Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Development ; 150(19)2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37791525

RESUMO

Our molecular understanding of the early stages of human inner ear development has been limited by the difficulty in accessing fetal samples at early gestational stages. As an alternative, previous studies have shown that inner ear morphogenesis can be partially recapitulated using induced pluripotent stem cells directed to differentiate into inner ear organoids (IEOs). Once validated and benchmarked, these systems could represent unique tools to complement and refine our understanding of human otic differentiation and model developmental defects. Here, we provide the first direct comparisons of the early human embryonic otocyst and fetal sensory organs with human IEOs. We use multiplexed immunostaining and single-cell RNA-sequencing to characterize IEOs at three key developmental steps, providing a new and unique signature of in vitro-derived otic placode, epithelium, neuroblasts and sensory epithelia. In parallel, we evaluate the expression and localization of crucial markers at these equivalent stages in human embryos. Together, our data indicate that the current state-of-the-art protocol enables the specification of bona fide otic tissue, supporting the further application of IEOs to inform inner ear biology and disease.


Assuntos
Orelha Interna , Células-Tronco Pluripotentes , Humanos , Gravidez , Feminino , Epitélio/metabolismo , Diferenciação Celular , Organoides
2.
Development ; 146(13)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31152002

RESUMO

The transcription factor sex determining region Y-box 2 (SOX2) is required for the formation of hair cells and supporting cells in the inner ear and is a widely used sensory marker. Paradoxically, we demonstrate via fate mapping that, initially, SOX2 primarily marks nonsensory progenitors in the mouse cochlea, and is not specific to all sensory regions until late otic vesicle stages. SOX2 fate mapping reveals an apical-to-basal gradient of SOX2 expression in the sensory region of the cochlea, reflecting the pattern of cell cycle exit. To understand SOX2 function, we undertook a timed-deletion approach, revealing that early loss of SOX2 severely impaired morphological development of the ear, whereas later deletions resulted in sensory disruptions. During otocyst stages, SOX2 shifted dramatically from a lateral to medial domain over 24-48 h, reflecting the nonsensory-to-sensory switch observed by fate mapping. Early loss or gain of SOX2 function led to changes in otic epithelial volume and progenitor proliferation, impacting growth and morphological development of the ear. Our study demonstrates a novel role for SOX2 in early otic morphological development, and provides insights into the temporal and spatial patterns of sensory specification in the inner ear.


Assuntos
Cóclea/embriologia , Orelha Interna/embriologia , Células Ciliadas Auditivas/fisiologia , Morfogênese/genética , Fatores de Transcrição SOXB1/fisiologia , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Cóclea/citologia , Orelha Interna/crescimento & desenvolvimento , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Feminino , Células Ciliadas Auditivas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Gravidez , Fatores de Transcrição SOXB1/genética , Fatores de Tempo
3.
Dev Biol ; 467(1-2): 39-50, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32891623

RESUMO

The Six1 transcription factor plays a major role in craniofacial development. Mutations in SIX1 and its co-factor, EYA1, are causative for about 50% of Branchio-otic/Branchio-oto-renal syndrome (BOR) patients, who are characterized by variable craniofacial, otic and renal malformations. We previously screened for other proteins that might interact with Six1 to identify additional genes that may play a role in BOR, and herein characterize the developmental role of one of them, Microspherule protein 1 (Mcrs1). We found that in cultured cells, Mcrs1 bound to Six1 and in both cultured cells and embryonic ectoderm reduced Six1-Eya1 transcriptional activation. Knock-down of Mcrs1 in embryos caused an expansion of the domains of neural plate genes and two genes expressed in both the neural plate and neural crest (zic1, zic2). In contrast, two other genes expressed in pre-migratory neural crest (foxd3, sox9) were primarily reduced. Cranial placode genes showed a mixture of expanded and diminished expression domains. At larval stages, loss of Mcrs1 resulted in a significant reduction of otic vesicle gene expression concomitant with a smaller otic vesicle volume. Experimentally increasing Mcrs1 above endogenous levels favored the expansion of neural border and neural crest gene domains over cranial placode genes; it also reduced otic vesicle gene expression but not otic vesicle volume. Co-expression of Mcrs1 and Six1 as well as double knock-down and rescue experiments establish a functional interaction between Mcrs1 and Six1 in the embryo, and demonstrate that this interaction has an important role in the development of craniofacial tissues including the otic vesicle.


Assuntos
Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Ligação a RNA/biossíntese , Crânio/embriologia , Proteínas de Xenopus/biossíntese , Animais , Ectoderma/embriologia , Crista Neural/embriologia , Xenopus laevis
4.
Development ; 145(24)2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30504125

RESUMO

Morphogenesis of the inner ear epithelium requires coordinated deployment of several signaling pathways, and disruptions cause abnormalities of hearing and/or balance. The FGFR2b ligands FGF3 and FGF10 are expressed throughout otic development and are required individually for normal morphogenesis, but their prior and redundant roles in otic placode induction complicates investigation of subsequent combinatorial functions in morphogenesis. To interrogate these roles and identify new effectors of FGF3 and FGF10 signaling at the earliest stages of otic morphogenesis, we used conditional gene ablation after otic placode induction, and temporal inhibition of signaling with a secreted, dominant-negative FGFR2b ectodomain. We show that both ligands are required continuously after otocyst formation for maintenance of otic neuroblasts and for patterning and proliferation of the epithelium, leading to normal morphogenesis of both the cochlear and vestibular domains. Furthermore, the first genome-wide identification of proximal targets of FGFR2b signaling in the early otocyst reveals novel candidate genes for inner ear development and function.


Assuntos
Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Morfogênese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Linhagem da Célula , Proliferação de Células , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Doxiciclina/farmacologia , Feminino , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 3 de Crescimento de Fibroblastos/metabolismo , Cistos Glanglionares/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Integrases/metabolismo , Ligantes , Masculino , Camundongos , Mutação/genética , Neurônios/citologia , Neurônios/metabolismo , Fator de Transcrição PAX2/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Vestíbulo do Labirinto/crescimento & desenvolvimento , Vestíbulo do Labirinto/metabolismo
5.
Dev Dyn ; 249(7): 867-883, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32384225

RESUMO

BACKGROUND: Sensorineural hearing loss is an understudied consequence of congenital Zika syndrome, and balance disorders are essentially unreported to date. Also lacking is information about the susceptibility and the pathogenesis of the developing inner ear following Zika virus (ZIKV) exposure. To address this, ZIKV was delivered directly into the otic cup/otocyst of chicken embryos and infection of inner ear tissues was evaluated using immunohistochemistry. RESULTS: After injections on embryonic days 2 to 5, ZIKV infection was observed in 90% of the samples harvested 2 to 8 days later; however, the degree of infection was highly variable across individuals. ZIKV was detected in all regions of the inner ear, associated ganglia, and in the surrounding periotic mesenchyme. Detection of virus peaked earlier in the ganglion and vestibular compartments, and later in the cochlea. ZIKV infection increased cell death robustly in the auditory ganglion, and modestly in the auditory sensory organ. Macrophage accumulation was found to overlap with dense viral infection in some tissues. Additionally, dysmorphogenesis of the semicircular canals and ganglion was observed for a subset of injection conditions. CONCLUSIONS: This article presents evidence of direct ZIKV infection of developing inner ear epithelium and shows previously unknown inner ear dysmorphogenesis phenotypes.


Assuntos
Orelha Interna/embriologia , Orelha Interna/virologia , Perda Auditiva Neurossensorial/embriologia , Infecção por Zika virus/virologia , Zika virus/metabolismo , Animais , Morte Celular , Embrião de Galinha , Galinhas , Cóclea , Orelha Interna/metabolismo , Epitélio/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Macrófagos/metabolismo , Fenótipo , Canais Semicirculares/embriologia , Canais Semicirculares/metabolismo , Fatores de Tempo , Infecção por Zika virus/metabolismo , Infecção por Zika virus/patologia
6.
Biochem Biophys Res Commun ; 526(2): 375-380, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32222280

RESUMO

Receptor tyrosine kinase EphA7 is specifically expressed in otic region in Xenopus early development. However, its role in otocyst development remains unknown. Knockdown of EphA7 by a specific morpholino oligonucleotide (MO) reduced the size of the otocyst and triggered otic epithelial cell extrusion. Interestingly, EphA7 depletion attenuated the membrane level of the tight junction protein Claudin6 (CLDN6). Utilizing the Cldn6 MO, we further confirmed that CLDN6 attenuation also led to otic epithelial cell extrusion. Our work suggested that EphA7 modulates the otic epithelial homeostasis through stabilizing the CLDN6 membrane level.


Assuntos
Claudinas/genética , Regulação da Expressão Gênica no Desenvolvimento , Receptor EphA7/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriologia , Animais , Técnicas de Silenciamento de Genes , Homeostase , Xenopus laevis/genética
7.
J Neurophysiol ; 122(6): 2272-2283, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577516

RESUMO

Children with congenital vestibular disorders show delayed motor development and challenges in maintaining posture and balance. Computed tomography images reveal that these children have abnormal inner ears in the form of a sac, with the semicircular canals missing or truncated. Little is known about how this inner ear abnormality affects central vestibular development. At present, mice with the chromodomain helicase DNA-binding protein 7 mutation are the most common model for studying congenital vestibular disorders, despite forming multiple diverse inner ear phenotypes and inducing abnormal cerebellar and visual system development. To identify the effects of a sac-like inner ear on central vestibular development, we have designed and implemented a new model, the anterior-posterior axis rotated otocyst (ARO) chick, which forms a sac-like inner ear in 85% of cases. The ARO chick is produced by anterior-posterior rotation of the otocyst at embryonic day 2. Here, we describe for the first time the 15% of ARO chicks that form three small semicircular canals and rename the ARO chicks forming sacs (ARO/s chicks). The basic features of the vestibular sensory organs in ARO/s chicks are similar to those found in patients' sacs, and ARO/s hatchlings experience balance and walking problems like patients. Thus, ARO/s chicks have a reproducible inner ear phenotype without abnormalities in vestibular-related structures, making the model a relatively simple one to evaluate the relationship between the sac-like inner ear pathology and formation of the central vestibular neural circuitry. Here, we describe unpublished details on the surgical approaches to produce ARO chicks, including pitfalls and difficulties to avoid.NEW & NOTEWORTHY This paper describes simple techniques for chick otocyst rotation resulting in a sac-like inner ear (85%), the common phenotype in congenital vestibular disorders. We now describe anterior-posterior axis rotated otocyst chicks, which form three small canals (15%), and rename chicks forming a sac (ARO/s chicks). Basic protocols and potential complications of otocyst rotation are described. With the use of ARO/s chicks, it will be possible to determine how the vestibular neural circuit is modified by sac-like inner ear formation.


Assuntos
Orelha Interna/patologia , Doenças Vestibulares/congênito , Doenças Vestibulares/patologia , Doenças Vestibulares/fisiopatologia , Animais , Embrião de Galinha , Modelos Animais de Doenças
8.
Development ; 143(12): 2228-37, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27151948

RESUMO

The inner ear consists of two otocyst-derived, structurally and functionally distinct components: the dorsal vestibular and ventral auditory compartments. BMP signaling is required to form the vestibular compartment, but how it complements other required signaling molecules and acts intracellularly is unknown. Using spatially and temporally controlled delivery of signaling pathway regulators to developing chick otocysts, we show that BMP signaling regulates the expression of Dlx5 and Hmx3, both of which encode transcription factors essential for vestibular formation. However, although BMP regulates Dlx5 through the canonical SMAD pathway, surprisingly, it regulates Hmx3 through a non-canonical pathway involving both an increase in cAMP-dependent protein kinase A activity and the GLI3R to GLI3A ratio. Thus, both canonical and non-canonical BMP signaling establish the precise spatiotemporal expression of Dlx5 and Hmx3 during dorsal vestibular development. The identification of the non-canonical pathway suggests an intersection point between BMP and SHH signaling, which is required for ventral auditory development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Orelha Interna/embriologia , Orelha Interna/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Animais , Galinhas , Cóclea/embriologia , Cóclea/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Modelos Biológicos , Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição Otx/metabolismo , Processamento de Proteína Pós-Traducional , Canais Semicirculares/embriologia , Canais Semicirculares/metabolismo , Proteínas Smad/metabolismo , Proteína Gli3 com Dedos de Zinco
9.
Dev Biol ; 420(1): 100-109, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27720745

RESUMO

During development of the inner ear, secreted morphogens act coordinately to establish otocyst dorsoventral polarity. Among these, Sonic hedgehog (SHH) plays a critical role in determining ventral polarity. However, how this extracellular signal is transduced intracellularly to establish ventral polarity is unknown. In this study, we show that cAMP dependent protein kinase A (PKA) is a key intracellular factor mediating SHH signaling through regulation of GLI3 processing. Gain-of-function experiments using targeted gene transfection by sonoporation or electroporation revealed that SHH signaling inactivates PKA, maintaining a basal level of PKA activity in the ventral otocyst. This, in turn, suppresses partial proteolytic processing of GLI3FL, resulting in a low GLI3R/GLI3FL ratio in the ventral otocyst and the expression of ventral-specific genes required for ventral otocyst morphogenesis. Thus, we identify a molecular mechanism that links extracellular and intracellular signaling, determines early ventral polarity of the inner ear, and has implications for understanding the integration of polarity signals in multiple organ rudiments regulated by gradients of signaling molecules.


Assuntos
Padronização Corporal , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Orelha Interna/metabolismo , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Transdução de Sinais , Animais , Polaridade Celular , Galinhas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Orelha Interna/citologia , Mesoderma/metabolismo , Camundongos , Modelos Biológicos , Fosforilação
10.
Dev Biol ; 414(1): 72-84, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27090805

RESUMO

The transcription factor Sox2 is both necessary and sufficient for the generation of sensory regions of the inner ear. It regulates expression of the Notch ligand Jag1 in prosensory progenitors, which signal to neighboring cells to up-regulate Sox2 and sustain prosensory identity. However, the expression pattern of Sox2 in the early inner ear is very broad, suggesting that Sox2-expressing progenitors form a wide variety of cell types in addition to generating the sensory regions of the ear. We used Sox2-CreER mice to follow the fates of Sox2-expressing cells at different stages in ear development. We find that Sox2-expressing cells in the early otocyst give rise to large numbers of non-sensory structures throughout the inner ear, and that Sox2 only becomes a truly prosensory marker at embryonic day (E)11.5. Our fate map reveals the organ of Corti derives from a central domain on the medial side of the otocyst and shows that a significant amount of the organ of Corti derives from a Sox2-negative population in this region.


Assuntos
Orelha Interna/citologia , Células-Tronco Neurais/citologia , Órgão Espiral/embriologia , Fatores de Transcrição SOXB1/análise , Animais , Antígenos de Diferenciação/análise , Linhagem da Célula , Orelha Interna/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Genes Reporter , Imageamento Tridimensional , Proteína Jagged-1/biossíntese , Proteína Jagged-1/genética , Proteínas Luminescentes/análise , Camundongos , Camundongos Transgênicos , Órgão Espiral/citologia , Receptores Notch/fisiologia , Transdução de Sinais/fisiologia
11.
Dev Dyn ; 244(2): 181-210, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25403746

RESUMO

BACKGROUND: Six1 plays an important role in the development of several vertebrate organs, including cranial sensory placodes, somites, and kidney. Although Six1 mutations cause one form of branchio-otic syndrome (BOS), the responsible gene in many patients has not been identified; genes that act downstream of Six1 are potential BOS candidates. RESULTS: We sought to identify novel genes expressed during placode, somite and kidney development by comparing gene expression between control and Six1-expressing ectodermal explants. The expression patterns of 19 of the significantly up-regulated and 11 of the significantly down-regulated genes were assayed from cleavage to larval stages. A total of 28/30 genes are expressed in the otocyst, a structure that is functionally disrupted in BOS, and 26/30 genes are expressed in the nephric mesoderm, a structure that is functionally disrupted in the related branchio-otic-renal (BOR) syndrome. We also identified the chick homologues of five genes and show that they have conserved expression patterns. CONCLUSIONS: Of the 30 genes selected for expression analyses, all are expressed at many of the developmental times and appropriate tissues to be regulated by Six1. Many have the potential to play a role in the disruption of hearing and kidney function seen in BOS/BOR patients.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Rim/embriologia , Crânio/embriologia , Somitos/embriologia , Proteínas de Xenopus/metabolismo , Animais , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Rim/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Crânio/citologia , Somitos/citologia , Proteínas de Xenopus/genética , Xenopus laevis
12.
Dev Biol ; 390(1): 51-67, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24583262

RESUMO

Control over ionic composition and volume of the inner ear luminal fluid endolymph is essential for normal hearing and balance. Mice deficient in either the EphB2 receptor tyrosine kinase or the cognate transmembrane ligand ephrin-B2 (Efnb2) exhibit background strain-specific vestibular-behavioral dysfunction and signs of abnormal endolymph homeostasis. Using various loss-of-function mouse models, we found that Efnb2 is required for growth and morphogenesis of the embryonic endolymphatic epithelium, a precursor of the endolymphatic sac (ES) and duct (ED), which mediate endolymph homeostasis. Conditional inactivation of Efnb2 in early-stage embryonic ear tissues disrupted cell proliferation, cell survival, and epithelial folding at the origin of the endolymphatic epithelium. This correlated with apparent absence of an ED, mis-localization of ES ion transport cells relative to inner ear sensory organs, dysplasia of the endolymph fluid space, and abnormally formed otoconia (extracellular calcite-protein composites) at later stages of embryonic development. A comparison of Efnb2 and Notch signaling-deficient mutant phenotypes indicated that these two signaling systems have distinct and non-overlapping roles in ES/ED development. Homozygous deletion of the Efnb2 C-terminus caused abnormalities similar to those found in the conditional Efnb2 null homozygote. Analyses of fetal Efnb2 C-terminus deletion heterozygotes found mis-localized ES ion transport cells only in the genetic background exhibiting vestibular dysfunction. We propose that developmental dysplasias described here are a gene dose-sensitive cause of the vestibular dysfunction observed in EphB-Efnb2 signaling-deficient mice.


Assuntos
Orelha Interna/metabolismo , Saco Endolinfático/metabolismo , Efrina-B2/genética , Epitélio/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular/genética , Orelha Interna/embriologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Saco Endolinfático/embriologia , Saco Endolinfático/ultraestrutura , Efrina-B2/metabolismo , Epitélio/embriologia , Epitélio/ultraestrutura , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica de Varredura , Morfogênese/genética , Gravidez , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética , Fatores de Tempo
13.
Methods ; 66(3): 447-53, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23792918

RESUMO

The inner ear transduces the mechanical stimuli that are associated with sound and balance perception. Missteps during its formation often result in deafness, and thus understanding otic development has a profound clinical relevance. The intricate complexity of the inner ear is derived from a simple epithelial sheet during embryogenesis. Study of this process in vitro has provided insight into the mechanisms of otic induction, patterning and differentiation. This article details methods for the culture of otic placode, otocyst, and basilar papilla, providing a toolkit for the investigation of multiple facets of otic organogenesis, for regeneration studies and for setting up small molecule screens to identify possible therapeutic targets.


Assuntos
Galinhas , Orelha Interna/embriologia , Técnicas de Cultura de Tecidos , Animais , Embrião de Galinha
14.
Viruses ; 15(1)2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36680287

RESUMO

The transmembrane protein Axl was proposed as an entry receptor for Zika virus (ZIKV) infection in vitro, but conflicting results from in vivo studies have made it difficult to establish Axl as a physiologically relevant ZIKV receptor. Both the functional redundancy of receptors and the experimental model used can lead to variable results. Therefore, it can be informative to explore alternative animal models to analyze ZIKV receptor candidates as an aid in discovering antivirals. This study used chicken embryos to examine the role of chicken Tyro3 (cTyro3), the equivalent of human Axl. Results show that endogenous cTyro3 mRNA expression overlaps with previously described hot spots of ZIKV infectivity in the brain and inner ear. We asked if ectopic expression or knockdown of cTyro3 influenced ZIKV infection in embryos. Tol2 vectors or replication-competent avian retroviruses were used in ovo to introduce full-length or truncated (presumed dominant-negative) cTyro3, respectively, into the neural tube on embryonic day two (E2). ZIKV was delivered to the brain 24 h later. cTyro3 manipulations did not alter ZIKV infection or cell death in the E5/E6 brain. Moreover, delivery of truncated cTyro3 variants to the E3 otocyst had no effect on inner ear formation on E6 or E10.


Assuntos
Orelha Interna , Infecção por Zika virus , Zika virus , Embrião de Galinha , Animais , Humanos , Receptor Tirosina Quinase Axl , Receptores Proteína Tirosina Quinases/metabolismo , Galinhas , Proteínas Proto-Oncogênicas/metabolismo , Receptores Virais/metabolismo , Encéfalo/metabolismo , Replicação Viral
15.
Cell Rep ; 38(12): 110542, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35320729

RESUMO

Inner ear vestibular and spiral ganglion neurons (VGNs and SGNs) are known to play pivotal roles in balance control and sound detection. However, the molecular mechanisms underlying otic neurogenesis at early embryonic ages have remained unclear. Here, we use single-cell RNA sequencing to reveal the transcriptomes of mouse otic tissues at three embryonic ages, embryonic day 9.5 (E9.5), E11.5, and E13.5, covering proliferating and undifferentiated otic neuroblasts and differentiating VGNs and SGNs. We validate the high quality of our studies by using multiple assays, including genetic fate mapping analysis, and we uncover several genes upregulated in neuroblasts or differentiating VGNs and SGNs, such as Shox2, Myt1, Casz1, and Sall3. Notably, our findings suggest a general cascaded differentiation trajectory during early otic neurogenesis. The comprehensive understanding of early otic neurogenesis provided by our study holds critical implications for both basic and translational research.


Assuntos
Orelha Interna , Células-Tronco Neurais , Animais , Camundongos , Neurogênese/genética , Neurônios , Transcriptoma/genética
16.
Anat Rec (Hoboken) ; 303(3): 451-460, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943808

RESUMO

The mammalian inner ear mediates hearing and balance and during development generates both cochleo-vestibular ganglion neurons and sensory epithelial receptor cells, that is, hair cells and support cells. Cell marking experiments have shown that both hair cells and support cells can originate from a common progenitor. Here, we demonstrate the lineage potential of individual otic epithelial cell clones using three cell lines established by a combination of limiting dilution and gene-marking techniques from an embryonic day 12 (E12) rat otocyst. Cell-type specific marker analyses of these clonal lines under proliferation and differentiation culture conditions demonstrate that during differentiation immature cell markers (Nanog and Nestin) were downregulated and hair cell (Myosin VIIa and Math1), support cell (p27Kip1 and cytokeratin) and neuronal cell (NF-H and NeuroD) markers were upregulated. Our results suggest that the otic epithelium of the E12 mammalian inner ear possess multipotent progenitor cells able to generate cell types of both sensory epithelial and neural cell lineages when cultured under a differentiation culture condition. Understanding the molecular mechanisms of proliferation and differentiation of multipotent otic progenitor cells may provide insights that could contribute to the development of a novel cell therapy with a potential to initiate or stimulate the sensorineural repair of damaged inner ear sensory receptors. Anat Rec, 303:451-460, 2020. © 2019 American Association for Anatomy.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Orelha Interna/citologia , Células Ciliadas Auditivas/citologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Orelha Interna/embriologia , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Miosina VIIa/metabolismo , Proteína Homeobox Nanog/metabolismo , Nestina/metabolismo , Neurônios/metabolismo , Ratos , Ratos Wistar , Células-Tronco/metabolismo
17.
Dis Model Mech ; 13(3)2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-31980437

RESUMO

Single-nucleotide mutations in human SIX1 result in amino acid substitutions in either the protein-protein interaction domain or the homeodomain, and cause ∼4% of branchio-otic (BOS) and branchio-oto-renal (BOR) cases. The phenotypic variation between patients with the same mutation, even within affected members of the same family, make it difficult to functionally distinguish between the different SIX1 mutations. We made four of the BOS/BOR substitutions in the Xenopus Six1 protein (V17E, R110W, W122R, Y129C), which is 100% identical to human in both the protein-protein interaction domain and the homeodomain, and expressed them in embryos to determine whether they cause differential changes in early craniofacial gene expression, otic gene expression or otic morphology. We confirmed that, similar to the human mutants, all four mutant Xenopus Six1 proteins access the nucleus but are transcriptionally deficient. Analysis of craniofacial gene expression showed that each mutant causes specific, often different and highly variable disruptions in the size of the domains of neural border zone, neural crest and pre-placodal ectoderm genes. Each mutant also had differential effects on genes that pattern the otic vesicle. Assessment of the tadpole inner ear demonstrated that while the auditory and vestibular structures formed, the volume of the otic cartilaginous capsule, otoliths, lumen and a subset of the hair cell-containing sensory patches were reduced. This detailed description of the effects of BOS/BOR-associated SIX1 mutations in the embryo indicates that each causes subtle changes in gene expression in the embryonic ectoderm and otocyst, leading to inner ear morphological anomalies.


Assuntos
Síndrome Brânquio-Otorrenal/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Mutação/genética , Crânio/embriologia , Sequência de Aminoácidos , Animais , Orelha , Células HEK293 , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Crista Neural/metabolismo , Membrana dos Otólitos/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Transcrição Gênica , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/genética
18.
J Assoc Res Otolaryngol ; 20(2): 133-149, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30564989

RESUMO

Many developmental disorders of the inner ear are manifested clinically as delayed motor development and challenges in maintaining posture and balance, indicating involvement of central vestibular circuits. How the vestibular circuitry is rewired in pediatric cases is poorly understood due to lack of a suitable animal model. Based on this, our lab designed and validated a chick embryo model to study vestibular development in congenital vestibular disorders. The developing inner ear or "otocyst" on the right side of 2-day-old chick embryos (E2) was surgically rotated 180° in the anterior-posterior axis, forming the "anterior-posterior axis rotated otocyst chick" or ARO chick. The ARO chick has a reproducible pathology of a sac with truncated or missing semicircular canals. A sac is the most common inner ear defect found in children with congenital vestibular disorders. In E13 ARO chicks, the sac contained all three cristae and maculae utriculi and sacculi, but the superior crista and macula utriculi were shortened in anterior-posterior extent. Also, the number of principal cells of the tangential vestibular nucleus, a major avian vestibular nucleus, was decreased 66 % on the rotated side. After hatching, no difference was detected between ARO and normal chicks in their righting reflex times. However, unlike normal chicks, ARO hatchlings had a constant, right head tilt, and after performing the righting reflex, ARO chicks stumbled and walked with a widened base. Identifying the structure and function of abnormally developed brain regions in ARO chicks may assist in improving treatments for patients with congenital vestibular disorder.


Assuntos
Embrião de Galinha , Modelos Animais de Doenças , Orelha Interna/embriologia , Doenças Vestibulares/congênito , Animais , Orelha Interna/inervação , Reflexo Vestíbulo-Ocular , Doenças Vestibulares/fisiopatologia
19.
Neurosci Lett ; 647: 153-158, 2017 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-28359931

RESUMO

The otocyst is an attractive target for studying treatment strategies for genetic hearing loss and for understanding inner ear development. We have previously reported that trans-uterine supplemental gene therapy in vivo into the otocysts of mice, which had a loss of function mutation in a causative gene of deafness, was able to prevent putative hearing loss. We herein set out to clarify the feasibility of allogenic cell transplantation into the mouse otocysts in vivo. We transplanted naive mouse-derived induced pluripotent stem cells (miPSCs) into the otocysts of wild type mice or connexin (Cx) 30 deficient mice, at embryonic day 11.5 (E11.5). The transplanted m-iPSCs survived in the lumens of the inner ears at E13.5 and E15.5 in wild type mice. In the Cx30 deficient mouse, the transplanted cells survived similarly, with some of the transplanted cells migrating into the lining cells of the lumens of the inner ears at E13.5 and showing tumorigenic cell proliferation at E15.5. In addition, engrafted cells appear to be able to differentiate after the cell transplantation. Our results suggest that otocyst transplanted cells survived and differentiated. A Cx30 deficiency may facilitate cell migration. These findings may offer some hope for cell transplantation therapy for profound genetic hearing loss caused by a Cxs deficiency.


Assuntos
Orelha Interna/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Animais , Carcinogênese , Diferenciação Celular , Movimento Celular , Proliferação de Células , Conexina 30 , Conexinas/genética , Orelha Interna/embriologia , Células Epiteliais/citologia , Estudos de Viabilidade , Camundongos Knockout
20.
Brain Struct Funct ; 222(5): 2071-2092, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27783221

RESUMO

The vertebrate inner ear is a complex three-dimensional sensorial structure with auditory and vestibular functions. The molecular patterning of the developing otic epithelium creates various positional identities, consequently leading to the stereotyped specification of each neurosensory and non-sensory element of the membranous labyrinth. The Iroquois (Iro/Irx) genes, clustered in two groups (A: Irx1, Irx2, and Irx4; and B: Irx3, Irx5, and Irx6), encode for transcriptional factors involved directly in numerous patterning processes of embryonic tissues in many phyla. This work presents a detailed study of the expression patterns of these six Irx genes during chick inner ear development, paying particular attention to the axial specification of the otic anlagen. The Irx genes seem to play different roles at different embryonic periods. At the otic vesicle stage (HH18), all the genes of each cluster are expressed identically. Both clusters A and B seem involved in the specification of the lateral and posterior portions of the otic anlagen. Cluster B seems to regulate a larger area than cluster A, including the presumptive territory of the endolymphatic apparatus. Both clusters seem also to be involved in neurogenic events. At stages HH24/25-HH27, combinations of IrxA and IrxB genes participate in the specification of most sensory patches and some non-sensory components of the otic epithelium. At stage HH34, the six Irx genes show divergent patterns of expression, leading to the final specification of the membranous labyrinth, as well as to cell differentiation.


Assuntos
Diferenciação Celular/fisiologia , Orelha Interna/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Embrião de Galinha , Galinhas , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Vertebrados/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA