Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38858073

RESUMEN

Neural cells are segregated into their distinct central nervous system (CNS) and peripheral nervous system (PNS) domains. However, at specialized regions of the nervous system known as transition zones (TZs), glial cells from both the CNS and PNS are uniquely present with other specialized TZ cells. Herein we review the current understanding of vertebrate TZ cells. The article discusses the distinct cells at vertebrate TZs with a focus on cells that are located on the peripheral side of the spinal cord TZs. In addition to the developmental origin and differentiation of these TZ cells, the functional importance and the role of TZ cells in disease are highlighted. This article also reviews the common and unique features of vertebrate TZs from zebrafish to mice. We propose challenges and open questions in the field that could lead to exciting insights in the field of glial biology.

2.
Sci Transl Med ; 16(753): eadj1597, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38924432

RESUMEN

Congenital pseudarthrosis of the tibia (CPT) is a severe pathology marked by spontaneous bone fractures that fail to heal, leading to fibrous nonunion. Half of patients with CPT are affected by the multisystemic genetic disorder neurofibromatosis type 1 (NF1) caused by mutations in the NF1 tumor suppressor gene, a negative regulator of RAS-mitogen-activated protein kinase (MAPK) signaling pathway. Here, we analyzed patients with CPT and Prss56-Nf1 knockout mice to elucidate the pathogenic mechanisms of CPT-related fibrous nonunion and explored a pharmacological approach to treat CPT. We identified NF1-deficient Schwann cells and skeletal stem/progenitor cells (SSPCs) in pathological periosteum as affected cell types driving fibrosis. Whereas NF1-deficient SSPCs adopted a fibrotic fate, NF1-deficient Schwann cells produced critical paracrine factors including transforming growth factor-ß and induced fibrotic differentiation of wild-type SSPCs. To counteract the elevated RAS-MAPK signaling in both NF1-deficient Schwann cells and SSPCs, we used MAPK kinase (MEK) and Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) inhibitors. Combined MEK-SHP2 inhibition in vivo prevented fibrous nonunion in the Prss56-Nf1 knockout mouse model, providing a promising therapeutic strategy for the treatment of fibrous nonunion in CPT.


Asunto(s)
Ratones Noqueados , Neurofibromina 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Seudoartrosis , Células de Schwann , Animales , Células de Schwann/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/patología , Seudoartrosis/patología , Seudoartrosis/metabolismo , Seudoartrosis/congénito , Neurofibromina 1/metabolismo , Neurofibromina 1/genética , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Células Madre/metabolismo , Células Madre/efectos de los fármacos , Tibia/patología , Ratones , Diferenciación Celular/efectos de los fármacos , Masculino , Neurofibromatosis 1/patología , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/complicaciones , Fibrosis , Femenino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores
3.
Elife ; 122023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38095361

RESUMEN

In addition to their roles in protecting nerves and increasing conduction velocity, peripheral glia plays key functions in blood vessel development by secreting molecules governing arteries alignment and maturation with nerves. Here, we show in mice that a specific, nerve-attached cell population, derived from boundary caps (BCs), constitutes a major source of mural cells for the developing skin vasculature. Using Cre-based reporter cell tracing and single-cell transcriptomics, we show that BC derivatives migrate into the skin along the nerves, detach from them, and differentiate into pericytes and vascular smooth muscle cells. Genetic ablation of this population affects the organization of the skin vascular network. Our results reveal the heterogeneity and extended potential of the BC population in mice, which gives rise to mural cells, in addition to previously described neurons, Schwann cells, and melanocytes. Finally, our results suggest that mural specification of BC derivatives takes place before their migration along nerves to the mouse skin.


Asunto(s)
Cresta Neural , Tubo Neural , Ratones , Animales , Cresta Neural/fisiología , Neuroglía , Células de Schwann , Piel , Diferenciación Celular/fisiología
4.
J Invest Dermatol ; 143(8): 1378-1387, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37330719

RESUMEN

Neurofibromatosis type 1 (NF1) is caused by a nonfunctional copy of the NF1 tumor suppressor gene that predisposes patients to the development of cutaneous neurofibromas (cNFs), the skin tumor that is the hallmark of this condition. Innumerable benign cNFs, each appearing by an independent somatic inactivation of the remaining functional NF1 allele, form in nearly all patients with NF1. One of the limitations in developing a treatment for cNFs is an incomplete understanding of the underlying pathophysiology and limitations in experimental modeling. Recent advances in preclinical in vitro and in vivo modeling have substantially enhanced our understanding of cNF biology and created unprecedented opportunities for therapeutic discovery. We discuss the current state of cNF preclinical in vitro and in vivo model systems, including two- and three-dimensional cell cultures, organoids, genetically engineered mice, patient-derived xenografts, and porcine models. We highlight the models' relationship to human cNFs and how they can be used to gain insight into cNF development and therapeutic discovery.


Asunto(s)
Neurofibroma , Neurofibromatosis 1 , Neoplasias Cutáneas , Ratones , Humanos , Animales , Porcinos , Neurofibromatosis 1/genética , Neurofibromatosis 1/terapia , Mutación , Neurofibroma/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Alelos
5.
Transl Res ; 261: 16-27, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37331503

RESUMEN

Cutaneous neurofibromas (cNFs) are a hallmark of patients with the neurofibromatosis type 1 (NF1) genetic disorder. These benign nerve sheath tumors, which can amount to thousands, develop from puberty onward, often cause pain and are considered by patients to be the primary burden of the disease. Mutations of NF1, encoding a negative regulator of the RAS signaling pathway, in the Schwann cell (SCs) lineage are considered to be at the origin of cNFs. The mechanisms governing cNFs development are poorly understood, and therapeutics to reduce cNFs are missing, mainly due to the lack of appropriate animal models. To address this, we designed the Nf1-KO mouse model that develops cNFs. Using this model, we found that cNFs development is a singular event and goes through 3 successive stages: initiation, progression, and stabilization characterized by changes in the proliferative and MAPK activities of tumor SCs. We found that skin trauma accelerated the development of cNFs and further used this model to explore the efficacy of the MEK inhibitor binimetinib to cure these tumors. We showed that while topically delivered binimetinib has a selective and minor effect on mature cNFs, the same drug prevents their development over long periods.


Asunto(s)
Neurofibroma , Neurofibromatosis 1 , Neoplasias Cutáneas , Humanos , Ratones , Animales , Neurofibromatosis 1/tratamiento farmacológico , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibroma/tratamiento farmacológico , Neurofibroma/genética , Bencimidazoles , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/prevención & control , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas Activadas por Mitógenos
6.
Elife ; 112022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35019839

RESUMEN

Peripheral nerves are vascularized by a dense network of blood vessels to guarantee their complex function. Despite the crucial role of vascularization to ensure nerve homeostasis and regeneration, the mechanisms governing nerve invasion by blood vessels remain poorly understood. We found, in mice, that the sciatic nerve invasion by blood vessels begins around embryonic day 16 and continues until birth. Interestingly, intra-nervous blood vessel density significantly decreases during post-natal period, starting from P10. We show that, while the axon guidance molecule Netrin-1 promotes nerve invasion by blood vessels via the endothelial receptor UNC5B during embryogenesis, myelinated Schwann cells negatively control intra-nervous vascularization during post-natal period.


Asunto(s)
Neovascularización Fisiológica , Fibras Nerviosas Mielínicas/fisiología , Netrina-1/genética , Células de Schwann/fisiología , Nervio Ciático/fisiología , Animales , Movimiento Celular , Femenino , Masculino , Ratones , Neovascularización Patológica , Regeneración Nerviosa , Netrina-1/metabolismo , Nervio Ciático/crecimiento & desarrollo
7.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34884531

RESUMEN

Central nervous system (CNS) lesions are a leading cause of death and disability worldwide. Three-dimensional neural cultures in biomaterials offer more physiologically relevant models for disease studies, toxicity screenings or in vivo transplantations. Herein, we describe the development and use of pullulan/dextran polysaccharide-based scaffolds for 3D neuronal culture. We first assessed scaffolding properties upon variation of the concentration (1%, 1.5%, 3% w/w) of the cross-linking agent, sodium trimetaphosphate (STMP). The lower STMP concentration (1%) allowed us to generate scaffolds with higher porosity (59.9 ± 4.6%), faster degradation rate (5.11 ± 0.14 mg/min) and lower elastic modulus (384 ± 26 Pa) compared with 3% STMP scaffolds (47 ± 2.1%, 1.39 ± 0.03 mg/min, 916 ± 44 Pa, respectively). Using primary cultures of embryonic neurons from PGKCre, Rosa26tdTomato embryos, we observed that in 3D culture, embryonic neurons remained in aggregates within the scaffolds and did not attach, spread or differentiate. To enhance neuronal adhesion and neurite outgrowth, we then functionalized the 1% STMP scaffolds with laminin. We found that treatment of the scaffold with a 100 µg/mL solution of laminin, combined with a subsequent freeze-drying step, created a laminin mesh network that significantly enhanced embryonic neuron adhesion, neurite outgrowth and survival. Such scaffold therefore constitutes a promising neuron-compatible and biodegradable biomaterial.


Asunto(s)
Materiales Biocompatibles/química , Técnicas de Cultivo Tridimensional de Células/métodos , Embrión de Mamíferos/citología , Neuronas/citología , Polisacáridos/química , Andamios del Tejido/química , Animales , Adhesión Celular , Supervivencia Celular , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Porosidad , Ingeniería de Tejidos
8.
Cell ; 180(4): 780-795.e25, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32059781

RESUMEN

The cerebral vasculature is a dense network of arteries, capillaries, and veins. Quantifying variations of the vascular organization across individuals, brain regions, or disease models is challenging. We used immunolabeling and tissue clearing to image the vascular network of adult mouse brains and developed a pipeline to segment terabyte-sized multichannel images from light sheet microscopy, enabling the construction, analysis, and visualization of vascular graphs composed of over 100 million vessel segments. We generated datasets from over 20 mouse brains, with labeled arteries, veins, and capillaries according to their anatomical regions. We characterized the organization of the vascular network across brain regions, highlighting local adaptations and functional correlates. We propose a classification of cortical regions based on the vascular topology. Finally, we analysed brain-wide rearrangements of the vasculature in animal models of congenital deafness and ischemic stroke, revealing that vascular plasticity and remodeling adopt diverging rules in different models.


Asunto(s)
Adaptación Fisiológica , Encéfalo/irrigación sanguínea , Capilares/anatomía & histología , Arterias Cerebrales/anatomía & histología , Venas Cerebrales/anatomía & histología , Remodelación Vascular , Animales , Capilares/patología , Arterias Cerebrales/patología , Venas Cerebrales/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Privación Sensorial , Estrés Psicológico/etiología , Estrés Psicológico/patología , Accidente Cerebrovascular/patología
9.
Acta Neuropathol ; 138(3): 457-476, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31011859

RESUMEN

Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinß1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.


Asunto(s)
Vasos Sanguíneos , Movimiento Celular/fisiología , Efrina-B3/metabolismo , Remielinización/fisiología , Células de Schwann/fisiología , Médula Espinal/metabolismo , Animales , Enfermedades Desmielinizantes/patología , Femenino , Fibronectinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/fisiología , Médula Espinal/patología
10.
Cancer Discov ; 9(1): 130-147, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30348676

RESUMEN

Patients carrying an inactive NF1 allele develop tumors of Schwann cell origin called neurofibromas (NF). Genetically engineered mouse models have significantly enriched our understanding of plexiform forms of NFs (pNF). However, this has not been the case for cutaneous neurofibromas (cNF), observed in all NF1 patients, as no previous model recapitulates their development. Here, we show that conditional Nf1 inactivation in Prss56-positive boundary cap cells leads to bona fide pNFs and cNFs. This work identifies subepidermal glia as a likely candidate for the cellular origin of cNFs and provides insights on disease mechanisms, revealing a long, multistep pathologic process in which inflammation-related signals play a pivotal role. This new mouse model is an important asset for future clinical and therapeutic investigations of NF1-associated neurofibromas. SIGNIFICANCE: Patients affected by NF1 develop numerous cNFs. We present a mouse model that faithfully recapitulates cNFs, identify a candidate cell type at their origin, analyze the steps involved in their formation, and show that their development is dramatically accelerated by skin injury. These findings have important clinical/therapeutic implications.This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
Neurofibroma/metabolismo , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Células de Schwann/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Mutación , Neurofibroma/etiología , Neurofibroma/genética , Neurofibroma/fisiopatología , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/genética , Neurofibromatosis 1/fisiopatología , Células de Schwann/fisiología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/fisiopatología
11.
PLoS Genet ; 14(3): e1007244, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29529029

RESUMEN

A mismatch between optical power and ocular axial length results in refractive errors. Uncorrected refractive errors constitute the most common cause of vision loss and second leading cause of blindness worldwide. Although the retina is known to play a critical role in regulating ocular growth and refractive development, the precise factors and mechanisms involved are poorly defined. We have previously identified a role for the secreted serine protease PRSS56 in ocular size determination and PRSS56 variants have been implicated in the etiology of both hyperopia and myopia, highlighting its importance in refractive development. Here, we use a combination of genetic mouse models to demonstrate that Prss56 mutations leading to reduced ocular size and hyperopia act via a loss of function mechanism. Using a conditional gene targeting strategy, we show that PRSS56 derived from Müller glia contributes to ocular growth, implicating a new retinal cell type in ocular size determination. Importantly, we demonstrate that persistent activity of PRSS56 is required during distinct developmental stages spanning the pre- and post-eye opening periods to ensure optimal ocular growth. Thus, our mouse data provide evidence for the existence of a molecule contributing to both the prenatal and postnatal stages of human ocular growth. Finally, we demonstrate that genetic inactivation of Prss56 rescues axial elongation in a mouse model of myopia caused by a null mutation in Egr1. Overall, our findings identify PRSS56 as a potential therapeutic target for modulating ocular growth aimed at preventing or slowing down myopia, which is reaching epidemic proportions.


Asunto(s)
Ojo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Errores de Refracción/genética , Serina Proteasas/metabolismo , Animales , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Ojo/citología , Ojo/embriología , Femenino , Humanos , Hiperopía/genética , Masculino , Ratones Mutantes , Ratones Transgénicos , Miopía/genética , Miopía/patología , Neuroglía/metabolismo , Refracción Ocular/genética , Refracción Ocular/fisiología , Errores de Refracción/prevención & control , Serina Proteasas/genética
12.
Development ; 145(1)2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29158447

RESUMEN

Although cardiac neural crest cells are required at early stages of arterial valve development, their contribution during valvular leaflet maturation remains poorly understood. Here, we show in mouse that neural crest cells from pre-otic and post-otic regions make distinct contributions to the arterial valve leaflets. Genetic fate-mapping analysis of Krox20-expressing neural crest cells shows a large contribution to the borders and the interleaflet triangles of the arterial valves. Loss of Krox20 function results in hyperplastic aortic valve and partially penetrant bicuspid aortic valve formation. Similar defects are observed in neural crest Krox20-deficient embryos. Genetic lineage tracing in Krox20-/- mutant mice shows that endothelial-derived cells are normal, whereas neural crest-derived cells are abnormally increased in number and misplaced in the valve leaflets. In contrast, genetic ablation of Krox20-expressing cells is not sufficient to cause an aortic valve defect, suggesting that adjacent cells can compensate this depletion. Our findings demonstrate a crucial role for Krox20 in arterial valve development and reveal that an excess of neural crest cells may be associated with bicuspid aortic valve.


Asunto(s)
Válvula Aórtica/anomalías , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Células Endoteliales/metabolismo , Enfermedades de las Válvulas Cardíacas/embriología , Miocardio/metabolismo , Cresta Neural/metabolismo , Animales , Válvula Aórtica/citología , Válvula Aórtica/embriología , Enfermedad de la Válvula Aórtica Bicúspide , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Células Endoteliales/citología , Ratones , Ratones Noqueados , Miocardio/citología , Cresta Neural/citología
13.
Curr Opin Neurobiol ; 47: 209-215, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29174469

RESUMEN

Broad plasticity of the peripheral glia is an emerging concept during development of the peripheral nervous system (PNS). Recent studies have identified the neural crest-derived boundary caps (BCs), as a multitask stem cell population of the developing PNS. BC progeny migrate along the nerves to provide the major glial component of nerve roots and nerve terminals in the skin. Strikingly, those two locations constitute the privileged sites for development of benign peripheral nerve sheath tumors called neurofibromas in patients with neurofibromatosis type 1 (NF1), making BCs attractive candidates for the 'cell of origin' of this disease. Here, we review these exciting findings, focusing on the origin and novel functions of BCs. We further discuss the heterogeneity of BCs, and address their implication in the pathogenesis of NF1.


Asunto(s)
Cresta Neural/citología , Células-Madre Neurales/citología , Neurofibromatosis 1/fisiopatología , Neuroglía/citología , Sistema Nervioso Periférico/citología , Animales , Humanos , Cresta Neural/patología , Cresta Neural/fisiología , Células-Madre Neurales/patología , Células-Madre Neurales/fisiología , Neuroglía/patología , Neuroglía/fisiología , Sistema Nervioso Periférico/patología , Sistema Nervioso Periférico/fisiología
14.
Science ; 357(6346)2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28684471

RESUMEN

Adrenaline is a fundamental circulating hormone for bodily responses to internal and external stressors. Chromaffin cells of the adrenal medulla (AM) represent the main neuroendocrine adrenergic component and are believed to differentiate from neural crest cells. We demonstrate that large numbers of chromaffin cells arise from peripheral glial stem cells, termed Schwann cell precursors (SCPs). SCPs migrate along the visceral motor nerve to the vicinity of the forming adrenal gland, where they detach from the nerve and form postsynaptic neuroendocrine chromaffin cells. An intricate molecular logic drives two sequential phases of gene expression, one unique for a distinct transient cellular state and another for cell type specification. Subsequently, these programs down-regulate SCP-gene and up-regulate chromaffin cell-gene networks. The AM forms through limited cell expansion and requires the recruitment of numerous SCPs. Thus, peripheral nerves serve as a stem cell niche for neuroendocrine system development.


Asunto(s)
Médula Suprarrenal/embriología , Diferenciación Celular , Células Cromafines/citología , Células Madre Multipotentes/citología , Células-Madre Neurales/citología , Células Neuroendocrinas/citología , Células de Schwann/citología , Médula Suprarrenal/citología , Animales , Diferenciación Celular/genética , Movimiento Celular , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Mutantes , Proteína Proteolipídica de la Mielina/genética , Cresta Neural/citología , Nervios Periféricos/citología , Factores de Transcripción SOXE/genética , Nicho de Células Madre/genética , Transcripción Genética
15.
Brain Struct Funct ; 221(9): 4411-4427, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26701169

RESUMEN

Adult neurogenesis in the mammalian brain is restricted to specific regions, such as the dentate gyrus (DG) in the hippocampus and the subventricular zone (SVZ) in the walls of the lateral ventricles. Here, we used a mouse line carrying a knock-in of Cre recombinase in the Prss56 gene, in combination with two Cre-inducible fluorescent reporters (Rosa26 mTmG and Rosa26 tdTom ), to perform genetic tracing of Prss56-expressing cells in the adult brain. We found reporter-positive cells in three neurogenic niches: the DG, the SVZ and the hypothalamus ventricular zone. In the prospective DG, Prss56 is expressed during embryogenesis in a subpopulation of radial glia. The pattern of migration and differentiation of reporter-positive cells during development recapitulates the successive steps of DG neurogenesis, including the formation of a subpopulation of adult neural stem cells (NSC). In the SVZ, Prss56 is expressed postnatally in a subpopulation of adult NSC mainly localized in the medial-ventral region of the lateral wall. This subpopulation preferentially gives rise to deep granule and Calbindin-positive periglomerular interneurons in the olfactory bulb. Finally, Prss56 is also expressed in a subpopulation of α2-tanycytes, which are potential adult NSCs of the hypothalamus ventricular zone. Our observations suggest that some α2-tanycytes translocate their soma into the parenchyma and may give rise to a novel cell type in this territory. Overall, this study establishes the Prss56 Cre line as an efficient and promising new tool to study multiple aspects of adult neurogenesis in the mouse.


Asunto(s)
Células Madre Adultas/fisiología , Encéfalo/fisiología , Células-Madre Neurales/fisiología , Neurogénesis , Serina Proteasas/metabolismo , Células Madre Adultas/metabolismo , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Movimiento Celular , Giro Dentado/embriología , Giro Dentado/metabolismo , Giro Dentado/fisiología , Células Ependimogliales/metabolismo , Células Ependimogliales/fisiología , Interneuronas/metabolismo , Interneuronas/fisiología , Ventrículos Laterales/embriología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Serina Proteasas/fisiología
16.
Stem Cell Reports ; 5(2): 278-90, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26212662

RESUMEN

While neurogenic stem cells have been identified in rodent and human skin, their manipulation and further characterization are hampered by a lack of specific markers. Here, we perform genetic tracing of the progeny of boundary cap (BC) cells, a neural-crest-derived cell population localized at peripheral nerve entry/exit points. We show that BC derivatives migrate along peripheral nerves to reach the skin, where they give rise to terminal glia associated with dermal nerve endings. Dermal BC derivatives also include cells that self-renew in sphere culture and have broad in vitro differentiation potential. Upon transplantation into adult mouse dorsal root ganglia, skin BC derivatives efficiently differentiate into various types of mature sensory neurons. Together, this work establishes the embryonic origin, pathway of migration, and in vivo neurogenic potential of a major component of skin stem-like cells. It provides genetic tools to study and manipulate this population of high interest for medical applications.


Asunto(s)
Células-Madre Neurales/citología , Neurogénesis , Neuroglía/citología , Piel/citología , Animales , Linaje de la Célula , Movimiento Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/fisiología , Células Receptoras Sensoriales/citología
17.
Dev Cell ; 33(3): 343-50, 2015 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-25942625

RESUMEN

Dorsal root ganglia (DRG) sensory neurons arise from heterogeneous precursors that differentiate in two neurogenic waves, respectively controlled by Neurog2 and Neurog1. We show here that transgenic mice expressing a Zeb1/2 dominant-negative form (DBZEB) exhibit reduced numbers of nociceptors and altered pain sensitivity. This reflects an early impairment of Neurog1-dependent neurogenesis due to the depletion of specific sensory precursor pools, which is slightly later partially compensated by the contribution of boundary cap cells (BCCs). Indeed, combined DBZEB expression and genetic BCCs ablation entirely deplete second wave precursors and, in turn, nociceptors, thus recapitulating the Neurog1(-/-) neuronal phenotype. Altogether, our results uncover roles for Zeb family members in the developing DRGs; they show that the Neurog1-dependent sensory neurogenesis can be functionally partitioned in two successive phases; and finally, they illustrate plasticity in the developing peripheral somatosensory system supported by the BCCs, thereby providing a rationale for sensory precursor diversity.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Plasticidad Neuronal/fisiología , Nociceptores/metabolismo , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Ganglios Espinales/embriología , Ganglios Espinales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Factores de Transcripción de Tipo Kruppel/genética , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neurogénesis/fisiología , Plasticidad Neuronal/genética , Proteínas Represoras/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
18.
Cardiovasc Res ; 104(3): 443-55, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25344368

RESUMEN

AIMS: Heart valve maturation is achieved by the organization of extracellular matrix (ECM) and the distribution of valvular interstitial cells. However, the factors that regulate matrix components required for valvular structure and function are unknown. Based on the discovery of its specific expression in cardiac valves, we aimed to uncover the role of Krox20 (Egr-2) during valve development and disease. METHODS AND RESULTS: Using series of mouse genetic tools, we demonstrated that loss of function of Krox20 caused significant hyperplasia of the semilunar valves, while atrioventricular valves appeared normal. This defect was associated with an increase in valvular interstitial cell number and ECM volume. Echo Doppler analysis revealed that adult mutant mice had aortic insufficiency. Defective aortic valves (AoVs) in Krox20(-/-) mice had features of human AoV disease, including excess of proteoglycan deposition and reduction of collagen fibres. Furthermore, examination of diseased human AoVs revealed decreased expression of KROX20. To identify downstream targets of Krox20, we examined expression of fibrillar collagens in the AoV leaflets at different stages in the mouse. We found significant down-regulation of Col1a1, Col1a2, and Col3a1 in the semilunar valves of Krox20 mutant mice. Utilizing in vitro and in vivo experiments, we demonstrated that Col1a1 and Col3a1 are direct targets of Krox20 activation in interstitial cells of the AoV. CONCLUSION: This study identifies a previously unknown function of Krox20 during heart valve development. These results indicate that Krox20-mediated activation of fibrillar Col1a1 and Col3a1 genes is crucial to avoid postnatal degeneration of the AoV leaflets.


Asunto(s)
Válvula Aórtica/embriología , Colágeno Tipo I/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/fisiología , Regulación del Desarrollo de la Expresión Génica , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Insuficiencia de la Válvula Aórtica/genética , Insuficiencia de la Válvula Aórtica/metabolismo , Insuficiencia de la Válvula Aórtica/patología , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Desarrollo Embrionario , Femenino , Colágenos Fibrilares/genética , Colágenos Fibrilares/metabolismo , Homeostasis , Humanos , Masculino , Mesodermo/metabolismo , Ratones , Regiones Promotoras Genéticas , Activación Transcripcional
19.
PLoS One ; 9(1): e81868, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465372

RESUMEN

Gene transcription is essential for learning, but the precise role of transcription factors that control expression of many other genes in specific learning paradigms is yet poorly understood. Zif268 (Krox24/Egr-1) is a transcription factor and an immediate-early gene associated with memory consolidation and reconsolidation, and induced in the striatum after addictive drugs exposure. In contrast, very little is known about its physiological role at early stages of operant learning. We investigated the role of Zif268 in operant conditioning for food. Zif268 expression was increased in all regions of the dorsal striatum and nucleus accumbens in mice subjected to the first session of operant conditioning. In contrast, Zif268 increase in the dorsomedial caudate-putamen and nucleus accumbens core was not detected in yoked mice passively receiving the food reward. This indicates that Zif268 induction in these structures is linked to experiencing or learning contingency, but not to reward delivery. When the task was learned (5 sessions), Zif268 induction disappeared in the nucleus accumbens and decreased in the medial caudate-putamen, whereas it remained high in the lateral caudate-putamen, previously implicated in habit formation. In transgenic mice expressing green fluorescent protein (GFP) in the striatonigral neurons, Zif268 induction occured after the first training session in both GFP-positive and negative neurons indicating an enhanced Zif268 expression in both striatonigral and striatopallidal neurons. Mutant mice lacking Zif268 expression obtained less rewards, but displayed a normal discrimination between reinforced and non-reinforced targets, and an unaltered approach to food delivery box. In addition, their motivation to obtain food rewards, evaluated in a progressive ratio schedule, was blunted. In conclusion, Zif268 participates in the processes underlying performance and motivation to execute food-conditioned instrumental task.


Asunto(s)
Condicionamiento Operante , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Plasticidad Neuronal , Adaptación Fisiológica , Animales , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Conducta Alimentaria/psicología , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Motivación , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Especificidad de Órganos , Activación Transcripcional
20.
J Neuropathol Exp Neurol ; 71(11): 938-47, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23095846

RESUMEN

Endoneurial fibroblast-like cells (EFLCs) have been described for more than 60 years, but the embryology, functions, and pathology of these cells are not well defined. Several hypotheses of their origin have been proposed. A previous study suggesting that they were of neural crest origin is supported by our data in humans. This lineage might account for EFLCs having multiple biologic functions and involvement in pathological processes. Here, we review what is known about the origin; functions in collagen synthesis, phagocytosis, inflammatory responses, and immune surveillance; and the pathological alterations of EFLCs based on the literature and on our personal observations.


Asunto(s)
Fibroblastos/citología , Cresta Neural/citología , Nervios Periféricos/citología , Fagocitos/citología , Animales , Linaje de la Célula/fisiología , Fibroblastos/clasificación , Fibroblastos/fisiología , Humanos , Fagocitos/clasificación , Fagocitos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...