Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 42(4): e112118, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36594367

ABSTRACT

Sensory-independent Ca2+ spiking regulates the development of mammalian sensory systems. In the immature cochlea, inner hair cells (IHCs) fire spontaneous Ca2+ action potentials (APs) that are generated either intrinsically or by intercellular Ca2+ waves in the nonsensory cells. The extent to which either or both of these Ca2+ signalling mechansims are required for IHC maturation is unknown. We find that intrinsic Ca2+ APs in IHCs, but not those elicited by Ca2+ waves, regulate the maturation and maintenance of the stereociliary hair bundles. Using a mouse model in which the potassium channel Kir2.1 is reversibly overexpressed in IHCs (Kir2.1-OE), we find that IHC membrane hyperpolarization prevents IHCs from generating intrinsic Ca2+ APs but not APs induced by Ca2+ waves. Absence of intrinsic Ca2+ APs leads to the loss of mechanoelectrical transduction in IHCs prior to hearing onset due to progressive loss or fusion of stereocilia. RNA-sequencing data show that pathways involved in morphogenesis, actin filament-based processes, and Rho-GTPase signaling are upregulated in Kir2.1-OE mice. By manipulating in vivo expression of Kir2.1 channels, we identify a "critical time period" during which intrinsic Ca2+ APs in IHCs regulate hair-bundle function.


Subject(s)
Hair Cells, Auditory, Inner , Signal Transduction , Animals , Hair Cells, Auditory, Inner/physiology , Action Potentials/physiology , Cochlea/physiology , Mammals
2.
Clin Immunol ; 251: 109326, 2023 06.
Article in English | MEDLINE | ID: mdl-37030525

ABSTRACT

Combined immunodeficiency diseases (CID) represent the most severe forms of inborn errors of immunity. Defective T cell development and/or function, leading to an impairment in adaptive immunity are responsible for these diseases. The DNA polymerase δ complex is important for genome duplication and maintenance and consists of the catalytic subunit POLD1, and the accessory subunits POLD2 and POLD3 which stabilizes the complex. Mutations in POLD1 and POLD2 have been recently shown to be associated with a syndromic CID characterized by T cell lymphopenia with or without intellectual deficiency and sensorineural hearing loss. Here we report a homozygous POLD3 variant (NM_006591.3; p.Ile10Thr) in a Lebanese patient, the product of a consanguineous family, presenting with a syndromic severe combined immunodeficiency (SCID) with neurodevelopmental delay and hearing loss. The homozygous POLD3Ile10Thr variant abolishes POLD3 as well as POLD1 and POLD2 expression. Our findings implicate POLD3 deficiency as a novel cause of syndromic SCID.


Subject(s)
Hearing Loss , Severe Combined Immunodeficiency , Humans , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/genetics , Mutation , Homozygote , Pedigree
3.
Am J Hum Genet ; 104(5): 914-924, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982611

ABSTRACT

Glypicans are a family of cell-surface heparan sulfate proteoglycans that regulate growth-factor signaling during development and are thought to play a role in the regulation of morphogenesis. Whole-exome sequencing of the Australian family that defined Keipert syndrome (nasodigitoacoustic syndrome) identified a hemizygous truncating variant in the gene encoding glypican 4 (GPC4). This variant, located in the final exon of GPC4, results in premature termination of the protein 51 amino acid residues prior to the stop codon, and in concomitant loss of functionally important N-linked glycosylation (Asn514) and glycosylphosphatidylinositol (GPI) anchor (Ser529) sites. We subsequently identified seven affected males from five additional kindreds with novel and predicted pathogenic variants in GPC4. Segregation analysis and X-inactivation studies in carrier females provided supportive evidence that the GPC4 variants caused the condition. Furthermore, functional studies of recombinant protein suggested that the truncated proteins p.Gln506∗ and p.Glu496∗ were less stable than the wild type. Clinical features of Keipert syndrome included a prominent forehead, a flat midface, hypertelorism, a broad nose, downturned corners of mouth, and digital abnormalities, whereas cognitive impairment and deafness were variable features. Studies of Gpc4 knockout mice showed evidence of the two primary features of Keipert syndrome: craniofacial abnormalities and digital abnormalities. Phylogenetic analysis demonstrated that GPC4 is most closely related to GPC6, which is associated with a bone dysplasia that has a phenotypic overlap with Keipert syndrome. Overall, we have shown that pathogenic variants in GPC4 cause a loss of function that results in Keipert syndrome, making GPC4 the third human glypican to be linked to a genetic syndrome.


Subject(s)
Deafness/congenital , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genetic Variation , Glypicans/genetics , Lower Extremity Deformities, Congenital/genetics , Lower Extremity Deformities, Congenital/pathology , Adult , Child , Child, Preschool , Deafness/genetics , Deafness/pathology , Female , Humans , Infant , Male , Pedigree , Phenotype , Young Adult
4.
J Physiol ; 598(18): 3891-3910, 2020 09.
Article in English | MEDLINE | ID: mdl-32608086

ABSTRACT

KEY POINTS: Age-related hearing loss (ARHL) is a very heterogeneous disease, resulting from cellular senescence, genetic predisposition and environmental factors (e.g. noise exposure). Currently, we know very little about age-related changes occurring in the auditory sensory cells, including those associated with the outer hair cells (OHCs). Using different mouse strains, we show that OHCs undergo several morphological and biophysical changes in the ageing cochlea. Ageing OHCs also exhibited the progressive loss of afferent and efferent synapses. We also provide evidence that the size of the mechanoelectrical transducer current is reduced in ageing OHCs, highlighting its possible contribution in cochlear ageing. ABSTRACT: Outer hair cells (OHCs) are electromotile sensory receptors that provide sound amplification within the mammalian cochlea. Although OHCs appear susceptible to ageing, the progression of the pathophysiological changes in these cells is still poorly understood. By using mouse strains with a different progression of hearing loss (C57BL/6J, C57BL/6NTac, C57BL/6NTacCdh23+ , C3H/HeJ), we have identified morphological, physiological and molecular changes in ageing OHCs (9-12 kHz cochlear region). We show that by 6 months of age, OHCs from all strains underwent a reduction in surface area, which was not a sign of degeneration. Although the ageing OHCs retained a normal basolateral membrane protein profile, they showed a reduction in the size of the K+ current and non-linear capacitance, a readout of prestin-dependent electromotility. Despite these changes, OHCs have a normal Vm and retain the ability to amplify sound, as distortion product otoacoustic emission thresholds were not affected in aged, good-hearing mice (C3H/HeJ, C57BL/6NTacCdh23+ ). The loss of afferent synapses was present in all strains at 15 months. The number of efferent synapses per OHCs, defined as postsynaptic SK2 puncta, was reduced in aged OHCs of all strains apart from C3H mice. Several of the identified changes occurred in aged OHCs from all mouse strains, thus representing a general trait in the pathophysiological progression of age-related hearing loss, possibly aimed at preserving functionality. We have also shown that the mechanoelectrical transduction (MET) current from OHCs of mice harbouring the Cdh23ahl allele is reduced with age, highlighting the possibility that changes in the MET apparatus could play a role in cochlear ageing.


Subject(s)
Hair Cells, Auditory, Outer , Otoacoustic Emissions, Spontaneous , Animals , Cadherins , Cochlea , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
5.
Eur J Neurosci ; 43(2): 148-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26386265

ABSTRACT

Functional maturation of afferent synaptic connections to inner hair cells (IHCs) involves pruning of excess synapses formed during development, as well as the strengthening and survival of the retained synapses. These events take place during the thyroid hormone (TH)-critical period of cochlear development, which is in the perinatal period for mice and in the third trimester for humans. Here, we used the hypothyroid Snell dwarf mouse (Pit1(dw)) as a model to study the role of TH in afferent type I synaptic refinement and functional maturation. We observed defects in afferent synaptic pruning and delays in calcium channel clustering in the IHCs of Pit1(dw) mice. Nevertheless, calcium currents and capacitance reached near normal levels in Pit1(dw) IHCs by the age of onset of hearing, despite the excess number of retained synapses. We restored normal synaptic pruning in Pit1(dw) IHCs by supplementing with TH from postnatal day (P)3 to P8, establishing this window as being critical for TH action on this process. Afferent terminals of older Pit1(dw) IHCs showed evidence of excitotoxic damage accompanied by a concomitant reduction in the levels of the glial glutamate transporter, GLAST. Our results indicate that a lack of TH during a critical period of inner ear development causes defects in pruning and long-term homeostatic maintenance of afferent synapses.


Subject(s)
Cochlea/growth & development , Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Inner/ultrastructure , Synapses/physiology , Synapses/ultrastructure , Triiodothyronine/physiology , Alcohol Oxidoreductases , Animals , Calcium Channels, L-Type/metabolism , Co-Repressor Proteins , Cochlea/drug effects , Cochlea/ultrastructure , DNA-Binding Proteins/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Hair Cells, Auditory, Inner/drug effects , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Synapses/drug effects , Transcription Factor Pit-1/genetics , Triiodothyronine/administration & dosage
6.
Eur J Neurosci ; 39(8): 1256-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24460873

ABSTRACT

Thrombospondins (TSPs) constitute a family of secreted extracellular matrix proteins that have been shown to be involved in the formation of synapses in the central nervous system. In this study, we show that TSP1 and TSP2 are expressed in the cochlea, and offer the first description of their putative roles in afferent synapse development and function in the inner ear. We examined mice with deletions of TSP1, TSP2 and both (TSP1/TSP2) for inner ear development and function. Immunostaining for synaptic markers indicated a significant decrease in the number of formed afferent synapses in the cochleae of TSP2 and TSP1/TSP2 knockout (KO) mice at postnatal day (P)29. In functional studies, TSP2 and TSP1/TSP2 KO mice showed elevated auditory brainstem response (ABR) thresholds as compared with wild-type littermates, starting at P15, with the most severe phenotype being seen for TSP1/TSP2 KO mice. TSP1/TSP2 KO mice also showed reduced wave I amplitudes of ABRs and vestibular evoked potentials, suggesting synaptic dysfunction in both the auditory and vestibular systems. Whereas ABR thresholds in TSP1 KO mice were relatively unaffected at early ages, TSP1/TSP2 KO mice showed the most severe phenotype among all of the genotypes tested, suggesting functional redundancy between the two genes. On the basis of the above results, we propose that TSPs play an important role in afferent synapse development and function of the inner ear.


Subject(s)
Ear, Inner/physiology , Evoked Potentials, Auditory , Neurons, Afferent/metabolism , Synapses/metabolism , Thrombospondin 1/metabolism , Thrombospondins/metabolism , Animals , Auditory Pathways/growth & development , Auditory Pathways/metabolism , Auditory Pathways/physiology , Ear, Inner/cytology , Ear, Inner/growth & development , Ear, Inner/metabolism , Gene Deletion , Mice , Neurons, Afferent/physiology , Sensory Thresholds , Synapses/physiology , Thrombospondin 1/genetics , Thrombospondins/genetics
7.
Cell Rep ; 43(4): 114025, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38564333

ABSTRACT

Type I spiral ganglion neurons (SGNs) convey sound information to the central auditory pathway by forming synapses with inner hair cells (IHCs) in the mammalian cochlea. The molecular mechanisms regulating the formation of the post-synaptic density (PSD) in the SGN afferent terminals are still unclear. Here, we demonstrate that brain-specific angiogenesis inhibitor 1 (BAI1) is required for the clustering of AMPA receptors GluR2-4 (glutamate receptors 2-4) at the PSD. Adult Bai1-deficient mice have functional IHCs but fail to transmit information to the SGNs, leading to highly raised hearing thresholds. Despite the almost complete absence of AMPA receptor subunits, the SGN fibers innervating the IHCs do not degenerate. Furthermore, we show that AMPA receptors are still expressed in the cochlea of Bai1-deficient mice, highlighting a role for BAI1 in trafficking or anchoring GluR2-4 to the PSDs. These findings identify molecular and functional mechanisms required for sound encoding at cochlear ribbon synapses.


Subject(s)
Cochlea , Hearing , Post-Synaptic Density , Receptors, AMPA , Receptors, G-Protein-Coupled , Spiral Ganglion , Animals , Receptors, AMPA/metabolism , Mice , Spiral Ganglion/metabolism , Hearing/physiology , Cochlea/metabolism , Post-Synaptic Density/metabolism , Mice, Knockout , Hair Cells, Auditory, Inner/metabolism , Mice, Inbred C57BL , Synapses/metabolism
8.
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
9.
Front Mol Neurosci ; 14: 686790, 2021.
Article in English | MEDLINE | ID: mdl-34025354

ABSTRACT

[This corrects the article DOI: 10.3389/fnmol.2020.00083.].

10.
J Neurosci ; 29(4): 1212-23, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19176829

ABSTRACT

The absence of thyroid hormone (TH) during late gestation and early infancy can cause irreparable deafness in both humans and rodents. A variety of rodent models have been used in an effort to identify the underlying molecular mechanism. Here, we characterize a mouse model of secondary hypothyroidism, pituitary transcription factor 1 (Pit1(dw)), which has profound, congenital deafness that is rescued by oral TH replacement. These mutants have tectorial membrane abnormalities, including a prominent Hensen's stripe, elevated beta-tectorin composition, and disrupted striated-sheet matrix. They lack distortion product otoacoustic emissions and cochlear microphonic responses, and exhibit reduced endocochlear potentials, suggesting defects in outer hair cell function and potassium recycling. Auditory system and hair cell physiology, histology, and anatomy studies reveal novel defects of hormone deficiency related to deafness: (1) permanently impaired expression of KCNJ10 in the stria vascularis of Pit1(dw) mice, which likely contributes to the reduced endocochlear potential, (2) significant outer hair cell loss in the mutants, which may result from cellular stress induced by the lower KCNQ4 expression and current levels in Pit1(dw) mutant outer hair cells, and (3) sensory and strial cell deterioration, which may have implications for thyroid hormone dysregulation in age-related hearing impairment. In summary, we suggest that these defects in outer hair cell and strial cell function are important contributors to the hearing impairment in Pit1(dw) mice.


Subject(s)
Deafness/etiology , Gene Expression Regulation/genetics , Hypothyroidism/complications , KCNQ Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Transcription Factor Pit-1/genetics , Age Factors , Animals , Animals, Newborn , Deafness/genetics , Deafness/pathology , Disease Models, Animal , Hair Cells, Auditory, Outer/diagnostic imaging , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/ultrastructure , Hypothyroidism/genetics , KCNQ Potassium Channels/genetics , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission/methods , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Otoacoustic Emissions, Spontaneous/genetics , Potassium Channels, Inwardly Rectifying/genetics , Stria Vascularis/pathology , Synaptophysin/genetics , Synaptophysin/metabolism , Tectorial Membrane/pathology , Tectorial Membrane/ultrastructure , Ultrasonography
11.
Front Mol Neurosci ; 13: 83, 2020.
Article in English | MEDLINE | ID: mdl-32523514

ABSTRACT

In the mature cochlea, each inner hair cell (IHC) is innervated by multiple spiral ganglion neurons of type I (SGNI). SGNIs are morphologically and electro-physiologically diverse. Also, they differ in their susceptibility to noise insult. However, the molecular underpinnings of their identity and physiological differences remain poorly understood. In this study, we developed a novel triple transgenic mouse, which enabled the isolation of pure populations of SGNIs and the analysis of a 96-gene panel via single-cell qPCR. We found three distinct populations of Type I SGNs, which were marked by their exclusive expression of Lmx1a, Slc4a4, or Mfap4/Fzd2, respectively, at postnatal days P3, P8, and P12. Our data suggest that afferent SGN subtypes are established genetically before the onset of hearing and that the expression of key physiological markers, such as ion channels, is heterogeneous and may be underlying the heterogeneous firing proprieties of SGNIs.

12.
Neuroscience ; 408: 68-80, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30928339

ABSTRACT

Thrombospondins (TSPs) are cell adhesion molecules that play an important role in the maintenance of hearing and afferent synaptic connections. Based on their reported function in restoring synaptic connections after stroke, we tested a potential role for TSP1 and TSP2 genes in repairing cochlear synapses following noise injury. We observed a tonotopic gradient in the expression of TSP1 and TSP2 mRNA in control mouse cochleae and an upregulation of these genes following noise exposure. Examining the functional sequelae of these changes revealed that afferent synaptic counts and auditory brainstem responses (ABRs) in noise-exposed TSP1 and TSP2 knockout (-/-) mice exhibited a worst recovery when compared to controls. Consistent with their tonotopic expression, TSP1-/- mice showed greater susceptibility to noise-induced hearing loss (NIHL) at 8 kHz and 16 kHz frequencies, whereas NIHL in TSP2-/- mice occurred only at mid and high frequencies. Further analysis of the ABR waveforms indicated peripheral neuronal damage in TSP2-/- but not in TSP1-/- mice. Noise trauma affecting mid to high frequencies triggered severe seizures in the TSP2-/- mice. We found that decreased susceptibility to audiogenic seizures in TSP1-/- mice was correlated with increased TSP2 protein levels in their inner ears, suggesting that TSP2 might functionally compensate for the loss of TSP1 in these mice. Our data indicate that TSP1 and TSP2 are both involved in susceptibility to NIHL, with TSP2 playing a more prominent role.


Subject(s)
Auditory Threshold/physiology , Cochlea/metabolism , Hearing Loss, Noise-Induced/metabolism , Thrombospondin 1/metabolism , Thrombospondins/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Evoked Potentials, Auditory, Brain Stem/physiology , Gene Expression Regulation , Hearing Loss, Noise-Induced/physiopathology , Mice , Mice, Knockout , Neurons/metabolism , Spiral Ganglion/metabolism , Thrombospondin 1/genetics , Thrombospondins/genetics
13.
Article in English | MEDLINE | ID: mdl-30617058

ABSTRACT

To provide a meaningful representation of the auditory landscape, mammalian cochlear hair cells are optimized to detect sounds over an incredibly broad range of frequencies and intensities with unparalleled accuracy. This ability is largely conferred by specialized ribbon synapses that continuously transmit acoustic information with high fidelity and sub-millisecond precision to the afferent dendrites of the spiral ganglion neurons. To achieve this extraordinary task, ribbon synapses employ a unique combination of molecules and mechanisms that are tailored to sounds of different frequencies. Here we review the current understanding of how the hair cell's presynaptic machinery and its postsynaptic afferent connections are formed, how they mature, and how their function is adapted for an accurate perception of sound.


Subject(s)
Hair Cells, Auditory/physiology , Neurons, Afferent/physiology , Synapses/physiology , Animals , Calcium Channels, L-Type/physiology , Humans , Membrane Potentials , Receptors, Glutamate/physiology
14.
J Assoc Res Otolaryngol ; 8(3): 329-37, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17619105

ABSTRACT

MYOSIN XV is a motor protein that interacts with the PDZ domain-containing protein WHIRLIN and transports WHIRLIN to the tips of the stereocilia. Shaker 2 (sh2) mice have a mutation in the motor domain of MYOSIN XV and exhibit congenital deafness and circling behavior, probably because of abnormally short stereocilia. Whirler (wi) mice have a similar phenotype caused by a deletion in the third PDZ domain of WHIRLIN. We compared the morphology of Whrn (wi/wi) and Myo15 (sh2/sh2) sensory hair cells and found that Myo15 (sh2/sh2) have more frequent pathology at the base of inner hair cells than Whrn (wi/wi), and shorter outer hair cell stereocilia. Considering the functional and morphologic similarities in the phenotypes caused by mutations in Myo15 and Whrn, and the physical interaction between their encoded proteins, we used a genetic approach to test for functional overlap. Double heterozygotes (Myo15 (sh2/+), Whrn (wi/+)) have normal hearing and no increase in hearing loss compared to normal littermates. Single and double mutants (Myo15 (sh2/sh2), Whrn (wi/wi)) exhibit abnormal persistence of kinocilia and microvilli, and develop abnormal cytoskeletal architecture. Double mutants are also similar to the single mutants in viability, circling behavior, and lack of a Preyer reflex. The morphology of cochlear hair cell stereocilia in double mutants reflects a dominance of the more severe Myo15 (sh2/sh2) phenotype over the Whrn (wi/wi) phenotype. This suggests that MYOSIN XV may interact with other proteins besides WHIRLIN that are important for hair cell maturation.


Subject(s)
Hair Cells, Auditory, Inner/pathology , Membrane Proteins/genetics , Mutation/genetics , Myosins/genetics , Actins/metabolism , Animals , Cilia/pathology , Cilia/physiology , Cytoskeleton/pathology , Cytoskeleton/physiology , Ear, Inner/pathology , Female , Hair Cells, Auditory, Inner/growth & development , Hair Cells, Auditory, Inner/physiology , Hearing/genetics , Hearing/physiology , Heterozygote , Homozygote , Male , Membrane Proteins/physiology , Mice , Mice, Inbred Strains , Myosins/physiology , Phenotype
15.
Sci Rep ; 7(1): 17354, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229982

ABSTRACT

Myosin-X (Myo10) is an unconventional myosin best known for its striking localization to the tips of filopodia. Despite the broad expression of Myo10 in vertebrate tissues, its functions at the organismal level remain largely unknown. We report here the generation of KO-first (Myo10 tm1a/tm1a ), floxed (Myo10 tm1c/tm1c ), and KO mice (Myo10 tm1d/tm1d ). Complete knockout of Myo10 is semi-lethal, with over half of homozygous KO embryos exhibiting exencephaly, a severe defect in neural tube closure. All Myo10 KO mice that survive birth exhibit a white belly spot, all have persistent fetal vasculature in the eye, and ~50% have webbed digits. Myo10 KO mice that survive birth can breed and produce litters of KO embryos, demonstrating that Myo10 is not absolutely essential for mitosis, meiosis, adult survival, or fertility. KO-first mice and an independent spontaneous deletion (Myo10 m1J/m1J ) exhibit the same core phenotypes. During retinal angiogenesis, KO mice exhibit a ~50% decrease in endothelial filopodia, demonstrating that Myo10 is required to form normal numbers of filopodia in vivo. The Myo10 mice generated here demonstrate that Myo10 has important functions in mammalian development and provide key tools for defining the functions of Myo10 in vivo.


Subject(s)
Myosins/physiology , Neovascularization, Pathologic , Neural Tube/physiopathology , Ophthalmic Artery/physiopathology , Pigmentation , Pseudopodia/pathology , Vitreous Body/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ophthalmic Artery/metabolism , Pseudopodia/metabolism , Vitreous Body/blood supply , Vitreous Body/metabolism
16.
Plast Reconstr Surg Glob Open ; 4(12): e1186, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28293529

ABSTRACT

BACKGROUND: Understanding the mechanisms in nerve damage can lead to better outcomes for neuronal rehabilitation. The purpose of our study was to assess the effect of major histocompatibility complex I deficiency and inhibition of the classical complement pathway (C1q) on functional recovery and cell survival in the facial motor nucleus (FMN) after crush injury in adult and juvenile mice. METHODS: A prospective blinded analysis of functional recovery and cell survival in the FMN after a unilateral facial nerve crush injury in juvenile and adult mice was undertaken between wild-type, C1q knockout (C1q-/-), and KbDb knockout (KbDb-/-) groups. Whisker function was quantified to assess functional recovery. Neuron counts were performed to determine neuron survival in the FMN after recovery. RESULTS: After facial nerve injury, all adult wild-type mice fully recovered. Juvenile mice recovered incompletely corresponding to a greater neuron loss in the FMN of juveniles compared with adults. The C1q-/- juvenile and adult groups did not differ from wild type. The KbDb-/- adults demonstrated 50% recovery of whisker movement and decreased cell survival in FMN. The KbDb-/- juvenile group did not demonstrate any difference from control group. CONCLUSION: Histocompatibility complex I plays a role for neuroprotection and enhanced facial nerve recovery in adult mice. Inhibition of the classical complement pathway alone does not affect functional recovery or neuronal survival. The alternative and mannose binding pathways pose alternative means for activating the final components of the pathway that may lead to acute nerve damage.

17.
Elife ; 42015 Aug 24.
Article in English | MEDLINE | ID: mdl-26302205

ABSTRACT

The precise assembly of inner ear hair cell stereocilia into rows of increasing height is critical for mechanotransduction and the sense of hearing. Yet, how the lengths of actin-based stereocilia are regulated remains poorly understood. Mutations of the molecular motor myosin 15 stunt stereocilia growth and cause deafness. We found that hair cells express two isoforms of myosin 15 that differ by inclusion of an 133-kDa N-terminal domain, and that these isoforms can selectively traffic to different stereocilia rows. Using an isoform-specific knockout mouse, we show that hair cells expressing only the small isoform remarkably develop normal stereocilia bundles. However, a critical subset of stereocilia with active mechanotransducer channels subsequently retracts. The larger isoform with the 133-kDa N-terminal domain traffics to these specialized stereocilia and prevents disassembly of their actin core. Our results show that myosin 15 isoforms can navigate between functionally distinct classes of stereocilia, and are independently required to assemble and then maintain the intricate hair bundle architecture.


Subject(s)
Hearing , Myosins/genetics , Myosins/metabolism , Stereocilia/metabolism , Stereocilia/physiology , Animals , Ear, Inner/physiology , Hair Cells, Auditory/physiology , Mice , Mice, Knockout , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport
18.
Eur J Hum Genet ; 10(3): 210-2, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11973626

ABSTRACT

We report the identification of a novel locus responsible for an autosomal recessive form of hearing loss (DFNB) segregating in a Palestinian consanguineous family from Jordan. The affected individuals suffer from profound prelingual sensorineural hearing impairment. A genetic linkage with polymorphic markers surrounding D9S1776 was detected, thereby identifying a novel deafness locus, DFNB31. This locus could be assigned to a 9q32-34 region of 15 cM between markers D9S289 and D9S1881. The whirler (wi) mouse mutant, characterised by deafness and circling behaviour, maps to the corresponding region on the murine chromosome 4, thus suggesting that DFNB31 and whirler may result from orthologous gene defects.


Subject(s)
Chromosomes, Human, Pair 9 , Genes, Recessive , Hearing Loss, Sensorineural/genetics , Alleles , Chromosome Mapping , Consanguinity , Female , Genetic Linkage , Genotype , Haplotypes , Homozygote , Humans , Lod Score , Male , Microsatellite Repeats , Mutation , Pedigree , Polymorphism, Genetic
19.
Eur J Hum Genet ; 10(6): 391-4, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12080392

ABSTRACT

Non-syndromic recessive deafness (NSRD) is the most commonly encountered form of hereditary hearing loss. The majority of NSRD cases in the Mediterranean area are linked to the DFNB1 locus (the connexin 26 GJB2 gene). Unrelated NSRD patients issued from 68 Jordanian families, were tested for mutations of the GJB2 gene by sequencing. Sixteen per cent of the families tested were linked to the DFNB1 locus. The 35delG was the only GJB2 mutation detected in these families. One of these families, presenting with four affected members and not linked to the gene, was subjected to a genome-wide search and was found to be mapped to 9q34.3 with a multipoint lodscore of 3.9. One candidate gene in the interval, coding for the chloride intracellular channel 3, CLIC3, was tested and excluded. The identification of a new NSRD locus, DFNB33, in one Jordanian family, shows the wide genetic heterogeneity that characterizes hearing impairment and the genetic diversity in Middle-Eastern populations.


Subject(s)
Chromosomes, Human, Pair 9 , Deafness/genetics , Chromosome Mapping , Connexin 26 , Connexins/genetics , Female , Genes, Recessive , Humans , Jordan , Lod Score , Male , Pedigree , Sequence Deletion
20.
PLoS One ; 9(5): e94549, 2014.
Article in English | MEDLINE | ID: mdl-24804771

ABSTRACT

Early cochlear development is marked by an exuberant outgrowth of neurites that innervate multiple targets. The establishment of mature cochlear neural circuits is, however, dependent on the pruning of inappropriate axons and synaptic connections. Such refinement also occurs in the central nervous system (CNS), and recently, genes ordinarily associated with immune and inflammatory processes have been shown to play roles in synaptic pruning in the brain. These molecules include the major histocompatibility complex class I (MHCI) genes, H2-K(b) and H2-D(b), and the complement cascade gene, C1qa. Since the mechanisms involved in synaptic refinement in the cochlea are not well understood, we investigated whether these immune system genes may be involved in this process and whether they are required for normal hearing function. Here we report that these genes are not necessary for normal synapse formation and refinement in the mouse cochlea. We further demonstrate that C1qa expression is not necessary for normal hearing in mice but the lack of expression of H2-K(b) and H2-D(b) causes hearing impairment. These data underscore the importance of the highly polymorphic family of MHCI genes in hearing in mice and also suggest that factors and mechanisms regulating synaptic refinement in the cochlea may be distinct from those in the CNS.


Subject(s)
Central Nervous System/metabolism , Cochlea/metabolism , Hearing/physiology , Animals , Central Nervous System/physiology , Cochlea/physiology , Gene Expression Regulation, Developmental , Hearing/genetics , Mice , Mice, Knockout , Synapses
SELECTION OF CITATIONS
SEARCH DETAIL