Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Development ; 142(3): 533-43, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25605781

RESUMO

The primary glial cells in the retina, the Müller glia, differentiate from retinal progenitors in the first postnatal week. CNTF/LIF/STAT3 signaling has been shown to promote their differentiation; however, another key glial differentiation signal, BMP, has not been examined during this period of Müller glial differentiation. In the course of our analysis of the BMP signaling pathway, we observed a transient wave of Smad1/5/8 signaling in the inner nuclear layer at the end of the first postnatal week, from postnatal day (P) 5 to P9, after the end of neurogenesis. To determine the function of this transient wave, we blocked BMP signaling during this period in vitro or in vivo, using either a BMP receptor antagonist or noggin (Nog). Either treatment leads to a reduction in expression of the Müller glia-specific genes Rlbp1 and Glul, and the failure of many of the Müller glia to repress the bipolar/photoreceptor gene Otx2. These changes in normal Müller glial differentiation result in permanent disruption of the retina, including defects in the outer limiting membrane, rosette formation and a reduction in functional acuity. Our results thus show that Müller glia require a transient BMP signal at the end of neurogenesis to fully repress the neural gene expression program and to promote glial gene expression.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/fisiologia , Células Ependimogliais/fisiologia , Neurogênese/fisiologia , Retina/crescimento & desenvolvimento , Transdução de Sinais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Imunoprecipitação da Cromatina , Primers do DNA/genética , Técnicas de Introdução de Genes , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
2.
Dev Biol ; 411(1): 1-14, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26826497

RESUMO

The inner ear contains six distinct sensory organs that each maintains some ability to regenerate hair cells into adulthood. In the postnatal cochlea, there appears to be a relationship between the developmental maturity of a region and its ability to regenerate as postnatal regeneration largely occurs in the apical turn, which is the last region to differentiate and mature during development. In the mature cristae there are also regional differences in regenerative ability, which led us to hypothesize that there may be a general relationship between the relative maturity of a region and the regenerative competence of that region in all of the inner ear sensory organs. By analyzing adult mouse cristae labeled embryonically with BrdU, we found that hair cell birth starts in the central region and progresses to the periphery with age. Since the peripheral region of the adult cristae also maintains active Notch signaling and some regenerative competence, these results are consistent with the hypothesis that the last regions to develop retain some of their regenerative ability into adulthood. Further, by analyzing embryonic day 14.5 inner ears we provide evidence for a wave of hair cell birth along the longitudinal axis of the cristae from the central regions to the outer edges. Together with the data from the adult inner ears labeled with BrdU as embryos, these results suggest that hair cell differentiation closely follows cell cycle exit in the cristae, unlike in the cochlea where they are uncoupled.


Assuntos
Ciclo Celular/fisiologia , Cóclea/embriologia , Células Ciliadas da Ampola/citologia , Células Ciliadas Auditivas/citologia , Organogênese/fisiologia , Animais , Bromodesoxiuridina/metabolismo , Diferenciação Celular , Feminino , Audição/fisiologia , Camundongos , Receptores Notch/metabolismo , Regeneração/fisiologia , Transdução de Sinais , Análise Espacial
3.
J Neurosci ; 32(37): 12876-84, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22973011

RESUMO

Hearing loss is becoming an increasingly prevalent problem affecting more than 250 million people worldwide. During development, fibroblast growth factors (FGFs) are required for inner ear development as well as hair cell formation in the mammalian cochlea and thus make attractive therapeutic candidates for the regeneration of sensory cells. Previous findings showed that Fgfr1 conditional knock out mice exhibited hair cell and support cell formation defects. Immunoblocking with Fgf20 antibody in vitro produced a similar phenotype. While hair cell differentiation in mice starts at embryonic day (E)14.5, beginning with the inner hair cells, Fgf20 expression precedes hair cell differentiation at E13.5 in the cochlea. This suggests a potential role for Fgf20 in priming the sensory epithelium for hair cell formation. Treatment of explants with a gamma-secretase inhibitor, DAPT, decreased Fgf20 mRNA, suggesting that Notch is upstream of Fgf20. Notch signaling also plays an early role in prosensory formation during cochlear development. In this report we show that during development, Notch-mediated regulation of prosensory formation in the cochlea occurs via Fgf20. Addition of exogenous FGF20 compensated for the block in Notch signaling and rescued Sox2, a prosensory marker, and Gfi1, an early hair cell marker in explant cultures. We hypothesized that Fgf20 plays a role in specification, amplification, or maintenance of Sox2 expression in prosensory progenitors of the developing mammalian cochlea.


Assuntos
Cóclea/embriologia , Cóclea/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurogênese/fisiologia , Receptores Notch/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Camundongos
4.
Proc Natl Acad Sci U S A ; 107(36): 15792-7, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20798046

RESUMO

During inner ear morphogenesis, the process of prosensory specification defines the specific regions of the otic epithelium that will give rise to the six separate inner ear organs essential for hearing and balance. The mechanism of prosensory specification is not fully understood, but there is evidence that the Notch intercellular signaling pathway plays a critical role. The Notch ligand Jagged1 (Jag1) is expressed in the prosensory domains, and mutation of Jag1 impairs sensory formation. Furthermore, pharmacological inhibition of Notch in vitro during prosensory specification disrupts the prosensory process. Additionally, activation of Notch by cDNA electroporation in chick otocysts results in formation of ectopic sensory patches. Here we test whether Notch activity is sufficient for prosensory specification in the mouse, using a Cre-/loxP approach to conditionally activate the Notch pathway in nonsensory regions of the inner ear epithelia during different stages of otic vesicle morphogenesis. We find that broad ectopic activation of Notch at very early developmental stages causes induction of prosensory markers throughout the entire otic epithelium. At later stages of development, activation of Notch in nonsensory regions leads to induction of sensory patches that later differentiate to form complete ectopic sensory structures. Activation of Notch in isolated nonsensory cells results in lateral induction of Jag1 expression in neighboring cells and spreading of prosensory specification to the adjacent cells through an intercellular mechanism. These results support a model where activation of Notch and propagation through lateral induction promote prosensory character in specific regions of the developing otocyst.


Assuntos
Células Ciliadas Auditivas Internas/citologia , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Morte Celular , Proliferação de Células , Células Ciliadas Auditivas Internas/metabolismo , Camundongos , Camundongos Knockout
5.
PLoS Genet ; 5(8): e1000607, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19680541

RESUMO

Mutations in the CLRN1 gene cause Usher syndrome type 3 (USH3), a human disease characterized by progressive blindness and deafness. Clarin 1, the protein product of CLRN1, is a four-transmembrane protein predicted to be associated with ribbon synapses of photoreceptors and cochlear hair cells, and recently demonstrated to be associated with the cytoskeleton. To study Clrn1, we created a Clrn1 knockout (KO) mouse and characterized the histological and functional consequences of Clrn1 deletion in the retina and cochlea. Clrn1 KO mice do not develop a retinal degeneration phenotype, but exhibit progressive loss of sensory hair cells in the cochlea and deterioration of the organ of Corti by 4 months. Hair cell stereocilia in KO animals were longer and disorganized by 4 months, and some Clrn1 KO mice exhibited circling behavior by 5-6 months of age. Clrn1 mRNA expression was localized in the retina using in situ hybridization (ISH), laser capture microdissection (LCM), and RT-PCR. Retinal Clrn1 transcripts were found throughout development and adulthood by RT-PCR, although expression peaked at P7 and declined to undetectable levels in adult retina by ISH. LCM localized Clrn1 transcripts to the retinas inner nuclear layer, and WT levels of retinal Clrn1 expression were observed in photoreceptor-less retinas. Examination of Clrn1 KO mice suggests that CLRN1 is unnecessary in the murine retina but essential for normal cochlear development and function. This may reflect a redundancy in the mouse retina not present in human retina. In contrast to mouse KO models of USH1 and USH2, our data indicate that Clrn1 expression in the retina is restricted to the Müller glia. This is a novel finding, as most retinal degeneration associated proteins are expressed in photoreceptors, not in glia. If CLRN1 expression in humans is comparable to the expression pattern observed in mice, this is the first report of an inner retinal protein that, when mutated, causes retinal degeneration.


Assuntos
Cóclea/crescimento & desenvolvimento , Células Ciliadas Auditivas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Retina/metabolismo , Animais , Cóclea/citologia , Cóclea/metabolismo , Modelos Animais de Doenças , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transporte Proteico , Retina/crescimento & desenvolvimento
6.
Hum Mol Genet ; 18(15): 2748-60, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19414487

RESUMO

Usher syndrome 3A (USH3A) is an autosomal recessive disorder characterized by progressive loss of hearing and vision due to mutation in the clarin-1 (CLRN1) gene. Lack of an animal model has hindered our ability to understand the function of CLRN1 and the pathophysiology associated with USH3A. Here we report for the first time a mouse model for ear disease in USH3A. Detailed evaluation of inner ear phenotype in the Clrn1 knockout mouse (Clrn1(-/-)) coupled with expression pattern of Clrn1 in the inner ear are presented here. Clrn1 was expressed as early as embryonic day 16.5 in the auditory and vestibular hair cells and associated ganglionic neurons, with its expression being higher in outer hair cells (OHCs) than inner hair cells. Clrn1(-/-) mice showed early onset hearing loss that rapidly progressed to severe levels. Two to three weeks after birth (P14-P21), Clrn1(-/-) mice showed elevated auditory-evoked brainstem response (ABR) thresholds and prolonged peak and interpeak latencies. By P21, approximately 70% of Clrn1(-/-) mice had no detectable ABR and by P30 these mice were deaf. Distortion product otoacoustic emissions were not recordable from Clrn1(-/-) mice. Vestibular function in Clrn1(-/-) mice mirrored the cochlear phenotype, although it deteriorated more gradually than cochlear function. Disorganization of OHC stereocilia was seen as early as P2 and by P21 OHC loss was observed. In sum, hair cell dysfunction and prolonged peak latencies in vestibular and cochlear evoked potentials in Clrn1(-/-) mice strongly indicate that Clrn1 is necessary for hair cell function and associated neural activation.


Assuntos
Células Ciliadas Auditivas/fisiologia , Proteínas de Membrana/metabolismo , Neurônios/fisiologia , Síndromes de Usher/genética , Síndromes de Usher/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndromes de Usher/metabolismo
7.
Dev Dyn ; 239(3): 1019-26, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20131355

RESUMO

Many studies have shown the importance of the fibroblast growth factor (FGF) family of factors in the development of the mammalian cochlea. There are four fibroblast growth factor receptors (FGFR1-4) and all four are expressed in the cochlea during development. While there are examples in the literature of expression patterns of some of the receptors at specific stages of cochlear development there has been no systematic study. We have assembled a full analysis of the patterns of receptor expression during cochlear development for all four Fgfrs using in situ hybridization. We have analyzed the expression patterns from embryonic day 13.5 through postnatal ages. We find that Fgfr1, 2, and 3 are expressed in the epithelium of the cochlear duct and Fgfr4 is limited in its expression to the mesenchyme surrounding the duct. We compare the receptor expression pattern to markers of the sensory domain (p27kip1) and the early hair cells (math1).


Assuntos
Cóclea/metabolismo , Ducto Coclear/embriologia , Fatores de Crescimento de Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/biossíntese , Animais , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Mesoderma/metabolismo , Camundongos , Microscopia de Fluorescência/métodos
8.
Dev Dyn ; 239(7): 2034-40, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20549727

RESUMO

RNA transfection methods have not proven to be as popular as DNA methods due to the highly transient nature of the RNA inside the cell. However, there are many advantages in using RNA for gene over-expression, such as the rapidity of expression, the ability to express in all cell types without the need for cell-type-specific promoters, and the ability to analyze the effects of gene over-expression in a transient manner. Therefore, we have developed a method (StabiLizingUtr: SLU) to stabilize the RNA for varying durations, using specific sequences from the 3'UTR of the Venezuelan equine encephalitis virus (VEEV). We have designed a plasmid for cloning genes upstream from repeated stabilizing sequences to generate mRNA with one or more VEEV-stabilizing sequence motifs. We demonstrate this method in several cell and tissue types, including the mammalian cochlea, a tissue that has been difficult to transfect with other methods.


Assuntos
Transfecção/métodos , Regiões 3' não Traduzidas/genética , Animais , Linhagem Celular , Eletroporação , Vírus da Encefalite Equina Venezuelana/genética , Humanos , Imuno-Histoquímica , Camundongos , Estabilidade de RNA/genética , RNA Mensageiro/genética , Ratos
9.
Elife ; 102021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34003106

RESUMO

This study provides transcriptomic characterization of the cells of the crista ampullaris, sensory structures at the base of the semicircular canals that are critical for vestibular function. We performed single-cell RNA-seq on ampullae microdissected from E16, E18, P3, and P7 mice. Cluster analysis identified the hair cells, support cells and glia of the crista as well as dark cells and other nonsensory epithelial cells of the ampulla, mesenchymal cells, vascular cells, macrophages, and melanocytes. Cluster-specific expression of genes predicted their spatially restricted domains of gene expression in the crista and ampulla. Analysis of cellular proportions across developmental time showed dynamics in cellular composition. The new cell types revealed by single-cell RNA-seq could be important for understanding crista function and the markers identified in this study will enable the examination of their dynamics during development and disease.


Assuntos
Perfilação da Expressão Gênica , RNA-Seq , Ductos Semicirculares/citologia , Ductos Semicirculares/fisiologia , Animais , Células Ciliadas da Ampola/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ductos Semicirculares/anatomia & histologia , Análise de Célula Única , Sequenciamento do Exoma
10.
Dev Dyn ; 238(9): 2163-78, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19191219

RESUMO

Delta gene expression in Drosophila is regulated by proneural basic helix-loop-helix (bHLH) transcription factors, such as acheate-scute. In vertebrates, multiple Delta-like and proneural bHLH genes are expressed during neurogenesis, especially in the retina. We recently uncovered a relationship between Acheate-scute like 1 (Ascl1), Delta-like genes, and Notch in chick retinal progenitors. Here, we report that mammalian retinal progenitors are also the primary source of Delta-like genes, likely signaling through Notch among themselves, while differentiating neurons expressed Jagged2. Ascl1 is coexpressed in Delta-like and Notch active progenitors, and required for normal Delta-like gene expression and Notch signaling. We also reveal a role for Ascl1 in the regulation of Hes6, a proneurogenic factor that inhibits Notch signaling to promote neural rather than glial differentiation. Thus, these results suggest a molecular mechanism whereby attenuated Notch levels coupled with reduced proneurogenic activity in progenitors leads to increased gliogenesis and decreased neurogenesis in the Ascl1-deficient retina.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptores Notch/metabolismo , Retina/metabolismo , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proteínas de Drosophila , Olho/embriologia , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Jagged-1 , Proteína Jagged-2 , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Reação em Cadeia da Polimerase , Receptores Notch/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Retina/citologia , Proteínas Serrate-Jagged , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
11.
Cell Rep ; 30(5): 1644-1659.e4, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32023475

RESUMO

To study the development of the human retina, we use single-cell RNA sequencing (RNA-seq) at key fetal stages and follow the development of the major cell types as well as populations of transitional cells. We also analyze stem cell (hPSC)-derived retinal organoids; although organoids have a very similar cellular composition at equivalent ages as the fetal retina, there are some differences in gene expression of particular cell types. Moreover, the inner retinal lamination is disrupted at more advanced stages of organoids compared with fetal retina. To determine whether the disorganization in the inner retina is due to the culture conditions, we analyze retinal development in fetal retina maintained under similar conditions. These retinospheres develop for at least 6 months, displaying better inner retinal lamination than retinal organoids. Our single-cell RNA sequencing (scRNA-seq) comparisons of fetal retina, retinal organoids, and retinospheres provide a resource for developing better in vitro models for retinal disease.


Assuntos
Feto/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Retina/citologia , Análise de Célula Única , Transcriptoma/genética , Células Cultivadas , Humanos , Mitose/genética , Esferoides Celulares/citologia , Fatores de Tempo
12.
J Neurosci ; 28(23): 5991-9, 2008 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-18524904

RESUMO

Tissue-specific deletion of Fgfr1 results in severe defects in the development of both hair cells and support cells (Pirvola et al., 2002). Despite the importance of Fgfr1 in this early phase of cochlear development, the timing for the requirement for FGF signaling at this stage is not known. Therefore, we investigated the requirement for FGF signaling at early stages of cochlear development using an FGF receptor inhibitor. We find that inhibition of FGF signaling from embryonic day 14 (E14) to E16 has a dramatic effect on the development of the sensory epithelium, causing a severe reduction in hair cells and support cells, similar to that reported for the Fgfr1 deletion. The effects of inhibition of FGF signaling on sensory specification are not explained by increases in cell death or changes in proliferation but lead to a rapid reduction in Pea3 and Erm and a loss of Math1 expression. We also show that a specific FGF, FGF20, is the likely ligand for FGFR1 at this sensory specification phase of cochlear development; Fgf20 is expressed at the right time and place to mediate sensory cell specification, and blocking FGF20 with a specific antibody inhibits hair cell and support cell development in a manner similar to the FGF receptor inhibitor. Our results thus define the period of FGF-dependent sensory cell specification and the ligand that mediates this step in cochlear development.


Assuntos
Cóclea/embriologia , Cóclea/metabolismo , Células Epiteliais/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Células Ciliadas Auditivas/metabolismo , Neurônios Aferentes/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Camundongos , Neurônios Aferentes/metabolismo , Técnicas de Cultura de Órgãos , Pirróis/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia
13.
Dev Biol ; 316(1): 87-99, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18291358

RESUMO

In cochlear development, the Notch signaling pathway is required for both the early prosensory phase and a later lateral inhibition phase. While it is known that Hes genes are important downstream mediators of Notch function in lateral inhibition, it is not known what genes function as mediators of the early prosensory function of Notch. We report that two members of the Hes-related gene family, Hesr1 and Hesr2, are expressed in the developing cochlea at a time and place that makes them excellent candidates as downstream mediators of Notch during prosensory specification. We also show that treatment of cochlear explant cultures at the time of prosensory specification with a small-molecule inhibitor of the Notch pathway mimics the results of conditional Jag1 deletion. This treatment also reduces Hesr1 and Hesr2 expression by as much as 80%. These results support the hypothesis that Hesr1 and Hesr2 are the downstream mediators of the prosensory function of Notch in early cochlear development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cóclea/embriologia , Morfogênese , Receptores Notch/metabolismo , Proteínas Repressoras/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Dipeptídeos/farmacologia , Deleção de Genes , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/citologia , Ácidos Hidroxâmicos/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Morfogênese/genética , Técnicas de Cultura de Órgãos , Receptores Notch/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Serrate-Jagged , Transdução de Sinais
14.
Sci Rep ; 9(1): 9060, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31227770

RESUMO

Hearing loss is often due to the absence or the degeneration of hair cells in the cochlea. Understanding the mechanisms regulating the generation of hair cells may therefore lead to better treatments for hearing disorders. To elucidate the transcriptional control mechanisms specifying the progenitor cells (i.e. prosensory cells) that generate the hair cells and support cells critical for hearing function, we compared chromatin accessibility using ATAC-seq in sorted prosensory cells (Sox2-EGFP+) and surrounding cells (Sox2-EGFP-) from E12, E14.5 and E16 cochlear ducts. In Sox2-EGFP+, we find greater accessibility in and near genes restricted in expression to the prosensory region of the cochlear duct including Sox2, Isl1, Eya1 and Pou4f3. Furthermore, we find significant enrichment for the consensus binding sites of Sox2, Six1 and Gata3-transcription factors required for prosensory development-in the open chromatin regions. Over 2,200 regions displayed differential accessibility with developmental time in Sox2-EGFP+ cells, with most changes in the E12-14.5 window. Open chromatin regions detected in Sox2-EGFP+ cells map to over 48,000 orthologous regions in the human genome that include regions in genes linked to deafness. Our results reveal a dynamic landscape of open chromatin in prosensory cells with potential implications for cochlear development and disease.


Assuntos
Cromatina/metabolismo , Cóclea/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Cóclea/embriologia , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Polimorfismo de Nucleotídeo Único , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/metabolismo
16.
J Assoc Res Otolaryngol ; 19(5): 483-491, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30171385

RESUMO

This study examines absolute hair cell numbers in the cristae of C57BL/6J mice and CBA/CaJ mice from weaning to adulthood as well as the dose required for 3,3'-iminodiproprionitrile (IDPN)-injury of the cristae in C57BL/6J mice and CBA/CaJ mice, the two mouse strains most commonly used by inner ear researchers. In cristae of CBA/CaJ and C57BL/6J mice, no loss of hair cells was observed up to 24 weeks. In both strains, dose-dependent loss of hair cells was observed 7 days after IDPN treatment of 2-month-old mice (IC50 = 16.1 mmol/kg in C57BL/6J mice vs. 25.21 mmol/kg in CBA/CaJ mice). Four-month-old C57BL/6J mice exposed to IDPN developed dose-dependent vestibular dysfunction as indicated by increased activity and circling behavior in open field tests and by failure to swim 7 days after treatment. IDPN-hair cell injury in C57BL/6J mice and CBA/CaJ mice represents a fast and predictable experimental model for the study of vestibular degeneration and a platform for the testing of vestibular therapies.


Assuntos
Células Ciliadas Auditivas/efeitos dos fármacos , Nitrilas/toxicidade , Animais , Contagem de Células , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA
17.
Dev Cell ; 43(6): 763-779.e4, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29233477

RESUMO

Clinical and genetic heterogeneity associated with retinal diseases makes stem-cell-based therapies an attractive strategy for personalized medicine. However, we have limited understanding of the timing of key events in the developing human retina, and in particular the factors critical for generating the unique architecture of the fovea and surrounding macula. Here we define three key epochs in the transcriptome dynamics of human retina from fetal day (D) 52 to 136. Coincident histological analyses confirmed the cellular basis of transcriptional changes and highlighted the dramatic acceleration of development in the fovea compared with peripheral retina. Human and mouse retinal transcriptomes show remarkable similarity in developmental stages, although morphogenesis was greatly expanded in humans. Integration of DNA accessibility data allowed us to reconstruct transcriptional networks controlling photoreceptor differentiation. Our studies provide insights into human retinal development and serve as a resource for molecular staging of human stem-cell-derived retinal organoids.


Assuntos
Neurogênese/fisiologia , Retina/citologia , Retina/fisiologia , Animais , Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Fóvea Central/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Macula Lutea/embriologia , Camundongos , Morfogênese , Neurogênese/genética , Neurônios/metabolismo , Retina/embriologia , Retina/crescimento & desenvolvimento , Análise de Sequência de RNA/métodos , Transcriptoma
18.
J Comp Neurol ; 496(2): 172-86, 2006 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-16538679

RESUMO

We carried out an analysis of the expression of Prox1, a homeo-domain transcription factor, during mouse inner ear development with particular emphasis on the auditory system. Prox1 is expressed in the otocyst beginning at embryonic day (E)11, in the developing vestibular sensory patches. Expression is down regulated in maturing (myosin VIIA immunoreactive) vestibular hair cells and subsequently in the underlying support cell layer by E16.5. In the auditory sensory epithelium, Prox1 is initially expressed at embryonic day 14.5 in a narrow stripe of cells at the base of the cochlea. This stripe encompasses the full thickness of the sensory epithelium, including developing hair cells and support cells. Over the next several days the stripe of expression extends to the apex, and as the sensory epithelium differentiates Prox1 becomes restricted to a subset of support cells. Double labeling for Prox1 and cell-type-specific markers revealed that the outer hair cells transiently express Prox1. After E18, Prox1 protein is no longer detectable in hair cells, but it continues to be expressed in support cells for the rest of embryogenesis and into the second postnatal week. During this time, Prox1 is not expressed in all support cell types in the organ of Corti, but is restricted to developing Deiters' and pillar cells. The expression is maintained in these cells into the second week of postnatal life, at which time Prox1 is dynamically down regulated. These studies form a baseline from which we can analyze the role of Prox1 in vertebrate sensory development.


Assuntos
Cóclea/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cóclea/embriologia , Cóclea/crescimento & desenvolvimento , Embrião de Mamíferos , Epitélio/embriologia , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas S100/metabolismo , Proteínas Supressoras de Tumor , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
19.
Curr Opin Neurobiol ; 13(1): 119-26, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12593990

RESUMO

The discovery of hair cell regeneration in the inner ear of birds provides new optimism that there may be a treatment for hearing and balance disorders. In this review we describe the process of hair cell regeneration in birds; including restoration of function, recovery of perception and what is currently known about molecular events, such as growth factors and signalling systems. We examine some of the key recent findings in both birds and mammals.


Assuntos
Aves/crescimento & desenvolvimento , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/lesões , Células Ciliadas Auditivas/crescimento & desenvolvimento , Células Ciliadas Auditivas/lesões , Regeneração Nervosa/fisiologia , Recuperação de Função Fisiológica/fisiologia , Animais , Aves/anatomia & histologia , Aves/fisiologia , Orelha Interna/ultraestrutura , Células Ciliadas Auditivas/ultraestrutura , Perda Auditiva Neurossensorial/fisiopatologia , Perda Auditiva Neurossensorial/terapia , Humanos , Células Labirínticas de Suporte/metabolismo , Células Labirínticas de Suporte/ultraestrutura , Modelos Animais , Transdução de Sinais/fisiologia
20.
J Comp Neurol ; 493(3): 460-74, 2005 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-16261529

RESUMO

Deprivation of auditory nerve input in young mice results in dramatic neuron death in the anteroventral cochlear nucleus (CN), while the same manipulation performed in older mice does not result in significant neuronal loss. The molecular basis underlying this critical period of susceptibility to loss of afferent input remains largely unknown. One possibility is that developmental differences in baseline mRNA expression of specific genes could predispose CN neurons to either death or survival after deafferentation. We used two microarray platforms to identify differentially expressed genes in the CN during and after this critical period. Results across platforms were also compared to each other. Three ages were examined; during the critical period (postnatal day (P)7), at the closing of the critical period (P14), and 1 week after the critical period (P21). Of all the genes surveyed (22,690 or 15,247), 1,082 were identified as significantly changed in expression during the critical period relative to after. Real-time reverse transcription polymerase chain reaction and immunohistochemistry confirmed and extended the microarray results for a subset of these genes. Further analysis of genes related to apoptotic pathways showed 6 out of 7 differentially expressed known pro-apoptotic genes had higher expression during the critical period. In contrast, 9 out of 11 differentially expressed known pro-survival genes increased after the critical period when CN neurons survive deprivation. This finding supports the concept that multiple neuroprotective mechanisms increase and pro-apoptotic factors decrease over development to protect mature neurons from stressful insults, making them less dependent on afferent input for survival.


Assuntos
Núcleo Coclear/citologia , Núcleo Coclear/crescimento & desenvolvimento , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/citologia , Animais , Apoptose/fisiologia , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Sobrevivência Celular/fisiologia , Núcleo Coclear/metabolismo , Período Crítico Psicológico , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios Aferentes/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA