Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 163(4): 894-906, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544938

ABSTRACT

A deficiency in pejvakin, a protein of unknown function, causes a strikingly heterogeneous form of human deafness. Pejvakin-deficient (Pjvk(-/-)) mice also exhibit variable auditory phenotypes. Correlation between their hearing thresholds and the number of pups per cage suggest a possible harmful effect of pup vocalizations. Direct sound or electrical stimulation show that the cochlear sensory hair cells and auditory pathway neurons of Pjvk(-/-) mice and patients are exceptionally vulnerable to sound. Subcellular analysis revealed that pejvakin is associated with peroxisomes and required for their oxidative-stress-induced proliferation. Pjvk(-/-) cochleas display features of marked oxidative stress and impaired antioxidant defenses, and peroxisomes in Pjvk(-/-) hair cells show structural abnormalities after the onset of hearing. Noise exposure rapidly upregulates Pjvk cochlear transcription in wild-type mice and triggers peroxisome proliferation in hair cells and primary auditory neurons. Our results reveal that the antioxidant activity of peroxisomes protects the auditory system against noise-induced damage.


Subject(s)
Hearing Loss, Noise-Induced/metabolism , Nerve Tissue Proteins/metabolism , Peroxisomes/metabolism , Proteins/metabolism , Animals , Auditory Pathways , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/pathology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , Oxidative Stress , Proteins/genetics
2.
Glia ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982826

ABSTRACT

During brain maturation, astrocytes establish complex morphologies unveiling intense structural plasticity. Connexin 30 (Cx30), a gap-junction channel-forming protein expressed postnatally, dynamically regulates during development astrocyte morphological properties by controlling ramification and extension of fine processes. However, the underlying mechanisms remain unexplored. Here, we found in vitro that Cx30 interacts with the actin cytoskeleton in astrocytes and inhibits its structural reorganization and dynamics during cell migration. This translates into an alteration of local physical surface properties, as assessed by correlative imaging using stimulated emission depletion (STED) super resolution imaging and atomic force microscopy (AFM). Specifically, Cx30 impaired astrocyte cell surface topology and cortical stiffness in motile astrocytes. As Cx30 alters actin organization, dynamics, and membrane physical properties, we assessed whether it controls astrocyte migration. We found that Cx30 reduced persistence and directionality of migrating astrocytes. Altogether, these data reveal Cx30 as a brake for astrocyte structural and mechanical plasticity.

3.
Proc Natl Acad Sci U S A ; 116(16): 8010-8017, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30936319

ABSTRACT

Noise overexposure causes oxidative stress, leading to auditory hair cell damage. Adaptive peroxisome proliferation involving pejvakin, a peroxisome-associated protein from the gasdermin family, has been shown to protect against this harmful oxidative stress. However, the role of pejvakin in peroxisome dynamics and homeostasis remains unclear. Here we show that sound overstimulation induces an early and rapid selective autophagic degradation of peroxisomes (pexophagy) in auditory hair cells from wild-type, but not pejvakin-deficient (Pjvk-/-), mice. Noise overexposure triggers recruitment of the autophagosome-associated protein MAP1LC3B (LC3B; microtubule-associated protein 1 light chain 3ß) to peroxisomes in wild-type, but not Pjvk-/-, mice. We also show that pejvakin-LC3B binding involves an LC3-interacting region within the predicted chaperone domain of pejvakin. In transfected cells and in vivo transduced auditory hair cells, cysteine mutagenesis experiments demonstrated the requirement for both C328 and C343, the two cysteine residues closest to the C terminus of pejvakin, for reactive oxygen species-induced pejvakin-LC3B interaction and pexophagy. The viral transduction of auditory hair cells from Pjvk-/- mice in vivo with both Pjvk and Lc3b cDNAs completely restored sound-induced pexophagy, fully prevented the development of oxidative stress, and resulted in normal levels of peroxisome proliferation, whereas Pjvk cDNA alone yielded only a partial correction of the defects. Overall, our results demonstrate that pexophagy plays a key role in noise-induced peroxisome proliferation and identify defective pexophagy as a cause of noise-induced hearing loss. They suggest that pejvakin acts as a redox-activated pexophagy receptor/adaptor, thereby identifying a previously unknown function of gasdermin family proteins.


Subject(s)
Hair Cells, Auditory , Hearing Loss, Noise-Induced , Macroautophagy/physiology , Proteins , Animals , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/physiology , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Noise-Induced/prevention & control , Mice , Microtubule-Associated Proteins/metabolism , Proteins/chemistry , Proteins/genetics , Proteins/metabolism
4.
Am J Hum Genet ; 98(6): 1266-1270, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259055

ABSTRACT

By genetic linkage analysis in a large consanguineous Iranian family with eleven individuals affected by severe to profound congenital deafness, we were able to define a 2.8 Mb critical interval (at chromosome 1p21.2-1p21.1) for an autosomal-recessive nonsyndromic deafness locus (DFNB). Whole-exome sequencing allowed us to identify a CDC14A biallelic nonsense mutation, c.1126C>T (p.Arg376(∗)), which was present in the eight clinically affected individuals still alive. Subsequent screening of 115 unrelated individuals affected by severe or profound congenital deafness of unknown genetic cause led us to identify another CDC14A biallelic nonsense mutation, c.1015C>T (p.Arg339(∗)), in an individual originating from Mauritania. CDC14A encodes a protein tyrosine phosphatase. Immunofluorescence analysis of the protein distribution in the mouse inner ear showed a strong labeling of the hair cells' kinocilia. By using a morpholino strategy to knockdown cdc14a in zebrafish larvae, we found that the length of the kinocilia was reduced in inner-ear hair cells. Therefore, deafness caused by loss-of-function mutations in CDC14A probably arises from a morphogenetic defect of the auditory sensory cells' hair bundles, whose differentiation critically depends on the proper growth of their kinocilium.


Subject(s)
Cilia/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/etiology , Mutation/genetics , Phosphoric Monoester Hydrolases/genetics , Severity of Illness Index , Adult , Aged , Animals , Cilia/metabolism , Female , Fluorescent Antibody Technique , Hair Cells, Auditory/enzymology , Hearing Loss, Sensorineural/pathology , Humans , Larva/genetics , Larva/growth & development , Male , Mice , Middle Aged , Pedigree , Protein Tyrosine Phosphatases , Young Adult , Zebrafish/genetics , Zebrafish/growth & development
5.
Proc Natl Acad Sci U S A ; 111(25): 9307-12, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24920589

ABSTRACT

A detrimental perceptive consequence of damaged auditory sensory hair cells consists in a pronounced masking effect exerted by low-frequency sounds, thought to occur when auditory threshold elevation substantially exceeds 40 dB. Here, we identified the submembrane scaffold protein Nherf1 as a hair-bundle component of the differentiating outer hair cells (OHCs). Nherf1(-/-) mice displayed OHC hair-bundle shape anomalies in the mid and basal cochlea, normally tuned to mid- and high-frequency tones, and mild (22-35 dB) hearing-threshold elevations restricted to midhigh sound frequencies. This mild decrease in hearing sensitivity was, however, discordant with almost nonresponding OHCs at the cochlear base as assessed by distortion-product otoacoustic emissions and cochlear microphonic potentials. Moreover, unlike wild-type mice, responses of Nherf1(-/-) mice to high-frequency (20-40 kHz) test tones were not masked by tones of neighboring frequencies. Instead, efficient maskers were characterized by their frequencies up to two octaves below the probe-tone frequency, unusually low intensities up to 25 dB below probe-tone level, and growth-of-masker slope (2.2 dB/dB) reflecting their compressive amplification. Together, these properties do not fit the current acknowledged features of a hypersensitivity of the basal cochlea to lower frequencies, but rather suggest a previously unidentified mechanism. Low-frequency maskers, we propose, may interact within the unaffected cochlear apical region with midhigh frequency sounds propagated there via a mode possibly using the persistent contact of misshaped OHC hair bundles with the tectorial membrane. Our findings thus reveal a source of misleading interpretations of hearing thresholds and of hypervulnerability to low-frequency sound interference.


Subject(s)
Auditory Perception/physiology , Hair Cells, Auditory, Outer/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sound , Animals , Hair Cells, Auditory, Outer/cytology , Mice , Mice, Knockout , Phosphoproteins/genetics , Sodium-Hydrogen Exchangers/genetics
6.
Proc Natl Acad Sci U S A ; 108(14): 5825-30, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21436032

ABSTRACT

The mechanotransducer channels of auditory hair cells are gated by tip-links, oblique filaments that interconnect the stereocilia of the hair bundle. Tip-links stretch from the tips of stereocilia in the short and middle rows to the sides of neighboring, taller stereocilia. They are made of cadherin-23 and protocadherin-15, products of the Usher syndrome type 1 genes USH1D and USH1F, respectively. In this study we address the role of sans, a putative scaffold protein and product of the USH1G gene. In Ush1g(-/-) mice, the cohesion of stereocilia is disrupted, and both the amplitude and the sensitivity of the transduction currents are reduced. In Ush1g(fl/fl)Myo15-cre(+/-) mice, the loss of sans occurs postnatally and the stereocilia remain cohesive. In these mice, there is a decrease in the amplitude of the total transducer current with no loss in sensitivity, and the tips of the stereocilia in the short and middle rows lose their prolate shape, features that can be attributed to the loss of tip-links. Furthermore, stereocilia from these rows undergo a dramatic reduction in length, suggesting that the mechanotransduction machinery has a positive effect on F-actin polymerization. Sans interacts with the cytoplasmic domains of cadherin-23 and protocadherin-15 in vitro and is absent from the hair bundle in mice defective for either of the two cadherins. Because sans localizes mainly to the tips of short- and middle-row stereocilia in vivo, we conclude that it belongs to a molecular complex at the lower end of the tip-link and plays a critical role in the maintenance of this link.


Subject(s)
Actins/metabolism , Hair Cells, Auditory/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Cadherin Related Proteins , Cadherins/metabolism , Cilia/metabolism , Electrophysiology , Fluorescent Antibody Technique , Genetic Vectors/genetics , Hair Cells, Auditory/ultrastructure , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Nerve Tissue Proteins/genetics , Polymerization , Protein Precursors/metabolism , Signal Transduction/genetics
7.
Nat Genet ; 34(4): 421-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12833159

ABSTRACT

The whirler mouse mutant (wi) does not respond to sound stimuli, and detailed ultrastructural analysis of sensory hair cells in the organ of Corti of the inner ear indicates that the whirler gene encodes a protein involved in the elongation and maintenance of stereocilia in both inner hair cells (IHCs) and outer hair cells (OHCs). BAC-mediated transgene correction of the mouse phenotype and mutation analysis identified the causative gene as encoding a novel PDZ protein called whirlin. The gene encoding whirlin also underlies the human autosomal recessive deafness locus DFNB31. In the mouse cochlea, whirlin is expressed in the sensory IHC and OHC stereocilia. Our findings suggest that this novel PDZ domain-containing molecule acts as an organizer of submembranous molecular complexes that control the coordinated actin polymerization and membrane growth of stereocilia.


Subject(s)
Deafness/genetics , Gene Expression , Membrane Proteins/genetics , Proteins/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cilia/physiology , Cilia/ultrastructure , DNA Mutational Analysis , DNA, Complementary/genetics , Genes, Recessive , Hair Cells, Auditory, Inner/ultrastructure , Hair Cells, Auditory, Outer/ultrastructure , Humans , Membrane Proteins/physiology , Mice , Mice, Mutant Strains , Mice, Transgenic , Molecular Sequence Data , Phenotype , Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Species Specificity
8.
Nat Commun ; 13(1): 4528, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35953476

ABSTRACT

Pten is one of the most frequently mutated tumour suppressor gene in cancer. PTEN is generally altered in invasive cancers such as glioblastomas, but its function in collective cell migration and invasion is not fully characterised. Herein, we report that the loss of PTEN increases cell speed during collective migration of non-tumourous cells both in vitro and in vivo. We further show that loss of PTEN promotes LKB1-dependent phosphorylation and activation of the major metabolic regulator AMPK. In turn AMPK increases VASP phosphorylation, reduces VASP localisation at cell-cell junctions and decreases the interjunctional transverse actin arcs at the leading front, provoking a weakening of cell-cell contacts and increasing migration speed. Targeting AMPK activity not only slows down PTEN-depleted cells, it also limits PTEN-null glioblastoma cell invasion, opening new opportunities to treat glioblastoma lethal invasiveness.


Subject(s)
AMP-Activated Protein Kinases , Glioblastoma , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Cell Movement , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplasm Invasiveness , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation
9.
Sci Rep ; 10(1): 16430, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009420

ABSTRACT

The hair bundle of cochlear hair cells is the site of auditory mechanoelectrical transduction. It is formed by three rows of stiff microvilli-like protrusions of graduated heights, the short, middle-sized, and tall stereocilia. In developing and mature sensory hair cells, stereocilia are connected to each other by various types of fibrous links. Two unconventional cadherins, protocadherin-15 (PCDH15) and cadherin-23 (CDH23), form the tip-links, whose tension gates the hair cell mechanoelectrical transduction channels. These proteins also form transient lateral links connecting neighboring stereocilia during hair bundle morphogenesis. The proteins involved in anchoring these diverse links to the stereocilia dense actin cytoskeleton remain largely unknown. We show that the long isoform of whirlin (L-whirlin), a PDZ domain-containing submembrane scaffold protein, is present at the tips of the tall stereocilia in mature hair cells, together with PCDH15 isoforms CD1 and CD2; L-whirlin localization to the ankle-link region in developing hair bundles moreover depends on the presence of PCDH15-CD1 also localizing there. We further demonstrate that L-whirlin binds to PCDH15 and CDH23 with moderate-to-high affinities in vitro. From these results, we suggest that L-whirlin is part of the molecular complexes bridging PCDH15-, and possibly CDH23-containing lateral links to the cytoskeleton in immature and mature stereocilia.


Subject(s)
Cadherins/metabolism , Cochlea/metabolism , Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Protein Precursors/metabolism , Animals , Cadherin Related Proteins , Cell Differentiation/physiology , Female , Male , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning/methods , Protein Isoforms/metabolism , Stereocilia/metabolism
10.
Curr Biol ; 12(13): 1106-11, 2002 Jul 09.
Article in English | MEDLINE | ID: mdl-12121617

ABSTRACT

Mutations in the gene encoding the gap junction protein connexin26 (Cx26) are responsible for the autosomal recessive isolated deafness, DFNB1, which accounts for half of the cases of prelingual profound hereditary deafness in Caucasian populations. To date, in vivo approaches to decipher the role of Cx26 in the inner ear have been hampered by the embryonic lethality of the Cx26 knockout mice. To overcome this difficulty, we performed targeted ablation of Cx26 specifically in one of the two cellular networks that it underlies in the inner ear, namely, the epithelial network. We show that homozygous mutant mice, Cx26(OtogCre), have hearing impairment, but no vestibular dysfunction. The inner ear developed normally. However, on postnatal day 14 (P14), i.e., soon after the onset of hearing, cell death appeared and eventually extended to the cochlear epithelial network and sensory hair cells. Cell death initially affected only the supporting cells of the genuine sensory cell (inner hair cell, IHC), thus suggesting that it could be triggered by the IHC response to sound stimulation. Altogether, our results demonstrate that the Cx26-containing epithelial gap junction network is essential for cochlear function and cell survival. We conclude that prevention of cell death in the sensory epithelium is essential for any attempt to restore the auditory function in DFNB1 patients.


Subject(s)
Apoptosis , Cochlea/physiology , Connexins/physiology , Gap Junctions/metabolism , Hearing Loss/metabolism , Animals , Cochlea/metabolism , Connexin 26 , Connexins/genetics , Ear, Inner/metabolism , Ear, Inner/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Mice , Mice, Transgenic , Phenotype , Potassium/metabolism
11.
J Cell Biol ; 212(2): 231-44, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26754646

ABSTRACT

The precise architecture of hair bundles, the arrays of mechanosensitive microvilli-like stereocilia crowning the auditory hair cells, is essential to hearing. Myosin IIIa, defective in the late-onset deafness form DFNB30, has been proposed to transport espin-1 to the tips of stereocilia, thereby promoting their elongation. We show that Myo3a(-/-)Myo3b(-/-) mice lacking myosin IIIa and myosin IIIb are profoundly deaf, whereas Myo3a-cKO Myo3b(-/-) mice lacking myosin IIIb and losing myosin IIIa postnatally have normal hearing. Myo3a(-/-)Myo3b(-/-) cochlear hair bundles display robust mechanoelectrical transduction currents with normal kinetics but show severe embryonic abnormalities whose features rapidly change. These include abnormally tall and numerous microvilli or stereocilia, ungraded stereocilia bundles, and bundle rounding and closure. Surprisingly, espin-1 is properly targeted to Myo3a(-/-)Myo3b(-/-) stereocilia tips. Our results uncover the critical role that class III myosins play redundantly in hair-bundle morphogenesis; they unexpectedly limit the elongation of stereocilia and of subsequently regressing microvilli, thus contributing to the early hair bundle shaping.


Subject(s)
Hair Cells, Auditory/physiology , Microvilli/physiology , Myosin Heavy Chains/physiology , Myosin Type III/physiology , Stereocilia/physiology , Amino Acid Sequence , Animals , Body Patterning , Deafness/genetics , HEK293 Cells , Hair Cells, Auditory/ultrastructure , Humans , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Microfilament Proteins/metabolism , Microvilli/ultrastructure , Molecular Sequence Data , Stereocilia/ultrastructure
12.
J Cell Biol ; 199(2): 381-99, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23045546

ABSTRACT

The mechanisms underlying retinal dystrophy in Usher syndrome type I (USH1) remain unknown because mutant mice lacking any of the USH1 proteins-myosin VIIa, harmonin, cadherin-23, protocadherin-15, sans-do not display retinal degeneration. We found here that, in macaque photoreceptor cells, all USH1 proteins colocalized at membrane interfaces (i) between the inner and outer segments in rods and (ii) between the microvillus-like calyceal processes and the outer segment basolateral region in rods and cones. This pattern, conserved in humans and frogs, was mediated by the formation of an USH1 protein network, which was associated with the calyceal processes from the early embryonic stages of outer segment growth onwards. By contrast, mouse photoreceptors lacked calyceal processes and had no USH1 proteins at the inner-outer segment interface. We suggest that USH1 proteins form an adhesion belt around the basolateral region of the photoreceptor outer segment in humans, and that defects in this structure cause the retinal degeneration in USH1 patients.


Subject(s)
Intercellular Junctions/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/ultrastructure , Usher Syndromes/metabolism , Animals , Anura , Cadherin Related Proteins , Cadherins/deficiency , Cadherins/genetics , Cadherins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cytoskeletal Proteins , Humans , Intercellular Junctions/ultrastructure , Macaca fascicularis , Mice , Myosin VIIa , Myosins/deficiency , Myosins/genetics , Myosins/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Precursors/deficiency , Protein Precursors/genetics , Protein Precursors/metabolism , Retina/metabolism , Retina/ultrastructure , Retinal Dystrophies/pathology , Swine , Usher Syndromes/pathology
13.
EMBO Mol Med ; 1(2): 125-38, 2009 May.
Article in English | MEDLINE | ID: mdl-20049712

ABSTRACT

Loud sound exposure is a significant cause of hearing loss worldwide. We asked whether a lack of vezatin, an ubiquitous adherens junction protein, could result in noise-induced hearing loss. Conditional mutant mice bearing non-functional vezatin alleles only in the sensory cells of the inner ear (hair cells) indeed exhibited irreversible hearing loss after only one minute exposure to a 105 dB broadband sound. In addition, mutant mice spontaneously underwent late onset progressive hearing loss and vestibular dysfunction related to substantial hair cell death. We establish that vezatin is an integral membrane protein with two adjacent transmembrane domains, and cytoplasmic N- and C-terminal regions. Late recruitment of vezatin at junctions between MDCKII cells indicates that the protein does not play a role in the formation of junctions, but rather participates in their stability. Moreover, we show that vezatin directly interacts with radixin in its actin-binding conformation. Accordingly, we provide evidence that vezatin associates with actin filaments at cell-cell junctions. Our results emphasize the overlooked role of the junctions between hair cells and their supporting cells in the auditory epithelium resilience to sound trauma.


Subject(s)
Adherens Junctions/metabolism , Carrier Proteins/metabolism , Cochlea/metabolism , Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Sound , Actins/metabolism , Aging/pathology , Animals , Calcium/metabolism , Carrier Proteins/chemistry , Cell Death , Cell Line , Cochlea/pathology , Cochlea/physiopathology , Cochlea/ultrastructure , Cytoskeletal Proteins/metabolism , Dogs , Hair Cells, Auditory/pathology , Hair Cells, Auditory/ultrastructure , Hair Cells, Vestibular/pathology , Hair Cells, Vestibular/ultrastructure , Hearing Loss/pathology , Hearing Loss/physiopathology , Integrases/metabolism , Membrane Proteins/chemistry , Mice , Mice, Mutant Strains , Noise , Otoacoustic Emissions, Spontaneous , Protein Binding , Protein Structure, Tertiary
14.
J Cell Sci ; 120(Pt 16): 2838-50, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17666436

ABSTRACT

Defects in myosin VIIa lead to developmental anomalies of the auditory and visual sensory cells. We sought proteins interacting with the myosin VIIa tail by using the yeast two-hybrid system. Here, we report on shroom2, a submembranous PDZ domain-containing protein that is associated with the tight junctions in multiple embryonic and adult epithelia. Shroom2 directly interacts with the C-terminal MyTH4-FERM domain of myosin VIIa and with F-actin. In addition, a shroom2 fragment containing the region of interaction with F-actin was able to protect actin filaments from cytochalasin-D-induced disruption in MDCK cells. Transfection experiments in MDCK and LE (L fibroblasts that express E-cadherin) cells led us to conclude that shroom2 is targeted to the cell-cell junctions in the presence of tight junctions only. In Ca(2+)-switch experiments on MDCK cells, ZO-1 (also known as TJP1) preceded GFP-tagged shroom2 at the differentiating tight junctions. ZO-1 directly interacts with the serine- and proline-rich region of shroom2 in vitro. Moreover, the two proteins colocalize in vivo at mature tight junctions, and could be coimmunoprecipitated from brain and cochlear extracts. We suggest that shroom2 and ZO-1 form a tight-junction-associated scaffolding complex, possibly linked to myosin VIIa, that bridges the junctional membrane to the underlying cytoskeleton, thereby contributing to the stabilization of these junctions.


Subject(s)
Actins/metabolism , Dyneins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myosins/metabolism , Phosphoproteins/metabolism , Tight Junctions/metabolism , Animals , Calcium Signaling , Cell Line , Cell Membrane/metabolism , Dogs , Embryonic Structures/cytology , Embryonic Structures/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Myosin VIIa , Protein Binding , Protein Structure, Tertiary , Protein Transport , Retina/cytology , Retina/metabolism , Zonula Occludens-1 Protein
15.
Cell ; 127(2): 277-89, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-17055430

ABSTRACT

The auditory inner hair cell (IHC) ribbon synapse operates with an exceptional temporal precision and maintains a high level of neurotransmitter release. However, the molecular mechanisms underlying IHC synaptic exocytosis are largely unknown. We studied otoferlin, a predicted C2-domain transmembrane protein, which is defective in a recessive form of human deafness. We show that otoferlin expression in the hair cells correlates with afferent synaptogenesis and find that otoferlin localizes to ribbon-associated synaptic vesicles. Otoferlin binds Ca(2+) and displays Ca(2+)-dependent interactions with the SNARE proteins syntaxin1 and SNAP25. Otoferlin deficient mice (Otof(-/-)) are profoundly deaf. Exocytosis in Otof(-/-) IHCs is almost completely abolished, despite normal ribbon synapse morphogenesis and Ca(2+) current. Thus, otoferlin is essential for a late step of synaptic vesicle exocytosis and may act as the major Ca(2+) sensor triggering membrane fusion at the IHC ribbon synapse.


Subject(s)
Cochlea/metabolism , Deafness/metabolism , Exocytosis , Hair Cells, Auditory, Inner/metabolism , Membrane Proteins/metabolism , Synapses/metabolism , Animals , Auditory Pathways/metabolism , Calcium/metabolism , Cochlea/growth & development , Deafness/genetics , Deafness/physiopathology , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory, Inner/ultrastructure , Humans , Membrane Fusion , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Synaptic Transmission , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Time Factors
16.
Dev Biol ; 280(2): 281-94, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15882573

ABSTRACT

Cadherin 23 is required for normal development of the sensory hair bundle, and recent evidence suggests it is a component of the tip links, filamentous structures thought to gate the hair cells' mechano-electrical transducer channels. Antibodies against unique peptide epitopes were used to study the properties of cadherin 23 and its spatio-temporal expression patterns in developing cochlear hair cells. In the rat, intra- and extracellular domain epitopes are readily detected in the developing hair bundle between E18 and P5, and become progressively restricted to the distal tip of the hair bundle. From P13 onwards, these epitopes are no longer detected in hair bundles, but immunoreactivity is observed in the apical, vesicle-rich, pericuticular region of the hair cell. In the P2-P3 mouse cochlea, immunogold labeling reveals cadherin 23 is associated with kinocilial links and transient lateral links located between and within stereociliary rows. At this stage, the cadherin 23 ectodomain epitope remains on the hair bundle following BAPTA or La(3+) treatment, but is lost following exposure to the protease subtilisin. In contrast, mechano-electrical transduction is abolished by BAPTA but unaffected by subtilisin. These results suggest cadherin 23 is associated with transient lateral links that have properties distinct from those of the tip-link.


Subject(s)
Cadherins/physiology , Cochlea/embryology , Egtazic Acid/analogs & derivatives , Gene Expression Regulation, Developmental , Hair Cells, Auditory/embryology , Animals , Cadherins/chemistry , Cells, Cultured , Cytoplasm/metabolism , Ear, Inner/metabolism , Egtazic Acid/pharmacology , Epitopes/chemistry , Immunohistochemistry , Indicators and Reagents/pharmacology , Lanthanum/metabolism , Mice , Microscopy, Electron , Microscopy, Fluorescence , Microscopy, Immunoelectron , Models, Genetic , Protein Structure, Tertiary , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subtilisin/chemistry , Subtilisin/metabolism , Time Factors
17.
Hum Mol Genet ; 14(3): 401-10, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15590698

ABSTRACT

Defects in myosin XVa and the PDZ domain-containing protein, whirlin, underlie deafness in humans and mice. Hair bundles of mutant mice defective for either protein have abnormally short stereocilia. Here, we show that whirlin, like myosin XVa, is present at the very tip of each stereocilium in the developing and mature hair bundles of the cochlear and vestibular system. We found that myosin XVa SH3-MyTH4 region binds to the short isoform of whirlin (PR-PDZ3) that can rescue the stereocilia growth defect in whirlin defective mice. Moreover, the C-terminal MyTH4-FERM region of myosin XVa binds to the PDZ1 and PDZ2 domains of the long whirlin isoform. We conclude that a direct myosin XVa-whirlin interaction at the stereocilia tip is likely to control the elongation of stereocilia. Whirlin, unlike myosin XVa, is also transiently localized in the basal region of developing stereocilia in rat vestibular and cochlear hair cells until P4 and P12, respectively. Notably, whirlin also interacts with myosin VIIa that is present along the entire length of the stereocilia. Finally, we show that the transmembrane netrin-G1 ligand (NGL-1) binds to the PDZ1 and PDZ2 domains of whirlin and has an extracellular region that homophilically self-interacts in a Ca2+-dependent manner. The interaction between whirlin and NGL-1 might be involved in the stabilization of interstereociliar links.


Subject(s)
Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Myosins/metabolism , Actins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cilia/genetics , Cilia/metabolism , Dogs , Hair Cells, Auditory/ultrastructure , Humans , Membrane Proteins/genetics , Mice , Myosins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Rats
18.
Cell ; 109(2): 217-28, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-12007408

ABSTRACT

The physiological role of anosmin-1, defective in the X chromosome-linked form of Kallmann syndrome, is not yet known. Here, we show that anti-anosmin-1 antibodies block the formation of the collateral branches of rat olfactory bulb output neurons (mitral and tufted cells) in organotypic cultures. Moreover, anosmin-1 greatly enhances axonal branching of these dissociated neurons in culture. In addition, coculture experiments with either piriform cortex or anosmin-1-producing CHO cells demonstrate that anosmin-1 is a chemoattractant for the axons of these neurons, suggesting that this protein, which is expressed in the piriform cortex, attracts their collateral branches in vivo. We conclude that anosmin-1 has a dual branch-promoting and guidance activity, which plays an essential role in the patterning of mitral and tufted cell axon collaterals to the olfactory cortex.


Subject(s)
Axons/metabolism , Body Patterning/physiology , Cell Differentiation/physiology , Chemotaxis/physiology , Extracellular Matrix Proteins , Kallmann Syndrome/genetics , Nerve Net/embryology , Nerve Tissue Proteins/metabolism , Olfactory Bulb/embryology , X Chromosome/genetics , Animals , Axons/ultrastructure , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Cricetinae , Female , Fetus , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Gene Expression Regulation, Developmental/physiology , Kallmann Syndrome/metabolism , Kallmann Syndrome/physiopathology , Nerve Net/cytology , Nerve Net/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/embryology , Olfactory Pathways/metabolism , Pregnancy , Rats , Rats, Wistar
19.
EMBO J ; 21(24): 6689-99, 2002 Dec 16.
Article in English | MEDLINE | ID: mdl-12485990

ABSTRACT

Deaf-blindness in three distinct genetic forms of Usher type I syndrome (USH1) is caused by defects in myosin VIIa, harmonin and cadherin 23. Despite being critical for hearing, the functions of these proteins in the inner ear remain elusive. Here we show that harmonin, a PDZ domain-containing protein, and cadherin 23 are both present in the growing stereocilia and that they bind to each other. Moreover, we demonstrate that harmonin b is an F-actin-bundling protein, which is thus likely to anchor cadherin 23 to the stereocilia microfilaments, thereby identifying a novel anchorage mode of the cadherins to the actin cytoskeleton. Moreover, harmonin b interacts directly with myosin VIIa, and is absent from the disorganized hair bundles of myosin VIIa mutant mice, suggesting that myosin VIIa conveys harmonin b along the actin core of the developing stereocilia. We propose that the shaping of the hair bundle relies on a functional unit composed of myosin VIIa, harmonin b and cadherin 23 that is essential to ensure the cohesion of the stereocilia.


Subject(s)
Cadherins/physiology , Carrier Proteins/physiology , Hair Cells, Auditory/metabolism , Myosins/physiology , Actins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cadherin Related Proteins , Cadherins/chemistry , Carrier Proteins/chemistry , Cell Cycle Proteins , Cell Differentiation , Cell Line , Cytoskeletal Proteins , DNA, Complementary/metabolism , Dyneins , Gene Library , HeLa Cells , Humans , Mice , Microscopy, Electron , Microscopy, Fluorescence , Myosin VIIa , Myosins/chemistry , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rats , Rats, Wistar , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL