Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Biomed Environ Sci ; 34(2): 110-118, 2021 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33685569

RESUMO

OBJECTIVE: The aim of this study was to explore the ototoxicity of toluene in the early development of zebrafish embryos/larvae. METHODS: Zebrafish were utilized to explore the ototoxicity of toluene. Locomotion analysis, immunofluorescence, and qPCR were used to understand the phenotypes and molecular mechanisms of toluene ototoxicity. RESULTS: The results demonstrated that at 2 mmol/L, toluene induced zebrafish larvae death at 120 hours post fertilization (hpf) at a rate of 25.79% and inhibited the rate of hatching at 72 hpf. Furthermore, toluene exposure inhibited the distance travelled and average swimming velocity of zebrafish larvae while increasing the frequency of movements. As shown by fluorescence staining of hair cells, toluene inhibited the formation of lateral line neuromasts and middle line 1 (Ml 1) neuromasts in 3 days post fertilization larvae in a concentration-dependent manner. Toluene altered the expression level of genes involved in ear development/function in zebrafish, among which the mRNA levels of cd164l2, tekt3, and pcsk5a were upregulated, while the level of otofb was downregulated, according to the qPCR results. CONCLUSION: This study indicated that toluene may affect the development of both the inner ear and lateral line systems in zebrafish, while the lateral line system may be more sensitive to toluene than the inner ear.


Assuntos
Orelha Interna/efeitos dos fármacos , Sistema da Linha Lateral/efeitos dos fármacos , Tolueno/toxicidade , Animais , Orelha Interna/crescimento & desenvolvimento , Embrião não Mamífero/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Sistema da Linha Lateral/crescimento & desenvolvimento , Locomoção/efeitos dos fármacos , Ototoxicidade/etiologia , Ototoxicidade/patologia , Ototoxicidade/fisiopatologia , Peixe-Zebra
3.
Sci Rep ; 9(1): 5912, 2019 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-30976015

RESUMO

Embryonic development requires the coordinated regulation of apoptosis, survival, autophagy, proliferation and differentiation programs. Senescence has recently joined the cellular processes required to master development, in addition to its well-described roles in cancer and ageing. Here, we show that senescent cells are present in a highly regulated temporal pattern in the developing vertebrate inner ear, first, surrounding the otic pore and, later, in the otocyst at the endolymphatic duct. Cellular senescence is associated with areas of increased apoptosis and reduced proliferation consistent with the induction of the process when the endolymphatic duct is being formed. Modulation of senescence disrupts otic vesicle morphology. Transforming growth factor beta (TGFß) signaling interacts with signaling pathways elicited by insulin-like growth factor type 1 (IGF-1) to jointly coordinate cellular dynamics required for morphogenesis and differentiation. Taken together, these results show that senescence is a natural occurring process essential for early inner ear development.


Assuntos
Diferenciação Celular , Senescência Celular , Orelha Interna/crescimento & desenvolvimento , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Organogênese , Fator de Crescimento Transformador beta2/metabolismo , Animais , Galinhas , Orelha Interna/metabolismo , Embrião de Mamíferos/metabolismo , Camundongos , Transdução de Sinais , Fator de Crescimento Transformador beta2/genética
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.
Hum Genet ; 137(6-7): 459-470, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29955957

RESUMO

Congenital inner ear malformations affecting both the osseous and membranous labyrinth can have a devastating impact on hearing and language development. With the exception of an enlarged vestibular aqueduct, non-syndromic inner ear malformations are rare, and their underlying molecular biology has thus far remained understudied. To identify molecular factors that might be important in the developing inner ear, we adopted a family-based trio exome sequencing approach in young unrelated subjects with severe inner ear malformations. We identified two previously unreported de novo loss-of-function variants in GREB1L [c.4368G>T;p.(Glu1410fs) and c.982C>T;p.(Arg328*)] in two affected subjects with absent cochleae and eighth cranial nerve malformations. The cochlear aplasia in these affected subjects suggests that a developmental arrest or problem at a very early stage of inner ear development exists, e.g., during the otic pit formation. Craniofacial Greb1l RNA expression peaks in mice during this time frame (E8.5). It also peaks in the developing inner ear during E13-E16, after which it decreases in adulthood. The crucial function of Greb1l in craniofacial development is also evidenced in knockout mice, which develop severe craniofacial abnormalities. In addition, we show that Greb1l-/- zebrafish exhibit a loss of abnormal sensory epithelia innervation. An important role for Greb1l in sensory epithelia innervation development is supported by the eighth cranial nerve deficiencies seen in both affected subjects. In conclusion, we demonstrate that GREB1L is a key player in early inner ear and eighth cranial nerve development. Abnormalities in cochleovestibular anatomy can provide challenges for cochlear implantation. Combining a molecular diagnosis with imaging techniques might aid the development of individually tailored therapeutic interventions in the future.


Assuntos
Surdez/genética , Doenças do Labirinto/genética , Proteínas de Neoplasias/genética , Proteínas/genética , Proteínas de Peixe-Zebra/genética , Animais , Surdez/fisiopatologia , Modelos Animais de Doenças , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/fisiopatologia , Células Epiteliais/patologia , Gânglios Parassimpáticos/crescimento & desenvolvimento , Gânglios Parassimpáticos/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Doenças do Labirinto/fisiopatologia , Proteínas de Membrana , Camundongos , Camundongos Knockout , Peixe-Zebra
6.
Hum Mutat ; 37(10): 991-1003, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27375115

RESUMO

Deafness in humans is a common neurosensory disorder and is genetically heterogeneous. Across diverse ethnic groups, mutations of MYO15A at the DFNB3 locus appear to be the third or fourth most common cause of autosomal-recessive, nonsyndromic deafness. In 49 of the 67 exons of MYO15A, there are currently 192 recessive mutations identified, including 14 novel mutations reported here. These mutations are distributed uniformly across MYO15A with one enigmatic exception; the alternatively spliced giant exon 2, encoding 1,233 residues, has 17 truncating mutations but no convincing deafness-causing missense mutations. MYO15A encodes three distinct isoform classes, one of which is 395 kDa (3,530 residues), the largest member of the myosin superfamily of molecular motors. Studies of Myo15 mouse models that recapitulate DFNB3 revealed two different pathogenic mechanisms of hearing loss. In the inner ear, myosin 15 is necessary both for the development and the long-term maintenance of stereocilia, mechanosensory sound-transducing organelles that extend from the apical surface of hair cells. The goal of this Mutation Update is to provide a comprehensive review of mutations and functions of MYO15A.


Assuntos
Surdez/genética , Surdez/patologia , Mutação , Miosinas/genética , Miosinas/metabolismo , Processamento Alternativo , Animais , Surdez/metabolismo , Modelos Animais de Doenças , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Orelha Interna/patologia , Éxons , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Estereocílios/metabolismo , Estereocílios/patologia
7.
Cell ; 163(6): 1348-59, 2015 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-26627734

RESUMO

Spontaneous electrical activity of neurons in developing sensory systems promotes their maturation and proper connectivity. In the auditory system, spontaneous activity of cochlear inner hair cells (IHCs) is initiated by the release of ATP from glia-like inner supporting cells (ISCs), facilitating maturation of central pathways before hearing onset. Here, we find that ATP stimulates purinergic autoreceptors in ISCs, triggering Cl(-) efflux and osmotic cell shrinkage by opening TMEM16A Ca(2+)-activated Cl(-) channels. Release of Cl(-) from ISCs also forces K(+) efflux, causing transient depolarization of IHCs near ATP release sites. Genetic deletion of TMEM16A markedly reduces the spontaneous activity of IHCs and spiral ganglion neurons in the developing cochlea and prevents ATP-dependent shrinkage of supporting cells. These results indicate that supporting cells in the developing cochlea have adapted a pathway used for fluid secretion in other organs to induce periodic excitation of hair cells.


Assuntos
Orelha Interna/crescimento & desenvolvimento , Células Ciliadas Auditivas/citologia , Trifosfato de Adenosina/metabolismo , Animais , Anoctamina-1 , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Orelha Interna/citologia , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Labirínticas de Suporte/citologia , Células Labirínticas de Suporte/metabolismo , Camundongos , Camundongos Knockout , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/metabolismo
8.
PLoS One ; 10(9): e0136832, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26375458

RESUMO

Hearing loss is one of the most prevalent human birth defects. Genetic factors contribute to the pathogenesis of deafness. It is estimated that one-third of deafness genes have already been identified. The current work is an attempt to find novel genes relevant to hearing loss using guilt-by-profiling and guilt-by-association bioinformatics analyses of approximately 80 known non-syndromic hereditary hearing loss (NSHL) genes. Among the 300 newly identified candidate deafness genes, slc26a2 were selected for functional studies in zebrafish. The slc26a2 gene was knocked down using an antisense morpholino (MO), and significant defects were observed in otolith patterns, semicircular canal morphology, and lateral neuromast distributions in morphants. Loss-of-function defects are caused primarily by apoptosis, and morphants are insensitive to sound stimulation and imbalanced swimming behaviours. Morphant defects were found to be partially rescued by co-injection of human SLC26A2 mRNA. All the results suggest that bioinformatics is capable of predicting new deafness genes and this showed slc26a2 is to be a critical otic gene whose dysfunction may induce hearing impairment.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Células Ciliadas Auditivas/citologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Proteínas de Transporte de Ânions/deficiência , Proteínas de Transporte de Ânions/genética , Apoptose , Sobrevivência Celular , Cílios , Biologia Computacional , Surdez/genética , Surdez/metabolismo , Surdez/fisiopatologia , Orelha Interna/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Audição , Humanos , Larva , Transportadores de Sulfato , Natação , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
9.
PLoS Genet ; 11(3): e1005037, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25781991

RESUMO

Neurons of the statoacoustic ganglion (SAG) transmit auditory and vestibular information from the inner ear to the hindbrain. SAG neuroblasts originate in the floor of the otic vesicle. New neuroblasts soon delaminate and migrate towards the hindbrain while continuing to proliferate, a phase known as transit amplification. SAG cells eventually come to rest between the ear and hindbrain before terminally differentiating. Regulation of these events is only partially understood. Fgf initiates neuroblast specification within the ear. Subsequently, Fgf secreted by mature SAG neurons exceeds a maximum threshold, serving to terminate specification and delay maturation of transit-amplifying cells. Notch signaling also limits SAG development, but how it is coordinated with Fgf is unknown. Here we show that transcription factor Tfap2a coordinates multiple signaling pathways to promote neurogenesis in the zebrafish inner ear. In both zebrafish and chick, Tfap2a is expressed in a ventrolateral domain of the otic vesicle that includes neurogenic precursors. Functional studies were conducted in zebrafish. Loss of Tfap2a elevated Fgf and Notch signaling, thereby inhibiting SAG specification and slowing maturation of transit-amplifying cells. Conversely, overexpression of Tfap2a inhibited Fgf and Notch signaling, leading to excess and accelerated SAG production. However, most SAG neurons produced by Tfap2a overexpression died soon after maturation. Directly blocking either Fgf or Notch caused less dramatic acceleration of SAG development without neuronal death, whereas blocking both pathways mimicked all observed effects of Tfap2a overexpression, including apoptosis of mature neurons. Analysis of genetic mosaics showed that Tfap2a acts non-autonomously to inhibit Fgf. This led to the discovery that Tfap2a activates expression of Bmp7a, which in turn inhibits both Fgf and Notch signaling. Blocking Bmp signaling reversed the effects of overexpressing Tfap2a. Together, these data support a model in which Tfap2a, acting through Bmp7a, modulates Fgf and Notch signaling to control the duration, amount and speed of SAG neural development.


Assuntos
Proteína Morfogenética Óssea 7/genética , Cistos Glanglionares/genética , Neurogênese/genética , Fator de Transcrição AP-2/biossíntese , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética , Animais , Proteína Morfogenética Óssea 7/biossíntese , Diferenciação Celular/genética , Galinhas , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Cistos Glanglionares/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética , Fator de Transcrição AP-2/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
10.
Otol Neurotol ; 36(4): 727-32, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25406873

RESUMO

HYPOTHESIS: A number of bone-related genes may be responsible for the unique suppression of perilabyrinthine bone remodeling. BACKGROUND: Bone remodeling is highly inhibited around the inner ear space most likely because of osteoprotegerin (OPG), which is a well-known potent inhibitor of osteoclast formation and function. However, other signaling molecules may also be responsible for the inhibition of bone remodeling within the otic capsule. METHODS: Microarray technology was used to determine bone-related genes differentially expressed between the lining tissues of the otic capsule (spiral ligament and stria vascularis) and the lining tissues from the middle ear of the rat. Data was analyzed with statistical bioinformatics tools. Gene expression levels of selected genes were validated using quantitative polymerase chain reaction. RESULTS: A total of 413 genes were identified when young inner bulla (growing) were compared with young otic capsule and 358 genes were identified when adult inner bulla (quiescent) were compared with adult otic capsule. Fourteen genes were involved in bone metabolism of which four genes have been previously discussed in the literature of perilabyrinthine bone biology. CONCLUSION: The gene expression of the otic capsule was significantly different from that of the middle ear. This study identified a number of differentially expressed bone-related mRNAs of potential significance and confirmed the OPG/receptor activator of nuclear factor kappa-B (RANK)/RANK ligand (RANKL) pathway as the key signaling system for the unique behavior of bone cells within the otic capsule. No differentially expressed up- or downstream messengers in the OPG/RANK/RANKL pathway were found.


Assuntos
Remodelação Óssea/fisiologia , Orelha Média/metabolismo , Expressão Gênica/fisiologia , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Animais , Osso e Ossos/metabolismo , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Orelha Média/crescimento & desenvolvimento , Regulação da Expressão Gênica , Osteoprotegerina/genética , Ratos , Transdução de Sinais/genética
11.
Curr Top Dev Biol ; 110: 75-127, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25248474

RESUMO

Proneural genes encode evolutionarily conserved basic-helix-loop-helix transcription factors. In Drosophila, proneural genes are required and sufficient to confer a neural identity onto naïve ectodermal cells, inducing delamination and subsequent neuronal differentiation. In vertebrates, proneural genes are expressed in cells that already have a neural identity, but they are still required and sufficient to initiate neurogenesis. In all organisms, proneural genes control neurogenesis by regulating Notch-mediated lateral inhibition and initiating the expression of downstream differentiation genes. The general mode of proneural gene function has thus been elucidated. However, the regulatory mechanisms that spatially and temporally control proneural gene function are only beginning to be deciphered. Understanding how proneural gene function is regulated is essential, as aberrant proneural gene expression has recently been linked to a variety of human diseases-ranging from cancer to neuropsychiatric illnesses and diabetes. Recent insights into proneural gene function in development and disease are highlighted herein.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Deficiências do Desenvolvimento/genética , Diabetes Mellitus/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Orelha Interna/crescimento & desenvolvimento , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Humanos , Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso/genética , Neurônios/citologia , Retina/fisiologia , Vertebrados/genética
12.
Dev Dyn ; 243(10): 1317-27, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24847848

RESUMO

BACKGROUND: The otic placode comprises the progenitors of the inner ear and the neurons that convey hearing and balance information to the brain. Transplantation studies in birds and amphibians demonstrate that when the otic placode is morphologically visible as a thickened patch of ectoderm, it is first committed to an otic fate. Fibroblast growth factor (FGF) signaling initiates induction of the otic placode, and levels of FGF signaling are fine-tuned by the Sprouty family of antagonists of receptor tyrosine kinase signaling. RESULTS: Here, we examined the size of the otic placode and cup by combinatorial inactivation of the Sprouty1 and Sprouty2 genes. Interestingly, in a Sprouty gene dosage series, early enlargement of the otic placode was progressively restored to normal. Restoration of otic size was preceded by normal levels of FGF signaling, reduced cell proliferation and reduced cell death. CONCLUSIONS: Our study demonstrates that excess otic placode cells, which form in response to increased FGF signaling, are not maintained in mammals. This suggests that growth plasticity exists in the mammalian otic placode and cup, and that FGF signaling may not be sufficient to induce the genetic program that maintains otic fate.


Assuntos
Orelha Interna/embriologia , Indução Embrionária , Células-Tronco Embrionárias/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Orelha/embriologia , Orelha/crescimento & desenvolvimento , Orelha Interna/crescimento & desenvolvimento , Embrião de Mamíferos , Indução Embrionária/genética , Fator 3 de Crescimento de Fibroblastos/genética , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Tamanho do Órgão , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases , Transdução de Sinais/genética
13.
Nat Protoc ; 9(6): 1229-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24784820

RESUMO

This protocol describes a culture system in which inner-ear sensory tissue is produced from mouse embryonic stem (ES) cells under chemically defined conditions. This model is amenable to basic and translational investigations into inner ear biology and regeneration. In this protocol, mouse ES cells are aggregated in 96-well plates in medium containing extracellular matrix proteins to promote epithelialization. During the first 14 d, a series of precisely timed protein and small-molecule treatments sequentially induce epithelia that represent the mouse embryonic non-neural ectoderm, preplacodal ectoderm and otic vesicle epithelia. Ultimately, these tissues develop into cysts with a pseudostratified epithelium containing inner ear hair cells and supporting cells after 16-20 d. Concurrently, sensory-like neurons generate synapse-like structures with the derived hair cells. We have designated the stem cell-derived epithelia harboring hair cells, supporting cells and sensory-like neurons as inner ear organoids. This method provides a reproducible and scalable means to generate inner ear sensory tissue in vitro.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Orelha Interna/crescimento & desenvolvimento , Células-Tronco Embrionárias/citologia , Organoides/crescimento & desenvolvimento , Animais , Células-Tronco Embrionárias/fisiologia , Proteínas da Matriz Extracelular/metabolismo , Células Ciliadas Auditivas/citologia , Camundongos
14.
Hum Mol Genet ; 23(12): 3289-98, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24482543

RESUMO

Cyclic AMP (cAMP) production, which is important for mechanotransduction within the inner ear, is catalyzed by adenylate cyclases (AC). However, knowledge of the role of ACs in hearing is limited. Previously, a novel autosomal recessive non-syndromic hearing impairment locus DFNB44 was mapped to chromosome 7p14.1-q11.22 in a consanguineous family from Pakistan. Through whole-exome sequencing of DNA samples from hearing-impaired family members, a nonsense mutation c.3112C>T (p.Arg1038*) within adenylate cyclase 1 (ADCY1) was identified. This stop-gained mutation segregated with hearing impairment within the family and was not identified in ethnically matched controls or within variant databases. This mutation is predicted to cause the loss of 82 amino acids from the carboxyl tail, including highly conserved residues within the catalytic domain, plus a calmodulin-stimulation defect, both of which are expected to decrease enzymatic efficiency. Individuals who are homozygous for this mutation had symmetric, mild-to-moderate mixed hearing impairment. Zebrafish adcy1b morphants had no FM1-43 dye uptake and lacked startle response, indicating hair cell dysfunction and gross hearing impairment. In the mouse, Adcy1 expression was observed throughout inner ear development and maturation. ADCY1 was localized to the cytoplasm of supporting cells and hair cells of the cochlea and vestibule and also to cochlear hair cell nuclei and stereocilia. Ex vivo studies in COS-7 cells suggest that the carboxyl tail of ADCY1 is essential for localization to actin-based microvilli. These results demonstrate that ADCY1 has an evolutionarily conserved role in hearing and that cAMP signaling is important to hair cell function within the inner ear.


Assuntos
Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Perda Auditiva/patologia , Adenilil Ciclases/química , Animais , Células COS , Chlorocebus aethiops , Códon sem Sentido , Citoplasma/metabolismo , Orelha Interna/crescimento & desenvolvimento , Feminino , Perda Auditiva/enzimologia , Humanos , Células Labirínticas de Suporte/metabolismo , Masculino , Camundongos , Peixe-Zebra/genética
15.
Stem Cells Dev ; 23(5): 502-14, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24172073

RESUMO

A loss of sensory hair cells or spiral ganglion neurons from the inner ear causes deafness, affecting millions of people. Currently, there is no effective therapy to repair the inner ear sensory structures in humans. Cochlear implantation can restore input, but only if auditory neurons remain intact. Efforts to develop stem cell-based treatments for deafness have demonstrated progress, most notably utilizing embryonic-derived cells. In an effort to bypass limitations of embryonic or induced pluripotent stem cells that may impede the translation to clinical applications, we sought to utilize an alternative cell source. Here, we show that adult human mesenchymal-like stem cells (MSCs) obtained from nasal tissue can repair spiral ganglion loss in experimentally lesioned cochlear cultures from neonatal rats. Stem cells engraft into gentamicin-lesioned organotypic cultures and orchestrate the restoration of the spiral ganglion neuronal population, involving both direct neuronal differentiation and secondary effects on endogenous cells. As a physiologic assay, nasal MSC-derived cells engrafted into lesioned spiral ganglia demonstrate responses to infrared laser stimulus that are consistent with those typical of excitable cells. The addition of a pharmacologic activator of the canonical Wnt/ß-catenin pathway concurrent with stem cell treatment promoted robust neuronal differentiation. The availability of an effective adult autologous cell source for inner ear tissue repair should contribute to efforts to translate cell-based strategies to the clinic.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Regeneração Nervosa , Adulto , Animais , Cóclea/crescimento & desenvolvimento , Cóclea/lesões , Cóclea/patologia , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/patologia , Humanos , Neurônios/patologia , Ratos , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/lesões , Gânglio Espiral da Cóclea/patologia , Via de Sinalização Wnt/genética
16.
PLoS Genet ; 9(9): e1003824, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086156

RESUMO

Lrig proteins are conserved transmembrane proteins that modulate a variety of signaling pathways from worm to humans. In mammals, there are three family members - Lrig1, Lrig2, and Lrig3--that are defined by closely related extracellular domains with a similar arrangement of leucine rich repeats and immunoglobulin domains. However, the intracellular domains show little homology. Lrig1 inhibits EGF signaling through internalization and degradation of ErbB receptors. Although Lrig3 can also bind ErbB receptors in vitro, it is unclear whether Lrig2 and Lrig3 exhibit similar functions to Lrig1. To gain insights into Lrig gene functions in vivo, we compared the expression and function of the Lrigs in the inner ear, which offers a sensitive system for detecting effects on morphogenesis and function. We find that all three family members are expressed in the inner ear throughout development, with Lrig1 and Lrig3 restricted to subsets of cells and Lrig2 expressed more broadly. Lrig1 and Lrig3 overlap prominently in the developing vestibular apparatus and simultaneous removal of both genes disrupts inner ear morphogenesis. This suggests that these two family members act redundantly in the otic epithelium. In contrast, although Lrig1 and Lrig2 are frequently co-expressed, Lrig1(-/-);Lrig2(-/-) double mutant ears show no enhanced structural abnormalities. At later stages, Lrig1 expression is sustained in non-sensory tissues, whereas Lrig2 levels are enhanced in neurons and sensory epithelia. Consistent with these distinct expression patterns, Lrig1 and Lrig2 mutant mice exhibit different forms of impaired auditory responsiveness. Notably, Lrig1(-/-);Lrig2(-/-) double mutant mice display vestibular deficits and suffer from a more severe auditory defect that is accompanied by a cochlear innervation phenotype not present in single mutants. Thus, Lrig genes appear to act both redundantly and independently, with Lrig2 emerging as the most functionally distinct family member.


Assuntos
Orelha Interna/crescimento & desenvolvimento , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Morfogênese/genética , Proteínas do Tecido Nervoso/genética , Animais , Citoplasma/genética , Citoplasma/metabolismo , Orelha Interna/metabolismo , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Epitélio , Regulação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transdução de Sinais , Vestíbulo do Labirinto/crescimento & desenvolvimento , Vestíbulo do Labirinto/metabolismo
17.
Neurosci Lett ; 555: 62-7, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24060674

RESUMO

Sensory cells contain ion channels involved in the organ-specific transduction mechanisms that convert different types of stimuli into electric energy. Here we focus on small-conductance calcium-activated potassium channel 1 (SK1) which plays an important role in all excitable cells acting as feedback regulators in after-hyperpolarization. This study was undertaken to analyze the pattern of expression of SK1 in the zebrafish peripheral nervous system and sensory organs using RT-PRC, Westernblot and immunohistochemistry. Expression of SK1 mRNA was observed at all developmental stages analyzed (from 10 to 100 days post fertilization, dpf), and the antibody used identified a protein with a molecular weight of 70kDa, at 100dpf (regarded to be adult). Cell expressing SK1 in adult animals were neurons of dorsal root and cranial nerve sensory ganglia, sympathetic neurons, sensory cells in neuromasts of the lateral line system and taste buds, crypt olfactory neurons and photoreceptors. Present results report for the first time the expression and the distribution of SK1 in the peripheral nervous system and sensory organs of adult zebrafish, and may contribute to set zebrafish as an interesting experimental model for calcium-activated potassium channels research. Moreover these findings are of potential interest because the potential role of SK as targets for the treatment of neurological diseases and sensory disorders.


Assuntos
Sistema Nervoso Periférico/metabolismo , Órgãos dos Sentidos/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Neurônios/metabolismo , Mucosa Olfatória/crescimento & desenvolvimento , Mucosa Olfatória/metabolismo , Especificidade de Órgãos , Sistema Nervoso Periférico/crescimento & desenvolvimento , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Órgãos dos Sentidos/crescimento & desenvolvimento , Papilas Gustativas/crescimento & desenvolvimento , Papilas Gustativas/metabolismo , Peixe-Zebra/crescimento & desenvolvimento
18.
Zebrafish ; 10(1): 52-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23461415

RESUMO

Prenatal alcohol exposure is known to have many profound detrimental effects on human fetal development (fetal alcohol spectrum disorders), which may manifest as lifelong disabilities. However, how alcohol affects the auditory/vestibular system is still largely unknown. This is the first study to investigate morphological effects of alcohol on the developing octavolateral system (the inner ear and lateral line) using the zebrafish, Danio rerio. Zebrafish embryos of 2 hours post fertilization (hpf) were treated in 2% alcohol for 48 hours and screened at 72 hpf for morphological defects of the inner ear and lateral line. Octavolateral organs from both alcohol-treated and control zebrafish were examined using light, confocal, and scanning electron microscopy. We observed several otolith phenotypes for alcohol-treated zebrafish including zero, one, two abnormal, two normal, and multiple otoliths. Results of this study show that alcohol treatment during early development impairs the inner ear (smaller ear, abnormal otoliths, and fewer sensory hair cells) and the lateral line (smaller neuromasts, fewer neuromasts and hair cells per neuromast, and shorter kinocilia of hair cells). Early embryonic alcohol exposure may also result in defects in hearing, balance, and hydrodynamic function of zebrafish.


Assuntos
Orelha Interna/efeitos dos fármacos , Etanol/farmacologia , Sistema da Linha Lateral/efeitos dos fármacos , Mecanorreceptores/efeitos dos fármacos , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Relação Dose-Resposta a Droga , Orelha Interna/embriologia , Orelha Interna/crescimento & desenvolvimento , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/embriologia , Larva/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Sistema da Linha Lateral/embriologia , Sistema da Linha Lateral/crescimento & desenvolvimento , Mecanorreceptores/fisiologia , Microscopia Confocal , Microscopia Eletrônica de Varredura
19.
PLoS One ; 8(2): e55796, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23405214

RESUMO

The serine protease CAP1/Prss8 is crucial for skin barrier function, lung alveolar fluid clearance and has been unveiled as diagnostic marker for specific cancer types. Here, we show that a constitutive knockout of CAP1/Prss8 leads to embryonic lethality. These embryos presented no specific defects, but it is during this period, and in particular at E13.5, that wildtype placentas show an increased expression of CAP1/Prss8, thus suggesting a placental defect in the knockout situation. The placentas of knockout embryos exhibited significantly reduced vascular development and incomplete cellular maturation. In contrary, epiblast-specific deletion of CAP1/Prss8 allowed development until birth. These CAP1/Prss8-deficient newborns presented abnormal epidermis, and died soon after birth due to impaired skin function. We thus conclude that a late placental insufficiency might be the primary cause of embryonic lethality in CAP1/Prss8 knockouts. This study highlights a novel and crucial role for CAP1/Prss8 in placental development and function.


Assuntos
Orelha Interna/crescimento & desenvolvimento , Perda do Embrião/genética , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Placentação/fisiologia , Serina Endopeptidases/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Embrião de Mamíferos/enzimologia , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real
20.
PLoS One ; 8(2): e55609, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23393595

RESUMO

All cellular phenomena and developmental events, including inner ear development, are modulated through harmonized signaling networks. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor, is a major signaling component involved in cross talk with key regulators of development; i.e., Wnt, Notch, and bone morphogenetic proteins. Although Pten function has been studied in various systems, its role in inner ear development is poorly understood. Here, we used inner ear-specific Pten conditional knockout mice and examined the characteristics of the inner ear. In a detailed analysis of the phenotype, reduced cochlear turning and widened epithelia were observed. Phalloidin staining of sensory epithelium revealed that hair cell patterns were disturbed; i.e., additional rows of hair cells were discovered. The neural abnormality revealed a reduction in and disorganization of nerve fibers, including apoptosis at the neural precursor stage. Pten deficiency induced increased phosphorylation of Akt at Ser473. The elevation of inhibitory glycogen synthase kinase 3ß Ser9 phosphorylation (pGSK3ß) was sustained until the neuronal differentiation stage at embryonic day 14.5, instead of pGSK3ß downregulation. This is the first report on the influence of Pten/Akt/GSK3ß signaling on the development of spiral ganglia. These results suggest that Pten is required for the maintenance of neuroblast number, neural precursors, and differentiation in the inner ear.


Assuntos
Orelha Interna/citologia , Orelha Interna/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Ciliadas Auditivas/metabolismo , Humanos , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA