Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Development ; 144(15): 2810-2823, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28684624

RESUMEN

In vertebrates, cranial placodes contribute to all sense organs and sensory ganglia and arise from a common pool of Six1/Eya2+ progenitors. Here we dissect the events that specify ectodermal cells as placode progenitors using newly identified genes upstream of the Six/Eya complex. We show in chick that two different tissues, namely the lateral head mesoderm and the prechordal mesendoderm, gradually induce placode progenitors: cells pass through successive transcriptional states, each identified by distinct factors and controlled by different signals. Both tissues initiate a common transcriptional state but over time impart regional character, with the acquisition of anterior identity dependent on Shh signalling. Using a network inference approach we predict the regulatory relationships among newly identified transcription factors and verify predicted links in knockdown experiments. Based on this analysis we propose a new model for placode progenitor induction, in which the initial induction of a generic transcriptional state precedes regional divergence.


Asunto(s)
Transducción de Señal/fisiología , Vertebrados/embriología , Animales , Comunicación Celular/genética , Comunicación Celular/fisiología , Embrión de Pollo , Pollos , Ectodermo/citología , Ectodermo/embriología , Ectodermo/metabolismo , Electroporación , Ganglios Sensoriales/citología , Ganglios Sensoriales/embriología , Ganglios Sensoriales/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Hibridación in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos , Codorniz , Órganos de los Sentidos/citología , Órganos de los Sentidos/embriología , Órganos de los Sentidos/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vertebrados/metabolismo
2.
Dev Biol ; 425(1): 85-99, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28315296

RESUMEN

Cranial sensory ganglia are components of the peripheral nervous system that possess a significant somatosensory role and include neurons within the trigeminal and epibranchial nerve bundles. Although it is well established that these ganglia arise from interactions between neural crest and neurogenic placode cells, the molecular basis of ganglia assembly is still poorly understood. Members of the Annexin protein superfamily play key roles in sensory nervous system development throughout metazoans. Annexin A6 is expressed in chick trigeminal and epibranchial placode cell-derived neuroblasts and neurons, but its function in cranial ganglia formation has not been elucidated. To this end, we interrogated the role of Annexin A6 using gene perturbation studies in the chick embryo. Our data reveal that placode cell-derived neuroblasts with reduced Annexin A6 levels ingress and migrate normally to the ganglionic anlage, where neural crest cell corridors correctly form around them. Strikingly, while Annexin A6-depleted placode cell-derived neurons still express mature neuronal markers, they fail to form two long processes, which are considered morphological features of mature neurons, and no longer innervate their designated targets due to the absence of this bipolar morphology. Moreover, overexpression of Annexin A6 causes some placode cell-derived neurons to form extra protrusions alongside these bipolar processes. These data demonstrate that the molecular program associated with neuronal maturation is distinct from that orchestrating changes in neuronal morphology, and, importantly, reveal Annexin A6 to be a key membrane scaffolding protein during sensory neuron membrane biogenesis. Collectively, our results provide novel insight into mechanisms underscoring morphological changes within placode cell-derived neurons that are essential for cranial gangliogenesis.


Asunto(s)
Anexina A6/metabolismo , Proteínas Aviares/metabolismo , Membrana Celular/metabolismo , Ganglios Sensoriales/metabolismo , Células Receptoras Sensoriales/metabolismo , Cráneo/inervación , Empalme Alternativo , Animales , Anexina A6/genética , Proteínas Aviares/genética , Secuencia de Bases , Embrión de Pollo , Pollos , Ganglios Sensoriales/citología , Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Immunoblotting , Microscopía Confocal , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Receptoras Sensoriales/citología , Homología de Secuencia de Ácido Nucleico
3.
Neural Dev ; 11: 3, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26819088

RESUMEN

BACKGROUND: The cranial sensory ganglia represent populations of neurons with distinct functions, or sensory modalities. The production of individual ganglia from distinct neurogenic placodes with different developmental pathways provides a powerful model to investigate the acquisition of specific sensory modalities. To date there is a limited range of gene markers available to examine the molecular pathways underlying this process. RESULTS: Transcriptional profiles were generated for populations of differentiated neurons purified from distinct cranial sensory ganglia using microdissection in embryonic chicken followed by FAC-sorting and RNAseq. Whole transcriptome analysis confirmed the division into somato- versus viscerosensory neurons, with additional evidence for subdivision of the somatic class into general and special somatosensory neurons. Cross-comparison of distinct ganglia transcriptomes identified a total of 134 markers, 113 of which are novel, which can be used to distinguish trigeminal, vestibulo-acoustic and epibranchial neuronal populations. In situ hybridisation analysis provided validation for 20/26 tested markers, and showed related expression in the target region of the hindbrain in many cases. CONCLUSIONS: One hundred thirty-four high-confidence markers have been identified for placode-derived cranial sensory ganglia which can now be used to address the acquisition of specific cranial sensory modalities.


Asunto(s)
Nervios Craneales/embriología , Nervios Craneales/metabolismo , Ganglios Sensoriales/embriología , Ganglios Sensoriales/metabolismo , Neuronas/fisiología , Transcriptoma , Animales , Diferenciación Celular , Embrión de Pollo , Neuronas/metabolismo
4.
Gene Expr Patterns ; 20(1): 71-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26712358

RESUMEN

Comparative genomic analysis of the nuclear receptor family suggests that the testicular receptor 2, Nr2c1, undergoes positive selection in the human-chimpanzee clade based upon a significant increase in nonsynonymous compared to synonymous substitutions. Previous in situ analyses of Nr2c1 lacked the temporal range and spatial resolution necessary to characterize cellular expression of this gene from early to mid gestation, when many nuclear receptors are key regulators of tissue specific stem or progenitor cells. Thus, we asked whether Nr2c1 protein is associated with stem cell populations in the mid-gestation mouse embryo. Nr2c1 is robustly expressed in the developing olfactory epithelium. Its expression in the olfactory epithelium shifts from multiple progenitor classes at early stages to primarily transit amplifying cells later in olfactory epithelium development. In the early developing central nervous system, Nr2c1 is limited to the anterior telencephalon/olfactory bulb anlagen, coincident with Nestin-positive neuroepithelial stem cells. Nr2c1 is also seen in additional cranial sensory specializations including cells surrounding the mystacial vibrissae, the retinal pigment epithelium and Scarpa's ganglion. Nr2c1 was also detected in a subset of mesenchymal cells in developing teeth and cranial bones. The timing and distribution of embryonic expression suggests that Nr2c1 is primarily associated with the early genesis of mammalian cranial sensory neurons and craniofacial skeletal structures. Thus, Nr2c1 may be a candidate for mediating parallel adaptive changes in cranial neural sensory specializations such as the olfactory epithelium, retina and mystacial vibrissae and in non-neural craniofacial features including teeth.


Asunto(s)
Miembro 1 del Grupo C de la Subfamilia 2 de Receptores Nucleares/biosíntesis , Mucosa Olfatoria/embriología , Cráneo/embriología , Células Madre/metabolismo , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Huesos Faciales/embriología , Huesos Faciales/metabolismo , Ganglios Sensoriales/embriología , Ganglios Sensoriales/metabolismo , Perfilación de la Expresión Génica , Ratones , Células-Madre Neurales/metabolismo , Bulbo Olfatorio/metabolismo , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Cráneo/citología , Cráneo/metabolismo , Telencéfalo/metabolismo , Diente/embriología , Diente/metabolismo
5.
Cell Tissue Res ; 364(1): 29-41, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26453396

RESUMEN

Pannexin1 (Panx1) is one of three members of the pannexin protein family. The expression of Panx1 mRNA has been extensively investigated from late embryonic to adult stages. In contrast, expression during early embryonic development is largely unknown. Our aim is to examine the temporal and spatial expression of Panx1 in mouse embryonic development by focusing on embryonic days (E) 9.5 to 12.5. Whole embryos are investigated in order to provide a comprehensive survey. Analyses were performed at the mRNA level by using reverse transcription plus the polymerase chain reaction and whole-mount in situ hybridization. Panx1 mRNA was detected in the heads and bodies of embryos at all developmental stages investigated (E9.5, E10.5, E11.5, E12.5). In particular, the nervous system expressed Panx1 at an early time point. Interestingly, Panx1 expression was found in afferent ganglia of the cranial nerves and spinal cord. This finding is of particular interest in the context of neuropathic pain and other Panx1-related neurological disorders. Our study shows, for the first time, that Panx1 is expressed in the central and peripheral nervous system during early developmental stages. The consequences of Panx1 deficiency or inhibition in a number of experimental paradigms might therefore be predicated on changes during early development.


Asunto(s)
Conexinas/biosíntesis , Embrión de Mamíferos/embriología , Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Animales , Conexinas/genética , Embrión de Mamíferos/citología , Ganglios Sensoriales/citología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética
6.
Genesis ; 52(10): 833-48, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25074687

RESUMEN

Matrix metalloproteinases (MMPs) are a large and complex family of zinc-dependent endoproteinases widely recognized for their roles in remodeling the extracellular matrix (ECM) during embryonic development, wound healing, and tissue homeostasis. Their misregulation is central to many pathologies, and they have therefore been the focus of biomedical research for decades. These proteases have also recently emerged as mediators of neural development and synaptic plasticity in vertebrates, however, understanding of the mechanistic basis of these roles and the molecular identities of the MMPs involved remains far from complete. We have identified a zebrafish orthologue of mmp25 (a.k.a. leukolysin; MT6-MMP), a membrane-type, furin-activated MMP associated with leukocytes and invasive carcinomas, but which we find is expressed by a subset of the sensory neurons during normal embryonic development. We detect high levels of Mmp25ß expression in the trigeminal, craniofacial, and posterior lateral line ganglia in the hindbrain, and in Rohon-Beard cells in the dorsal neural tube during the first 48 h of embryonic development. Knockdown of Mmp25ß expression with morpholino oligonucleotides results in larvae that are uncoordinated and insensitive to touch, and which exhibit defects in the development of sensory neural structures. Using in vivo zymography, we observe that Mmp25ß morphant embryos show reduced Type IV collagen degradation in regions of the head traversed by elongating axons emanating from the trigeminal ganglion, suggesting that Mmp25ß may play a pivotal role in mediating ECM remodeling in the vicinity of these elongating axons.


Asunto(s)
Ganglios Sensoriales/enzimología , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Células Receptoras Sensoriales/enzimología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Colágeno Tipo IV/metabolismo , Embrión no Mamífero/enzimología , Desarrollo Embrionario , Matriz Extracelular/enzimología , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Metaloproteinasas de la Matriz Asociadas a la Membrana/genética , Especificidad de Órganos , Homología de Secuencia de Aminoácido , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
7.
Dev Biol ; 385(2): 405-16, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24513489

RESUMEN

Cranial neurogenic placodes and the neural crest make essential contributions to key adult characteristics of all vertebrates, including the paired peripheral sense organs and craniofacial skeleton. Neurogenic placode development has been extensively characterized in representative jawed vertebrates (gnathostomes) but not in jawless fishes (agnathans). Here, we use in vivo lineage tracing with DiI, together with neuronal differentiation markers, to establish the first detailed fate-map for placode-derived sensory neurons in a jawless fish, the sea lamprey Petromyzon marinus, and to confirm that neural crest cells in the lamprey contribute to the cranial sensory ganglia. We also show that a pan-Pax3/7 antibody labels ophthalmic trigeminal (opV, profundal) placode-derived but not maxillomandibular trigeminal (mmV) placode-derived neurons, mirroring the expression of gnathostome Pax3 and suggesting that Pax3 (and its single Pax3/7 lamprey ortholog) is a pan-vertebrate marker for opV placode-derived neurons. Unexpectedly, however, our data reveal that mmV neuron precursors are located in two separate domains at neurula stages, with opV neuron precursors sandwiched between them. The different branches of the mmV nerve are not comparable between lampreys and gnatho-stomes, and spatial segregation of mmV neuron precursor territories may be a derived feature of lampreys. Nevertheless, maxillary and mandibular neurons are spatially segregated within gnathostome mmV ganglia, suggesting that a more detailed investigation of gnathostome mmV placode development would be worthwhile. Overall, however, our results highlight the conservation of cranial peripheral sensory nervous system development across vertebrates, yielding insight into ancestral vertebrate traits.


Asunto(s)
Ganglios Sensoriales/embriología , Petromyzon/embriología , Animales , Linaje de la Célula , Ganglios Sensoriales/citología , Cresta Neural/embriología , Neuronas/citología , Factores de Transcripción Paired Box/inmunología , Cráneo
8.
Dev Biol ; 389(1): 28-38, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24491819

RESUMEN

In the vertebrate head, the peripheral components of the sensory nervous system are derived from two embryonic cell populations, the neural crest and cranial sensory placodes. Both arise in close proximity to each other at the border of the neural plate: neural crest precursors abut the future central nervous system, while placodes originate in a common preplacodal region slightly more lateral. During head morphogenesis, complex events organise these precursors into functional sensory structures, raising the question of how their development is coordinated. Here we review the evidence that neural crest and placode cells remain in close proximity throughout their development and interact repeatedly in a reciprocal manner. We also review recent controversies about the relative contribution of the neural crest and placodes to the otic and olfactory systems. We propose that a sequence of mutual interactions between the neural crest and placodes drives the coordinated morphogenesis that generates functional sensory systems within the head.


Asunto(s)
Ectodermo/embriología , Ganglios Sensoriales/embriología , Cabeza/inervación , Cresta Neural/embriología , Animales , Comunicación Celular/genética , Comunicación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Ectodermo/citología , Ectodermo/metabolismo , Ganglios Sensoriales/citología , Ganglios Sensoriales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Modelos Neurológicos , Morfogénesis/genética , Morfogénesis/fisiología , Cresta Neural/citología , Cresta Neural/metabolismo
9.
Development ; 140(17): 3595-600, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23942515

RESUMEN

The majority of cranial sensory neurons originate in placodes in the surface ectoderm, migrating to form ganglia that connect to the central nervous system (CNS). Interactions between inward-migrating sensory neuroblasts and emigrant cranial neural crest cells (NCCs) play a role in coordinating this process, but how the relationship between these two cell populations is established is not clear. Here, we demonstrate that NCCs generate corridors delineating the path of migratory neuroblasts between the placode and CNS in both chick and mouse. In vitro analysis shows that NCCs are not essential for neuroblast migration, yet act as a superior substrate to mesoderm, suggesting provision of a corridor through a less-permissive mesodermal territory. Early organisation of NCC corridors occurs prior to sensory neurogenesis and can be recapitulated in vitro; however, NCC extension to the placode requires placodal neurogenesis, demonstrating reciprocal interactions. Together, our data indicate that NCC corridors impose physical organisation for precise ganglion formation and connection to the CNS, providing a local environment to enclose migrating neuroblasts and axonal processes as they migrate through a non-neural territory.


Asunto(s)
Movimiento Celular/fisiología , Ganglios Sensoriales/embriología , Cresta Neural/fisiología , Células-Madre Neurales/fisiología , Células Receptoras Sensoriales/fisiología , Cráneo/embriología , Animales , Embrión de Pollo , Ganglios Sensoriales/citología , Hibridación in Situ , Ratones , Microscopía Confocal , Cráneo/citología
10.
Proc Natl Acad Sci U S A ; 110(31): 12709-14, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858437

RESUMEN

Skin melanocytes arise from two sources: either directly from neural crest progenitors or indirectly from neural crest-derived Schwann cell precursors after colonization of peripheral nerves. The relationship between these two melanocyte populations and the factors controlling their specification remains poorly understood. Direct lineage tracing reveals that neural crest and Schwann cell progenitor-derived melanocytes are differentially restricted to the epaxial and hypaxial body domains, respectively. Furthermore, although both populations are initially part of the Foxd3 lineage, hypaxial melanocytes lose Foxd3 at late stages upon separation from the nerve, whereas we recently found that epaxial melanocytes segregate earlier from Foxd3-positive neural progenitors while still residing in the dorsal neural tube. Gain- and loss-of-function experiments in avians and mice, respectively, reveal that Foxd3 is both sufficient and necessary for regulating the balance between melanocyte and Schwann cell development. In addition, Foxd3 is also sufficient to regulate the switch between neuronal and glial fates in sensory ganglia. Together, we propose that differential fate acquisition of neural crest-derived cells depends on their progressive segregation from the Foxd3-positive lineage.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Factores de Transcripción Forkhead/metabolismo , Melanocitos/metabolismo , Cresta Neural/embriología , Células-Madre Neurales/metabolismo , Proteínas Represoras/metabolismo , Células de Schwann/metabolismo , Animales , Embrión de Pollo , Pollos , Ganglios Sensoriales/citología , Ganglios Sensoriales/embriología , Melanocitos/citología , Ratones , Cresta Neural/citología , Células-Madre Neurales/citología , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células de Schwann/citología
11.
PLoS One ; 8(2): e57960, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23469119

RESUMEN

Cell fate commitment of spinal progenitor neurons is initiated by long-range, midline-derived, morphogens that regulate an array of transcription factors that, in turn, act sequentially or in parallel to control neuronal differentiation. Included among these are transcription factors that regulate the expression of receptors for guidance cues, thereby determining axonal trajectories. The Ig/FNIII superfamily molecules TAG1/Axonin1/CNTN2 (TAG1) and Neurofascin (Nfasc) are co-expressed in numerous neuronal cell types in the CNS and PNS - for example motor, DRG and interneurons - both promote neurite outgrowth and both are required for the architecture and function of nodes of Ranvier. The genes encoding TAG1 and Nfasc are adjacent in the genome, an arrangement which is evolutionarily conserved. To study the transcriptional network that governs TAG1 and Nfasc expression in spinal motor and commissural neurons, we set out to identify cis elements that regulate their expression. Two evolutionarily conserved DNA modules, one located between the Nfasc and TAG1 genes and the second directly 5' to the first exon and encompassing the first intron of TAG1, were identified that direct complementary expression to the CNS and PNS, respectively, of the embryonic hindbrain and spinal cord. Sequential deletions and point mutations of the CNS enhancer element revealed a 130bp element containing three conserved E-boxes required for motor neuron expression. In combination, these two elements appear to recapitulate a major part of the pattern of TAG1 expression in the embryonic nervous system.


Asunto(s)
Contactina 2/genética , Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Médula Espinal/embriología , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Moléculas de Adhesión Celular/metabolismo , Embrión de Pollo , Secuencia Conservada , Elementos E-Box/genética , Evolución Molecular , Ganglios Sensoriales/citología , Ganglios Sensoriales/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Ratones , Datos de Secuencia Molecular , Neuronas Motoras/metabolismo , Mutagénesis , Factores de Crecimiento Nervioso/metabolismo , Especificidad de Órganos , Ratas , Rombencéfalo/citología , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Raíces Nerviosas Espinales/metabolismo
12.
Dev Biol ; 370(1): 3-23, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22790010

RESUMEN

In the vertebrate head, crucial parts of the sense organs and sensory ganglia develop from special regions, the cranial placodes. Despite their cellular and functional diversity, they arise from a common field of multipotent progenitors and acquire distinct identity later under the influence of local signalling. Here we present the gene regulatory network that summarises our current understanding of how sensory cells are specified, how they become different from other ectodermal derivatives and how they begin to diversify to generate placodes with different identities. This analysis reveals how sequential activation of sets of transcription factors subdivides the ectoderm over time into smaller domains of progenitors for the central nervous system, neural crest, epidermis and sensory placodes. Within this hierarchy the timing of signalling and developmental history of each cell population is of critical importance to determine the ultimate outcome. A reoccurring theme is that local signals set up broad gene expression domains, which are further refined by mutual repression between different transcription factors. The Six and Eya network lies at the heart of sensory progenitor specification. In a positive feedback loop these factors perpetuate their own expression thus stabilising pre-placodal fate, while simultaneously repressing neural and neural crest specific factors. Downstream of the Six and Eya cassette, Pax genes in combination with other factors begin to impart regional identity to placode progenitors. While our review highlights the wealth of information available, it also points to the lack information on the cis-regulatory mechanisms that control placode specification and of how the repeated use of signalling input is integrated.


Asunto(s)
Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Cabeza/inervación , Sistema Nervioso Periférico/embriología , Células Receptoras Sensoriales/fisiología , Vertebrados/embriología , Animales , Diferenciación Celular/fisiología , Ectodermo/fisiología , Cabeza/embriología , Modelos Biológicos , Células-Madre Neurales/citología , Factores de Transcripción/metabolismo
13.
Dev Biol ; 363(1): 84-94, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22210003

RESUMEN

Macrophage migration inhibitory factor (MIF) plays versatile roles in the immune system. MIF is also widely expressed during embryonic development, particularly in the nervous system, although its roles in neural development are only beginning to be understood. Evidence from frogs, mice and zebrafish suggests that MIF has a major role as a neurotrophin in the early development of sensory systems, including the auditory system. Here we show that the zebrafish mif pathway is required for both sensory hair cell (HC) and sensory neuronal cell survival in the ear, for HC differentiation, semicircular canal formation, statoacoustic ganglion (SAG) development, and lateral line HC differentiation. This is consistent with our findings that MIF is expressed in the developing mammalian and avian auditory systems and promotes mouse and chick SAG neurite outgrowth and neuronal survival, demonstrating key instructional roles for MIF in vertebrate otic development.


Asunto(s)
Oído Interno/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores de Crecimiento Nervioso/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Diferenciación Celular/genética , Oído Interno/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Ganglios Sensoriales/embriología , Ganglios Sensoriales/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Ciliadas Auditivas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Pirimidinas/farmacología , Receptores Inmunológicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Semicirculares/embriología , Canales Semicirculares/metabolismo , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Factores de Tiempo , Pez Cebra/embriología
14.
PLoS One ; 6(11): e27150, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22096531

RESUMEN

BACKGROUND: Lysophospholipids such as lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are important signaling molecules that can regulate a wide range of cellular responses. We discovered that Sphingosine kinase 1 (Sphk1), a key enzyme that converts sphingosine to S1P, is expressed in neurons and progenitor cells in nascent trigeminal and dorsal root ganglia during mouse embryogenesis. METHODS AND FINDINGS: Sphk1 null mouse embryos do not display overt deficits owing to compensation by Sphk2. Thus, we analyzed embryos that are deficient in both Sphk1 and Sphk2 (which essentially eliminates S1P function) in order to investigate the role(s) of Sphk1 during sensory ganglia formation. While animals lacking 1-3 alleles of Sphk1 and Sphk2 had no obvious phenotype, embryos without both genes displayed clear developmental defects. The complete absence of Sphk1 and Sphk2 resulted in trigeminal and dorsal root ganglia with fewer neurons and progenitor cells. The profound loss in cell number could be attributed to a decrease in cell proliferation as well as an increase in apoptosis. Furthermore, Sphk1/2 double mutants displayed an overall reduction in other sphingolipids as well as an imbalance of S1P/sphingosine and S1P/ceramide ratio, thereby favoring cell death and reducing cell growth. CONCLUSIONS: Together, these results provide strong in vivo evidence that sphingosine kinase/S1P signaling plays an important role in regulating early events during development of sensory ganglia.


Asunto(s)
Ganglios Sensoriales/citología , Ganglios Sensoriales/metabolismo , Lisofosfolípidos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Células Madre/citología , Células Madre/metabolismo , Animales , Ganglios Sensoriales/embriología , Genotipo , Hibridación in Situ , Ratones , Ratones Mutantes , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Transducción de Señal/genética , Esfingosina/metabolismo
15.
Dev Biol ; 358(1): 147-55, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21806979

RESUMEN

Fibroblast growth factor (FGF) signalling has important roles in the development of the embryonic pharyngeal (branchial) arches, but its effects on innervation of the arches and associated structures have not been studied extensively. We investigated the consequences of deleting two receptor tyrosine kinase (RTK) antagonists of the Sprouty (Spry) gene family on the early development of the branchial nerves. The morphology of the facial, glossopharyngeal and vagus nerves are abnormal in Spry1-/-;Spry2-/- embryos. We identify specific defects in the epibranchial placodes and neural crest, which contribute sensory neurons and glia to these nerves. A dissection of the tissue-specific roles of these genes in branchial nerve development shows that Sprouty gene deletion in the pharyngeal epithelia can affect both placode formation and neural crest fate. However, epithelial-specific gene deletion only results in defects in the facial nerve and not the glossopharyngeal and vagus nerves, suggesting that the facial nerve is most sensitive to perturbations in RTK signalling. Reducing the Fgf8 gene dosage only partially rescued defects in the glossopharyngeal nerve and was not sufficient to rescue facial nerve defects, suggesting that FGF8 is functionally redundant with other RTK ligands during facial nerve development.


Asunto(s)
Región Branquial/inervación , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/fisiología , Fosfoproteínas/fisiología , Transducción de Señal/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Región Branquial/embriología , Nervio Facial/anomalías , Factor 8 de Crecimiento de Fibroblastos/genética , Genotipo , Nervio Glosofaríngeo/anomalías , Haploinsuficiencia , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Cresta Neural/embriología , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas , Nervio Vago/anomalías
16.
Genesis ; 49(4): 278-94, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21381180

RESUMEN

Neurogenic placodes are transient, thickened patches of embryonic vertebrate head ectoderm that give rise to the paired peripheral sense organs and most neurons in cranial sensory ganglia. We present the first analysis of gene expression during neurogenic placode development in a basal actinopterygian (ray-finned fish), the North American paddlefish (Polyodon spathula). Pax3 expression in the profundal placode confirms its homology with the ophthalmic trigeminal placode of amniotes. We report the conservation of expression of Pax2 and Pax8 in the otic and/or epibranchial placodes, Phox2b in epibranchial placode-derived neurons, Sox3 during epibranchial and lateral line placode development, and NeuroD in developing cranial sensory ganglia. We identify Sox3 as a novel marker for developing fields of electrosensory ampullary organs and for ampullary organs themselves. Sox3 is also the first molecular marker for actinopterygian ampullary organs. This is consistent with, though does not prove, a lateral line placode origin for actinopterygian ampullary organs.


Asunto(s)
Ectodermo/embriología , Peces/embriología , Ganglios Sensoriales/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción Paired Box/metabolismo , Filogenia , Órganos de los Sentidos/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ectodermo/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Especificidad de la Especie , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Cell Adh Migr ; 4(4): 622-30, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20930541

RESUMEN

Neural crest cells (NCCs) are migratory cells that delaminate from the neural tube early in development and then disseminate throughout the embryo to give rise to a wide variety of cell types that are key to the vertebrate body plan. During their journey from the neural tube to their peripheral targets, NCCs progressively differentiate, raising the question when the fate of an individual NCC is sealed. One hypothesis suggests that the fate of a NCC is specified by target-derived signals emanating from the environment they migrate through, while another hypothesis proposes that NCCs are already specified to differentiate along select lineages at the time they are born in the neural tube, with environmental signals helping them to realize their prespecified fate potential. Alternatively, both mechanisms may cooperate to drive NCC diversity. This review highlights recent advances in our understanding of prespecification during trunk NCC development.


Asunto(s)
Diferenciación Celular , Cresta Neural/citología , Abdomen/embriología , Abdomen/inervación , Animales , Sistema Nervioso Autónomo/citología , Sistema Nervioso Autónomo/embriología , Movimiento Celular , Rastreo Celular , Ganglios Sensoriales/embriología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cresta Neural/embriología , Neuropilinas/metabolismo , Células Receptoras Sensoriales/citología , Transducción de Señal , Somitos/citología , Somitos/embriología , Tórax/embriología , Tórax/inervación
18.
J Neurosci ; 30(24): 8234-44, 2010 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-20554875

RESUMEN

The lateral line system of teleosts has recently become a model system to study patterning and morphogenesis. However, its embryonic origins are still not well understood. In zebrafish, the posterior lateral line (PLL) system is formed in two waves, one that generates the embryonic line of seven to eight neuromasts and 20 afferent neurons and a second one that generates three additional lines during larval development. The embryonic line originates from a postotic placode that produces both a migrating sensory primordium and afferent neurons. Nothing is known about the origin and innervation of the larval lines. Here we show that a "secondary" placode can be detected at 24 h postfertilization (hpf), shortly after the primary placode has given rise to the embryonic primordium and ganglion. The secondary placode generates two additional sensory primordia, primD and primII, as well as afferent neurons. The primary and secondary placodes require retinoic acid signaling at the same stage of late gastrulation, suggesting that they share a common origin. Neither primary nor secondary neurons show intrinsic specificity for neuromasts derived from their own placode, but the sequence of neuromast deposition ensures that neuromasts are primarily innervated by neurons derived from the cognate placode. The delayed formation of secondary afferent neurons accounts for the capability of the fish to form a new PLL ganglion after ablation of the embryonic ganglion at 24 hpf.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Sistema de la Línea Lateral/citología , Sistema de la Línea Lateral/embriología , Neuronas/fisiología , Pez Cebra/fisiología , Técnicas de Ablación/métodos , Aminoácidos/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/fisiología , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Embrión no Mamífero , Ganglios Sensoriales/citología , Ganglios Sensoriales/embriología , Proteínas Fluorescentes Verdes/genética , Larva/crecimiento & desarrollo , Neuronas/clasificación , Transducción de Señal/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , p-Aminoazobenceno/análogos & derivados , p-Aminoazobenceno/farmacología
19.
Neuroreport ; 21(10): 731-5, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20531234

RESUMEN

Phosphatase and tensin homolog (PTEN) is a tumor suppressor gene that controls neural stem cell renewal and differentiation and is a potential target for regeneration in the optic nerve. Here we show that it has a critical pattern of expression in the mammalian developing auditory system. PTEN was expressed in the cochlear-vestibular ganglion at embryonic day 10.5 and then progressively in hair cells as they differentiated from the base to the apex of the cochlea. By postnatal day 7, PTEN was downregulated in hair cells and subsequently in the neurons. This very specific, transient expression pattern suggests that PTEN plays a crucial role in the differentiation of the sensory neurons and hair cells and that it is a potential therapeutic target for hearing regeneration.


Asunto(s)
Cóclea/crecimiento & desarrollo , Ganglios Sensoriales/crecimiento & desarrollo , Células Ciliadas Auditivas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Células Receptoras Sensoriales/metabolismo , Ganglio Espiral de la Cóclea/crecimiento & desarrollo , Animales , Vías Auditivas/embriología , Vías Auditivas/crecimiento & desarrollo , Vías Auditivas/metabolismo , Cóclea/embriología , Cóclea/metabolismo , Epitelio/embriología , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Ganglios Sensoriales/embriología , Ganglios Sensoriales/metabolismo , Ratones , Ratones Endogámicos ICR , Ganglio Espiral de la Cóclea/embriología , Ganglio Espiral de la Cóclea/metabolismo
20.
Clin Anat ; 23(6): 726-31, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20564350

RESUMEN

The use of superficial fibular nerve (Sfn) as a potential donor nerve in nerve grafting has been introduced. The limited availability of donor nerves has paved the way for nerve allografting. We studied the sensory portion of Sfn in 60 limbs from 30 fetuses. Three distinct patterns of the nerve were designated as Types 1, 2, and 3 by us. Type 1 (66.67%) comprised Sfn piercing fascia cruris then branching into Mdn and Idn. Type 2 (21.67%) was a pattern where Sfn penetrated deep fascia then continued undivided over the dorsum of foot. Type 3 (11.67%) was where Mdn and Idn penetrated deep fascia independently. The study provided quantitative measurement data of the sensory portion of Sfn and its branching nerves with respect to osseous landmarks like the head of fibula and the malleoli. Such data may be of help in defining nerve segments suitable for harvesting in nerve grafts from fetuses.


Asunto(s)
Trasplante de Tejido Fetal/métodos , Feto/cirugía , Pie/embriología , Ganglios Sensoriales/embriología , Nervio Peroneo/embriología , Nervio Peroneo/trasplante , Femenino , Pie/inervación , Edad Gestacional , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...