Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nature ; 605(7909): 298-303, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35508658

RESUMEN

The cochlea uses two types of mechanosensory cell to detect sounds. A single row of inner hair cells (IHCs) synapse onto neurons to transmit sensory information to the brain, and three rows of outer hair cells (OHCs) selectively amplify auditory inputs1. So far, two transcription factors have been implicated in the specific differentiation of OHCs, whereas, to our knowledge, none has been identified in the differentiation of IHCs2-4. One such transcription factor for OHCs, INSM1, acts during a crucial embryonic period to consolidate the OHC fate, preventing OHCs from transdifferentiating into IHCs2. In the absence of INSM1, embryonic OHCs misexpress a core set of IHC-specific genes, which we predict are involved in IHC differentiation. Here we find that one of these genes, Tbx2, is a master regulator of IHC versus OHC differentiation in mice. Ablation of Tbx2 in embryonic IHCs results in their development as OHCs, expressing early OHC markers such as Insm1 and eventually becoming completely mature OHCs in the position of IHCs. Furthermore, Tbx2 is epistatic to Insm1: in the absence of both genes, cochleae generate only OHCs, which suggests that TBX2 is necessary for the abnormal transdifferentiation of INSM1-deficient OHCs into IHCs, as well as for normal IHC differentiation. Ablation of Tbx2 in postnatal, largely differentiated IHCs makes them transdifferentiate directly into OHCs, replacing IHC features with those of mature and not embryonic OHCs. Finally, ectopic expression of Tbx2 in OHCs results in their transdifferentiation into IHCs. Hence, Tbx2 is both necessary and sufficient to make IHCs distinct from OHCs and maintain this difference throughout development.


Asunto(s)
Diferenciación Celular , Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Externas , Animales , Diferenciación Celular/genética , Cóclea/citología , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Externas/citología , Ratones , Proteínas de Dominio T Box
2.
Proc Natl Acad Sci U S A ; 120(11): e2217891120, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36893263

RESUMEN

Prestin (SLC26A5)-mediated voltage-driven elongations and contractions of sensory outer hair cells within the organ of Corti are essential for mammalian cochlear amplification. However, whether this electromotile activity directly contributes on a cycle-by-cycle basis is currently controversial. By restoring motor kinetics in a mouse model expressing a slowed prestin missense variant, this study provides experimental evidence acknowledging the importance of fast motor action to mammalian cochlear amplification. Our results also demonstrate that the point mutation in prestin disrupting anion transport in other proteins of the SLC26 family does not alter cochlear function, suggesting that the potential weak anion transport of prestin is not essential in the mammalian cochlea.


Asunto(s)
Proteínas de Transporte de Anión , Proteínas , Ratones , Animales , Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Proteínas/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Mamíferos/metabolismo , Aniones/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo
3.
Nature ; 565(7737): E2, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30518865

RESUMEN

In Figs. 1e and 2g of this Letter, the labels 'actin' and 'VGLUT3', respectively, should have been in red instead of green font. This has been corrected online.

4.
J Physiol ; 602(6): 1199-1210, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38431907

RESUMEN

DFNB61 is a recessively inherited nonsyndromic hearing loss caused by mutations in SLC26A5, the gene that encodes the voltage-driven motor protein, prestin. Prestin is abundantly expressed in the auditory outer hair cells that mediate cochlear amplification. Two DFNB61-associated SLC26A5 variants, p.W70X and p.R130S, were identified in patients who are compound heterozygous for these nonsense and missense changes (SLC26A5W70X/R130S ). Our recent study showed that mice homozygous for p.R130S (Slc26a5R130S/R130S ) suffer from hearing loss that is ascribed to significantly reduced motor kinetics of prestin. Given that W70X-prestin is nonfunctional, compound heterozygous Slc26a5R130S/- mice were used as a model for human SLC26A5W70X/R130S . By examining the pathophysiological consequences of p.R130S prestin when it is the sole allele for prestin protein production, we determined that this missense change results in progressive outer hair cell loss in addition to its effects on prestin's motor action. Thus, this study defines the pathogenic roles of p.R130S prestin and identifies a limited time window for potential clinical intervention. KEY POINTS: The voltage-driven motor protein, prestin, is encoded by SLC26A5 and expressed abundantly in cochlear outer hair cells (OHCs). The importance of prestin for normal hearing was demonstrated in mice lacking prestin; however, none of the specific SLC26A5 variants identified to date in human patients has been experimentally demonstrated to be pathogenic. In this study we used both cell lines and a mouse model to define the pathogenic role of compound heterozygous p.W70X (c.209G>A) and p.R130S (c.390A>C) SLC26A5 variants identified in patients with moderate to profound hearing loss. As in patients, mice carrying one copy of p.R130S Slc26a5 showed OHC dysfunction and progressive degeneration, which results in congenital progressive hearing loss. This is the first functional study reporting pathogenic SLC26A5 variants and pointing to the presence of a therapeutic time window for potential clinical interventions targeting the affected OHCs before they are lost.


Asunto(s)
Sordera , Pérdida Auditiva , Animales , Humanos , Ratones , Células Ciliadas Auditivas Externas/fisiología , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Mutación , Proteínas/genética
5.
Nature ; 563(7733): 691-695, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30305733

RESUMEN

The mammalian cochlea contains two types of mechanosensory hair cell that have different and critical functions in hearing. Inner hair cells (IHCs), which have an elaborate presynaptic apparatus, signal to cochlear neurons and communicate sound information to the brain. Outer hair cells (OHCs) mechanically amplify sound-induced vibrations, providing enhanced sensitivity to sound and sharp tuning. Cochlear hair cells are solely generated during development, and hair cell death-most often of OHCs-is the most common cause of deafness. OHCs and IHCs, together with supporting cells, originate in embryos from the prosensory region of the otocyst, but how hair cells differentiate into two different types is unknown1-3. Here we show that Insm1, which encodes a zinc finger protein that is transiently expressed in nascent OHCs, consolidates their fate by preventing trans-differentiation into IHCs. In the absence of INSM1, many hair cells that are born as OHCs switch fates to become mature IHCs. To identify the genetic mechanisms by which Insm1 operates, we compared the transcriptomes of immature IHCs and OHCs, and of OHCs with and without INSM1. In OHCs that lack INSM1, a set of genes is upregulated, most of which are normally preferentially expressed by IHCs. The homeotic cell transformation of OHCs without INSM1 into IHCs reveals a mechanism by which these neighbouring mechanosensory cells begin to differ: INSM1 represses a core set of early IHC-enriched genes in embryonic OHCs and makes them unresponsive to an IHC-inducing gradient, so that they proceed to mature as OHCs. Without INSM1, some of the OHCs in which these few IHC-enriched transcripts are upregulated trans-differentiate into IHCs, identifying candidate genes for IHC-specific differentiation.


Asunto(s)
Transdiferenciación Celular/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Externas/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Animales , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética , Proteínas Represoras , Factores de Transcripción/metabolismo , Transcriptoma/genética , Regulación hacia Arriba/genética
6.
Proc Natl Acad Sci U S A ; 117(24): 13571-13579, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32482850

RESUMEN

Synchronized beating of cilia on multiciliated cells (MCCs) generates a directional flow of mucus across epithelia. This motility requires a "9 + 2" microtubule (MT) configuration in axonemes and the unidirectional array of basal bodies of cilia on the MCCs. However, it is not fully understood what components are needed for central MT-pair assembly as they are not continuous with basal bodies in contrast to the nine outer MT doublets. In this study, we discovered that a homozygous knockdown mouse model for MT minus-end regulator calmodulin-regulated spectrin-associated protein 3 (CAMSAP3), Camsap3tm1a/tm1a , exhibited multiple phenotypes, some of which are typical of primary ciliary dyskinesia (PCD), a condition caused by motile cilia defects. Anatomical examination of Camsap3tm1a/tm1a mice revealed severe nasal airway blockage and abnormal ciliary morphologies in nasal MCCs. MCCs from different tissues exhibited defective synchronized beating and ineffective generation of directional flow likely underlying the PCD-like phenotypes. In normal mice, CAMSAP3 localized to the base of axonemes and at the basal bodies in MCCs. However, in Camsap3tm1a/tm1a , MCCs lacked CAMSAP3 at the ciliary base. Importantly, the central MT pairs were missing in the majority of cilia, and the polarity of the basal bodies was disorganized. These phenotypes were further confirmed in MCCs of Xenopus embryos when CAMSAP3 expression was knocked down by morpholino injection. Taken together, we identified CAMSAP3 as being important for the formation of central MT pairs, proper orientation of basal bodies, and synchronized beating of motile cilia.


Asunto(s)
Cuerpos Basales/metabolismo , Cilios/metabolismo , Trastornos de la Motilidad Ciliar/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Animales , Axonema/metabolismo , Polaridad Celular , Trastornos de la Motilidad Ciliar/genética , Células Epiteliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Xenopus
7.
Biophys J ; 120(21): 4777-4785, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34555361

RESUMEN

Studies of genetic disorders of sensorineural hearing loss have been instrumental in delineating mechanisms that underlie the remarkable sensitivity and selectivity that are hallmarks of mammalian hearing. For example, genetic modifications of TECTA and TECTB, which are principal proteins that comprise the tectorial membrane (TM), have been shown to alter auditory thresholds and frequency tuning in ways that can be understood in terms of changes in the mechanical properties of the TM. Here, we investigate effects of genetic modification targeting CEACAM16, a third important TM protein. Loss of CEACAM16 has been recently shown to lead to progressive reductions in sensitivity. Whereas age-related hearing losses have previously been linked to changes in sensory receptor cells, the role of the TM in progressive hearing loss is largely unknown. Here, we show that TM stiffness and viscosity are significantly reduced in adult mice that lack functional CEACAM16 relative to age-matched wild-type controls. By contrast, these same mechanical properties of TMs from juvenile mice that lack functional CEACAM16 are more similar to those of wild-type mice. Thus, changes in hearing phenotype align with changes in TM material properties and can be understood in terms of the same TM wave properties that were previously used to characterize modifications of TECTA and TECTB. These results demonstrate that CEACAM16 is essential for maintaining TM mechanical and wave properties, which in turn are necessary for sustaining the remarkable sensitivity and selectivity of mammalian hearing with increasing age.


Asunto(s)
Moléculas de Adhesión Celular , Pérdida Auditiva , Membrana Tectoria , Factores de Edad , Animales , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/metabolismo , Proteínas de la Matriz Extracelular , Audición , Ratones , Viscosidad
8.
J Neurosci ; 38(13): 3177-3189, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29453205

RESUMEN

Acquired hearing loss is the predominant neurodegenerative condition associated with aging in humans. Although mutations on several genes are known to cause congenital deafness in newborns, few genes have been implicated in age-related hearing loss (ARHL), perhaps because its cause is likely polygenic. Here, we generated mice lacking lysosomal calcium channel mucolipins 3 and 1 and discovered that both male and female mice suffered a polygenic form of hearing loss. Whereas mucolipin 1 is ubiquitously expressed in all cells, mucolipin 3 is expressed in a small subset of cochlear cells, hair cells (HCs) and marginal cells of the stria vascularis, and very few other cell types. Mice lacking both mucolipins 3 and 1, but not either one alone, experienced hearing loss as early as at 1 month of age. The severity of hearing impairment progressed from high to low frequencies and increased with age. Early onset of ARHL in these mice was accompanied by outer HC (OHC) loss. Adult mice conditionally lacking mucolipins in HCs exhibited comparable auditory phenotypes, thereby revealing that the reason for OHC loss is mucolipin codeficiency in the HCs and not in the stria vascularis. Furthermore, we observed that OHCs lacking mucolipins contained abnormally enlarged lysosomes aggregated at the apical region of the cell, whereas other organelles appeared normal. We also demonstrated that these aberrant lysosomes in OHCs lost their membrane integrity through lysosomal membrane permeabilization, a known cause of cellular toxicity that explains why and how OHCs die, leading to premature ARHL.SIGNIFICANCE STATEMENT Presbycusis, or age-related hearing loss (ARHL), is a common characteristic of aging in mammals. Although many genes have been identified to cause deafness from birth in both humans and mice, only a few are known to associate with progressive ARHL, the most prevalent form of deafness. We have found that mice lacking two lysosomal channels, mucolipins 3 and 1, suffer accelerated ARHL due to auditory outer hair cell degeneration, the most common cause of hearing loss and neurodegenerative condition in humans. Lysosomes lacking mucolipins undergo organelle membrane permeabilization and promote cytotoxicity with age, revealing a novel mechanism of outer hair cell degeneration and ARHL. These results underscore the importance of lysosomes in hair cell survival and the maintenance of hearing.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Presbiacusia/genética , Canales de Potencial de Receptor Transitorio/genética , Animales , Femenino , Eliminación de Gen , Células Ciliadas Auditivas/patología , Lisosomas/metabolismo , Lisosomas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Presbiacusia/patología
9.
J Neurosci ; 34(4): 1325-32, 2014 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24453323

RESUMEN

The solute carrier gene family 26 (SLC26) encodes membrane proteins with diverse physiological roles but with the common feature of halide involvement. Here, we present bioinformatic and biochemical evidence that SLC26 proteins have intrinsically disordered regions (IDRs) in their C-terminal domains and that these regions contain calmodulin (CaM) binding sites. The veracity of these predictions and the functional consequences of CaM binding were examined in prestin, SLC26A5, as a model for the SLC26 family and as one of the most investigated and best understood members. We found that CaM binds directly to the IDR in the C-terminal domain of prestin in a calcium-obligate manner. Using both isolated murine outer hair cells (OHCs) and a heterologous expression system, we also found that this calcium-obligate CaM binding shifts the operating point of the protein to more hyperpolarized potentials with consequent alteration of the function of the prestin. Because calcium is the main intracellular second messenger used by the efferent medial olivocochlear (MOC) pathway of the auditory system and CaM is abundant in OHCs, the CaM-prestin interaction may be involved in the MOC-mediated modulation of cochlear amplification. However, this regulatory mechanism is not likely to be restricted to cochlear OHCs, in light of both clear bioinformatic evidence and the fact that calcium and CaM are ubiquitous intracellular second messengers used by virtually all cell types. Hence, the calcium/CaM-dependent regulatory mechanism described herein is likely applicable to most, if not all, SLC26 paralogs.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Calmodulina/metabolismo , Proteínas Motoras Moleculares/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Anión/química , Secuencia de Bases , Sitios de Unión , Calcio/metabolismo , Femenino , Células Ciliadas Auditivas Externas/metabolismo , Humanos , Masculino , Ratones , Proteínas Motoras Moleculares/química , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Transportadores de Sulfato
10.
J Neurosci ; 34(31): 10325-38, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25080593

RESUMEN

α-Tectorin (TECTA), ß-tectorin (TECTB), and carcinoembryonic antigen-related cell adhesion molecule 16 (CEACAM) are secreted glycoproteins that are present in the tectorial membrane (TM), an extracellular structure overlying the hearing organ of the inner ear, the organ of Corti. Previous studies have shown that TECTA and TECTB are both required for formation of the striated-sheet matrix within which collagen fibrils of the TM are imbedded and that CEACAM16 interacts with TECTA. To learn more about the structural and functional significance of CEACAM16, we created a Ceacam16-null mutant mouse. In the absence of CEACAM16, TECTB levels are reduced, a clearly defined striated-sheet matrix does not develop, and Hensen's stripe, a prominent feature in the basal two-thirds of the TM in WT mice, is absent. CEACAM16 is also shown to interact with TECTB, indicating that it may stabilize interactions between TECTA and TECTB. Although brain-stem evoked responses and distortion product otoacoustic emissions are, for most frequencies, normal in young mice lacking CEACAM16, stimulus-frequency and transiently evoked emissions are larger. We also observed spontaneous otoacoustic emissions (SOAEs) in 70% of the homozygous mice. This incidence is remarkable considering that <3% of WT controls have SOAEs. The predominance of SOAEs >15 kHz correlates with the loss of Hensen's stripe. Results from mice lacking CEACAM16 are consistent with the idea that the organ of Corti evolved to maximize the gain of the cochlear amplifier while preventing large oscillations. Changes in TM structure appear to influence the balance between energy generation and dissipation such that the system becomes unstable.


Asunto(s)
Moléculas de Adhesión Celular/deficiencia , Proteínas de la Matriz Extracelular/metabolismo , Órgano Espiral/citología , Emisiones Otoacústicas Espontáneas/fisiología , Membrana Tectoria/fisiología , Estimulación Acústica , Animales , Moléculas de Adhesión Celular/genética , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Inmunoprecipitación , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Emisiones Otoacústicas Espontáneas/genética , Técnicas de Placa-Clamp , Membrana Tectoria/ultraestructura , beta-Galactosidasa/metabolismo
11.
J Biol Chem ; 288(4): 2452-63, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23212912

RESUMEN

Outer hair cells (OHCs) are a mammalian innovation for mechanically amplifying sound energy to overcome the viscous damping of the cochlear partition. Although the voltage-dependent OHC membrane motor, prestin, has been demonstrated to be essential for mammalian cochlear amplification, the molecular mechanism by which prestin converts electrical energy into mechanical displacement/force remains elusive. Identifying mutations that alter the motor function of prestin provides vital information for unraveling the energy transduction mechanism of prestin. We show that the V499G/Y501H mutation does not deprive prestin of its voltage-induced motor activity, but it does significantly impair the fast motor kinetics and voltage operating range. Furthermore, mutagenesis studies suggest that Val-499 is the primary site responsible for these changes. We also show that V499G/Y501H prestin forms heteromers with wild-type prestin and that the fast motor kinetics of wild-type prestin is not affected by heteromer formation with V499G/Y501H prestin. These results suggest that prestin subunits are individually functional within a given multimer.


Asunto(s)
Proteínas de Transporte de Anión/genética , Células Ciliadas Auditivas Externas/metabolismo , Proteínas Motoras Moleculares/genética , Mutación , Algoritmos , Secuencia de Aminoácidos , Animales , Electrofisiología/métodos , Células HEK293 , Humanos , Inmunoprecipitación , Cinética , Ratones , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Transportadores de Sulfato , Tirosina/química , Valina/química
12.
Proc Natl Acad Sci U S A ; 108(10): 4218-23, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368133

RESUMEN

We report on a secreted protein found in mammalian cochlear outer hair cells (OHC) that is a member of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family of adhesion proteins. Ceacam16 mRNA is expressed in OHC, and its protein product localizes to the tips of the tallest stereocilia and the tectorial membrane (TM). This specific localization suggests a role in maintaining the integrity of the TM as well as in the connection between the OHC stereocilia and TM, a linkage essential for mechanical amplification. In agreement with this role, CEACAM16 colocalizes and coimmunoprecipitates with the TM protein α-tectorin. In addition, we show that mutation of CEACAM16 leads to autosomal dominant nonsyndromic deafness (ADNSHL) at the autosomal dominant hearing loss (DFNA4) locus. In aggregate, these data identify CEACAM16 as an α-tectorin-interacting protein that concentrates at the point of attachment of the TM to the stereocilia and, when mutated, results in ADNSHL at the DFNA4 locus.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Genes Dominantes , Pérdida Auditiva/metabolismo , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Secuencia de Aminoácidos , Animales , Western Blotting , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Proteínas Ligadas a GPI/metabolismo , Pérdida Auditiva/genética , Humanos , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética
13.
bioRxiv ; 2024 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-38826260

RESUMEN

Prestin's voltage-driven motor activity confers sound-elicited somatic electromotility in auditory outer hair cells (OHCs) and is essential for the exquisite sensitivity and frequency selectivity of mammalian hearing. Lack of prestin results in hearing threshold shifts across frequency, supporting the causal association of variants in the prestin-coding gene, SLC26A5 , with human hearing loss, DFNB61. However, cochlear function can tolerate reductions in prestin-mediated OHC electromotility. We found that two deafness-associated prestin variants, p.A100T and p.P119S, do not deprive prestin of its fast motor function but significantly reduce membrane expression, leading to large reductions in OHC electromotility that were only ∼30% of wildtype (WT). Mice harboring these missense variants suffered congenital hearing loss that was worse at high frequencies; however, they retained WT-like auditory brainstem response thresholds at 8 kHz, which is processed at the apex of the mouse cochlea. This observation suggests the increasing importance of prestin-driven cochlear amplification at higher frequencies relevant to mammalian hearing. The observation also suggests the promising clinical possibility that small enhancements of OHC electromotility could significantly ameliorate DFNB61 hearing loss in human patients. SIGNIFICANCE: Prestin is abundantly expressed in the auditory outer hair cells and is essential for normal cochlear operation. Hence, reduction of prestin expression is often taken as indicative of reduced cochlear function in diseased or aged ears. However, this assumption overlooks the fact that cochlear function can tolerate large reductions in prestin motor activity. DFNB61 mouse models generated and characterized in this study provide an opportunity to gauge the amount of prestin motor activity needed to sustain normal hearing sensitivity. This knowledge is crucial not only for understanding the pathogenic roles of deafness-associated variants that impair OHC electromotility but also for unraveling how prestin contributes to cochlear amplification.

14.
J Assoc Res Otolaryngol ; 24(4): 413-428, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37464091

RESUMEN

Normal hearing is associated with cochlear nonlinearity. When two tones (f1 and f2) are presented, the intracochlear response contains additional components that can be recorded from the ear canal as distortion product otoacoustic emissions (DPOAEs). Although the most prominent intermodulation distortion component is at 2f1-f2, other cubic distortion products are also generated. Because these measurements are noninvasive, they are used in humans and in animal models to detect hearing loss. This study evaluated how loss of sensitivity affects DPOAEs with frequencies above and below the stimulating primaries, i.e., for upper sideband (USB) components like 2f2-f1 and for lower sideband (LSB) components like 2f1-f2. DPOAEs were recorded in several mouse mutants with varying degrees of hearing loss associated with structural changes to the tectorial membrane (TM), or with loss of outer hair cell (OHC) somatic electromotility due to lack of prestin or to the expression of a non-functional prestin. In mice with changes in sensitivity, magnitude reductions were observed for 2f1-f2 relative to controls with mice lacking prestin showing the greatest changes. In contrast, 2f2-f1 was minimally affected by reductions in cochlear gain due to changes in the TM or by the loss of OHC somatic electromotility. In addition, TM mutants with spontaneous otoacoustic emissions (SOAEs) generated larger responses than controls at 2f2-f1 when its frequency was similar to that for the SOAEs. Although cochlear pathologies appear to affect USB and LSB DPOAEs in different ways, both 2f1-f2 and 2f2-f1 reflect nonlinearities associated with the transducer channels. However, in mice, the component at 2f2-f1 does not appear to receive enhancement due to prestin's motor action.


Asunto(s)
Sordera , Pérdida Auditiva , Humanos , Animales , Ratones , Emisiones Otoacústicas Espontáneas/fisiología , Cóclea/fisiología , Células Ciliadas Auditivas Externas , Estimulación Acústica
15.
bioRxiv ; 2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37662362

RESUMEN

DFNB61 is a recessively inherited nonsyndromic hearing loss caused by mutations in SLC26A5 , the gene that encodes the voltage-driven motor protein, prestin. Prestin is abundantly expressed in the auditory outer hair cells that mediate cochlear amplification. Two DFNB61-associated SLC26A5 variants, p.W70X and p.R130S, were identified in patients who are compound heterozygous for these nonsense and missense changes ( SLC26A5 W70X/R130S ). Our recent study showed that mice homozygous for p.R130S ( Slc26a5 R130S/R130S ) suffer from hearing loss that is ascribed to significantly reduced motor kinetics of prestin. Given that W70X-prestin is nonfunctional, compound heterozygous Slc26a5 R130S/- mice were used as a model for human SLC26A5 W70X/R130S . By examining the pathophysiological consequences of p.R130S prestin when it is the sole allele for prestin protein production, we determined that this missense change results in progressive outer hair cell loss in addition to its effects on prestin's motor action. Thus, this study fully defines the pathogenic roles for the p.R130S prestin, which points to the presence of a limited time window for potential clinical intervention.

16.
Hear Res ; 423: 108376, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34848118

RESUMEN

Prestin (SLC26A5) is a membrane-based voltage-dependent motor protein responsible for outer hair cell (OHC) somatic electromotility. Its importance for mammalian cochlear amplification has been demonstrated using mouse models lacking prestin (prestin-KO) and expressing dysfunctional prestin, prestinV499G/Y501H (499-prestin-KI). However, it is still not elucidated how prestin contributes to the mechanical amplification process in the cochlea. In this study, we characterized several prestin mouse models in which prestin activity in OHCs was variously manipulated. We found that near-normal cochlear function can be maintained even when prestin activity is significantly reduced, suggesting that the relationship between OHC electromotility and the peripheral sensitivity to sound may not be linear. This result is counterintuitive given the large threshold shifts in prestin-KO and 499-prestin-KI mice, as reported in previous studies. To reconcile these apparently opposing observations, we entertain a voltage- and turgor pressure-based cochlear amplification mechanism that requires prestin but is insensitive to significant reductions in prestin protein expression. This article is part of the Special Issue Outer hair cell Edited by Joseph Santos-Sacchi and Kumar Navaratnam.


Asunto(s)
Células Ciliadas Auditivas Externas , Proteínas Motoras Moleculares , Animales , Cóclea/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Audición , Mamíferos/metabolismo , Ratones , Proteínas Motoras Moleculares/metabolismo , Actividad Motora
17.
Hear Res ; 423: 108428, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34987016

RESUMEN

Outer hair cells (OHCs) are innervated by both medial olivocochlear (MOC) efferents and type II afferents, which also innervate supporting cells to form a local neural network. It has also been demonstrated that prestin provides the molecular basis for OHC somatic electromotility, amplifying movements within the organ of Corti. Although not anticipated, early-onset OHC loss was found in two prestin transgenic mouse models that either lack prestin protein or lack electromotility. To uncover the molecular pathways that evoke OHC death, we profiled the coding transcriptome of OHCs from wildtype (WT), prestin-knockout (KO), and 499-knockin (KI) mice using single-cell RNA sequencing (scRNA-seq). scRNA-Seq transcriptomics and pathway analyses did not reveal common pathways associated with OHC loss observed in prestin-KO and 499-KI mice. Clustering enrichment analysis showed that increased gene expression in OHCs from prestin-KO mice was associated with lipid metabolic processes and cell death pathways. These mRNA profiles likely contribute to the OHC loss observed in prestin-KO mice and support the notion that prestin is also a structural protein, important for the normal plasma membrane compartmentalization that is essential to establish MOC efferent synapses. In contrast, the mRNA profile of OHCs from 499-KI mice did not provide a rational explanation of the early-onset OHC loss in this mutant. OHCs from 499-KI mice have normal plasma membrane compartmentalization and normal OHC-MOC contacts. However, 499 prestin lacks electromotility and appears to change the local neural network around OHCs, as more synaptic markers were found near neighboring supporting cells when compared to WT and prestin-KO mice. Thus, OHCs in prestin-KOs (no prestin protein, no electromotility) and 499-KIs (prestin protein present, no electromotility) may influence local neuronal networks in different ways. Collectively, our data suggest that prestin and its motile properties are important for OHC survival and the maintenance of local afferent/efferent circuits, as well as for its role in cochlear amplification. This article is part of the Special Issue Outer hair cell Edited by Joseph Santos-Sacchi and Kumar Navaratnam.


Asunto(s)
Células Ciliadas Auditivas Externas , Proteínas Motoras Moleculares , Animales , Cóclea/metabolismo , Células Ciliadas Auditivas Externas/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , ARN Mensajero/metabolismo
18.
Biochim Biophys Acta ; 1803(7): 796-804, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20359505

RESUMEN

Prestin is the motor protein responsible for cochlear outer hair cell (OHC) somatic electromotility. Eliminating this abundant basolateral membrane protein not only causes loss of frequency selectivity and hearing sensitivity, but also leads to OHC death. A membrane-based yeast two-hybrid approach was used to screen an OHC-enriched cDNA (complementary Deoxyribonucleic Acid) library in order to identify prestin-associated proteins. Several proteins were recognized as potential prestin partners, including vesicle-associated membrane protein associated protein A (VAPA or VAP-33). VAPA is an integral membrane protein that plays an important role in membrane trafficking, endoplasmic reticulum homeostasis, and the stress-signaling system. The connection between VAPA and prestin was confirmed through co-immunoprecipitation experiments. This new finding prompted the investigation of the interaction between VAPA and prestin in outer hair cells. By comparing VAPA expression between wild-type OHCs and OHCs derived from prestin-knockout mice, we found that VAPA is expressed in OHCs and the quantity of VAPA expressed is related to the presence of prestin. In other words, less VAPA protein is found in OHCs lacking prestin. Thus, prestin appears to modify the expression of VAPA protein in OHCs. Intriguingly, more prestin protein appears at the plasma membrane when VAPA is co-expressed with prestin. These data suggest that VAPA could be involved in prestin's transportation inside OHCs and may facilitate the targeting of this abundant OHC protein to the plasma membrane.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Motoras Moleculares/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Membrana Celular/metabolismo , Biblioteca de Genes , Células Ciliadas Auditivas Externas/citología , Células Ciliadas Auditivas Externas/fisiología , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas Motoras Moleculares/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Técnicas del Sistema de Dos Híbridos , Proteínas de Transporte Vesicular
19.
Hear Res ; 409: 108314, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34332206

RESUMEN

Cochlear function depends on the operation of a coupled feedback loop, incorporating outer hair cells (OHCs), and structured to assure that inner hair cells (IHCs) convey frequency specific acoustic information to the brain, even at very low sound levels. Although our knowledge of OHC function and its contribution to cochlear amplification has expanded, the importance of the tectorial membrane (TM) to the processing of mechanical inputs has not been fully elucidated. In addition, there are a surprising number of genetic mutations that affect TM structure and that produce hearing loss in humans. By synthesizing old and new results obtained on several mouse mutants, we learned that animals with abnormal TMs are prone to generate spontaneous otoacoustic emissions (SOAE), which are uncommon in most wildtype laboratory animals. Because SOAEs are not produced in TM mutants or in humans when threshold shifts exceed approximately 25 dB, some degree of cochlear amplification is required. However, amplification by itself is not sufficient because normal mice are rarely spontaneous emitters. Since SOAEs reflect active cochlear operation, TM mutants are valuable for studying the oscillatory nature of the amplification process and the structures associated with its stabilization. Inasmuch as the mouse models were selected to mirror human auditory disorders, using SOAEs as a noninvasive clinical tool may assist the classification of individuals with genetic defects that influence the active mechanisms responsible for sensitivity and frequency selectivity, the hallmarks of mammalian hearing.


Asunto(s)
Emisiones Otoacústicas Espontáneas , Membrana Tectoria , Estimulación Acústica , Animales , Biomarcadores , Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Externas , Ratones
20.
Hear Res ; 400: 108143, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33340968

RESUMEN

The global standing-wave model for generation of spontaneous otoacoustic emissions (SOAEs) suggests that they are amplitude-stabilized standing waves and that the spacing between SOAEs corresponds to the interval over which the phase changes by one cycle as determined from the phase-gradient delays of stimulus frequency otoacoustic emissions (SFOAEs). Because data characterizing the relationship between spontaneous and evoked emissions in nonhuman mammals are limited, we examined SOAEs and SFOAEs in tectorial membrane (TM) mutants and their controls. Computations indicate that the spacing between adjacent SOAEs is predicted by the SFOAE phase-gradient delays for TM mutants lacking Ceacam16, where SOAE frequencies are greater than ~20 kHz and the mutants retain near-normal hearing when young. Mice with a missense mutation in Tecta (TectaY1870C/+), as well as mice lacking Otoancorin (Otoa-/-), were also examined. Although these mutants exhibit hearing loss, they generate SOAEs with average frequencies of 11 kHz in TectaY1870C/+ and 6 kHz in Otoa-/-. In these animals, the spacing between adjacent SOAEs is larger than predicted by the SFOAE phase delays. It is also demonstrated that mice do not exhibit the strong frequency-dependence in signal coding that characterizes species with good low-frequency hearing. In fact, a transition occurs near the apical end of the mouse cochlea rather than at the mid-point along the cochlear partition. Hence, disagreements with the standing-wave model are not easily explained by a transition in tuning ratios between apical and basal regions of the cochlea, especially for SOAEs generated in TectaY1870C/+mice.


Asunto(s)
Membrana Tectoria , Animales , Sordera , Audición , Ratones , Mutación Missense , Emisiones Otoacústicas Espontáneas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA