Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Cell ; 151(6): 1283-95, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23217710

RESUMEN

Hair cells are mechanosensors for the perception of sound, acceleration, and fluid motion. Mechanotransduction channels in hair cells are gated by tip links, which connect the stereocilia of a hair cell in the direction of their mechanical sensitivity. The molecular constituents of the mechanotransduction channels of hair cells are not known. Here, we show that mechanotransduction is impaired in mice lacking the tetraspan TMHS. TMHS binds to the tip-link component PCDH15 and regulates tip-link assembly, a process that is disrupted by deafness-causing Tmhs mutations. TMHS also regulates transducer channel conductance and is required for fast channel adaptation. TMHS therefore resembles other ion channel regulatory subunits such as the transmembrane alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor regulatory proteins (TARPs) of AMPA receptors that facilitate channel transport and regulate the properties of pore-forming channel subunits. We conclude that TMHS is an integral component of the hair cell's mechanotransduction machinery that functionally couples PCDH15 to the transduction channel.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Audición , Mecanotransducción Celular , Proteínas de la Membrana/metabolismo , Animales , Proteínas Relacionadas con las Cadherinas , Cadherinas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Ratones , Ratones Noqueados , Precursores de Proteínas/metabolismo , Estereocilios/metabolismo
2.
Dev Biol ; 457(1): 91-103, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31550482

RESUMEN

Little is known about the role of TBX1 in post-otocyst stages of inner ear development. Here, we report on mice with a missense mutation of Tbx1 that are viable with fully developed but abnormally formed inner ears. Mutant mice are deaf due to an undeveloped stria vascularis and show vestibular dysfunction associated with abnormal semicircular canal formation. We show that TBX1 is expressed in endolymph-producing strial marginal cells and vestibular dark cells of the inner ear and is an upstream regulator of Esrrb, which previously was shown to control the developmental fate of these cells. We also show that TBX1 is expressed in sensory cells of the crista ampullaris, which may relate to the semicircular canal abnormalities observed in mutant mice. Inner ears of mutant embryos have a non-resorbed fusion plate in the posterior semicircular canal and a single ampulla connecting anterior and lateral canals. We hypothesize that the TBX1 missense mutation prevents binding with specific co-regulatory proteins. These findings reveal previously unknown functions of TBX1 during later stages of inner ear development.


Asunto(s)
Oído Interno/embriología , Mutación Missense , Canales Semicirculares/embriología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Oído Interno/citología , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Morfogénesis , Receptores de Estrógenos/metabolismo , Canales Semicirculares/anomalías , Estría Vascular/citología , Proteínas de Dominio T Box/química , Técnicas del Sistema de Dos Híbridos , Secuenciación del Exoma
3.
J Neurosci ; 38(4): 843-857, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29222402

RESUMEN

Sensory hair cells require control of physical properties of their apical plasma membranes for normal development and function. Members of the ADP-ribosylation factor (ARF) small GTPase family regulate membrane trafficking and cytoskeletal assembly in many cells. We identified ELMO domain-containing protein 1 (ELMOD1), a guanine nucleoside triphosphatase activating protein (GAP) for ARF6, as the most highly enriched ARF regulator in hair cells. To characterize ELMOD1 control of trafficking, we analyzed mice of both sexes from a strain lacking functional ELMOD1 [roundabout (rda)]. In rda/rda mice, cuticular plates of utricle hair cells initially formed normally, then degenerated after postnatal day 5; large numbers of vesicles invaded the compromised cuticular plate. Hair bundles initially developed normally, but the cell's apical membrane lifted away from the cuticular plate, and stereocilia elongated and fused. Membrane trafficking in type I hair cells, measured by FM1-43 dye labeling, was altered in rda/rda mice. Consistent with the proposed GAP role for ELMOD1, the ARF6 GTP/GDP ratio was significantly elevated in rda/rda utricles compared with controls, and the level of ARF6-GTP was correlated with the severity of the rda/rda phenotype. These results suggest that conversion of ARF6 to its GDP-bound form is necessary for final stabilization of the hair bundle.SIGNIFICANCE STATEMENT Assembly of the mechanically sensitive hair bundle of sensory hair cells requires growth and reorganization of apical actin and membrane structures. Hair bundles and apical membranes in mice with mutations in the Elmod1 gene degenerate after formation, suggesting that the ELMOD1 protein stabilizes these structures. We show that ELMOD1 is a GTPase-activating protein in hair cells for the small GTP-binding protein ARF6, known to participate in actin assembly and membrane trafficking. We propose that conversion of ARF6 into the GDP-bound form in the apical domain of hair cells is essential for stabilizing apical actin structures like the hair bundle and ensuring that the apical membrane forms appropriately around the stereocilia.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Células Ciliadas Vestibulares/metabolismo , Factor 6 de Ribosilación del ADP , Animales , Femenino , Guanosina Trifosfato/metabolismo , Células Ciliadas Vestibulares/ultraestructura , Hidrólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas , Estereocilios/metabolismo , Estereocilios/ultraestructura
4.
Dev Biol ; 443(2): 153-164, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30217595

RESUMEN

Hundreds of thousands of cis-regulatory DNA sequences are predicted in vertebrate genomes, but unlike genes themselves, few have been characterized at the functional level or even unambiguously paired with a target gene. Here we serendipitously identified and started investigating the first reported long-range regulatory region for the Nr2f1 (Coup-TFI) transcription factor gene. NR2F1 is temporally and spatially regulated during development and required for patterning and regionalization in the nervous system, including sensory hair cell organization in the auditory epithelium of the cochlea. Analyzing the deaf wanderer (dwnd) spontaneous mouse mutation, we traced back the cause of its associated circling behavior to a 53 kb deletion removing five exons and adjacent intronic regions of the poorly characterized Mctp1 gene. Interestingly, loss of Mctp1 function cannot account for the hearing loss, inner ear dysmorphology and sensory hair cell disorganization observed in dwnd mutants. Instead, we found that the Mctp1dwnd deletion affects the Nr2f1 gene located 1.4 Mb away, downregulating transcription and protein expression in the embryonic cochlea. Remarkably, the Mctp1dwnd allele failed to complement a targeted inactivation allele of Nr2f1, and transheterozygotes or Mctp1dwnd homozygotes exhibit the same morphological defects observed in inner ears of Nr2f1 mutants without sharing their early life lethality. Defects include improper separation of the utricle and saccule in the vestibule not described previously, which can explain the circling behavior that first brought the spontaneous mutation to attention. By contrast, mice homozygous for a targeted inactivation of Mctp1 have normal hearing and inner ear structures. We conclude that the 53 kb Mctp1dwnd deletion encompasses a long-range cis-regulatory region essential for proper Nr2f1 expression in the embryonic inner ear, providing a first opportunity to investigate Nr2f1 function in postnatal inner ears. This work adds to the short list of long-range regulatory regions characterized as essential to drive expression of key developmental control genes.


Asunto(s)
Factor de Transcripción COUP I/genética , Factor de Transcripción COUP I/metabolismo , Oído Interno/embriología , Animales , Factor de Transcripción COUP I/fisiología , Sordera/genética , Oído Interno/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
Hum Mol Genet ; 26(19): 3722-3735, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28934385

RESUMEN

Mutations of the human ATP6V1B1 gene cause distal renal tubular acidosis (dRTA; OMIM #267300) often associated with sensorineural hearing impairment; however, mice with a knockout mutation of Atp6v1b1 were reported to exhibit a compensated acidosis and normal hearing. We discovered a new spontaneous mutation (vortex, symbol vtx) of Atp6v1b1 in an MRL/MpJ (MRL) colony of mice. In contrast to the reported phenotype of the knockout mouse, which was developed on a primarily C57BL/6 (B6) strain background, MRL-Atp6v1b1vtx/vtx mutant mice exhibit profound hearing impairment, which is associated with enlarged endolymphatic compartments of the inner ear. Mutant mice have alkaline urine but do not exhibit overt metabolic acidosis, a renal phenotype similar to that of the Atpbv1b1 knockout mouse. The abnormal inner ear phenotype of MRL- Atp6v1b1vtx/vtx mice was lost when the mutation was transferred onto the C57BL/6J (B6) background, indicating the influence of strain-specific genetic modifiers. To genetically map modifier loci in Atp6v1b1vtx/vtx mice, we analysed ABR thresholds of progeny from a backcross segregating MRL and B6 alleles. We found statistically significant linkage with a locus on Chr 13 that accounts for about 20% of the hearing threshold variation in the backcross mice. The important effect that genetic background has on the inner ear phenotype of Atp6v1b1 mutant mice provides insight into the hearing loss variability associated with dRTA caused by ATP6V1B1 mutations. Because MRL-Atp6v1b1vxt/vtx mice do not recapitulate the metabolic acidosis of dRTA patients, they provide a new genetic model for nonsyndromic deafness with enlarged vestibular aqueduct (EVA; OMIM #600791).


Asunto(s)
Sordera/genética , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Acidosis/genética , Acidosis/metabolismo , Acidosis Tubular Renal/genética , Acidosis Tubular Renal/metabolismo , Animales , Sordera/metabolismo , Modelos Animales de Enfermedad , Oído Interno/patología , Femenino , Ligamiento Genético , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Acueducto Vestibular/metabolismo , Acueducto Vestibular/fisiología
6.
Mamm Genome ; 30(1-2): 5-22, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30778664

RESUMEN

Maternal and fetal sources of thyroid hormone are important for the development of many organ systems. Thyroid hormone deficiency causes variable intellectual disability and hearing impairment in mouse and man, but the basis for this variation is not clear. To explore this variation, we studied two thyroid hormone-deficient mouse mutants with mutations in pituitary-specific transcription factors, POU1F1 and PROP1, that render them unable to produce thyroid stimulating hormone. DW/J-Pou1f1dw/dw mice have profound deafness and both neurosensory and conductive hearing impairment, while DF/B-Prop1df/df mice have modest elevations in hearing thresholds consistent with developmental delay, eventually achieving normal hearing ability. The thyroid glands of Pou1f1 mutants are more severely affected than those of Prop1df/df mice, and they produce less thyroglobulin during the neonatal period critical for establishing hearing. We previously crossed DW/J-Pou1f1dw/+ and Cast/Ei mice and mapped a major locus on Chromosome 2 that protects against hypothyroidism-induced hearing impairment in Pou1f1dw/dw mice: modifier of dw hearing (Mdwh). Here we refine the location of Mdwh by genotyping 196 animals with 876 informative SNPs, and we conduct novel mapping with a DW/J-Pou1f1dw/+ and 129/P2 cross that reveals 129/P2 mice also have a protective Mdwh locus. Using DNA sequencing of DW/J and DF/B strains, we determined that the genes important for thyroid gland function within Mdwh vary in amino acid sequence between strains that are susceptible or resistant to hypothyroidism-induced hearing impairment. These results suggest that the variable effects of congenital hypothyroidism on the development of hearing ability are attributable to genetic variation in postnatal thyroid gland folliculogenesis and function.


Asunto(s)
Susceptibilidad a Enfermedades , Variación Genética , Pérdida Auditiva/etiología , Hipotiroidismo/complicaciones , Glándula Tiroides/embriología , Glándula Tiroides/metabolismo , Alelos , Animales , Animales Recién Nacidos , Biomarcadores , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Modelos Animales de Enfermedad , Oído Medio/embriología , Oído Medio/metabolismo , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/tratamiento farmacológico , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Glándula Tiroides/patología , Hormonas Tiroideas/farmacología
7.
Proc Natl Acad Sci U S A ; 111(21): E2200-9, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24825892

RESUMEN

The rhomboid 5 homolog 2 (Rhbdf2) gene encodes an inactive rhomboid (iRhom) protease, iRhom2, one of a family of enzymes containing a long cytosolic N terminus and a dormant peptidase domain of unknown function. iRhom2 has been implicated in epithelial regeneration and cancer growth through constitutive activation of epidermal growth factor receptor (EGFR) signaling. However, little is known about the physiological substrates for iRhom2 or the molecular mechanisms underlying these functions. We show that iRhom2 is a short-lived protein whose stability can be increased by select mutations in the N-terminal domain. In turn, these stable variants function to augment the secretion of EGF family ligands, including amphiregulin, independent of metalloprotease a disintegrin and metalloproteinase 17 (ADAM17) activity. In vivo, N-terminal iRhom2 mutations induce accelerated wound healing as well as accelerated tumorigenesis, but they do not drive spontaneous tumor development. This work underscores the physiological prominence of iRhom2 in controlling EGFR signaling events involved in wound healing and neoplastic growth, and yields insight into the function of key iRhom2 domains.


Asunto(s)
Proteínas Portadoras/genética , Receptores ErbB/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Estabilidad Proteica , Transducción de Señal/fisiología , Anfirregulina , Animales , Células COS , Chlorocebus aethiops , Clonación Molecular , Familia de Proteínas EGF , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Estimación de Kaplan-Meier , Ratones , Mutagénesis , Mutación/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Cicatrización de Heridas/genética
8.
Mamm Genome ; 27(5-6): 200-12, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27090238

RESUMEN

Adaptor protein (AP) complexes function in the intracellular sorting and vesicular transport of membrane proteins. The clathrin-associated AP-1 complex functions at the trans-Golgi network and endosomes, and some forms of this complex are thought to mediate the sorting of proteins in plasma membranes of polarized epithelial cells. A null mutation of the mouse Ap1g1 gene, which encodes the gamma-1 subunit of the AP-1 complex, causes embryonic lethality when homozygous, indicating its critical importance in early development but precluding studies of its possible roles during later stages. Here, we describe our analyses of a new spontaneous mutation of Ap1g1 named "figure eight" (symbol fgt) and show that it is an in-frame deletion of 6 bp, which results in the elimination of two amino acids of the encoded protein. In contrast to Ap1g1 (-/-) null mice, mice homozygous for the recessive fgt mutation are viable with adult survival similar to controls. Although Ap1g1 is ubiquitously expressed, the phenotype of Ap1g1 (fgt) mutant mice is primarily restricted to abnormalities in sensory epithelial cells of the inner ear, pigmented epithelial cells of the retina, follicular epithelial cells of the thyroid gland, and the germinal epithelium of the testis, suggesting that impaired AP-1 sorting and targeting of membrane proteins in these polarized cells may underlie the observed pathologies. Ap1g1 (fgt) mutant mice provide a new animal model to study the in vivo roles of gamma-1 adaptin and the AP-1 complex throughout development and to investigate factors that underlie its associated phenotypic abnormalities.


Asunto(s)
Anomalías Múltiples/genética , Complejo 1 de Proteína Adaptadora/genética , Subunidades gamma de Complejo de Proteína Adaptadora/genética , Red trans-Golgi/genética , Anomalías Múltiples/patología , Animales , Polaridad Celular/genética , Modelos Animales de Enfermedad , Oído Interno/anomalías , Humanos , Masculino , Ratones , Mutación , Retina/anomalías , Testículo/anomalías , Glándula Tiroides/anomalías , Red trans-Golgi/metabolismo
9.
Mamm Genome ; 26(7-8): 338-47, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26092689

RESUMEN

Inbred mouse strains serve as important models for human presbycusis or age-related hearing loss. We previously mapped a locus (ahl8) contributing to the progressive hearing loss of DBA/2J (D2) mice and later showed that a missense variant of the Fscn2 gene, unique to the D2 inbred strain, was responsible for the ahl8 effect. Although ahl8 can explain much of the hearing loss difference between C57BL/6J (B6) and D2 strain mice, other loci also contribute. Here, we present results of our linkage analyses to map quantitative trait loci (QTLs) that modify the severity of hearing loss associated with the D2 strain Fscn2 (ahl8) allele. We searched for modifier loci by analyzing 31 BXD recombinant inbred (RI) lines fixed for the predisposing D2-derived Fscn2 (ahl8/ahl8) genotype and found a statistically significant linkage association of threshold means with a QTL on Chr 5, which we designated M5ahl8. The highest association (LOD 4.6) was with markers at the 84-90 Mb position of Chr 5, which could explain about 46 % of the among-RI strain variation in auditory brainstem response (ABR) threshold means. The semidominant nature of the modifying effect of M5ahl8 on the Fscn2 (ahl8/ahl8) phenotype was demonstrated by analysis of a backcross involving D2 and B6.D2-Chr11D/LusJ strain mice. The Chr 5 map position of M5ahl8 and the D2 origin of its susceptibility allele correspond to Tmc1m4, a previously reported QTL that modifies outer hair cell degeneration in Tmc1 (Bth) mutant mice, suggesting that M5ahl8 and Tmc1m4 may represent the same gene affecting maintenance of stereocilia structure and function during aging.


Asunto(s)
Envejecimiento/genética , Proteínas Portadoras/genética , Cromosomas de los Mamíferos/química , Predisposición Genética a la Enfermedad , Proteínas de Microfilamentos/genética , Presbiacusia/genética , Sitios de Carácter Cuantitativo , Envejecimiento/metabolismo , Envejecimiento/patología , Alelos , Animales , Proteínas Portadoras/metabolismo , Mapeo Cromosómico , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Expresión Génica , Ligamiento Genético , Genotipo , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteínas de Microfilamentos/metabolismo , Fenotipo , Presbiacusia/metabolismo , Presbiacusia/patología , Índice de Severidad de la Enfermedad , Especificidad de la Especie
10.
J Neurosci ; 33(19): 8114-21, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23658152

RESUMEN

Stereocilia are actin-based protrusions on auditory sensory hair cells that are deflected by sound waves to initiate the conversion of mechanical energy to neuronal signals. Stereocilia maintenance is essential because auditory hair cells are not renewed in mammals. This process requires both ß-actin and γ-actin as knock-out mice lacking either isoform develop distinct stereocilia pathology during aging. In addition, stereocilia integrity may hinge on immobilizing actin, which outside of a small region at stereocilia tips turns over with a very slow, months-long half-life. Here, we establish that ß-actin and the actin crosslinking protein fascin-2 cooperate to maintain stereocilia length and auditory function. We observed that mice expressing mutant fascin-2 (p.R109H) or mice lacking ß-actin share a common phenotype including progressive, high-frequency hearing loss together with shortening of a defined subset of stereocilia in the hair cell bundle. Fascin-2 binds ß-actin and γ-actin filaments with similar affinity in vitro and fascin-2 does not depend on ß-actin for localization in vivo. Nevertheless, double-mutant mice lacking ß-actin and expressing fascin-2 p.R109H have a more severe phenotype suggesting that each protein has a different function in a common stereocilia maintenance pathway. Because the fascin-2 p.R109H mutant binds but fails to efficiently crosslink actin filaments, we propose that fascin-2 crosslinks function to slow actin depolymerization at stereocilia tips to maintain stereocilia length.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Células Ciliadas Auditivas/citología , Proteínas de Microfilamentos/metabolismo , Estereocilios/fisiología , Estimulación Acústica , Actinas/deficiencia , Actinas/genética , Envejecimiento/genética , Animales , Benzofuranos , Cadherinas/genética , Proteínas Portadoras/genética , Electroencefalografía , Receptor alfa de Estrógeno/genética , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Femenino , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/ultraestructura , Pérdida Auditiva de Alta Frecuencia/genética , Pérdida Auditiva de Alta Frecuencia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Microscopía de Fuerza Atómica , Mutación/genética , Fenotipo , Unión Proteica/genética , Quinolinas , Estereocilios/ultraestructura
11.
Am J Hum Genet ; 86(2): 148-60, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20137774

RESUMEN

Recessive mutations at the mouse pirouette (pi) locus result in hearing loss and vestibular dysfunction due to neuroepithelial defects in the inner ear. Using a positional cloning strategy, we have identified mutations in the gene Grxcr1 (glutaredoxin cysteine-rich 1) in five independent allelic strains of pirouette mice. We also provide sequence data of GRXCR1 from humans with profound hearing loss suggesting that pirouette is a model for studying the mechanism of nonsyndromic deafness DFNB25. Grxcr1 encodes a 290 amino acid protein that contains a region of similarity to glutaredoxin proteins and a cysteine-rich region at its C terminus. Grxcr1 is expressed in sensory epithelia of the inner ear, and its encoded protein is localized along the length of stereocilia, the actin-filament-rich mechanosensory structures at the apical surface of auditory and vestibular hair cells. The precise architecture of hair cell stereocilia is essential for normal hearing. Loss of function of Grxcr1 in homozygous pirouette mice results in abnormally thin and slightly shortened stereocilia. When overexpressed in transfected cells, GRXCR1 localizes along the length of actin-filament-rich structures at the dorsal-apical surface and induces structures with greater actin filament content and/or increased lengths in a subset of cells. Our results suggest that deafness in pirouette mutants is associated with loss of GRXCR1 function in modulating actin cytoskeletal architecture in the developing stereocilia of sensory hair cells.


Asunto(s)
Oído Interno/fisiopatología , Sitios Genéticos/genética , Glutarredoxinas/genética , Mutación/genética , Citoesqueleto de Actina , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Secuencia Conservada , Análisis Mutacional de ADN , Evolución Molecular , Femenino , Regulación de la Expresión Génica , Glutarredoxinas/química , Pérdida Auditiva/genética , Pérdida Auditiva/fisiopatología , Humanos , Masculino , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Linaje , Estructura Terciaria de Proteína , Transporte de Proteínas
12.
Nat Genet ; 35(1): 21-3, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12910270

RESUMEN

Age-related hearing loss (AHL) in common inbred mouse strains is a genetically complex quantitative trait. We found a synonymous single-nucleotide polymorphism in exon 7 of Cdh23 that shows significant association with AHL and the deafness modifier mdfw (modifer of deafwaddler). The hypomorphic Cdh23(753A) allele causes in-frame skipping of exon 7. Altered adhesion or reduced stability of CDH23 may confer susceptibility to AHL. Homozygosity at Cdh23(753A) or in combination with heterogeneous secondary factors is a primary determinant of AHL in mice.


Asunto(s)
Cadherinas/genética , Pérdida Auditiva Sensorineural/genética , Envejecimiento , Alelos , Animales , Cadherinas/fisiología , Clonación de Organismos , Patrón de Herencia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos , Polimorfismo de Nucleótido Simple
13.
Nat Genet ; 30(4): 401-5, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11925566

RESUMEN

Once a mutation in the gene tub was identified as the cause of obesity, retinal degeneration and hearing loss in tubby mice, it became increasingly evident that the members of the tub gene family (tulps) influence maintenance and function of the neuronal cell lineage. Suggested molecular functions of tubby-like proteins include roles in vesicular trafficking, mediation of insulin signaling and gene transcription. The mechanisms through which tub functions in neurons, however, have yet to be elucidated. Here we report the positional cloning of an auditory quantitative trait locus (QTL), the modifier of tubby hearing 1 gene (moth1), whose wildtype alleles from strains AKR/J, CAST/Ei and 129P2/OlaHsd protect tubby mice from hearing loss. Through a transgenic rescue experiment, we verified that sequence polymorphisms in the neuron-specific microtubule-associated protein 1a gene (Mtap1a) observed in the susceptible strain C57BL/6J (B6) are crucial for the hearing-loss phenotype. We also show that these polymorphisms change the binding efficiency of MTAP1A to postsynaptic density molecule 95 (PSD95), a core component in the cytoarchitecture of synapses. This indicates that at least some of the observed polymorphisms are functionally important and that the hearing loss in C57BL/6J-tub/tub (B6-tub/tub) mice may be caused by impaired protein interactions involving MTAP1A. We therefore propose that tub may be associated with synaptic function in neuronal cells.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales , Alelos , Animales , Línea Celular , Clonación Molecular , ADN Complementario/metabolismo , Homólogo 4 de la Proteína Discs Large , Biblioteca de Genes , Marcadores Genéticos , Guanilato-Quinasas , Immunoblotting , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Modelos Genéticos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Fenotipo , Polimorfismo Genético , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Carácter Cuantitativo Heredable , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sinapsis/metabolismo , Transcripción Genética , Transgenes
14.
Blood ; 115(6): 1267-76, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19846887

RESUMEN

The spontaneous mouse mutation "thrombocytopenia and cardiomyopathy" (trac) causes macrothrombocytopenia, prolonged bleeding times, anemia, leukopenia, infertility, cardiomyopathy, and shortened life span. Homozygotes show a 20-fold decrease in platelet numbers and a 3-fold increase in platelet size with structural alterations and functional impairments in activation and aggregation. Megakaryocytes in trac/trac mice are present in increased numbers, have poorly developed demarcation membrane systems, and have decreased polyploidy. The thrombocytopenia is not intrinsic to defects at the level of hematopoietic progenitor cells but is associated with a microenvironmental abnormality. The trac mutation maps to mouse chromosome 17, syntenic with human chromosome 2p21-22. A G to A mutation in exon 10 of the adenosine triphosphate (ATP)-binding cassette subfamily G, member 5 (Abcg5) gene, alters a tryptophan codon (UGG) to a premature stop codon (UAG). Crosses with mice doubly transgenic for the human ABCG5 and ABCG8 genes rescued platelet counts and volumes. ABCG5 and ABCG8 form a functional complex that limits dietary phytosterol accumulation. Phytosterolemia in trac/trac mice confirmed a functional defect in the ABCG5/ABCG8 transport system. The trac mutation provides a new clinically significant animal model for human phytosterolemia and provides a new means for studying the role of phytosterols in hematologic diseases and testing therapeutic interventions.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Cardiomiopatías/genética , Modelos Animales de Enfermedad , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/fisiología , Mutación/genética , Fitosteroles/metabolismo , Sitoesteroles/metabolismo , Trombocitopenia/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Tiempo de Sangría , Cardiomiopatías/patología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Cruzamientos Genéticos , Femenino , Feto/citología , Feto/metabolismo , Errores Innatos del Metabolismo Lipídico/patología , Lipoproteínas/genética , Hígado/citología , Hígado/metabolismo , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Trombocitopenia/patología
15.
J Neurosci ; 30(29): 9683-94, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20660251

RESUMEN

The quantitative trait locus ahl8 is a key contributor to the early-onset, age-related hearing loss of DBA/2J mice. A nonsynonymous nucleotide substitution in the mouse fascin-2 gene (Fscn2) is responsible for this phenotype, confirmed by wild-type BAC transgene rescue of hearing loss in DBA/2J mice. In chickens and mice, FSCN2 protein is abundant in hair-cell stereocilia, the actin-rich structures comprising the mechanically sensitive hair bundle, and is concentrated toward stereocilia tips of the bundle's longest stereocilia. FSCN2 expression increases when these stereocilia differentially elongate, suggesting that FSCN2 controls filament growth, stiffens exposed stereocilia, or both. Because ahl8 accelerates hearing loss only in the presence of mutant cadherin 23, a component of hair-cell tip links, mechanotransduction and actin crosslinking must be functionally interrelated.


Asunto(s)
Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Células Ciliadas Auditivas Internas/metabolismo , Pérdida Auditiva/genética , Proteínas de Microfilamentos/genética , Mutación Missense , Actinas/genética , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Cadherinas/genética , Cadherinas/metabolismo , Embrión de Pollo , Progresión de la Enfermedad , Potenciales Evocados Auditivos , Ratones , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Polimorfismo Genético , Sáculo y Utrículo/ultraestructura , Xenopus laevis
16.
Hear Res ; 409: 108327, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34388681

RESUMEN

CACHD1 recently was shown to be an α2δ-like subunit that can modulate the activity of some types of voltage-gated calcium channels, including the low-voltage activated, T-type CaV3 channels. CACHD1 is widely expressed in the central nervous system but its biological functions and relationship to disease states are unknown. Here, we report that mice with deleterious Cachd1 mutations are hearing impaired and have balance defects, demonstrating that CACHD1 is functionally important in the peripheral auditory and vestibular organs of the inner ear. The vestibular dysfunction of Cachd1 mutant mice, exhibited by leaning and head tilting behaviors, is related to a deficiency of calcium carbonate crystals (otoconia) in the saccule and utricle. The auditory dysfunction, shown by ABR threshold elevations and reduced DPOAEs, is associated with reduced endocochlear potentials and increased endolymph calcium concentrations. Paint-fills of mutant inner ears from prenatal and newborn mice revealed dilation of the membranous labyrinth caused by an enlarged volume of endolymph. These pathologies all can be related to a disturbance of calcium homeostasis in the endolymph of the inner ear, presumably caused by the loss of CACHD1 regulatory effects on voltage-gated calcium channel activity. Cachd1 expression in the cochlea appears stronger in late embryonic stages than in adults, suggesting an early role in establishing endolymph calcium concentrations. Our findings provide new insights into CACHD1 function and suggest the involvement of voltage-gated calcium channels in endolymph homeostasis, essential for normal auditory and vestibular function.


Asunto(s)
Audición , Homeostasis , Animales , Animales Recién Nacidos , Calcio , Canales de Calcio , Femenino , Ratones , Embarazo , Vestíbulo del Laberinto
17.
Genesis ; 48(10): 578-84, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20715164

RESUMEN

DSCAMs are cell adhesion molecules that play several important roles in neurodevelopment. Mouse alleles of Dscam identified to date do not survive on an inbred C57BL/6 background, complicating analysis of DSCAM-dependent developmental processes because of phenotypic variability related to the segregating backgrounds needed for postnatal survival. A novel spontaneous allele of Dscam, hereafter referred to as Dscam²(J), has been identified. This allele contains a four base pair duplication in exon 19, leading to a frameshift and truncation of the open reading frame. Mice homozygous for the Dscam²(J) mutant allele survive into adulthood on the C3H/HeJ background on which the mutation was identified. Using the Dscam²(J) allele, retinal phenotypes that have variable severity on a segregating background were examined. A neurite lamination defect similar to that described in chick was discovered in mice. These results indicate that, in the retina, additional DSCAM-dependent processes can be found by analysis of mutations on different genetic backgrounds.


Asunto(s)
Alelos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Ratones Endogámicos C3H , Fenotipo , Animales , Emparejamiento Base , Exones , Mutación del Sistema de Lectura , Homocigoto , Ratones , Ratones Noqueados , Mutación/genética , Neuritas/fisiología , Retina/citología , Retina/metabolismo
18.
BMC Dev Biol ; 9: 6, 2009 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19161597

RESUMEN

BACKGROUND: The evolutionarily conserved Notch signalling pathway regulates multiple developmental processes in a wide variety of organisms. One critical posttranslational modification of Notch for its function in vivo is the addition of O-linked fucose residues by protein O-fucosyltransferase 1 (POFUT1). In addition, POFUT1 acts as a chaperone and is required for Notch trafficking. Mouse embryos lacking POFUT1 function die with a phenotype indicative of global inactivation of Notch signalling. O-linked fucose residues on Notch can serve as substrates for further sugar modification by Fringe (FNG) proteins. Notch modification by Fringe differently affects the ability of ligands to activate Notch receptors in a context-dependent manner indicating a complex modulation of Notch activity by differential glycosylation. Whether the context-dependent effects of Notch receptor glycosylation by FNG reflect different requirements of distinct developmental processes for O-fucosylation by POFUT1 is unclear. RESULTS: We have identified and characterized a spontaneous mutation in the mouse Pofut1 gene, referred to as "compact axial skeleton" (cax). Cax carries an insertion of an intracisternal A particle retrotransposon into the fourth intron of the Pofut1 gene and represents a hypomorphic Pofut1 allele that reduces transcription and leads to reduced Notch signalling. Cax mutant embryos have somites of variable size, showed partly abnormal Lfng expression and, consistently defective anterior-posterior somite patterning and axial skeleton development but had virtually no defects in several other Notch-regulated early developmental processes outside the paraxial mesoderm that we analyzed. CONCLUSION: Notch-dependent processes apparently differ with respect to their requirement for levels of POFUT1. Normal Lfng expression and anterior-posterior somite patterning is highly sensitive to reduced POFUT1 levels in early mammalian embryos, whereas other early Notch-dependent processes such as establishment of left-right asymmetry or neurogenesis are not. Thus, it appears that in the presomitic mesoderm (PSM) Notch signalling is particularly sensitive to POFUT1 levels. Reduced POFUT1 levels might affect Notch trafficking or overall O-fucosylation. Alternatively, reduced O-fucosylation might preferentially affect sites that are substrates for LFNG and thus important for somite formation and patterning.


Asunto(s)
Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Mesodermo/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Tipificación del Cuerpo/fisiología , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Inmunohistoquímica , Mesodermo/embriología , Mesodermo/enzimología , Ratones , Mutación
19.
BMC Dev Biol ; 9: 27, 2009 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-19379485

RESUMEN

BACKGROUND: The H6 homeobox genes Hmx1, Hmx2, and Hmx3 (also known as Nkx5-3; Nkx5-2 and Nkx5-1, respectively), compose a family within the NKL subclass of the ANTP class of homeobox genes. Hmx gene family expression is mostly limited to sensory organs, branchial (pharyngeal) arches, and the rostral part of the central nervous system. Targeted mutation of either Hmx2 or Hmx3 in mice disrupts the vestibular system. These tandemly duplicated genes have functional overlap as indicated by the loss of the entire vestibular system in double mutants. Mutants have not been described for Hmx1, the most divergent of the family. RESULTS: Dumbo (dmbo) is a semi-lethal mouse mutation that was recovered in a forward genetic mutagenesis screen. Mutants exhibit enlarged ear pinnae with a distinctive ventrolateral shift. Here, we report on the basis of this phenotype and other abnormalities in the mutant, and identify the causative mutation as being an allele of Hmx1. Examination of dumbo skulls revealed only subtle changes in cranial bone morphology, namely hyperplasia of the gonial bone and irregularities along the caudal border of the squamous temporal bone. Other nearby otic structures were unaffected. The semilethality of dmbo/dmbo mice was found to be ~40%, occured perinatally, and was associated with exencephaly. Surviving mutants of both sexes exhibited reduced body mass from ~3 days postpartum onwards. Most dumbo adults were microphthalmic. Recombinant animals and specific deletion-bearing mice were used to map the dumbo mutation to a 1.8 Mb region on Chromosome 5. DNA sequencing of genes in this region revealed a nonsense mutation in the first exon of H6 Homeobox 1 (Hmx1; also Nkx5-3). An independent spontaneous allele called misplaced ears (mpe) was also identified, confirming Hmx1 as the responsible mutant gene. CONCLUSION: The divergence of Hmx1 from its paralogs is reflected by different and diverse developmental roles exclusive of vestibular involvement. Additionally, these mutant Hmx1 alleles represent the first mouse models of a recently-discovered Oculo-Auricular syndrome caused by mutation of the orthologous human gene.


Asunto(s)
Peso Corporal/genética , Anomalías Craneofaciales/genética , Mutación , Factores de Transcripción/genética , Alelos , Animales , Animales Recién Nacidos , Secuencia de Bases , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Análisis Mutacional de ADN , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Anomalías del Ojo/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Pruebas Auditivas , Proteínas de Homeodominio/efectos adversos , Proteínas de Homeodominio/genética , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/efectos adversos , Proteínas del Tejido Nervioso/genética , Fenotipo
20.
Mamm Genome ; 20(8): 462-75, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19669234

RESUMEN

Podosome-type adhesions are actin-based membrane protrusions involved in cell-matrix adhesion and extracellular matrix degradation. Despite growing knowledge of many proteins associated with podosome-type adhesions, much remains unknown concerning the function of podosomal proteins at the level of the whole animal. In this study, the spontaneous mouse mutant nee was used to identify a component of podosome-type adhesions that is essential for normal postnatal growth and development. Mice homozygous for the nee allele exhibited runted growth, craniofacial and skeletal abnormalities, ocular anterior segment dysgenesis, and hearing impairment. Adults also exhibited infertility and a form of lipodystrophy. Using genetic mapping and DNA sequencing, the cause of nee phenotypes was identified as a 1-bp deletion within the Sh3pxd2b gene on mouse Chromosome 11. Whereas the wild-type Sh3pxd2b gene is predicted to encode a protein with one PX domain and four SH3 domains, the nee mutation is predicted to cause a frameshift and a protein truncation altering a portion of the third SH3 domain and deleting all of the fourth SH3 domain. The SH3PXD2B protein is believed to be an important component of podosomes likely to mediate protein-protein interactions with membrane-spanning metalloproteinases. Testing this directly, SH3PXD2B localized to podosomes in constitutively active Src-transfected fibroblasts and through its last SH3 domain associated with a transmembrane member of a disintegrin and metalloproteinase family of proteins, ADAM15. These results identify SH3PXD2B as a podosomal-adaptor protein required for postnatal growth and development, particularly within physiologic contexts involving extracellular matrix regulation.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Ratones/crecimiento & desarrollo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Extensiones de la Superficie Celular/química , Extensiones de la Superficie Celular/genética , Mapeo Cromosómico , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Femenino , Masculino , Ratones/genética , Ratones/metabolismo , Datos de Secuencia Molecular , Mutación , Proteínas de Transferencia de Fosfolípidos/química , Proteínas de Transferencia de Fosfolípidos/genética , Transporte de Proteínas , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA