Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Development ; 150(21)2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37938828

RESUMEN

Cranial ganglia are aggregates of sensory neurons that mediate distinct types of sensation. The statoacoustic ganglion (SAG) develops into several lobes that are spatially arranged to connect appropriately with hair cells of the inner ear. To investigate the cellular behaviours involved in the 3D organization of the SAG, we use high-resolution confocal imaging of single-cell, labelled zebrafish neuroblasts (NBs), photoconversion, photoablation, and genetic perturbations. We show that otic NBs delaminate out of the otic epithelium in an epithelial-mesenchymal transition-like manner, rearranging apical polarity and primary cilia proteins. We also show that, once delaminated, NBs require RhoGTPases in order to perform active migration. Furthermore, tracking of recently delaminated NBs revealed their directed migration and coalescence around a small population of pioneer SAG neurons. These pioneer SAG neurons, not from otic placode origin, populate the coalescence region before otic neurogenesis begins and their ablation disrupts delaminated NB migratory pathways, consequentially affecting SAG shape. Altogether, this work shows for the first time the role of pioneer SAG neurons in orchestrating SAG development.


Asunto(s)
Oído Interno , Pez Cebra , Animales , Pez Cebra/genética , Diferenciación Celular/genética , Oído Interno/metabolismo , Células Ciliadas Auditivas/fisiología , Células Receptoras Sensoriales
2.
Dev Dyn ; 252(1): 81-103, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35972036

RESUMEN

Sensory neurons of the head are the ones that transmit the information about the external world to our brain for its processing. Axons from cranial sensory neurons sense different chemoattractant and chemorepulsive molecules during the journey and in the target tissue to establish the precise innervation with brain neurons and/or receptor cells. Here, we aim to unify and summarize the available information regarding molecular mechanisms guiding the different afferent sensory axons of the head. By putting the information together, we find the use of similar guidance cues in different sensory systems but in distinct combinations. In vertebrates, the number of genes in each family of guidance cues has suffered a great expansion in the genome, providing redundancy, and robustness. We also discuss recently published data involving the role of glia and mechanical forces in shaping the axon paths. Finally, we highlight the remaining questions to be addressed in the field.


Asunto(s)
Orientación del Axón , Axones , Animales , Axones/fisiología , Células Receptoras Sensoriales , Neuroglía , Órganos de los Sentidos
3.
Mol Cell ; 58(5): 755-66, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25959397

RESUMEN

Protein function is often regulated and controlled by posttranslational modifications, such as oxidation. Although oxidation has been mainly considered to be uncontrolled and nonenzymatic, many enzymatic oxidations occur on enzyme-selected lysine residues; for instance, LOXL2 oxidizes lysines by converting the ε-amino groups into aldehyde groups. Using an unbiased proteomic approach, we have identified methylated TAF10, a member of the TFIID complex, as a LOXL2 substrate. LOXL2 oxidation of TAF10 induces its release from its promoters, leading to a block in TFIID-dependent gene transcription. In embryonic stem cells, this results in the inactivation of the pluripotency genes and loss of the pluripotent capacity. During zebrafish development, the absence of LOXL2 resulted in the aberrant overexpression of the neural progenitor gene Sox2 and impaired neural differentiation. Thus, lysine oxidation of the transcription factor TAF10 is a controlled protein modification and demonstrates a role for protein oxidation in regulating pluripotency genes.


Asunto(s)
Aminoácido Oxidorreductasas/fisiología , Diferenciación Celular , Células-Madre Neurales/fisiología , Procesamiento Proteico-Postraduccional , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/fisiología , Animales , Epigénesis Genética , Células HEK293 , Humanos , Metilación , Oxidación-Reducción , Factor de Transcripción TFIID/metabolismo , Pez Cebra
4.
Semin Cell Dev Biol ; 65: 47-59, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27686400

RESUMEN

The vertebrate inner ear is a precision sensory organ, acting as both a microphone to receive sound and an accelerometer to detect gravity and motion. It consists of a series of interlinked, fluid-filled chambers containing patches of sensory epithelia, each with a specialised function. The ear contains many different differentiated cell types with distinct morphologies, from the flask-shaped hair cells found in thickened sensory epithelium, to the thin squamous cells that contribute to non-sensory structures, such as the semicircular canal ducts. Nearly all cell types of the inner ear, including the afferent neurons that innervate it, are derived from the otic placode, a region of cranial ectoderm that develops adjacent to the embryonic hindbrain. As the ear develops, the otic epithelia grow, fold, fuse and rearrange to form the complex three-dimensional shape of the membranous labyrinth. Much of our current understanding of the processes of inner ear morphogenesis comes from genetic and pharmacological manipulations of the developing ear in mouse, chicken and zebrafish embryos. These traditional approaches are now being supplemented with exciting new techniques-including force measurements and light-sheet microscopy-that are helping to elucidate the mechanisms that generate this intricate organ system.


Asunto(s)
Linaje de la Célula/genética , Ectodermo/citología , Células Epiteliales/citología , Células Ciliadas Auditivas/citología , Células Laberínticas de Soporte/citología , Organogénesis/genética , Animales , Diferenciación Celular , Movimiento Celular , Embrión de Pollo , Ectodermo/metabolismo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas/metabolismo , Células Laberínticas de Soporte/metabolismo , Ratones , Especificidad de la Especie , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra
5.
J Anat ; 232(3): 431-439, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29235648

RESUMEN

There is growing evidence of a direct influence of vasculature on the development of neurons in the brain. The development of the cranial vasculature has been well described in zebrafish but its anatomical relationship with the adjacent developing sensory ganglia has not been addressed. Here, by 3D imaging of fluorescently labelled blood vessels and sensory ganglia, we describe for the first time the spatial organization of the cranial vasculature in relation to the cranial ganglia during zebrafish development. We show that from 24 h post-fertilization (hpf) onwards, the statoacoustic ganglion (SAG) develops in direct contact with two main blood vessels, the primordial hindbrain channel and the lateral dorsal aortae (LDA). At 48 hpf, the LDA is displaced medially, losing direct contact with the SAG. The relationship of the other cranial ganglia with the vasculature is evident for the medial lateral line ganglion and for the vagal ganglia that grow along the primary head sinus (PHS). We also observed that the innervation of the anterior macula runs over the PHS vessel. Our spatiotemporal anatomical map of the cranial ganglia and the head vasculature indicates physical interactions between both systems and suggests a possible functional interaction during development.


Asunto(s)
Vasos Sanguíneos/embriología , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Nervios Craneales/irrigación sanguínea , Pez Cebra/embriología , Animales , Nervios Craneales/embriología , Ganglios/irrigación sanguínea , Ganglios/embriología
6.
J Neurosci ; 35(47): 15752-66, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26609166

RESUMEN

During development, otic sensory progenitors give rise to hair cells and supporting cells. In mammalian adults, differentiated and quiescent sensory cells are unable to generate new hair cells when these are lost due to various insults, leading to irreversible hearing loss. Retinoic acid (RA) has strong regenerative capacity in several organs, but its role in hair cell regeneration is unknown. Here, we use genetic and pharmacological inhibition to show that the RA pathway is required for hair cell regeneration in zebrafish. When regeneration is induced by laser ablation in the inner ear or by neomycin treatment in the lateral line, we observe rapid activation of several components of the RA pathway, with dynamics that position RA signaling upstream of other signaling pathways. We demonstrate that blockade of the RA pathway impairs cell proliferation of supporting cells in the inner ear and lateral line. Moreover, in neuromast, RA pathway regulates the transcription of p27(kip) and sox2 in supporting cells but not fgf3. Finally, genetic cell-lineage tracing using Kaede photoconversion demonstrates that de novo hair cells derive from FGF-active supporting cells. Our findings reveal that RA has a pivotal role in zebrafish hair cell regeneration by inducing supporting cell proliferation, and shed light on the underlying transcriptional mechanisms involved. This signaling pathway might be a promising approach for hearing recovery. SIGNIFICANCE STATEMENT: Hair cells are the specialized mechanosensory cells of the inner ear that capture auditory and balance sensory input. Hair cells die after acoustic trauma, ototoxic drugs or aging diseases, leading to progressive hearing loss. Mammals, in contrast to zebrafish, lack the ability to regenerate hair cells. Here, we find that retinoic acid (RA) pathway is required for hair cell regeneration in vivo in the zebrafish inner ear and lateral line. RA pathway is activated very early upon hair cell loss, promotes cell proliferation of progenitor cells, and regulates two key genes, p27(kip) and sox2. Our results position RA as an essential signal for hair cell regeneration with relevance in future regenerative strategies in mammals.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Ciliadas Auditivas/metabolismo , Regeneración Nerviosa/fisiología , Factores de Transcripción SOX/metabolismo , Transducción de Señal/fisiología , Tretinoina/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Femenino , Masculino , Factores de Transcripción SOX/antagonistas & inhibidores , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores
7.
Dev Biol ; 389(1): 50-67, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24508480

RESUMEN

For both the intricate morphogenetic layout of the sensory cells in the ear and the elegantly radial arrangement of the sensory neurons in the nose, numerous signaling molecules and genetic determinants are required in concert to generate these specialized neuronal populations that help connect us to our environment. In this review, we outline many of the proteins and pathways that play essential roles in the differentiation of otic and olfactory neurons and their integration into their non-neuronal support structures. In both cases, well-known signaling pathways together with region-specific factors transform thickened ectodermal placodes into complex sense organs containing numerous, diverse neuronal subtypes. Olfactory and otic placodes, in combination with migratory neural crest stem cells, generate highly specialized subtypes of neuronal cells that sense sound, position and movement in space, odors and pheromones throughout our lives.


Asunto(s)
Oído Interno/embriología , Ectodermo/embriología , Neurogénesis/fisiología , Vías Olfatorias/embriología , Órganos de los Sentidos/embriología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Oído Interno/citología , Oído Interno/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Neurogénesis/genética , Vías Olfatorias/citología , Vías Olfatorias/metabolismo , Órganos de los Sentidos/citología , Órganos de los Sentidos/metabolismo , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo
8.
J Cell Sci ; 126(Pt 1): 53-9, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23178937

RESUMEN

In a genome-wide expression profile search for genes required for Drosophila R7 photoreceptor development we found ß amyloid protein precursor-like (Appl), the ortholog of human APP, which is a key factor in the pathogenesis of Alzheimer's disease. We analyzed Appl expression in the eye imaginal disc and found that is highly accumulated in R7 photoreceptor cells. The R7 photoreceptor is responsible for UV light detection. To explore the link between high expression of Appl and R7 function, we have analyzed Appl null mutants and found reduced preference for UV light, probably because of mistargeted R7 axons. Moreover, axon mistargeting and inappropriate light discrimination are enhanced in combination with neurotactin mutants. R7 differentiation is triggered by the inductive interaction between R8 and R7 precursors, which results in a burst of Ras1/MAPK, activated by the tyrosine kinase receptor Sevenless. Therefore, we examined whether Ras1/MAPK is responsible for the high Appl expression. Inhibition of Ras1 signaling leads to reduced Appl expression, whereas constitutive activation drives ectopic Appl expression. We show that Appl is directly regulated by the Ras/MAPK pathway through a mechanism mediated by PntP2, an ETS transcription factor that specifically binds ETS sites in the Appl regulatory region. We also found that zebrafish appb expression increased after ectopic fgfr activation in the neural tube of zebrafish embryos, suggesting a conserved regulatory mechanism.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Fotorreceptoras/citología , Células Fotorreceptoras/metabolismo , Proteínas ras/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas ras/genética
9.
Development ; 138(3): 397-408, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21205785

RESUMEN

Proper spatial control of neurogenesis in the inner ear ensures the precise innervation of mechanotransducing cells and the propagation of auditory and equilibrium stimuli to the brain. Members of the Hairy and enhancer of split (Hes) gene family regulate neurogenesis by inhibiting neuronal differentiation and maintaining neural stem cell pools in non-neurogenic zones. Remarkably, their role in the spatial control of neurogenesis in the ear is unknown. In this study, we identify her9, a zebrafish ortholog of Hes1, as a key gene in regulating otic neurogenesis through the definition of the posterolateral non-neurogenic field. First, her9 emerges as a novel otic patterning gene that represses proneural function and regulates the extent of the neurogenic domain. Second, we place Her9 downstream of Tbx1, linking these two families of transcription factors for the first time in the inner ear and suggesting that the reported role of Tbx1 in repressing neurogenesis is in part mediated by the bHLH transcriptional repressor Her9. Third, we have identified retinoic acid (RA) signaling as the upstream patterning signal of otic posterolateral genes such as tbx1 and her9. Finally, we show that at the level of the cranial otic field, opposing RA and Hedgehog signaling position the boundary between the neurogenic and non-neurogenic compartments. These findings permit modeling of the complex genetic cascade that underlies neural patterning of the otic vesicle.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Proteínas de Dominio T Box/metabolismo , Tretinoina/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proliferación Celular/efectos de los fármacos , Oído Interno , Embrión no Mamífero/efectos de los fármacos , Inmunohistoquímica , Hibridación in Situ , Proteínas de Dominio T Box/genética , Tretinoina/farmacología , Pez Cebra , Proteínas de Pez Cebra/genética
10.
Cell Rep ; 37(7): 110004, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34788624

RESUMEN

Polyphosphate (polyP) is a polymer of hundreds of phosphate residues present in all organisms. In mammals, polyP is involved in crucial physiological processes, including coagulation, inflammation, and stress response. However, after decades of research, the metabolic enzymes are still unknown. Here, we purify and identify Nudt3, a NUDIX family member, as the enzyme responsible for polyP phosphatase activity in mammalian cells. We show that Nudt3 shifts its substrate specificity depending on the cation; specifically, Nudt3 is active on polyP when Zn2+ is present. Nudt3 has in vivo polyP phosphatase activity in human cells, and importantly, we show that cells with altered polyP levels by modifying Nudt3 protein amount present reduced viability upon oxidative stress and increased DNA damage, suggesting that polyP and Nudt3 play a role in oxidative stress protection. Finally, we show that Nudt3 is involved in the early stages of embryo development in zebrafish.


Asunto(s)
Ácido Anhídrido Hidrolasas/metabolismo , Estrés Oxidativo/fisiología , Polifosfatos/metabolismo , Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/fisiología , Animales , Células HEK293 , Humanos , Masculino , Mamíferos/metabolismo , Oxidación-Reducción , Monoéster Fosfórico Hidrolasas/fisiología , Ratas , Ratas Sprague-Dawley , Especificidad por Sustrato/fisiología , Pez Cebra , Zinc/metabolismo
11.
Hum Genet ; 128(4): 411-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668882

RESUMEN

POU3F4 encodes a POU-domain transcription factor required for inner ear development. Defects in POU3F4 function are associated with X-linked deafness type 3 (DFN3). Multiple deletions affecting up to ~900-kb upstream of POU3F4 are found in DFN3 patients, suggesting the presence of essential POU3F4 enhancers in this region. Recently, an inner ear enhancer was reported that is absent in most DFN3 patients with upstream deletions. However, two indications suggest that additional enhancers in the POU3F4 upstream region are required for POU3F4 function during inner ear development. First, there is at least one DFN3 deletion that does not eliminate the reported enhancer. Second, the expression pattern driven by this enhancer does not fully recapitulate Pou3f4 expression in the inner ear. Here, we screened a 1-Mb region upstream of the POU3F4 gene for additional cis-regulatory elements and searched for novel DFN3 mutations in the identified POU3F4 enhancers. We found several novel enhancers for otic vesicle expression. Some of these also drive expression in kidney, pancreas and brain, tissues that are known to express Pou3f4. In addition, we report a new and smallest deletion identified so far in a DFN3 family which eliminates 3.9 kb, comprising almost exclusively the previous reported inner ear enhancer. We suggest that multiple enhancers control the expression of Pou3f4 in the inner ear and these may contribute to the phenotype observed in DFN3 patients. In addition, the novel deletion demonstrates that the previous reported enhancer, although not sufficient, is essential for POU3F4 function during inner ear development.


Asunto(s)
Región de Flanqueo 5'/genética , Oído Interno/metabolismo , Elementos de Facilitación Genéticos/genética , Audición/genética , Factores del Dominio POU/genética , Animales , Secuencia de Bases , Análisis Mutacional de ADN , Oído Interno/crecimiento & desarrollo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Salud de la Familia , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Pérdida Auditiva/genética , Humanos , Hibridación in Situ , Masculino , Microscopía Fluorescente , Linaje , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Xenopus/embriología , Xenopus/genética
12.
Curr Opin Cell Biol ; 67: 79-85, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32950922

RESUMEN

Vertebrates sense a large variety of sensory stimuli that ranges from temperature, volatile and nonvolatile chemicals, touch, pain, light, sound and gravity. To achieve this, they use specialized cells present in sensory organs and cranial ganglia. Much of our understanding of the transcription factors and mechanisms responsible for sensory cell specification comes from cell-lineage tracing and genetic experiments in different species, but recent advances in single-cell transcriptomics, high-resolution imaging and systems biology approaches have allowed to study these processes in an unprecedented resolution. Here I will point to the transcription factor programs driving cell diversity in the different sensory organs of vertebrates to then discuss in vivo data of how cell specification is coupled with tissue morphogenesis.


Asunto(s)
Diferenciación Celular , Cráneo/citología , Vertebrados/fisiología , Animales , Linaje de la Célula , Reprogramación Celular , Humanos , Morfogénesis
13.
Cell Rep ; 32(2): 107903, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668260

RESUMEN

In many organs, stem cell function depends on communication with their niche partners. Cranial sensory neurons develop in close proximity to blood vessels; however, whether vasculature is an integral component of their niches is yet unknown. Here, two separate roles for vasculature in cranial sensory neurogenesis in zebrafish are uncovered. The first involves precise spatiotemporal endothelial-neuroblast cytoneme contacts and Dll4-Notch signaling to restrain neuroblast proliferation. The second, instead, requires blood flow to trigger a transcriptional response that modifies neuroblast metabolic status and induces sensory neuron differentiation. In contrast, no role of sensory neurogenesis in vascular development is found, suggesting unidirectional signaling from vasculature to sensory neuroblasts. Altogether, we demonstrate that the cranial vasculature constitutes a niche component of the sensory ganglia that regulates the pace of their growth and differentiation dynamics.


Asunto(s)
Circulación Sanguínea/fisiología , Vasos Sanguíneos/citología , Ciclo Celular , Diferenciación Celular , Células Receptoras Sensoriales/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Circulación Sanguínea/efectos de los fármacos , Tipificación del Cuerpo/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Recuento de Células , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Oxígeno/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Receptores Notch/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Transducción de Señal/efectos de los fármacos , Cráneo/irrigación sanguínea , Tiazolidinas/farmacología , Transcripción Genética/efectos de los fármacos , Nervio Vestibulococlear/citología , Nervio Vestibulococlear/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
14.
Dev Biol ; 322(1): 109-20, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18674529

RESUMEN

The otic placode generates the auditory and vestibular sense organs and their afferent neurons; however, how auditory and vestibular fates are specified is unknown. We have generated a fate map of the otic placode and show that precursors for vestibular and auditory cells are regionally segregated in the otic epithelium. The anterior-lateral portion of the otic placode generates vestibular neurons, whereas the posterior-medial region gives rise to auditory neurons. Precursors for vestibular and auditory sense organs show the same distribution. Thus, different regions of the otic placode correspond to particular sense organs and their innervating neurons. Neurons from contiguous domains rarely intermingle suggesting that the regional organisation of the otic placode dictates positional cues to otic neurons. But, in addition, vestibular and cochlear neurogenesis also follows a stereotyped temporal pattern. Precursors from the anterior-lateral otic placode delaminate earlier than those from its medial-posterior portion. The expression of the proneural genes NeuroM and NeuroD reflects the sequence of neuroblast formation and differentiation. Both genes are transiently expressed in vestibular and then in cochlear neuroblasts, while differentiated neurons express Islet1, Tuj1 and TrkC, but not NeuroM or NeuroD. Together, our results indicate that the position of precursors within the otic placode confers identity to sensory organs and to the corresponding otic neurons. In addition, positional information is integrated with temporal cues that coordinate neurogenesis and sensory differentiation.


Asunto(s)
Cóclea/embriología , Neuronas Aferentes/citología , Vestíbulo del Laberinto/embriología , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Proteínas Aviares/biosíntesis , Proteínas Aviares/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/fisiología , Linaje de la Célula , Movimiento Celular/fisiología , Embrión de Pollo , Cóclea/citología , Cóclea/inervación , Epitelio/embriología , Epitelio/inervación , Colorantes Fluorescentes , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas Aferentes/fisiología , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Células Madre/citología , Células Madre/metabolismo , Vestíbulo del Laberinto/citología , Vestíbulo del Laberinto/inervación
15.
Mech Dev ; 124(7-8): 631-45, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17532192

RESUMEN

Otic neuronal precursors are the first cells to be specified and do so in the anterior domain of the otic placode, the proneural domain. In the present study, we have explored the early events of otic proneural regionalization in relation to the activity of the Notch signaling pathway. The proneural domain was characterized by the expression of Sox3, Fgf10 and members of the Notch pathway such as Delta1, Hes5 and Lunatic Fringe. The complementary non-neural domain expressed two patterning genes, Lmx1b and Iroquois1, and the members of the Notch pathway, Serrate1 and Hairy1. Fate map studies and double injections with DiI/DiO showed that labeled cells remained confined to anterior or posterior territories with limited cell intermingling. To explore whether Notch signaling pathway plays a role in the initial regionalization of the otic placode, Notch activity was blocked by a gamma-secretase inhibitor (DAPT). Notch blockade induced the expansion of non-neural genes, Lmx1 and Iroquois1, into the proneural domain. Combined gene expression and DiI experiments showed that these effects were not due to migration of non-neural cells into the proneural domain, suggesting that Notch activity regulates the expression of non-neural genes. This was further confirmed by the electroporation of a dominant-negative form of the Mastermind-like1 gene that caused the up-regulation of Lmx1 within the proneural domain. In addition, Notch pathway was involved in neuronal precursor selection, probably by a classical mechanism of lateral inhibition. We propose that the regionalization of the otic domain into a proneural and a non-neural territory is a very early event in otic development, and that Notch signaling activity is required to exclude the expression of non-neural genes from the proneural territory.


Asunto(s)
Oído Interno/embriología , Receptores Notch/fisiología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Tipificación del Cuerpo , Embrión de Pollo , Oído Interno/metabolismo , Regulación del Desarrollo de la Expresión Génica , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Triglicéridos/farmacología , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/farmacología
16.
Int J Dev Biol ; 51(6-7): 483-93, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17891711

RESUMEN

Hair-cells, supporting cells and sensory neurons are the main specialized cell-types responsible for mechanotransduction in the inner ear. They derive from precursors expressing proneural genes and recent data has underlined the importance of SoxB1 genes as upstream activators of proneural genes during cranial placode development. Here we review the steps of establishing a proneural field and propose several models for how early otic regionalization into a proneural territory is achieved.


Asunto(s)
Oído Interno/citología , Oído Interno/embriología , Animales , Oído Interno/metabolismo , Embrión no Mamífero/metabolismo , Inducción Embrionaria , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas/metabolismo , Células Laberínticas de Soporte/citología , Células Laberínticas de Soporte/metabolismo , Mecanotransducción Celular , Modelos Biológicos , Neuronas Aferentes/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
J Comp Neurol ; 503(4): 487-500, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17534940

RESUMEN

The generation of the mechanosensory elements of the inner ear during development proceeds in a precise temporal and spatial pattern. First, neurosensory precursors form sensory neurons. Then, prosensory patches emerge and give rise to hair and supporting cells. Hair cells are innervated by cochleovestibular neurons that convey sound and balance information to the brain. SOX2 is an HMG transcription factor characteristic of the stem-cell genetic network responsible for progenitor self-renewal and commitment, and its loss of function generates defects in ear sensory epithelia. The present study shows that SOX2 protein is expressed in a spatially and temporally restricted manner throughout development of the chick inner ear. SOX2 is first expressed in the neurogenic region that gives rise to sensory neurons. SOX2 is then restricted to the prosensory patches in E4 and E5 embryos, as revealed by double and parallel labelling with SOX2 and Tuj1, MyoVIIa, or Islet1. Proliferating cell nuclear antigen labelling showed that SOX2 is expressed in proliferating cells during those stages. By E5, SOX2 is also expressed in the Schwann cells of the cochleovestibular ganglion, but not in the otic neurons. At E8 and E17, beyond stages of sensory cell specification, SOX2 is transiently expressed in hair cells, but its level remains high in supporting cells. SOX3 is concomitantly expressed with SOX2 in the neurogenic domain of the otic cup, but not in prosensory patches. Our data are consistent with a role for SOX2 in specifying a population of otic progenitors committed to a neural fate, giving rise to neurons and hair cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Oído Interno , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas HMGB/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Neuronas Aferentes/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Diferenciación Celular , Embrión de Pollo , Proteínas de Unión al ADN/genética , Oído Interno/citología , Oído Interno/embriología , Oído Interno/metabolismo , Proteínas HMGB/genética , Proteínas del Grupo de Alta Movilidad/genética , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Modelos Anatómicos , Proteínas del Tejido Nervioso/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Transcripción SOXB1 , Factores de Transcripción/genética
19.
Elife ; 62017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28537554

RESUMEN

Neural patterning involves regionalised cell specification. Recent studies indicate that cell dynamics play instrumental roles in neural pattern refinement and progression, but the impact of cell behaviour and morphogenesis on neural specification is not understood. Here we combine 4D analysis of cell behaviours with dynamic quantification of proneural expression to uncover the construction of the zebrafish otic neurogenic domain. We identify pioneer cells expressing neurog1 outside the otic epithelium that migrate and ingress into the epithelialising placode to become the first otic neuronal progenitors. Subsequently, neighbouring cells express neurog1 inside the placode, and apical symmetric divisions amplify the specified pool. Interestingly, pioneer cells delaminate shortly after ingression. Ablation experiments reveal that pioneer cells promote neurog1 expression in other otic cells. Finally, ingression relies on the epithelialisation timing controlled by FGF activity. We propose a novel view for otic neurogenesis integrating cell dynamics whereby ingression of pioneer cells instructs neuronal specification.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Oído/embriología , Epitelio/embriología , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/embriología , Células Neuroepiteliales/fisiología , Neurogénesis , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Tipificación del Cuerpo , Movimiento Celular
20.
Nat Commun ; 6: 7355, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26077034

RESUMEN

Many organ functions rely on epithelial cavities with particular shapes. Morphogenetic anomalies in these cavities lead to kidney, brain or inner ear diseases. Despite their relevance, the mechanisms regulating lumen dimensions are poorly understood. Here, we perform live imaging of zebrafish inner ear development and quantitatively analyse the dynamics of lumen growth in 3D. Using genetic, chemical and mechanical interferences, we identify two new morphogenetic mechanisms underlying anisotropic lumen growth. The first mechanism involves thinning of the epithelium as the cells change their shape and lose fluids in concert with expansion of the cavity, suggesting an intra-organ fluid redistribution process. In the second mechanism, revealed by laser microsurgery experiments, mitotic rounding cells apicobasally contract the epithelium and mechanically contribute to expansion of the lumen. Since these mechanisms are axis specific, they not only regulate lumen growth but also the shape of the cavity.


Asunto(s)
Forma de la Célula , Oído Interno/embriología , Células Epiteliales/citología , Mitosis , Animales , Oído Interno/citología , Embrión no Mamífero , Imagenología Tridimensional , Líquido Intracelular/metabolismo , Organogénesis , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA