Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33903231

ABSTRACT

The cochlea of our auditory system is an intricate structure deeply embedded in the temporal bone. Compared with other sensory organs such as the eye, the cochlea has remained poorly accessible for investigation, for example, by imaging. This limitation also concerns the further development of technology for restoring hearing in the case of cochlear dysfunction, which requires quantitative information on spatial dimensions and the sensorineural status of the cochlea. Here, we employed X-ray phase-contrast tomography and light-sheet fluorescence microscopy and their combination for multiscale and multimodal imaging of cochlear morphology in species that serve as established animal models for auditory research. We provide a systematic reference for morphological parameters relevant for cochlear implant development for rodent and nonhuman primate models. We simulate the spread of light from the emitters of the optical implants within the reconstructed nonhuman primate cochlea, which indicates a spatially narrow optogenetic excitation of spiral ganglion neurons.


Subject(s)
Cochlea/diagnostic imaging , Cochlear Implantation , Hearing Loss, Sensorineural/therapy , Neurons/metabolism , Animals , Cochlea/pathology , Cochlear Implants , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/physiopathology , Humans , Neurons/pathology , Optogenetics , Spiral Ganglion/diagnostic imaging , Spiral Ganglion/pathology
2.
Mol Ther ; 30(1): 105-118, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34174443

ABSTRACT

Myosin VI(MYO6) is an unconventional myosin that is vital for auditory and vestibular function. Pathogenic variants in the human MYO6 gene cause autosomal-dominant or -recessive forms of hearing loss. Effective treatments for Myo6 mutation causing hearing loss are limited. We studied whether adeno-associated virus (AAV)-PHP.eB vector-mediated in vivo delivery of Staphylococcus aureus Cas9 (SaCas9-KKH)-single-guide RNA (sgRNA) complexes could ameliorate hearing loss in a Myo6WT/C442Y mouse model that recapitulated the phenotypes of human patients. The in vivo editing efficiency of the AAV-SaCas9-KKH-Myo6-g2 system on Myo6C442Y is 4.05% on average in Myo6WT/C442Y mice, which was ∼17-fold greater than editing efficiency of Myo6WT alleles. Rescue of auditory function was observed up to 5 months post AAV-SaCas9-KKH-Myo6-g2 injection in Myo6WT/C442Y mice. Meanwhile, shorter latencies of auditory brainstem response (ABR) wave I, lower distortion product otoacoustic emission (DPOAE) thresholds, increased cell survival rates, more regular hair bundle morphology, and recovery of inward calcium levels were also observed in the AAV-SaCas9-KKH-Myo6-g2-treated ears compared to untreated ears. These findings provide further reference for in vivo genome editing as a therapeutic treatment for various semi-dominant forms of hearing loss and other semi-dominant diseases.


Subject(s)
Gene Editing , Hearing Loss , Animals , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing , Hearing Loss/genetics , Hearing Loss/therapy , Humans , Mice , RNA, Guide, Kinetoplastida
3.
PLoS Biol ; 17(4): e3000194, 2019 04.
Article in English | MEDLINE | ID: mdl-30973865

ABSTRACT

Adult-onset hearing loss is very common, but we know little about the underlying molecular pathogenesis impeding the development of therapies. We took a genetic approach to identify new molecules involved in hearing loss by screening a large cohort of newly generated mouse mutants using a sensitive electrophysiological test, the auditory brainstem response (ABR). We review here the findings from this screen. Thirty-eight unexpected genes associated with raised thresholds were detected from our unbiased sample of 1,211 genes tested, suggesting extreme genetic heterogeneity. A wide range of auditory pathophysiologies was found, and some mutant lines showed normal development followed by deterioration of responses, revealing new molecular pathways involved in progressive hearing loss. Several of the genes were associated with the range of hearing thresholds in the human population and one, SPNS2, was involved in childhood deafness. The new pathways required for maintenance of hearing discovered by this screen present new therapeutic opportunities.


Subject(s)
Auditory Perception/genetics , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing Loss/genetics , Acoustic Stimulation/methods , Adult , Animals , Anion Transport Proteins/genetics , Child , Electrophysiological Phenomena/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Genetic Association Studies , Hearing/genetics , Hearing Loss/metabolism , Humans , Male , Mice , Mice, Inbred C57BL
4.
J Neurosci ; 40(15): 2976-2992, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32152201

ABSTRACT

Hepatocyte growth factor (HGF) is a multifunctional protein that signals through the MET receptor. HGF stimulates cell proliferation, cell dispersion, neuronal survival, and wound healing. In the inner ear, levels of HGF must be fine-tuned for normal hearing. In mice, a deficiency of HGF expression limited to the auditory system, or an overexpression of HGF, causes neurosensory deafness. In humans, noncoding variants in HGF are associated with nonsyndromic deafness DFNB39 However, the mechanism by which these noncoding variants causes deafness was unknown. Here, we reveal the cause of this deafness using a mouse model engineered with a noncoding intronic 10 bp deletion (del10) in Hgf Male and female mice homozygous for del10 exhibit moderate-to-profound hearing loss at 4 weeks of age as measured by tone burst auditory brainstem responses. The wild type (WT) 80 mV endocochlear potential was significantly reduced in homozygous del10 mice compared with WT littermates. In normal cochlea, endocochlear potentials are dependent on ion homeostasis mediated by the stria vascularis (SV). Previous studies showed that developmental incorporation of neural crest cells into the SV depends on signaling from HGF/MET. We show by immunohistochemistry that, in del10 homozygotes, neural crest cells fail to infiltrate the developing SV intermediate layer. Phenotyping and RNAseq analyses reveal no other significant abnormalities in other tissues. We conclude that, in the inner ear, the noncoding del10 mutation in Hgf leads to developmental defects of the SV and consequently dysfunctional ion homeostasis and a reduction in the EP, recapitulating human DFNB39 nonsyndromic deafness.SIGNIFICANCE STATEMENT Hereditary deafness is a common, clinically and genetically heterogeneous neurosensory disorder. Previously, we reported that human deafness DFNB39 is associated with noncoding variants in the 3'UTR of a short isoform of HGF encoding hepatocyte growth factor. For normal hearing, HGF levels must be fine-tuned as an excess or deficiency of HGF cause deafness in mouse. Using a Hgf mutant mouse with a small 10 bp deletion recapitulating a human DFNB39 noncoding variant, we demonstrate that neural crest cells fail to migrate into the stria vascularis intermediate layer, resulting in a significantly reduced endocochlear potential, the driving force for sound transduction by inner ear hair cells. HGF-associated deafness is a neurocristopathy but, unlike many other neurocristopathies, it is not syndromic.


Subject(s)
Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing Loss, Sensorineural/genetics , Hepatocyte Growth Factor/genetics , Neural Crest/growth & development , Stria Vascularis/pathology , Animals , Cell Count , Ear, Inner/abnormalities , Female , Hair Cells, Auditory , Hearing Loss, Sensorineural/pathology , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Crest/pathology , RNA Probes
5.
Am J Med Genet A ; 185(4): 1131-1141, 2021 04.
Article in English | MEDLINE | ID: mdl-33529473

ABSTRACT

Smith-Lemli-Opitz syndrome (SLOS) is an autosomal recessive multiple congenital malformation and intellectual disability syndrome resulting from variants in DHCR7. Auditory characteristics of persons with SLOS have been described in limited case reports but have not been systematically evaluated. The objective of this study is to describe the auditory phenotype in SLOS. Age- and ability-appropriate hearing evaluations were conducted on 32 patients with SLOS. A subset of 21 had auditory brainstem response testing, from which an auditory neural phenotype is described. Peripheral or retrocochlear auditory dysfunction was observed in at least one ear of 65.6% (21) of the patients in our SLOS cohort. The audiometric phenotype was heterogeneous and included conductive, mixed, and sensorineural hearing loss. The most common presentation was a slight to mild conductive hearing loss, although profound sensorineural hearing loss was also observed. Abnormal auditory brainstem responses indicative of retrocochlear dysfunction were identified in 21.9% of the patients. Many were difficult to test behaviorally and required objective assessment methods to estimate hearing sensitivity. Individuals with SLOS are likely to have hearing loss that may impact communication, including speech and language development. Routine audiologic surveillance should be conducted to ensure prompt management of hearing loss.


Subject(s)
Auditory Diseases, Central/genetics , Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , Smith-Lemli-Opitz Syndrome/diagnosis , Adolescent , Adult , Audiometry , Auditory Diseases, Central/physiopathology , Child , Child, Preschool , Cochlear Nerve/physiopathology , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Hearing Loss, Sensorineural/physiopathology , Humans , Infant , Male , Mutation/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phenotype , Smith-Lemli-Opitz Syndrome/genetics , Smith-Lemli-Opitz Syndrome/physiopathology , Young Adult
6.
J Neurosci ; 39(24): 4797-4813, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30936239

ABSTRACT

Fragile X syndrome (FXS) is characterized by hypersensitivity to sensory stimuli, including environmental sounds. We compared the auditory brainstem response (ABR) recorded in vivo in mice lacking the gene (Fmr1-/y ) for fragile X mental retardation protein (FMRP) with that in wild-type animals. We found that ABR wave I, which represents input from the auditory nerve, is reduced in Fmr1-/y animals, but only at high sound levels. In contrast, wave IV, which represents the activity of auditory brainstem nuclei is enhanced at all sound levels, suggesting that loss of FMRP alters the central processing of auditory signals. Current-clamp recordings of neurons in the medial nucleus of the trapezoid body in the auditory brainstem revealed that, in contrast to neurons from wild-type animals, sustained depolarization triggers repetitive firing rather than a single action potential. In voltage-clamp recordings, K+ currents that activate at positive potentials ("high-threshold" K+ currents), which are required for high-frequency firing and are carried primarily by Kv3.1 channels, are elevated in Fmr1-/y mice, while K+ currents that activate near the resting potential and inhibit repetitive firing are reduced. We therefore tested the effects of AUT2 [((4-({5-[(4R)-4-ethyl-2,5-dioxo-1-imidazolidinyl]-2-pyridinyl}oxy)-2-(1-methylethyl) benzonitrile], a compound that modulates Kv3.1 channels. AUT2 reduced the high-threshold K+ current and increased the low-threshold K+ currents in neurons from Fmr1-/y animals by shifting the activation of the high-threshold current to more negative potentials. This reduced the firing rate and, in vivo, restored wave IV of the ABR. Our results from animals of both sexes suggest that the modulation of the Kv3.1 channel may have potential for the treatment of sensory hypersensitivity in patients with FXS.SIGNIFICANCE STATEMENT mRNA encoding the Kv3.1 potassium channel was one of the first described targets of the fragile X mental retardation protein (FMRP). Fragile X syndrome is caused by loss of FMRP and, in humans and mice, causes hypersensitivity to auditory stimuli. We found that components of the auditory brain response (ABR) corresponding to auditory brainstem activity are enhanced in mice lacking FMRP. This is accompanied by hyperexcitability and altered potassium currents in auditory brainstem neurons. Treatment with a drug that alters the voltage dependence of Kv3.1 channels normalizes the imbalance of potassium currents, as well as ABR responses in vivo, suggesting that such compounds may be effective in treating some symptoms of fragile X syndrome.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Shaw Potassium Channels/metabolism , Animals , Auditory Pathways , Auditory Perception , Brain Stem/drug effects , Cochlear Nucleus/physiology , Electrophysiological Phenomena , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Hydantoins/pharmacology , In Vitro Techniques , Male , Mice , Mice, Knockout , Patch-Clamp Techniques , Pyridines/pharmacology
7.
J Cell Mol Med ; 24(16): 9101-9113, 2020 08.
Article in English | MEDLINE | ID: mdl-34008309

ABSTRACT

Noise-induced hearing loss (NIHL) is one of the most frequent disabilities in industrialized countries. Evidence shows that hair cell loss in the auditory end organ is responsible for the majority of various ear pathological conditions. The functional roles of the receptor tyrosine kinase ROR1 have been underscored in various tumours. In this study, we evaluated the ability of ROR1 to influence cochlear hair cell loss of guinea pigs with NIHL. The NIHL model was developed in guinea pigs, with subsequent measurement of the auditory brainstem response (ABR). Gain-of-function experiments were employed to explore the role of ROR1 in NIHL. The interaction between ROR1 and Wnt5a and their functions in the cochlear hair cell loss were further analysed in response to alteration of ROR1 and Wnt5a. Guinea pigs with NIHL demonstrated elevated ABR threshold and down-regulated ROR1, Wnt5a and NF-κB p65. The up-regulation of ROR1 was shown to decrease the cochlear hair cell loss and the expression of pro-apoptotic gene (Bax, p53) in guinea pig cochlea, but promoted the expression of anti-apoptotic gene (Bcl-2) and the fluorescence intensity of cleaved-caspase-3. ROR1 interacted with Wnt5a to activate the NF-κB signalling pathway through inducing phosphorylation and translocation of p65. Furthermore, Wnt5a overexpression decreased the cochlear hair cell loss. Collectively, this study suggested the protection of overexpression of ROR1 against cochlear hair cell loss in guinea pigs with NIHL via the Wnt5a-dependent NF-κB signalling pathway.


Subject(s)
Hair Cells, Auditory/pathology , Hearing Loss/prevention & control , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Animals , Apoptosis/genetics , Cells, Cultured , Disease Models, Animal , Down-Regulation , Ectopic Gene Expression , Evoked Potentials, Auditory, Brain Stem/genetics , Guinea Pigs , Hair Cells, Auditory/physiology , Hearing Loss/etiology , Hearing Loss/pathology , Male , NF-kappa B/metabolism , Noise/adverse effects , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt-5a Protein/metabolism
8.
Neural Plast ; 2020: 8889264, 2020.
Article in English | MEDLINE | ID: mdl-32587610

ABSTRACT

Inbred mouse models are widely used to study age-related hearing loss (AHL). Many genes associated with AHL have been mapped in a variety of strains. However, little is known about gene variants that have the converse function-protective genes that confer strong resistance to hearing loss. Previously, we reported that C57BL/6J (B6) and DBA/2J (D2) strains share a common hearing loss allele in Cdh23. The cadherin 23 (Cdh23) gene is a key contributor to early-onset hearing loss in humans. In this study, we tested hearing across a large family of 54 BXD strains generated from B6 to D2 crosses. Five of 54 strains maintain the normal threshold (20 dB SPL) even at 2 years old-an age at which both parental strains are essentially deaf. Further analyses revealed an age-related hearing protection (ahp) locus on chromosome 16 (Chr 16) at 57~76 Mb with a maximum LOD of 5.7. A small number of BXD strains at 2 years with good hearing correspond roughly to the percentage of humans who have good hearing at 90 years old. Further studies to define candidate genes in the ahp locus and related molecular mechanisms involved in age-related resilience or resistance to AHL are warranted.


Subject(s)
Alleles , Auditory Threshold/physiology , Cadherins/genetics , Chromosomes, Mammalian , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing Loss/genetics , Hearing/physiology , Animals , Genetic Predisposition to Disease , Genotype , Mice , Phenotype
9.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003463

ABSTRACT

Aging is associated with functional and morphological changes in the sensory organs, including the auditory system. Mitophagy, a process that regulates the turnover of dysfunctional mitochondria, is impaired with aging. This study aimed to investigate the effect of aging on mitophagy in the central auditory system using an age-related hearing loss mouse model. C57BL/6J mice were divided into the following four groups based on age: 1-, 6-, 12-, and 18-month groups. The hearing ability was evaluated by measuring the auditory brainstem response (ABR) thresholds. The mitochondrial DNA damage level and the expression of mitophagy-related genes, and proteins were investigated by real-time polymerase chain reaction and Western blot analyses. The colocalization of mitophagosomes and lysosomes in the mouse auditory cortex and inferior colliculus was analyzed by immunofluorescence analysis. The expression of genes involved in mitophagy, such as PINK1, Parkin, and BNIP3 in the mouse auditory cortex and inferior colliculus, was investigated by immunohistochemical staining. The ABR threshold increased with aging. In addition to the mitochondrial DNA integrity, the mRNA levels of PINK1, Parkin, NIX, and BNIP3, as well as the protein levels of PINK1, Parkin, BNIP3, COX4, LC3B, mitochondrial oxidative phosphorylation (OXPHOS) subunits I-IV in the mouse auditory cortex significantly decreased with aging. The immunofluorescence analysis revealed that the colocalization of mitophagosomes and lysosomes in the mouse auditory cortex and inferior colliculus decreased with aging. The immunohistochemical analysis revealed that the expression of PINK1, Parkin, and BNIP3 decreased in the mouse auditory cortex and inferior colliculus with aging. These findings indicate that aging-associated impaired mitophagy may contribute to the cellular changes observed in an aged central auditory system, which result in age-related hearing loss. Thus, the induction of mitophagy can be a potential therapeutic strategy for age-related hearing loss.


Subject(s)
Aging/genetics , Mitochondria/genetics , Mitophagy/genetics , Presbycusis/genetics , Aging/pathology , Animals , Auditory Diseases, Central/genetics , Auditory Diseases, Central/physiopathology , DNA, Mitochondrial/genetics , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Lysosomes/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Phosphorylation , Presbycusis/physiopathology
10.
J Neuroinflammation ; 16(1): 61, 2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30866963

ABSTRACT

BACKGROUND: Microglia play a central role in most neurological disorders, but the impact of microgliosis on brain environment and clinical functions is not fully understood. Mice lacking multifunctional protein-2 (MFP2), a pivotal enzyme in peroxisomal ß-oxidation, develop a fatal disorder characterized by motor problems similar to the milder form of MFP2 deficiency in humans. The hallmark of disease in mice is the chronic proliferation of microglia in the brain, but molecular pathomechanisms that drive rapid clinical deterioration in human and mice remain unknown. In the present study, we identified the effects of specific deletion of MFP2 from microglia in the brain on immune responses, neuronal functioning, and behavior. METHODS: We created a novel Cx3cr1-Mfp2-/- mouse model and studied the impact of MFP2 deficiency on microglial behavior at different ages using immunohistochemistry and real-time PCR. Pro- and anti-inflammatory responses of Mfp2-/- microglia were assessed in vitro and in vivo after stimulation with IL-1ß/INFγ and IL-4 (in vitro) and LPS and IL-4 (in vivo). Facial nerve axotomy was unilaterally performed in Cx3cr1-Mfp2-/- and control mice, and microglial functioning in response to neuronal injury was subsequently analyzed by histology and real-time PCR. Finally, neuronal function, motor function, behavior, and cognition were assessed using brainstem auditory evoked potentials, grip strength and inverted grid test, open field exploration, and passive avoidance learning, respectively. RESULTS: We found that Mfp2-/- microglia in a genetically intact brain environment adopt an inflammatory activated and proliferative state. In addition, we found that acute inflammatory and neuronal injury provoked normal responses of Mfp2-/- microglia in Cx3cr1-Mfp2-/- mice during the post-injury period. Despite chronic pro-inflammatory microglial reactivity, Cx3cr1-Mfp2-/- mice exhibited normal neuronal transmission, clinical performance, and cognition. CONCLUSION: Our data demonstrate that MFP2 deficiency in microglia causes intrinsic dysregulation of their inflammatory profile, which is not harmful to neuronal function, motor function, and cognition in mice during their first year of life.


Subject(s)
Brain/pathology , Inflammation/pathology , Microglia/drug effects , Microglia/metabolism , Peroxisomal Multifunctional Protein-2/deficiency , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , CX3C Chemokine Receptor 1/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Facial Nerve Diseases/complications , Facial Nerve Diseases/pathology , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hand Strength/physiology , Inflammation/chemically induced , Interleukin-4/administration & dosage , Lipopolysaccharides/toxicity , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/pathology , Peroxisomal Multifunctional Protein-2/genetics
11.
Neurochem Res ; 44(6): 1494-1507, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30659505

ABSTRACT

Autism is a wide spread neurodevelopmental disorder with growing morbidity rates, affecting more boys than girls worldwide. Activity-dependent neuroprotective protein (ADNP) was recently recognized as a leading gene accounted for 0.17% of autism spectrum disorder (ASD) cases globally. Respectively, mutations in the human ADNP gene (ADNP syndrome), cause multi-system body dysfunctions with apparent ASD-related traits, commencing as early as childhood. The Adnp haploinsufficient (Adnp+/-) mouse model was researched before in relations to Alzheimer's disease and autism. Adnp+/- mice suffer from deficient social memory, vocal and motor impediments, irregular tooth eruption and short stature, all of which corresponds with reported phenotypes in patients with the ADNP syndrome. Recently, a more elaborated description of the ADNP syndrome was published, presenting impediments such as hearing disabilities in > 10% of the studied children. Irregular auditory brainstem response (ABR) has been connected to ASD-related cases and has been suggested as a potential hallmark for autism, allowing diagnosis of ASD risk and early intervention. Herein, we present detriment hearing in the Adnp+/- mice with atypical ABR and significant protein expression irregularities that coincides with ASD and hearing loss studies in the brain.


Subject(s)
Autism Spectrum Disorder/complications , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing Loss/etiology , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Animals , Auditory Cortex , Autism Spectrum Disorder/genetics , Choline O-Acetyltransferase/metabolism , Female , Glutamate Decarboxylase/metabolism , Hair Cells, Auditory/cytology , Hearing Loss/genetics , Male , Mice , Mutation
12.
J Neurosci ; 37(8): 2073-2085, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28115485

ABSTRACT

Cellular asymmetries play crucial roles in development and organ function. The planar cell polarity (PCP) signaling pathway is involved in the establishment of cellular asymmetry within the plane of a cell sheet. Inner ear sensory hair cells (HCs), which have several rows of staircase-like stereocilia and one kinocilium located at the vertex of the stereocilia protruding from the apical surface of each HC, exhibit a typical form of PCP. Although connections between cilia and PCP signaling in vertebrate development have been reported, their precise nature is not well understood. During inner ear development, several ciliary proteins are known to play a role in PCP formation. In the current study, we investigated a functional role for intestinal cell kinase (Ick), which regulates intraflagellar transport (IFT) at the tip of cilia, in the mouse inner ear. A lack of Ick in the developing inner ear resulted in PCP defects in the cochlea, including misorientation or misshaping of stereocilia and aberrant localization of the kinocilium and basal body in the apical and middle turns, leading to auditory dysfunction. We also observed abnormal ciliary localization of Ift88 in both HCs and supporting cells. Together, our results show that Ick ciliary kinase is essential for PCP formation in inner ear HCs, suggesting that ciliary transport regulation is important for PCP signaling.SIGNIFICANCE STATEMENT The cochlea in the inner ear is the hearing organ. Planar cell polarity (PCP) in hair cells (HCs) in the cochlea is essential for mechanotransduction and refers to the asymmetric structure consisting of stereociliary bundles and the kinocilium on the apical surface of the cell body. We reported previously that a ciliary kinase, Ick, regulates intraflagellar transport (IFT). Here, we found that loss of Ick leads to abnormal localization of the IFT component in kinocilia, PCP defects in HCs, and hearing dysfunction. Our study defines the association of ciliary transport regulation with PCP formation in HCs and hearing function.


Subject(s)
Cell Polarity/genetics , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory, Inner/physiology , Hearing/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Antigens/metabolism , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Embryo, Mammalian , Evoked Potentials, Auditory, Brain Stem/genetics , Hair Cells, Auditory, Inner/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otoacoustic Emissions, Spontaneous/genetics , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Protein Serine-Threonine Kinases/genetics
13.
J Neurochem ; 146(4): 459-473, 2018 08.
Article in English | MEDLINE | ID: mdl-29675997

ABSTRACT

Previous studies have convincingly argued that reactive oxygen species (ROS) contribute to the development of several major types of sensorineural hearing loss, such as noise-induced hearing loss (NIHL), drug-induced hearing loss, and age-related hearing loss. However, the underlying molecular mechanisms induced by ROS in these pathologies remain unclear. To resolve this issue, we established an in vivo model of ROS overproduction by generating a transgenic (TG) mouse line expressing the human NADPH oxidase 4 (NOX4, NOX4-TG mice), which is a constitutively active ROS-producing enzyme that does not require stimulation or an activator. Overproduction of ROS was detected at the cochlea of the inner ear in NOX4-TG mice, but they showed normal hearing function under baseline conditions. However, they demonstrated hearing function vulnerability, especially at high-frequency sounds, upon exposure to intense noise, which was accompanied by loss of cochlear outer hair cells (OHCs). The vulnerability to loss of hearing function and OHCs was rescued by treatment with the antioxidant Tempol. Additionally, we found increased protein levels of the heat-shock protein 47 (HSP47) in models using HEK293 cells, including H2 O2 treatment and cells with stable and transient expression of NOX4. Furthermore, the up-regulated levels of Hsp47 were observed in both the cochlea and heart of NOX4-TG mice. Thus, antioxidant therapy is a promising approach for the treatment of NIHL. Hsp47 may be an endogenous antioxidant factor, compensating for the chronic ROS overexposure in vivo, and counteracting ROS-related hearing loss.


Subject(s)
Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/physiopathology , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aldehydes/metabolism , Animals , Cochlea/metabolism , Cochlea/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Gene Expression Regulation/genetics , HEK293 Cells , HSP47 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/metabolism , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/pathology , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , NADPH Oxidase 4/metabolism , Transfection
14.
Mol Ther ; 25(2): 379-391, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28082074

ABSTRACT

Adeno-associated virus (AAV) is a safe and effective vector for gene therapy for retinal disorders. Gene therapy for hearing disorders is not as advanced, in part because gene delivery to sensory hair cells of the inner ear is inefficient. Although AAV transduces the inner hair cells of the mouse cochlea, outer hair cells remain refractory to transduction. Here, we demonstrate that a vector, exosome-associated AAV (exo-AAV), is a potent carrier of transgenes to all inner ear hair cells. Exo-AAV1-GFP is more efficient than conventional AAV1-GFP, both in mouse cochlear explants in vitro and with direct cochlear injection in vivo. Exo-AAV shows no toxicity in vivo, as assayed by tests of auditory and vestibular function. Finally, exo-AAV1 gene therapy partially rescues hearing in a mouse model of hereditary deafness (lipoma HMGIC fusion partner-like 5/tetraspan membrane protein of hair cell stereocilia [Lhfpl5/Tmhs-/-]). Exo-AAV is a powerful gene delivery system for hair cell research and may be useful for gene therapy for deafness.


Subject(s)
Dependovirus/genetics , Exosomes/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , Hair Cells, Auditory, Inner/metabolism , Hearing/genetics , Animals , Cells, Cultured , Dependovirus/classification , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Gene Expression , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Male , Mice , Mice, Knockout , Phenotype , Transduction, Genetic , Transgenes
15.
Cogn Behav Neurol ; 31(2): 53-78, 2018 06.
Article in English | MEDLINE | ID: mdl-29927797

ABSTRACT

Atypical responses to sound are common in individuals with autism spectrum disorder (ASD), and growing evidence suggests an underlying auditory brainstem pathology. This review of the literature provides a comprehensive account of the structural and functional evidence for auditory brainstem abnormalities in ASD. The studies reviewed were published between 1975 and 2016 and were sourced from multiple online databases. Indices of both the quantity and quality of the studies reviewed are considered. Findings show converging evidence for auditory brainstem pathology in ASD, although the specific functions and anatomical structures involved remain equivocal. Two main trends emerge from the literature: (1) abnormalities occur mainly at higher levels of the auditory brainstem, according to structural imaging and electrophysiology studies; and (2) brainstem abnormalities appear to be more common in younger than older children with ASD. These findings suggest delayed maturation of neural transmission pathways between lower and higher levels of the brainstem and are consistent with the auditory disorders commonly observed in ASD, including atypical sound sensitivity, poor sound localization, and difficulty listening in background noise. Limitations of existing studies are discussed, and recommendations for future research are offered.


Subject(s)
Autism Spectrum Disorder/pathology , Brain Stem/pathology , Evoked Potentials, Auditory, Brain Stem/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Young Adult
16.
Mol Cell Neurosci ; 80: 123-133, 2017 04.
Article in English | MEDLINE | ID: mdl-28286294

ABSTRACT

An important hallmark of various neurodegenerative disorders is the proliferation and activation of microglial cells, the resident immune cells of the central nervous system (CNS). Mice that lack multifunctional protein-2 (MFP2), the key enzyme in peroxisomal ß-oxidation, develop excessive microgliosis that positively correlates with behavioral deficits whereas no neuronal loss occurs. However, the precise contribution of neuroinflammation to the fatal neuropathology of MFP2 deficiency remains largely unknown. Here, we first attempted to suppress the inflammatory response by administering various anti-inflammatory drugs but they failed to reduce microgliosis. Subsequently, Mfp2-/- mice were treated with the selective colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 as microglial proliferation and survival is dependent on CSF1R signaling. This resulted in the elimination of >95% of microglia from control mice but only 70% of the expanded microglial population from Mfp2-/- mice. Despite microglial diminution in Mfp2-/- brain, inflammatory markers remained unaltered and residual microglia persisted in a reactive state. CSF1R inhibition did not prevent neuronal dysfunction, cognitive decline and clinical deterioration of Mfp2-/- mice. Collectively, the unaltered inflammatory profile despite suppressed microgliosis concurrent with persevering clinical decline strengthens our hypothesis that neuroinflammation importantly contributes to the Mfp2-/- phenotype.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalitis , Gliosis/etiology , Peroxisomal Multifunctional Protein-2/deficiency , Acoustic Stimulation , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Differentiation/metabolism , Avoidance Learning/drug effects , Avoidance Learning/physiology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Encephalitis/complications , Encephalitis/genetics , Encephalitis/pathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Muscle Strength/drug effects , Muscle Strength/genetics , Peroxisomal Multifunctional Protein-2/genetics , Severity of Illness Index
17.
J Neurosci ; 36(27): 7198-209, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27383594

ABSTRACT

UNLABELLED: During selective attention, subjects voluntarily focus their cognitive resources on a specific stimulus while ignoring others. Top-down filtering of peripheral sensory responses by higher structures of the brain has been proposed as one of the mechanisms responsible for selective attention. A prerequisite to accomplish top-down modulation of the activity of peripheral structures is the presence of corticofugal pathways. The mammalian auditory efferent system is a unique neural network that originates in the auditory cortex and projects to the cochlear receptor through the olivocochlear bundle, and it has been proposed to function as a top-down filter of peripheral auditory responses during attention to cross-modal stimuli. However, to date, there is no conclusive evidence of the involvement of olivocochlear neurons in selective attention paradigms. Here, we trained wild-type and α-9 nicotinic receptor subunit knock-out (KO) mice, which lack cholinergic transmission between medial olivocochlear neurons and outer hair cells, in a two-choice visual discrimination task and studied the behavioral consequences of adding different types of auditory distractors. In addition, we evaluated the effects of contralateral noise on auditory nerve responses as a measure of the individual strength of the olivocochlear reflex. We demonstrate that KO mice have a reduced olivocochlear reflex strength and perform poorly in a visual selective attention paradigm. These results confirm that an intact medial olivocochlear transmission aids in ignoring auditory distraction during selective attention to visual stimuli. SIGNIFICANCE STATEMENT: The auditory efferent system is a neural network that originates in the auditory cortex and projects to the cochlear receptor through the olivocochlear system. It has been proposed to function as a top-down filter of peripheral auditory responses during attention to cross-modal stimuli. However, to date, there is no conclusive evidence of the involvement of olivocochlear neurons in selective attention paradigms. Here, we studied the behavioral consequences of adding different types of auditory distractors in a visual selective attention task in wild-type and α-9 nicotinic receptor knock-out (KO) mice. We demonstrate that KO mice perform poorly in the selective attention paradigm and that an intact medial olivocochlear transmission aids in ignoring auditory distractors during attention.


Subject(s)
Attention/physiology , Auditory Pathways/physiology , Auditory Perception/physiology , Choice Behavior/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Receptors, Nicotinic/deficiency , Acoustic Stimulation , Analysis of Variance , Animals , Cochlear Nerve/physiology , Electroencephalography , Evoked Potentials, Auditory, Brain Stem/genetics , Male , Mice , Mice, Knockout , Photic Stimulation , Receptors, Nicotinic/genetics , Time Factors
18.
J Neurosci ; 36(23): 6175-85, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27277796

ABSTRACT

UNLABELLED: Congenital sensory deprivation can lead to reorganization of the deprived cortical regions by another sensory system. Such cross-modal reorganization may either compete with or complement the "original" inputs to the deprived area after sensory restoration and can thus be either adverse or beneficial for sensory restoration. In congenital deafness, a previous inactivation study documented that supranormal visual behavior was mediated by higher-order auditory fields in congenitally deaf cats (CDCs). However, both the auditory responsiveness of "deaf" higher-order fields and interactions between the reorganized and the original sensory input remain unknown. Here, we studied a higher-order auditory field responsible for the supranormal visual function in CDCs, the auditory dorsal zone (DZ). Hearing cats and visual cortical areas served as a control. Using mapping with microelectrode arrays, we demonstrate spatially scattered visual (cross-modal) responsiveness in the DZ, but show that this did not interfere substantially with robust auditory responsiveness elicited through cochlear implants. Visually responsive and auditory-responsive neurons in the deaf auditory cortex formed two distinct populations that did not show bimodal interactions. Therefore, cross-modal plasticity in the deaf higher-order auditory cortex had limited effects on auditory inputs. The moderate number of scattered cross-modally responsive neurons could be the consequence of exuberant connections formed during development that were not pruned postnatally in deaf cats. Although juvenile brain circuits are modified extensively by experience, the main driving input to the cross-modally (visually) reorganized higher-order auditory cortex remained auditory in congenital deafness. SIGNIFICANCE STATEMENT: In a common view, the "unused" auditory cortex of deaf individuals is reorganized to a compensatory sensory function during development. According to this view, cross-modal plasticity takes over the unused cortex and reassigns it to the remaining senses. Therefore, cross-modal plasticity might conflict with restoration of auditory function with cochlear implants. It is unclear whether the cross-modally reorganized auditory areas lose auditory responsiveness. We show that the presence of cross-modal plasticity in a higher-order auditory area does not reduce auditory responsiveness of that area. Visual reorganization was moderate, spatially scattered and there were no interactions between cross-modally reorganized visual and auditory inputs. These results indicate that cross-modal reorganization is less detrimental for neurosensory restoration than previously thought.


Subject(s)
Auditory Cortex/pathology , Cochlear Implantation , Deafness/physiopathology , Deafness/therapy , Neurons/physiology , Acoustic Stimulation , Analysis of Variance , Animals , Auditory Cortex/physiopathology , Brain Mapping , Cats , Cochlear Implants , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Male , Models, Biological , Photic Stimulation
19.
J Neurosci ; 36(25): 6803-19, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27335410

ABSTRACT

UNLABELLED: The PKR-like endoplasmic reticulum kinase (PERK) pathway of the unfolded protein response (UPR) is protective against toxic accumulations of misfolded proteins in the endoplasmic reticulum, but is thought to drive cell death via the transcription factor, CHOP. However, in many cell types, CHOP is an obligate step in the PERK pathway, which frames the conundrum of a prosurvival pathway that kills cells. Our laboratory and others have previously demonstrated the prosurvival activity of the PERK pathway in oligodendrocytes. In the current study, we constitutively overexpress CHOP in myelinating cells during development and into adulthood under normal or UPR conditions. We show that this transcription factor does not drive apoptosis. Indeed, we observe no detriment in mice at multiple levels from single cells to mouse behavior and life span. In light of these data and other studies, we reinterpret PERK pathway function in the context of a stochastic vulnerability model, which governs the likelihood that cells undergo cell death upon cessation of UPR protection and while attempting to restore homeostasis. SIGNIFICANCE STATEMENT: Herein, we tackle the biggest controversy in the UPR literature: the function of the transcription factor CHOP as a protective or a prodeath factor. This manuscript is timely in light of the 2014 Lasker award for the UPR. Our in vivo data show that CHOP is not a prodeath protein, and we demonstrate that myelinating glial cells function normally in the presence of high CHOP expression from development to adulthood. Further, we propose a simplified view of UPR-mediated cell death after CHOP induction. We anticipate our work may turn the tide of the dogmatic view of CHOP and cause a reinvestigation of its function in different cell types. Accordingly, we believe our work will be a watershed for the UPR field.


Subject(s)
Nerve Fibers, Myelinated/metabolism , Phenotype , Stress, Physiological/physiology , Transcription Factor CHOP/metabolism , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Cell Line, Tumor , Evoked Potentials, Auditory, Brain Stem/genetics , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Optic Nerve/pathology , Psychomotor Performance/physiology , Signal Transduction/genetics , Spinal Cord/pathology , Transcription Factor CHOP/genetics
20.
J Neurosci ; 36(35): 9201-16, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27581460

ABSTRACT

UNLABELLED: Neuroplastin (Nptn) is a member of the Ig superfamily and is expressed in two isoforms, Np55 and Np65. Np65 regulates synaptic transmission but the function of Np55 is unknown. In an N-ethyl-N-nitrosaurea mutagenesis screen, we have now generated a mouse line with an Nptn mutation that causes deafness. We show that Np55 is expressed in stereocilia of outer hair cells (OHCs) but not inner hair cells and affects interactions of stereocilia with the tectorial membrane. In vivo vibrometry demonstrates that cochlear amplification is absent in Nptn mutant mice, which is consistent with the failure of OHC stereocilia to maintain stable interactions with the tectorial membrane. Hair bundles show morphological defects as the mutant mice age and while mechanotransduction currents can be evoked in early postnatal hair cells, cochlea microphonics recordings indicate that mechanontransduction is affected as the mutant mice age. We thus conclude that differential splicing leads to functional diversification of Nptn, where Np55 is essential for OHC function, while Np65 is implicated in the regulation of synaptic function. SIGNIFICANCE STATEMENT: Amplification of input sound signals, which is needed for the auditory sense organ to detect sounds over a wide intensity range, depends on mechanical coupling of outer hair cells to the tectorial membrane. The current study shows that neuroplastin, a member of the Ig superfamily, which has previously been linked to the regulation of synaptic plasticity, is critical to maintain a stable mechanical link of outer hair cells with the tectorial membrane. In vivo recordings demonstrate that neuroplastin is essential for sound amplification and that mutation in neuroplastin leads to auditory impairment in mice.


Subject(s)
Hair Cells, Auditory, Outer/cytology , Mechanotransduction, Cellular/physiology , Membrane Glycoproteins/metabolism , Stereocilia/physiology , Acoustic Stimulation , Animals , Animals, Newborn , DNA Mutational Analysis , Deafness/genetics , Deafness/pathology , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory, Inner/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Mutation/genetics , Otoacoustic Emissions, Spontaneous/genetics , Patch-Clamp Techniques , Physical Stimulation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/genetics , RNA, Messenger/metabolism , Stereocilia/ultrastructure , Tomography, Optical Coherence , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL