Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Protein Pept Lett ; 29(7): 567-573, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35546749

RESUMO

Protein tyrosine phosphatase receptor-type Q (PTPRQ), a member of the type III tyrosine phosphatase receptor (R3 PTPR) family, is composed of three domains, including 18 extracellular fibronectin type III (FN3) repeats, a transmembrane helix, and a cytoplasmic phosphotyrosine phosphatase (PTP) domain. PTPRQ was initially identified as a transcript upregulated in glomerular mesangial cells in a rat model of glomerulonephritis. Subsequently, studies found that PTPRQ has phosphotyrosine phosphatase and phosphatidylinositol phosphatase activities and can regulate cell proliferation, apoptosis, differentiation, and survival. Further in vivo studies showed that PTPRQ is necessary for the maturation of cochlear hair bundles and is considered a potential gene for deafness. In the recent two decades, 21 mutations in PTPRQ have been linked to autosomal recessive hearing loss (DFNB84) and autosomal dominant hearing loss (DFNA73). Recent mutations, deletions, and amplifications of PTPRQ have been observed in many types of cancers, which indicate that PTPRQ might play an essential role in the development of many cancers. In this review, we briefly describe PTPRQ structure and enzyme activity and focus on the correlation between PTPRQ and human disease. A profound understanding of PTPRQ could be helpful in the identification of new therapeutic targets to treat associated diseases.


Assuntos
Cóclea/metabolismo , Perda Auditiva , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Animais , Cóclea/crescimento & desenvolvimento , Fibronectinas , Perda Auditiva/genética , Humanos , Fosfatidilinositóis , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/química
2.
Development ; 148(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34061174

RESUMO

During embryonic development, the otic epithelium and surrounding periotic mesenchymal cells originate from distinct lineages and coordinate to form the mammalian cochlea. Epithelial sensory precursors within the cochlear duct first undergo terminal mitosis before differentiating into sensory and non-sensory cells. In parallel, periotic mesenchymal cells differentiate to shape the lateral wall, modiolus and pericochlear spaces. Previously, Wnt activation was shown to promote proliferation and differentiation of both otic epithelial and mesenchymal cells. Here, we fate-mapped Wnt-responsive epithelial and mesenchymal cells in mice and found that Wnt activation resulted in opposing cell fates. In the post-mitotic cochlear epithelium, Wnt activation via ß-catenin stabilization induced clusters of proliferative cells that dedifferentiated and lost epithelial characteristics. In contrast, Wnt-activated periotic mesenchyme formed ectopic pericochlear spaces and cell clusters showing a loss of mesenchymal and gain of epithelial features. Finally, clonal analyses via multi-colored fate-mapping showed that Wnt-activated epithelial cells proliferated and formed clonal colonies, whereas Wnt-activated mesenchymal cells assembled as aggregates of mitotically quiescent cells. Together, we show that Wnt activation drives transition between epithelial and mesenchymal states in a cell type-dependent manner.


Assuntos
Cóclea/embriologia , Epitélio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Desdiferenciação Celular , Diferenciação Celular , Proliferação de Células , Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Wnt , beta Catenina/metabolismo
3.
Curr Med Sci ; 41(1): 153-157, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33582920

RESUMO

K+ cycling in the cochlea is critical to maintain hearing. Many sodium-potassium pumps are proved to participate in K+ cycling, such as Na/K-ATPase. The α2-Na/K-ATPase is an important isoform of Na/K-ATPase. The expression of α2-Na/K-ATPase in the cochlea is not clear. In this study, we used C57BL/6 mice as a model of presbycusis and implemented immunohistochemistry staining and quantitative real time-PCR, and the α2-Na/K-ATPase expression pattern was confirmed in the inner ear. It was found α2-Na/K-ATPase was expressed widely in cochlea and its mRNA and protein expression was gradually reduced with aging (4-, 14-, 26- and 48-weeks old mice). We suspected that, the down-regulation of α2-Na/K-ATPase expression might be associated with the remodeling of K+ cycling, degeneration of morphological structure and decrease of hearing function in aging C57 mice. In conclusion, we speculated that the reduction of α2-Na/K-ATPase might play an important role in the pathogenesis of age-related hearing loss.


Assuntos
Envelhecimento/metabolismo , Cóclea/metabolismo , Perda Auditiva Neurossensorial/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , Envelhecimento/patologia , Animais , Cóclea/crescimento & desenvolvimento , Perda Auditiva Neurossensorial/genética , Camundongos , Camundongos Endogâmicos C57BL , ATPase Trocadora de Sódio-Potássio/metabolismo
4.
J Neurosci ; 41(4): 594-612, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33303678

RESUMO

Spontaneous bursts of electrical activity in the developing auditory system arise within the cochlea before hearing onset and propagate through future sound-processing circuits of the brain to promote maturation of auditory neurons. Studies in isolated cochleae revealed that this intrinsically generated activity is initiated by ATP release from inner supporting cells (ISCs), resulting in activation of purinergic autoreceptors, K+ efflux, and subsequent depolarization of inner hair cells. However, it is unknown when this activity emerges or whether different mechanisms induce activity during distinct stages of development. Here we show that spontaneous electrical activity in mouse cochlea from both sexes emerges within ISCs during the late embryonic period, preceding the onset of spontaneous correlated activity in inner hair cells and spiral ganglion neurons, which begins at birth and follows a base to apex developmental gradient. At all developmental ages, pharmacological inhibition of P2Y1 purinergic receptors dramatically reduced spontaneous activity in these three cell types. Moreover, in vivo imaging within the inferior colliculus revealed that auditory neurons within future isofrequency zones exhibit coordinated neural activity at birth. The frequency of these discrete bursts increased progressively during the postnatal prehearing period yet remained dependent on P2RY1. Analysis of mice with disrupted cholinergic signaling in the cochlea indicate that this efferent input modulates, rather than initiates, spontaneous activity before hearing onset. Thus, the auditory system uses a consistent mechanism involving ATP release from ISCs and activation of P2RY1 autoreceptors to elicit coordinated excitation of neurons that will process similar frequencies of sound.SIGNIFICANCE STATEMENT In developing sensory systems, groups of neurons that will process information from similar sensory space exhibit highly correlated electrical activity that is critical for proper maturation and circuit refinement. Defining the period when this activity is present, the mechanisms responsible and the features of this activity are crucial for understanding how spontaneous activity influences circuit development. We show that, from birth to hearing onset, the auditory system relies on a consistent mechanism to elicit correlate firing of neurons that will process similar frequencies of sound. Targeted disruption of this activity will increase our understanding of how these early circuits mature and may provide insight into processes responsible for developmental disorders of the auditory system.


Assuntos
Vias Auditivas/crescimento & desenvolvimento , Vias Auditivas/fisiologia , Receptores Purinérgicos/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Cóclea/crescimento & desenvolvimento , Cóclea/fisiologia , Feminino , Células Ciliadas Auditivas/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Colículos Inferiores/fisiologia , Células Labirínticas de Suporte/fisiologia , Masculino , Camundongos , Sistema Nervoso Parassimpático/efeitos dos fármacos , Sistema Nervoso Parassimpático/fisiologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y1/fisiologia , Retina/fisiologia , Gânglio Espiral da Cóclea/fisiologia
5.
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
6.
Neural Plast ; 2018: 7513258, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30410537

RESUMO

Inner ear formation requires that a series of cell fate decisions and morphogenetic events occur in a precise temporal and spatial pattern. Previous studies have shown that transcription factors, including Pax2, Sox2, and Prox1, play important roles during the inner ear development. However, the temporospatial expression patterns among these transcription factors are poorly understood. In the current study, we present a comprehensive description of the temporal and spatial expression profiles of Pax2, Sox2, and Prox1 during auditory and vestibular sensory organ development in mice. Using immunohistochemical analyses, we show that Sox2 and Pax2 are both expressed in the prosensory cells (the developing hair cells), but Sox2 is later restricted to only the supporting cells of the organ of Corti. In the vestibular sensory organ, however, the Pax2 expression is localized in hair cells at postnatal day 7, while Sox2 is still expressed in both the hair cells and supporting cells at that time. Prox1 was transiently expressed in the presumptive hair cells and developing supporting cells, and lower Prox1 expression was observed in the vestibular sensory organ compared to the organ of Corti. The different expression patterns of these transcription factors in the developing auditory and vestibular sensory organs suggest that they play different roles in the development of the sensory epithelia and might help to shape the respective sensory structures.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Vestibulares/metabolismo , Proteínas de Homeodomínio/biossíntese , Fator de Transcrição PAX2/biossíntese , Fatores de Transcrição SOXB1/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Animais , Animais Recém-Nascidos , Diferenciação Celular/fisiologia , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Orelha Interna , Feminino , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição PAX2/genética , Gravidez , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética
7.
Hum Genet ; 137(6-7): 437-446, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29860631

RESUMO

Identification of genes with variants causing non-syndromic hearing loss (NSHL) is challenging due to genetic heterogeneity. The difficulty is compounded by technical limitations that in the past prevented comprehensive gene identification. Recent advances in technology, using targeted capture and next-generation sequencing (NGS), is changing the face of gene identification and making it possible to rapidly and cost-effectively sequence the whole human exome. Here, we characterize a five-generation Chinese family with progressive, postlingual autosomal dominant nonsyndromic hearing loss (ADNSHL). By combining population-specific mutation arrays, targeted deafness genes panel, whole exome sequencing (WES), we identified PDE1C (Phosphodiesterase 1C) c.958G>T (p.A320S) as the disease-associated variant. Structural modeling insights into p.A320S strongly suggest that the sequence alteration will likely affect the substrate-binding pocket of PDE1C. By whole-mount immunofluorescence on postnatal day 3 mouse cochlea, we show its expression in outer (OHC) and inner (IHC) hair cells cytosol co-localizing with Lamp-1 in lysosomes. Furthermore, we provide evidence that the variant alters the PDE1C hydrolytic activity for both cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Collectively, our findings indicate that the c.958G>T variant in PDE1C may disrupt the cross talk between cGMP-signaling and cAMP pathways in Ca2+ homeostasis.


Assuntos
Cóclea/crescimento & desenvolvimento , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/genética , Surdez/genética , Proteínas de Membrana Lisossomal/genética , Animais , Povo Asiático/genética , Cóclea/metabolismo , Cóclea/fisiopatologia , AMP Cíclico/genética , Surdez/fisiopatologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Dominantes , Genótipo , Homeostase/genética , Humanos , Lisossomos/genética , Masculino , Camundongos , Mutação , Linhagem , Sequenciamento do Exoma
8.
J Comp Neurol ; 526(3): 425-438, 2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-29055051

RESUMO

Type II spiral ganglion neurons (SGNs) are small caliber, unmyelinated afferents that extend dendritic arbors hundreds of microns along the cochlear spiral, contacting many outer hair cells (OHCs). Despite these many contacts, type II afferents are insensitive to sound and only weakly depolarized by glutamate release from OHCs. Recent studies suggest that type II afferents may be cochlear nociceptors, and can be excited by ATP released during tissue damage, by analogy to somatic pain-sensing C-fibers. The present work compares the expression patterns among cochlear type II afferents of two genes found in C-fibers: calcitonin-related polypeptide alpha (Calca/Cgrpα), specific to pain-sensing C-fibers, and tyrosine hydroxylase (Th), specific to low-threshold mechanoreceptive C-fibers, which was shown previously to be a selective biomarker of type II versus type I cochlear afferents (Vyas et al., ). Whole-mount cochlear preparations from 3-week- to 2-month-old CGRPα-EGFP (GENSAT) mice showed expression of Cgrpα in a subset of SGNs with type II-like peripheral dendrites extending beneath OHCs. Double labeling with other molecular markers confirmed that the labeled SGNs were neither type I SGNs nor olivocochlear efferents. Cgrpα starts to express in type II SGNs before hearing onset, but the expression level declines in the adult. The expression patterns of Cgrpα and Th formed opposing gradients, with Th being preferentially expressed in apical and Cgrpα in basal type II afferent neurons, indicating heterogeneity among type II afferent neurons. The expression of Th and Cgrpα was not mutually exclusive and co-expression could be observed, most abundantly in the middle cochlear turn.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Receptoras Sensoriais/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Vias Aferentes/crescimento & desenvolvimento , Vias Aferentes/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Audição/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miosinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Tubulina (Proteína)/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo
9.
Eur J Histochem ; 61(3): 2706, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-29046047

RESUMO

Kölliker's organ is a temporary but indispensable structure in the development of the cochlea. Supporting cells (SCs) within it release adenosine 5'-triphosphate (ATP), which may play a crucial role in cochlear development before the onset of hearing. To reveal the apoptosis of Kölliker's organ in new-born rats, we studied the morphological changes and expression of apoptosis-related factors during early postnatal development. We found SCs in Kölliker's organ decreased in number and changed in appearance along the cochlea apex-to-base gradient, and the expression of caspase-3, caspase-8, caspase-9 and bcl-2 in Kölliker's organ of the cochlea fluctuated along the course of postnatal development, with an expression peak at postnatal day 3. This study demonstrates a time-dependent degeneration of Kölliker's organ during postnatal cochlea development, which might be triggered by endogenous factors.


Assuntos
Apoptose/genética , Cóclea/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Animais , Animais Recém-Nascidos , Western Blotting , Cóclea/anatomia & histologia , Imunofluorescência , Imuno-Histoquímica , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/metabolismo
10.
Cell Rep ; 19(2): 307-320, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402854

RESUMO

Hearing loss is widespread and persistent because mature mammalian auditory hair cells (HCs) are nonregenerative. In mice, the ability to regenerate HCs from surrounding supporting cells (SCs) declines abruptly after postnatal maturation. We find that combining p27Kip1 deletion with ectopic ATOH1 expression surmounts this age-related decline, leading to conversion of SCs to HCs in mature mouse cochleae and after noise damage. p27Kip1 deletion, independent of canonical effects on Rb-family proteins, upregulated GATA3, a co-factor for ATOH1 that is lost from SCs with age. Co-activation of GATA3 or POU4F3 and ATOH1 promoted conversion of SCs to HCs in adult mice. Activation of POU4F3 alone also converted mature SCs to HCs in vivo. These data illuminate a genetic pathway that initiates auditory HC regeneration and suggest p27Kip1, GATA3, and POU4F3 as additional therapeutic targets for ATOH1-mediated HC regeneration.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/genética , Fator de Transcrição GATA3/biossíntese , Perda Auditiva/genética , Proteínas de Homeodomínio/biossíntese , Fator de Transcrição Brn-3C/biossíntese , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células/genética , Cóclea/crescimento & desenvolvimento , Cóclea/patologia , Fator de Transcrição GATA3/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Perda Auditiva/patologia , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Regeneração/genética , Transdução de Sinais/genética , Fator de Transcrição Brn-3C/genética
11.
PLoS One ; 12(1): e0170568, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28118374

RESUMO

Primary auditory neurons (PANs) connect cochlear sensory hair cells in the mammalian inner ear to cochlear nucleus neurons in the brainstem. PANs develop from neuroblasts delaminated from the proneurosensory domain of the otocyst and keep maturing until the onset of hearing after birth. There are two types of PANs: type I, which innervate the inner hair cells (IHCs), and type II, which innervate the outer hair cells (OHCs). Glial cells surrounding these neurons originate from neural crest cells and migrate to the spiral ganglion. Several transcription factors are known to regulate the development and differentiation of PANs. Here we systematically examined the spatiotemporal expression of five transcription factors: Sox2, Sox10, Gata3, Mafb, and Prox1 from early delamination at embryonic day (E) 10.5 to adult. We found that Sox2 and Sox10 were initially expressed in the proneurosensory cells in the otocyst (E10.5). By E12.75 both Sox2 and Sox10 were downregulated in the developing PANs; however, Sox2 expression transiently increased in the neurons around birth. Furthermore, both Sox2 and Sox10 continued to be expressed in spiral ganglion glial cells. We also show that Gata3 and Prox1 were first expressed in all developing neurons, followed by a decrease in expression of Gata3 and Mafb in type I PANs and Prox1 in type II PANs as they matured. Moreover, we describe two subtypes of type II neurons based on Peripherin expression. These results suggest that Sox2, Gata3 and Prox1 play a role during neurogenesis as well as maturation of the PANs.


Assuntos
Cóclea/embriologia , Fator de Transcrição GATA3/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Neurogênese , Fatores de Transcrição SOXB1/biossíntese , Células Receptoras Sensoriais/metabolismo , Gânglio Espiral da Cóclea/embriologia , Proteínas Supressoras de Tumor/biossíntese , Animais , Núcleo Celular/metabolismo , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Fator de Transcrição GATA3/genética , Técnicas de Introdução de Genes , Genes Reporter , Idade Gestacional , Proteínas de Homeodomínio/genética , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neuroglia/metabolismo , Periferinas/biossíntese , Periferinas/genética , Proteínas Recombinantes de Fusão/biossíntese , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXE/biossíntese , Fatores de Transcrição SOXE/genética , Células Receptoras Sensoriais/classificação , Gânglio Espiral da Cóclea/metabolismo , Proteínas Supressoras de Tumor/genética
12.
J Nanosci Nanotechnol ; 17(4): 2390-393, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29648421

RESUMO

TMEM16A is a Ca²âº-activated Cl⁻ channel found in secretory glands, GI and respiratory tracts, and sensory organs, playing a major physiological role in fluid secretion, autonomous GI motility, and sensory transduction. In addition, overexpression of TMEM16A has been associated with cancer cell proliferation and invasion. Suppression of upregulated TMEM16A has been proposed as an effective anti-cancer strategy. While searching for a potential TMEM16A inhibitor, components of rice bran attracted our attention due to their anti-cancer potential in colon cancer cells, a type of cells known to overexpressing TMEM16A. Here, it was tested whether rice bran extract exhibits anti-TMEM16A activity. Rice bran extract was tested in the neonatal rat cochlear tissues where TMEM16A-involved spontaneous activity is generated as a part of normal development of the auditory pathway. Rice bran extract readily inhibited the TMEM16A-involved activity in the cochlear tissues and the effect was reversible upon washout. Taken together, rice bran extract appears to contain a putative TMEM16A inhibitor and the rice byproduct might serve as a source of a new anti-cancer agent.


Assuntos
Anoctamina-1/metabolismo , Cóclea/efeitos dos fármacos , Oryza/química , Extratos Vegetais/farmacologia , Animais , Animais Recém-Nascidos , Anoctamina-1/antagonistas & inibidores , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cóclea/crescimento & desenvolvimento , Fibras na Dieta , Ratos , Ratos Sprague-Dawley
13.
PLoS One ; 11(12): e0167850, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27959894

RESUMO

Ca2+ signaling has been implicated in the initial pathophysiologic mechanisms underlying the cochlea's response to acoustic overstimulation. Intracellular Ca2+ signaling (ICS) waves, which occur in glia and retinal cells in response to injury to activate cell regulatory pathways, have been proposed as an early event in cochlear injury. Disruption of ICS activity is thought to underlie Connexin 26-associated hearing loss, the most common genetic form of deafness, and downstream sequelae of ICS wave activity, such as MAP kinase pathway activation, have been implicated in noise-induced hearing loss. However, ICS waves have only been observed in neonatal cochlear cultures and are thought to be quiescent after the onset of hearing. In this study, we employ an acute explant model of an adult, hearing cochlea that retains many in vivo physiologic features to investigate Ca2+ changes in response to sound. We find that both slow monotonic changes in intracellular Ca2+ concentration as well as discrete ICS waves occur with acoustic overstimulation. The ICS waves share many intrinsic features with their better-described neonatal counterparts, including ATP and gap-junction dependence, and propagation velocity and distance. This identification of ICS wave activity in the adult, hearing cochlea thus confirms and characterizes an important early detection mechanism for cochlear trauma and provides a target for interventions for noise-induced and Connexin 26-associated hearing loss.


Assuntos
Sinalização do Cálcio , Cóclea/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Cóclea/crescimento & desenvolvimento , Cóclea/fisiologia , Feminino , Junções Comunicantes/metabolismo , Gerbillinae , Camundongos , Camundongos Endogâmicos C57BL , Som
14.
Proc Natl Acad Sci U S A ; 113(46): E7194-E7201, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27807138

RESUMO

Spatially and temporally coordinated variations of the cytosolic free calcium concentration ([Ca2+]c) play a crucial role in a variety of tissues. In the developing sensory epithelium of the mammalian cochlea, elevation of extracellular adenosine trisphosphate concentration ([ATP]e) triggers [Ca2+]c oscillations and propagation of intercellular inositol 1,4,5-trisphosphate (IP3)-dependent Ca2+ waves. What remains uncertain is the relative contribution of gap junction channels and connexin hemichannels to these fundamental mechanisms, defects in which impair hearing acquisition. Another related open question is whether [Ca2+]c oscillations require oscillations of the cytosolic IP3 concentration ([IP3]c) in this system. To address these issues, we performed Ca2+ imaging experiments in the lesser epithelial ridge of the mouse cochlea around postnatal day 5 and constructed a computational model in quantitative adherence to experimental data. Our results indicate that [Ca2+]c oscillations are governed by Hopf-type bifurcations within the experimental range of [ATP]e and do not require [IP3]c oscillations. The model replicates accurately the spatial extent and propagation speed of intercellular Ca2+ waves and predicts that ATP-induced ATP release is the primary mechanism underlying intercellular propagation of Ca2+ signals. The model also uncovers a discontinuous transition from propagating regimes (intercellular Ca2+ wave speed > 11 µm⋅s-1) to propagation failure (speed = 0), which occurs upon lowering the maximal ATP release rate below a minimal threshold value. The approach presented here overcomes major limitations due to lack of specific connexin channel inhibitors and can be extended to other coupled cellular systems.


Assuntos
Trifosfato de Adenosina/metabolismo , Sinalização do Cálcio , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Modelos Biológicos , Animais , Animais Recém-Nascidos , Camundongos
15.
Sci Rep ; 6: 31668, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27550540

RESUMO

Here we present spatio-temporal localization of Kremen1, a transmembrane receptor, in the mammalian cochlea, and investigate its role in the formation of sensory organs in mammal and fish model organisms. We show that Kremen1 is expressed in prosensory cells during cochlear development and in supporting cells of the adult mouse cochlea. Based on this expression pattern, we investigated whether Kremen1 functions to modulate cell fate decisions in the prosensory domain of the developing cochlea. We used gain and loss-of-function experiments to show that Kremen1 is sufficient to bias cells towards supporting cell fate, and is implicated in suppression of hair cell formation. In addition to our findings in the mouse cochlea, we examined the effects of over expression and loss of Kremen1 in the zebrafish lateral line. In agreement with our mouse data, we show that over expression of Kremen1 has a negative effect on the number of mechanosensory cells that form in the zebrafish neuromasts, and that fish lacking Kremen1 protein develop more hair cells per neuromast compared to wild type fish. Collectively, these data support an inhibitory role for Kremen1 in hair cell fate specification.


Assuntos
Cóclea/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/metabolismo , Sistema da Linha Lateral/metabolismo , Proteínas de Membrana/genética , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Cóclea/embriologia , Cóclea/crescimento & desenvolvimento , Sistema da Linha Lateral/embriologia , Sistema da Linha Lateral/crescimento & desenvolvimento , Mecanorreceptores/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Neurogênese/genética , Interferência de RNA , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
16.
J Neurosci ; 36(33): 8734-45, 2016 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-27535918

RESUMO

UNLABELLED: The generation of hair cells (HCs) from the differentiation of proliferating supporting cells (SCs) appears to be an ideal approach for replacing lost HCs in the cochlea and is promising for restoring hearing after damage to the organ of Corti. We show here that extensive proliferation of SCs followed by mitotic HC generation is achieved through a genetic reprogramming process involving the activation of ß-catenin to upregulate Wnt signaling, the deletion of Notch1 to downregulate Notch signaling, and the overexpression of Atoh1 in Sox2(+) SCs in neonatal mouse cochleae. We used RNA sequencing to compare the transcripts of the cochleae from control mice and from mice with ß-catenin activation, Notch1 deletion, and ß-catenin activation combined with Notch1 deletion in Sox2(+) SCs. We identified the genes involved in the proliferation and transdifferentiation process that are either controlled by individual signaling pathways or by the combination of Wnt and Notch signaling. Moreover, the proliferation of SCs induced by Notch1 deletion disappears after deleting ß-catenin in Notch1 knock-out Sox2(+) cells, which further demonstrates that Notch signaling is an upstream and negative regulator of Wnt signaling. SIGNIFICANCE STATEMENT: We show here that the extensive proliferation of supporting cells (SCs) and the subsequent mitotic hair cell (HC) generation is achieved through a genetic reprogramming process involving activation of ß-catenin to upregulate Wnt signaling, deletion of Notch1 to downregulate Notch signaling, and overexpression of Atoh1 in Sox2(+) SCs in neonatal mice cochleae. By comparing the transcripts of the cochleae among controls, ß-catenin activation, Notch1 deletion, and ß-catenin activation combined with Notch1 deletion group, we identified multiple genes involved in the proliferation and transdifferentiation process that are either controlled by individual signaling pathways or by the combination of Wnt and Notch signaling. This provides a better understanding of the mechanisms behind mitotic HC generation and might provide new approaches to stimulating mitotic HC regeneration.


Assuntos
Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Cóclea/citologia , Regulação da Expressão Gênica/genética , Células Ciliadas Auditivas/fisiologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Ciclo Celular/genética , Diferenciação Celular/fisiologia , Cóclea/crescimento & desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miosina VIIa , Miosinas/genética , Miosinas/metabolismo , Compostos de Fenilureia/metabolismo , Receptor Notch3/genética , Receptor Notch3/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/genética , beta Catenina/metabolismo
17.
J Neurosci ; 36(31): 8200-9, 2016 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-27488639

RESUMO

UNLABELLED: The stria vascularis is a nonsensory structure that is essential for auditory hair cell function by maintaining potassium concentration of the scala media. During mouse embryonic development, a subpopulation of neural crest cell-derived melanocytes migrates and incorporates into a subregion of the cochlear epithelium, forming the intermediate cell layer of the stria vascularis. The relation of this developmental process to stria vascularis function is currently unknown. In characterizing the molecular differentiation of developing peripheral auditory structures, we discovered that hepatocyte growth factor (Hgf) is expressed in the future stria vascularis of the cochlear epithelium. Its receptor tyrosine kinase, c-Met, is expressed in the cochlear epithelium and melanocyte-derived intermediate cells in the stria vascularis. Genetic dissection of HGF signaling via c-MET reveals that the incorporation of the melanocytes into the future stria vascularis of the cochlear duct requires c-MET signaling. In addition, inactivation of either the ligand or receptor developmentally resulted in a profound hearing loss at young adult stages. These results suggest a novel connection between HGF signaling and deafness via melanocyte deficiencies. SIGNIFICANCE STATEMENT: We found the roles of hepatocyte growth factor (HGF) signaling in stria vascularis development for the first time and that lack of HGF signaling in the inner ear leads to profound hearing loss in the mouse. Our findings reveal a novel mechanism that may underlie human deafness DFNB39 and DFNB97. Our findings reveal an additional example of context-dependent c-MET signaling diversity, required here for proper cellular invasion developmentally that is essential for specific aspects of auditory-related organogenesis.


Assuntos
Perda Auditiva/metabolismo , Audição , Fator de Crescimento de Hepatócito/metabolismo , Melanócitos/metabolismo , Melanócitos/fisiologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Cóclea/embriologia , Cóclea/crescimento & desenvolvimento , Cóclea/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Perda Auditiva/embriologia , Perda Auditiva/patologia , Masculino , Camundongos , Transdução de Sinais , Estria Vascular/patologia
18.
Laryngoscope ; 126(11): E379-E385, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27328420

RESUMO

OBJECTIVES/HYPOTHESIS: The purpose of this study was to investigate the effect of early postnatal neurotrophin-3 (NT3) support on hearing acquisition. STUDY DESIGN: A prospective experimental animal study. METHODS: Adenoviral (Ad) vectors expressing green fluorescence protein (GFP) alone or in combination with NT3 were injected into the scala tympani through the round window of 5-postnatal-day-old (P5) rats. Changes in NT3 mRNA level, hearing thresholds, and morphological studies were done after the viral vector injection. RESULTS: NT3 mRNA was significantly increased in the Ad-GFP-NT3 group compared to the normal-developmental group and Ad-GFP alone group. GFP was widely expressed in the cochlea such as in the hair cells, supporting cell area, and spiral ganglion neurons. Auditory brainstem response thresholds were significantly lower in the Ad-GFP-NT3 group compared to the normal-developmental group and Ad-GFP alone group at P15. CONCLUSIONS: These results show that early postnatal NT3 overexpression may accelerate the acquisition of hearing in rats. LEVEL OF EVIDENCE: NA Laryngoscope, 126:E379-E385, 2016.


Assuntos
Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/administração & dosagem , Audição/genética , Neurotrofina 3/genética , RNA Mensageiro/metabolismo , Animais , Cóclea/crescimento & desenvolvimento , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Estudos Prospectivos , Ratos , Ratos Sprague-Dawley , Janela da Cóclea , Rampa do Tímpano
19.
Eur J Histochem ; 59(2): 2486, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-26150157

RESUMO

Inositol 1, 4, 5-trisphosphate receptor (IP3R) has been established to be essential for hearing. However, the expression of IP3R in the cochlea in the period of auditory development remains unknown. We investigated the expression of IP3R in the developing rat cochlea using immunohistochemistry and real-time reverse transcription polymerase chain reaction (RT-PCR). We observed its presence in the developing rat cochlea, and changes in IP3R protein expressions from the early post-natal period to adult. At birth (post-natal day 0, P0), IP3R expression was only found in Hensen's cell. IP3R immunoreactivity first appeared in the sensory hair cells in the organ of Corti at P2. This localization was confirmed by means of double-labeling experiments with Myosin VIIA, a marker for cochlear hair cells. Colocalization of IP3R and Myosin VIIA from P2 to the second post-natal week suggested early expression of IP3R in developing inner and outer hair cells. Claudius' cells near the spiral ligament were labelled for IP3R from P8 onwards. Transient IP3R expression was observed in the stria vascularis in early post-natal rat from P4 to P8. Spiral ganglion neurons also exhibited weaker IP3R fluorescence signals during post-natal development. The results of RT-PCR demonstrated that all three IP3R isoforms (IP3R1, IP3R2, and IP3R3) were present in rat cochlea during four different developmental stages of cochlea, from P0 to P28. Present immunohistochemical evidence for both change and maintenance of expression of IP3R during post-natal development of the rat cochlea indicated the possible involvement of IP3R-mediated calcium signaling in cochlear development.


Assuntos
Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Animais , Western Blotting , Sinalização do Cálcio/fisiologia , Feminino , Células Ciliadas Auditivas Internas/metabolismo , Imuno-Histoquímica , Masculino , Microscopia Confocal , Miosina VIIa , Miosinas/metabolismo , Órgão Espiral/crescimento & desenvolvimento , Órgão Espiral/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/metabolismo , Estria Vascular/metabolismo
20.
Artigo em Chinês | MEDLINE | ID: mdl-25989665

RESUMO

OBJECTIVE: To study the apoptosis/proliferation of Kölliker organ supporting cells and to understand the prompting apoptosis factors in vivo in the supporting cells in the Kölliker organ by changing the environment of the cultured supporting cells in the Kliker organ in vitro, via the separation, culture and purification of the supporting cells in the K6lliker organ. METHOD: A combinatorial approach of enzymatic digestion and mechanical separation was employed to isolate and culture in vitro pure Kölliker organ supporting cells. The purity was tested by flow cytometry assay. And K6lliker organ supporting cells were harvested to detect the rate and cycle of apoptosis by flow cytometry after Annexin V/PI staining, to test the cell growth curve by MTT assay, and to observe the differential expressions of the Bcl-2, Caspase-3, Caspase-8 and Caspase-9 through the Realtime PCR and Western blot. The calcium, potassium and glutamate concentrations in the culture medium of these cells in vitro were changed to detect the survival rate of cells by MTT assay. RESULT: The purity of K6lliker organ supporting cells by flow cytometry assay was 96. 56%. And these cells showed no significant difference in apoptosis, but an evident linear growth. The results of Realtime PCR and Western blot showed that the expression of Bcl-2, Caspase-3, Caspase-8 and Caspase-9 mRNA and protein in all different time points kept stable. Furthermore, the elevation of extracellular Ca2+ might contribute to decrease the cell viability of supporting cells. And K+ participated regulation of cell viability in a concentration-depending way. However, glutamate appeared to be a protective factor in high concentration. CONCLUSION: There is no significant apoptosis in vitro of the supporting cells in the Kölliker organ of rats, showing a linear growth. The Ca2+ in high concentration might contribute to the apoptosis factor of these cells. However, the K+ and glutamate appear to be protective factors in high concentration.


Assuntos
Apoptose , Proliferação de Células , Cóclea/citologia , Animais , Animais Recém-Nascidos , Caspase 3 , Ciclo Celular , Sobrevivência Celular , Cóclea/crescimento & desenvolvimento , Citometria de Fluxo , Técnicas In Vitro , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA