Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37665322

RESUMEN

One-carbon/folate (1C) metabolism supplies methyl groups required for DNA and histone methylation, and is involved in the maintenance of self-renewal in stem cells. Dihydrofolate reductase (DHFR), a key enzyme in 1C metabolism, is highly expressed in human and mouse neural progenitors at the early stages of neocortical development. Here, we have investigated the role of DHFR in the developing neocortex and report that reducing its activity in human neural organoids and mouse embryonic neocortex accelerates indirect neurogenesis, thereby affecting neuronal composition of the neocortex. Furthermore, we show that decreasing DHFR activity in neural progenitors leads to a reduction in one-carbon/folate metabolites and correlates with modifications of H3K4me3 levels. Our findings reveal an unanticipated role for DHFR in controlling specific steps of neocortex development and indicate that variations in 1C metabolic cues impact cell fate transitions.


Asunto(s)
Neocórtex , Neurogénesis , Tetrahidrofolato Deshidrogenasa , Animales , Humanos , Ratones , Carbono , Ácido Fólico , Neurogénesis/genética , Tetrahidrofolato Deshidrogenasa/genética
2.
J Neurosci ; 43(7): 1154-1165, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36596698

RESUMEN

During development, cortical neurons are produced in a temporally regulated sequence from apical progenitors, directly or indirectly, through the production of intermediate basal progenitors. The balance between these major progenitor types is critical for the production of the proper number and types of neurons, and it is thus important to decipher the cellular and molecular cues controlling this equilibrium. Here we address the role of a cell cycle regulator, the CDC25B phosphatase, in this process. We show that, in the developing mouse neocortex of both sex, deleting CDC25B in apical progenitors leads to a transient increase in the production of TBR1+ neurons at the expense of TBR2+ basal progenitors. This phenotype is associated with lengthening of the G2 phase of the cell cycle, the total cell cycle length being unaffected. Using in utero electroporation and cortical slice cultures, we demonstrate that the defect in TBR2+ basal progenitor production requires interaction with CDK1 and is because of the G2 phase lengthening in CDC25B mutants. Together, this study identifies a new role for CDC25B and G2 phase length in direct versus indirect neurogenesis at early stages of cortical development.SIGNIFICANCE STATEMENT This study is the first analysis of the function of CDC25B, a G2/M regulator, in the developing neocortex. We show that removing CDC25B function leads to a transient increase in neuronal differentiation at early stages, occurring simultaneously with a decrease in basal intermediate progenitors (bIPs). Conversely, a CDC25B gain of function promotes production of bIPs, and this is directly related to CDC25B's ability to regulate CDK1 activity. This imbalance of neuron/progenitor production is linked to a G2 phase lengthening in apical progenitors; and using pharmacological treatments on cortical slice cultures, we show that shortening the G2 phase is sufficient to enhance bIP production. Our results reveal the importance of G2 phase length regulation for neural progenitor fate determination.


Asunto(s)
Neocórtex , Células-Madre Neurales , Neurogénesis , Animales , Ratones , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/metabolismo
3.
Dev Dyn ; 252(3): 363-376, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36153792

RESUMEN

BACKGROUND: The apical surface (AS) of epithelial cells is highly specialized; it is important for morphogenetic processes that are essential to shape organs and tissues and it plays a role in morphogen and growth factor signaling. Apical progenitors in the mammalian neocortex are pseudoepithelial cells whose apical surface lines the ventricle. Whether changes in their apical surface sizes are important for cortical morphogenesis and/or other aspects of neocortex development has not been thoroughly addressed. RESULTS: Here we show that apical progenitors are heterogeneous with respect to their apical surface area. In Efnb1 mutants, the size of the apical surface is modified and this correlates with discrete alterations of tissue organization without impacting apical progenitors proliferation. CONCLUSIONS: Altogether, our data reveal heterogeneity in apical progenitors AS area in the developing neocortex and shows a role for Ephrin B1 in controlling AS size. Our study also indicates that changes in AS size do not have strong repercussion on apical progenitor behavior.


Asunto(s)
Neocórtex , Neuronas , Animales , Neuronas/metabolismo , Transducción de Señal , Efrina-B1/metabolismo , Mamíferos/metabolismo
4.
BMC Dev Biol ; 20(1): 12, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404061

RESUMEN

BACKGROUND: During mammalian cerebral cortex development, different types of projection neurons are produced in a precise temporal order and in stereotypical numbers. The mechanisms regulating timely generation of neocortex projection neurons and ensuring production in sufficient numbers of each neuronal identity are only partially understood. RESULTS: Here, we show that ephrin-B2, a member of the Eph:ephrin cell-to-cell communication pathway, sets the neurogenic tempo in the neocortex. Indeed, conditional mutant embryos for ephrin-B2 exhibit a transient delay in neurogenesis and acute stimulation of Eph signaling by in utero injection of synthetic ephrin-B2 led to a transient increase in neuronal production. Using genetic approaches we show that ephrin-B2 acts on neural progenitors to control their differentiation in a juxtacrine manner. Unexpectedly, we observed that perinatal neuron numbers recovered following both loss and gain of ephrin-B2, highlighting the ability of neural progenitors to adapt their behavior to the state of the system in order to produce stereotypical numbers of neurons. CONCLUSIONS: Altogether, our data uncover a role for ephrin-B2 in embryonic neurogenesis and emphasize the plasticity of neuronal production in the neocortex.


Asunto(s)
Efrina-B2/metabolismo , Neocórtex/citología , Neocórtex/metabolismo , Neuronas/citología , Neuronas/metabolismo , Animales , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Efrina-B2/genética , Femenino , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Neurogénesis/genética , Neurogénesis/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
5.
J Pathol ; 239(3): 250-61, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27015986

RESUMEN

CDKN1C encodes the cyclin-CDK inhibitor p57(Kip2) (p57), a negative regulator of the cell cycle and putative tumour suppressor. Genetic and epigenetic alterations causing loss of p57 function are the most frequent cause of Beckwith-Wiedemann syndrome (BWS), a genetic disorder characterized by multiple developmental anomalies and increased susceptibility to tumour development during childhood. So far, BWS development has been attributed entirely to the deregulation of proliferation caused by loss of p57-mediated CDK inhibition. However, a fraction of BWS patients have point mutations in CDKN1C located outside of the CDK inhibitory region, suggesting the involvement of other parts of the protein in the disease. To test this possibility, we generated knock-in mice deficient for p57-mediated cyclin-CDK inhibition (p57(CK) (-) ), the only clearly defined function of p57. Comparative analysis of p57(CK) (-) and p57(KO) mice provided clear evidence for CDK-independent roles of p57 and revealed that BWS is not caused entirely by CDK deregulation, as several features of BWS are caused by the loss of CDK-independent roles of p57. Thus, while the genetic origin of BWS is well understood, our results underscore that the underlying molecular mechanisms remain largely unclear. To probe these mechanisms further, we determined the p57 interactome. Several partners identified are involved in genetic disorders with features resembling those caused by CDKN1C mutation, suggesting that they could be involved in BWS pathogenesis and revealing a possible connection between seemingly distinct syndromes. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica/genética , Proteínas Supresoras de Tumor/genética , Secuencia de Aminoácidos , Animales , Síndrome de Beckwith-Wiedemann/patología , Ciclo Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fenotipo , Alineación de Secuencia , Proteínas Supresoras de Tumor/metabolismo
6.
Development ; 140(10): 2082-92, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23578932

RESUMEN

Apical neural progenitors are polarized cells for which the apical membrane is the site of cell-cell and cell-extracellular matrix adhesion events that are essential for maintaining the integrity of the developing neuroepithelium. Apical adhesion is important for several aspects of the nervous system development, including morphogenesis and neurogenesis, yet the mechanisms underlying its regulation remain poorly understood. Here, we show that ephrin B1, a cell surface protein that engages in cell signaling upon binding cognate Eph receptors, controls normal morphogenesis of the developing cortex. Efnb1-deficient embryos exhibit morphological alterations of the neuroepithelium that correlate with neural tube closure defects. Using loss-of-function experiments by ex vivo electroporation, we demonstrate that ephrin B1 is required in apical progenitors (APs) to maintain their apical adhesion. Mechanistically, we show that ephrin B1 controls cell-ECM adhesion by promoting apical localization of integrin ß1 and we identify ADP-ribosylation factor 6 (Arf6) as an important effector of ephrin B1 reverse signaling in apical adhesion of APs. Our results provide evidence for an important role for ephrin B1 in maintaining the structural integrity of the developing cortex and highlight the importance of tightly controlling apical cell-ECM adhesion for neuroepithelial development.


Asunto(s)
Efrina-B1/fisiología , Neuronas/citología , Células Madre/citología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Animales , Tipificación del Cuerpo , Encéfalo/embriología , Adhesión Celular , Comunicación Celular , Membrana Celular/metabolismo , Células Cultivadas , Electroporación , Efrina-B1/metabolismo , Femenino , Masculino , Ratones , Ratones Transgénicos , Tubo Neural/embriología , Factores de Tiempo
7.
Nature ; 465(7297): 483-6, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20445537

RESUMEN

In development, tissue regeneration or certain diseases, angiogenic growth leads to the expansion of blood vessels and the lymphatic vasculature. This involves endothelial cell proliferation as well as angiogenic sprouting, in which a subset of cells, termed tip cells, acquires motile, invasive behaviour and extends filopodial protrusions. Although it is already appreciated that angiogenesis is triggered by tissue-derived signals, such as vascular endothelial growth factor (VEGF) family growth factors, the resulting signalling processes in endothelial cells are only partly understood. Here we show with genetic experiments in mouse and zebrafish that ephrin-B2, a transmembrane ligand for Eph receptor tyrosine kinases, promotes sprouting behaviour and motility in the angiogenic endothelium. We link this pro-angiogenic function to a crucial role of ephrin-B2 in the VEGF signalling pathway, which we have studied in detail for VEGFR3, the receptor for VEGF-C. In the absence of ephrin-B2, the internalization of VEGFR3 in cultured cells and mutant mice is defective, which compromises downstream signal transduction by the small GTPase Rac1, Akt and the mitogen-activated protein kinase Erk. Our results show that full VEGFR3 signalling is coupled to receptor internalization. Ephrin-B2 is a key regulator of this process and thereby controls angiogenic and lymphangiogenic growth.


Asunto(s)
Efrina-B2/metabolismo , Linfangiogénesis , Neovascularización Fisiológica , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Pérdida del Embrión , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Endocitosis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Efrina-B2/deficiencia , Efrina-B2/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Linfangiogénesis/genética , Vasos Linfáticos , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Neuropéptidos/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor EphB4/deficiencia , Receptor EphB4/genética , Receptor EphB4/metabolismo , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
8.
Dev Biol ; 383(2): 264-74, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24056079

RESUMEN

Axon fasciculation is one of the processes controlling topographic innervation during embryonic development. While axon guidance steers extending axons in the accurate direction, axon fasciculation allows sets of co-extending axons to grow in tight bundles. The Eph:ephrin family has been involved both in axon guidance and fasciculation, yet it remains unclear how these two distinct types of responses are elicited. Herein we have characterized the role of ephrin-B1, a member of the ephrinB family in sensory and motor innervation of the limb. We show that ephrin-B1 is expressed in sensory axons and in the limb bud mesenchyme while EphB2 is expressed in motor and sensory axons. Loss of ephrin-B1 had no impact on the accurate dorso-ventral innervation of the limb by motor axons, yet EfnB1 mutants exhibited decreased fasciculation of peripheral motor and sensory nerves. Using tissue-specific excision of EfnB1 and in vitro experiments, we demonstrate that ephrin-B1 controls fasciculation of axons via a surround repulsion mechanism involving growth cone collapse of EphB2-expressing axons. Altogether, our results highlight the complex role of Eph:ephrin signaling in the development of the sensory-motor circuit innervating the limb.


Asunto(s)
Axones/fisiología , Efrina-B1/metabolismo , Neuronas Motoras/fisiología , Receptores de la Familia Eph/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Efrina-B2/metabolismo , Extremidades/embriología , Extremidades/inervación , Ganglios Espinales/metabolismo , Conos de Crecimiento/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/metabolismo , Mutación/genética , Células Receptoras Sensoriales/metabolismo
9.
Circ Res ; 110(5): 688-700, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22302788

RESUMEN

RATIONALE: Cardiac tissue cohesion relying on highly ordered cardiomyocytes (CM) interactions is critical because most cardiomyopathies are associated with tissue remodeling and architecture alterations. OBJECTIVE: Eph/ephrin system constitutes a ubiquitous system coordinating cellular communications which recently emerged as a major regulator in adult organs. We examined if eph/ephrin could participate in cardiac tissue cyto-organization. METHODS AND RESULTS: We reported the expression of cardiac ephrin-B1 in both endothelial cells and for the first time in CMs where ephrin-B1 localized specifically at the lateral membrane. Ephrin-B1 knock-out (KO) mice progressively developed cardiac tissue disorganization with loss of adult CM rod-shape and sarcomeric and intercalated disk structural disorganization confirmed in CM-specific ephrin-B1 KO mice. CMs lateral membrane exhibited abnormal structure by electron microscopy and notably increased stiffness by atomic force microscopy. In wild-type CMs, ephrin-B1 interacted with claudin-5/ZO-1 complex at the lateral membrane, whereas the complex disappeared in KO/CM-specific ephrin-B1 KO mice. Ephrin-B1 deficiency resulted in decreased mRNA expression of CM basement membrane components and disorganized fibrillar collagen matrix, independently of classical integrin/dystroglycan system. KO/CM-specific ephrin-B1 KO mice exhibited increased left ventricle diameter and delayed atrioventricular conduction. Under pressure overload stress, KO mice were prone to death and exhibited striking tissue disorganization. Finally, failing CMs displayed downregulated ephrin-B1/claudin-5 gene expression linearly related to the ejection fraction. CONCLUSIONS: Ephrin-B1 is necessary for cardiac tissue architecture cohesion by stabilizing the adult CM morphology through regulation of its lateral membrane. Because decreased ephrin-B1 is associated with molecular/functional cardiac defects, it could represent a new actor in the transition toward heart failure.


Asunto(s)
Comunicación Celular/fisiología , Efrina-B1/fisiología , Proteínas de la Membrana/fisiología , Miocitos Cardíacos/fisiología , Animales , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Células Cultivadas , Colágeno/fisiología , Colágeno/ultraestructura , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Endotelio Vascular/ultraestructura , Efrina-B1/deficiencia , Efrina-B1/genética , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Miocitos Cardíacos/citología , Miocitos Cardíacos/ultraestructura , Sarcómeros/diagnóstico por imagen , Sarcómeros/fisiología , Ultrasonografía
10.
Genesis ; 49(10): 811-20, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21735541

RESUMEN

Genetic studies have shown that ephrin-B2 and its cognate EphB4 receptor are necessary for normal embryonic angiogenesis. Moreover, there is overwhelming evidence that ephrin-B2 is involved in tumor vascularization, yet its role in adult angiogenesis has been difficult to track genetically. Here, we report the generation of transgenic mice that over-express EfnB2 specifically in endothelial cells (ECs). We show that exogenous expression of EfnB2 under the control of the Tie2 promoter/enhancer regions in ECs does not affect viability or growth of the transgenic animals. We further show that targeted expression of EfnB2 in ECs is not sufficient to rescue severe cardiovascular defects at mid-gestation stages but rescues early embryonic lethality associated with loss-of-function mutation in EfnB2. This mouse model will be useful to study the role of ephrin-B2 in physiological and pathological angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Efrina-B2/genética , Efrina-B2/metabolismo , Neovascularización Fisiológica , Animales , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Efrina-B2/biosíntesis , Genotipo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas Receptoras/genética , Receptor EphB4/genética , Receptor EphB4/metabolismo , Receptor TIE-2 , Transducción de Señal/fisiología
11.
Brain Res Bull ; 174: 153-160, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34139316

RESUMEN

Chromosome 13q deletions encompassing EFNB2, which encodes the transmembrane protein ephrin-B2, are likely to cause syndromic forms of sensorineural hearing loss of unclear origin. Thus, unravelling the pathogenic mechanisms could help to improve therapeutic strategies. In the cochlea, adjacent non-sensory epithelial cells are connected via gap junction channels, the activity of which is critical to maintain cochlear homeostasis. Here we show that ephrin-B2 promotes the assembly of connexin 30 (Cx30) gap junction plaques (GJPs) between adjacent non-sensory Deiters' cells. An in situ proximity ligation assay revealed that ephrin-B2 preferentially interacts with Cx30 in the periphery of the GJPs, i.e. where newly synthesized connexin hemichannels accrue to the GJP. Moreover, we observed that heterozygous mice encoding an Efnb2 null allele display excessive clathrin-mediated internalization of Cx30 GJPs in early postnatal stages. Finally, an in vitro organotypic assay revealed that ectopic activation of ephrin-B2 reverse signalling promotes the internalization of Cx30 GJPs. These data argue in favor of a cell-autonomous, Eph receptor-independent role of ephrin-B2 in the assembly of Cx30 GJPs. According to recent observations, early GJP degradation could certainly play a role in the pathogenic process leading to progressive sensorineural hearing loss due to Efnb2/EFNB2 haploinsufficiency.


Asunto(s)
Cóclea/patología , Sinapsis Eléctricas/patología , Endocitosis/genética , Efrina-B2/genética , Animales , Conexina 30/biosíntesis , Conexina 30/genética , Efrina-B2/farmacología , Haploinsuficiencia , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Heterocigoto , Ratones , Ratones Noqueados , Transducción de Señal/genética
12.
Cancer Lett ; 503: 129-137, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33545223

RESUMEN

Brain tumors are a heterogeneous group of benign and malignant tumors arising from the brain parenchyma and its surrounding structures, with in general a poor clinical outcome due to high recurrence. One of the underlying causes for this somber prognostic is the presence of brain tumor initiating cells (BTIC) endowed with self-renewal potential, multi-lineage differentiation and resistance to treatment. One promising therapeutic avenue for brain tumors is targeting BTIC self-renewal potential and forcing their differentiation. A compelling candidate is one-carbon metabolism shown to play a key role in maintaining stem cell self-renewal in several lineages. Here, we focus on dihydrofolate reductase (DHFR), a key enzyme in one-carbon metabolism, and demonstrate this enzyme's overexpression in several human brain tumors and its expression in human BTIC. We show that DHFR inhibition, either by Methotrexate (MTX) or EphB activation with synthetic ligands, reduces the tumorigenic potential of 4 human BTIC lines, by reducing their self-renewal capacities both in vitro and in a cerebral organoid glioma (GLICO) model. Our data indicate that driving BTIC differentiation by inhibiting DHFR may provide a new therapeutic approach to treating highly refractory aggressive tumors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Metotrexato/farmacología , Células Madre Neoplásicas/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Organoides/citología , Organoides/efectos de los fármacos , Organoides/patología , Pronóstico
13.
Cereb Cortex Commun ; 1(1): tgaa063, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-34296126

RESUMEN

The mammalian neocortex is composed of different subtypes of projection neurons that are generated sequentially during embryogenesis by differentiation of neural progenitors. While molecular mechanisms that control neuronal production in the developing neocortex have been extensively studied, the dynamics and absolute numbers of the different progenitor and neuronal populations are still poorly characterized. Here, we describe a medium throughput approach based on flow cytometry and well-known identity markers of cortical subpopulations to collect quantitative data over the course of mouse neocortex development. We collected a complete dataset in a physiological developmental context on two progenitor and two neuron populations, including relative proportions and absolute numbers. Our study reveals unexpected total numbers of Tbr2+ progenitors. In addition, we show that polyploid neurons are present throughout neocortex development.

14.
PLoS Biol ; 4(10): e315, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16968134

RESUMEN

Mutations in X-linked ephrin-B1 in humans cause craniofrontonasal syndrome (CFNS), a disease that affects female patients more severely than males. Sorting of ephrin-B1-positive and -negative cells following X-inactivation has been observed in ephrin-B1(+/-) mice; however, the mechanisms by which mosaic ephrin-B1 expression leads to cell sorting and phenotypic defects remain unknown. Here we show that ephrin-B1(+/-) mice exhibit calvarial defects, a phenotype autonomous to neural crest cells that correlates with cell sorting. We have traced the causes of calvarial defects to impaired differentiation of osteogenic precursors. We show that gap junction communication (GJC) is inhibited at ectopic ephrin boundaries and that ephrin-B1 interacts with connexin43 and regulates its distribution. Moreover, we provide genetic evidence that GJC is implicated in the calvarial defects observed in ephrin-B1(+/-) embryos. Our results uncover a novel role for Eph/ephrins in regulating GJC in vivo and suggest that the pleiotropic defects seen in CFNS patients are due to improper regulation of GJC in affected tissues.


Asunto(s)
Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Efrinas/metabolismo , Uniones Comunicantes , Animales , Diferenciación Celular , Conexina 43/metabolismo , Anomalías Craneofaciales/patología , Modelos Animales de Enfermedad , Efrina-B1/biosíntesis , Efrina-B2/biosíntesis , Efrinas/fisiología , Femenino , Masculino , Ratones , Ratones Transgénicos , Mutación , Síndrome
15.
Biochem Biophys Res Commun ; 375(3): 362-6, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18694724

RESUMEN

Eph receptor tyrosine kinases and their membrane-bound ligand ephrins form an essential cell communication system. Both ephrin classes have been shown to localize within cell surface lipid rafts, yet regulate different biological processes. In order to provide insight into this distinct behavior, we examined ephrin-A5 and B1 localization and signaling in murine fibroblasts and tissues. Results indicated that ephrin-A5 was constitutively present in detergent-resistant membrane fractions, while ephrin-B1 displayed translocation to membrane fractions upon stimulation. Ephrin-A5 and B1 were present in detergent-resistant membrane fractions with different buoyancies in vitro and in different raft fractions in vivo. Moreover, ephrin-A5 and B1 differentially influenced actin reorganization. Finally, microarray analysis revealed unique patterns of gene expression between the two ephrin classes. We thus demonstrate that distinct localization and compartmentalization provide insight into the subcellular basis for differential signaling observed in ephrin-A and B classes.


Asunto(s)
Efrina-A5/metabolismo , Efrina-B1/metabolismo , Microdominios de Membrana/metabolismo , Animales , Citoesqueleto/ultraestructura , Efrina-A5/análisis , Efrina-A5/clasificación , Efrina-B1/análisis , Efrina-B1/clasificación , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Microdominios de Membrana/química , Microdominios de Membrana/ultraestructura , Ratones , Células 3T3 NIH
16.
Epigenet Insights ; 11: 2516865718820946, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30627699

RESUMEN

Balancing self-renewal with differentiation is crucial for neural stem cells (NSC) functions to ensure tissue development and homeostasis. Over the last years, multiple studies have highlighted the coupling of either metabolic or epigenetic reprogramming to NSC fate decisions. Metabolites are essential as they provide the energy and building blocks for proper cell function. Moreover, metabolites can also function as substrates and/or cofactors for epigenetic modifiers. It is becoming more evident that metabolic alterations and epigenetics rewiring are highly intertwined; however, their relation regarding determining NSC fate is not well understood. In this review, we summarize the major metabolic pathways and epigenetic modifications that play a role in NSC. We then focus on the notion that nutrients availability can function as a switch to modify the epigenetic machinery and drive NSC sequential differentiation during embryonic neurogenesis.

17.
Cell Rep ; 23(10): 2864-2873.e7, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29874574

RESUMEN

Metabolic pathways, once seen as a mere consequence of cell states, have emerged as active players in dictating different cellular events such as proliferation, self-renewal, and differentiation. Several studies have reported a role for folate-dependent one-carbon (1C) metabolism in stem cells; however, its exact mode of action and how it interacts with other cues are largely unknown. Here, we report a link between the Eph:ephrin cell-cell communication pathway and 1C metabolism in controlling neural stem cell differentiation. Transcriptional and functional analyses following ephrin stimulation revealed alterations in folate metabolism-related genes and enzymatic activity. In vitro and in vivo data indicate that Eph-B forward signaling alters the methylation state of H3K4 by regulating 1C metabolism and locks neural stem cell in a differentiation-ready state. Our study highlights a functional link between cell-cell communication, metabolism, and epigenomic remodeling in the control of stem cell self-renewal.


Asunto(s)
Carbono/metabolismo , Diferenciación Celular , Efrinas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Epigénesis Genética , Histonas/metabolismo , Patrón de Herencia/genética , Metilación , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Tetrahidrofolato Deshidrogenasa/metabolismo
18.
Curr Biol ; 28(11): 1768-1782.e4, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29779877

RESUMEN

The corpus callosum is the largest commissure in the brain, whose main function is to ensure communication between homotopic regions of the cerebral cortex. During fetal development, corpus callosum axons (CCAs) grow toward and across the brain midline and then away on the contralateral hemisphere to their targets. A particular feature of this circuit, which raises a key developmental question, is that the outgoing trajectory of post-crossing CCAs is mirror-symmetric with the incoming trajectory of pre-crossing axons. Here, we show that post-crossing CCAs switch off their response to axon guidance cues, among which the secreted Semaphorin-3C (Sema3C), that act as attractants for pre-crossing axons on their way to the midline. This change is concomitant with an upregulation of the surface protein Ephrin-B1, which acts in CCAs to inhibit Sema3C signaling via interaction with the Neuropilin-1 (Nrp1) receptor. This silencing activity is independent of Eph receptors and involves a N-glycosylation site (N-139) in the extracellular domain of Ephrin-B1. Together, our results reveal a molecular mechanism, involving interaction between the two unrelated guidance receptors Ephrin-B1 and Nrp1, that is used to control the navigation of post-crossing axons in the corpus callosum.


Asunto(s)
Axones/fisiología , Cuerpo Calloso/fisiología , Efrina-B1/genética , Regulación del Desarrollo de la Expresión Génica , Neuropilina-1/genética , Semaforinas/genética , Animales , Efrina-B1/metabolismo , Silenciador del Gen , Ratones , Neuropilina-1/metabolismo , Semaforinas/metabolismo
19.
Neural Dev ; 12(1): 10, 2017 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-28595615

RESUMEN

BACKGROUND: In the vertebrate spinal cord, motor neurons (MN) are generated in stereotypical numbers from a pool of dedicated progenitors (pMN) whose number depends on signals that control their specification but also their proliferation and differentiation rates. Although the initial steps of pMN specification have been extensively studied, how pMN numbers are regulated over time is less well characterized. RESULTS: Here, we show that ephrinB2 and ephrinB3 are differentially expressed in progenitor domains in the ventral spinal cord with several Eph receptors more broadly expressed. Genetic loss-of-function analyses show that ephrinB2 and ephrinB3 inversely control pMN numbers and that these changes in progenitor numbers correlate with changes in motor neuron numbers. Detailed phenotypic analyses by immunostaining and genetic interaction studies between ephrinB2 and Shh indicate that changes in pMN numbers in ephrin mutants are due to alteration in progenitor identity at late stages of development. CONCLUSIONS: Altogether our data reveal that Eph:ephrin signaling is required to control progenitor identities in the ventral spinal cord.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Neuronas Motoras/metabolismo , Células-Madre Neurales/metabolismo , Receptores de la Familia Eph/metabolismo , Médula Espinal/embriología , Médula Espinal/metabolismo , Animales , Recuento de Células , Proteínas Hedgehog/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
20.
J Cell Biol ; 214(5): 555-69, 2016 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-27551053

RESUMEN

Cytokinesis is the last step of cell division, culminating in the physical separation of daughter cells at the end of mitosis. Cytokinesis is a tightly regulated process that until recently was mostly viewed as a cell-autonomous event. Here, we investigated the role of Ephrin/Eph signaling, a well-known local cell-to-cell communication pathway, in cell division. We show that activation of Eph signaling in vitro leads to multinucleation and polyploidy, and we demonstrate that this is caused by alteration of the ultimate step of cytokinesis, abscission. Control of abscission requires Eph kinase activity, and Src and citron kinase (CitK) are downstream effectors in the Eph-induced signal transduction cascade. CitK is phosphorylated on tyrosines in neural progenitors in vivo, and Src kinase directly phosphorylates CitK. We have identified the specific tyrosine residues of CitK that are phosphorylated and show that tyrosine phosphorylation of CitK impairs cytokinesis. Finally, we show that, similar to CitK, Ephrin/Eph signaling controls neuronal ploidy in the developing neocortex. Our study indicates that CitK integrates intracellular and extracellular signals provided by the local environment to coordinate completion of cytokinesis.


Asunto(s)
Citocinesis , Efrina-B2/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Tirosina/metabolismo , Secuencia de Aminoácidos , Animales , Muerte Celular , Femenino , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Masculino , Ratones , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Transducción de Señal , Telofase , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA