Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Biol Chem ; 299(10): 105243, 2023 10.
Article in English | MEDLINE | ID: mdl-37690683

ABSTRACT

Myosin-7a is an actin-based motor protein essential for vision and hearing. Mutations of myosin-7a cause type 1 Usher syndrome, the most common and severe form of deafblindness in humans. The molecular mechanisms that govern its mechanochemistry remain poorly understood, primarily because of the difficulty of purifying stable intact protein. Here, we recombinantly produce the complete human myosin-7a holoenzyme in insect cells and characterize its biochemical and motile properties. Unlike the Drosophila ortholog that primarily associates with calmodulin (CaM), we found that human myosin-7a utilizes a unique combination of light chains including regulatory light chain, CaM, and CaM-like protein 4. Our results further reveal that CaM-like protein 4 does not function as a Ca2+ sensor but plays a crucial role in maintaining the lever arm's structural-functional integrity. Using our recombinant protein system, we purified two myosin-7a splicing isoforms that have been shown to be differentially expressed along the cochlear tonotopic axis. We show that they possess distinct mechanoenzymatic properties despite differing by only 11 amino acids at their N termini. Using single-molecule in vitro motility assays, we demonstrate that human myosin-7a exists as an autoinhibited monomer and can move processively along actin when artificially dimerized or bound to cargo adaptor proteins. These results suggest that myosin-7a can serve multiple roles in sensory systems such as acting as a transporter or an anchor/force sensor. Furthermore, our research highlights that human myosin-7a has evolved unique regulatory elements that enable precise tuning of its mechanical properties suitable for mammalian auditory functions.


Subject(s)
Actins , Deaf-Blind Disorders , Myosin VIIa , Humans , Actins/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Myosin VIIa/genetics , Myosin VIIa/metabolism , Calmodulin/metabolism , Calcium-Binding Proteins/metabolism
2.
ESC Heart Fail ; 8(3): 2310-2315, 2021 06.
Article in English | MEDLINE | ID: mdl-33835720

ABSTRACT

We report a novel cardiomyopathy associated to Usher syndrome and related to combined mutation of MYO7A and Calreticulin genes. A 37-year-old man with deafness and vision impairment because of retinitis pigmentosa since childhood and a MYO7A gene mutation suggesting Usher syndrome, developed a dilated cardiomyopathy with ventricular tachyarrhythmias and recurrent syncope. At magnetic resonance cardiomyopathy was characterized by left ventricular dilatation with hypo-contractility and mitral prolapse with valve regurgitation. At left ventricular endomyocardial biopsy, it was documented cardiomyocyte disconnection because of cytoskeletal disorganization of cell-to-cell contacts, including intercalated discs, and mitochondrial damage and dysfunction with significant reduction of adenosine triphosphate production in patient cultured fibroblasts. At an extensive analysis by next-generation-sequencing of 4183 genes potentially related to the cardiomyopathy a pathogenic mutation of calreticulin was found. The cardiomyopathy appeared to be functionally and electrically stabilized by a combination therapy including carvedilol and amiodarone at a follow-up of 18 months.


Subject(s)
Cardiomyopathy, Dilated , Usher Syndromes , Adult , Calreticulin/genetics , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Child , DNA Mutational Analysis , Humans , Male , Mutation , Myosin VIIa , Myosins/genetics , Pedigree , Usher Syndromes/diagnosis , Usher Syndromes/genetics
3.
Cell Rep ; 35(3): 109016, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882317

ABSTRACT

The mammalian cochlea cannot regenerate functional hair cells (HCs) spontaneously. Atoh1 overexpression as well as other strategies are unable to generate functional HCs. Here, we simultaneously upregulated the expression of Gfi1, Pou4f3, and Atoh1 in postnatal cochlear supporting cells (SCs) in vivo, which efficiently converted SCs into HCs. The newly regenerated HCs expressed HC markers Myo7a, Calbindin, Parvalbumin, and Ctbp2 and were innervated by neurites. Importantly, many new HCs expressed the mature and terminal marker Prestin or vesicular glutamate transporter 3 (vGlut3), depending on the subtypes of the source SCs. Finally, our patch-clamp analysis showed that the new HCs in the medial region acquired a large K+ current, fired spikes transiently, and exhibited signature refinement of ribbon synapse functions, in close resemblance to native wild-type inner HCs. We demonstrated that co-upregulating Gfi1, Pou4f3, and Atoh1 enhances the efficiency of HC generation and promotes the functional maturation of new HCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , Hair Cells, Auditory/metabolism , Homeodomain Proteins/genetics , Labyrinth Supporting Cells/metabolism , Organogenesis/genetics , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics , Action Potentials/physiology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calbindins/genetics , Calbindins/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Homeodomain Proteins/metabolism , Ion Transport , Labyrinth Supporting Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Myosin VIIa/genetics , Myosin VIIa/metabolism , Neurites/metabolism , Neurites/ultrastructure , Parvalbumins/genetics , Parvalbumins/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Signal Transduction , Transcription Factor Brn-3C/metabolism , Transcription Factors/metabolism
4.
Int J Pediatr Otorhinolaryngol ; 112: 1-5, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30055715

ABSTRACT

BACKGROUND: Consanguinity rate is high in Algeria, and the population is thus at high risk for genetic diseases transmitted on an autosomal recessive mode. Inherited congenital hearing impairment (HI) is a highly heterogeneous disorder, which affects approximately 1 in 800 Algerian newborns. Several hundreds of genes responsible for deafness have been reported among which more than one hundred are responsible for isolated deafness, of which 19 have already been reported to be involved in the Algerian population. This study focuses on patients from the Ghardaïa province, an ethnically and geographically isolated region of Southern Algeria that has the highest consanguinity rate in the country (56%). METHODS: Eleven families, with at least two related members experiencing moderate to profound congenital HI, were recruited and screened for mutations in known HI genes. RESULTS: A preliminary screening for common mutations in GJB2 and GJB6 identified the prevalent GJB2:c.35delG mutation in four families. Targeted exome sequencing further identified the causal mutations in the remaining seven families: CIB2:c.97C > T; p.(Arg33*), MYO7A:c.470+1G > A; p.(?), and SLC26A4:c.410C > T; p.(Ser137Leu) biallelic mutations in two families each, and a TECTA:c.2743 A > G; p.(Ile915Val) monoallelic mutation in the only family with autosomal dominant transmission of the HI. Of note, the missense mutations of SLC26A4 and TECTA had not been previously reported. CONCLUSION: These results further substantiate the genetic heterogeneity of HI, even in reportedly isolated populations. However, several families may harbor the same mutations as a result of a long history of marriages between relatives. This study has important implications for the HI molecular diagnosis strategy, and to develop genetic counseling for families originating from the Ghardaïa province of Algeria.


Subject(s)
Genetic Heterogeneity , Hearing Loss/genetics , Algeria , Calcium-Binding Proteins/genetics , Connexin 26 , Connexins/genetics , Consanguinity , Extracellular Matrix Proteins/genetics , Female , GPI-Linked Proteins/genetics , Genetic Markers , Humans , Male , Membrane Transport Proteins/genetics , Mutation , Myosin VIIa , Myosins/genetics , Sulfate Transporters
5.
PLoS One ; 13(7): e0200210, 2018.
Article in English | MEDLINE | ID: mdl-29979748

ABSTRACT

Hearing loss is the most common sensorineural disorder, affecting over 5% of the population worldwide. Its most frequent cause is the loss of hair cells (HCs), the mechanosensory receptors of the cochlea. HCs transduce incoming sounds into electrical signals that activate auditory neurons, which in turn send this information to the brain. Although some spontaneous HC regeneration has been observed in neonatal mammals, the very small pool of putative progenitor cells that have been identified in the adult mammalian cochlea is not able to replace the damaged HCs, making any hearing impairment permanent. To date, guided differentiation of human cells to HC-like cells has only been achieved using either embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs). However, use of such cell types suffers from a number of important disadvantages, such as the risk of tumourigenicity if transplanted into the host´s tissue. We have obtained cells expressing hair cell markers from cultures of human fibroblasts by overexpression of GFI1, Pou4f3 and ATOH1 (GPA), three genes that are known to play a critical role in the development of HCs. Immunocytochemical, qPCR and RNAseq analyses demonstrate the expression of genes typically expressed by HCs in the transdifferentiated cells. Our protocol represents a much faster approach than the methods applied to ESCs and iPSCs and validates the combination of GPA as a set of genes whose activation leads to the direct conversion of human somatic cells towards the hair cell lineage. Our observations are expected to contribute to the development of future therapies aimed at the regeneration of the auditory organ and the restoration of hearing.


Subject(s)
Cell Transdifferentiation/physiology , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Lineage/physiology , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Hair Cells, Auditory/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Myosin VIIa , Myosins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Transcription Factors/genetics , Tretinoin/pharmacology
6.
Cell Death Dis ; 9(4): 437, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29740017

ABSTRACT

Degeneration or loss of inner ear hair cells (HCs) is irreversible and results in sensorineural hearing loss (SHL). Human-induced pluripotent stem cells (hiPSCs) have been employed in disease modelling and cell therapy. Here, we propose a transcription factor (TF)-driven approach using ATOH1 and regulatory factor of x-box (RFX) genes to generate HC-like cells from hiPSCs. Our results suggest that ATOH1/RFX1/RFX3 could significantly increase the differentiation capacity of iPSCs into MYO7AmCherry-positive cells, upregulate the mRNA expression levels of HC-related genes and promote the differentiation of HCs with more mature stereociliary bundles. To model the molecular and stereociliary structural changes involved in HC dysfunction in SHL, we further used ATOH1/RFX1/RFX3 to differentiate HC-like cells from the iPSCs from patients with myoclonus epilepsy associated with ragged-red fibres (MERRF) syndrome, which is caused by A8344G mutation of mitochondrial DNA (mtDNA), and characterised by myoclonus epilepsy, ataxia and SHL. Compared with isogenic iPSCs, MERRF-iPSCs possessed ~42-44% mtDNA with A8344G mutation and exhibited significantly elevated reactive oxygen species (ROS) production and CAT gene expression. Furthermore, MERRF-iPSC-differentiated HC-like cells exhibited significantly elevated ROS levels and MnSOD and CAT gene expression. These MERRF-HCs that had more single cilia with a shorter length could be observed only by using a non-TF method, but those with fewer stereociliary bundle-like protrusions than isogenic iPSCs-differentiated-HC-like cells could be further observed using ATOH1/RFX1/RFX3 TFs. We further analysed and compared the whole transcriptome of M1ctrl-HCs and M1-HCs after treatment with ATOH1 or ATOH1/RFX1/RFX3. We revealed that the HC-related gene transcripts in M1ctrl-iPSCs had a significantly higher tendency to be activated by ATOH1/RFX1/RFX3 than M1-iPSCs. The ATOH1/RFX1/RFX3 TF-driven approach for the differentiation of HC-like cells from iPSCs is an efficient and promising strategy for the disease modelling of SHL and can be employed in future therapeutic strategies to treat SHL patients.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Hair Cells, Auditory, Inner/metabolism , MERRF Syndrome/pathology , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X1/genetics , Adolescent , Basic Helix-Loop-Helix Transcription Factors/metabolism , Catalase/genetics , Catalase/metabolism , Cilia/physiology , DNA, Mitochondrial/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Female , Hair Cells, Auditory, Inner/cytology , Humans , Induced Pluripotent Stem Cells/cytology , MERRF Syndrome/complications , Myosin VIIa/genetics , Myosin VIIa/metabolism , Point Mutation , Reactive Oxygen Species/metabolism , Regulatory Factor X Transcription Factors/metabolism , Regulatory Factor X1/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
7.
J Biol Chem ; 293(3): 819-829, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29167268

ABSTRACT

Mutations in the MYO7A gene, encoding the motor protein myosin VIIa, can cause Usher 1B, a deafness/blindness syndrome in humans, and the shaker-1 phenotype, characterized by deafness, head tossing, and circling behavior, in mice. Myosin VIIa is responsible for tension bearing and the transduction mechanism in the stereocilia and for melanosome transport in the retina, in line with the phenotypic outcomes observed in mice. However, the effect of the shaker-1 mutation, a R502P amino acid substitution, on the motor function is unclear. To explore this question, we determined the kinetic properties and the effect on the filopodial tip localization of the recombinant mouse myosin VIIa-5IQ-SAH R502P (myoVIIa-sh1) construct. Interestingly, although residue 502 is localized to a region thought to be involved in interacting with actin, the kinetic parameters for actin binding changed only slightly for the mutant construct. However, the rate constant for ATP hydrolysis (k+H + k-H) was reduced by ∼200-fold from 12 s-1 to 0.05 s-1, making the hydrolysis step the rate-limiting step of the ATPase cycle in the presence and absence of actin. Given that wild-type mouse myosin VIIa is a slow, high-duty ratio, monomeric motor, this altered hydrolysis rate would reduce activity to extremely low levels. Indeed, the translocation to the filopodial tips was hampered by the diminished motor function of a dimeric construct of the shaker-1 mutant. We conclude that the diminished motor activity of this mutant is most likely responsible for impaired hearing in the shaker-1 mice.


Subject(s)
Adenosine Triphosphate/metabolism , Myosins/genetics , Myosins/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Mice , Mutation/genetics , Myosin VIIa , Retina/metabolism
8.
Structure ; 25(4): 579-591.e4, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28262393

ABSTRACT

Several unconventional myosins contain a highly charged single α helix (SAH) immediately following the calmodulin (CaM) binding IQ motifs, functioning to extend lever arms of these myosins. How such SAH is connected to the IQ motifs and whether the conformation of the IQ motifs-SAH segments are regulated by Ca2+ fluctuations are not known. Here, we demonstrate by solving its crystal structure that the predicted SAH of myosin VIIa (Myo7a) forms a stable SAH. The structure of Myo7a IQ5-SAH segment in complex with apo-CaM reveals that the SAH sequence can extend the length of the Myo7a lever arm. Although Ca2+-CaM remains bound to IQ5-SAH, the Ca2+-induced CaM binding mode change softens the conformation of the IQ5-SAH junction, revealing a Ca2+-induced lever arm flexibility change for Myo7a. We further demonstrate that the last IQ motif of several other myosins also binds to both apo- and Ca2+-CaM, suggesting a common Ca2+-induced conformational regulation mechanism.


Subject(s)
Calcium/metabolism , Myosins/chemistry , Myosins/metabolism , Apolipoproteins/metabolism , Binding Sites , Calmodulin/metabolism , HeLa Cells , Humans , Models, Molecular , Myosin VIIa , Protein Binding , Protein Domains , Protein Structure, Secondary
9.
Mol Vis ; 22: 827-35, 2016.
Article in English | MEDLINE | ID: mdl-27440999

ABSTRACT

PURPOSE: Usher syndrome accounts for about 50% of all hereditary deaf-blindness cases. The most severe form of this syndrome, Usher syndrome type I (USH1), is characterized by profound congenital sensorineural deafness, vestibular dysfunction, and retinitis pigmentosa. Six USH1 genes have been identified, MYO7A, CDH23, PCDH15, USH1C, SANS, and CIB2, encoding myosin VIIA, cadherin-23, protocadherin-15, harmonin, scaffold protein containing ankyrin repeats and a sterile alpha motif (SAM) domain, and calcium- and integrin-binding member 2, respectively. METHODS: In the present study, we recruited four Tunisian families with a diagnosis of USH1, together with healthy unrelated controls. Affected members underwent detailed audiologic and ocular examinations. We used the North African Deafness (NADf) chip to search for known North African mutations associated with USH. Then, we selected microsatellite markers covering USH1 known loci to genotype the DNA samples. Finally, we performed DNA sequencing of three known USH1 genes: MYO7A, PCDH15, and USH1C. RESULTS: Four biallelic mutations, all single base changes, were found in the MYO7A, USH1C, and PCDH15 genes. These mutations consist of a previously reported splicing defect c.470+1G>A in MYO7A, three novel variants, including two nonsense (p.Arg3X and p.Arg134X) in USH1C and PCDH15, respectively, and one frameshift (p.Lys615Asnfs*6) in MYO7A. CONCLUSIONS: We found a remarkable genetic heterogeneity in the studied families with USH1 with a variety of mutations, among which three were novel. These novel mutations will be included in the NADf mutation screening chip that will allow a higher diagnosis efficiency of this extremely genetically heterogeneous disease. Ultimately, efficient molecular diagnosis of USH in a patient's early childhood is of utmost importance, allowing better educational and therapeutic management.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cadherins/genetics , Codon, Nonsense , Frameshift Mutation , Myosins/genetics , Usher Syndromes/diagnosis , Usher Syndromes/genetics , Adolescent , Adult , Cadherin Related Proteins , Cell Cycle Proteins , Consanguinity , Cytoskeletal Proteins , DNA Mutational Analysis , Electroretinography , Female , Genetic Testing , Humans , Male , Middle Aged , Molecular Diagnostic Techniques , Myosin VIIa , Pedigree , Sequence Analysis, DNA , Tunisia , Young Adult
10.
Neurotox Res ; 29(4): 594-604, 2016 May.
Article in English | MEDLINE | ID: mdl-26913517

ABSTRACT

Mild maternal iron deficiency anemia (IDA) adversely affects the development of cochlear hair cells of the young offspring, but the mechanisms underlying the association are incompletely understood. The aim of this study was to evaluate whether mild maternal IDA in guinea pigs could interrupt inner hair cell (IHC) ribbon synapse density and outer hair cell motility of the offspring. Here, we established a dietary restriction model that allows us to study quantitative changes in the number of IHC ribbon synapses and hearing impairment in response to mild maternal IDA in young guinea pig. The offspring were weaned on postnatal day (PND) 9 and then were given the iron-sufficient diet. On PND 24, pups were examined the hearing function by auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) measurements. Then, the cochleae were harvested for assessment of the number of IHC ribbon synapses by immunofluorescence, the morphology of cochlear hair cells, and spiral ganglion cells (SGCs) by scanning electron microscope and hematoxylin-eosin staining, the location, and expression of vesicular glutamate transporter (VGLUT) 3, myosin VIIa, and prestin by immunofluorescence and blotting. Here, we show that mild maternal IDA in guinea pigs induced elevated ABR threshold shifts, declined DPOAE level shifts, and reduced the number of ribbon synapses, impaired the morphology of cochlear hair cells and SGCs in offsprings. In addition, downregulation of VGLUT3 and myosin VIIa, and upregulation of prestin were observed in the cochlea of offsprings from mild maternal IDA in guinea pigs. These data indicate that mild maternal IDA in guinea pigs induced hearing impairment in offsprings, and this deficit may be attributed to the reduction of ribbon synapse density and dysregulation of VGLUT3, myosin VIIa, and prestin.


Subject(s)
Anemia, Iron-Deficiency/complications , Hearing Loss/etiology , Hearing Loss/pathology , Myosins/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Synapses/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Acoustic Stimulation , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Guinea Pigs , Hair Cells, Auditory, Inner/cytology , Male , Myosin VIIa , Otoacoustic Emissions, Spontaneous , Pregnancy , Psychoacoustics , Spiral Ganglion/cytology , Spiral Ganglion/ultrastructure , Synapses/ultrastructure , Up-Regulation/physiology
11.
Eur J Histochem ; 59(2): 2486, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-26150157

ABSTRACT

Inositol 1, 4, 5-trisphosphate receptor (IP3R) has been established to be essential for hearing. However, the expression of IP3R in the cochlea in the period of auditory development remains unknown. We investigated the expression of IP3R in the developing rat cochlea using immunohistochemistry and real-time reverse transcription polymerase chain reaction (RT-PCR). We observed its presence in the developing rat cochlea, and changes in IP3R protein expressions from the early post-natal period to adult. At birth (post-natal day 0, P0), IP3R expression was only found in Hensen's cell. IP3R immunoreactivity first appeared in the sensory hair cells in the organ of Corti at P2. This localization was confirmed by means of double-labeling experiments with Myosin VIIA, a marker for cochlear hair cells. Colocalization of IP3R and Myosin VIIA from P2 to the second post-natal week suggested early expression of IP3R in developing inner and outer hair cells. Claudius' cells near the spiral ligament were labelled for IP3R from P8 onwards. Transient IP3R expression was observed in the stria vascularis in early post-natal rat from P4 to P8. Spiral ganglion neurons also exhibited weaker IP3R fluorescence signals during post-natal development. The results of RT-PCR demonstrated that all three IP3R isoforms (IP3R1, IP3R2, and IP3R3) were present in rat cochlea during four different developmental stages of cochlea, from P0 to P28. Present immunohistochemical evidence for both change and maintenance of expression of IP3R during post-natal development of the rat cochlea indicated the possible involvement of IP3R-mediated calcium signaling in cochlear development.


Subject(s)
Cochlea/growth & development , Cochlea/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Animals , Blotting, Western , Calcium Signaling/physiology , Female , Hair Cells, Auditory, Inner/metabolism , Immunohistochemistry , Male , Microscopy, Confocal , Myosin VIIa , Myosins/metabolism , Organ of Corti/growth & development , Organ of Corti/metabolism , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Spiral Ganglion/growth & development , Spiral Ganglion/metabolism , Stria Vascularis/metabolism
12.
Neurosci Lett ; 597: 183-8, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25956034

ABSTRACT

To evaluate whether cochlear inner hair cells (IHCs) ribbon synapse plasticity would be interrupted by insulin resistance (IR) due to dietary iron overload, we established an IR model in C57Bl/6 male mice with an iron-enriched diet for 16 weeks. Glucose levels were measured at weeks 4, 8, 12, 16. Glucose tolerance test and insulin tolerance test were performed at week 16 after overnight fasting. Then, auditory brainstem responses (ABRs) measurements were performed for hearing threshold shifts. After ABR measurements, cochleae were harvested for assessment of the number of IHC ribbon synapses by immunostaining, the morphology of cochlear hair cells and spiral ganglion neurons (SGNs) by transmission electron microscopy or immunostaining. Here, we show that IR due to dietary iron overload decreased the number of ribbon synapses, and induced moderate ABR threshold elevations. Besides, additional components including outer hair cells (OHCs), IHCs, and SGNs were unaffected. Moreover, IR did not disrupt the expression of vesicular glutamate transporter 3 (VGLUT3), myosin VIIa and prestin in hair cells. These results indicate that IHC ribbon synapses may be more susceptible to IR due to dietary iron overload.


Subject(s)
Hair Cells, Auditory, Inner/physiology , Insulin Resistance , Iron Overload/metabolism , Iron, Dietary/administration & dosage , Synapses/physiology , Amino Acid Transport Systems, Acidic/metabolism , Animals , Evoked Potentials, Auditory, Brain Stem , Iron Overload/physiopathology , Male , Mice, Inbred C57BL , Molecular Motor Proteins/metabolism , Myosin VIIa , Myosins/metabolism , Neuronal Plasticity
13.
J Biol Chem ; 290(28): 17587-98, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26001786

ABSTRACT

Human myosin VIIA (HM7A) is responsible for human Usher syndrome type 1B, which causes hearing and visual loss in humans. Here we studied the regulation of HM7A. The actin-activated ATPase activity of full-length HM7A (HM7AFull) was lower than that of tail-truncated HM7A (HM7AΔTail). Deletion of the C-terminal 40 amino acids and mutation of the basic residues in this region (R2176A or K2179A) abolished the inhibition. Electron microscopy revealed that HM7AFull is a monomer in which the tail domain bends back toward the head-neck domain to form a compact structure. This compact structure is extended at high ionic strength or in the presence of Ca(2+). Although myosin VIIA has five isoleucine-glutamine (IQ) motifs, the neck length seems to be shorter than the expected length of five bound calmodulins. Supporting this observation, the IQ domain bound only three calmodulins in Ca(2+), and the first IQ motif failed to bind calmodulin in EGTA. These results suggest that the unique IQ domain of HM7A is important for the tail-neck interaction and, therefore, regulation. Cellular studies revealed that dimer formation of HM7A is critical for its translocation to filopodial tips and that the tail domain (HM7ATail) markedly reduced the filopodial tip localization of the HM7AΔTail dimer, suggesting that the tail-inhibition mechanism is operating in vivo. The translocation of the HM7AFull dimer was significantly less than that of the HM7AΔTail dimer, and R2176A/R2179A mutation rescued the filopodial tip translocation. These results suggest that HM7A can transport its cargo molecules, such as USH1 proteins, upon release of the tail-dependent inhibition.


Subject(s)
Myosins/chemistry , Myosins/metabolism , Amino Acid Sequence , Binding Sites , Calmodulin/metabolism , HeLa Cells , Humans , Microscopy, Electron, Transmission , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Myosin VIIa , Myosins/genetics , Protein Multimerization , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sequence Homology, Amino Acid , Usher Syndromes/genetics , Usher Syndromes/metabolism
14.
J Comp Neurol ; 523(8): 1258-80, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25560461

ABSTRACT

In the vestibular periphery of nearly every vertebrate, cholinergic vestibular efferent neurons give rise to numerous presynaptic varicosities that target hair cells and afferent processes in the sensory neuroepithelium. Although pharmacological studies have described the postsynaptic actions of vestibular efferent stimulation in several species, characterization of efferent innervation patterns and the relative distribution of efferent varicosities among hair cells and afferents are also integral to understanding how efferent synapses operate. Vestibular efferent markers, however, have not been well characterized in the turtle, one of the animal models used by our laboratory. Here we sought to identify reliable efferent neuronal markers in the vestibular periphery of turtle, to use these markers to understand how efferent synapses are organized, and to compare efferent neuronal labeling patterns in turtle with two other amniotes using some of the same markers. Efferent fibers and varicosities were visualized in the semicircular canal of red-eared turtles (Trachemys scripta elegans), zebra finches (Taeniopygia guttata), and mice (Mus musculus) utilizing fluorescent immunohistochemistry with antibodies against choline acetyltransferase (ChAT). Vestibular hair cells and afferents were counterstained using antibodies to myosin VIIa and calretinin. In all species, ChAT labeled a population of small diameter fibers giving rise to numerous spherical varicosities abutting type II hair cells and afferent processes. That these ChAT-positive varicosities represent presynaptic release sites were demonstrated by colabeling with antibodies against the synaptic vesicle proteins synapsin I, SV2, or syntaxin and the neuropeptide calcitonin gene-related peptide. Comparisons of efferent innervation patterns among the three species are discussed.


Subject(s)
Neurons, Efferent/cytology , Semicircular Canals/innervation , Turtles/anatomy & histology , Animals , Blotting, Western , Calbindin 2/metabolism , Cell Size , Choline O-Acetyltransferase/metabolism , Female , Finches/anatomy & histology , Finches/metabolism , Fluorescent Antibody Technique , Hair Cells, Vestibular/cytology , Hair Cells, Vestibular/metabolism , Male , Mice/anatomy & histology , Mice/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Myosin VIIa , Myosins/metabolism , Neurons, Efferent/metabolism , Semicircular Canals/metabolism , Species Specificity , Synapses/metabolism , Turtles/metabolism
15.
J Neurosci ; 34(31): 10453-8, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080603

ABSTRACT

The neuroactive peptide calcitonin-gene related peptide (CGRP) is known to act at efferent synapses and their targets in hair cell organs, including the cochlea and lateral line. CGRP is also expressed in vestibular efferent neurons as well as a number of central vestibular neurons. Although CGRP-null (-/-) mice demonstrate a significant reduction in cochlear nerve sound-evoked activity compared with wild-type mice, it is unknown whether and how the loss of CGRP influence vestibular system function. Vestibular function was assessed by quantifying the vestibulo-ocular reflex (VOR) in alert mice. The loss of CGRP in (-/-) mice was associated with a reduction of the VOR gain of ≈50% without a concomitant change in phase. Using immunohistochemistry, we confirmed that, although CGRP staining was absent in the vestibular end-organs of null (-/-) mice, cholinergic staining appeared normal, suggesting that the overall gross development of vestibular efferent innervation was unaltered. We further confirmed that the observed deficit in vestibular function of null (-/-) mice was not the result of nontargeted effects at the level of the extraocular motor neurons and/or their innervation of extraocular muscles. Analysis of the relationship between vestibular quick phase amplitude and peak velocity revealed that extraocular motor function was unchanged, and immunohistochemistry revealed no abnormalities in motor endplates. Together, our findings show that the neurotransmitter CGRP plays a key role in ensuring VOR efficacy.


Subject(s)
Calcitonin Gene-Related Peptide/deficiency , Reflex, Vestibulo-Ocular/genetics , Analysis of Variance , Animals , Botulinum Toxins, Type A/metabolism , Calbindin 2/metabolism , Calcitonin Gene-Related Peptide/genetics , Choline O-Acetyltransferase/metabolism , Eye Movements/genetics , Female , Gene Expression Regulation/genetics , Male , Mice , Mice, Knockout , Myosin VIIa , Myosins/metabolism , Vestibule, Labyrinth/metabolism
16.
Mol Biol Rep ; 41(9): 5645-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903972

ABSTRACT

Valvular calcification precedes the development of valvular stenosis and may represent an important early phenotype for valvular heart disease. It is known that development of valvular calcification is likely to occur among members of a family. However, the knowledge about the role of genomic predictive markers in valvular calcification is still elusive. Aims of this review are to assess the impact of gene polymorphisms on risk and severity of aortic stenosis and mitral annular calcification. According to the results of the investigations carried out, all polymorphisms may be divided into the three groups conferring the level of evidence of their association with valvular stenosis. It is possible to conclude that apoB (XbaI, rs1042031, and rs6725189), ACE (rs4340), IL10 (rs1800896 and rs1800872), and LPA (rs10455872) gene polymorphisms may be associated with valvular calcific stenosis with a relatively high level of evidence. A number of other polymorphisms, such as PvuII polymorphism within the ORα gene, rs1042636 polymorphism within the CaSR gene, rs3024491, rs3021094, rs1554286, and rs3024498 polymorphisms within the IL10 gene, rs662 polymorphism within the PON1 gene, rs2276288 polymorphism within the MYO7A gene, rs5194 polymorphism within the AGTR1 gene, rs2071307 polymorphism within the ELN gene, rs17659543 and rs13415097 polymorphisms within the IL1F9 gene may correlate with a risk of calcific valve stenosis with moderate level of evidence. Finally, rs1544410 polymorphism within the VDR gene, E2 and E4 alleles within the apoE gene, rs6254 polymorphism within the PTH gene, and rs1800871 polymorphism within the IL10 gene may be associated with aortic stenosis with low level of evidence.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve/pathology , Calcinosis/genetics , Genetic Predisposition to Disease , Vascular Calcification/genetics , Alleles , Aortic Valve Stenosis/pathology , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Aryldialkylphosphatase/genetics , Calcinosis/pathology , Evaluation Studies as Topic , Humans , Interleukin-1/genetics , Interleukin-10/genetics , Myosin VIIa , Myosins/genetics , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Receptor, Angiotensin, Type 1/genetics , Receptors, Calcitriol/genetics , Receptors, Calcium-Sensing/genetics , Vascular Calcification/pathology
17.
Exp Eye Res ; 122: 65-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24698764

ABSTRACT

Mutations in myosin VIIa (MYO7A) cause Usher Syndrome 1B (USH1B), a disease characterized by the combination of sensorineural hearing loss and visual impairment termed retinitis pigmentosa (RP). Although the shaker-1 mouse model of USH1B exists, only minor defects in the retina have been observed during its lifespan. Previous studies of the zebrafish mariner mutant, which also carries a mutation in myo7aa, revealed balance and hearing defects in the mutants but the retinal phenotype has not been described. We found elevated cell death in the outer nuclear layer (ONL) of myo7aa(-/-) mutants. While myo7aa(-/-) mutants retained visual behaviors in the optokinetic reflex (OKR) assay, electroretinogram (ERG) recordings revealed a significant decrease in both a- and b-wave amplitudes in mutant animals, but not a change in ERG threshold sensitivity. Immunohistochemistry showed mislocalization of rod and blue cone opsins and reduced expression of rod-specific markers in the myo7aa(-/-) ONL, providing further evidence that the photoreceptor degeneration observed represents the initial stages of the RP. Further, constant light exposure resulted in widespread photoreceptor degeneration and the appearance of large holes in the retinal pigment epithelium (RPE). No differences were observed in the retinomotor movements of the photoreceptors or in melanosome migration within the RPE, suggesting that myo7aa(-/-) does not function in these processes in teleosts. These results indicate that the zebrafish myo7aa(-/-) mutant is a useful animal model for the RP seen in humans with USH1B.


Subject(s)
Codon, Nonsense , Myosins/genetics , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cell Death , Dark Adaptation , Disease Models, Animal , Electroretinography , Immunohistochemistry , In Situ Nick-End Labeling , Light , Melanosomes/physiology , Microscopy, Electron, Transmission , Myosin VIIa , Nystagmus, Optokinetic/physiology , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/metabolism , Rod Opsins/metabolism , Usher Syndromes/genetics , Usher Syndromes/metabolism , Usher Syndromes/pathology
18.
J Neurosci ; 34(16): 5505-14, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741041

ABSTRACT

Tip links between adjacent stereocilia are believed to gate mechano-electrical transducer (MET) channels and mediate the electrical responses of sensory hair cells. We found that mouse auditory hair cells that lack tip links due to genetic mutations or exposure to the Ca(2+) chelator BAPTA can, however, still respond to mechanical stimuli. These MET currents have unusual properties and are predominantly of the opposite polarity relative to those measured when tip links are present. There are other striking differences, for example, the channels are usually all closed when the hair cell is not stimulated and the currents in response to strong stimuli can be substantially larger than normal. These anomalous MET currents can also be elicited early in development, before the onset of mechano-electrical transduction with normal response polarity. Current-voltage curves of the anomalous MET currents are linear and do not show the rectification characteristic of normal MET currents. The permeant MET channel blocker dihydrostreptomycin is two orders of magnitude less effective in blocking the anomalous MET currents. The findings suggest the presence of a large population of MET channels with pore properties that are distinct from those of normal MET channels. These channels are not gated by hair-bundle links and can be activated under a variety of conditions in which normal tip-link-mediated transduction is not operational.


Subject(s)
Cell Membrane Permeability/physiology , Hair Cells, Auditory/physiology , Ion Channels/physiology , Mechanotransduction, Cellular/physiology , Animals , Animals, Newborn , Cadherin Related Proteins , Cadherins/genetics , Cell Membrane Permeability/genetics , Chelating Agents/pharmacology , Dihydrostreptomycin Sulfate/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Embryo, Mammalian , Female , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , In Vitro Techniques , Ion Channels/drug effects , Male , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myosin VIIa , Myosins/genetics , Organ of Corti/cytology , Protein Precursors/genetics
19.
J Neurosci ; 33(47): 18409-24, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259566

ABSTRACT

Auditory neuropathy is a form of hearing loss in which cochlear inner hair cells fail to correctly encode or transmit acoustic information to the brain. Few genes have been implicated in the adult-onset form of this disease. Here we show that mice lacking the transcription factor Foxo3 have adult onset hearing loss with the hallmark characteristics of auditory neuropathy, namely, elevated auditory thresholds combined with normal outer hair cell function. Using histological techniques, we demonstrate that Foxo3-dependent hearing loss is not due to a loss of cochlear hair cells or spiral ganglion neurons, both of which normally express Foxo3. Moreover, Foxo3-knock-out (KO) inner hair cells do not display reductions in numbers of synapses. Instead, we find that there are subtle structural changes in and surrounding inner hair cells. Confocal microscopy in conjunction with 3D modeling and quantitative analysis show that synaptic localization is altered in Foxo3-KO mice and Myo7a immunoreactivity is reduced. TEM demonstrates apparent afferent degeneration. Strikingly, acoustic stimulation promotes Foxo3 nuclear localization in vivo, implying a connection between cochlear activity and synaptic function maintenance. Together, these findings support a new role for the canonical damage response factor Foxo3 in contributing to the maintenance of auditory synaptic transmission.


Subject(s)
Cochlea/pathology , Forkhead Transcription Factors/genetics , Hearing Loss, Central/genetics , Hearing Loss, Central/pathology , Mutation/genetics , Synapses/pathology , Acoustic Stimulation , Age Factors , Alcohol Oxidoreductases , Animals , Animals, Newborn , Calcium-Binding Proteins/metabolism , Co-Repressor Proteins , Cochlea/growth & development , Cochlea/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Inner/ultrastructure , Hearing Loss, Central/physiopathology , Imaging, Three-Dimensional , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Myosin VIIa , Myosins/metabolism , Phosphoproteins/metabolism , Receptors, AMPA/metabolism , Synapses/genetics , Synapses/ultrastructure
20.
Physiol Genomics ; 45(21): 987-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24022220

ABSTRACT

Usher syndrome (USH) is a neurosensory disorder affecting both hearing and vision in humans. Linkage studies of families of USH patients, studies in animals, and characterization of purified proteins have provided insight into the molecular mechanisms of hearing. To date, 11 USH proteins have been identified, and evidence suggests that all of them are crucial for the function of the mechanosensory cells of the inner ear, the hair cells. Most USH proteins are localized to the stereocilia of the hair cells, where mechano-electrical transduction (MET) of sound-induced vibrations occurs. Therefore, elucidation of the functions of USH proteins in the stereocilia is a prerequisite to understanding the exact mechanisms of MET.


Subject(s)
Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Stereocilia/metabolism , Usher Syndromes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins , Cytoskeletal Proteins , Ear, Inner/pathology , Ear, Inner/physiopathology , Hair Cells, Auditory/pathology , Humans , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Mutation , Myosin VIIa , Myosins/genetics , Myosins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Stereocilia/pathology , Usher Syndromes/genetics , Usher Syndromes/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL