Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
EMBO Mol Med ; 11(9): e10288, 2019 09.
Article in English | MEDLINE | ID: mdl-31448880

ABSTRACT

Hearing relies on mechanically gated ion channels present in the actin-rich stereocilia bundles at the apical surface of cochlear hair cells. Our knowledge of the mechanisms underlying the formation and maintenance of the sound-receptive structure is limited. Utilizing a large-scale forward genetic screen in mice, genome mapping and gene complementation tests, we identified Clrn2 as a new deafness gene. The Clrn2clarinet/clarinet mice (p.Trp4* mutation) exhibit a progressive, early-onset hearing loss, with no overt retinal deficits. Utilizing data from the UK Biobank study, we could show that CLRN2 is involved in human non-syndromic progressive hearing loss. Our in-depth morphological, molecular and functional investigations establish that while it is not required for initial formation of cochlear sensory hair cell stereocilia bundles, clarin-2 is critical for maintaining normal bundle integrity and functioning. In the differentiating hair bundles, lack of clarin-2 leads to loss of mechano-electrical transduction, followed by selective progressive loss of the transducing stereocilia. Together, our findings demonstrate a key role for clarin-2 in mammalian hearing, providing insights into the interplay between mechano-electrical transduction and stereocilia maintenance.


Subject(s)
Hearing Loss/metabolism , Stereocilia/metabolism , Adult , Aged , Animals , Cohort Studies , Female , Hair Cells, Auditory/metabolism , Hearing , Hearing Loss/genetics , Hearing Loss/physiopathology , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Middle Aged , Stereocilia/genetics
2.
Proc Natl Acad Sci U S A ; 116(22): 11000-11009, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31097578

ABSTRACT

The pathogenic variant c.144T>G (p.N48K) in the clarin1 gene (CLRN1) results in progressive loss of vision and hearing in Usher syndrome IIIA (USH3A) patients. CLRN1 is predicted to be an essential protein in hair bundles, the mechanosensory structure of hair cells critical for hearing and balance. When expressed in animal models, CLRN1 localizes to the hair bundle, whereas glycosylation-deficient CLRN1N48K aggregates in the endoplasmic reticulum, with only a fraction reaching the bundle. We hypothesized that the small amount of CLRN1N48K that reaches the hair bundle does so via an unconventional secretory pathway and that activation of this pathway could be therapeutic. Using genetic and pharmacological approaches, we find that clarin1 knockout (clrn1KO/KO ) zebrafish that express the CLRN1c.144T>G pathogenic variant display progressive hair cell dysfunction, and that CLRN1N48K is trafficked to the hair bundle via the GRASP55 cargo-dependent unconventional secretory pathway (GCUSP). On expression of GRASP55 mRNA, or on exposure to the drug artemisinin (which activates GCUSP), the localization of CLRN1N48K to the hair bundles was enhanced. Artemisinin treatment also effectively restored hair cell mechanotransduction and attenuated progressive hair cell dysfunction in clrn1KO/KO larvae that express CLRN1c.144T>G , highlighting the potential of artemisinin to prevent sensory loss in CLRN1c.144T>G patients.


Subject(s)
Hair Cells, Auditory/physiology , Mechanotransduction, Cellular/genetics , Membrane Proteins , Secretory Pathway/genetics , Animals , Animals, Genetically Modified , Artemisinins/pharmacology , Hair Cells, Auditory/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Zebrafish
3.
Sci Rep ; 7(1): 13480, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29044151

ABSTRACT

Usher syndrome type III (USH3) characterized by progressive loss of vision and hearing is caused by mutations in the clarin-1 gene (CLRN1). Clrn1 knockout (KO) mice develop hair cell defects by postnatal day 2 (P2) and are deaf by P21-P25. Early onset profound hearing loss in KO mice and lack of information about the cochlear cell type that requires Clrn1 expression pose challenges to therapeutic investigation. We generated KO mice harboring a transgene, TgAC1, consisting of Clrn1-UTR (Clrn1 cDNA including its 5' and 3' UTR) under the control of regulatory elements (Atoh1 3' enhancer/ß-globin basal promoter) to direct expression of Clrn1 in hair cells during development and down regulate it postnatally. The KO-TgAC1 mice displayed delayed onset progressive hearing loss associated with deterioration of the hair bundle structure, leading to the hypothesis that hair cell expression of Clrn1 is essential for postnatal preservation of hair cell structure and hearing. Consistent with that hypothesis, perinatal transfection of hair cells in KO-TgAC1 mice with a single injection of AAV-Clrn1-UTR vector showed correlative preservation of the hair bundle structure and hearing through adult life. Further, the efficacy of AAV-Clrn1 vector was significantly attenuated, revealing the potential importance of UTR in gene therapy.


Subject(s)
Hearing Loss/diagnosis , Hearing Loss/etiology , Usher Syndromes/complications , Animals , Base Sequence , Dependovirus/genetics , Disease Models, Animal , Gene Expression , Gene Order , Genetic Vectors/genetics , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Hearing Loss/prevention & control , Humans , Immunohistochemistry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Organ Specificity , Phenotype , Protein Transport , Transduction, Genetic , Usher Syndromes/diagnosis , Usher Syndromes/etiology
4.
Front Cell Neurosci ; 11: 393, 2017.
Article in English | MEDLINE | ID: mdl-29311828

ABSTRACT

Cisplatin and other related platinum antineoplastic drugs are commonly used in the treatment of a variety of cancers in both adults and children but are often associated with severe side effects, including hearing loss. Cisplatin's ototoxic effects are multifaceted, culminating in irreversible damage to the mechanosensory hair cells in the inner ear. Platinum drugs act on cancerous cells by forming nuclear DNA adducts, which may initiate signaling leading to cell cycle arrest or apoptosis. Moreover, it was reported that cisplatin may induce mitochondrial DNA damage in non-cancerous cells. Therefore, protecting mitochondria may alleviate cisplatin-induced insult to non-proliferating cells. Thus, it is important to identify agents that shield the mitochondria from cisplatin-induced insult without compromising the anti-tumor actions of the platinum-based drugs. In this study we tested the protective properties of mitochondrial division inhibitor, mdivi-1, a derivative of quinazolinone and a regulator of mitochondrial fission. Interestingly, it has been reported that mdivi-1 increases the apoptosis of cells that are resistant to cisplatin. The ability of mdivi-1 to protect hair cells against cisplatin-induced toxicity was evaluated in a fish model. Wild-type (Tübingen strain), cdh23 mutant, and transgenic pvalb3b::GFP zebrafish stably expressing GFP in the hair cells were used in this study. Larvae at 5-6 days post fertilization were placed in varying concentrations of cisplatin (50-200 µM) and/or mdivi-1 (1-10 µM) for 16 h. To evaluate hair cell's viability the number of hair bundles per neuromast were counted. To assess hair cell function, we used the FM1-43 uptake assay and recordings of neuromast microphonic potentials. The results showed that mdivi-1 protected hair cells of lateral line neuromasts when they were challenged by 50 µM of cisplatin: viability of hair cells increased almost twice from 19% ± 1.8% to 36% ± 2.0% (p < 0.001). No protection was observed when higher concentrations of cisplatin were used. In addition, our data were in accord with previously reported results that functional mechanotransduction strongly potentiates cisplatin-induced hair cell toxicity. Together, our results suggest that mitochondrial protection may prevent cisplatin-induced damage to hair cells.

5.
Nat Chem Biol ; 12(6): 444-51, 2016 06.
Article in English | MEDLINE | ID: mdl-27110679

ABSTRACT

Usher syndrome type III (USH3), characterized by progressive deafness, variable balance disorder and blindness, is caused by destabilizing mutations in the gene encoding the clarin-1 (CLRN1) protein. Here we report a new strategy to mitigate hearing loss associated with a common USH3 mutation CLRN1(N48K) that involves cell-based high-throughput screening of small molecules capable of stabilizing CLRN1(N48K), followed by a secondary screening to eliminate general proteasome inhibitors, and finally an iterative process to optimize structure-activity relationships. This resulted in the identification of BioFocus 844 (BF844). To test the efficacy of BF844, we developed a mouse model that mimicked the progressive hearing loss associated with USH3. BF844 effectively attenuated progressive hearing loss and prevented deafness in this model. Because the CLRN1(N48K) mutation causes both hearing and vision loss, BF844 could in principle prevent both sensory deficiencies in patients with USH3. Moreover, the strategy described here could help identify drugs for other protein-destabilizing monogenic disorders.


Subject(s)
Disease Models, Animal , Membrane Proteins/antagonists & inhibitors , Pyrazoles/pharmacology , Pyridazines/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Usher Syndromes/drug therapy , Animals , High-Throughput Screening Assays , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyridazines/therapeutic use , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Usher Syndromes/genetics
6.
J Neurosci ; 35(28): 10188-201, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26180195

ABSTRACT

Usher syndrome type III (USH3) is characterized by progressive loss of hearing and vision, and varying degrees of vestibular dysfunction. It is caused by mutations that affect the human clarin-1 protein (hCLRN1), a member of the tetraspanin protein family. The missense mutation CLRN1(N48K), which affects a conserved N-glycosylation site in hCLRN1, is a common causative USH3 mutation among Ashkenazi Jews. The affected individuals hear at birth but lose that function over time. Here, we developed an animal model system using zebrafish transgenesis and gene targeting to provide an explanation for this phenotype. Immunolabeling demonstrated that Clrn1 localized to the hair cell bundles (hair bundles). The clrn1 mutants generated by zinc finger nucleases displayed aberrant hair bundle morphology with diminished function. Two transgenic zebrafish that express either hCLRN1 or hCLRN1(N48K) in hair cells were produced to examine the subcellular localization patterns of wild-type and mutant human proteins. hCLRN1 localized to the hair bundles similarly to zebrafish Clrn1; in contrast, hCLRN1(N48K) largely mislocalized to the cell body with a small amount reaching the hair bundle. We propose that this small amount of hCLRN1(N48K) in the hair bundle provides clarin-1-mediated function during the early stages of life; however, the presence of hCLRN1(N48K) in the hair bundle diminishes over time because of intracellular degradation of the mutant protein, leading to progressive loss of hair bundle integrity and hair cell function. These findings and genetic tools provide an understanding and path forward to identify therapies to mitigate hearing loss linked to the CLRN1 mutation. SIGNIFICANCE STATEMENT: Mutations in the clarin-1 gene affect eye and ear function in humans. Individuals with the CLRN1(N48K) mutation are born able to hear but lose that function over time. Here, we develop an animal model system using zebrafish transgenesis and gene targeting to provide an explanation for this phenotype. This approach illuminates the role of clarin-1 and the molecular mechanism linked to the CLRN1(N48K) mutation in sensory hair cells of the inner ear. Additionally, the investigation provided an in vivo model to guide future drug discovery to rescue the hCLRN1(N48K) in hair cells.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory/pathology , Membrane Proteins/metabolism , Usher Syndromes/pathology , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Auditory Pathways/metabolism , Auditory Pathways/pathology , Body Patterning/drug effects , Body Patterning/genetics , Cadherins/genetics , Disease Models, Animal , Endodeoxyribonucleases/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Genotype , Hearing Loss/genetics , Humans , Larva , Male , Membrane Proteins/genetics , Mutation/genetics , Postural Balance/genetics , Sequence Analysis, Protein , Synapses/metabolism , Synapses/pathology , Usher Syndromes/complications , Usher Syndromes/genetics , Vision Disorders/etiology , Zebrafish , Zebrafish Proteins/genetics
7.
J Neurosci ; 33(10): 4395-404, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23467356

ABSTRACT

In hair cells of the inner ear, sound or head movement increases tension in fine filaments termed tip links, which in turn convey force to mechanosensitive ion channels to open them. Tip links are formed by a tetramer of two cadherin proteins: protocadherin 15 (PCDH15) and cadherin 23 (CDH23), which have 11 and 27 extracellular cadherin (EC) repeats, respectively. Mutations in either protein cause inner ear disorders in mice and humans. We showed recently that these two cadherins bind tip-to-tip in a "handshake" mode that involves the EC1 and EC2 repeats of both proteins. However, a paucity of appropriate animal models has slowed our understanding both of the interaction and of how mutations of residues within the predicted interface compromise tip link integrity. Here, we present noddy, a new mouse model for hereditary deafness. Identified in a forward genetic screen, noddy homozygotes lack inner ear function. Mapping and sequencing showed that noddy mutant mice harbor an isoleucine-to-asparagine (I108N) mutation in the EC1 repeat of PCDH15. Residue I108 interacts with CDH23 EC2 in the handshake and its mutation impairs the interaction in vitro. The noddy mutation allowed us to determine the consequences of blocking the handshake in vivo: tip link formation and bundle morphology are disrupted, and mechanotransduction channels fail to remain open at rest. These results offer new insights into the interaction between PCDH15 and CDH23 and help explain the etiology of human deafness linked to mutations in the tip-link interface.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Hair Cells, Auditory/metabolism , Labyrinth Diseases , Mechanotransduction, Cellular/physiology , Mutation, Missense/genetics , Protein Precursors/genetics , Age Factors , Animals , Animals, Newborn , Cadherin Related Proteins , Calcium/metabolism , Cells, Cultured , Electroencephalography , Ethylnitrosourea/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genotype , Hair Cells, Auditory/pathology , Hair Cells, Auditory/ultrastructure , Hearing Loss/chemically induced , Hearing Loss/genetics , Labyrinth Diseases/chemically induced , Labyrinth Diseases/genetics , Labyrinth Diseases/pathology , Labyrinth Diseases/physiopathology , Mice , Mice, Transgenic , Microscopy, Atomic Force , Mutagens/pharmacology , Mutation, Missense/drug effects , Phenotype , Polymorphism, Single Nucleotide/genetics , Protein Binding/drug effects , Protein Binding/genetics , Pyridinium Compounds , Quaternary Ammonium Compounds
SELECTION OF CITATIONS
SEARCH DETAIL
...