Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Vis Exp ; (210)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39248532

ABSTRACT

Myosin-7a is an actin-based motor protein vital for auditory and visual processes. Mutations in myosin-7a lead to Usher syndrome type 1, the most common and severe form of deaf-blindness in humans. It is hypothesized that myosin-7a forms a transmembrane adhesion complex with other Usher proteins, essential for the structural-functional integrity of photoreceptor and cochlear hair cells. However, due to the challenges in obtaining pure, intact protein, the exact functional mechanisms of human myosin-7a remain elusive, with limited structural and biomechanical studies available. Recent studies have shown that mammalian myosin-7a is a multimeric motor complex consisting of a heavy chain and three types of light chains: regulatory light chain (RLC), calmodulin, and calmodulin-like protein 4 (CALML4). Unlike calmodulin, CALML4 does not bind to calcium ions. Both the calcium-sensitive, and insensitive calmodulins are critical for mammalian myosin-7a for proper fine-tuning of its mechanical properties. Here, we describe a detailed method to produce recombinant human myosin-7a holoenzyme using the MultiBac Baculovirus protein expression system. This yields milligram quantities of high-purity full-length protein, allowing for its biochemical and biophysical characterization. We further present a protocol for assessing its mechanical and motile properties using tailored in vitro motility assays and fluorescence microscopy. The availability of the intact human myosin-7a protein, along with the detailed functional characterization protocol described here, paves the way for further investigations into the molecular aspects of myosin-7a in vision and hearing.


Subject(s)
Myosin VIIa , Humans , Myosin VIIa/metabolism , Myosin VIIa/genetics , Myosins/chemistry , Myosins/metabolism , Myosins/genetics , Myosins/isolation & purification , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Sf9 Cells , Spodoptera
2.
J Physiol ; 602(16): 3995-4025, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39037943

ABSTRACT

The hair bundle of cochlear hair cells comprises specialized microvilli, the stereocilia, which fulfil the role of mechanotransduction. Genetic defects and environmental noise challenge the maintenance of hair bundle structure, critically contributing to age-related hearing loss. Stereocilia fusion is a major component of the hair bundle pathology in mature hair cells, but its role in hearing loss and its molecular basis are poorly understood. Here, we utilized super-resolution expansion microscopy to examine the molecular anatomy of outer hair cell stereocilia fusion in mouse models of age-related hearing loss, heightened endoplasmic reticulum stress and prolonged noise exposure. Prominent stereocilia fusion in our model of heightened endoplasmic reticulum stress, Manf (Mesencephalic astrocyte-derived neurotrophic factor)-inactivated mice in a background with Cadherin 23 missense mutation, impaired mechanotransduction and calcium balance in stereocilia. This was indicated by reduced FM1-43 dye uptake through the mechanotransduction channels, reduced neuroplastin/PMCA2 expression and increased expression of the calcium buffer oncomodulin inside stereocilia. Sparse BAIAP2L2 and myosin 7a expression was retained in the fused stereocilia but mislocalized away from their functional sites at the tips. These hair bundle abnormalities preceded cell soma degeneration, suggesting a sequela from stereociliary molecular perturbations to cell death signalling. In the age-related hearing loss and noise-exposure models, stereocilia fusion was more restricted within the bundles, yet both models exhibited oncomodulin upregulation at the fusion sites, implying perturbed calcium homeostasis. We conclude that stereocilia fusion is linked with the failure to maintain cellular proteostasis and with disturbances in stereociliary calcium balance. KEY POINTS: Stereocilia fusion is a hair cell pathology causing hearing loss. Inactivation of Manf, a component of the endoplasmic reticulum proteostasis machinery, has a cell-intrinsic mode of action in triggering outer hair cell stereocilia fusion and the death of these cells. The genetic background with Cadherin 23 missense mutation contributes to the high susceptibility of outer hair cells to stereocilia fusion, evidenced in Manf-inactivated mice and in the mouse models of early-onset hearing loss and noise exposure. Endoplasmic reticulum stress feeds to outer hair cell stereocilia bundle pathology and impairs the molecular anatomy of calcium regulation. The maintenance of the outer hair cell stereocilia bundle cohesion is challenged by intrinsic and extrinsic stressors, and understanding the underlying mechanisms will probably benefit the development of interventions to promote hearing health.


Subject(s)
Cadherins , Hair Cells, Auditory, Outer , Mechanotransduction, Cellular , Stereocilia , Animals , Stereocilia/metabolism , Stereocilia/pathology , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Mice , Cadherins/metabolism , Cadherins/genetics , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , Male , Calcium/metabolism , Myosin VIIa/metabolism , Female , Hearing Loss/pathology , Hearing Loss/genetics , Hearing Loss/metabolism , Mutation, Missense , Calcium-Binding Proteins
3.
Mol Biol Rep ; 51(1): 683, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796585

ABSTRACT

BACKGROUND: Usher syndrome 1 (USH1) is the most severe subtype of Usher syndrome characterized by severe sensorineural hearing impairment, retinitis pigmentosa, and vestibular areflexia. USH1 is usually induced by variants in MYO7A, a gene that encodes the myosin-VIIa protein. Myosin-VIIA is effectively involved in intracellular molecular traffic essential for the proper function of the cochlea, the retinal photoreceptors, and the retinal pigmented epithelial cells. METHODS AND RESULTS: In this study, we report a new homozygous missense variant (NM_000260.4: c.1657 C > T p.(His553Tyr)) in MYO7A of a 28-year-old female with symptoms consistent with USH1. This variant, c.1657 C > T p.(His553Tyr) is positioned in the highly conserved myosin-VIIA motor domain. Previous studies showed that variants in this domain might disrupt the ability of the protein to bind to actin and thus cause the disorder. CONCLUSIONS: Our findings contribute to our understanding of the phenotypic and mutational spectrum of USH1 associated with autosomal recessive MYO7A variants and emphasize the important role of molecular testing in accurately diagnosing this syndrome. More advanced research is required to understand the functional effect of the identified variant and the genotype-phonotype correlations of MYO7A-related Usher syndrome 1.


Subject(s)
Homozygote , Mutation, Missense , Myosin VIIa , Usher Syndromes , Usher Syndromes/genetics , Myosin VIIa/metabolism , Myosin VIIa/genetics , Humans , Female , Mutation, Missense/genetics , Adult , Myosins/genetics , Pedigree
4.
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
5.
Vision Res ; 212: 108311, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37586294

ABSTRACT

Usher syndrome type 1B (USH1B) is a deaf-blindness disorder, caused by mutations in the MYO7A gene, which encodes the heavy chain of an unconventional actin-based motor protein. Here, we examined the two retinal isoforms of MYO7A, IF1 and IF2. We compared 3D models of the two isoforms and noted that the 38-amino acid region that is present in IF1 but absent from IF2 affects the C lobe of the FERM1 domain and the opening of a cleft in this potentially important protein binding domain. Expression of each of the two isoforms of human MYO7A and pig and mouse Myo7a was detected in the RPE and neural retina. Quantification by qPCR showed that the expression of IF2 was typically âˆ¼ 7-fold greater than that of IF1. We discuss the implications of these findings for any USH1B gene therapy strategy. Given the current incomplete knowledge of the functions of each isoform, both isoforms should be considered for targeting both the RPE and the neural retina in gene augmentation therapies.


Subject(s)
Usher Syndromes , Humans , Mice , Animals , Swine , Usher Syndromes/genetics , Usher Syndromes/therapy , Usher Syndromes/metabolism , Myosin VIIa/genetics , Myosin VIIa/metabolism , Retina/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Mutation , Genetic Therapy
6.
Sci Rep ; 11(1): 13893, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230535

ABSTRACT

Several studies have shown how different cell lines can influence the differentiation of stem cells through co-culture systems. The House Ear Institute-Organ of Corti 1 (HEI-OC1) is considered an important cell line for in vitro auditory research. However, it is unknown if HEI-OC1 cells can promote the differentiation of embryonic stem cells (ESCs). In this study, we investigated whether co-culture of ESCs with HEI-OC1 cells promotes differentiation. To this end, we developed a co-culture system of mouse ESCs with HEI-OC1 cells. Dissociated or embryonic bodies (EBs) of ESCs were introduced to a conditioned and inactivated confluent layer of HEI-OC1 cells for 14 days. The dissociated ESCs coalesced into an EB-like form that was smaller than the co-cultured EBs. Contact co-culture generated cells expressing several otic progenitor markers as well as hair cell specific markers. ESCs and EBs were also cultured in non-contact setup but using conditioned medium from HEI-OC1 cells, indicating that soluble factors alone could have a similar effect. The ESCs did not form into aggregates but were still Myo7a-positive, while the EBs degenerated. However, in the fully differentiated EBs, evidence to prove mature differentiation of inner ear hair cell was still rudimentary. Nevertheless, these results suggest that cellular interactions between ESCs and HEI-OC1 cells may both stimulate ESC differentiation.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Hair Cells, Auditory/cytology , Animals , Biomarkers/metabolism , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Epithelium/metabolism , Gene Expression Regulation/drug effects , Mice , Myosin VIIa/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism
7.
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
8.
Acta Ophthalmol ; 99(8): 922-930, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33576163

ABSTRACT

PURPOSE: We aimed to establish correlations between the clinical features of a cohort of Usher syndrome (USH) patients with pathogenic variants in MYO7A, type of pathogenic variant, and location on the protein domain. METHODS: Sixty-two USH patients from 46 families with biallelic variants in MYO7A were examined for visual and audiological features. Participants were evaluated based on self-reported ophthalmological history and ophthalmological investigations (computerized visual field testing, best-corrected visual acuity, and ophthalmoscopic and electrophysiological examination). Optical coherence tomography and fundus autofluorescence imaging were performed when possible. Auditory and vestibular functions were evaluated. Patients were classified according to the type of variant and the protein domain where the variants were located. RESULTS: Most patients displayed a typical USH1 phenotype, that is, prelingual severe-profound sensorineural hearing loss, prepubertal retinitis pigmentosa (RP) and vestibular dysfunction. No statistically significant differences were observed for the variables analysed except for the onset of hearing loss due to the existence of two USH2 cases, defined as postlingual sensorineural hearing loss, postpubertal onset of RP, and absence of vestibular dysfunction, and one atypical case of USH. CONCLUSION: We were unable to find a correlation between genotype and phenotype for MYO7A. However, our findings could prove useful for the assessment of efficacy in clinical trials, since the type of MYO7A variant does not seem to change the onset, severity or course of visual disease.


Subject(s)
Clinical Trials as Topic , DNA/genetics , Genetic Association Studies/methods , Mutation, Missense , Myosin VIIa/genetics , Usher Syndromes/genetics , Adolescent , Adult , Aged , Child , DNA Mutational Analysis , Female , Fluorescein Angiography/methods , Fundus Oculi , Genotype , Humans , Male , Middle Aged , Myosin VIIa/metabolism , Pedigree , Phenotype , Retina/diagnostic imaging , Retrospective Studies , Tomography, Optical Coherence/methods , Usher Syndromes/diagnosis , Young Adult
9.
Nat Commun ; 12(1): 563, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33495456

ABSTRACT

Myosin-7a, despite being monomeric in isolation, plays roles in organizing actin-based cell protrusions such as filopodia, microvilli and stereocilia, as well as transporting cargoes within them. Here, we identify a binding protein for Drosophila myosin-7a termed M7BP, and describe how M7BP assembles myosin-7a into a motile complex that enables cargo translocation and actin cytoskeletal remodeling. M7BP binds to the autoinhibitory tail of myosin-7a, extending the molecule and activating its ATPase activity. Single-molecule reconstitution show that M7BP enables robust motility by complexing with myosin-7a as 2:2 translocation dimers in an actin-regulated manner. Meanwhile, M7BP tethers actin, enhancing complex's processivity and driving actin-filament alignment during processive runs. Finally, we show that myosin-7a-M7BP complex assembles actin bundles and filopodia-like protrusions while migrating along them in living cells. Together, these findings provide insights into the mechanisms by which myosin-7a functions in actin protrusions.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Myosin VIIa/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Microscopy, Fluorescence/methods , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Myosin VIIa/chemistry , Myosin VIIa/genetics , Protein Binding , Protein Multimerization , Pseudopodia/genetics , Pseudopodia/physiology , Stereocilia/genetics , Stereocilia/physiology
10.
FEBS J ; 288(1): 325-353, 2021 01.
Article in English | MEDLINE | ID: mdl-32323465

ABSTRACT

Cochlear development is a complex process with precise spatiotemporal patterns. A detailed understanding of this process is important for studies of congenital hearing loss and regenerative medicine. However, much of our understanding of cochlear development is based on rodent models. Animal models that bridge the gap between humans and rodents are needed. In this study, we investigated the development of hearing organs in a small New World monkey species, the common marmoset (Callithrix jacchus). We describe the general stages of cochlear development in comparison with those of humans and mice. Moreover, we examined more than 25 proteins involved in cochlear development and found that expression patterns were generally conserved between rodents and primates. However, several proteins involved in supporting cell processes and neuronal development exhibited interspecific expression differences. Human fetal samples for studies of primate-specific cochlear development are extremely rare, especially for late developmental stages. Our results support the use of the common marmoset as an effective alternative for analyses of primate cochlear development.


Subject(s)
Callithrix/genetics , Cochlea/metabolism , Gene Expression Regulation, Developmental , Models, Animal , Organogenesis/genetics , Animals , Aquaporin 4/genetics , Aquaporin 4/metabolism , Calbindin 1/genetics , Calbindin 1/metabolism , Callithrix/embryology , Callithrix/growth & development , Callithrix/metabolism , Cochlea/anatomy & histology , Cochlea/cytology , Cochlea/growth & development , Conserved Sequence , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Embryo, Mammalian , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Myosin VIIa/genetics , Myosin VIIa/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Peripherins/genetics , Peripherins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Species Specificity , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Tubulin/genetics , Tubulin/metabolism
11.
Dev Biol ; 470: 121-135, 2021 02.
Article in English | MEDLINE | ID: mdl-33248112

ABSTRACT

Actin filament crosslinking, bundling and molecular motor proteins are necessary for the assembly of epithelial projections such as microvilli, stereocilia, hairs, and bristles. Mutations in such proteins cause defects in the shape, structure, and function of these actin - based protrusions. One protein necessary for stereocilia formation, Myosin VIIA, is an actin - based motor protein conserved throughout phylogeny. In Drosophila melanogaster, severe mutations in the MyoVIIA homolog crinkled (ck) are "semi - lethal" with only a very small percentage of flies surviving to adulthood. Such survivors show morphological defects related to actin bundling in hairs and bristles. To better understand ck/MyoVIIA's function in bundled - actin structures, we used dominant female sterile approaches to analyze the loss of maternal and zygotic (M/Z) ck/MyoVIIA in the morphogenesis of denticles, small actin - based projections on the ventral epidermis of Drosophila embryos. M/Z ck mutants displayed severe defects in denticle morphology - actin filaments initiated in the correct location, but failed to elongate and bundle to form normal projections. Using deletion mutant constructs, we demonstrated that both of the C - terminal MyTH4 and FERM domains are necessary for proper denticle formation. Furthermore, we show that ck/MyoVIIA interacts genetically with dusky - like (dyl), a member of the ZPD family of proteins that links the extracellular matrix to the plasma membrane, and when mutated also disrupts normal denticle formation. Loss of either protein alone does not alter the localization of the other; however, loss of the two proteins together dramatically enhances the defects in denticle shape observed when either protein alone was absent. Our data indicate that ck/MyoVIIA plays a key role in the formation and/or organization of actin filament bundles, which drive proper shape of cellular projections.


Subject(s)
Actin Cytoskeleton/ultrastructure , Cell Surface Extensions/ultrastructure , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Myosin VIIa/metabolism , Actin Cytoskeleton/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Epidermis/embryology , Female , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Morphogenesis , Mutant Proteins/metabolism , Mutation , Myosin VIIa/genetics
12.
Nat Commun ; 11(1): 2066, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32350269

ABSTRACT

Mutations in myosin-VIIa (MYO7A) cause Usher syndrome type 1, characterized by combined deafness and blindness. MYO7A is proposed to function as a motor that tensions the hair cell mechanotransduction (MET) complex, but conclusive evidence is lacking. Here we report that multiple MYO7A isoforms are expressed in the mouse cochlea. In mice with a specific deletion of the canonical isoform (Myo7a-ΔC mouse), MYO7A is severely diminished in inner hair cells (IHCs), while expression in outer hair cells is affected tonotopically. IHCs of Myo7a-ΔC mice undergo normal development, but exhibit reduced resting open probability and slowed onset of MET currents, consistent with MYO7A's proposed role in tensioning the tip link. Mature IHCs of Myo7a-ΔC mice degenerate over time, giving rise to progressive hearing loss. Taken together, our study reveals an unexpected isoform diversity of MYO7A expression in the cochlea and highlights MYO7A's essential role in tensioning the hair cell MET complex.


Subject(s)
Hair Cells, Auditory, Inner/metabolism , Mechanotransduction, Cellular , Myosin VIIa/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Gene Deletion , Hair Cells, Auditory, Inner/ultrastructure , Hearing Loss/metabolism , Hearing Loss/pathology , Mice, Inbred C57BL , Myosin VIIa/chemistry , Myosin VIIa/genetics , Protein Isoforms/metabolism , Protein Transport , Stereocilia/metabolism , Stereocilia/ultrastructure
13.
Hear Res ; 390: 107951, 2020 05.
Article in English | MEDLINE | ID: mdl-32244147

ABSTRACT

The retinoblastoma family of pocket proteins (pRBs), composed of Rb1, p107, and p130 are negative regulators of cell-cycle progression. The deletion of any individual pRB in the auditory system triggers hair cells' (HCs) and supporting cells' (SCs) proliferation to different extents. Nevertheless, accessing their combined role in the inner ear through conditional or complete knockout methods is limited by the early mortality of the triple knockout. In quiescent cells, hyperphosphorylation and inactivation of the pRBs are maintained through the activity of the Cyclin-D1-cdk4/6 complex. Cyclin D1 (CycD1) is expressed in the embryonic and neonatal inner ear. In the mature organ of Corti (OC), CycD1 expression is significantly downregulated, paralleling the OC mitotic quiescence. Earlier studies showed that CycD1 overexpression leads to cell-cycle reactivation in cultures of inner ear explants. Here, we characterize a Cre-activated, Doxycycline (Dox)-controlled, conditional CycD1 overexpression model, which when bred to a tetracycline-controlled transcriptional activator and the Atoh1-cre mouse lines, allow for transient CycD1 overexpression and pRBs' downregulation in the inner ear in a reversible fashion. Analyses of postnatal mice's inner ears at various time points revealed the presence of supernumerary cells throughout the length of the cochlea and in the vestibular end-organs. Notably, most supernumerary cells were observed in the inner hair cells' (IHCs) region, expressed myosin VIIa (M7a), and showed no signs of apoptosis at any of the time points analyzed. Auditory and vestibular phenotypes were similar between the different genotypes and treatment groups. The fact that no significant differences were observed in auditory and vestibular function supports the notion that the supernumerary cells detected in the adult mice cochlea and macular end-organs may not impair auditory functions.


Subject(s)
Cell Proliferation , Cyclin D1/metabolism , Ear, Inner/metabolism , Hair Cells, Auditory, Inner/metabolism , Mitosis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cyclin D1/genetics , Ear, Inner/cytology , Evoked Potentials, Auditory, Brain Stem , Female , Male , Mice, Transgenic , Myosin VIIa/metabolism , Otoacoustic Emissions, Spontaneous , Phosphorylation , Retinoblastoma Protein/metabolism , Signal Transduction , Time Factors , Up-Regulation , Vestibular Evoked Myogenic Potentials
14.
Anat Rec (Hoboken) ; 303(3): 451-460, 2020 03.
Article in English | MEDLINE | ID: mdl-31943808

ABSTRACT

The mammalian inner ear mediates hearing and balance and during development generates both cochleo-vestibular ganglion neurons and sensory epithelial receptor cells, that is, hair cells and support cells. Cell marking experiments have shown that both hair cells and support cells can originate from a common progenitor. Here, we demonstrate the lineage potential of individual otic epithelial cell clones using three cell lines established by a combination of limiting dilution and gene-marking techniques from an embryonic day 12 (E12) rat otocyst. Cell-type specific marker analyses of these clonal lines under proliferation and differentiation culture conditions demonstrate that during differentiation immature cell markers (Nanog and Nestin) were downregulated and hair cell (Myosin VIIa and Math1), support cell (p27Kip1 and cytokeratin) and neuronal cell (NF-H and NeuroD) markers were upregulated. Our results suggest that the otic epithelium of the E12 mammalian inner ear possess multipotent progenitor cells able to generate cell types of both sensory epithelial and neural cell lineages when cultured under a differentiation culture condition. Understanding the molecular mechanisms of proliferation and differentiation of multipotent otic progenitor cells may provide insights that could contribute to the development of a novel cell therapy with a potential to initiate or stimulate the sensorineural repair of damaged inner ear sensory receptors. Anat Rec, 303:451-460, 2020. © 2019 American Association for Anatomy.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Ear, Inner/cytology , Hair Cells, Auditory/cytology , Neurons/cytology , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Ear, Inner/embryology , Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Myosin VIIa/metabolism , Nanog Homeobox Protein/metabolism , Nestin/metabolism , Neurons/metabolism , Rats , Rats, Wistar , Stem Cells/metabolism
15.
Gene Expr Patterns ; 35: 119092, 2020 01.
Article in English | MEDLINE | ID: mdl-31918020

ABSTRACT

Myosins make up a large super family of motor proteins responsible for actin-based motility in most eukaryotic cells. Myosin VIIA is essential for the development and function of sensory hair cells in the inner ear. The role of Myosin VIIA in the development of cochleovestibular ganglion (CVG) neurons in the mouse is largely unknown. Neurons of the CVG innervate sensory hair cells of the cochlea and vestibular organs to transmit hearing and balance information respectively to the brain. The aim of this study was to characterize the expression of MYOSIN VIIA in the CVG of mouse embryos. Spatiotemporal expression of MYOSIN VIIA was characterized in embryonic (E) mouse inner ear neurons from E9.5 to postnatal (P) day 0. At early stages, when otic neurons begin to delaminate to form the CVG, MYOSIN VIIA was co-expressed with TuJ1, ISLET1 and NEUROD in the otic epithelium and CVG. When CVG neurons were migrating and exiting mitosis, MYSOSIN VIIA was downregulated in a subset of neurons, which were NEUROD-negative and GATA3-positive. After segregation of the CVG, MYOSIN VIIA was observed in a subset of vestibular neurons marked by TUJ1 and absent in cochlear neurons, marked by GATA3. The differential expression of MYOSIN VIIA may indicate a role in inner ear neuron migration and specific labeling of vestibular neurons.


Subject(s)
Myosin VIIa/genetics , Neurogenesis , Neurons/metabolism , Vestibulocochlear Nerve/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/embryology , Brain/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Inbred ICR , Myosin VIIa/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Tubulin/genetics , Tubulin/metabolism , Vestibulocochlear Nerve/embryology
16.
Cell Rep ; 29(4): 974-986.e4, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31644917

ABSTRACT

Hair cell stereocilia tip-links function to sense mechanical forces generated by sound waves and maintain the structure of stereocilia by rooting the tail of cadherins to highly dense structures known as tip-link densities. Although the molecular components are largely known, the mechanisms underlying the tip-link density formation are unknown. Here, we show that Myosin VIIB (MYO7B), USH1C, and ANKS4B, which form a specific complex stabilizing tip-links in intestine microvilli, could form dense condensates via liquid-liquid phase separation in vitro and in cells. The MYO7A, USH1C, and USH1G complex also undergoes phase separation in cells. Formation of the MYO7A/USH1C/USH1G and MYO7B/USH1C/ANKS4B condensates requires strong and multivalent interactions between proteins in both tripartite complexes. Point mutations of MYO7A found in Usher syndrome patients weaken or even disrupt the multivalent interactions of the MYO7A/USH1C/USH1G complex and impair its phase separation. Thus, the stereocilia tip-link densities may form via phase separation of the MYO7A/USH1C/USH1G complex.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Myosin VIIa/metabolism , Nerve Tissue Proteins/metabolism , Binding Sites , Carrier Proteins/chemistry , Cell Cycle Proteins/chemistry , Cytoskeletal Proteins/chemistry , HEK293 Cells , HeLa Cells , Humans , Mutation , Myosin VIIa/chemistry , Myosin VIIa/genetics , Nerve Tissue Proteins/chemistry , Protein Binding
17.
Climacteric ; 22(4): 361-369, 2019 08.
Article in English | MEDLINE | ID: mdl-30612476

ABSTRACT

Objective: This study aimed to test the hearing function and to investigate the effect of estrogen on prestin and MYO7A expression in hair cells of the cochlea in ovariectomized rats. Study design: We compared the hearing function systematically in normal female Sprague-Dawley rats and in ovariectomized rats with or without exogenous ß-estradiol administration by auditory brainstem response measurements and distortion product otoacoustic emissions. In addition, a correlation analysis between the functional parameters and cochlear histology was carried out. Results: There was a significant auditory threshold shift in the high-frequency range in the ovariectomized rats. Prestin and MYO7A expression was lowered in the cochlea of ovariectomized rats. These effects could be recovered by subcutaneous administration of ß-estradiol. Conclusion: Low estrogen levels may lead to reduced expression of prestin and MYO7A in cochlea, leading to a menopause-related hearing loss.


Subject(s)
Hearing Loss/metabolism , Menopause , Myosin VIIa/metabolism , Sulfate Transporters/metabolism , Animals , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Ovariectomy , Rats , Rats, Sprague-Dawley
18.
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
SELECTION OF CITATIONS
SEARCH DETAIL