106 results on '"Mary Ann Cheatham"'
Search Results
2. The susceptibility of cochlear outer hair cells to cyclodextrin is not related to their electromotile activity
- Author
-
Yingjie Zhou, Satoe Takahashi, Kazuaki Homma, Chongwen Duan, Jason Zheng, Mary Ann Cheatham, and Jing Zheng
- Subjects
Prestin ,Niemann-pick type C1 ,HPβCD ,Cholesterol ,Salicylate ,Electromotility ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Niemann-Pick Type C1 (NPC1) disease is a fatal neurovisceral disorder caused by dysfunction of NPC1 protein, which plays a role in intracellular cholesterol trafficking. The cholesterol-chelating agent, 2-hydroxypropyl-β-cyclodextrin (HPβCD), is currently undergoing clinical trials for treatment of this disease. Though promising in alleviating neurological symptoms, HPβCD causes irreversible hearing loss in NPC1 patients and outer hair cell (OHC) death in animal models. We recently found that HPβCD-induced OHC death can be significantly alleviated in a mouse model lacking prestin, an OHC-specific motor protein required for the high sensitivity and sharp frequency selectivity of mammalian hearing. Since cholesterol status is known to influence prestin’s electromotility, we examined how prestin contributes to HPβCD-induced OHC death in the disease context using the NPC1 knockout (KO) mouse model (NPC1-KO). We found normal expression and localization of prestin in NPC1-KO OHCs. Whole-cell patch-clamp recordings revealed a significant depolarization of the voltage-operating point of prestin in NPC1-KO mice, suggesting reduced levels of cholesterol in the lateral membrane of OHCs that lack NPC1. OHC loss and elevated thresholds were found for high frequency regions in NPC1-KO mice, whose OHCs retained their sensitivity to HPβCD. To investigate whether prestin’s electromotile function contributes to HPβCD-induced OHC death, the prestin inhibitor salicylate was co-administered with HPβCD to WT and NPC1-KO mice. Neither oral nor intraperitoneal administration of salicylate mitigated HPβCD-induced OHC loss. To further determine the contribution of prestin’s electromotile function, a mouse model expressing a virtually nonelectromotile prestin protein (499-prestin) was subjected to HPβCD treatment. 499-prestin knockin mice showed no resistance to HPβCD-induced OHC loss. As 499-prestin maintains its ability to bind cholesterol, our data imply that HPβCD-induced OHC death is ascribed to the structural role of prestin in maintaining the OHC’s lateral membrane, rather than its motor function.
- Published
- 2018
- Full Text
- View/download PDF
3. Accelerated Age-Related Degradation of the Tectorial Membrane in the Ceacam16βgal/βgal Null Mutant Mouse, a Model for Late-Onset Human Hereditary Deafness DFNB113
- Author
-
Richard J. Goodyear, Mary Ann Cheatham, Souvik Naskar, Yingjie Zhou, Richard T. Osgood, Jing Zheng, and Guy P. Richardson
- Subjects
cochlea ,hearing ,deafness ,tectorial membrane ,CEACAM16 ,spontaneous otoacoustic emissions ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
CEACAM16 is a non-collagenous protein of the tectorial membrane, an extracellular structure of the cochlea essential for normal hearing. Dominant and recessive mutations in CEACAM16 have been reported to cause postlingual and progressive forms of deafness in humans. In a previous study of young Ceacam16βgal/βgal null mutant mice on a C57Bl/6J background, the incidence of spontaneous otoacoustic emissions (SOAEs) was greatly increased relative to Ceacam16+/+ and Ceacam16+/βgal mice, but auditory brain-stem responses (ABRs) and distortion product otoacoustic emissions (DPOAEs) were near normal, indicating auditory thresholds were not significantly affected. To determine if the loss of CEACAM16 leads to hearing loss at later ages in this mouse line, cochlear structure and auditory function were examined in Ceacam16+/+, Ceacam16+/βgal and Ceacam16βgal/βgal mice at 6 and 12 months of age and compared to that previously described at 1 month. Analysis of older Ceacam16βgal/βgal mice reveals a progressive loss of matrix from the core of the tectorial membrane that is more extensive in the apical, low-frequency regions of the cochlea. In Ceacam16βgal/βgal mice at 6–7 months, the DPOAE magnitude at 2f1-f2 and the incidence of SOAEs both decrease relative to young animals. By ∼12 months, SOAEs and DPOAEs are not detected in Ceacam16βgal/βgal mice and ABR thresholds are increased by up to ∼40 dB across frequency, despite a complement of hair cells similar to that present in Ceacam16+/+ mice. Although SOAE incidence decreases with age in Ceacam16βgal/βgal mice, it increases in aging heterozygous Ceacam16+/βgal mice and is accompanied by a reduction in the accumulation of CEACAM16 in the tectorial membrane relative to controls. An apically-biased loss of matrix from the core of the tectorial membrane, similar to that observed in young Ceacam16βgal/βgal mice, is also seen in Ceacam16+/+ and Ceacam16+/βgal mice, and other strains of wild-type mice, but at much later ages. The loss of Ceacam16 therefore accelerates age-related degeneration of the tectorial membrane leading, as in humans with mutations in CEACAM16, to a late-onset progressive form of hearing loss.
- Published
- 2019
- Full Text
- View/download PDF
4. Prestin Contributes to Membrane Compartmentalization and Is Required for Normal Innervation of Outer Hair Cells
- Author
-
Satoe Takahashi, Willy Sun, Yingjie Zhou, Kazuaki Homma, Bechara Kachar, Mary Ann Cheatham, and Jing Zheng
- Subjects
prestin ,outer hair cells ,efferent innervation ,PMCA2 ,KCNQ4 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Outer hair cells (OHC) act as amplifiers and their function is modified by medial olivocochlear (MOC) efferents. The unique OHC motor protein, prestin, provides the molecular basis for somatic electromotility, which is required for sensitivity and frequency selectivity, the hallmarks of mammalian hearing. Prestin proteins are the major component of the lateral membrane of mature OHCs, which separates apical and basal domains. To investigate the contribution of prestin to this unique arrangement, we compared the distribution of membrane proteins in OHCs of wildtype (WT) and prestin-knockout (KO) mice. In WT, the apical protein PMCA2 was exclusively localized to the hair bundles, while it was also found at the lateral membrane in KOs. Similarly, a basal protein KCNQ4 did not coalesce at the base of OHCs but was widely dispersed in mice lacking prestin. Since the expression levels of PMCA2 and KCNQ4 remained unchanged in KOs, the data indicate that prestin is required for the normal distribution of apical and basal membrane proteins in OHCs. Since OHC synapses predominate in the basal subnuclear region, we also examined the synaptic architecture in prestin-KO mice. Although neurite densities were not affected, MOC efferent terminals in prestin-KO mice were no longer constrained to the basal pole as in WT. This trend was evident as early as at postnatal day 12. Furthermore, terminals were often enlarged and frequently appeared as singlets when compared to the multiple clusters of individual terminals in WT. This abnormality in MOC synaptic morphology in prestin-KO mice is similar to defects in mice lacking MOC pathway proteins such as α9/α10 nicotinic acetylcholine receptors and BK channels, indicating a role for prestin in the proper establishment of MOC synapses. To investigate the contribution of prestin’s electromotility, we also examined OHCs from a mouse model that expresses non-functional prestin (499-prestin). We found no changes in PMCA2 localization and MOC synaptic morphology in OHCs from 499-prestin mice. Taken together, these results indicate that prestin, independent of its motile function, plays an important structural role in membrane compartmentalization, which is required for the formation of normal efferent-OHC synapses in mature OHCs.
- Published
- 2018
- Full Text
- View/download PDF
5. Marshalin, a microtubule minus-end binding protein, regulates cytoskeletal structure in the organ of Corti
- Author
-
Jing Zheng, David Furness, Chongwen Duan, Katharine K. Miller, Roxanne M. Edge, Jessie Chen, Kazuaki Homma, Carole M. Hackney, Peter Dallos, and Mary Ann Cheatham
- Subjects
Microtubule minus-end binding protein ,Noncentrosomal MTOC ,Cochlea ,CAMSAP3 ,Nezha ,Patronin ,Science ,Biology (General) ,QH301-705.5 - Abstract
Summary Dramatic structural changes in microtubules (MT) and the assembly of complicated intercellular connections are seen during the development of the cellular matrix of the sense organ for hearing, the organ of Corti. This report examines the expression of marshalin, a minus-end binding protein, during this process of cochlear development. We discovered that marshalin is abundantly expressed in both sensory hair cells and supporting cells. In the adult, prominent marshalin expression is observed in the cuticular plates of hair cells and in the noncentrosomal MT organization centers (MTOC) of Deiters' and pillar cells. Based upon differences in marshalin expression patterns seen in the organ of Corti, we identified eight isoforms ranging from 863 to 1280 amino acids. mRNAs/proteins associated with marshalin's isoforms are detected at different times during development. These isoforms carry various protein–protein interacting domains, including coiled-coil (CC), calponin homology (CH), proline-rich (PR), and MT-binding domains, referred to as CKK. We, therefore, examined membranous organelles and structural changes in the cytoskeleton induced by expressing two of these marshalin isoforms in vitro. Long forms containing CC and PR domains induce thick, spindle-shaped bundles, whereas short isoforms lacking CC and PR induce more slender variants that develop into densely woven networks. Together, these data suggest that marshalin is closely associated with noncentrosomal MTOCs, and may be involved in MT bundle formation in supporting cells. As a scaffolding protein with multiple isoforms, marshalin is capable of modifying cytoskeletal networks, and consequently organelle positioning, through interactions with various protein partners present in different cells.
- Published
- 2013
- Full Text
- View/download PDF
6. Prestin-Dependence of Outer Hair Cell Survival and Partial Rescue of Outer Hair Cell Loss in PrestinV499G/Y501H Knockin Mice.
- Author
-
Mary Ann Cheatham, Roxanne M Edge, Kazuaki Homma, Emily L Leserman, Peter Dallos, and Jing Zheng
- Subjects
Medicine ,Science - Abstract
A knockin (KI) mouse expressing mutated prestinV499G/Y501H (499 prestin) was created to study cochlear amplification. Recordings from isolated outer hair cells (OHC) in this mutant showed vastly reduced electromotility and, as a consequence, reduced hearing sensitivity. Although 499 prestin OHCs were normal in stiffness and longer than OHCs lacking prestin, accelerated OHC death was unexpectedly observed relative to that documented in prestin knockout (KO) mice. These observations imply an additional role of prestin in OHC maintenance besides its known requirement for mammalian cochlear amplification. In order to gain mechanistic insights into prestin-associated OHC loss, we implemented several interventions to improve survival. First, 499 prestin KI's were backcrossed to Bak KO mice, which lack the mitochondrial pro-apoptotic gene Bak. Because oxidative stress is implicated in OHC death, another group of 499 prestin KI mice was fed the antioxidant diet, Protandim. 499 KI mice were also backcrossed onto the FVB murine strain, which retains excellent high-frequency hearing well into adulthood, to reduce the compounding effect of age-related hearing loss associated with the original 499 prestin KIs. Finally, a compound heterozygous (chet) mouse expressing one copy of 499 prestin and one copy of KO prestin was also created to reduce quantities of 499 prestin protein. Results show reduction in OHC death in chets, and in 499 prestin KIs on the FVB background, but only a slight improvement in OHC survival for mice receiving Protandim. We also report that improved OHC survival in 499 prestin KIs had little effect on hearing phenotype, reaffirming the original contention about the essential role of prestin's motor function in cochlear amplification.
- Published
- 2015
- Full Text
- View/download PDF
7. Prestin’s fast motor kinetics is essential for mammalian cochlear amplification
- Author
-
Satoe Takahashi, Yingjie Zhou, Takashi Kojima, Mary Ann Cheatham, and Kazuaki Homma
- Subjects
Multidisciplinary - Abstract
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.
- Published
- 2023
8. Tbx2 is a master regulator of inner versus outer hair cell differentiation
- Author
-
Jaime García-Añoveros, John C. Clancy, Chuan Zhi Foo, Ignacio García-Gómez, Yingjie Zhou, Kazuaki Homma, Mary Ann Cheatham, and Anne Duggan
- Subjects
Hair Cells, Auditory, Outer ,Mice ,Multidisciplinary ,Hair Cells, Auditory, Inner ,Animals ,Cell Differentiation ,T-Box Domain Proteins ,Article ,Cochlea - Abstract
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 inputs(1). 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 IHCs(2–4). One such transcription factor for OHCs, INSM1, acts during a crucial embryonic period to consolidate the OHC fate, preventing OHCs from transdifferentiating into IHCs(2). 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.
- Published
- 2022
9. CAMSAP3 facilitates basal body polarity and the formation of the central pair of microtubules in motile cilia
- Author
-
Satoe Takahashi, Eva J. Brotslaw, Mary Ann Cheatham, Daniele Procissi, Brian J. Mitchell, Jing Zheng, Alan M. Robinson, Claus Peter Richter, Aisha Ahmad, and Emma Ferrer
- Subjects
Axoneme ,Morpholino ,Xenopus ,primary ciliary dyskinesia ,Motility ,central MT pair ,Biology ,Microtubules ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Microtubule ,CAMSAP3 ,medicine ,Animals ,Humans ,Basal body ,Cilia ,motile cilia ,030304 developmental biology ,Primary ciliary dyskinesia ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,basal body orientation ,Cilium ,Cell Polarity ,Epithelial Cells ,Biological Sciences ,medicine.disease ,Basal Bodies ,Cell biology ,PNAS Plus ,Motile cilium ,Microtubule-Associated Proteins ,Ciliary base ,030217 neurology & neurosurgery ,Developmental Biology ,Ciliary Motility Disorders - Abstract
Significance Cilia are composed of hundreds of proteins whose identities and functions are far from being completely understood. In this study, we determined that calmodulin-regulated spectrin-associated protein 3 (CAMSAP3) plays an important role for the function of motile cilia in multiciliated cells (MCCs). Global knockdown of CAMSAP3 protein expression in mice resulted in defects in ciliary structures, polarity, and synchronized beating in MCCs. These animals also displayed signs and symptoms reminiscent of primary ciliary dyskinesia (PCD), including a mild form of hydrocephalus, subfertility, and impaired mucociliary clearance that leads to hyposmia, anosmia, rhinosinusitis, and otitis media. Functional characterization of CAMSAP3 enriches our understanding of the molecular mechanisms underlying the generation and function of motile cilia in MCCs., 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.
- Published
- 2020
10. Hair Cells: From Molecules to Function, Volume II
- Author
-
Brad Walters, David Z. He, Mary Ann Cheatham, and Bernd Fritzsch
- Published
- 2022
11. Prestin and electromotility may serve multiple roles in cochlear outer hair cells
- Author
-
Jing Zheng, Satoe Takahashi, Yingjie Zhou, and Mary Ann Cheatham
- Subjects
Mice, Knockout ,Hair Cells, Auditory, Outer ,Mice ,Molecular Motor Proteins ,Animals ,Mice, Transgenic ,RNA, Messenger ,Sensory Systems ,Cochlea - Abstract
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.
- Published
- 2021
12. Spontaneous otoacoustic emissions are biomarkers for mice with tectorial membrane defects
- Author
-
Mary Ann Cheatham
- Subjects
Spontaneous Otoacoustic Emissions ,Frequency selectivity ,Hair Cells, Auditory, Inner ,Tectorial Membrane ,Hearing loss ,Tectorial membrane ,Otoacoustic Emissions, Spontaneous ,Wild type ,Biology ,Sensory Systems ,Article ,Hair Cells, Auditory, Outer ,Mice ,medicine.anatomical_structure ,Acoustic Stimulation ,otorhinolaryngologic diseases ,medicine ,Animals ,sense organs ,medicine.symptom ,TECTA ,Outer hair cells ,Neuroscience ,Cochlea ,Biomarkers - Abstract
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.
- Published
- 2021
13. How much prestin motor activity is required for normal hearing?
- Author
-
Kazuaki Homma, Mary Ann Cheatham, and Satoe Takahashi
- Subjects
Mammals ,biology ,Chemistry ,Molecular Motor Proteins ,Motor Activity ,Sensory Systems ,Cochlear function ,Protein expression ,Cell biology ,Cochlea ,Motor protein ,Hair Cells, Auditory, Outer ,Mice ,medicine.anatomical_structure ,Hearing ,medicine ,biology.protein ,Animals ,sense organs ,Hair cell ,Motor activity ,Prestin activity ,Prestin - Abstract
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.
- Published
- 2021
14. Age-related degradation of tectorial membrane dynamics with loss of CEACAM16
- Author
-
Dennis M. Freeman, Daniel Filizzola, Mary Ann Cheatham, Amer Mansour, Jonathan B. Sellon, and Roozbeh Ghaffari
- Subjects
Extracellular Matrix Proteins ,Tectorial Membrane ,Chemistry ,Tectorial membrane ,Viscosity ,Biophysics ,Age Factors ,Progressive hearing loss ,medicine.disease ,Phenotype ,Mice ,medicine.anatomical_structure ,Hearing ,Age related ,medicine ,Animals ,Sensorineural hearing loss ,TECTA ,Hearing Loss ,Auditory thresholds ,Cell Adhesion Molecules - Abstract
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. While 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.
- Published
- 2021
15. Comparing spontaneous and stimulus frequency otoacoustic emissions in mice with tectorial membrane defects
- Author
-
Mary Ann Cheatham
- Subjects
0301 basic medicine ,Tectorial Membrane ,Tectorial membrane ,Hearing loss ,Otoacoustic Emissions, Spontaneous ,Mutation, Missense ,Mouse Cochlea ,Deafness ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Hearing ,medicine ,otorhinolaryngologic diseases ,Animals ,TECTA ,Cochlea ,Otoancorin ,Spontaneous Otoacoustic Emissions ,Chemistry ,Sensory Systems ,030104 developmental biology ,medicine.anatomical_structure ,Biophysics ,Stimulus frequency ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
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.
- Published
- 2020
16. Trans-differentiation of outer hair cells into inner hair cells in the absence of INSM1
- Author
-
Freddie Márquez, Jorge A. Cantu, Teerawat Wiwatpanit, Chuan Zhi Foo, Matthew J. Schipma, Anne Duggan, Sarah M. Lorenzen, Ann K. Hogan, Jaime García-Añoveros, Mary Ann Cheatham, and John C. Clancy
- Subjects
Male ,0301 basic medicine ,Cell ,Biology ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,otorhinolaryngologic diseases ,medicine ,Animals ,Cochlea ,Regulation of gene expression ,Hair Cells, Auditory, Inner ,Multidisciplinary ,Transdifferentiation ,Gene Expression Regulation, Developmental ,Embryo, Mammalian ,Embryonic stem cell ,Up-Regulation ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Repressor Proteins ,Hair Cells, Auditory, Outer ,030104 developmental biology ,medicine.anatomical_structure ,Organ Specificity ,Cell Transdifferentiation ,Female ,sense organs ,Hair cell ,Transcriptome ,Homeotic gene ,Transcription Factors - Abstract
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. Conditional deletion of Insm1 in mice demonstrates that INSM1 is the key switch that causes the maturation of outer hair cells in the cochlea, with its absence resulting in an increase in inner hair cells instead.
- Published
- 2018
17. The susceptibility of cochlear outer hair cells to cyclodextrin is not related to their electromotile activity
- Author
-
Jing Zheng, Mary Ann Cheatham, Satoe Takahashi, Jason Zheng, Chongwen Duan, Yingjie Zhou, and Kazuaki Homma
- Subjects
0301 basic medicine ,Patch-Clamp Techniques ,lcsh:RC346-429 ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Prestin ,Cell Line, Transformed ,Mice, Knockout ,Mice, Inbred BALB C ,biology ,Chemistry ,Molecular Motor Proteins ,Anti-Inflammatory Agents, Non-Steroidal ,Age Factors ,Intracellular Signaling Peptides and Proteins ,Depolarization ,Niemann-Pick Disease, Type C ,Salicylates ,Cell biology ,2-Hydroxypropyl-beta-cyclodextrin ,medicine.anatomical_structure ,Cholesterol ,HPβCD ,Electromotility ,Hair cell ,medicine.symptom ,Hearing loss ,Green Fluorescent Proteins ,Context (language use) ,Transfection ,Niemann-pick type C1 ,Pathology and Forensic Medicine ,Motor protein ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Niemann-Pick C1 Protein ,medicine ,Evoked Potentials, Auditory, Brain Stem ,Animals ,lcsh:Neurology. Diseases of the nervous system ,Salicylate ,Proteins ,Mice, Inbred C57BL ,Disease Models, Animal ,Hair Cells, Auditory, Outer ,030104 developmental biology ,Gene Expression Regulation ,Mutation ,biology.protein ,Neurology (clinical) ,sense organs ,NPC1 ,030217 neurology & neurosurgery - Abstract
Niemann-Pick Type C1 (NPC1) disease is a fatal neurovisceral disorder caused by dysfunction of NPC1 protein, which plays a role in intracellular cholesterol trafficking. The cholesterol-chelating agent, 2-hydroxypropyl-β-cyclodextrin (HPβCD), is currently undergoing clinical trials for treatment of this disease. Though promising in alleviating neurological symptoms, HPβCD causes irreversible hearing loss in NPC1 patients and outer hair cell (OHC) death in animal models. We recently found that HPβCD-induced OHC death can be significantly alleviated in a mouse model lacking prestin, an OHC-specific motor protein required for the high sensitivity and sharp frequency selectivity of mammalian hearing. Since cholesterol status is known to influence prestin’s electromotility, we examined how prestin contributes to HPβCD-induced OHC death in the disease context using the NPC1 knockout (KO) mouse model (NPC1-KO). We found normal expression and localization of prestin in NPC1-KO OHCs. Whole-cell patch-clamp recordings revealed a significant depolarization of the voltage-operating point of prestin in NPC1-KO mice, suggesting reduced levels of cholesterol in the lateral membrane of OHCs that lack NPC1. OHC loss and elevated thresholds were found for high frequency regions in NPC1-KO mice, whose OHCs retained their sensitivity to HPβCD. To investigate whether prestin’s electromotile function contributes to HPβCD-induced OHC death, the prestin inhibitor salicylate was co-administered with HPβCD to WT and NPC1-KO mice. Neither oral nor intraperitoneal administration of salicylate mitigated HPβCD-induced OHC loss. To further determine the contribution of prestin’s electromotile function, a mouse model expressing a virtually nonelectromotile prestin protein (499-prestin) was subjected to HPβCD treatment. 499-prestin knockin mice showed no resistance to HPβCD-induced OHC loss. As 499-prestin maintains its ability to bind cholesterol, our data imply that HPβCD-induced OHC death is ascribed to the structural role of prestin in maintaining the OHC’s lateral membrane, rather than its motor function.
- Published
- 2018
18. Codeficiency of Lysosomal Mucolipins 3 and 1 in Cochlear Hair Cells Diminishes Outer Hair Cell Longevity and Accelerates Age-Related Hearing Loss
- Author
-
Aisha Ahmad, Natalie N. Remis, Jaime García-Añoveros, Mary Ann Cheatham, Yingjie Zhou, John C. Clancy, and Teerawat Wiwatpanit
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Cell type ,TRPML ,Hearing loss ,Presbycusis ,Degeneration (medical) ,Biology ,Mice ,03 medical and health sciences ,Transient Receptor Potential Channels ,Lysosome ,Internal medicine ,Hair Cells, Auditory ,otorhinolaryngologic diseases ,medicine ,Animals ,Research Articles ,General Neuroscience ,medicine.disease ,Organelle membrane ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Female ,sense organs ,Hair cell ,medicine.symptom ,Lysosomes ,Gene Deletion - Abstract
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 STATEMENTPresbycusis, 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.
- Published
- 2018
19. Identifying the Origin of Effects of Contralateral Noise on Transient Evoked Otoacoustic Emissions in Unanesthetized Mice
- Author
-
Jonathan H. Siegel, Yingyue Xu, and Mary Ann Cheatham
- Subjects
Male ,medicine.medical_specialty ,Efferent ,Ear, Middle ,Stimulation ,Stimulus (physiology) ,Audiology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Hearing ,medicine ,Animals ,Auditory system ,030223 otorhinolaryngology ,Acoustic reflex ,Diagnostic Techniques, Otological ,Chemistry ,Vestibulocochlear Nerve ,Reflex, Acoustic ,Sensory Systems ,Peripheral ,medicine.anatomical_structure ,Otorhinolaryngology ,Middle ear ,Reflex ,Female ,sense organs ,Noise ,030217 neurology & neurosurgery ,Research Article - Abstract
Descending neural pathways in the mammalian auditory system are known to modulate the function of the peripheral auditory system. These pathways include the medial olivocochlear (MOC) efferent innervation to outer hair cells (OHCs) and the acoustic reflex pathways mediating middle ear muscle (MEM) contractions. Based on measurements in humans (Marks and Siegel, companion paper), we applied a sensitive method to attempt to differentiate MEM and MOC reflexes using contralateral acoustic stimulation in mice under different levels of anesthesia. Separation of these effects is based on the knowledge that OHC-generated transient evoked otoacoustic emissions (TEOAE) are delayed relative to the stimulus, and that the MOC reflex affects the emission through its innervation of OHC. In contrast, the MEM-mediated changes in middle ear reflectance alter both the stimulus (with a short delay) and the emission. Using this approach, time averages to transient stimuli were evaluated to determine if thresholds for a contralateral effect on the delayed emission, indicating potential MOC activation, could be observed in the absence of a change in the stimulus pressure. This outcome was not observed in the majority of cases. There were also no statistically significant differences between MEM and putative MOC thresholds, and variability was high for both thresholds regardless of anesthesia level. Since the two reflex pathways could not be differentiated on the basis of activation thresholds, it was concluded that the MEM reflex dominates changes in TEOAEs induced by contralateral noise. This result complicates the identification of purely MOC-induced changes on OAEs in mice unless the MEM reflex is inactivated surgically or pharmacologically.
- Published
- 2017
20. Deletion of exons 17 and 18 in prestin’s STAS domain results in loss of function
- Author
-
Satoe Takahashi, Kazuaki Homma, Jian Zuo, Yingjie Zhou, Jing Zheng, Mary Ann Cheatham, and Tetsuji Yamashita
- Subjects
Male ,0301 basic medicine ,Calmodulin ,Protein domain ,lcsh:Medicine ,Article ,Motor protein ,Mice ,03 medical and health sciences ,Exon ,Medical research ,0302 clinical medicine ,Protein Domains ,Animals ,Humans ,Gene Knock-In Techniques ,Binding site ,lcsh:Science ,Prestin ,Cochlea ,Sequence Deletion ,Multidisciplinary ,biology ,Chemistry ,lcsh:R ,Exons ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Sulfate Transporters ,RNA splicing ,biology.protein ,lcsh:Q ,Female ,sense organs ,030217 neurology & neurosurgery - Abstract
Cochlear outer hair cells (OHC) express the motor protein, prestin, which is required for sensitivity and frequency selectivity. Because our previous work showed that a calmodulin binding site (CBS) was located in prestin’s C-terminal, specifically within the intrinsically disordered region, we sought to delete the IDR to study the functional significance of calcium-dependent, calmodulin binding on OHC function. Although the construct lacking the IDR (∆IDR prestin) demonstrated wildtype-like nonlinear capacitance (NLC) in HEK293T cells, the phenotype in ∆IDR prestin knockins (KI) was similar to that in prestin knockouts: thresholds were elevated, NLC was absent and OHCs were missing from basal regions of the cochlea. Although ∆IDR prestin mRNA was measured, no prestin protein was detected. At the mRNA level, both of prestin’s exons 17 and 18 were entirely removed, rather than the smaller region encoding the IDR. Our hybrid exon that contained the targeted deletion (17–18 ∆IDR) failed to splice in vitro and prestin protein lacking exons 17 and 18 aggregated and failed to target the cell membrane. Hence, the absence of prestin protein in ∆IDR KI OHCs may be due to the unexpected splicing of the hybrid 17–18 ∆IDR exon followed by rapid degradation of nonfunctional prestin protein.
- Published
- 2019
21. Spontaneous otoacoustic emissions in TectaY1870C/+ mice reflect changes in cochlear amplification and how it is controlled by the tectorial membrane
- Author
-
Mary Ann Cheatham, Peter Dallos, Richard J. Goodyear, Yingjie Zhou, and Guy P. Richardson
- Subjects
Stereocilia (inner ear) ,QP0431 ,QP0351 ,Audiology ,Mice ,0302 clinical medicine ,TECTA ,Spontaneous Otoacoustic Emissions ,0303 health sciences ,Extracellular Matrix Proteins ,Chemistry ,General Neuroscience ,General Medicine ,QP0461 ,New Research ,cochlear amplifier ,harmonic distortion ,medicine.anatomical_structure ,Sensory and Motor Systems ,Hair cell ,spontaneous otoacoustic emissions ,medicine.symptom ,Psychoacoustics ,medicine.medical_specialty ,Cochlear amplifier ,outer hair cells ,Tectorial Membrane ,Tectorial membrane ,Hearing loss ,Otoacoustic Emissions, Spontaneous ,Mice, Transgenic ,GPI-Linked Proteins ,Statistics, Nonparametric ,03 medical and health sciences ,medicine ,Evoked Potentials, Auditory, Brain Stem ,otorhinolaryngologic diseases ,Animals ,Ear canal ,Cysteine ,030304 developmental biology ,Auditory Threshold ,QP ,Mice, Inbred C57BL ,Hair Cells, Auditory, Outer ,Acoustic Stimulation ,8.1 ,Mutation ,Tyrosine ,sense organs ,QP0448 ,030217 neurology & neurosurgery - Abstract
Spontaneous otoacoustic emissions (SOAEs) recorded from the ear canal in the absence of sound reflect cochlear amplification, an outer hair cell (OHC) process required for the extraordinary sensitivity and frequency selectivity of mammalian hearing. Although wild-type mice rarely emit, those with mutations that influence the tectorial membrane (TM) show an incidence of SOAEs similar to that in humans. In this report, we characterized mice with a missense mutation inTecta,a gene required for the formation of the striated-sheet matrix within the core of the TM. Mice heterozygous for the Y1870C mutation (TectaY1870C/+) are prolific emitters, despite a moderate hearing loss. Additionally, Kimura’s membrane, into which the OHC stereocilia insert, separates from the main body of the TM, except at apical cochlear locations. Multimodal SOAEs are also observed inTectaY1870C/+mice where energy is present at frequencies that are integer multiples of a lower-frequency SOAE (the primary). Second-harmonic SOAEs, at twice the frequency of a lower-frequency primary, are the most frequently observed. These secondary SOAEs are found in spatial regions where stimulus-evoked OAEs are small or in the noise floor. Introduction of high-level suppressors just above the primary SOAE frequency reduce or eliminate both primary and second-harmonic SOAEs. In contrast, second-harmonic SOAEs are not affected by suppressors, either above or below the second-harmonic SOAE frequency, even when they are much larger in amplitude. Hence, second-harmonic SOAEs do not appear to be spatially separated from their primaries, a finding that has implications for cochlear mechanics and the consequences of changes to TM structure.
- Published
- 2018
22. Author Correction: Trans-differentiation of outer hair cells into inner hair cells in the absence of INSM1
- Author
-
Ann K. Hogan, Jorge A. Cantu, Matthew J. Schipma, Teerawat Wiwatpanit, Anne Duggan, Mary Ann Cheatham, John C. Clancy, Jaime García-Añoveros, Freddie Márquez, Chuan Zhi Foo, and Sarah M. Lorenzen
- Subjects
Multidisciplinary ,Chemistry ,otorhinolaryngologic diseases ,Inner hair cells ,sense organs ,Outer hair cells ,Trans differentiation ,Actin ,Article ,Cell biology - Abstract
The mammalian cochlea contains two types of mechanosensory hair cells (HCs) that play different and critical roles in hearing. Inner hair cells (IHCs), with an elaborate presynaptic apparatus, signal to cochlear neurons and communicate sound information to the brain. Outer hair cells (OHCs) mechanically amplify sound-induced vibrations, enabling enhanced sensitivity to sound and sharp tuning. Cochlear HCs are solely generated during development and their death, most often of OHCs, is the main cause of deafness. OHCs and IHCs, together with supporting cells, originate embryonically from the prosensory region of the otocyst, but how HCs differentiate into two different types is unknown1–3. Here we show that Insm1, which encodes a zinc finger protein transiently expressed in nascent OHCs, consolidates their fate by preventing trans-differentiation into IHCs. In the absence of INSM1 many HCs born embryonically as OHCs switch fates to become mature IHCs. In order to identify the genetic mechanisms by which Insm1 operates, we compared transcriptomes of immature IHCs vs OHCs, as well as OHCs with and without INSM1. OHCs lacking INSM1 upregulate a set of genes, most of which are normally preferentially expressed by IHCs. The homeotic cell transformation of OHCs without INSM1 into IHCs reveals for the first time a mechanism by which these neighboring 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 upregulating these few IHC-enriched transcripts trans-differentiate into IHCs, revealing the first candidate genes for IHC-specific differentiation.
- Published
- 2018
23. The R130S mutation significantly affects the function of prestin, the outer hair cell motor protein
- Author
-
Mary Ann Cheatham, Satoe Takahashi, Jing Zheng, and Kazuaki Homma
- Subjects
Anions ,Models, Molecular ,0301 basic medicine ,Protein Conformation ,Hearing loss ,Anion Transport Proteins ,Gene Expression ,Article ,Cell Line ,Motor protein ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Gene expression ,medicine ,Humans ,Missense mutation ,Amino Acid Sequence ,Prestin ,Alleles ,Genetics (clinical) ,Genetics ,biology ,Biological Transport ,Electrophysiological Phenomena ,Cell biology ,Transport protein ,Hair Cells, Auditory, Outer ,Protein Transport ,030104 developmental biology ,medicine.anatomical_structure ,Amino Acid Substitution ,Sulfate Transporters ,Multigene Family ,Mutation ,biology.protein ,Molecular Medicine ,sense organs ,Heterologous expression ,Hair cell ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
A missense mutation, R130S, was recently found in the prestin gene, SLC26A5, of patients with moderate to severe hearing loss (DFNB61). In order to define the pathology of hearing loss associated with this missense mutation, a recombinant prestin construct harboring the R130S mutation (R130S-prestin) was generated, and its functional consequences examined in a heterologous expression system. We found that R130S-prestin targets the plasma membrane but less efficiently compared to wild-type. The voltage operating point and voltage sensitivity of the motor function of R130S-prestin were similar to wild-type prestin. However, the motor activity of R130S-prestin is greatly reduced at higher voltage stimulus frequencies, indicating a reduction in motor kinetics. Our study thus provides experimental evidence that supports a causal relationship between the R130S mutation in the prestin gene and hearing loss found in patients with this missense mutation.Membrane targeting of prestin is impaired by the R130S missense mutation. The fast motor kinetics of prestin is impaired by the R130S missense mutation. Our study strongly suggests that the prestin R130S missense mutation is pathogenic.
- Published
- 2016
24. The V499G/Y501H Mutation Impairs Fast Motor Kinetics of Prestin and Has Significance for Defining Functional Independence of Individual Prestin Subunits
- Author
-
Jing Zheng, Chongwen Duan, Peter Dallos, Mary Ann Cheatham, and Kazuaki Homma
- Subjects
Anion Transport Proteins ,Molecular Sequence Data ,Heteromer ,Biology ,medicine.disease_cause ,Biochemistry ,Mice ,Molecular motor ,medicine ,Animals ,Humans ,Immunoprecipitation ,Amino Acid Sequence ,Prestin ,Molecular Biology ,Mutation ,Molecular Motor Proteins ,HEK 293 cells ,Valine ,Cell Biology ,Protein Structure, Tertiary ,Transport protein ,Cell biology ,Electrophysiology ,Hair Cells, Auditory, Outer ,Kinetics ,HEK293 Cells ,medicine.anatomical_structure ,Membrane protein ,Sulfate Transporters ,biology.protein ,Tyrosine ,sense organs ,Hair cell ,Molecular Biophysics ,Algorithms ,Protein Binding - Abstract
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. Background: Prestin converts electrical energy into mechanical work. Results: The V499G/Y501H mutation significantly impairs fast motor kinetics of prestin. Conclusion: Impaired kinetics is attributable to mutation at the Val-499 site that is conserved among SLC26 proteins regardless of their function as motors or transporters. Significance: V499G/Y501H mutated prestin provides clues to the molecular mechanisms underlying somatic electromotility and thus cochlear amplification.
- Published
- 2013
25. Cadherin 23-C Regulates Microtubule Networks by Modifying CAMSAP3’s Function
- Author
-
Vincent J. Mui, Kazuaki Homma, Satoe Takahashi, Mary Ann Cheatham, Samuel K. Rosenberg, and Jing Zheng
- Subjects
0301 basic medicine ,Gene isoform ,Usher syndrome ,Mutation, Missense ,Cadherin Related Proteins ,Biology ,Microtubules ,Article ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,CDH23 ,Microtubule ,In vivo ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Multidisciplinary ,Cadherin ,Binding protein ,Cadherins ,medicine.disease ,Molecular biology ,eye diseases ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Amino Acid Substitution ,Organ of Corti ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery - Abstract
Cadherin-related 23 (CDH23) is an adhesive protein important for hearing and vision, while CAMSAP3/Marshalin is a microtubule (MT) minus-end binding protein that regulates MT networks. Although both CDH23 and CAMSAP3/Marshalin are expressed in the organ of Corti and carry several protein-protein interaction domains, no functional connection between these two proteins has been proposed. In this report, we demonstrate that the C isoform of CDH23 (CDH23-C) directly binds to CAMSAP3/Marshalin and modifies its function by inhibiting CAMSAP3/Marshalin-induced bundle formation, a process that requires a tubulin-binding domain called CKK. We further identified a conserved N-terminal region of CDH23-C that binds to the CKK domain. This CKK binding motif (CBM) is adjacent to the domain that interacts with harmonin, a binding partner of CDH23 implicated in deafness. Because the human Usher Syndrome 1D-associated mutation, CDH23 R3175H, maps to the CBM, we created a matched mutation in mouse CDH23-C at R55H. Both in vivo and in vitro assays decreased the ability of CDH23-C to interact with CAMSAP3/Marshalin, indicating that the interaction between CDH23 and CAMSAP3/Marshalin plays a vital role in hearing and vision. Together, our data suggest that CDH23-C is a CAMSAP3/Marshalin-binding protein that can modify MT networks indirectly through its interaction with CAMSAP3/Marshalin.
- Published
- 2016
26. Progress in cochlear physiology after Békésy
- Author
-
John J. Guinan, Alec N. Salt, and Mary Ann Cheatham
- Subjects
medicine.medical_specialty ,Tectorial Membrane ,Hearing loss ,Tectorial membrane ,Stereocilia (inner ear) ,Anion Transport Proteins ,Otoacoustic Emissions, Spontaneous ,Presbycusis ,Audiology ,Biology ,History, 21st Century ,Mechanotransduction, Cellular ,Models, Biological ,Vibration ,Article ,Hearing ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Prestin ,Cochlea ,History, 20th Century ,medicine.disease ,Sensory Systems ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Acoustic Stimulation ,Hearing Loss, Noise-Induced ,Sulfate Transporters ,Cochlear Microphonic Potentials ,biology.protein ,sense organs ,Hair cell ,medicine.symptom ,Cochlear microphonic potential - Abstract
In the fifty years since Békésy was awarded the Nobel Prize, cochlear physiology has blossomed. Many topics that are now current are things Békésy could not have imagined. In this review we start by describing progress in understanding the origin of cochlear gross potentials, particularly the cochlear microphonic, an area in which Békésy had extensive experience. We then review progress in areas of cochlear physiology that were mostly unknown to Békésy, including: (1) stereocilia mechano-electrical transduction, force production, and response amplification, (2) outer hair cell (OHC) somatic motility and its molecular basis in prestin, (3) cochlear amplification and related micromechanics, including the evidence that prestin is the main motor for cochlear amplification, (4) the influence of the tectorial membrane, (5) cochlear micromechanics and the mechanical drives to inner hair cell stereocilia, (6) otoacoustic emissions, and (7) olivocochlear efferents and their influence on cochlear physiology. We then return to a subject that Békésy knew well: cochlear fluids and standing currents, as well as our present understanding of energy dependence on the lateral wall of the cochlea. Finally, we touch on cochlear pathologies including noise damage and aging, with an emphasis on where the field might go in the future.
- Published
- 2012
27. Carcinoembryonic antigen-related cell adhesion molecule 16 interacts with α-tectorin and is mutated in autosomal dominant hearing loss (DFNA4)
- Author
-
Todd E. Scheetz, Richard J.H. Smith, Jennifer Drummond, Tao Yang, P. Kevin Legan, Peter Dallos, Jing Zheng, Steve Scherer, Guy P. Richardson, Richard J. Goodyear, Katharine K. Miller, Mary Ann Cheatham, A. Eliot Shearer, Adam P. DeLuca, and Michael S. Hildebrand
- Subjects
Tectorial membrane ,Blotting, Western ,Molecular Sequence Data ,GPI-Linked Proteins ,Extracellular matrix ,Mice ,Myosin ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Nonsyndromic deafness ,Hearing Loss ,Prestin ,Peptide sequence ,In Situ Hybridization ,Cochlea ,Genes, Dominant ,Myosin Type II ,Extracellular Matrix Proteins ,Multidisciplinary ,Myosin Heavy Chains ,biology ,Cell adhesion molecule ,Biological Sciences ,medicine.disease ,Molecular biology ,medicine.anatomical_structure ,Mutation ,biology.protein ,sense organs ,Cell Adhesion Molecules - Abstract
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.
- Published
- 2011
28. Interaction between the motor protein prestin and the transporter protein VAPA
- Author
-
Katharine K. Miller, Kazuaki Homma, Soma Sengupta, Mary Ann Cheatham, Jing Zheng, Peter Dallos, and Roxanne Edge
- Subjects
Recombinant Fusion Proteins ,Protein trafficking ,Vesicular Transport Proteins ,Article ,Cell Line ,Cell membrane ,Motor protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Two-Hybrid System Techniques ,medicine ,Animals ,Humans ,Prestin ,Integral membrane protein ,Molecular Biology ,VAPA ,030304 developmental biology ,Gene Library ,0303 health sciences ,biology ,Endoplasmic reticulum ,Molecular Motor Proteins ,Cell Membrane ,Membrane Proteins ,Cell Biology ,Transport protein ,Cell biology ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Membrane protein ,Outer hair cell ,biology.protein ,Hair cell ,sense organs ,Carrier Proteins ,030217 neurology & neurosurgery - Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
29. Interaction between CFTR and prestin (SLC26A5)
- Author
-
Salvador Aguinaga, Katharine K. Miller, Jing Zheng, Kazuaki Homma, Charles T. Anderson, Soma Sengupta, Mary Ann Cheatham, Peter Dallos, and Guo Guang Du
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Immunoprecipitation ,Biophysics ,Cystic Fibrosis Transmembrane Conductance Regulator ,Nonlinear capacitance ,Biology ,Chloride ,Biochemistry ,Article ,Cell Line ,Motor protein ,Cell membrane ,Mice ,Chlorides ,Cyclic AMP ,medicine ,Animals ,Humans ,RNA, Messenger ,CFTR ,Prestin ,Epithelial polarity ,Molecular Motor Proteins ,Cell Membrane ,Cell Biology ,respiratory system ,digestive system diseases ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Solute carrier family ,Cell biology ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Outer hair cell ,Chloride channel ,biology.protein ,sense organs ,SLC26A ,Ion Channel Gating - Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-activated chloride channel that is present in a variety of epithelial cell types, and usually expressed in the luminal membrane. In contrast, prestin (SLC26A5) is a voltage-dependent motor protein, which is present in the basolateral membrane of cochlear outer hair cells (OHCs), and plays an important role in the frequency selectivity and sensitivity of mammalian hearing. By using in situ hybridization and immunofluorescence, we found that both mRNA and protein of CFTR are present in OHCs, and that CFTR localizes in both the apical and the lateral membranes. CFTR was not detected in the lateral membrane of inner hair cells (IHCs) or in that of OHCs derived from prestin-knockout mice, i.e., in instances where prestin is not expressed. These results suggest that prestin may interact physically with CFTR in the lateral membrane of OHCs. Immunoprecipitation experiments confirmed a prestin–CFTR interaction. Because chloride is important for prestin function and for the efferent-mediated inhibition of cochlear output, the prestin-directed localization of CFTR to the lateral membrane of OHCs has a potential physiological significance. Aside from its role as a chloride channel, CFTR is known as a regulator of multiple protein functions, including those of the solute carrier family 26 (SLC26). Because prestin is in the SLC26 family, several members of which interact with CFTR, we explored the potential modulatory relationship associated with a direct, physical interaction between prestin and CFTR. Electrophysiological experiments demonstrated that cAMP-activated CFTR is capable of enhancing voltage-dependent charge displacement, a signature of OHC motility, whereas prestin does not affect the chloride conductance of CFTR.
- Published
- 2010
- Full Text
- View/download PDF
30. EHD4 and CDH23 Are Interacting Partners in Cochlear Hair Cells
- Author
-
Mary Ann Cheatham, Jonathan Chou, Peter Dallos, Katharine K. Miller, Hamid Band, Soma Sengupta, Khurram Naik, Manju George, Jing Zheng, and Mayumi Naramura
- Subjects
Stereocilia (inner ear) ,Vesicular Transport Proteins ,Gene Expression ,Endocytic recycling ,Biology ,Kidney ,Biochemistry ,Cell Line ,Mice ,Molecular Basis of Cell and Developmental Biology ,CDH23 ,Hearing ,Two-Hybrid System Techniques ,Hair Cells, Auditory ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Inner ear ,Molecular Biology ,Cochlea ,Mice, Knockout ,Genetics ,Cadherin ,Nuclear Proteins ,Cell Biology ,Cadherins ,Transmembrane protein ,Protein Structure, Tertiary ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,medicine.anatomical_structure ,sense organs ,Hair cell - Abstract
Cadherin 23 (CDH23), a transmembrane protein localized near the tips of hair cell stereocilia in the mammalian inner ear, is important for delivering mechanical signals to the mechano-electric transducer channels. To identify CDH23-interacting proteins, a membrane-based yeast two-hybrid screen of an outer hair cell (OHC) cDNA library was performed. EHD4, a member of the C-terminal EH domain containing a protein family involved in endocytic recycling, was identified as a potential interactor. To confirm the interaction, we first demonstrated the EHD4 mRNA expression in hair cells using in situ hybridization. Next, we showed that EHD4 co-localizes and co-immunoprecipitates with CDH23 in mammalian cells. Interestingly, the co-immunoprecipitation was found to be calcium-sensitive. To investigate the role of EHD4 in hearing, compound action potentials were measured in EHD4 knock-out (KO) mice. Although EHD4 KO mice have normal hearing sensitivity, analysis of mouse cochlear lysates revealed a 2-fold increase in EHD1, but no increase in EHD2 or EHD3, in EHD4 KO cochleae compared with wild type, suggesting that a compensatory increase in EHD1 levels may account for the absence of a hearing defect in EHD4 KO mice. Taken together, these data indicate that EHD4 is a novel CDH23-interacting protein that could regulate CDH23 trafficking/localization in a calcium-sensitive manner.
- Published
- 2009
31. Prestin-Based Outer Hair Cell Motility Is Necessary for Mammalian Cochlear Amplification
- Author
-
Jing Zheng, Jian Zuo, Peter Dallos, Xudong Wu, David Z.Z. He, Wendy H.Y. Cheng, Mary Ann Cheatham, Shuping Jia, Charles T. Anderson, Xiang Wang, Soma Sengupta, and Jiangang Gao
- Subjects
Somatic cell ,Neuroscience(all) ,Motility ,Action Potentials ,Biology ,Mechanotransduction, Cellular ,Article ,MOLNEURO ,Motor protein ,Mice ,Hearing ,Cell Movement ,medicine ,otorhinolaryngologic diseases ,Humans ,Animals ,Cilia ,Prestin ,Mammals ,Mice, Knockout ,General Neuroscience ,Molecular Motor Proteins ,Auditory Threshold ,Mice, Mutant Strains ,Cell biology ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Organ of Corti ,Knockout mouse ,Models, Animal ,biology.protein ,Hair cell ,sense organs ,Transduction (physiology) ,Neuroscience - Abstract
It is a central tenet of cochlear neurobiology that mammalian ears rely on a local, mechanical amplification process for their high sensitivity and sharp frequency selectivity. While there is general agreement that outer hair cells provide the amplification, two mechanisms have been proposed: stereociliary motility and somatic motility. The latter is driven by the motor protein prestin. Electrophysiological phenotyping of a prestin knockout mouse intimated that somatic motility is the amplifier. However, outer hair cells of knockout mice have significantly altered mechanical properties, which makes this mouse model unsatisfactory. Here we study a new mouse model without alteration to outer hair cell and organ of Corti mechanics or to mechano-electric transduction, but with diminished prestin function. These animals have knockout-like behavior, demonstrating that prestin-based electromotility is required for cochlear amplification.
- Published
- 2008
- Full Text
- View/download PDF
32. Glucose transporter 5 is undetectable in outer hair cells and does not contribute to cochlear amplification
- Author
-
Yiling Yu, Shuping Jia, Xudong Wu, Jian Zuo, Jiangang Gao, Xiang Wang, David Z.Z. He, Jing Zheng, Peter Dallos, and Mary Ann Cheatham
- Subjects
Male ,medicine.medical_specialty ,Glucose Transport Proteins, Facilitative ,Motility ,Fructose ,Biology ,Mechanotransduction, Cellular ,Article ,Mice ,Hearing ,Microscopy, Electron, Transmission ,Cell Movement ,Internal medicine ,Testis ,otorhinolaryngologic diseases ,medicine ,Animals ,Inner ear ,Cilia ,RNA, Messenger ,Prestin ,Molecular Biology ,Cells, Cultured ,Cochlea ,Mice, Knockout ,Chimera ,Glucose Transporter Type 5 ,Molecular Motor Proteins ,Stem Cells ,General Neuroscience ,Glucose transporter ,Transporter ,Mice, Inbred C57BL ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Endocrinology ,biology.protein ,sense organs ,Neurology (clinical) ,Hair cell ,Energy Metabolism ,GLUT5 ,Developmental Biology - Abstract
Glucose transporter 5 (Glut5) is a high-affinity fructose transporter. It was proposed to be a motor protein or part of the motor complex required for cochlear amplification in outer hair cells (OHCs). Here we show that, in contrast to previous reports, Glut5 is undetectable, and possibly absent, in OHCs harvested from wildtype mice. Further, Glut5-deficient mice display normal OHC morphology and motor function (i.e., nonlinear capacitance and electromotility) and normal cochlear sensitivity and frequency selectivity. We conclude that Glut5 is not required for OHC motility or cochlear amplification.
- Published
- 2008
33. Prestin-based outer hair cell electromotility in knockin mice does not appear to adjust the operating point of a cilia-based amplifier
- Author
-
Keiji Matsuda, Jian Zuo, Jiangang Gao, Shuping Jia, Peter Dallos, Xiang Wang, Mary Ann Cheatham, Kristin Huynh, Sal Aguinaga, David Z.Z. He, Jing Zheng, Manish Patel, and Xudong Wu
- Subjects
Somatic cell ,Stereocilia (inner ear) ,Molecular Sequence Data ,Motility ,Mice, Transgenic ,Cell Separation ,Cell Line ,Motor protein ,Mice ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Cilia ,Prestin ,Cell Shape ,Cochlea ,Multidisciplinary ,Amplifiers, Electronic ,Base Sequence ,biology ,Molecular Motor Proteins ,Proteins ,Anatomy ,Biological Sciences ,Hyperpolarization (biology) ,Cell biology ,Electrophysiology ,Mice, Inbred C57BL ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Nonlinear Dynamics ,Mutation ,Auditory Perception ,biology.protein ,sense organs ,Hair cell - Abstract
The remarkable sensitivity and frequency selectivity of the mammalian cochlea is attributed to a unique amplification process that resides in outer hair cells (OHCs). Although the mammalian-specific somatic motility is considered a substrate of cochlear amplification, it has also been proposed that somatic motility in mammals simply acts as an operating-point adjustment for the ubiquitous stereocilia-based amplifier. To address this issue, we created a mouse model in which a mutation (C1) was introduced into the OHC motor protein prestin, based on previous results in transfected cells. In C1/C1 knockin mice, localization of C1-prestin, as well as the length and number of OHCs, were all normal. In OHCs isolated from C1/C1 mice, nonlinear capacitance and somatic motility were both shifted toward hyperpolarization, so that, compared with WT controls, the amplitude of cycle-by-cycle (alternating, or AC) somatic motility remained the same, but the unidirectional (DC) component reversed polarity near the OHC's presumed in vivo resting membrane potential. No physiological defects in cochlear sensitivity or frequency selectivity were detected in C1/C1 or C1/+ mice. Hence, our results do not support the idea that OHC somatic motility adjusts the operating point of a stereocilia-based amplifier. However, they are consistent with the notion that the AC component of OHC somatic motility plays a dominant role in mammalian cochlear amplification.
- Published
- 2007
34. Evaluation of an Independent Prestin Mouse Model Derived from the 129S1 Strain
- Author
-
Mary Ann Cheatham, Jian Zuo, Charles T. Anderson, Peter Dallos, Guo Guang Du, Allen F. Ryan, Jing Zheng, Roxanne Edge, and K. H. Huynh
- Subjects
Heterozygote ,Cochlear amplifier ,Pathology ,medicine.medical_specialty ,Genotype ,Physiology ,Mice, Transgenic ,Motor protein ,Mice ,Speech and Hearing ,medicine ,Animals ,RNA, Messenger ,Prestin ,Cochlea ,Mice, Knockout ,Microscopy, Confocal ,Round window ,biology ,Molecular Motor Proteins ,Homozygote ,Auditory Threshold ,Exons ,Mice, Mutant Strains ,Sensory Systems ,Compound muscle action potential ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Hair Cells, Auditory, Outer ,Phenotype ,medicine.anatomical_structure ,Otorhinolaryngology ,Gene Targeting ,Knockout mouse ,Cochlear Microphonic Potentials ,biology.protein ,sense organs ,Hair cell - Abstract
Studies using the prestin knockout mouse indicate that removal of the outer hair cell (OHC) motor protein is associated with loss of sensitivity, frequency selectivity and somatic electromotility. Here we provide data obtained from another prestin mouse model that was produced commercially. In vivo electrical recordings from the round window indicate that the phenotype is similar to that of the original knockout generated by the Zuo group at St. Jude Children’s Research Hospital. Hence, compound action potential (CAP) thresholds are shifted in a frequency-dependent manner and CAP tuning curves at 12 kHz are flat for masker frequencies between 3 and 18 kHz. Although CAP input-output functions at 6 kHz show a shift in sensitivity at low levels, responses approach wild-type magnitudes at high levels where the cochlear amplifier has less influence. In order to confirm that the loss of sensitivity and frequency selectivity is due to loss of prestin, we performed immunohistochemistry using a prestin antibody. Cochlear segments from homozygous mutant mice showed no fluorescence, while wild-type mice displayed a fluorescent signal targeted to the OHC’s lateral membrane. Absence of prestin protein was confirmed using LDS-PAGE/Western blot analysis. These results indicate that the loss of function phenotype is associated with loss of prestin protein. Lack of prestin protein also results in a shortening of OHC length to ∼60% of wild-type, similar to that reported previously by Liberman’s group. The linkage shown between the loss of prestin protein and abnormal cochlear function validates the original knockout and attests to the importance of OHC motor function in the auditory periphery.
- Published
- 2007
35. Susceptibility of outer hair cells to cholesterol chelator 2-hydroxypropyl-β-cyclodextrine is prestin-dependent
- Author
-
Kazuaki Homma, Satoe Takahashi, Shinichi Nishimura, Aisha Ahmad, Yingjie Zhou, Jessie Chen, Chongwen Duan, Jing Zheng, and Mary Ann Cheatham
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Hearing Loss, Sensorineural ,Gene Expression ,Time-Lapse Imaging ,Article ,Motor protein ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Ototoxicity ,Internal medicine ,medicine ,Animals ,Humans ,Prestin ,Chelating Agents ,Mice, Knockout ,Multidisciplinary ,biology ,Cell Death ,Cholesterol ,Molecular Motor Proteins ,beta-Cyclodextrins ,Wild type ,Brain ,Niemann-Pick Disease, Type C ,medicine.disease ,In vitro ,3. Good health ,2-Hydroxypropyl-beta-cyclodextrin ,Hair Cells, Auditory, Outer ,030104 developmental biology ,Endocrinology ,Neuroprotective Agents ,chemistry ,Biochemistry ,biology.protein ,sense organs ,Disease Susceptibility ,NPC1 ,Niemann–Pick disease ,030217 neurology & neurosurgery - Abstract
Niemann-Pick type C1 disease (NPC1) is a fatal genetic disorder caused by impaired intracellular cholesterol trafficking. Recent studies reported ototoxicity of 2-hydroxypropyl- β-cyclodextrin (HPβCD), a cholesterol chelator and the only promising treatment for NPC1. Because outer hair cells (OHCs) are the only cochlear cells affected by HPβCD, we investigated whether prestin, an OHC-specific motor protein, might be involved. Single, high-dose administration of HPβCD resulted in OHC death in prestin wildtype (WT) mice whereas OHCs were largely spared in prestin knockout (KO) mice in the basal region, implicating prestin’s involvement in ototoxicity of HPβCD. We found that prestin can interact with cholesterol in vitro, suggesting that HPβCD-induced ototoxicity may involve disruption of this interaction. Time-lapse analysis revealed that OHCs isolated from WT animals rapidly deteriorated upon HPβCD treatment while those from prestin-KOs tolerated the same regimen. These results suggest that a prestin-dependent mechanism contributes to HPβCD ototoxicity.
- Published
- 2015
36. Separating medial olivocochlear from acoustic reflex effects on transient evoked otoacoustic emissions in unanesthetized mice
- Author
-
Yingyue Xu, Mary Ann Cheatham, and Jonathan H. Siegel
- Subjects
medicine.medical_specialty ,Contraction (grammar) ,Broadband noise ,Chemistry ,Efferent ,Middle ear muscle ,Audiology ,Cochlear function ,Peripheral ,medicine.anatomical_structure ,embryonic structures ,otorhinolaryngologic diseases ,medicine ,Auditory system ,sense organs ,Acoustic reflex - Abstract
Descending neural pathways in the mammalian auditory system are believed to modulate the function of the peripheral auditory system [3, 8, 10]. These pathways include the medial olivocochlear (MOC) efferent innervation to the cochlear outer hair cells (OHCs) and the acoustic reflex pathways mediating middle ear muscle (MEM) contractions. The MOC effects can be monitored noninvasively using otoacoustic emissions (OAEs) [5, 6], which are acoustic byproducts of cochlear function [7]. In this study, we applied a sensitive method to determine when and to what degree contralateral MEM suppression contaminated MOC efferent effects on TEOAEs in unanesthetized mice. The lowest contralateral broadband noise evoking MEM contractions varied across animals. Examples of potential MOC-mediated TEOAE suppression with contralateral noise below MEM contraction thresholds were seen, but this behavior did not occur in the majority of cases.
- Published
- 2015
37. Prestin and the cochlear amplifier
- Author
-
Jing Zheng, Mary Ann Cheatham, and Peter Dallos
- Subjects
Cochlear amplifier ,medicine.medical_specialty ,Physiology ,Stereocilia (inner ear) ,Stereovilli ,Audiology ,Biology ,Motor protein ,otorhinolaryngologic diseases ,medicine ,biology.protein ,sense organs ,Mechanotransduction ,Outer hair cells ,Molecular Motor Proteins ,Prestin ,Neuroscience - Abstract
In non-mammalian, hair cell-bearing sense organs amplification is associated with mechano-electric transducer channels in the stereovilli (commonly called stereocilia). Because mammals possess differentiated outer hair cells (OHC), they also benefit from a novel electromotile process, powered by the motor protein, prestin. Here we consider new work pertaining to this protein and its potential role as the mammalian cochlear amplifier.
- Published
- 2006
38. Cochlear function in mice with only one copy of theprestingene
- Author
-
Mary Ann Cheatham, E. Navarrete, Peter Dallos, Guo Guang Du, Jiangang Gao, Jing Zheng, Jian Zuo, and K. H. Huynh
- Subjects
Genetics ,medicine.diagnostic_test ,Physiology ,Somatic cell ,Immunocytochemistry ,Wild type ,Heterozygote advantage ,Biology ,Compound muscle action potential ,Cell biology ,medicine.anatomical_structure ,Western blot ,medicine ,biology.protein ,sense organs ,Hair cell ,Prestin - Abstract
Targeted deletion of the prestin gene reduces cochlear sensitivity and eliminates both frequency selectivity and outer hair cell (OHC) somatic electromotility. In addition, it has been reported by Liberman and colleagues that F2 generation heterozygotes exhibit a 6 dB reduction in sensitivity, as well as a decrease in protein and electromotility. Considering that the active process is non-linear, a halving of somatic electromotility would be expected to produce a much larger change in sensitivity. We therefore re-evaluated comparisons between heterozygotes and wildtype mice using both in vivo and in vitro electrophysiology, as well as molecular biology. Data reported here for F3–F5 generation mice indicate that compound action potential thresholds and tuning curves, as well as the cochlear microphonic, are similar in heterozygotes and wildtype controls. Measurements of non-linear capacitance in isolated OHCs demonstrate that charge density, as well as the voltage dependence and sensitivity of motor function, is indistinguishable in the two genotypes, as is somatic electromotility. In addition, both immunocytochemistry and western blot analysis in young adult mice suggest that prestin protein in heterozygotes is near normal. In contrast, prestin mRNA is always less than in wildtype mice at all ages tested. Results from F3–F5 generation mice suggest that one copy of the prestin gene is capable of compensating for the deleted copy and that heterozygous mice do not suffer peripheral hearing impairment.
- Published
- 2005
39. Cochlear function inPrestinknockout mice
- Author
-
Jian Zuo, K. H. Huynh, Jiangang Gao, Mary Ann Cheatham, and Peter Dallos
- Subjects
Cochlear amplifier ,medicine.medical_specialty ,biology ,Physiology ,Chemistry ,Audiology ,Compound muscle action potential ,Cell biology ,medicine.anatomical_structure ,Combination tone ,Knockout mouse ,biology.protein ,medicine ,sense organs ,Hair cell ,Prestin ,Transduction (physiology) ,Intermodulation - Abstract
Gross-potential recordings in mice lacking the Prestin gene indicate that compound action potential (CAP) thresholds are shifted by ∼45 dB at 5 kHz and by ∼60 dB at 33 kHz. However, in order to conclude that outer hair cell (OHC) electromotility is associated with the cochlear amplifier, frequency selectivity must be evaluated and the integrity of the OHC's forward transducer ascertained. The present report demonstrates no frequency selectivity in CAP tuning curves recorded in homozygotes. In addition, CAP input–output functions indicate that responses in knockout mice approach those in controls at high levels where the amplifier has little influence. Although the cochlear microphonic in knockout mice remains ∼12 dB below that in wild-type mice even at the highest levels, this deficit is thought to reflect hair cell losses in mice lacking prestin. A change in OHC forward transduction is not implied because knockout mice display non-linear responses similar to those in controls. For example, homozygotes exhibit a bipolar summating potential (SP) with positive responses at high frequencies; negative responses at low frequencies. Measurement of intermodulation distortion also shows that the cubic difference tone, 2f1–f2, is ∼20 dB down from the primaries in both homozygotes and their controls. Because OHCs are the sole generators of the negative SP and because 2f1–f2 is also thought to originate in OHC transduction, these data support the idea that forward transduction is not degraded in OHCs lacking prestin. Finally, application of AM1-43, which initially enters hair cells through their transducer channels, produces fluorescence in wild-type and knockout mice indicating transducer channel activity in both inner and outer hair cells.
- Published
- 2004
40. Mouse outer hair cells lacking the α9 ACh receptor are motile
- Author
-
Douglas E. Vetter, David Z.Z. He, Mary Ann Cheatham, and Malini Pearce
- Subjects
medicine.medical_specialty ,Patch-Clamp Techniques ,Protein subunit ,Efferent ,Action Potentials ,Mice, Inbred Strains ,Receptors, Nicotinic ,Neurotransmission ,Biology ,Inhibitory postsynaptic potential ,Efferent nerve ,Membrane Potentials ,Mice ,chemistry.chemical_compound ,Developmental Neuroscience ,Cell Movement ,Internal medicine ,medicine ,Animals ,Neurotransmitter ,Cells, Cultured ,Cell Size ,Acetylcholine receptor ,Mice, Knockout ,Electric Conductivity ,Electric Stimulation ,Cell biology ,Hair Cells, Auditory, Outer ,Endocrinology ,chemistry ,sense organs ,Acetylcholine ,Developmental Biology ,medicine.drug - Abstract
Efferent nerve fibers form chemical synapses at the bases of outer hair cells (OHC), with acetylcholine (ACh) being their principal neurotransmitter. The activation of ACh receptors on OHCs is known to influence cochlear function. These efferent effects exhibit an unusual pharmacology and are generally known to be inhibitory. Recent evidence suggests that an ACh receptor subunit, known as alpha9, plays a dominant role in mediating the olivocochlear neurotransmission to OHCs. In this investigation, we attempt to determine the possible role(s) of the alpha9 subunit in regulating OHC function by examining OHC electromotility and compound action potentials (CAP) in mice carrying a null mutation for the alpha9 gene. Results indicate that cochlear sensitivity, based on CAP thresholds, is similar for homozygous mutant and wild-type mice. Electromotility is also present in OHCs, independent of whether the alpha9 subunit is present or absent.
- Published
- 2004
41. Loss of the tectorial membrane protein CEACAM16 enhances spontaneous, stimulus-frequency, and transiently evoked otoacoustic emissions
- Author
-
Kazuaki Homma, Mary Ann Cheatham, Guy P. Richardson, Jonathan H. Siegel, Souvik Naskar, Peter Dallos, Richard J. Goodyear, Julia Korchagina, Jing Zheng, and P K Legan
- Subjects
Cochlear amplifier ,Patch-Clamp Techniques ,Tectorial Membrane ,Tectorial membrane ,Otoacoustic Emissions, Spontaneous ,Matrix (biology) ,Biology ,Membrane Potentials ,Mice ,Microscopy, Electron, Transmission ,medicine ,Evoked Potentials, Auditory, Brain Stem ,Animals ,Humans ,Immunoprecipitation ,Inner ear ,TECTA ,Organ of Corti ,Cochlea ,Mice, Knockout ,Extracellular Matrix Proteins ,Cell adhesion molecule ,General Neuroscience ,Anatomy ,Articles ,beta-Galactosidase ,Mice, Inbred C57BL ,medicine.anatomical_structure ,HEK293 Cells ,Acoustic Stimulation ,Gene Expression Regulation ,Biophysics ,sense organs ,Cell Adhesion Molecules - Abstract
α-Tectorin (TECTA), β-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 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.
- Published
- 2014
42. Functional regulation of the SLC26-family protein prestin by calcium/calmodulin
- Author
-
Peter Dallos, Mary Ann Cheatham, Chongwen Duan, Jing Zheng, Kazuaki Homma, and Jacob P. Keller
- Subjects
Male ,Patch-Clamp Techniques ,Calmodulin ,Anion Transport Proteins ,Molecular Sequence Data ,Biology ,Mice ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,Prestin ,Binding Sites ,Base Sequence ,General Neuroscience ,Molecular Motor Proteins ,Articles ,Transport protein ,Cell biology ,Solute carrier family ,Hair Cells, Auditory, Outer ,Membrane protein ,Biochemistry ,Sulfate Transporters ,Second messenger system ,biology.protein ,Calcium ,Female ,sense organs ,Intracellular - Abstract
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.
- Published
- 2014
43. Inner hair cell response patterns: Implications for low-frequency hearing
- Author
-
Peter Dallos and Mary Ann Cheatham
- Subjects
Frequency response ,medicine.medical_specialty ,Sound Spectrography ,Acoustics and Ultrasonics ,Acoustics ,media_common.quotation_subject ,Guinea Pigs ,Receptor potential ,Audiology ,Asymmetry ,Hearing ,Species Specificity ,Arts and Humanities (miscellaneous) ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Pitch Perception ,media_common ,Physics ,Hair Cells, Auditory, Inner ,Low frequency hearing ,Auditory Threshold ,Cochlea ,Helicotrema ,medicine.anatomical_structure ,Evoked Potentials, Auditory ,Middle ear ,sense organs ,Hair cell ,Guinea pig cochlea - Abstract
Inner hair cell (IHC) responses to tone-burst stimuli were measured from three locations in the apical half of the guinea pig cochlea. In addition to the measurement of ac receptor potentials, average intracellular voltages, reflecting both ac and dc components of the receptor potential, were computed and compared to determine how bandwidth changes with level. Companion phase measures were also obtained and evaluated. Data collected from turn 2, where best frequency (BF) is approximately 4000 Hz, indicate that frequency response functions are asymmetrical with steeper slopes above the best frequency of the cell. However, in turn 4, where BF is around 250 Hz, the opposite behavior is observed and the steepest slopes are measured below BF. The data imply that cochlear filters are generally asymmetrical with steeper slopes above BF. High-pass filtering by the middle ear serves to reduce this asymmetry in turn 3 and to reverse it in turn 4. Apical response patterns are used to assess the degree to which the middle ear transfer function, the IHC's velocity dependence and the shunting effect of the helicotrema influence low-frequency hearing in guinea pigs. Implications for low-frequency hearing in man are also discussed.
- Published
- 2001
44. Use of the Pinna Reflex as a Test of Hearing in Mutant Mice
- Author
-
Malini Pearce, Claus Peter Richter, Mary Ann Cheatham, Ko Onodera, and Jordan A. Shavit
- Subjects
medicine.medical_specialty ,Physiology ,Mice ,Speech and Hearing ,Hearing ,Internal medicine ,Reflex ,Evoked Potentials, Auditory, Brain Stem ,otorhinolaryngologic diseases ,medicine ,Animals ,Inner ear ,Ear, External ,Cochlear Nerve ,Electrodes ,Cochlea ,Vestibular system ,Round window ,biology ,business.industry ,Pinna ,Wild type ,Auditory Threshold ,Anatomy ,biology.organism_classification ,Mice, Mutant Strains ,Sensory Systems ,Endocrinology ,medicine.anatomical_structure ,Acoustic Stimulation ,Round Window, Ear ,Otorhinolaryngology ,Organ of Corti ,business - Abstract
Although it is a gross measure, the pinna reflex test is easily administered and is, therefore, incorporated as a general screening tool in mutagenesis programs. Our recent application of this approach indicated that mutant mice lacking one of the small Maf proteins, in this case MafG, failed to exhibit a pinna reflex. In contrast, littermate controls, with the same mixed 129/CD1 background, and including both wild type and heterozygous mutant animals, passed the test. Because previous studies indicate that mafG is expressed in both cochlear and vestibular parts of the mouse inner ear, the source of this ‘presumed deafness’ was further assessed by making round window recordings to determine compound action potential thresholds. Auditory brainstem responses were also acquired to assess function along portions of the central auditory pathway. In all cases, responses in homozygous mutants (–/–) were comparable to those obtained from littermate controls, either wild type (+/+) or heterozygous mutants (+/–). Gross anatomy of the organ of Corti was also found to be similar in all three groups of mice. Hence, the lack of a pinna reflex must relate to nonauditory causes.
- Published
- 2001
45. The dynamic range of inner hair cell and organ of Corti responses
- Author
-
Peter Dallos and Mary Ann Cheatham
- Subjects
Physics ,Hair Cells, Auditory, Inner ,Acoustics and Ultrasonics ,Acoustics ,Guinea Pigs ,Transducers ,Receptor potential ,Signal ,Biomechanical Phenomena ,Cochlea ,Intensity (physics) ,Synapse ,medicine.anatomical_structure ,Arts and Humanities (miscellaneous) ,Organ of Corti ,otorhinolaryngologic diseases ,Psychophysics ,medicine ,Animals ,Hair cell ,Neuroscience - Abstract
Inner hair cell (IHC) and organ of Corti (OC) responses are measured from the apical three turns of the guinea pig cochlea, allowing access to regions with best, or most sensitive, frequencies at approximately 250, 1000, and 4000 Hz. In addition to measuring both ac and dc receptor potentials, the average value of the half-wave rectified response (AVEHR) is computed to better reflect the signal that induces transmitter release. This measure facilitates comparisons with single-unit responses in the auditory nerve. Although IHC ac responses exhibit compressive growth, response magnitudes at high levels depend on stimulus frequency. For example, IHCs with moderate and high best frequencies (BF) exhibit more linear responses below the BF of the cell, where higher sound-pressure levels are required to approach saturation. Because a similar frequency dependence is observed in extracellular OC responses, this phenomenon may originate in cochlear mechanics. At the most apical recording location, however, the pattern documented at the base of the cochlea is not seen in IHCs with low BFs around 250 Hz. In fact, more linear behavior is measured above the BF of the cell. These frequency-dependent features require modification of cochlear models that do not provide for longitudinal variations and generally depend on a single stage of saturation located at the synapse. Finally, behavior of dc and AVEHR responses suggests that a single IHC is capable of coding intensity over a large dynamic range [Patuzzi and Sellick, J. Acoust. Soc. Am. 74, 1734-1741 (1983); Smith et al., in Hearing--Physiological Bases and Psychophysics (Springer, Berlin, 1983); Smith, in Auditory Function (Wiley, New York, 1988)] and that information compiled over wide areas along the cochlear partition is not essential for loudness perception, consistent with psychophysical results [Viemeister, Hearing Res. 34, 267-274 (1988)].
- Published
- 2000
46. Response phase: A view from the inner hair cell
- Author
-
Peter Dallos and Mary Ann Cheatham
- Subjects
Neurons ,Hair Cells, Auditory, Inner ,Time Factors ,Acoustics and Ultrasonics ,Chemistry ,Acoustics ,Stereocilia (inner ear) ,Guinea Pigs ,Phase (waves) ,Receptor potential ,Depolarization ,Basilar Membrane ,Basilar membrane ,medicine.anatomical_structure ,Arts and Humanities (miscellaneous) ,otorhinolaryngologic diseases ,medicine ,Biophysics ,Animals ,sense organs ,Hair cell ,Cochlea ,Ion channel - Abstract
Inner hair cell (IHC) responses are recorded from the apical three turns of the guinea pig cochlea in order to define the relationship between hair cell depolarization and position of the basilar membrane. At low frequencies, inner hair cell depolarization is generally observed near basilar membrane velocity to scala vestibuli, reflecting the putative freestanding nature of the IHC's stereocilia. While this is consistent with previous IHC results, independent of location, and with neural responses for fibers with low best frequencies, it is inconsistent with single-unit results from the base of the cochlea, where response phase is associated with basilar membrane velocity to scala tympani. Results suggest that the temporal disparity between IHC and neural data from the base of the cochlea may relate to several factors that influence transmembrane voltage in IHCs. First, extracellular voltages (Ingvarsson, 1981; Sellick et al., 1982; Russell and Sellick, 1983) can potentially affect low- and high-frequency regions differently because electrical interactions are more likely in the base of the cochlea than in the apex (Dallos, 1983, 1985). Second, waveform distortion and kinetic properties associated with voltage-dependent ion channels in the IHC's basolateral membrane can both influence response phase by adding harmonic components and lagging the receptor potential by as much as 90 deg. Third, the velocity dependence of IHCs in the apex appears to extend to higher frequencies than the velocity dependence demonstrated for IHCs in the base of the cochlea. These features, which influence the timing of discharges in the auditory nerve, are compared and evaluated.
- Published
- 1999
47. The level dependence of response phase: Observations from cochlear hair cells
- Author
-
Mary Ann Cheatham and Peter Dallos
- Subjects
Cochlear microphonic ,Acoustics and Ultrasonics ,Chemistry ,Acoustics ,Depolarization ,Stimulus (physiology) ,Models, Biological ,Basilar Membrane ,Basilar membrane ,medicine.anatomical_structure ,Acoustic Stimulation ,Arts and Humanities (miscellaneous) ,Organ of Corti ,Hair Cells, Auditory ,otorhinolaryngologic diseases ,medicine ,Biophysics ,Traveling wave ,Humans ,sense organs ,Hair cell ,Guinea pig cochlea - Abstract
Hair cell responses are recorded from third turn of the guinea pig cochlea in order to define the relationship between hair cell depolarization and position of the basilar membrane. Because the latter is determined locally, using the cochlear microphonic recorded in the organ of Corti (OC) fluid space, no corrections are required to compensate traveling wave and/or synaptic delays. At low levels, inner hair cells (IHC) depolarize near basilar membrane velocity to scala vestibuli reflecting the free standing nature of their stereocilia. At high levels, the time of depolarization changes rapidly from velocity to scala vestibuli to the scala tympani phase of the basilar membrane response. This change in response phase, recorded in the fundamental component of the IHC response, is associated with a decrease in response magnitude. The absence of this behavior in OC and outer hair cell responses implies that basilar membrane mechanics may not be responsible for these response patterns. Because these features are reminiscent of the magnitude notches and the large phase shifts observed in single unit responses at high stimulus levels, they provide the IHC correlates of these phenomena.
- Published
- 1998
48. Intermodulation components in inner hair cell and organ of Corti responses
- Author
-
Mary Ann Cheatham and Peter Dallos
- Subjects
Physics ,Acoustics and Ultrasonics ,Acoustics ,Guinea Pigs ,Basilar Membrane ,Cochlea ,Basilar membrane ,medicine.anatomical_structure ,Acoustic Stimulation ,Arts and Humanities (miscellaneous) ,Organ of Corti ,Distortion ,Hair Cells, Auditory ,Psychophysics ,medicine ,Animals ,sense organs ,Hair cell ,Guinea pig cochlea ,Displacement (fluid) ,Intermodulation - Abstract
Two-tone responses are recorded from inner hair cells and from the organ of Corti fluid space in second and third turns of the guinea pig cochlea where best frequencies (BF) are approximately 4000 and 1000 Hz, respectively. This allows both ac and dc response components to be obtained and facilitates comparisons with psychophysical investigations that have traditionally been conducted at low and moderate frequencies. The measurements of ac responses in the organ of Corti fluid space also allow comparisons with mechanical results because the cochlear microphonic is proportional to basilar membrane displacement. By using a constant frequency ratio (f2/f1) of 1.4, local distortion products generated at the recording location are prominent when the two primaries are near the BF of the cell. However, when the primary pairs increase above BF, quadratic and cubic difference tones are recorded even when responses to the primaries are not measurable. The presence of these traveling distortion products is consistent with the idea that both f2-f1 and 2f1-f2 have their own traveling waves. Notches in the existence regions of quadratic and cubic difference tones were also observed and found to be influenced by mutual suppression between the two inputs.
- Published
- 1997
49. Low-frequency modulation of inner hair cell and organ of Corti responses in the guinea pig cochlea
- Author
-
Peter Dallos and Mary Ann Cheatham
- Subjects
medicine.medical_specialty ,Guinea Pigs ,Receptor potential ,Audiology ,Stimulus (physiology) ,Biology ,Low frequency modulation ,Membrane Potentials ,medicine ,Animals ,Organ of Corti ,Cochlea ,Hair Cells, Auditory, Inner ,Vestibulocochlear Nerve ,Sensory Systems ,Basilar membrane ,medicine.anatomical_structure ,Acoustic Stimulation ,Cochlear Microphonic Potentials ,Evoked Potentials, Auditory ,Biophysics ,sense organs ,Hair cell ,Guinea pig cochlea - Abstract
Low-frequency tones are used to study changes in responsiveness as a function of phase in inner hair cell (IHC) and organ of Corti (OC) responses recorded from second turn of the guinea pig cochlea. In these experiments a 40 Hz stimulus is combined with a variable frequency probe to determine the degree to which tones at and below best frequency (BF) are modulated. Changes in responsiveness produced by the low-frequency input are quantified and related to position of the basilar membrane which is estimated using the phase of the cochlear microphonic measured in the OC fluid space. Results obtained when 40 Hz is presented at its lowest effective level demonstrate that ac responses to low-level BF probes are reduced for basilar membrane displacements to scala tympani while probe tones well below BF are modulated in the opposite direction. The transition between these two response patterns occurs when the overall DC produced in the OC by the two-tone input changes from positive to negative. Because of this association, the frequency dependence exhibited in the bias results may be linked to mechanisms responsible for generating the two polarities of the summating potential and the DC receptor potentials that it reflects. An attempt is also made to relate bias-induced changes in hair cell receptor potentials to modulations in single-unit rate responses. In other words, to address variations in the temporal relationships between excitation and suppression measured in the auditory nerve.
- Published
- 1997
50. Prestin-Dependence of Outer Hair Cell Survival and Partial Rescue of Outer Hair Cell Loss in PrestinV499G/Y501H Knockin Mice
- Author
-
Kazuaki Homma, Peter Dallos, Mary Ann Cheatham, Roxanne Edge, Emily L. Leserman, and Jing Zheng
- Subjects
Mice, 129 Strain ,Cell Survival ,Transgene ,Drug Evaluation, Preclinical ,Mutation, Missense ,lcsh:Medicine ,Mice, Transgenic ,Mitochondrion ,medicine.disease_cause ,Antioxidants ,Motor protein ,medicine ,Animals ,Gene Knock-In Techniques ,Hearing Loss ,lcsh:Science ,Prestin ,Cochlea ,Genetics ,Mutation ,Multidisciplinary ,biology ,Molecular Motor Proteins ,lcsh:R ,Cell biology ,Mice, Inbred C57BL ,Hair Cells, Auditory, Outer ,medicine.anatomical_structure ,Apoptosis ,biology.protein ,lcsh:Q ,sense organs ,Hair cell ,Drugs, Chinese Herbal ,Research Article - Abstract
A knockin (KI) mouse expressing mutated prestin V499G/Y501H (499 prestin) was created to study cochlear amplification. Recordings from isolated outer hair cells (OHC) in this mutant showed vastly reduced electromotility and, as a consequence, reduced hearing sensitivity. Although 499 prestin OHCs were normal in stiffness and longer than OHCs lacking prestin, accelerated OHC death was unexpectedly observed relative to that documented in prestin knockout (KO) mice. These observations imply an additional role of prestin in OHC maintenance besides its known requirement for mammalian cochlear amplification. In order to gain mechanistic insights into prestin-associated OHC loss, we implemented several interventions to improve survival. First, 499 prestin KI’s were backcrossed to Bak KO mice, which lack the mitochondrial pro-apoptotic gene Bak. Because oxidative stress is implicated in OHC death, another group of 499 prestin KI mice was fed the antioxidant diet, Protandim. 499 KI mice were also backcrossed onto the FVB murine strain, which retains excellent high-frequency hearing well into adulthood, to reduce the compounding effect of age-related hearing loss associated with the original 499 prestin KIs. Finally, a compound heterozygous (chet) mouse expressing one copy of 499 prestin and one copy of KO prestin was also created to reduce quantities of 499 prestin protein. Results show reduction in OHC death in chets, and in 499 prestin KIs on the FVB background, but only a slight improvement in OHC survival for mice receiving Protandim. We also report that improved OHC survival in 499 prestin KIs had little effect on hearing phenotype, reaffirming the original contention about the essential role of prestin’s motor function in cochlear amplification.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.