Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 134(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33674448

RESUMEN

Hearing loss affects ∼10% of adults worldwide. Most sensorineural hearing loss is caused by the progressive loss of mechanosensitive hair cells (HCs) in the cochlea. The molecular mechanisms underlying HC maintenance and loss remain poorly understood. LBH, a transcription co-factor implicated in development, is abundantly expressed in outer hair cells (OHCs). We used Lbh-null mice to identify its role in HCs. Surprisingly, Lbh deletion did not affect differentiation and the early development of HCs, as nascent HCs in Lbh knockout mice had normal looking stereocilia. The stereocilia bundle was mechanosensitive and OHCs exhibited the characteristic electromotility. However, Lbh-null mice displayed progressive hearing loss, with stereocilia bundle degeneration and OHC loss as early as postnatal day 12. RNA-seq analysis showed significant gene enrichment of biological processes related to transcriptional regulation, cell cycle, DNA damage/repair and autophagy in Lbh-null OHCs. In addition, Wnt and Notch pathway-related genes were found to be dysregulated in Lbh-deficient OHCs. Our study implicates, for the first time, loss of LBH function in progressive hearing loss, and demonstrates a critical requirement of LBH in promoting HC survival in adult mice.


Asunto(s)
Pérdida Auditiva , Factores de Transcripción , Animales , Cóclea , Células Ciliadas Auditivas Externas , Ratones , Estereocilios
2.
Proc Natl Acad Sci U S A ; 113(21): 5993-8, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27162350

RESUMEN

Hair cells of the inner ear, the mechanosensory receptors, convert sound waves into neural signals that are passed to the brain via the auditory nerve. Little is known about the molecular mechanisms that govern the development of hair cell-neuronal connections. We ascertained a family with autosomal recessive deafness associated with a common cavity inner ear malformation and auditory neuropathy. Via whole-exome sequencing, we identified a variant (c.2207G>C, p.R736T) in ROR1 (receptor tyrosine kinase-like orphan receptor 1), cosegregating with deafness in the family and absent in ethnicity-matched controls. ROR1 is a tyrosine kinase-like receptor localized at the plasma membrane. At the cellular level, the mutation prevents the protein from reaching the cellular membrane. In the presence of WNT5A, a known ROR1 ligand, the mutated ROR1 fails to activate NF-κB. Ror1 is expressed in the inner ear during development at embryonic and postnatal stages. We demonstrate that Ror1 mutant mice are severely deaf, with preserved otoacoustic emissions. Anatomically, mutant mice display malformed cochleae. Axons of spiral ganglion neurons show fasciculation defects. Type I neurons show impaired synapses with inner hair cells, and type II neurons display aberrant projections through the cochlear sensory epithelium. We conclude that Ror1 is crucial for spiral ganglion neurons to innervate auditory hair cells. Impairment of ROR1 function largely affects development of the inner ear and hearing in humans and mice.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Mutación , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Ganglio Espiral de la Cóclea/metabolismo , Animales , Axones/metabolismo , Axones/patología , Línea Celular , Células Ciliadas Auditivas/patología , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Humanos , Ratones , Ratones Mutantes , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Ganglio Espiral de la Cóclea/patología , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
3.
Hum Genet ; 137(6-7): 437-446, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29860631

RESUMEN

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.


Asunto(s)
Cóclea/crecimiento & desarrollo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/genética , Sordera/genética , Proteínas de Membrana de los Lisosomas/genética , Animales , Pueblo Asiatico/genética , Cóclea/metabolismo , Cóclea/fisiopatología , AMP Cíclico/genética , Sordera/fisiopatología , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genes Dominantes , Genotipo , Homeostasis/genética , Humanos , Lisosomas/genética , Masculino , Ratones , Mutación , Linaje , Secuenciación del Exoma
4.
J Cell Physiol ; 232(4): 743-758, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27704564

RESUMEN

Ear is a complex system where appropriate ionic composition is essential for maintaining the tissue homeostasis and hearing function. Ion transporters and channels present in the auditory system plays a crucial role in maintaining proper ionic composition in the ear. The extracellular fluid, called endolymph, found in the cochlea of the mammalian inner ear is particularly unique due to its electrochemical properties. At an endocochlear potential of about +80 mV, signaling initiated by acoustic stimuli at the level of the hair cells is dependent on the unusually high potassium (K+ ) concentration of endolymph. There are ion channels and transporters that exists in the ear to ensure that K+ is continually being cycled into the stria media endolymph. This review is focused on the discussion of the molecular and genetic basis of previously and newly recognized ion channels and transporters that support sensory hair cell excitation based on recent knock-in and knock-out studies of these channels. This article also addresses the molecular and genetic defects and the pathophysiology behind Meniere's disease as well as how the dysregulation of these ion transporters can result in severe defects in hearing or even deafness. Understanding the role of ion channels and transporters in the auditory system will facilitate in designing effective treatment modalities against ear disorders including Meniere's disease and hearing loss. J. Cell. Physiol. 232: 743-758, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Vías Auditivas/metabolismo , Canales Iónicos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Humanos , Modelos Biológicos , Modelos Moleculares , Mutación/genética
5.
J Cell Physiol ; 232(10): 2710-2721, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27869308

RESUMEN

Ear is a sensitive organ involved in hearing and balance function. The complex signaling network in the auditory system plays a crucial role in maintaining normal physiological function of the ear. The inner ear comprises a variety of host signaling pathways working in synergy to deliver clear sensory messages. Any disruption, as minor as it can be, has the potential to affect this finely tuned system with temporary or permanent sequelae including vestibular deficits and hearing loss. Mutations linked to auditory symptoms, whether inherited or acquired, are being actively researched for ways to reverse, silence, or suppress them. In this article, we discuss recent advancements in understanding the pathways involved in auditory system signaling, from hair cell development through transmission to cortical centers. Our review discusses Notch and Wnt signaling, cell to cell communication through connexin and pannexin channels, and the detrimental effects of reactive oxygen species on the auditory system. There has been an increased interest in the auditory community to explore the signaling system in the ear for hair cell regeneration. Understanding signaling pathways in the auditory system will pave the way for the novel avenues to regenerate sensory hair cells and restore hearing function. J. Cell. Physiol. 232: 2710-2721, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Ciliadas Auditivas/metabolismo , Audición , Receptores Notch/metabolismo , Regeneración , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Vías Auditivas/metabolismo , Vías Auditivas/patología , Conexinas/metabolismo , Células Ciliadas Auditivas/patología , Humanos , Células Laberínticas de Soporte/metabolismo , Células Laberínticas de Soporte/patología , NADPH Oxidasas/metabolismo , Fenotipo
6.
J Cell Physiol ; 232(9): 2359-2372, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27512962

RESUMEN

Neurotransmitters, including catecholamines and serotonin, play a crucial role in maintaining homeostasis in the human body. Studies on these neurotransmitters mainly revolved around their role in the "fight or flight" response, transmitting signals across a chemical synapse and modulating blood flow throughout the body. However, recent research has demonstrated that neurotransmitters can play a significant role in the gastrointestinal (GI) physiology. Norepinephrine (NE), epinephrine (E), dopamine (DA), and serotonin have recently been a topic of interest because of their roles in the gut physiology and their potential roles in GI and central nervous system pathophysiology. These neurotransmitters are able to regulate and control not only blood flow, but also affect gut motility, nutrient absorption, GI innate immune system, and the microbiome. Furthermore, in pathological states, such as inflammatory bowel disease (IBD) and Parkinson's disease, the levels of these neurotransmitters are dysregulated, therefore causing a variety of GI symptoms. Research in this field has shown that exogenous manipulation of catecholamine serum concentrations can help in decreasing symptomology and/or disease progression. In this review article, we discuss the current state-of-the-art research and literature regarding the role of neurotransmitters in regulation of normal GI physiology, their impact on several disease processes, and novel work focused on the use of exogenous hormones and/or psychotropic medications to improve disease symptomology. J. Cell. Physiol. 232: 2359-2372, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Bacterias/metabolismo , Encéfalo/metabolismo , Catecolaminas/metabolismo , Sistema Nervioso Entérico/metabolismo , Microbioma Gastrointestinal , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/microbiología , Serotonina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/fisiopatología , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/microbiología , Enfermedades del Sistema Nervioso Central/fisiopatología , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/fisiopatología , Interacciones Huésped-Patógeno , Humanos , Ácido gamma-Aminobutírico/metabolismo
7.
Hum Mol Genet ; 24(9): 2482-91, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25601850

RESUMEN

Hearing loss is the most common sensory deficit in humans. We show that a point mutation in DCDC2 (DCDC2a), a member of doublecortin domain-containing protein superfamily, causes non-syndromic recessive deafness DFNB66 in a Tunisian family. Using immunofluorescence on rat inner ear neuroepithelia, DCDC2a was found to localize to the kinocilia of sensory hair cells and the primary cilia of nonsensory supporting cells. DCDC2a fluorescence is distributed along the length of the kinocilium with increased density toward the tip. DCDC2a-GFP overexpression in non-polarized COS7 cells induces the formation of long microtubule-based cytosolic cables suggesting a role in microtubule formation and stabilization. Deafness mutant DCDC2a expression in hair cells and supporting cells causes cilium structural defects, such as cilium branching, and up to a 3-fold increase in length ratios. In zebrafish, the ortholog dcdc2b was found to be essential for hair cell development, survival and function. Our results reveal DCDC2a to be a deafness gene and a player in hair cell kinocilia and supporting cell primary cilia length regulation likely via its role in microtubule formation and stabilization.


Asunto(s)
Cilios/metabolismo , Genes Recesivos , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Sensorineural/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación Missense , Secuencia de Aminoácidos , Animales , Línea Celular , Mapeo Cromosómico , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Proteína Doblecortina , Femenino , Expresión Génica , Genes Reporteros , Homocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Alineación de Secuencia , Pez Cebra
8.
Proc Natl Acad Sci U S A ; 111(27): 9864-8, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958875

RESUMEN

In a large consanguineous Turkish kindred with recessive nonsyndromic, prelingual, profound hearing loss, we identified in the gene FAM65B (MIM611410) a splice site mutation (c.102-1G>A) that perfectly cosegregates with the phenotype in the family. The mutation leads to exon skipping and deletion of 52-amino acid residues of a PX membrane localization domain. FAM65B is known to be involved in myotube formation and in regulation of cell adhesion, polarization, and migration. We show that wild-type Fam65b is expressed during embryonic and postnatal development stages in murine cochlea, and that the protein localizes to the plasma membranes of the stereocilia of inner and outer hair cells of the inner ear. The wild-type protein targets the plasma membrane, whereas the mutant protein accumulates in cytoplasmic inclusion bodies and does not reach the membrane. In zebrafish, knockdown of fam65b leads to significant reduction of numbers of saccular hair cells and neuromasts and to hearing loss. We conclude that FAM65B is a plasma membrane-associated protein of hair cell stereocilia that is essential for hearing.


Asunto(s)
Audición/fisiología , Proteínas/fisiología , Estereocilios/fisiología , Animales , Moléculas de Adhesión Celular , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Audición/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Ratones , Linaje , Proteínas/genética , Proteínas/metabolismo , Empalme del ARN , Fracciones Subcelulares/metabolismo , Turquía , Pez Cebra
9.
Hum Mutat ; 37(5): 481-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26841241

RESUMEN

Hereditary hearing loss (HL) is characterized by both allelic and locus genetic heterogeneity. Both recessive and dominant forms of HL may be caused by different mutations in the same deafness gene. In a family with post-lingual progressive non-syndromic deafness, whole-exome sequencing of genomic DNA from five hearing-impaired relatives revealed a single variant, p.Gly488Glu (rs145970949:G>A) in MYO3A, co-segregating with HL as an autosomal dominant trait. This amino acid change, predicted to be pathogenic, alters a highly conserved residue in the motor domain of MYO3A. The mutation severely alters the ATPase activity and motility of the protein in vitro, and the mutant protein fails to accumulate in the filopodia tips in COS7 cells. However, the mutant MYO3A was able to reach the tips of organotypic inner ear culture hair cell stereocilia, raising the possibility of a local effect on positioning of the mechanoelectrical transduction (MET) complex at the stereocilia tips. To address this hypothesis, we investigated the interaction of MYO3A with the cytosolic tail of the integral tip-link protein protocadherin 15 (PCDH15), a core component of MET complex. Interestingly, we uncovered a novel interaction between MYO3A and PCDH15 shedding new light on the function of myosin IIIA at stereocilia tips.


Asunto(s)
Cadherinas/metabolismo , Sordera/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/genética , Miosina Tipo III/metabolismo , Polimorfismo de Nucleótido Simple , Sustitución de Aminoácidos , Animales , Células COS , Proteínas Relacionadas con las Cadherinas , Células Cultivadas , Niño , Preescolar , Chlorocebus aethiops , Sordera/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Linaje
10.
J Struct Biol ; 194(2): 139-46, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26806019

RESUMEN

All inner ear organs possess extracellular matrix appendices over the sensory epithelia that are crucial for their proper function. The tectorial membrane (TM) is a gelatinous acellular membrane located above the hearing sensory epithelium and is composed mostly of type II collagen, and α and ß tectorins. TM molecules self-assemble in the endolymph fluid environment, interacting medially with the spiral limbus and distally with the outer hair cell stereocilia. Here, we used immunogold labeling in freeze-substituted mouse cochleae to assess the fine localization of both tectorins in distinct TM regions. We observed that the TM adheres to the spiral limbus through a dense thin matrix enriched in α- and ß-tectorin, both likely bound to the membranes of interdental cells. Freeze-etching images revealed that type II collagen fibrils were crosslinked by short thin filaments (4±1.5nm, width), resembling another collagen type protein, or chains of globular elements (15±3.2nm, diameter). Gold-particles for both tectorins also localized adjacent to the type II collagen fibrils, suggesting that these globules might be composed essentially of α- and ß-tectorins. Finally, the presence of gold-particles at the TM lower side suggests that the outer hair cell stereocilia membrane has a molecular partner to tectorins, probably stereocilin, allowing the physical connection between the TM and the organ of Corti.


Asunto(s)
Colágeno Tipo II/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Membrana/metabolismo , Órgano Espiral/metabolismo , Membrana Tectoria/metabolismo , Animales , Colágeno Tipo II/genética , Colágeno Tipo II/ultraestructura , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/ultraestructura , Grabado por Congelación , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/ultraestructura , Expresión Génica , Cobayas , Inmunohistoquímica , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Ratones , Microscopía Electrónica de Transmisión , Miosinas/deficiencia , Miosinas/genética , Órgano Espiral/ultraestructura , Unión Proteica , Ratas , Membrana Tectoria/ultraestructura
11.
J Cell Physiol ; 231(8): 1656-70, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26627116

RESUMEN

The P2X purinergic receptors are cation-selective channels gated by extracellular adenosine 5'-triphosphate (ATP). These purinergic receptors are found in virtually all mammalian cell types and facilitate a number of important physiological processes. Within the past few years, the characterization of crystal structures of the zebrafish P2X4 receptor in its closed and open states has provided critical insights into the mechanisms of ligand binding and channel activation. Understanding of this gating mechanism has facilitated to design and interpret new modeling and structure-function experiments to better elucidate how different agonists and antagonists can affect the receptor with differing levels of potency. This review summarizes the current knowledge on the structure, activation, allosteric modulators, function, and location of the different P2X receptors. Moreover, an emphasis on the P2X2 receptors has been placed in respect to its role in the auditory system. In particular, the discovery of three missense mutations in P2X2 receptors could become important areas of study in the field of gene therapy to treat progressive and noise-induced hearing loss. J. Cell. Physiol. 231: 1656-1670, 2016. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Adenosina Trifosfato/metabolismo , Vías Auditivas/metabolismo , Pérdida Auditiva Provocada por Ruido/metabolismo , Audición , Activación del Canal Iónico , Receptores Purinérgicos P2X/metabolismo , Transducción de Señal , Animales , Vías Auditivas/efectos de los fármacos , Vías Auditivas/fisiopatología , Predisposición Genética a la Enfermedad , Audición/efectos de los fármacos , Pérdida Auditiva Provocada por Ruido/genética , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Provocada por Ruido/terapia , Humanos , Activación del Canal Iónico/efectos de los fármacos , Ligandos , Modelos Moleculares , Mutación Missense , Fenotipo , Conformación Proteica , Agonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X/química , Receptores Purinérgicos P2X/efectos de los fármacos , Receptores Purinérgicos P2X/genética , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
12.
J Cell Physiol ; 231(12): 2599-621, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27187048

RESUMEN

Matrix metalloproteinases (MMPs) are a diverse group of proteolytic enzymes and play an important role in the degradation and remodeling of the extracellular matrix (ECM). In normal physiological conditions, MMPs are usually minimally expressed. Despite their low expression, MMPs have been implicated in many cellular processes ranging from embryological development to apoptosis. The activity of MMPs is controlled at three different stages: (1) transcription; (2) zymogen activation; and (3) inhibition of active forms by tissue inhibitor metalloproteinases (TIMPs). They can collectively degrade any component of ECM and basement membrane, and their excessive activity has been linked to numerous pathologies mainly including, but not limited to, tumor invasion and metastasis. The lack of information about several MMPs and the steady stream of new discoveries suggest that there is much more to be studied in this field. In particular, there is a need for controlling their expression in disease states. Various studies over the past 30 years have found that each MMP has a specific mode of activation, action, and inhibition. Drugs specifically targeting individual MMPs could revolutionize the treatment of a great number of health conditions and tremendously reduce their burden. In this review article, we have summarized the recent advances in understanding the role of MMPs in physiological and pathological conditions. J. Cell. Physiol. 231: 2599-2621, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enfermedad , Metaloproteinasas de la Matriz/metabolismo , Animales , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Modelos Moleculares
13.
Hum Genet ; 135(9): 971-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27384229

RESUMEN

Human genetics research employs the two opposing approaches of forward and reverse genetics. While forward genetics identifies and links a mutation to an observed disease etiology, reverse genetics induces mutations in model organisms to study their role in disease. In most cases, causality for mutations identified by forward genetics is confirmed by reverse genetics through the development of genetically engineered animal models and an assessment of whether the model can recapitulate the disease. While many technological advances have helped improve these approaches, some gaps still remain. CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated), which has emerged as a revolutionary genetic engineering tool, holds great promise for closing such gaps. By combining the benefits of forward and reverse genetics, it has dramatically expedited human genetics research. We provide a perspective on the power of CRISPR-based forward and reverse genetics tools in human genetics and discuss its applications using some disease examples.


Asunto(s)
Investigación Biomédica , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Genética Médica , Genética Inversa , Humanos
14.
Hum Genet ; 135(5): 513-524, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27023905

RESUMEN

The high prevalence/incidence of hearing loss (HL) in humans makes it the most common sensory defect. The majority of the cases are of genetic origin. Non-syndromic hereditary HL is extremely heterogeneous. Genetic approaches have been instrumental in deciphering genes that are crucial for auditory function. In this study, we first used NADf chip to exclude the implication of known North-African mutations in HL in a large consanguineous Tunisian family (FT13) affected by autosomal recessive non-syndromic HL (ARNSHL). We then performed genome-wide linkage analysis and assigned the deafness gene locus to ch:5q23.2-31.1, corresponding to the DFNB60 ARNSHL locus. Moreover, we performed whole exome sequencing on FT13 patient DNA and uncovered amino acid substitution p.Cys113Tyr in SLC22A4, a transporter of organic cations, cosegregating with HL in FT13 and therefore the cause of ARNSHL DFNB60. We also screened a cohort of small Tunisian HL families and uncovered an additional deaf proband of consanguineous parents that is homozygous for p.Cys113Tyr carried by the same microsatellite marker haplotype as in FT13, indicating that this mutation is ancestral. Using immunofluorescence, we found that Slc22a4 is expressed in stria vascularis (SV) endothelial cells of rodent cochlea and targets their apical plasma membrane. We also found Slc22a4 transcripts in our RNA-seq library from purified primary culture of mouse SV endothelial cells. Interestingly, p.Cys113Tyr mutation affects the trafficking of the transporter and severely alters ergothioneine uptake. We conclude that SLC22A4 is an organic cation transporter of the SV endothelium that is essential for hearing, and its mutation causes DFNB60 form of HL.


Asunto(s)
Cóclea/patología , Consanguinidad , Endotelio/patología , Genes Recesivos/genética , Pérdida Auditiva/genética , Mutación/genética , Proteínas de Transporte de Catión Orgánico/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cóclea/metabolismo , Endotelio/metabolismo , Exoma/genética , Femenino , Células HEK293 , Pérdida Auditiva/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Linaje , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Ratas , Ratas Sprague-Dawley , Homología de Secuencia de Aminoácido , Simportadores
15.
Proc Natl Acad Sci U S A ; 110(6): 2228-33, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23345450

RESUMEN

Age-related hearing loss and noise-induced hearing loss are major causes of human morbidity. Here we used genetics and functional studies to show that a shared cause of these disorders may be loss of function of the ATP-gated P2X(2) receptor (ligand-gated ion channel, purinergic receptor 2) that is expressed in sensory and supporting cells of the cochlea. Genomic analysis of dominantly inherited, progressive sensorineural hearing loss DFNA41 in a six-generation kindred revealed a rare heterozygous allele, P2RX2 c.178G > T (p.V60L), at chr12:133,196,029, which cosegregated with fully penetrant hearing loss in the index family, and also appeared in a second family with the same phenotype. The mutation was absent from more than 7,000 controls. P2RX2 p.V60L abolishes two hallmark features of P2X(2) receptors: ATP-evoked inward current response and ATP-stimulated macropore permeability, measured as loss of ATP-activated FM1-43 fluorescence labeling. Coexpression of mutant and WT P2X(2) receptor subunits significantly reduced ATP-activated membrane permeability. P2RX2-null mice developed severe progressive hearing loss, and their early exposure to continuous moderate noise led to high-frequency hearing loss as young adults. Similarly, among family members heterozygous for P2RX2 p.V60L, noise exposure exacerbated high-frequency hearing loss in young adulthood. Our results suggest that P2X(2) function is required for life-long normal hearing and for protection from exposure to noise.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/genética , Pérdida Auditiva Sensorineural/genética , Mutación Missense , Receptores Purinérgicos P2X2/genética , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos , Femenino , Genes Dominantes , Pérdida Auditiva Provocada por Ruido/etiología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/fisiopatología , Heterocigoto , Humanos , Activación del Canal Iónico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Linaje , Penetrancia , Receptores Purinérgicos P2X2/deficiencia , Receptores Purinérgicos P2X2/fisiología , Homología de Secuencia de Aminoácido , Adulto Joven
16.
Mol Genet Genomics ; 290(4): 1327-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25633957

RESUMEN

Hearing loss (HL) is a major public health issue. It is clinically and genetically heterogeneous.The identification of the causal mutation is important for early diagnosis, clinical follow-up, and genetic counseling. HL due to mutations in COL11A2, encoding collagen type XI alpha-2, can be non-syndromic autosomal-dominant or autosomal-recessive, and also syndromic as in Otospondylomegaepiphyseal Dysplasia, Stickler syndrome type III, and Weissenbacher-Zweymuller syndrome. However, thus far only one mutation co-segregating with autosomal recessive non-syndromic hearing loss (ARNSHL) in a single family has been reported. In this study, whole exome sequencing of two consanguineous families with ARNSHL from Tunisia and Turkey revealed two novel causative COL11A2 mutations, c.109G > T (p.Ala37Ser) and c.2662C > A (p.Pro888Thr). The variants identified co-segregated with deafness in both families. All homozygous individuals in those families had early onset profound hearing loss across all frequencies without syndromic findings. The variants are predicted to be damaging the protein function. The p.Pro888Thr mutation affects a -Gly-X-Y- triplet repeat motif. The novel p.Ala37Ser is the first missense mutation located in the NC4 domain of the COL11A2 protein. Structural model suggests that this mutation will likely obliterate, or at least partially compromise, the ability of NC4 domain to interact with its cognate ligands. In conclusion, we confirm that COL11A2 mutations cause ARNSHL and broaden the mutation spectrum that may shed new light on genotype-phenotype correlation for the associated phenotypes and clinical follow-up.


Asunto(s)
Colágeno Tipo XI/genética , Genes Recesivos , Predisposición Genética a la Enfermedad/genética , Pérdida Auditiva Sensorineural/genética , Mutación Missense , Secuencia de Aminoácidos , Secuencia de Bases , Colágeno Tipo XI/química , Consanguinidad , Exoma/genética , Salud de la Familia , Femenino , Frecuencia de los Genes , Genotipo , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN/métodos , Homología de Secuencia de Aminoácido
17.
Proc Natl Acad Sci U S A ; 108(28): 11476-81, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21709241

RESUMEN

In the most accepted model for hair cell mechanotransduction, a cluster of myosin motors located at the stereocilia upper tip-link density (UTLD) keeps the tip-link under tension at rest. Both myosin VIIa (MYO7A) and myosin 1c have been implicated in mechanotransduction based on functional studies. However, localization studies are conflicting, leaving open the question of which myosin localizes at the UTLD and generates the tip-link resting tension. Using immunofluorescence, we now show that MYO7A and sans, a MYO7A-interacting protein, cluster at the UTLD. Analysis of the immunofluorescence intensity indicates that eight or more MYO7A molecules are present at each UTLD, consistent with a direct role for MYO7A in maintaining tip-link tension. MYO7A and sans localization at the UTLD is confirmed by transfection of hair cells with GFP-tagged constructs for these proteins. Cotransfection studies in a heterologous system show that MYO7A, sans, and the UTLD protein harmonin-b form a tripartite complex and that each protein is capable of interacting with one another independently. We propose that MYO7A, sans, and harmonin-b form the core components of the UTLD molecular complex. In this complex, MYO7A is likely the motor element that pulls on CDH23 to exert tension on the tip-link.


Asunto(s)
Cilios/metabolismo , Mecanotransducción Celular/fisiología , Miosinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Chlorocebus aethiops , Cilios/ultraestructura , Proteínas del Citoesqueleto , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Cobayas , Células Ciliadas Vestibulares/metabolismo , Células Ciliadas Vestibulares/ultraestructura , Humanos , Ratones , Ratones Mutantes , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Datos de Secuencia Molecular , Complejos Multiproteicos , Miosina VIIa , Miosinas/química , Miosinas/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Órgano Espiral/metabolismo , Órgano Espiral/ultraestructura , Dominios y Motivos de Interacción de Proteínas , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Síndromes de Usher/fisiopatología
18.
J Neurosci ; 32(41): 14288-93, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23055499

RESUMEN

Usher syndrome is the leading cause of genetic deaf-blindness. Monoallelic mutations in PDZD7 increase the severity of Usher type II syndrome caused by mutations in USH2A and GPR98, which respectively encode usherin and GPR98. PDZ domain-containing 7 protein (PDZD7) is a paralog of the scaffolding proteins harmonin and whirlin, which are implicated in Usher type 1 and type 2 syndromes. While usherin and GPR98 have been reported to form hair cell stereocilia ankle-links, harmonin localizes to the stereocilia upper tip-link density and whirlin localizes to both tip and ankle-link regions. Here, we used mass spectrometry to show that PDZD7 is expressed in chick stereocilia at a comparable molecular abundance to GPR98. We also show by immunofluorescence and by overexpression of tagged proteins in rat and mouse hair cells that PDZD7 localizes to the ankle-link region, overlapping with usherin, whirlin, and GPR98. Finally, we show in LLC-PK1 cells that cytosolic domains of usherin and GPR98 can bind to both whirlin and PDZD7. These observations are consistent with PDZD7 being a modifier and candidate gene for USH2, and suggest that PDZD7 is a second scaffolding component of the ankle-link complex.


Asunto(s)
Proteínas Portadoras/química , Redes Reguladoras de Genes/fisiología , Dominios PDZ/fisiología , Estereocilios/química , Síndromes de Usher , Secuencia de Aminoácidos , Animales , Células COS , Proteínas Portadoras/genética , Embrión de Pollo , Chlorocebus aethiops , Femenino , Humanos , Células LLC-PK1 , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Ratas , Estereocilios/genética , Estereocilios/metabolismo , Porcinos , Síndromes de Usher/genética , Síndromes de Usher/metabolismo
19.
BMC Neurosci ; 14: 96, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-24011083

RESUMEN

BACKGROUND: Myelinated axons are organized into distinct subcellular and molecular regions. Without proper organization, electrical nerve conduction is delayed, resulting in detrimental physiological outcomes. One such region is the paranode where axo-glial septate junctions act as a molecular fence to separate the sodium (Na+) channel-enriched node from the potassium (K+) channel-enriched juxtaparanode. A significant lack of knowledge remains as to cytoskeletal proteins which stabilize paranodal domains and underlying cytoskeleton. Whirlin (Whrn) is a PDZ domain-containing cytoskeletal scaffold whose absence in humans results in Usher Syndromes or variable deafness-blindness syndromes. Mutant Whirlin (Whrn) mouse model studies have linked such behavioral deficits to improper localization of critical transmembrane protein complexes in the ear and eye. Until now, no reports exist about the function of Whrn in myelinated axons. RESULTS: RT-PCR and immunoblot analyses revealed expression of Whrn mRNA and Whrn full-length protein, respectively, in several stages of central and peripheral nervous system development. Comparing wild-type mice to Whrn knockout (Whrn-/-) mice, we observed no significant differences in the expression of standard axonal domain markers by immunoblot analysis but observed and quantified a novel paranodal compaction phenotype in 4 to 8 week-old Whrn-/- nerves. The paranodal compaction phenotype and associated cytoskeletal disruption was observed in Whrn-/- mutant sciatic nerves and spinal cord fibers from early (2 week-old) to late (1 year-old) stages of development. Light and electron microscopic analyses of Whrn knockout mice reveal bead-like swellings in cerebellar Purkinje axons containing mitochondria and vesicles by both. These data suggest that Whrn plays a role in proper cytoskeletal organization in myelinated axons. CONCLUSIONS: Domain organization in myelinated axons remains a complex developmental process. Here we demonstrate that loss of Whrn disrupts proper axonal domain organization. Whrn likely contributes to the stabilization of paranodal myelin loops and axonal cytoskeleton through yet unconfirmed cytoskeletal proteins. Paranodal abnormalities are consistently observed throughout development (2 wk-1 yr) and similar between central and peripheral nervous systems. In conclusion, our observations suggest that Whrn is not required for the organization of axonal domains, but once organized, Whrn acts as a cytoskeletal linker to ensure proper paranodal compaction and stabilization of the axonal cytoskeleton in myelinated axons.


Asunto(s)
Citoesqueleto/ultraestructura , Proteínas de la Membrana/metabolismo , Fibras Nerviosas Mielínicas/ultraestructura , Animales , Axones/metabolismo , Axones/ultraestructura , Citoesqueleto/metabolismo , Electrofisiología , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Fibras Nerviosas Mielínicas/metabolismo , Nódulos de Ranvier/metabolismo , Nódulos de Ranvier/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Biol Chem ; 285(46): 35770-82, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20826793

RESUMEN

Myosin IIIa (Myo3A) transports cargo to the distal end of actin protrusions and contains a kinase domain that is thought to autoregulate its activity. Because Myo3A tends to cluster at the tips of actin protrusions, we investigated whether intermolecular phosphorylation could regulate Myo3A biochemical activity, cellular localization, and cellular function. Inactivation of Myo3A 2IQ kinase domain with the point mutation K50R did not alter maximal ATPase activity, whereas phosphorylation of Myo3A 2IQ resulted in reduced maximal ATPase activity and actin affinity. The rate and degree of Myo3A 2IQ autophosphorylation was unchanged by the presence of actin but was found to be dependent upon Myo3A 2IQ concentration within the range of 0.1 to 1.2 µm, indicating intermolecular autophosphorylation. In cultured cells, we observed that the filopodial tip localization of Myo3A lacking the kinase domain decreased when co-expressed with kinase-active, full-length Myo3A. The cellular consequence of reduced Myo3A tip localization was decreased filopodial density along the cell periphery, identifying a novel cellular function for Myo3A in mediating the formation and stability of actin-based protrusions. Our results suggest that Myo3A motor activity is regulated through a mechanism involving concentration-dependent autophosphorylation. We suggest that this regulatory mechanism plays an essential role in mediating the transport and actin bundle formation/stability functions of Myo3A.


Asunto(s)
Actinas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Ciliadas Vestibulares/metabolismo , Humanos , Microscopía Fluorescente , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Órgano Espiral/metabolismo , Fosforilación , Unión Proteica , Seudópodos/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA