Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Med ; 25(7): 1123-1130, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270503

RESUMEN

Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.


Asunto(s)
Alelos , Edición Génica , Pérdida Auditiva Sensorineural/prevención & control , Proteínas de la Membrana/genética , Animales , Proteína 9 Asociada a CRISPR/fisiología , Línea Celular , Células Cultivadas , Dependovirus/genética , Modelos Animales de Enfermedad , Pérdida Auditiva Sensorineural/genética , Humanos , Ratones , Ratones Endogámicos C57BL
2.
Nat Commun ; 10(1): 734, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30737404

RESUMEN

The original version of this Article contained errors in Fig. 5. In panels i and j the three rightmost x-axis labels inadvertently read 'Tmc1' instead of 'Tmc2'. These errors have been corrected in both the PDF and HTML versions of the Article.

3.
Nat Commun ; 10(1): 236, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670701

RESUMEN

Fifty percent of inner ear disorders are caused by genetic mutations. To develop treatments for genetic inner ear disorders, we designed gene replacement therapies using synthetic adeno-associated viral vectors to deliver the coding sequence for Transmembrane Channel-Like (Tmc) 1 or 2 into sensory hair cells of mice with hearing and balance deficits due to mutations in Tmc1 and closely related Tmc2. Here we report restoration of function in inner and outer hair cells, enhanced hair cell survival, restoration of cochlear and vestibular function, restoration of neural responses in auditory cortex and recovery of behavioral responses to auditory and vestibular stimulation. Secondarily, we find that inner ear Tmc gene therapy restores breeding efficiency, litter survival and normal growth rates in mouse models of genetic inner ear dysfunction. Although challenges remain, the data suggest that Tmc gene therapy may be well suited for further development and perhaps translation to clinical application.


Asunto(s)
Sordera/genética , Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Pérdida Auditiva/genética , Enfermedades del Laberinto/genética , Proteínas de la Membrana/genética , Animales , Sordera/terapia , Células Ciliadas Auditivas/fisiología , Células Ciliadas Vestibulares/fisiología , Pérdida Auditiva/terapia , Enfermedades del Laberinto/terapia , Ratones , Ratones Mutantes
4.
Sci Rep ; 8(1): 12124, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30108254

RESUMEN

Recent work has demonstrated that transmembrane channel-like 1 protein (TMC1) is an essential component of the sensory transduction complex in hair cells of the inner ear. A closely related homolog, TMC2, is expressed transiently in the neonatal mouse cochlea and can enable sensory transduction in Tmc1-null mice during the first postnatal week. Both TMC1 and TMC2 are expressed at adult stages in mouse vestibular hair cells. The extent to which TMC1 and TMC2 can substitute for each other is unknown. Several biophysical differences between TMC1 and TMC2 suggest these proteins perform similar but not identical functions. To investigate these differences, and whether TMC2 can substitute for TMC1 in mature hair cells, we generated a knock-in mouse model allowing Cre-inducible expression of Tmc2. We assayed for changes in hair cell sensory transduction and auditory and vestibular function in Tmc2 knockin mice (Tm[Tmc2]) in the presence or absence of endogenous Tmc1, Tmc2 or both. Our results show that expression of Tm[TMC2] restores sensory transduction in vestibular hair cells and transiently in cochlear hair cells in the absence of TMC1. The cellular rescue leads to recovery of balance but not auditory function. We conclude that TMC1 provides some additional necessary function, not provided by TMC2.


Asunto(s)
Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Vestibulares/metabolismo , Proteínas de la Membrana/metabolismo , Equilibrio Postural/fisiología , Animales , Técnicas de Sustitución del Gen , Audición/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Modelos Animales , Transgenes/genética
5.
Neuron ; 99(4): 736-753.e6, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30138589

RESUMEN

The proteins that form the permeation pathway of mechanosensory transduction channels in inner-ear hair cells have not been definitively identified. Genetic, anatomical, and physiological evidence support a role for transmembrane channel-like protein (TMC) 1 in hair cell sensory transduction, yet the molecular function of TMC proteins remains unclear. Here, we provide biochemical evidence suggesting TMC1 assembles as a dimer, along with structural and sequence analyses suggesting similarity to dimeric TMEM16 channels. To identify the pore region of TMC1, we used cysteine mutagenesis and expressed mutant TMC1 in hair cells of Tmc1/2-null mice. Cysteine-modification reagents rapidly and irreversibly altered permeation properties of mechanosensory transduction. We propose that TMC1 is structurally similar to TMEM16 channels and includes ten transmembrane domains with four domains, S4-S7, that line the channel pore. The data provide compelling evidence that TMC1 is a pore-forming component of sensory transduction channels in auditory and vestibular hair cells.


Asunto(s)
Células Ciliadas Auditivas Internas/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/química , Proteínas de la Membrana/fisiología , Porinas/química , Porinas/fisiología , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Estructura Secundaria de Proteína
6.
Elife ; 72018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-30019672

RESUMEN

Human vestibular sensory epithelia in explant culture were incubated in gentamicin to ablate hair cells. Subsequent transduction of supporting cells with ATOH1 using an Ad-2 viral vector resulted in generation of highly significant numbers of cells expressing the hair cell marker protein myosin VIIa. Cells expressing myosin VIIa were also generated after blocking the Notch signalling pathway with TAPI-1 but less efficiently. Transcriptomic analysis following ATOH1 transduction confirmed up-regulation of 335 putative hair cell marker genes, including several downstream targets of ATOH1. Morphological analysis revealed numerous cells bearing dense clusters of microvilli at the apical surfaces which showed some hair cell-like characteristics confirming a degree of conversion of supporting cells. However, no cells bore organised hair bundles and several expected hair cell markers genes were not expressed suggesting incomplete differentiation. Nevertheless, the results show a potential to induce conversion of supporting cells in the vestibular sensory tissues of humans.


Asunto(s)
Epitelio/fisiología , Células Ciliadas Vestibulares/fisiología , Regeneración/fisiología , Adenoviridae/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Epitelio/ultraestructura , Regulación de la Expresión Génica , Gentamicinas/efectos adversos , Proteínas Fluorescentes Verdes/metabolismo , Células Ciliadas Vestibulares/patología , Células Ciliadas Vestibulares/ultraestructura , Humanos , Miosina VIIa , Miosinas/metabolismo , Receptores Notch/metabolismo , Sáculo y Utrículo/fisiología , Sáculo y Utrículo/ultraestructura , Transducción de Señal , Transducción Genética
7.
Nat Biotechnol ; 35(3): 264-272, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28165476

RESUMEN

Because there are currently no biological treatments for hearing loss, we sought to advance gene therapy approaches to treat genetic deafness. We focused on Usher syndrome, a devastating genetic disorder that causes blindness, balance disorders and profound deafness, and studied a knock-in mouse model, Ush1c c.216G>A, for Usher syndrome type IC (USH1C). As restoration of complex auditory and balance function is likely to require gene delivery systems that target auditory and vestibular sensory cells with high efficiency, we delivered wild-type Ush1c into the inner ear of Ush1c c.216G>A mice using a synthetic adeno-associated viral vector, Anc80L65, shown to transduce 80-90% of sensory hair cells. We demonstrate recovery of gene and protein expression, restoration of sensory cell function, rescue of complex auditory function and recovery of hearing and balance behavior to near wild-type levels. The data represent unprecedented recovery of inner ear function and suggest that biological therapies to treat deafness may be suitable for translation to humans with genetic inner ear disorders.


Asunto(s)
Proteínas Portadoras/genética , Terapia Genética/métodos , Pérdida Auditiva Sensorineural/terapia , Síndromes de Usher/genética , Síndromes de Usher/terapia , Enfermedades Vestibulares/terapia , Animales , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Femenino , Técnicas de Sustitución del Gen , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación , Plásmidos/genética , Recuperación de la Función/genética , Resultado del Tratamiento , Enfermedades Vestibulares/diagnóstico , Enfermedades Vestibulares/genética
8.
Sci Transl Med ; 7(295): 295ra108, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26157030

RESUMEN

Genetic hearing loss accounts for up to 50% of prelingual deafness worldwide, yet there are no biologic treatments currently available. To investigate gene therapy as a potential biologic strategy for restoration of auditory function in patients with genetic hearing loss, we tested a gene augmentation approach in mouse models of genetic deafness. We focused on DFNB7/11 and DFNA36, which are autosomal recessive and dominant deafnesses, respectively, caused by mutations in transmembrane channel-like 1 (TMC1). Mice that carry targeted deletion of Tmc1 or a dominant Tmc1 point mutation, known as Beethoven, are good models for human DFNB7/11 and DFNA36. We screened several adeno-associated viral (AAV) serotypes and promoters and identified AAV2/1 and the chicken ß-actin (Cba) promoter as an efficient combination for driving the expression of exogenous Tmc1 in inner hair cells in vivo. Exogenous Tmc1 or its closely related ortholog, Tmc2, were capable of restoring sensory transduction, auditory brainstem responses, and acoustic startle reflexes in otherwise deaf mice, suggesting that gene augmentation with Tmc1 or Tmc2 is well suited for further development as a strategy for restoration of auditory function in deaf patients who carry TMC1 mutations.


Asunto(s)
Sordera/terapia , Terapia Genética , Audición , Proteínas de la Membrana/genética , Animales , Sordera/genética , Dependovirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
9.
Hear Res ; 311: 17-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24423408

RESUMEN

Transmembrane channel-like (TMC) proteins 1 and 2 are necessary for hair cell mechanotransduction but their precise function is controversial. A growing body of evidence supports a direct role for TMC1 and TMC2 as components of the transduction complex. However, a number of important questions remain and alternate hypotheses have been proposed. Here we present an historical overview of the identification and cloning of Tmc genes, a discussion of mutations in TMC1 that cause deafness in mice and humans and a brief review of other members of the Tmc gene superfamily. We also examine expression of Tmc mRNAs and localization of the protein products. The review focuses on potential functions of TMC proteins and the evidence from Beethoven mice that suggests a direct role for TMC1 in hair cell mechanotransduction. Data that support alternate interpretations are also considered. The article concludes with a discussion of outstanding questions and future directions for TMC research. This article is part of a Special Issue entitled .


Asunto(s)
Percepción Auditiva , Células Ciliadas Auditivas/metabolismo , Audición , Mecanotransducción Celular , Proteínas de la Membrana/metabolismo , Estimulación Acústica , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Trastornos de la Audición/genética , Trastornos de la Audición/metabolismo , Trastornos de la Audición/fisiopatología , Trastornos de la Audición/psicología , Humanos , Potenciales de la Membrana , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Datos de Secuencia Molecular
10.
Neuron ; 79(3): 504-15, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23871232

RESUMEN

Sensory transduction in auditory and vestibular hair cells requires expression of transmembrane channel-like (Tmc) 1 and 2 genes, but the function of these genes is unknown. To investigate the hypothesis that TMC1 and TMC2 proteins are components of the mechanosensitive ion channels that convert mechanical information into electrical signals, we recorded whole-cell and single-channel currents from mouse hair cells that expressed Tmc1, Tmc2, or mutant Tmc1. Cells that expressed Tmc2 had high calcium permeability and large single-channel currents, while cells with mutant Tmc1 had reduced calcium permeability and reduced single-channel currents. Cells that expressed Tmc1 and Tmc2 had a broad range of single-channel currents, suggesting multiple heteromeric assemblies of TMC subunits. The data demonstrate TMC1 and TMC2 are components of hair cell transduction channels and contribute to permeation properties. Gradients in TMC channel composition may also contribute to variation in sensory transduction along the tonotopic axis of the mammalian cochlea.


Asunto(s)
Fenómenos Biofísicos/fisiología , Células Ciliadas Auditivas/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/metabolismo , Estimulación Acústica , Adenoviridae/genética , Factores de Edad , Animales , Percepción Auditiva/fisiología , Fenómenos Biofísicos/efectos de los fármacos , Fenómenos Biofísicos/genética , Calcio/metabolismo , Calcio/farmacología , Recuento de Células , Células Cultivadas , Relación Dosis-Respuesta a Droga , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Células Ciliadas Auditivas/metabolismo , Técnicas In Vitro , Mecanotransducción Celular/genética , Potenciales de la Membrana/genética , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Mutación/genética , Órgano Espiral/citología , Técnicas de Placa-Clamp , Transducción Genética
11.
J Clin Invest ; 121(12): 4796-809, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22105175

RESUMEN

Inner ear hair cells convert the mechanical stimuli of sound, gravity, and head movement into electrical signals. This mechanotransduction process is initiated by opening of cation channels near the tips of hair cell stereocilia. Since the identity of these ion channels is unknown, and mutations in the gene encoding transmembrane channel-like 1 (TMC1) cause hearing loss without vestibular dysfunction in both mice and humans, we investigated the contribution of Tmc1 and the closely related Tmc2 to mechanotransduction in mice. We found that Tmc1 and Tmc2 were expressed in mouse vestibular and cochlear hair cells and that GFP-tagged TMC proteins localized near stereocilia tips. Tmc2 expression was transient in early postnatal mouse cochlear hair cells but persisted in vestibular hair cells. While mice with a targeted deletion of Tmc1 (Tmc1(Δ) mice) were deaf and those with a deletion of Tmc2 (Tmc2(Δ) mice) were phenotypically normal, Tmc1(Δ)Tmc2(Δ) mice had profound vestibular dysfunction, deafness, and structurally normal hair cells that lacked all mechanotransduction activity. Expression of either exogenous TMC1 or TMC2 rescued mechanotransduction in Tmc1(Δ)Tmc2(Δ) mutant hair cells. Our results indicate that TMC1 and TMC2 are necessary for hair cell mechanotransduction and may be integral components of the mechanotransduction complex. Our data also suggest that persistent TMC2 expression in vestibular hair cells may preserve vestibular function in humans with hearing loss caused by TMC1 mutations.


Asunto(s)
Sordera/genética , Células Ciliadas Auditivas Internas/fisiología , Células Ciliadas Vestibulares/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/fisiología , Animales , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Femenino , Colorantes Fluorescentes/metabolismo , Prueba de Complementación Genética , Gentamicinas/metabolismo , Células Ciliadas Auditivas Internas/ultraestructura , Células Ciliadas Vestibulares/ultraestructura , Masculino , Mecanotransducción Celular/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , ARN Mensajero/biosíntesis , Estereocilios/fisiología , Estereocilios/ultraestructura
12.
Gene Expr Patterns ; 10(2-3): 113-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20085825

RESUMEN

The zebrafish provides a useful experimental system for investigations of aural development. To permit the controlled expression of transgenes in developing hair cells, we isolated the genomic control regions of the parvalbumin 3a (pvalb3a) and parvalbumin 3b (pvalb3b) genes. Deletion analysis and somatic-cell transgenesis restricted the cis-acting control regions for hair cells to as little as 484base pairs for pvalb3a and 650base pairs for pvalb3b. Using both meganuclease-mediated and standard methods, we produced transgenic animals that transmit transgenes through their germ lines. These fish express GFP in hair cells in the inner ear and lateral line. Two stable transgenic lines express GFP prior to hair-bundle formation, so the associated promoter constructs are suitable for manipulating gene expression during bundle development. We additionally identified a transgenic line that offers variable labeling of supporting cells.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Parvalbúminas/genética , Transgenes , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Pez Cebra/embriología , Pez Cebra/genética
13.
J Assoc Res Otolaryngol ; 11(1): 27-37, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19834762

RESUMEN

TRP genes encode a diverse family of ion channels which have been implicated in many sensory functions. Because several TRP channels have similar properties to the elusive hair cell transduction channel, recent attention has focused on TRP gene expression in the inner ear. At least four TRP genes are known to be expressed in hair cells: TRPC3, TRPV4, TRPA1, and TRPML3. However, there is little evidence supporting any of these as a component of the transduction complex. Other less well-characterized TRP channels are expressed in the inner ear, in particular, within the organ of Corti. Because of their potential role in sensory function, we investigated the developmental expression of RNA that encodes all 33 TRP subunits as well as several splice variants. We designed a quantitative PCR screen using cochlear samples acquired before, during, and after the time when mechanotransduction is acquired in sensory hair cells (embryonic day 17 to postnatal day 8). Cochleas, which included the organ of Corti, stria vascularis, and Reissner's membrane, were subdivided into four equal quadrants which allowed for regional comparison during development. Expression of RNA transcripts that encoded 33 TRP subunits plus several splice forms and beta-actin were quantified in 28 samples for a total of 1,092 individual measurements, each done in triplicate. We detected RNA that encoded all TRP channels except two: TRPC7 and TRPM8. The largest changes in RNA expression were for TRPA1 (>100-fold), TRPP3 (>50-fold), and TRPC5.2 (>20-fold) which suggested that these subunits may contribute to normal cochlear function. Furthermore, the screen revealed TRPP3 and PKD1L3 RNA expression patterns that were correlated with the acquisition of sensory transduction in outer hair cells (Lelli et al., J Neurophysiol. 101:2961-2973, 2009). Numerous spatiotemporal expression gradients were identified many of which may contribute to the normal functional development of the mouse cochlea.


Asunto(s)
Cóclea/embriología , Cóclea/fisiología , Regulación del Desarrollo de la Expresión Génica , Canales de Potencial de Receptor Transitorio/genética , Empalme Alternativo/genética , Animales , Femenino , Ratones , Familia de Multigenes/genética , Reacción en Cadena de la Polimerasa , Embarazo , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPV/genética
14.
J Neurophysiol ; 101(6): 2961-73, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19339464

RESUMEN

Inner ear hair cells are exquisite mechanosensors that transduce nanometer scale deflections of their sensory hair bundles into electrical signals. Several essential elements must be precisely assembled during development to confer the unique structure and function of the mechanotransduction apparatus. Here we investigated the functional development of the transduction complex in outer hair cells along the length of mouse cochlea acutely excised between embryonic day 17 (E17) and postnatal day 8 (P8). We charted development of the stereociliary bundle using scanning electron microscopy; FM1-43 uptake, which permeates hair cell transduction channels, mechanotransduction currents evoked by rapid hair bundle deflections, and mRNA expression of possible components of the transduction complex. We demonstrated that uptake of FM1-43 first occurred in the basal portion of the cochlea at P0 and progressed toward the apex over the subsequent week. Electrophysiological recordings obtained from 234 outer hair cells between E17 and P8 from four cochlear regions revealed a correlation between the pattern of FM1-43 uptake and the acquisition of mechanotransduction. We found a spatiotemporal gradient in the properties of transduction including onset, amplitude, operating range, time course, and extent of adaptation. We used quantitative RT-PCR to examine relative mRNA expression of several hair cell myosins and candidate tip-link molecules. We found spatiotemporal expression patterns for mRNA that encodes cadherin 23, protocadherin 15, myosins 3a, 7a, 15a, and PMCA2 that preceded the acquisition of transduction. The spatiotemporal expression patterns of myosin 1c and PMCA2 mRNA were correlated with developmental changes in several properties of mechanotransduction.


Asunto(s)
Cóclea , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Ciliadas Auditivas Externas/fisiología , Mecanotransducción Celular/fisiología , Adaptación Fisiológica , Animales , Animales Recién Nacidos , Cóclea/citología , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Embrión de Mamíferos , Células Ciliadas Auditivas Externas/ultraestructura , Potenciales de la Membrana/fisiología , Ratones , Microscopía Confocal/métodos , Microscopía Electrónica de Rastreo/métodos , Miosinas/clasificación , Miosinas/genética , Miosinas/metabolismo , Técnicas de Placa-Clamp , Estimulación Física/métodos , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , ARN Mensajero/metabolismo
15.
Proc Natl Acad Sci U S A ; 103(24): 9069-74, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16754885

RESUMEN

The development of the vertebrate inner ear depends on the precise expression of fibroblast growth factors. In a mutagenesis screen for zebrafish with abnormalities of inner-ear development and behavior, we isolated a mutant line, ru622, whose phenotypic characteristics resembled those of null mutants for the gene encoding fibroblast growth factor 8 (Fgf8): an inconsistent startle response, circular swimming, fused otoliths, and abnormal semicircular canals. Positional cloning disclosed that the mutant gene encodes the transcriptional corepressor Atrophin2. Both the Fgf8 protein and zebrafish "similar expression to fgf genes" protein (Sef), an antagonist of fibroblast growth factors induced by Fgf8 itself, were found to be overexpressed in ru622 mutants. We therefore hypothesized that an excess of Sef eliminates Fgf8 signals and produces an fgf8 null phenotype in ru622 mutants. In support of this idea, we could rescue larvae whose atrophin2 expression had been diminished with morpholinos by reducing the expression of Sef as well. We propose that Atrophin2 plays a role in the feedback regulation of Fgf8 signaling. When mutation of the atrophin2 gene results in the overexpression of both Fgf8 and Sef, the excessive Sef inhibits Fgf8 signaling. The resultant imbalance of Fgf8 and Sef signals then underlies the abnormal aural development observed in ru622.


Asunto(s)
Oído Interno , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso , Proteínas Represoras , Transducción de Señal/fisiología , Pez Cebra/fisiología , Animales , Conducta Animal/fisiología , Oído Interno/anatomía & histología , Oído Interno/embriología , Oído Interno/crecimiento & desarrollo , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Reflejo/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Pez Cebra/anatomía & histología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
J Mol Biol ; 327(2): 453-63, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12628250

RESUMEN

The bacterial flagellar motor is a tiny molecular machine that uses a transmembrane flux of H(+) or Na(+) ions to drive flagellar rotation. In proton-driven motors, the membrane proteins MotA and MotB interact via their transmembrane regions to form a proton channel. The sodium-driven motors that power the polar flagellum of Vibrio species contain homologs of MotA and MotB, called PomA and PomB. They require the unique proteins MotX and MotY. In this study, we investigated how ion selectivity is determined in proton and sodium motors. We found that Escherichia coli MotA/B restore motility in DeltapomAB Vibrio alginolyticus. Most hypermotile segregants isolated from this weakly motile strain contain mutations in motB. We constructed proteins in which segments of MotB were fused to complementary portions of PomB. A chimera joining the N terminus of PomB to the periplasmic C terminus of MotB (PotB7(E)) functioned with PomA as the stator of a sodium motor, with or without MotX/Y. This stator (PomA/PotB7(E)) supported sodium-driven motility in motA or motB E.coli cells, and the swimming speed was even higher than with the original stator of E.coli MotA/B. We conclude that the cytoplasmic and transmembrane domains of PomA/B are sufficient for sodium-driven motility. However, MotA expressed with a B subunit containing the N terminus of MotB fused to the periplasmic domain of PomB (MomB7(E)) supported sodium-driven motility in a MotX/Y-dependent fashion. Thus, although the periplasmic domain of PomB is not necessary for sodium-driven motility in a PomA/B motor, it can convert a MotA/B proton motor into a sodium motor.


Asunto(s)
Proteínas Bacterianas/metabolismo , Escherichia coli/fisiología , Flagelos/fisiología , Hidrógeno , Proteínas Motoras Moleculares/fisiología , Canales de Sodio/metabolismo , Sodio , Vibrio/fisiología , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Escherichia coli/genética , Flagelos/genética , Concentración de Iones de Hidrógeno , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Plásmidos , Homología de Secuencia de Aminoácido , Canales de Sodio/genética , Transformación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA