Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 26.
Article in English | MEDLINE | ID: mdl-38826418

ABSTRACT

Hair cells (HCs) are the sensory receptors of the auditory and vestibular systems in the inner ears of vertebrates that selectively transduce mechanical stimuli into electrical activity. Although all HCs have the hallmark stereocilia bundle for mechanotransduction, HCs in non-mammals and mammals differ in their molecular specialization in the apical, basolateral and synaptic membranes. HCs of non-mammals, such as zebrafish (zHCs), are electrically tuned to specific frequencies and possess an active process in the stereocilia bundle to amplify sound signals. Mammalian cochlear HCs, in contrast, are not electrically tuned and achieve amplification by somatic motility of outer HCs (OHCs). To understand the genetic mechanisms underlying differences among adult zebrafish and mammalian cochlear HCs, we compared their RNA-seq-characterized transcriptomes, focusing on protein-coding orthologous genes related to HC specialization. There was considerable shared expression of gene orthologs among the HCs, including those genes associated with mechanotransduction, ion transport/channels, and synaptic signaling. For example, both zebrafish and mouse HCs express Tmc1, Lhfpl5, Tmie, Cib2, Cacna1d, Cacnb2, Otof, Pclo and Slc17a8. However, there were some notable differences in expression among zHCs, OHCs, and inner HCs (IHCs), which likely underlie the distinctive physiological properties of each cell type. Tmc2 and Cib3 were not detected in adult mouse HCs but tmc2a and b and cib3 were highly expressed in zHCs. Mouse HCs express Kcna10, Kcnj13, Kcnj16, and Kcnq4, which were not detected in zHCs. Chrna9 and Chrna10 were expressed in mouse HCs. In contrast, chrna10 was not detected in zHCs. OHCs highly express Slc26a5 which encodes the motor protein prestin that contributes to OHC electromotility. However, zHCs have only weak expression of slc26a5, and subsequently showed no voltage dependent electromotility when measured. Notably, the zHCs expressed more paralogous genes including those associated with HC-specific functions and transcriptional activity, though it is unknown whether they have functions similar to their mammalian counterparts. There was overlap in the expressed genes associated with a known hearing phenotype. Our analyses unveil substantial differences in gene expression patterns that may explain phenotypic specialization of zebrafish and mouse HCs. This dataset also includes several protein-coding genes to further the functional characterization of HCs and study of HC evolution from non-mammals to mammals.

2.
Sci Adv ; 10(25): eadk2299, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38896614

ABSTRACT

Noise-induced hearing loss (NIHL) is a common sensorineural hearing impairment that lacks U.S. Food and Drug Administration-approved drugs. To fill the gap in effective screening models, we used an in silico transcriptome-based drug screening approach, identifying 22 biological pathways and 64 potential small molecule treatments for NIHL. Two of these, afatinib and zorifertinib [epidermal growth factor receptor (EGFR) inhibitors], showed efficacy in zebrafish and mouse models. Further tests with EGFR knockout mice and EGF-morpholino zebrafish confirmed their protective role against NIHL. Molecular studies in mice highlighted EGFR's crucial involvement in NIHL and the protective effect of zorifertinib. When given orally, zorifertinib was found in the perilymph with favorable pharmacokinetics. In addition, zorifertinib combined with AZD5438 (a cyclin-dependent kinase 2 inhibitor) synergistically prevented NIHL in zebrafish. Our results underscore the potential for in silico transcriptome-based drug screening in diseases lacking efficient models and suggest EGFR inhibitors as potential treatments for NIHL, meriting clinical trials.


Subject(s)
ErbB Receptors , Hearing Loss, Noise-Induced , Transcriptome , Zebrafish , Animals , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , ErbB Receptors/genetics , Mice , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/genetics , Disease Models, Animal , Computer Simulation , Protein Kinase Inhibitors/pharmacology , Humans , Drug Evaluation, Preclinical , Mice, Knockout , Gene Expression Profiling
3.
Hear Res ; 436: 108831, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37393735

ABSTRACT

Pediatric auditory neuropathy spectrum disorder is a particular type of hearing loss caused by abnormal sound transmission from the cochlea to the brain. It is due to defective peripheral synaptic function or improper neuronal conduction. Using trio whole-exome sequencing, we have identified novel biallelic variants in the PLEC gene in three individuals with profound deafness from two unrelated families. Among them, one pediatric patient diagnosed with auditory neuropathy spectrum disorder had a good cochlear implantation outcome. The other two adult patients were diagnosed with non-syndromic hearing loss. Studies in mice and zebrafish confirmed that plectin is developmentally expressed in the inner ear. Moreover, plectin's knockdown resulted in a reduction of synaptic mitochondrial potential and loss of ribbon synapses, reinforcing the idea of a role for plectin in neuronal transmission. Altogether, the results presented here, point to a new unconventional role for plectin in the inner ear. Contrary to the well-characterized association of plectin to skin and muscle diseases, we found that specific plectin mutations can result in hearing loss with no other clinical manifestations. This is important because 1) it provides evidence of plectin's involvement in inner ear function and 2) it will help clinicians at the time of diagnosis and treatment.


Subject(s)
Deafness , Hearing Loss , Mice , Animals , Plectin/genetics , Zebrafish/genetics , Hearing Loss/genetics
4.
bioRxiv ; 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37333346

ABSTRACT

Noise-Induced Hearing Loss (NIHL) represents a widespread disease for which no therapeutics have been approved by the Food and Drug Administration (FDA). Addressing the conspicuous void of efficacious in vitro or animal models for high throughput pharmacological screening, we utilized an in silico transcriptome-oriented drug screening strategy, unveiling 22 biological pathways and 64 promising small molecule candidates for NIHL protection. Afatinib and zorifertinib, both inhibitors of the Epidermal Growth Factor Receptor (EGFR), were validated for their protective efficacy against NIHL in experimental zebrafish and murine models. This protective effect was further confirmed with EGFR conditional knockout mice and EGF knockdown zebrafish, both demonstrating protection against NIHL. Molecular analysis using Western blot and kinome signaling arrays on adult mouse cochlear lysates unveiled the intricate involvement of several signaling pathways, with particular emphasis on EGFR and its downstream pathways being modulated by noise exposure and Zorifertinib treatment. Administered orally, Zorifertinib was successfully detected in the perilymph fluid of the inner ear in mice with favorable pharmacokinetic attributes. Zorifertinib, in conjunction with AZD5438 - a potent inhibitor of cyclin dependent kinase 2 - produced synergistic protection against NIHL in the zebrafish model. Collectively, our findings underscore the potential application of in silico transcriptome-based drug screening for diseases bereft of efficient screening models and posit EGFR inhibitors as promising therapeutic agents warranting clinical exploration for combatting NIHL. Highlights: In silico transcriptome-based drug screens identify pathways and drugs against NIHL.EGFR signaling is activated by noise but reduced by zorifertinib in mouse cochleae.Afatinib, zorifertinib and EGFR knockout protect against NIHL in mice and zebrafish.Orally delivered zorifertinib has inner ear PK and synergizes with a CDK2 inhibitor.

5.
Eur J Med Chem ; 226: 113849, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34560429

ABSTRACT

Cyclin-dependent kinase 2 (CDK2) is a potential therapeutic target for the treatment of hearing loss and cancer. Previously, we identified AZD5438 and AT7519-7 as potent inhibitors of CDK2, however, they also targeted additional kinases, leading to unwanted toxicities. Proteolysis Targeting Chimeras (PROTACs) are a new promising class of small molecules that can effectively direct specific proteins to proteasomal degradation. Herein we report the design, synthesis, and characterization of PROTACs of AT7519-7 and AZD5438 and the identification of PROTAC-8, an AZD5438-PROTAC, that exhibits selective, partial CDK2 degradation. Furthermore, PROTAC-8 protects against cisplatin ototoxicity and kainic acid excitotoxicity in zebrafish. Molecular dynamics simulations reveal the structural requirements for CDK2 degradation. Together, PROTAC-8 is among the first-in-class PROTACs with in vivo therapeutic activities and represents a new lead compound that can be further developed for better efficacy and selectivity for CDK2 degradation against hearing loss and cancer.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Hearing Loss, Noise-Induced/drug therapy , Imidazoles/pharmacology , Protective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line , Cisplatin/antagonists & inhibitors , Cisplatin/pharmacology , Cyclin-Dependent Kinase 2/metabolism , Dose-Response Relationship, Drug , Hearing Loss, Noise-Induced/metabolism , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Molecular Dynamics Simulation , Molecular Structure , Protective Agents/chemical synthesis , Protective Agents/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Zebrafish
6.
JCI Insight ; 6(5)2021 03 08.
Article in English | MEDLINE | ID: mdl-33476306

ABSTRACT

Hair cell loss is the leading cause of hearing and balance disorders in humans. It can be caused by many factors, including noise, aging, and therapeutic agents. Previous studies have shown the therapeutic potential of quinoxaline against drug-induced ototoxicity. Here, we screened a library of 68 quinoxaline derivatives for protection against aminoglycoside-induced damage of hair cells from the zebrafish lateral line. We identified quinoxaline-5-carboxylic acid (Qx28) as the best quinoxaline derivative that provides robust protection against both aminoglycosides and cisplatin in zebrafish and mouse cochlear explants. FM1-43 and aminoglycoside uptake, as well as antibiotic efficacy studies, revealed that Qx28 is neither blocking the mechanotransduction channels nor interfering with aminoglycoside antibacterial activity, suggesting that it may be protecting the hair cells by directly counteracting the ototoxin's mechanism of action. Only when animals were incubated with higher doses of Qx28 did we observe a partial blockage of the mechanotransduction channels. Finally, we assessed the regulation of the NF-κB pathway in vitro in mouse embryonic fibroblasts and in vivo in zebrafish larvae. Those studies showed that Qx28 protects hair cells by blocking NF-κB canonical pathway activation. Thus, Qx28 is a promising and versatile otoprotectant that can act across different species and toxins.


Subject(s)
Cell Death/drug effects , Hearing Loss , Ototoxicity/drug therapy , Protective Agents/pharmacology , Quinoxalines/pharmacology , Animals , Cells, Cultured , Fibroblasts , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Mice , Zebrafish
7.
Sci Adv ; 6(49)2020 12.
Article in English | MEDLINE | ID: mdl-33268358

ABSTRACT

Hearing loss caused by noise, aging, antibiotics, and chemotherapy affects 10% of the world population, yet there are no Food and Drug Administration (FDA)-approved drugs to prevent it. Here, we screened 162 small-molecule kinase-specific inhibitors for reduction of cisplatin toxicity in an inner ear cell line and identified dabrafenib (TAFINLAR), a BRAF kinase inhibitor FDA-approved for cancer treatment. Dabrafenib and six additional kinase inhibitors in the BRAF/MEK/ERK cellular pathway mitigated cisplatin-induced hair cell death in the cell line and mouse cochlear explants. In adult mice, oral delivery of dabrafenib repressed ERK phosphorylation in cochlear cells, and protected from cisplatin- and noise-induced hearing loss. Full protection was achieved in mice with co-treatment with oral AZD5438, a CDK2 kinase inhibitor. Our study explores a previously unidentified cellular pathway and molecular target BRAF kinase for otoprotection and may advance dabrafenib into clinics to benefit patients with cisplatin- and noise-induced ototoxicity.


Subject(s)
Antineoplastic Agents , Deafness , Hearing Loss , Animals , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Hair Cells, Auditory , Hearing Loss/etiology , Hearing Loss/prevention & control , Humans , Mice , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
8.
Sci Rep ; 9(1): 8720, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31197194

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

9.
Sci Rep ; 8(1): 15119, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30310154

ABSTRACT

Hair cell (HC) death is the leading cause of hearing and balance disorders in humans. It can be triggered by multiple insults, including noise, aging, and treatment with certain therapeutic drugs. As society becomes more technologically advanced, the source of noise pollution and the use of drugs with ototoxic side effects are rapidly increasing, posing a threat to our hearing health. Although the underlying mechanism by which ototoxins affect auditory function varies, they share common intracellular byproducts, particularly generation of reactive oxygen species. Here, we described the therapeutic effect of the heterocyclic compound quinoxaline (Qx) against ototoxic insults in zebrafish HCs. Animals incubated with Qx were protected against the deleterious effects of cisplatin and gentamicin, and partially against neomycin. In the presence of Qx, there was a reduction in the number of TUNEL-positive HCs. Since Qx did not block the mechanotransduction channels, based on FM1-43 uptake and microphonic potentials, this implies that Qx's otoprotective effect is at the intracellular level. Together, these results unravel a novel therapeutic role for Qx as an otoprotective drug against the deleterious side effects of cisplatin and aminoglycosides, offering an alternative option for patients treated with these compounds.

10.
J Cell Sci ; 130(21): 3698-3712, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28883094

ABSTRACT

The way an organism perceives its surroundings depends on sensory systems and the highly specialized cilia present in the neurosensory cells. Here, we describe the existence of an integrin α8 (Itga8) and protocadherin-15a (Pcdh15a) ciliary complex in neuromast hair cells in a zebrafish model. Depletion of the complex via downregulation or loss-of-function mutation leads to a dysregulation of cilia biogenesis and endocytosis. At the molecular level, removal of the complex blocks the access of Rab8a into the cilia as well as normal recruitment of ciliary cargo by centriolar satellites. These defects can be reversed by the introduction of a constitutively active form of Rhoa, suggesting that Itga8-Pcdh15a complex mediates its effect through the activation of this small GTPase and probably by the regulation of actin cytoskeleton dynamics. Our data points to a novel mechanism involved in the regulation of sensory cilia development, with the corresponding implications for normal sensory function.


Subject(s)
Cadherins/genetics , Cilia/metabolism , Hair Cells, Auditory/metabolism , Integrins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cadherin Related Proteins , Cadherins/antagonists & inhibitors , Cadherins/deficiency , Centrioles/metabolism , Centrioles/ultrastructure , Cilia/ultrastructure , Disease Models, Animal , Endocytosis , Gene Expression Regulation, Developmental , Hair Cells, Auditory/ultrastructure , Humans , Integrins/antagonists & inhibitors , Integrins/deficiency , Larva/genetics , Larva/growth & development , Larva/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Mutation , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Usher Syndromes/genetics , Usher Syndromes/metabolism , Usher Syndromes/pathology , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
11.
Hear Res ; 341: 100-108, 2016 11.
Article in English | MEDLINE | ID: mdl-27553900

ABSTRACT

Alport syndrome, a type IV collagen disorder, manifests as glomerular disease associated with hearing loss with thickening of the glomerular and strial capillary basement membranes (SCBMs). We have identified a role for endothelin-1 (ET-1) activation of endothelin A receptors (ETARs) in glomerular pathogenesis. Here we explore whether ET-1 plays a role in strial pathology. Wild type (WT) and Alport mice were treated with the ETAR antagonist, sitaxentan. The stria vascularis was analyzed for SCBM thickness and for extracellular matrix (ECM) proteins. Additional WT and Alport mice were exposed to noise or hypoxia and the stria analyzed for hypoxia-related and ECM genes. A strial marginal cell line cultured under hypoxic conditions, or stimulated with ET-1 was analyzed for expression of hypoxia-related and ECM transcripts. Noise exposure resulted in significantly elevated ABR thresholds in Alport mice relative to wild type littermates. Alport stria showed elevated expression of collagen α1(IV), laminin α2, and laminin α5 proteins relative to WT. SCBM thickening and elevated ECM protein expression was ameliorated by ETAR blockade. Stria from normoxic Alport mice and hypoxic WT mice showed upregulation of hypoxia-related, ECM, and ET-1 transcripts. Both ET-1 stimulation and hypoxia up-regulated ECM transcripts in cultured marginal cells. We conclude that ET-1 mediated activation of ETARs on strial marginal cells results in elevated expression of ECM genes and thickening of the SCBMs in Alport mice. SCBM thickening results in hypoxic stress further elevating ECM and ET-1 gene expression, exacerbating strial pathology.


Subject(s)
Endothelin-1/metabolism , Extracellular Matrix/genetics , Nephritis, Hereditary/physiopathology , Stria Vascularis/cytology , Animals , Basement Membrane/pathology , Body Temperature , Capillaries/pathology , Cell Line , Collagen Type IV/metabolism , Disease Models, Animal , Gene Expression Regulation , Hypoxia/pathology , Isoxazoles/chemistry , Laminin/metabolism , Mice , Oxidative Stress , Phenotype , Stria Vascularis/metabolism , Thiophenes/chemistry
12.
J Cell Biol ; 207(3): 375-91, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25365995

ABSTRACT

Clarin-1 is a four-transmembrane protein expressed by hair cells and photoreceptors. Mutations in its corresponding gene are associated with Usher syndrome type 3, characterized by late-onset and progressive hearing and vision loss in humans. Mice carrying mutations in the clarin-1 gene have hair bundle dysmorphology and a delay in synapse maturation. In this paper, we examined the expression and function of clarin-1 in zebrafish hair cells. We observed protein expression as early as 1 d postfertilization. Knockdown of clarin-1 resulted in inhibition of FM1-43 incorporation, shortening of the kinocilia, and mislocalization of ribeye b clusters. These phenotypes were fully prevented by co-injection with clarin-1 transcript, requiring its C-terminal tail. We also observed an in vivo interaction between clarin-1 and Pcdh15a. Altogether, our results suggest that clarin-1 is functionally important for mechanotransduction channel activity and for proper localization of synaptic components, establishing a critical role for clarin-1 at the apical and basal poles of hair cells.


Subject(s)
Hair Cells, Auditory/physiology , Mechanotransduction, Cellular , Membrane Proteins/physiology , Presynaptic Terminals/metabolism , Zebrafish Proteins/physiology , Animals , Cadherin Related Proteins , Cadherins/metabolism , Cilia/metabolism , Cilia/ultrastructure , Endocytosis , HeLa Cells , Humans , Protein Transport , Secretory Vesicles/metabolism , Usher Syndromes/genetics , Zebrafish , Zebrafish Proteins/metabolism
13.
PLoS One ; 9(6): e99083, 2014.
Article in English | MEDLINE | ID: mdl-24915008

ABSTRACT

It has been known for some time that laminins containing α1 and α2 chains, which are normally restricted to the mesangial matrix, accumulate in the glomerular basement membranes (GBM) of Alport mice, dogs, and humans. We show that laminins containing the α2 chain, but not those containing the α1 chain activates focal adhesion kinase (FAK) on glomerular podocytes in vitro and in vivo. CD151-null mice, which have weakened podocyte adhesion to the GBM rendering these mice more susceptible to biomechanical strain in the glomerulus, also show progressive accumulation of α2 laminins in the GBM, and podocyte FAK activation. Analysis of glomerular mRNA from both models demonstrates significant induction of MMP-9, MMP-10, MMP-12, MMPs linked to GBM destruction in Alport disease models, as well as the pro-inflammatory cytokine IL-6. SiRNA knockdown of FAK in cultured podocytes significantly reduced expression of MMP-9, MMP-10 and IL-6, but not MMP-12. Treatment of Alport mice with TAE226, a small molecule inhibitor of FAK activation, ameliorated fibrosis and glomerulosclerosis, significantly reduced proteinuria and blood urea nitrogen levels, and partially restored GBM ultrastructure. Glomerular expression of MMP-9, MMP-10 and MMP-12 mRNAs was significantly reduced in TAE226 treated animals. Collectively, this work identifies laminin α2-mediated FAK activation in podocytes as an important early event in Alport glomerular pathogenesis and suggests that FAK inhibitors, if safe formulations can be developed, might be employed as a novel therapeutic approach for treating Alport renal disease in its early stages.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Laminin/metabolism , Nephritis, Hereditary/enzymology , Nephritis, Hereditary/etiology , Animals , Biomechanical Phenomena/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Gene Knockdown Techniques , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/enzymology , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/ultrastructure , I-kappa B Proteins/metabolism , Interleukin-6/metabolism , Kinetics , Matrix Metalloproteinases/metabolism , Mice, Knockout , Morpholines/pharmacology , Morpholines/therapeutic use , NF-KappaB Inhibitor alpha , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/pathology , Podocytes/enzymology , Podocytes/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Tetraspanin 24/metabolism
14.
PLoS One ; 9(4): e94272, 2014.
Article in English | MEDLINE | ID: mdl-24705452

ABSTRACT

Usher syndrome type 1B is a combined deaf-blindness condition caused by mutations in the MYO7A gene. Loss of functional myosin VIIa in the retinal pigment epithelia (RPE) and/or photoreceptors leads to blindness. We evaluated the impact of subretinally delivered UshStat, a recombinant EIAV-based lentiviral vector expressing human MYO7A, on photoreceptor function in the shaker1 mouse model for Usher type 1B that lacks a functional Myo7A gene. Subretinal injections of EIAV-CMV-GFP, EIAV-RK-GFP (photoreceptor specific), EIAV-CMV-MYO7A (UshStat) or EIAV-CMV-Null (control) vectors were performed in shaker1 mice. GFP and myosin VIIa expression was evaluated histologically. Photoreceptor function in EIAV-CMV-MYO7A treated eyes was determined by evaluating α-transducin translocation in photoreceptors in response to low light intensity levels, and protection from light induced photoreceptor degeneration was measured. The safety and tolerability of subretinally delivered UshStat was evaluated in macaques. Expression of GFP and myosin VIIa was confirmed in the RPE and photoreceptors in shaker1 mice following subretinal delivery of the EIAV-CMV-GFP/MYO7A vectors. The EIAV-CMV-MYO7A vector protected the shaker1 mouse photoreceptors from acute and chronic intensity light damage, indicated by a significant reduction in photoreceptor cell loss, and restoration of the α-transducin translocation threshold in the photoreceptors. Safety studies in the macaques demonstrated that subretinal delivery of UshStat is safe and well-tolerated. Subretinal delivery of EIAV-CMV-MYO7A (UshStat) rescues photoreceptor phenotypes in the shaker1 mouse. In addition, subretinally delivered UshStat is safe and well-tolerated in macaque safety studies These data support the clinical development of UshStat to treat Usher type 1B syndrome.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Infectious Anemia Virus, Equine/genetics , Usher Syndromes/genetics , Usher Syndromes/therapy , Animals , Cell Line , Disease Models, Animal , Female , Gene Order , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Humans , Macaca , Male , Mice , Mice, Knockout , Myosin VIIa , Myosins/genetics , Phenotype , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Protein Transport , Retina/metabolism , Retina/pathology , Transducin/metabolism
15.
Exp Eye Res ; 118: 145-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24211856

ABSTRACT

Usher syndrome combines congenital hearing loss and retinitis pigmentosa (RP). Mutations in the whirlin gene (DFNB31/WHRN) cause a subtype of Usher syndrome (USH2D). Whirler mice have a defective whirlin gene. They have inner ear defects but usually do not develop retinal degeneration. Here we report that, in whirler mouse photoreceptors, the light-activated rod transducin translocation is delayed and its activation threshold is shifted to a higher level. Rhodopsin mis-localization is observed in rod inner segments. Continuous moderate light exposure can induce significant rod photoreceptor degeneration. Whirler mice reared under a 1500 lux light/dark cycle also develop severe photoreceptor degeneration. Previously, we have reported that shaker1 mice, a USH1B model, show moderate light-induced photoreceptor degeneration with delayed transducin translocation. Here, we further show that, in both whirler and shaker1 mice, short-term moderate light/dark changes can induce rod degeneration as severe as that induced by continuous light exposure. The results from shaker1 and whirler mice suggest that defective transducin translocation may be functionally related to light-induced degeneration, and these two symptoms may be caused by defects in Usher protein function in rods. Furthermore, these results indicate that both Usher syndrome mouse models possess a light-induced retinal phenotype and may share a closely related pathobiological mechanism.


Subject(s)
Adaptation, Ocular/physiology , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/physiology , Transducin/genetics , Translocation, Genetic , Animals , Blotting, Western , Cell Count , Immunohistochemistry , Male , Mice , Photoperiod , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Transducin/metabolism
16.
Int J Biochem Cell Biol ; 46: 80-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24239741

ABSTRACT

The 10 different genes associated with the deaf/blind disorder, Usher syndrome, encode a number of structurally and functionally distinct proteins, most expressed as multiple isoforms/protein variants. Functional characterization of these proteins suggests a role in stereocilia development in cochlear hair cells, likely owing to adhesive interactions in hair bundles. In mature hair cells, homodimers of the Usher cadherins, cadherin 23 and protocadherin 15, interact to form a structural fiber, the tip link, and the linkages that anchor the taller stereocilia's actin cytoskeleton core to the shorter adjacent stereocilia and the elusive mechanotransduction channels, explaining the deafness phenotype when these molecular interactions are perturbed. The conundrum is that photoreceptors lack a synonymous mechanotransduction apparatus, and so a common theory for Usher protein function in the two neurosensory cell types affected in Usher syndrome is lacking. Recent evidence linking photoreceptor cell dysfunction in the shaker 1 mouse model for Usher syndrome to light-induced protein translocation defects, combined with localization of an Usher protein interactome at the periciliary region of the photoreceptors suggests Usher proteins might regulate protein trafficking between the inner and outer segments of photoreceptors. A distinct Usher protein complex is trafficked to the ribbon synapses of hair cells, and synaptic defects have been reported in Usher mutants in both hair cells and photoreceptors. This review aims to clarify what is known about Usher protein function at the synaptic and apical poles of hair cells and photoreceptors and the prospects for identifying a unifying pathobiological mechanism to explain deaf/blindness in Usher syndrome.


Subject(s)
Hair Cells, Auditory/pathology , Photoreceptor Cells/pathology , Usher Syndromes/genetics , Usher Syndromes/pathology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Hair Cells, Auditory/metabolism , Humans , Photoreceptor Cells/metabolism , Usher Syndromes/metabolism
17.
Am J Pathol ; 183(4): 1269-1280, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23911822

ABSTRACT

Alport syndrome, hereditary glomerulonephritis with hearing loss, results from mutations in type IV collagen COL4A3, COL4A4, or COL4A5 genes. The mechanism for delayed glomerular disease onset is unknown. Comparative analysis of Alport mice and CD151 knockout mice revealed progressive accumulation of laminin 211 in the glomerular basement membrane. We show mesangial processes invading the capillary loops of both models as well as in human Alport glomeruli, as the likely source of this laminin. L-NAME salt-induced hypertension accelerated mesangial cell process invasion. Cultured mesangial cells showed reduced migratory potential when treated with either integrin-linked kinase inhibitor or Rac1 inhibitor, or by deletion of integrin α1. Treatment of Alport mice with Rac1 inhibitor or deletion of integrin α1 reduced mesangial cell process invasion of the glomerular capillary tuft. Laminin α2-deficient Alport mice show reduced mesangial process invasion, and cultured laminin α2-null cells showed reduced migratory potential, indicating a functional role for mesangial laminins in progression of Alport glomerular pathogenesis. Collectively, these findings predict a role for biomechanical insult in the induction of integrin α1ß1-dependent Rac1-mediated mesangial cell process invasion of the glomerular capillary tuft as an initiation mechanism of Alport glomerular pathology.


Subject(s)
Capillaries/pathology , Glomerular Mesangium/blood supply , Glomerular Mesangium/pathology , Integrin alpha1beta1/metabolism , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , rac1 GTP-Binding Protein/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Biomechanical Phenomena/drug effects , Capillaries/drug effects , Capillaries/metabolism , Capillaries/physiopathology , Cell Movement/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Deletion , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/physiopathology , Glomerular Basement Membrane/ultrastructure , Glomerular Mesangium/physiopathology , Glomerular Mesangium/ultrastructure , Humans , Hypertension/complications , Hypertension/metabolism , Hypertension/pathology , Hypertension/physiopathology , Laminin/metabolism , Mice , Mice, Knockout , Nephritis, Hereditary/complications , Nephritis, Hereditary/physiopathology , Protein Transport/drug effects , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
18.
PLoS One ; 8(3): e58832, 2013.
Article in English | MEDLINE | ID: mdl-23555598

ABSTRACT

The transducin GTPase-accelerating protein complex, which determines the photoresponse duration of photoreceptors, is composed of RGS9-1, Gß5L and R9AP. Here we report that RGS9-1 and Gß5L change their distribution in rods during light/dark adaptation. Upon prolonged dark adaptation, RGS9-1 and Gß5L are primarily located in rod inner segments. But very dim-light exposure quickly translocates them to the outer segments. In contrast, their anchor protein R9AP remains in the outer segment at all times. In the dark, Gß5L's interaction with R9AP decreases significantly and RGS9-1 is phosphorylated at S(475) to a significant degree. Dim light exposure leads to quick de-phosphorylation of RGS9-1. Furthermore, after prolonged dark adaptation, RGS9-1 and transducin Gα are located in different cellular compartments. These results suggest a previously unappreciated mechanism by which prolonged dark adaptation leads to increased light sensitivity in rods by dissociating RGS9-1 from R9AP and redistributing it to rod inner segments.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , Light , Membrane Proteins/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Dark Adaptation/physiology , Female , Male , Membrane Proteins/chemistry , Mice , Phosphorylation , Protein Binding , Protein Transport/radiation effects , Retinal Photoreceptor Cell Inner Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Rod Photoreceptor Cells/radiation effects , Serine/metabolism
19.
J Neurosci ; 32(40): 13841-59, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035094

ABSTRACT

Usher syndrome is a genetically heterogeneous disorder characterized by hearing and balance dysfunction and progressive retinitis pigmentosa. Mouse models carrying mutations for the nine Usher-associated genes have splayed stereocilia, and some show delayed maturation of ribbon synapses suggesting these proteins may play different roles in terminal differentiation of auditory hair cells. The presence of the Usher proteins at the basal and apical aspects of the neurosensory epithelia suggests the existence of regulated trafficking through specific transport proteins and routes. Immature mouse cochleae and UB/OC-1 cells were used in this work to address whether specific variants of PCDH15 and VLGR1 are being selectively transported to opposite poles of the hair cells. Confocal colocalization studies between apical and basal vesicular markers and the different PCDH15 and VLGR1 variants along with sucrose density gradients and the use of vesicle trafficking inhibitors show the existence of Usher protein complexes in at least two vesicular subpools. The apically trafficked pool colocalized with the early endosomal vesicle marker, rab5, while the basally trafficked pool associated with membrane microdomains and SNAP25. Moreover, coimmunoprecipitation experiments between SNAP25 and VLGR1 show a physical interaction of these two proteins in organ of Corti and brain. Collectively, these findings establish the existence of a differential vesicular trafficking mechanism for specific Usher protein variants in mouse cochlear hair cells, with the apical variants playing a potential role in endosomal recycling and stereocilia development/maintenance, and the basolateral variants involved in vesicle docking and/or fusion through SNAP25-mediated interactions.


Subject(s)
Cadherins/metabolism , Cell Polarity/physiology , Hair Cells, Auditory/ultrastructure , Protein Precursors/metabolism , Receptors, G-Protein-Coupled/metabolism , Transport Vesicles/physiology , ADP-Ribosylation Factor 1/analysis , Animals , Brain Chemistry , Cadherin Related Proteins , Cadherins/biosynthesis , Cadherins/genetics , Cell Compartmentation , Cell Differentiation , Disease Models, Animal , Gene Knockdown Techniques , Hair Cells, Auditory/metabolism , Immunoprecipitation , Mice , Mice, Neurologic Mutants , Mutation , Organ of Corti/chemistry , Organ of Corti/ultrastructure , Protein Interaction Mapping , Protein Precursors/biosynthesis , Protein Precursors/genetics , Protein Transport/drug effects , RNA Interference , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Structure-Activity Relationship , Synaptosomal-Associated Protein 25/analysis , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/metabolism , Transport Vesicles/chemistry , Usher Syndromes/metabolism , rab5 GTP-Binding Proteins/analysis
20.
PLoS One ; 7(2): e30573, 2012.
Article in English | MEDLINE | ID: mdl-22363448

ABSTRACT

The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1-/- mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzer(av3J) mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.


Subject(s)
Cadherins/metabolism , Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Protein Precursors/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/metabolism , Adsorption , Animals , Antibodies/immunology , Cadherin Related Proteins , Cell Line , Cell Polarity , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Multiprotein Complexes/metabolism , Protein Isoforms/metabolism , Synapses/ultrastructure , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL