proliferación Celular , Proteínas del Citoesqueleto/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal/inmunología
8.
Nature ; 487(7408): 496-9, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22763436

RESUMEN

Tissue development and regeneration depend on cell-cell interactions and signals that target stem cells and their immediate progeny. However, the cellular behaviours that lead to a properly regenerated tissue are not well understood. Using a new, non-invasive, intravital two-photon imaging approach we study physiological hair-follicle regeneration over time in live mice. By these means we have monitored the behaviour of epithelial stem cells and their progeny during physiological hair regeneration and addressed how the mesenchyme influences their behaviour. Consistent with earlier studies, stem cells are quiescent during the initial stages of hair regeneration, whereas the progeny are more actively dividing. Moreover, stem cell progeny divisions are spatially organized within follicles. In addition to cell divisions, coordinated cell movements of the progeny allow the rapid expansion of the hair follicle. Finally, we show the requirement of the mesenchyme for hair regeneration through targeted cell ablation and long-term tracking of live hair follicles. Thus, we have established an in vivo approach that has led to the direct observation of cellular mechanisms of growth regulation within the hair follicle and that has enabled us to precisely investigate functional requirements of hair-follicle components during the process of physiological regeneration.


Asunto(s)
Folículo Piloso/citología , Regeneración/fisiología , Células Madre/citología , Animales , División Celular , Movimiento Celular , Supervivencia Celular , Rastreo Celular , Dermis/citología , Terapia por Láser , Mesodermo/citología , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica
9.
Proc Natl Acad Sci U S A ; 108(29): 11924-9, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730140

RESUMEN

Individual cell types are defined by architecturally and functionally specialized cortical domains. The Ezrin, Radixin, and Moesin (ERM) proteins play a major role in organizing cortical domains by assembling membrane protein complexes and linking them to the cortical actin cytoskeleton. Many studies have focused on the individual roles of the ERM proteins in stabilizing the membrane-cytoskeleton interface, controlling the distribution and function of apical membrane complexes, regulating the small GTPase Rho, or establishing cell-cell junctions. We previously found that deletion of the mouse Ezrin gene yields severe defects in apical integrity throughout the developing intestinal epithelium, resulting in incomplete villus morphogenesis and neonatal death. However, the molecular function of Ezrin in building the apical surface of the intestinal epithelium was not clear. By deleting Ezrin in the adult mouse intestinal epithelium, we provide evidence that Ezrin performs multiple molecular functions that collaborate to build the functional apical surface of the intestinal epithelium in vivo. The loss of Ezrin-mediated apical integrity in the adult intestine yields severe morphological consequences during intestinal homeostasis, including defects in cell geometry, extrusion, junctional remodeling, and spindle orientation. Surprisingly, deletion of Ezrin either before or after villus morphogenesis yields villus fusion, revealing a previously unrecognized step in intestinal homeostasis. Our studies indicate that the function of Ezrin in building and maintaining the apical domain is essential not only for intestinal morphogenesis but also for homeostasis in the mature intestine.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Homeostasis/fisiología , Mucosa Intestinal/ultraestructura , Morfogénesis/fisiología , Animales , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Técnicas Histológicas , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiología , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión
10.
PLoS One ; 5(8): e12404, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20806059

RESUMEN

BACKGROUND: Ezrin/radixin/moesin (ERM) proteins are highly homologous proteins that function to link cargo molecules to the actin cytoskeleton. Ezrin and moesin are both expressed in mature lymphocytes, where they play overlapping roles in cell signaling and polarity, but their role in lymphoid development has not been explored. METHODOLOGY/PRINCIPAL FINDINGS: We characterized ERM protein expression in lymphoid tissues and analyzed the requirement for ezrin expression in lymphoid development. In wildtype mice, we found that most cells in the spleen and thymus express both ezrin and moesin, but little radixin. ERM protein expression in the thymus was differentially regulated, such that ezrin expression was highest in immature thymocytes and diminished during T cell development. In contrast, moesin expression was low in early thymocytes and upregulated during T cell development. Mice bearing a germline deletion of ezrin exhibited profound defects in the size and cellularity of the spleen and thymus, abnormal thymic architecture, diminished hematopoiesis, and increased proportions of granulocytic precursors. Further analysis using fetal liver chimeras and thymic transplants showed that ezrin expression is dispensable in hematopoietic and stromal lineages, and that most of the defects in lymphoid development in ezrin(-/-) mice likely arise as a consequence of nutritional stress. CONCLUSIONS/SIGNIFICANCE: We conclude that despite high expression in lymphoid precursor cells, ezrin is dispensable for lymphoid development, most likely due to redundancy with moesin.


Asunto(s)
Proteínas del Citoesqueleto/genética , Regulación del Desarrollo de la Expresión Génica , Linfocitos T/citología , Timo/crecimiento & desarrollo , Timo/metabolismo , Animales , Recuento de Células , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/metabolismo , Hematopoyesis , Masculino , Proteínas de la Membrana/genética , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Bazo/citología , Bazo/metabolismo , Células del Estroma/metabolismo , Linfocitos T/metabolismo , Timo/citología , Factores de Tiempo , Regulación hacia Arriba
11.
Genes Dev ; 24(16): 1718-30, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20675406

RESUMEN

The molecular signals that control the maintenance and activation of liver stem/progenitor cells are poorly understood, and the role of liver progenitor cells in hepatic tumorigenesis is unclear. We report here that liver-specific deletion of the neurofibromatosis type 2 (Nf2) tumor suppressor gene in the developing or adult mouse specifically yields a dramatic, progressive expansion of progenitor cells throughout the liver without affecting differentiated hepatocytes. All surviving mice eventually developed both cholangiocellular and hepatocellular carcinoma, suggesting that Nf2(-/-) progenitors can be a cell of origin for these tumors. Despite the suggested link between Nf2 and the Hpo/Wts/Yki signaling pathway in Drosophila, and recent studies linking the corresponding Mst/Lats/Yap pathway to mammalian liver tumorigenesis, our molecular studies suggest that Merlin is not a major regulator of YAP in liver progenitors, and that the overproliferation of Nf2(-/-) liver progenitors is instead driven by aberrant epidermal growth factor receptor (EGFR) activity. Indeed, pharmacologic inhibition of EGFR blocks the proliferation of Nf2(-/-) liver progenitors in vitro and in vivo, consistent with recent studies indicating that the Nf2-encoded protein Merlin can control the abundance and signaling of membrane receptors such as EGFR. Together, our findings uncover a critical role for Nf2/Merlin in controlling homeostasis of the liver stem cell niche.


Asunto(s)
Homeostasis/fisiología , Neoplasias Hepáticas/fisiopatología , Hígado/fisiopatología , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Células Madre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/fisiopatología , Proteínas de Ciclo Celular , Proliferación Celular , Células Cultivadas , Colangiocarcinoma/genética , Colangiocarcinoma/fisiopatología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Hepatomegalia/genética , Hepatomegalia/fisiopatología , Homeostasis/genética , Hígado/citología , Neoplasias Hepáticas/genética , Masculino , Ratones , Neurofibromatosis 2/genética , Fosfoproteínas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Proteínas Señalizadoras YAP
12.
J Immunol ; 182(2): 1021-32, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124745

RESUMEN

The highly homologous proteins ezrin, radixin, and moesin link proteins to the actin cytoskeleton. The two family members expressed in T cells, ezrin and moesin, are implicated in promoting T cell activation and polarity. To elucidate the contributions of ezrin and moesin, we conducted a systematic analysis of their function during T cell activation. In response to TCR engagement, ezrin and moesin were phosphorylated in parallel at the regulatory threonine, and both proteins ultimately localized to the distal pole complex (DPC). However, ezrin exhibited unique behaviors, including tyrosine phosphorylation and transient localization to the immunological synapse before movement to the DPC. To ask whether these differences reflect unique requirements for ezrin vs moesin in T cell signaling, we generated mice with conditional deletion of ezrin in mature T cells. Ezrin-/- T cells exhibited normal immunological synapse organization based upon localization of protein kinase C-theta, talin, and phospho-ZAP70. DPC localization of CD43 and RhoGDP dissociation inhibitor, as well as the novel DPC protein Src homology region 2 domain-containing phosphatase-1, was also unaffected. However, recruitment of three novel DPC proteins, ezrin binding protein of 50 kDa, Csk binding protein, and the p85 subunit of PI3K was partially perturbed. Biochemical analysis of ezrin-/- T cells or T cells suppressed for moesin using small interfering RNA showed intact early TCR signaling, but diminished levels of IL-2. The defects in IL-2 production were more pronounced in T cells deficient for both ezrin and moesin. These cells also exhibited diminished phospholipase C-gamma1 phosphorylation and calcium flux. We conclude that despite their unique movement and phosphorylation patterns, ezrin and moesin function together to promote T cell activation.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Activación de Linfocitos/inmunología , Proteínas de Microfilamentos/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Línea Celular , Células Cultivadas , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Humanos , Sinapsis Inmunológicas/genética , Células Jurkat , Activación de Linfocitos/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/inmunología
13.
Exp Eye Res ; 82(4): 720-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16289046

RESUMEN

Ezrin, a member of the ezrin/moesin/radixin (ERM) family, localizes to microvilli of epithelia in vivo, where it functions as a bridge between actin filaments and plasma membrane proteins. In the eye, ezrin has been localized to both apical microvilli of Müller cells and retinal pigment epithelium (RPE) apical microvilli and basal infoldings. In the present study, we analyze these structures in the eyes of early postnatal ezrin knockout mice. This analysis indicates that the loss of ezrin leads to substantial reductions in the apical microvilli and basal infoldings in RPE cells and in the Müller cell apical microvilli. The absence of apical microvilli in the RPE is accompanied by the presence of microvilli-like inclusions (MIs) in the RPE cytoplasm. Finally, photoreceptors in the ezrin knockout animals show substantial retardation in development as compared to their wild type littermates.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Epitelio Pigmentado Ocular/patología , Animales , Animales Recién Nacidos , Biomarcadores/análisis , Citoplasma/patología , Proteínas del Citoesqueleto/análisis , Células Epiteliales/patología , Inmunohistoquímica/métodos , Ratones , Ratones Noqueados , Microscopía Electrónica/métodos , Microvellosidades/metabolismo , Microvellosidades/patología , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Epitelio Pigmentado Ocular/metabolismo
14.
Dev Cell ; 6(6): 855-64, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15177033

RESUMEN

Ezrin, Radixin, and Moesin (the ERM proteins) supply regulated linkage between membrane proteins and the actin cytoskeleton. The study of mammalian ERM proteins has been hampered by presumed functional overlap. We have found that Ezrin, the only ERM detected in epithelial cells of the developing intestine, provides an essential role in configuring the mouse intestinal epithelium. Surprisingly, Ezrin is not absolutely required for the formation of brush border microvilli or for the establishment or maintenance of epithelial polarity. Instead, Ezrin organizes the apical terminal web region, which is critical for the poorly understood process of de novo lumen formation and expansion during villus morphogenesis. Our data also suggest that Ezrin controls the localization and/or function of certain apical membrane proteins that support normal intestinal function. These in vivo studies highlight the critical function of Ezrin in the formation of a multicellular epithelium rather than an individual epithelial cell.


Asunto(s)
Diferenciación Celular/genética , Células Epiteliales/metabolismo , Insuficiencia de Crecimiento/genética , Mucosa Intestinal/anomalías , Organogénesis/genética , Fosfoproteínas/fisiología , Animales , Animales Recién Nacidos , Comunicación Celular/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Proteínas del Citoesqueleto , Células Epiteliales/ultraestructura , Insuficiencia de Crecimiento/patología , Feto , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Fenotipo , Fosfoproteínas/deficiencia , Fosfoproteínas/genética
15.
Genes Dev ; 17(9): 1090-100, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12695331

RESUMEN

Mutation of the Neurofibromatosis 2 (NF2) tumor suppressor gene leads to cancer development in humans and mice. Recent studies suggest that Nf2 loss also contributes to tumor metastasis. The Nf2-encoded protein, merlin, is related to the ERM (ezrin, radixin, and moesin) family of membrane:cytoskeleton-associated proteins. However, the cellular mechanism whereby merlin controls cell proliferation from this location is not known. Here we show that the major cellular consequence of Nf2 deficiency in primary cells is an inability to undergo contact-dependent growth arrest and to form stable cadherin-containing cell:cell junctions. Merlin colocalizes and interacts with adherens junction (AJ) components in confluent wild-type cells, suggesting that the lack of AJs and contact-dependent growth arrest in Nf2(-/-) cells directly results from the absence of merlin at sites of cell:cell contact. Our studies indicate that merlin functions as a tumor and metastasis suppressor by controlling cadherin-mediated cell:cell contact.


Asunto(s)
Uniones Adherentes/fisiología , Metástasis de la Neoplasia , Neoplasias/genética , Neurofibromina 2/deficiencia , Animales , Ratones , Neoplasias/etiología , Neurofibromina 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA