Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Hear Res ; 426: 108626, 2022 12.
Article in English | MEDLINE | ID: mdl-36228456

ABSTRACT

The human stria vascularis has been examined both by scanning and transmission electron microscopy in normal controls and from individuals who had received loop diuretics, aminoglycoside antibiotics or some combination of the two prior to their deaths. The tissues were preserved by perilymphatic perfusion of fixative within an hour of death and preservation was adequate. The normal ultrastructure is described and does not differ significantly from that found in experimental animals. The loop diuretics are associated with structural changes that cannot be distinguished from those found in animals treated with large doses of the same drugs. The aminoglycosides caused some changes, but the patients had been in renal failure and this probably contributed to the structural alterations. The combination of a loop diuretic and aminoglycoside was associated with a range of alterations from mild to severe. Overall, the three treatment groups had a series of ultrastructural changes resembling those found in animal models thereby justifying the use of experimental animals to predict human ototoxicity.


Subject(s)
Aminoglycosides , Stria Vascularis , Animals , Humans , Aminoglycosides/toxicity , Sodium Potassium Chloride Symporter Inhibitors , Anti-Bacterial Agents/pharmacology
2.
JCI Insight ; 7(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-35132964

ABSTRACT

Norrie disease is caused by mutation of the NDP gene, presenting as congenital blindness followed by later onset of hearing loss. Protecting patients from hearing loss is critical for maintaining their quality of life. This study aimed to understand the onset of pathology in cochlear structure and function. By investigating patients and juvenile Ndp-mutant mice, we elucidated the sequence of onset of physiological changes (in auditory brainstem responses, distortion product otoacoustic emissions, endocochlear potential, blood-labyrinth barrier integrity) and determined the cellular, histological, and ultrastructural events leading to hearing loss. We found that cochlear vascular pathology occurs earlier than previously reported and precedes sensorineural hearing loss. The work defines a disease mechanism whereby early malformation of the cochlear microvasculature precedes loss of vessel integrity and decline of endocochlear potential, leading to hearing loss and hair cell death while sparing spiral ganglion cells. This provides essential information on events defining the optimal therapeutic window and indicates that early intervention is needed. In an era of advancing gene therapy and small-molecule technologies, this study establishes Ndp-mutant mice as a platform to test such interventions and has important implications for understanding the progression of hearing loss in Norrie disease.


Subject(s)
Blindness/congenital , Disease Management , Evoked Potentials, Auditory, Brain Stem/physiology , Forecasting , Genetic Diseases, X-Linked/physiopathology , Hearing Loss, Sensorineural/physiopathology , Hearing/physiology , Nervous System Diseases/physiopathology , Retinal Degeneration/physiopathology , Spasms, Infantile/physiopathology , Adolescent , Adult , Animals , Blindness/complications , Blindness/physiopathology , Blindness/therapy , Child , Child, Preschool , Disease Models, Animal , Female , Follow-Up Studies , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/therapy , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/etiology , Humans , Male , Mice , Mice, Mutant Strains , Nervous System Diseases/complications , Nervous System Diseases/therapy , Retinal Degeneration/complications , Retinal Degeneration/therapy , Spasms, Infantile/complications , Spasms, Infantile/therapy , Young Adult
3.
J Neurosci Res ; 98(9): 1745-1763, 2020 09.
Article in English | MEDLINE | ID: mdl-31762086

ABSTRACT

The aging cochlea is subjected to a number of pathological changes to play a role in the onset of age-related hearing loss (ARHL). Although ARHL has often been thought of as the result of the loss of hair cells, it is in fact a disorder with a complex etiology, arising from the changes to both the organ of Corti and its supporting structures. In this study, we examine two aging pathologies that have not been studied in detail despite their apparent prevalence; the fusion, elongation, and engulfment of cochlear inner hair cell stereocilia, and the changes that occur to the tectorial membrane (TM), a structure overlying the organ of Corti that modulates its physical properties in response to sound. Our work demonstrates that similar pathological changes occur in these two structures in the aging cochleae of both mice and humans, examines the ultrastructural changes that underlie stereocilial fusion, and identifies the lost TM components that lead to changes in membrane structure. We place these changes into the context of the wider pathology of the aging cochlea, and identify how they may be important in particular for understanding the more subtle hearing pathologies that precede auditory threshold loss in ARHL.


Subject(s)
Aging/physiology , Cochlea/pathology , Hearing Loss/etiology , Stereocilia/pathology , Tectorial Membrane/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cochlea/ultrastructure , Female , Hair Cells, Auditory , Hearing , Humans , Male , Mice , Mice, Inbred CBA , Middle Aged , Organ of Corti , Stereocilia/ultrastructure , Tectorial Membrane/physiology , Tectorial Membrane/ultrastructure
4.
Biol Open ; 8(1)2019 Jan 07.
Article in English | MEDLINE | ID: mdl-30504133

ABSTRACT

The prevalence and importance of hearing damage caused by noise levels not previously thought to cause permanent hearing impairment has become apparent in recent years. The damage to, and loss of, afferent terminals of auditory nerve fibres at the cochlear inner hair cell has been well established, but the effects of noise exposure and terminal loss on the inner hair cell are less known. Using three-dimensional structural studies in mice we have examined the consequences of afferent terminal damage on inner hair cell morphology and intracellular structure. We identified a structural phenotype in the pre-synaptic regions of these damaged hair cells that persists for four weeks after noise exposure, and demonstrates a specific dysregulation of the synaptic vesicle recycling pathway. We show evidence of a failure in regeneration of vesicles from small membrane cisterns in damaged terminals, resulting from a failure of separation of small vesicle buds from the larger cisternal membranes.

5.
Elife ; 72018 07 18.
Article in English | MEDLINE | ID: mdl-30019672

ABSTRACT

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


Subject(s)
Epithelium/physiology , Hair Cells, Vestibular/physiology , Regeneration/physiology , Adenoviridae/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelium/ultrastructure , Gene Expression Regulation , Gentamicins/adverse effects , Green Fluorescent Proteins/metabolism , Hair Cells, Vestibular/pathology , Hair Cells, Vestibular/ultrastructure , Humans , Myosin VIIa , Myosins/metabolism , Receptors, Notch/metabolism , Saccule and Utricle/physiology , Saccule and Utricle/ultrastructure , Signal Transduction , Transduction, Genetic
6.
Cell Physiol Biochem ; 47(4): 1509-1532, 2018.
Article in English | MEDLINE | ID: mdl-29940568

ABSTRACT

BACKGROUND/AIMS: From invertebrates to mammals, Gαi proteins act together with their common binding partner Gpsm2 to govern cell polarization and planar organization in virtually any polarized cell. Recently, we demonstrated that Gαi3-deficiency in pre-hearing murine cochleae pointed to a role of Gαi3 for asymmetric migration of the kinocilium as well as the orientation and shape of the stereociliary ("hair") bundle, a requirement for the progression of mature hearing. We found that the lack of Gαi3 impairs stereociliary elongation and hair bundle shape in high-frequency cochlear regions, linked to elevated hearing thresholds for high-frequency sound. How these morphological defects translate into hearing phenotypes is not clear. METHODS: Here, we studied global and conditional Gnai3 and Gnai2 mouse mutants deficient for either one or both Gαi proteins. Comparative analyses of global versus Foxg1-driven conditional mutants that mainly delete in the inner ear and telencephalon in combination with functional tests were applied to dissect essential and redundant functions of different Gαi isoforms and to assign specific defects to outer or inner hair cells, the auditory nerve, satellite cells or central auditory neurons. RESULTS: Here we report that lack of Gαi3 but not of the ubiquitously expressed Gαi2 elevates hearing threshold, accompanied by impaired hair bundle elongation and shape in high-frequency cochlear regions. During the crucial reprogramming of the immature inner hair cell (IHC) synapse into a functional sensory synapse of the mature IHC deficiency for Gαi2 or Gαi3 had no impact. In contrast, double-deficiency for Gαi2 and Gαi3 isoforms results in abnormalities along the entire tonotopic axis including profound deafness associated with stereocilia defects. In these mice, postnatal IHC synapse maturation is also impaired. In addition, the analysis of conditional versus global Gαi3-deficient mice revealed that the amplitude of ABR wave IV was disproportionally elevated in comparison to ABR wave I indicating that Gαi3 is selectively involved in generation of neural gain during auditory processing. CONCLUSION: We propose a so far unrecognized complexity of isoform-specific and overlapping Gαi protein functions particular during final differentiation processes.


Subject(s)
Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hair Cells, Auditory, Inner/metabolism , Hearing/physiology , Nerve Tissue Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Forkhead Transcription Factors/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Hair Cells, Auditory, Inner/cytology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics
8.
Nat Commun ; 8: 14907, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387217

ABSTRACT

Mutations in GPSM2 cause Chudley-McCullough syndrome (CMCS), an autosomal recessive neurological disorder characterized by early-onset sensorineural deafness and brain anomalies. Here, we show that mutation of the mouse orthologue of GPSM2 affects actin-rich stereocilia elongation in auditory and vestibular hair cells, causing deafness and balance defects. The G-protein subunit Gαi3, a well-documented partner of Gpsm2, participates in the elongation process, and its absence also causes hearing deficits. We show that Gpsm2 defines an ∼200 nm nanodomain at the tips of stereocilia and this localization requires the presence of Gαi3, myosin 15 and whirlin. Using single-molecule tracking, we report that loss of Gpsm2 leads to decreased outgrowth and a disruption of actin dynamics in neuronal growth cones. Our results elucidate the aetiology of CMCS and highlight a new molecular role for Gpsm2/Gαi3 in the regulation of actin dynamics in epithelial and neuronal tissues.


Subject(s)
Actins/metabolism , Agenesis of Corpus Callosum/genetics , Arachnoid Cysts/genetics , Carrier Proteins/genetics , Growth Cones/metabolism , Hair Cells, Auditory/metabolism , Hair Cells, Vestibular/metabolism , Hearing Loss, Sensorineural/genetics , Neurons/metabolism , Stereocilia/metabolism , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/physiopathology , Animals , Arachnoid Cysts/metabolism , Arachnoid Cysts/physiopathology , Cell Cycle Proteins , Deafness/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation , Myosins/metabolism , Postural Balance , Sensation Disorders/genetics
9.
PLoS Genet ; 13(3): e1006692, 2017 03.
Article in English | MEDLINE | ID: mdl-28346477

ABSTRACT

Behavioural anomalies suggesting an inner ear disorder were observed in a colony of transgenic mice. Affected animals were profoundly deaf. Severe hair bundle defects were identified in all outer and inner hair cells (OHC, IHC) in the cochlea and in hair cells of vestibular macular organs, but hair cells in cristae were essentially unaffected. Evidence suggested the disorder was likely due to gene disruption by a randomly inserted transgene construct. Whole-genome sequencing identified interruption of the SorCS2 (Sortilin-related VPS-10 domain containing protein) locus. Real-time-qPCR demonstrated disrupted expression of SorCS2 RNA in cochlear tissue from affected mice and this was confirmed by SorCS2 immuno-labelling. In all affected hair cells, stereocilia were shorter than normal, but abnormalities of bundle morphology and organisation differed between hair cell types. Bundles on OHC were grossly misshapen with significantly fewer stereocilia than normal. However, stereocilia were organised in rows of increasing height. Bundles on IHC contained significantly more stereocilia than normal with some longer stereocilia towards the centre, or with minimal height differentials. In early postnatal mice, kinocilia (primary cilia) of IHC and of OHC were initially located towards the lateral edge of the hair cell surface but often became surrounded by stereocilia as bundle shape and apical surface contour changed. In macular organs the kinocilium was positioned in the centre of the cell surface throughout maturation. There was disruption of the signalling pathway controlling intrinsic hair cell apical asymmetry. LGN and Gαi3 were largely absent, and atypical Protein Kinase C (aPKC) lost its asymmetric distribution. The results suggest that SorCS2 plays a role upstream of the intrinsic polarity pathway and that there are differences between hair cell types in the deployment of the machinery that generates a precisely organised hair bundle.


Subject(s)
Gene Expression Regulation , Hair Cells, Auditory, Inner/metabolism , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/genetics , Stereocilia/genetics , Age Factors , Animals , Hair Cells, Auditory, Inner/pathology , Hearing Loss/genetics , Hearing Loss/metabolism , Hearing Loss/physiopathology , Immunohistochemistry , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Scanning , Nerve Tissue Proteins/metabolism , Organ of Corti/metabolism , Organ of Corti/physiopathology , Organ of Corti/ultrastructure , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stereocilia/metabolism , Stereocilia/pathology
10.
Nat Biotechnol ; 35(3): 280-284, 2017 03.
Article in English | MEDLINE | ID: mdl-28165475

ABSTRACT

Efforts to develop gene therapies for hearing loss have been hampered by the lack of safe, efficient, and clinically relevant delivery modalities. Here we demonstrate the safety and efficiency of Anc80L65, a rationally designed synthetic vector, for transgene delivery to the mouse cochlea. Ex vivo transduction of mouse organotypic explants identified Anc80L65 from a set of other adeno-associated virus (AAV) vectors as a potent vector for the cochlear cell targets. Round window membrane injection resulted in highly efficient transduction of inner and outer hair cells in mice, a substantial improvement over conventional AAV vectors. Anc80L65 round window injection was well tolerated, as indicated by sensory cell function, hearing and vestibular function, and immunologic parameters. The ability of Anc80L65 to target outer hair cells at high rates, a requirement for restoration of complex auditory function, may enable future gene therapies for hearing and balance disorders.


Subject(s)
Cochlea/physiology , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Plasmids/genetics , Transduction, Genetic/methods , Animals , Cochlea/virology , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage
11.
J Cell Sci ; 128(14): 2529-40, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26045447

ABSTRACT

The ways in which cell architecture is modelled to meet cell function is a poorly understood facet of cell biology. To address this question, we have studied the cytoarchitecture of a cell with highly specialised organisation, the cochlear inner hair cell (IHC), using multiple hierarchies of three-dimensional (3D) electron microscopy analyses. We show that synaptic terminal distribution on the IHC surface correlates with cell shape, and the distribution of a highly organised network of membranes and mitochondria encompassing the infranuclear region of the cell. This network is juxtaposed to a population of small vesicles, which represents a potential new source of neurotransmitter vesicles for replenishment of the synapses. Structural linkages between organelles that underlie this organisation were identified by high-resolution imaging. Taken together, these results describe a cell-encompassing network of membranes and mitochondria present in IHCs that support efficient coding and transmission of auditory signals. Such techniques also have the potential for clarifying functionally specialised cytoarchitecture of other cell types.


Subject(s)
Hair Cells, Auditory, Inner/ultrastructure , Imaging, Three-Dimensional , Synaptic Vesicles/ultrastructure , Animals , Guinea Pigs , Hair Cells, Auditory, Inner/metabolism , Mice , Microscopy, Electron , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism
12.
Neurobiol Aging ; 36(6): 2068-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818177

ABSTRACT

Balance disequilibrium is a significant contributor to falls in the elderly. The most common cause of balance dysfunction is loss of sensory cells from the vestibular sensory epithelia of the inner ear. However, inaccessibility of inner ear tissue in humans severely restricts possibilities for experimental manipulation to develop therapies to ameliorate this loss. We provide a structural and functional analysis of human vestibular sensory epithelia harvested at trans-labyrinthine surgery. We demonstrate the viability of the tissue and labeling with specific markers of hair cell function and of ion homeostasis in the epithelium. Samples obtained from the oldest patients revealed a significant loss of hair cells across the tissue surface, but we found immature hair bundles present in epithelia harvested from patients >60 years of age. These results suggest that the environment of the human vestibular sensory epithelium could be responsive to stimulation of developmental pathways to enhance hair cell regeneration, as has been demonstrated successfully in the vestibular organs of adult mice.


Subject(s)
Aging/pathology , Hair Cells, Vestibular/pathology , Vestibule, Labyrinth/cytology , Vestibule, Labyrinth/pathology , Aged , Animals , Cell Survival , Cells, Cultured , Epithelium/pathology , Epithelium/physiology , Hair Cells, Vestibular/physiology , Humans , Mice, Inbred C57BL , Mice, Inbred CBA , Nerve Regeneration , Regenerative Medicine , Stereocilia , Vestibule, Labyrinth/physiology
13.
Hum Mol Genet ; 24(3): 609-24, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25217574

ABSTRACT

In the mammalian inner ear, bicellular and tricellular tight junctions (tTJs) seal the paracellular space between epithelial cells. Tricellulin and immunoglobulin-like (Ig-like) domain containing receptor 1 (ILDR1, also referred to as angulin-2) localize to tTJs of the sensory and non-sensory epithelia in the organ of Corti and vestibular end organs. Recessive mutations of TRIC (DFNB49) encoding tricellulin and ILDR1 (DFNB42) cause human nonsyndromic deafness. However, the pathophysiology of DFNB42 deafness remains unknown. ILDR1 was recently reported to be a lipoprotein receptor mediating the secretion of the fat-stimulated cholecystokinin (CCK) hormone in the small intestine, while ILDR1 in EpH4 mouse mammary epithelial cells in vitro was shown to recruit tricellulin to tTJs. Here we show that two different mouse Ildr1 mutant alleles have early-onset severe deafness associated with a rapid degeneration of cochlear hair cells (HCs) but have a normal endocochlear potential. ILDR1 is not required for recruitment of tricellulin to tTJs in the cochlea in vivo; however, tricellulin becomes mislocalized in the inner ear sensory epithelia of ILDR1 null mice after the first postnatal week. As revealed by freeze-fracture electron microscopy, ILDR1 contributes to the ultrastructure of inner ear tTJs. Taken together, our data provide insight into the pathophysiology of human DFNB42 deafness and demonstrate that ILDR1 is crucial for normal hearing by maintaining the structural and functional integrity of tTJs, which are critical for the survival of auditory neurosensory HCs.


Subject(s)
Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/pathology , Receptors, Cell Surface/genetics , Tight Junctions/pathology , Animals , Disease Models, Animal , Hair Cells, Auditory/metabolism , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Humans , MARVEL Domain Containing 2 Protein/metabolism , Mice , Mutation , Receptors, Cell Surface/metabolism , Tight Junctions/metabolism
14.
Hum Mol Genet ; 24(1): 37-49, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25124451

ABSTRACT

Hearing relies on the mechanosensory inner and outer hair cells (OHCs) of the organ of Corti, which convert mechanical deflections of their actin-rich stereociliary bundles into electrochemical signals. Several actin-associated proteins are essential for stereocilia formation and maintenance, and their absence leads to deafness. One of the most abundant actin-bundling proteins of stereocilia is plastin 1, but its function has never been directly assessed. Here, we found that plastin 1 knock-out (Pls1 KO) mice have a moderate and progressive form of hearing loss across all frequencies. Auditory hair cells developed normally in Pls1 KO, but in young adult animals, the stereocilia of inner hair cells were reduced in width and length. The stereocilia of OHCs were comparatively less affected; however, they also showed signs of degeneration in ageing mice. The hair bundle stiffness and the acquisition of the electrophysiological properties of hair cells were unaffected by the absence of plastin 1, except for a significant change in the adaptation properties, but not the size of the mechanoelectrical transducer currents. These results show that in contrast to other actin-bundling proteins such as espin, harmonin or Eps8, plastin 1 is dispensable for the initial formation of stereocilia. However, the progressive hearing loss and morphological defects of hair cells in adult Pls1 KO mice point at a specific role for plastin 1 in the preservation of adult stereocilia and optimal hearing. Hence, mutations in the human PLS1 gene may be associated with relatively mild and progressive forms of hearing loss.


Subject(s)
Hair Cells, Auditory, Inner/pathology , Hearing Loss/physiopathology , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Stereocilia/pathology , Age Factors , Animals , Hearing Loss/genetics , Hearing Loss/pathology , Humans , Mice , Mice, Knockout , Mutation
15.
Cell Tissue Res ; 360(3): 633-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25381570

ABSTRACT

Normal development, function and repair of the sensory epithelia in the inner ear are all dependent on gap junctional intercellular communication. Mutations in the connexin genes GJB2 and GJB6 (encoding CX26 and CX30) result in syndromic and non-syndromic deafness via various mechanisms. Clinical vestibular defects, however, are harder to connect with connexin dysfunction. Cx26 and Cx30 proteins are widely expressed in the epithelial and connective tissues of the cochlea, where they may form homomeric or heteromeric gap junction channels in a cell-specific and spatiotemporally complex fashion. Despite the study of mutant channels and animal models for both recessive and dominant autosomal deafness, it is still unclear why gap junctions are essential for auditory function, and why Cx26 and Cx30 do not compensate for each other in vivo. Cx26 appears to be essential for normal development of the auditory sensory epithelium, but may be dispensable during normal hearing. Cx30 appears to be essential for normal repair following sensory cell loss. The specific modes of intercellular signalling mediated by inner ear gap junction channels remain undetermined, but they are hypothesised to play essential roles in the maintenance of ionic and metabolic homeostasis in the inner ear. Recent studies have highlighted involvement of gap junctions in the transfer of essential second messengers between the non-sensory cells, and have proposed roles for hemichannels in normal hearing. Here, we summarise the current knowledge about the molecular and functional properties of inner ear gap junctions, and about tissue pathologies associated with connexin mutations.


Subject(s)
Connexins/metabolism , Ear, Inner/metabolism , Gap Junctions/metabolism , Potassium/metabolism , Animals , Biophysical Phenomena , Connexin 26 , Connexins/genetics , Homeostasis , Humans
16.
J Neurosci ; 34(48): 15851-60, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25429127

ABSTRACT

The loss of auditory hair cells triggers repair responses within the population of nonsensory supporting cells. When hair cells are irreversibly lost from the mammalian cochlea, supporting cells expand to fill the resulting lesions in the sensory epithelium, an initial repair process that is dependent on gap junctional intercellular communication (GJIC). In the chicken cochlea (the basilar papilla or BP), dying hair cells are extruded from the epithelium and supporting cells expand to fill the lesions and then replace hair cells via mitotic and/or conversion mechanisms. Here, we investigated the involvement of GJIC in the initial epithelial repair process in the aminoglycoside-damaged BP. Gentamicin-induced hair cell loss was associated with a decrease of chicken connexin43 (cCx43) immunofluorescence, yet cCx30-labeled gap junction plaques remained. Fluorescence recovery after photobleaching experiments confirmed that the GJIC remained robust in gentamicin-damaged explants, but regionally asymmetric coupling was no longer evident. Dye injections in slice preparations from undamaged BP explants identified cell types with characteristic morphologies along the neural-abneural axis, but these were electrophysiologically indistinct. In gentamicin-damaged BP, supporting cells expanded to fill space formerly occupied by hair cells and displayed more variable electrophysiological phenotypes. When GJIC was inhibited during the aminoglycoside damage paradigm, the epithelial repair response halted. Dying hair cells were retained within the sensory epithelium and supporting cells remained unexpanded. These observations suggest that repair of the auditory epithelium shares common mechanisms across vertebrate species and emphasize the importance of functional gap junctions in maintaining a homeostatic environment permissive for subsequent hair cell regeneration.


Subject(s)
Cell Communication/physiology , Gap Junctions/pathology , Gap Junctions/physiology , Hair Cells, Auditory/pathology , Hair Cells, Auditory/physiology , Animals , Birds , Cells, Cultured , Chickens , Cochlea/pathology , Cochlea/physiology , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , HeLa Cells , Humans , Male , Organ Culture Techniques
17.
PLoS Genet ; 10(10): e1004688, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25356849

ABSTRACT

Spinster homolog 2 (Spns2) acts as a Sphingosine-1-phosphate (S1P) transporter in zebrafish and mice, regulating heart development and lymphocyte trafficking respectively. S1P is a biologically active lysophospholipid with multiple roles in signalling. The mechanism of action of Spns2 is still elusive in mammals. Here, we report that Spns2-deficient mice rapidly lost auditory sensitivity and endocochlear potential (EP) from 2 to 3 weeks old. We found progressive degeneration of sensory hair cells in the organ of Corti, but the earliest defect was a decline in the EP, suggesting that dysfunction of the lateral wall was the primary lesion. In the lateral wall of adult mutants, we observed structural changes of marginal cell boundaries and of strial capillaries, and reduced expression of several key proteins involved in the generation of the EP (Kcnj10, Kcnq1, Gjb2 and Gjb6), but these changes were likely to be secondary. Permeability of the boundaries of the stria vascularis and of the strial capillaries appeared normal. We also found focal retinal degeneration and anomalies of retinal capillaries together with anterior eye defects in Spns2 mutant mice. Targeted inactivation of Spns2 in red blood cells, platelets, or lymphatic or vascular endothelial cells did not affect hearing, but targeted ablation of Spns2 in the cochlea using a Sox10-Cre allele produced a similar auditory phenotype to the original mutation, suggesting that local Spns2 expression is critical for hearing in mammals. These findings indicate that Spns2 is required for normal maintenance of the EP and hence for normal auditory function, and support a role for S1P signalling in hearing.


Subject(s)
Anion Transport Proteins/genetics , Cochlea/pathology , Ear, Inner/pathology , Hearing Loss/genetics , Age of Onset , Animals , Anion Transport Proteins/deficiency , Anion Transport Proteins/metabolism , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Cochlea/metabolism , Connexin 26 , Connexins , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss/metabolism , Hearing Loss/pathology , Lysophospholipids/metabolism , Mice , Organogenesis/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Stria Vascularis/pathology , Zebrafish
18.
PLoS One ; 8(11): e80104, 2013.
Article in English | MEDLINE | ID: mdl-24244619

ABSTRACT

22q11.2 Deletion Syndrome (22q11DS) arises from an interstitial chromosomal microdeletion encompassing at least 30 genes. This disorder is one of the most significant known cytogenetic risk factors for schizophrenia, and can also cause heart abnormalities, cognitive deficits, hearing difficulties, and a variety of other medical problems. The Df1/+ hemizygous knockout mouse, a model for human 22q11DS, recapitulates many of the deficits observed in the human syndrome including heart defects, impaired memory, and abnormal auditory sensorimotor gating. Here we show that Df1/+ mice, like human 22q11DS patients, have substantial rates of hearing loss arising from chronic middle ear infection. Auditory brainstem response (ABR) measurements revealed significant elevation of click-response thresholds in 48% of Df1/+ mice, often in only one ear. Anatomical and histological analysis of the middle ear demonstrated no gross structural abnormalities, but frequent signs of otitis media (OM, chronic inflammation of the middle ear), including excessive effusion and thickened mucosa. In mice for which both in vivo ABR thresholds and post mortem middle-ear histology were obtained, the severity of signs of OM correlated directly with the level of hearing impairment. These results suggest that abnormal auditory sensorimotor gating previously reported in mouse models of 22q11DS could arise from abnormalities in auditory processing. Furthermore, the findings indicate that Df1/+ mice are an excellent model for increased risk of OM in human 22q11DS patients. Given the frequently monaural nature of OM in Df1/+ mice, these animals could also be a powerful tool for investigating the interplay between genetic and environmental causes of OM.


Subject(s)
DiGeorge Syndrome/genetics , Ear, Middle/physiopathology , Hearing Loss/genetics , Otitis Media with Effusion/genetics , Animals , Auditory Threshold , DiGeorge Syndrome/complications , DiGeorge Syndrome/microbiology , DiGeorge Syndrome/physiopathology , Disease Models, Animal , Ear, Middle/microbiology , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Evoked Potentials, Auditory, Brain Stem , Female , Gene-Environment Interaction , Hearing Loss/complications , Hearing Loss/microbiology , Hearing Loss/physiopathology , Hemizygote , Humans , Lactococcus/growth & development , Lactococcus/isolation & purification , Male , Mice , Otitis Media with Effusion/complications , Otitis Media with Effusion/microbiology , Otitis Media with Effusion/physiopathology , Pantoea/growth & development , Pantoea/isolation & purification , Severity of Illness Index
19.
J Clin Invest ; 123(9): 4036-49, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23979167

ABSTRACT

The two compositionally distinct extracellular cochlear fluids, endolymph and perilymph, are separated by tight junctions that outline the scala media and reticular lamina. Mutations in TRIC (also known as MARVELD2), which encodes a tricellular tight junction protein known as tricellulin, lead to nonsyndromic hearing loss (DFNB49). We generated a knockin mouse that carries a mutation orthologous to the TRIC coding mutation linked to DFNB49 hearing loss in humans. Tricellulin was absent from the tricellular junctions in the inner ear epithelia of the mutant animals, which developed rapidly progressing hearing loss accompanied by loss of mechanosensory cochlear hair cells, while the endocochlear potential and paracellular permeability of a biotin-based tracer in the stria vascularis were unaltered. Freeze-fracture electron microscopy revealed disruption of the strands of intramembrane particles connecting bicellular and tricellular junctions in the inner ear epithelia of tricellulin-deficient mice. These ultrastructural changes may selectively affect the paracellular permeability of ions or small molecules, resulting in a toxic microenvironment for cochlear hair cells. Consistent with this hypothesis, hair cell loss was rescued in tricellulin-deficient mice when generation of normal endolymph was inhibited by a concomitant deletion of the transcription factor, Pou3f4. Finally, comprehensive phenotypic screening showed a broader pathological phenotype in the mutant mice, which highlights the non-redundant roles played by tricellulin.


Subject(s)
Hair Cells, Auditory, Outer/metabolism , Hearing Loss/metabolism , MARVEL Domain Containing 2 Protein/deficiency , Tight Junctions/metabolism , Animals , Female , Hearing Loss/pathology , MARVEL Domain Containing 2 Protein/genetics , Male , Membrane Potentials , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Mutation, Missense , Organ of Corti/pathology , Stria Vascularis/metabolism , Stria Vascularis/pathology , Vestibule, Labyrinth/metabolism , Vestibule, Labyrinth/pathology
20.
J Cell Sci ; 126(Pt 7): 1703-12, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23424196

ABSTRACT

A role for connexin (Cx)30 in epithelial repair following injury was examined in the organ of Corti, the sensory epithelium of the cochlea. In this tissue, lesions caused by loss of the sensory hair cells are closed by the supporting cells that surround each one. Gap junctions in which Cx30 is the predominant connexin are large and numerous between supporting cells. In mice carrying a deletion in the gene (Gjb6) that encodes Cx30, the size and number of gap junction plaques, and the extent of dye transfer, between supporting cells was greatly reduced compared with normal animals. This corresponded with unique peculiarities of the lesion closure events during the progressive hair cell loss that occurs in these animals in comparison with other models of hair cell loss, whether acquired or as a result of a mutation. Only one, rather than all, of the supporting cells that contacted an individual dying hair closed the lesion, indicating disturbance of the co-ordination of cellular responses. The cell shape changes that the supporting cells normally undergo during repair of the organ of Corti did not occur. Also, there was disruption of the migratory activities that normally lead to the replacement of a columnar epithelium with a squamous-like one. These observations demonstrate a role for Cx30 and intercellular communication in regulating repair responses in an epithelial tissue.


Subject(s)
Cochlea/metabolism , Connexins/metabolism , Animals , Cell Communication/genetics , Cell Communication/physiology , Cochlea/ultrastructure , Connexin 30 , Connexins/genetics , Gap Junctions/metabolism , Gap Junctions/ultrastructure , In Vitro Techniques , Mice , Mice, Knockout , Microscopy, Electron , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...