Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 553(7687): 217-221, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29258297

ABSTRACT

Although genetic factors contribute to almost half of all cases of deafness, treatment options for genetic deafness are limited. We developed a genome-editing approach to target a dominantly inherited form of genetic deafness. Here we show that cationic lipid-mediated in vivo delivery of Cas9-guide RNA complexes can ameliorate hearing loss in a mouse model of human genetic deafness. We designed and validated, both in vitro and in primary fibroblasts, genome editing agents that preferentially disrupt the dominant deafness-associated allele in the Tmc1 (transmembrane channel-like gene family 1) Beethoven (Bth) mouse model, even though the mutant Tmc1Bth allele differs from the wild-type allele at only a single base pair. Injection of Cas9-guide RNA-lipid complexes targeting the Tmc1Bth allele into the cochlea of neonatal Tmc1Bth/+ mice substantially reduced progressive hearing loss. We observed higher hair cell survival rates and lower auditory brainstem response thresholds in injected ears than in uninjected ears or ears injected with control complexes that targeted an unrelated gene. Enhanced acoustic startle responses were observed among injected compared to uninjected Tmc1Bth/+ mice. These findings suggest that protein-RNA complex delivery of target gene-disrupting agents in vivo is a potential strategy for the treatment of some types of autosomal-dominant hearing loss.


Subject(s)
CRISPR-Associated Proteins/administration & dosage , Gene Editing/methods , Genes, Dominant/genetics , Genetic Therapy/methods , Hearing Loss/genetics , Acoustic Stimulation , Alleles , Animals , Animals, Newborn , Auditory Threshold , Base Sequence , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/therapeutic use , CRISPR-Cas Systems , Cell Survival , Cochlea/cytology , Cochlea/metabolism , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Fibroblasts , Hair Cells, Auditory/cytology , Hearing Loss/physiopathology , Hearing Loss/prevention & control , Humans , Liposomes , Male , Membrane Proteins/genetics , Mice , Reflex, Startle
2.
Mol Ther ; 28(12): 2662-2676, 2020 12 02.
Article in English | MEDLINE | ID: mdl-32818431

ABSTRACT

Usher syndrome is a syndromic form of hereditary hearing impairment that includes sensorineural hearing loss and delayed-onset retinitis pigmentosa (RP). Type 1 Usher syndrome (USH1) is characterized by congenital profound sensorineural hearing impairment and vestibular areflexia, with adolescent-onset RP. Systemic treatment with antisense oligonucleotides (ASOs) targeting the human USH1C c.216G>A splicing mutation in a knockin mouse model of USH1 restores hearing and balance. Herein, we explore the effect of delivering ASOs locally to the ear to treat hearing and vestibular dysfunction associated with Usher syndrome. Three localized delivery strategies were investigated in USH1C mice: inner ear injection, trans-tympanic membrane injection, and topical tympanic membrane application. We demonstrate, for the first time, that ASOs delivered directly to the ear correct Ush1c expression in inner ear tissue, improve cochlear hair cell transduction currents, restore vestibular afferent irregularity, spontaneous firing rate, and sensitivity to head rotation, and successfully recover hearing thresholds and balance behaviors in USH1C mice. We conclude that local delivery of ASOs to the middle and inner ear reach hair cells and can rescue both hearing and balance. These results also demonstrate the therapeutic potential of ASOs to treat hearing and balance deficits associated with Usher syndrome and other ear diseases.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Ear, Middle/drug effects , Genetic Therapy/methods , Hair Cells, Auditory/drug effects , Mutation , Oligonucleotides, Antisense/administration & dosage , Usher Syndromes/genetics , Usher Syndromes/therapy , Vestibule, Labyrinth/drug effects , Administration, Topical , Animals , Animals, Newborn , Disease Models, Animal , Female , Gene Knock-In Techniques , Hair Cells, Auditory/metabolism , Hearing/drug effects , Injections , Male , Mice , Mice, Inbred C57BL , Tympanic Membrane/drug effects , Vestibule, Labyrinth/metabolism
3.
Am J Hum Genet ; 98(6): 1101-1113, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27236922

ABSTRACT

Hearing impairment is the most common sensory deficit. It is frequently caused by the expression of an allele carrying a single dominant missense mutation. Herein, we show that a single intracochlear injection of an artificial microRNA carried in a viral vector can slow progression of hearing loss for up to 35 weeks in the Beethoven mouse, a murine model of non-syndromic human deafness caused by a dominant gain-of-function mutation in Tmc1 (transmembrane channel-like 1). This outcome is noteworthy because it demonstrates the feasibility of RNA-interference-mediated suppression of an endogenous deafness-causing allele to slow progression of hearing loss. Given that most autosomal-dominant non-syndromic hearing loss in humans is caused by this mechanism of action, microRNA-based therapeutics might be broadly applicable as a therapy for this type of deafness.


Subject(s)
Auditory Pathways , Hearing Loss/prevention & control , Membrane Proteins/physiology , MicroRNAs/genetics , Mutation, Missense/genetics , Animals , Dependovirus/genetics , Hearing Loss/etiology , Hearing Loss/pathology , Humans , Mechanotransduction, Cellular , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C3H , Mice, Knockout , MicroRNAs/administration & dosage , RNA Interference
4.
Hum Mol Genet ; 23(9): 2374-90, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24334608

ABSTRACT

Usher syndrome type 2 (USH2) is the predominant form of USH, a leading genetic cause of combined deafness and blindness. PDZD7, a paralog of two USH causative genes, USH1C and USH2D (WHRN), was recently reported to be implicated in USH2 and non-syndromic deafness. It encodes a protein with multiple PDZ domains. To understand the biological function of PDZD7 and the pathogenic mechanism caused by PDZD7 mutations, we generated and thoroughly characterized a Pdzd7 knockout mouse model. The Pdzd7 knockout mice exhibit congenital profound deafness, as assessed by auditory brainstem response, distortion product otoacoustic emission and cochlear microphonics tests, and normal vestibular function, as assessed by their behaviors. Lack of PDZD7 leads to the disorganization of stereocilia bundles and a reduction in mechanotransduction currents and sensitivity in cochlear outer hair cells. At the molecular level, PDZD7 determines the localization of the USH2 protein complex, composed of USH2A, GPR98 and WHRN, to ankle links in developing cochlear hair cells, likely through its direct interactions with these three proteins. The localization of PDZD7 to the ankle links of cochlear hair bundles also relies on USH2 proteins. In photoreceptors of Pdzd7 knockout mice, the three USH2 proteins largely remain unchanged at the periciliary membrane complex. The electroretinogram responses of both rod and cone photoreceptors are normal in knockout mice at 1 month of age. Therefore, although the organization of the USH2 complex appears different in photoreceptors, it is clear that PDZD7 plays an essential role in organizing the USH2 complex at ankle links in developing cochlear hair cells. GenBank accession numbers: KF041446, KF041447, KF041448, KF041449, KF041450, KF041451.


Subject(s)
Gene Deletion , Hair Cells, Auditory/metabolism , Hearing Loss/genetics , Hearing Loss/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Usher Syndromes/metabolism , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Usher Syndromes/genetics
5.
Pflugers Arch ; 467(1): 85-94, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25074487

ABSTRACT

Mutations of the transmembrane channel-like 1 (TMC1) gene can cause dominant and recessive forms of deafness in humans and mice. TMC1 is one of eight mammalian TMC genes of unknown function. The multi-pass transmembrane topologic structure of the proteins they encode suggests roles as a receptor, transporter, channel, or pump. Tmc1 and the closely related Tmc2 gene are expressed in neurosensory hair cells of the auditory and vestibular end organs of the mouse inner ear. Recent studies have demonstrated that Tmc1 and Tmc2 are specifically required for mechanoelectrical transduction in hair cells. The exact role of these proteins in mechanoelectrical transduction is unknown. TMC1 and TMC2 are viable candidates for the mechanoelectrical transduction channel of hair cells, whose component molecules have eluded identification for over 30 years. We expect that studies of TMC proteins will yield insights into molecular components and mechanisms of mechanosensation in auditory and vestibular hair cells, as well as in other tissues and organs.


Subject(s)
Hair Cells, Auditory/physiology , Hair Cells, Vestibular/physiology , Hearing/physiology , Ion Channels/physiology , Mechanotransduction, Cellular/physiology , Postural Balance/physiology , Animals , Humans , Ion Channel Gating/physiology
6.
Nat Commun ; 14(1): 2400, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37100771

ABSTRACT

Usher syndrome type 1 F (USH1F), caused by mutations in the protocadherin-15 gene (PCDH15), is characterized by congenital deafness, lack of balance, and progressive blindness. In hair cells, the receptor cells of the inner ear, PCDH15 is a component of tip links, fine filaments which pull open mechanosensory transduction channels. A simple gene addition therapy for USH1F is challenging because the PCDH15 coding sequence is too large for adeno-associated virus (AAV) vectors. We use rational, structure-based design to engineer mini-PCDH15s in which 3-5 of the 11 extracellular cadherin repeats are deleted, but which still bind a partner protein. Some mini-PCDH15s can fit in an AAV. An AAV encoding one of these, injected into the inner ears of mouse models of USH1F, produces a mini-PCDH15 which properly forms tip links, prevents the degeneration of hair cell bundles, and rescues hearing. Mini-PCDH15s may be a useful therapy for the deafness of USH1F.


Subject(s)
Ear, Inner , Usher Syndromes , Animals , Mice , Cadherins/metabolism , Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Hearing/genetics , Usher Syndromes/genetics , Usher Syndromes/therapy , Cadherin Related Proteins/metabolism
7.
Sci Total Environ ; 834: 155166, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35413348

ABSTRACT

The Anoxic/Oxic (A/O) process involves recirculating mixed liquor between its A and O tanks so that nitrate produced in the O tank can be used to for denitrification with influent COD in the A tank. Because biomass is recirculated along with nitrate, A/O operation leads to similar microbial communities in the A and O tanks, which may decrease the rates of denitrification and nitrification in each tank. Here, bench-scale experiments simulated this aspect of the A/O process by exchanging biomass between an anoxic flask and an oxic cylinder at exchange ratios of 0%, 20%, 30%, and 50%. Nitrification and denitrification rates were only 40% and 19% for 50% biomass exchange of that for no biomass exchange. Phylogenetic analysis documented that the microbial communities became much more similar with biomass exchange, and the finding was consistent with community composition in a full-scale A/O process in a municipal wastewater treatment plant. A two-stage vertical baffled bioreactor (VBBR) realized efficient total­nitrogen removal in recirculation without biomass exchange. Average removals of COD and TN were respectively 6% and 22% higher for the two-stage VBBR than the conventional A/O process, but its hydraulic retention time (HRT) was 55% to 70% of the volume of a conventional A/O process treating the same influent wastewater. The VBBR was more efficient because its anoxic biofilm was enriched in denitrifying bacteria, while its oxic biofilm was enriched in nitrifying bacteria. For example, the phylum Chloroflexi was greater in the An-VBBR, while the phylum Proteobacteria was greater in the Ox-VBBR.


Subject(s)
Denitrification , Nitrates , Biomass , Bioreactors , Nitrification , Nitrogen/analysis , Phylogeny , Sewage , Wastewater
8.
Sci Adv ; 8(28): eabo1126, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35857511

ABSTRACT

The transmembrane (TM) channel-like 1 (TMC1) and TMC2 proteins play a central role in auditory transduction, forming ion channels that convert sound into electrical signals. However, the molecular mechanism of their gating remains unknown. Here, using predicted structural models as a guide, we probed the effects of 12 mutations on the mechanical gating of the transduction currents in native hair cells of Tmc1/2-null mice expressing virally introduced TMC1 variants. Whole-cell electrophysiological recordings revealed that mutations within the pore-lining TM4 and TM6 helices modified gating, reducing the force sensitivity or shifting the open probability of the channels, or both. For some of the mutants, these changes were accompanied by a change in single-channel conductance. Our observations are in line with a model wherein conformational changes in the TM4 and TM6 helices are involved in the mechanical gating of the transduction channel.

9.
Hear Res ; 394: 107882, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31980281

ABSTRACT

Viral delivery of exogenous coding sequences into the inner ear has the potential for therapeutic benefit for patients suffering genetic or acquired hearing loss. To devise improved strategies for viral delivery, we investigated two injection techniques, round window membrane injection or a novel utricle injection method, for their ability to safely and efficiently transduce sensory hair cells and neurons of the mouse inner ear. In addition, we evaluated three synthetic AAV vectors (Anc80L65, AAV9-PHP.B, AAV2.7m8) encoding enhanced green fluorescent protein (eGFP) and three promoters (Cmv, Synapsin, Gfap) for their ability to transduce and drive expression in desired cell types. We found the utricle injection method with AAV9-PHP.B and a Cmv promoter was the most efficient combination for driving robust eGFP expression in both inner and outer hair cells. We found eGFP expression levels rose over 3-5 days post-injection, a viral dose of 1.5 × 109 gc yielded half maximal eGFP expression and that the utricle injection method yielded transduced hair cells even when delivered as late as postnatal day 16. Sensory transduction and auditory thresholds were unaltered in injected mice relative to uninjected wild-type controls. Vestibular end organs were also transduced without affecting balance behavior. The Synapsin promoter and the Gfap promoter drove strong eGFP expression in inner ear neurons and supporting cells, respectively. We conclude the AAV9-PHP.B vector and the utricle injection method are well-suited for delivery of exogenous gene constructs into inner ears of mouse models of auditory and vestibular dysfunction.


Subject(s)
Ear, Inner , Animals , Cytomegalovirus Infections , Dependovirus/genetics , Genetic Therapy , Genetic Vectors , Hair Cells, Auditory, Outer , Mice , Saccule and Utricle , Synapsins/genetics
10.
Sci Transl Med ; 12(546)2020 06 03.
Article in English | MEDLINE | ID: mdl-32493795

ABSTRACT

Most genetic diseases arise from recessive point mutations that require correction, rather than disruption, of the pathogenic allele to benefit patients. Base editing has the potential to directly repair point mutations and provide therapeutic restoration of gene function. Mutations of transmembrane channel-like 1 gene (TMC1) can cause dominant or recessive deafness. We developed a base editing strategy to treat Baringo mice, which carry a recessive, loss-of-function point mutation (c.A545G; resulting in the substitution p.Y182C) in Tmc1 that causes deafness. Tmc1 encodes a protein that forms mechanosensitive ion channels in sensory hair cells of the inner ear and is required for normal auditory function. We found that sensory hair cells of Baringo mice have a complete loss of auditory sensory transduction. To repair the mutation, we tested several optimized cytosine base editors (CBEmax variants) and guide RNAs in Baringo mouse embryonic fibroblasts. We packaged the most promising CBE, derived from an activation-induced cytidine deaminase (AID), into dual adeno-associated viruses (AAVs) using a split-intein delivery system. The dual AID-CBEmax AAVs were injected into the inner ears of Baringo mice at postnatal day 1. Injected mice showed up to 51% reversion of the Tmc1 c.A545G point mutation to wild-type sequence (c.A545A) in Tmc1 transcripts. Repair of Tmc1 in vivo restored inner hair cell sensory transduction and hair cell morphology and transiently rescued low-frequency hearing 4 weeks after injection. These findings provide a foundation for a potential one-time treatment for recessive hearing loss and support further development of base editing to correct pathogenic point mutations.


Subject(s)
Deafness , Membrane Proteins , Animals , Deafness/genetics , Deafness/therapy , Fibroblasts , Hair Cells, Auditory , Hearing/genetics , Humans , Mice
11.
Nanotechnology ; 20(12): 125101, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19420458

ABSTRACT

With the aim of improving the amount and delivery efficiency of genes taken by carbon nanotubes into human cancer cells, different generations of polyamidoamine dendrimer modified multi-walled carbon nanotubes (dMNTs) were fabricated, and characterized by high-resolution transmission electron microscopy, atomic force microscopy, x-ray photoelectron spectroscopy, Raman spectroscopy, Fourier transform infrared spectroscopy and thermogravimetric analysis, revealing the presence of dendrimer capped on the surface of carbon nanotubes. The dMNTs fully conjugated with FITC-labeled antisense c-myc oligonucleotides (asODN), those resultant asODN-dMNTs composites were incubated with human breast cancer cell line MCF-7 cells and MDA-MB-435 cells, and liver cancer cell line HepG2 cells, and confirmed to enter into tumor cells within 15 min by laser confocal microscopy. These composites inhibited the cell growth in time- and dose-dependent means, and down-regulated the expression of the c-myc gene and C-Myc protein. Compared with the composites of CNT-NH(2)-asODN and dendrimer-asODN, no. 5 generation of dendrimer-modified MNT-asODN composites exhibit maximal transfection efficiencies and inhibition effects on tumor cells. The intracellular gene transport and uptake via dMNTs should be generic for the mammalian cell lines. The dMNTs have potentials in applications such as gene or drug delivery for cancer therapy and molecular imaging.


Subject(s)
Gene Transfer Techniques , Nanotubes, Carbon/chemistry , Polyamines/chemistry , Analysis of Variance , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Dendrimers , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanotubes, Carbon/ultrastructure , Neoplasms , Polymerase Chain Reaction , Spectrum Analysis
12.
Cancer Res ; 67(17): 8156-63, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804728

ABSTRACT

Magnetic nanoparticles (MNP) with a diameter of 8 nm were modified with different generations of polyamidoamine (PAMAM) dendrimers and mixed with antisense survivin oligodeoxynucleotide (asODN). The MNP then formed asODN-dendrimer-MNP composites, which we incubated with human tumor cell lines such as human breast cancer MCF-7, MDA-MB-435, and liver cancer HepG2 and then analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, quantitative reverse transcription-PCR, Western blotting, laser confocal microscopy, and high-resolution transmission electron microscopy. Results showed that the asODN-dendrimer-MNP composites were successfully synthesized, can enter into tumor cells within 15 min, caused marked down-regulation of the survivin gene and protein, and inhibited cell growth in dose- and time-dependent means. No.5 generation of asODN-dendrimer-MNP composites exhibits the highest efficiency for cellular transfection and inhibition. These results show that PAMAM dendrimer-modified MNPs may be a good gene delivery system and have potential applications in cancer therapy and molecular imaging diagnosis.


Subject(s)
Dendrimers/pharmacology , Dendrimers/therapeutic use , Gene Transfer Techniques , Genetic Therapy/methods , Metal Nanoparticles/therapeutic use , Neoplasms/therapy , Cell Proliferation , Cell Survival , Dendrimers/chemistry , HL-60 Cells , Humans , Inhibitor of Apoptosis Proteins , Magnetics , Microtubule-Associated Proteins/genetics , Models, Biological , Neoplasm Proteins/genetics , Oligodeoxyribonucleotides, Antisense/administration & dosage , Survivin , Treatment Outcome , Tumor Cells, Cultured
13.
Nat Commun ; 10(1): 734, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737404

ABSTRACT

The original version of this Article contained errors in Fig. 5. In panels i and j the three rightmost x-axis labels inadvertently read 'Tmc1' instead of 'Tmc2'. These errors have been corrected in both the PDF and HTML versions of the Article.

14.
Nat Commun ; 10(1): 236, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670701

ABSTRACT

Fifty percent of inner ear disorders are caused by genetic mutations. To develop treatments for genetic inner ear disorders, we designed gene replacement therapies using synthetic adeno-associated viral vectors to deliver the coding sequence for Transmembrane Channel-Like (Tmc) 1 or 2 into sensory hair cells of mice with hearing and balance deficits due to mutations in Tmc1 and closely related Tmc2. Here we report restoration of function in inner and outer hair cells, enhanced hair cell survival, restoration of cochlear and vestibular function, restoration of neural responses in auditory cortex and recovery of behavioral responses to auditory and vestibular stimulation. Secondarily, we find that inner ear Tmc gene therapy restores breeding efficiency, litter survival and normal growth rates in mouse models of genetic inner ear dysfunction. Although challenges remain, the data suggest that Tmc gene therapy may be well suited for further development and perhaps translation to clinical application.


Subject(s)
Deafness/genetics , Genetic Predisposition to Disease , Genetic Therapy/methods , Hearing Loss/genetics , Labyrinth Diseases/genetics , Membrane Proteins/genetics , Animals , Deafness/therapy , Hair Cells, Auditory/physiology , Hair Cells, Vestibular/physiology , Hearing Loss/therapy , Labyrinth Diseases/therapy , Mice , Mice, Mutant Strains
15.
Nat Med ; 25(7): 1123-1130, 2019 07.
Article in English | MEDLINE | ID: mdl-31270503

ABSTRACT

Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.


Subject(s)
Alleles , Gene Editing , Hearing Loss, Sensorineural/prevention & control , Membrane Proteins/genetics , Animals , CRISPR-Associated Protein 9/physiology , Cell Line , Cells, Cultured , Dependovirus/genetics , Disease Models, Animal , Hearing Loss, Sensorineural/genetics , Humans , Mice , Mice, Inbred C57BL
16.
Anal Chem ; 80(21): 7996-8001, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18816142

ABSTRACT

A highly selective, ultrasensitive, fluorescence detection method for DNA and antigen based on self-assembly of multiwalled carbon nanotubes (CNTs) and CdSe quantum dots (QDs) via oligonucleotide hybridization is reported. Mercaptoalkyloligonucleotide molecules bind to the quantum dots, while amineoalkyloligonucleotides bind to CNTs with -COCl surface groups. QDs and CNTs further assemble into nanohybrids through DNA hybridization in the presence of target complementary oligonucleotides. The method is achieved with good repeatability with the detection limit of 0.2 pM DNA molecules and 0.01 nM antigen molecules. This novel detection system can also be used for multicomponent detection and antigen-antibody immunoreaction. The novel system has great potential in applications such as ultrasensitive pathogen DNA or antigen or antibody detection, molecular imaging, and photoelectrical biosensors.


Subject(s)
Antigens/analysis , Antigens/chemistry , DNA/analysis , DNA/chemistry , Immunoassay/methods , Nanotubes, Carbon/chemistry , Quantum Dots , Antibodies/immunology , Antigens/immunology , Base Sequence , Substrate Specificity
17.
Front Cell Neurosci ; 12: 41, 2018.
Article in English | MEDLINE | ID: mdl-29515374

ABSTRACT

Hair cells of the inner ear transduce mechanical stimuli like sound or head movements into electrical signals, which are propagated to the central nervous system. The hair-cell mechanotransduction channel remains unidentified. We tested whether three transient receptor channel (TRP) family members, TRPV6, TRPM6 and TRPM7, were necessary for transduction. TRPV6 interacted with USH1C (harmonin), a scaffolding protein that participates in transduction. Using a cysteine-substitution knock-in mouse line and methanethiosulfonate (MTS) reagents selective for this allele, we found that inhibition of TRPV6 had no effect on transduction in mouse cochlear hair cells. TRPM6 and TRPM7 each interacted with the tip-link component PCDH15 in cultured eukaryotic cells, which suggested they might be part of the transduction complex. Cochlear hair cell transduction was not affected by manipulations of Mg2+, however, which normally perturbs TRPM6 and TRPM7. To definitively examine the role of these two channels in transduction, we showed that deletion of either or both of their genes selectively in hair cells had no effect on auditory function. We suggest that TRPV6, TRPM6 and TRPM7 are unlikely to be the pore-forming subunit of the hair-cell transduction channel.

18.
Sci Rep ; 8(1): 12125, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108230

ABSTRACT

Mouse Tmc1 and Tmc2 are required for sensory transduction in cochlear and vestibular hair cells. Homozygous Tmc1∆/∆ mice are deaf, Tmc2∆/∆ mice have normal hearing, and double homozygous Tmc1∆/∆; Tmc2∆/∆ mice have deafness and profound vestibular dysfunction. These phenotypes are consistent with their different spatiotemporal expression patterns. Tmc1 expression is persistent in cochlear and vestibular hair cells, whereas Tmc2 expression is transient in cochlear hair cells but persistent in vestibular hair cells. On the basis of these findings, we hypothesized that persistent Tmc2 expression in mature cochlear hair cells could restore auditory function in Tmc1∆/∆ mice. To express Tmc2 in mature cochlear hair cells, we generated a transgenic mouse line, Tg[PTmc1::Tmc2], in which Tmc2 cDNA is expressed under the control of the Tmc1 promoter. The Tg[PTmc1::Tmc2] transgene slightly but significantly restored hearing in young Tmc1∆/∆ mice, though hearing thresholds were elevated with age. The elevation of hearing thresholds was associated with deterioration of sensory transduction in inner hair cells and loss of outer hair cell function. Although sensory transduction was retained in outer hair cells, their stereocilia eventually degenerated. These results indicate distinct roles and requirements for Tmc1 and Tmc2 in mature cochlear hair cells.


Subject(s)
Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/pathology , Membrane Proteins/metabolism , Stereocilia/pathology , Animals , Disease Models, Animal , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Hair Cells, Vestibular/metabolism , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Hearing Tests , Homozygote , Humans , Mechanotransduction, Cellular , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Mutation , Patch-Clamp Techniques , Promoter Regions, Genetic/genetics , Stereocilia/ultrastructure
19.
Sci Rep ; 8(1): 12124, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108254

ABSTRACT

Recent work has demonstrated that transmembrane channel-like 1 protein (TMC1) is an essential component of the sensory transduction complex in hair cells of the inner ear. A closely related homolog, TMC2, is expressed transiently in the neonatal mouse cochlea and can enable sensory transduction in Tmc1-null mice during the first postnatal week. Both TMC1 and TMC2 are expressed at adult stages in mouse vestibular hair cells. The extent to which TMC1 and TMC2 can substitute for each other is unknown. Several biophysical differences between TMC1 and TMC2 suggest these proteins perform similar but not identical functions. To investigate these differences, and whether TMC2 can substitute for TMC1 in mature hair cells, we generated a knock-in mouse model allowing Cre-inducible expression of Tmc2. We assayed for changes in hair cell sensory transduction and auditory and vestibular function in Tmc2 knockin mice (Tm[Tmc2]) in the presence or absence of endogenous Tmc1, Tmc2 or both. Our results show that expression of Tm[TMC2] restores sensory transduction in vestibular hair cells and transiently in cochlear hair cells in the absence of TMC1. The cellular rescue leads to recovery of balance but not auditory function. We conclude that TMC1 provides some additional necessary function, not provided by TMC2.


Subject(s)
Hair Cells, Auditory, Inner/metabolism , Hair Cells, Vestibular/metabolism , Membrane Proteins/metabolism , Postural Balance/physiology , Animals , Gene Knock-In Techniques , Hearing/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Animal , Transgenes/genetics
20.
Neuron ; 99(4): 736-753.e6, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30138589

ABSTRACT

The proteins that form the permeation pathway of mechanosensory transduction channels in inner-ear hair cells have not been definitively identified. Genetic, anatomical, and physiological evidence support a role for transmembrane channel-like protein (TMC) 1 in hair cell sensory transduction, yet the molecular function of TMC proteins remains unclear. Here, we provide biochemical evidence suggesting TMC1 assembles as a dimer, along with structural and sequence analyses suggesting similarity to dimeric TMEM16 channels. To identify the pore region of TMC1, we used cysteine mutagenesis and expressed mutant TMC1 in hair cells of Tmc1/2-null mice. Cysteine-modification reagents rapidly and irreversibly altered permeation properties of mechanosensory transduction. We propose that TMC1 is structurally similar to TMEM16 channels and includes ten transmembrane domains with four domains, S4-S7, that line the channel pore. The data provide compelling evidence that TMC1 is a pore-forming component of sensory transduction channels in auditory and vestibular hair cells.


Subject(s)
Hair Cells, Auditory, Inner/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/chemistry , Membrane Proteins/physiology , Porins/chemistry , Porins/physiology , Animals , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL