Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.683
Filter
1.
Sci Rep ; 14(1): 15296, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961203

ABSTRACT

Blast wave exposure, a leading cause of hearing loss and balance dysfunction among military personnel, arises primarily from direct mechanical damage to the mechanosensory hair cells and supporting structures or indirectly through excessive oxidative stress. We previously reported that HK-2, an orally active, multifunctional redox modulator (MFRM), was highly effective in reducing both hearing loss and hair cells loss in rats exposed to a moderate intensity workday noise that likely damages the cochlea primarily from oxidative stress versus direct mechanical trauma. To determine if HK-2 could also protect cochlear and vestibular cells from damage caused primarily from direct blast-induced mechanical trauma versus oxidative stress, we exposed rats to six blasts of 186 dB peak SPL. The rats were divided into four groups: (B) blast alone, (BEP) blast plus earplugs, (BHK-2) blast plus HK-2 and (BEPHK-2) blast plus earplugs plus HK-2. HK-2 was orally administered at 50 mg/kg/d from 7-days before to 30-day after the blast exposure. Cochlear and vestibular tissues were harvested 60-d post-exposure and evaluated for loss of outer hair cells (OHC), inner hair cells (IHC), auditory nerve fibers (ANF), spiral ganglion neurons (SGN) and vestibular hair cells in the saccule, utricle and semicircular canals. In the untreated blast-exposed group (B), massive losses occurred to OHC, IHC, ANF, SGN and only the vestibular hair cells in the striola region of the saccule. In contrast, rats treated with HK-2 (BHK-2) sustained significantly less OHC (67%) and IHC (57%) loss compared to the B group. OHC and IHC losses were smallest in the BEPHK-2 group, but not significantly different from the BEP group indicating lack of protective synergy between EP and HK-2. There was no loss of ANF, SGN or saccular hair cells in the BHK-2, BEP and BEPHK-2 groups. Thus, HK-2 not only significantly reduced OHC and IHC damage, but completely prevented loss of ANF, SGN and saccule hair cells. The powerful protective effects of this oral MFRM make HK-2 an extremely promising candidate for human clinical trials.


Subject(s)
Blast Injuries , Hair Cells, Vestibular , Spiral Ganglion , Animals , Spiral Ganglion/drug effects , Spiral Ganglion/pathology , Rats , Blast Injuries/prevention & control , Hair Cells, Vestibular/drug effects , Hair Cells, Vestibular/metabolism , Male , Oxidation-Reduction , Rats, Sprague-Dawley , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Oxidative Stress/drug effects , Hearing Loss, Noise-Induced/prevention & control , Hearing Loss, Noise-Induced/pathology
2.
STAR Protoc ; 5(2): 103118, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38852155

ABSTRACT

The avian inner ear can naturally regenerate sensory hair cells and is therefore an ideal candidate for investigating mechanisms leading to hair cell regeneration and functional recovery. Here, we present a surgical protocol for eliminating auditory hair cells via sisomicin injection into the lateral semicircular canal. We describe steps for multiplex mRNA detection in chicken basilar papilla and utricle sections. We then detail procedures for integrating immunohistochemistry for concurrent mRNA and protein visualization, complemented by S-phase labeling with EdU. For complete details on the use and execution of this protocol, please refer to Benkafadar et al., Benkafadar et al., Sato et al., Janesick et al., Scheibinger et al.1,2,3,4,5.


Subject(s)
Chickens , Hair Cells, Auditory , Immunohistochemistry , RNA, Messenger , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/analysis , Immunohistochemistry/methods , Hair Cells, Auditory/metabolism
3.
J Otolaryngol Head Neck Surg ; 53: 19160216241258431, 2024.
Article in English | MEDLINE | ID: mdl-38888945

ABSTRACT

IMPORTANCE: Mesenchymal stem cells (MSCs) have the capability of providing ongoing paracrine support to degenerating tissues. Since MSCs can be extracted from a broad range of tissues, their specific surface marker profiles and growth factor secretions can be different. We hypothesized that MSCs derived from different sources might also have different neuroprotective potential. OBJECTIVE: In this study, we extracted MSCs from rodent olfactory mucosa and compared their neuroprotective effects on auditory hair cell survival with MSCs extracted from rodent adipose tissue. METHODS: Organ of Corti explants were dissected from 41 cochlea and incubated with olfactory mesenchymal stem cells (OMSCs) and adipose mesenchymal stem cells (AMSCs). After 72 hours, Corti explants were fixed, stained, and hair cells counted. Growth factor concentrations were determined in the supernatant and cell lysate using Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS: Co-culturing of organ of Corti explants with OMSCs resulted in a significant increase in inner and outer hair cell stereocilia survival, compared to control. Comparisons between both stem cell lines, showed that co-culturing with OMSCs resulted in superior inner and outer hair cell stereocilia survival rates over co-culturing with AMSCs. Assessment of growth factor secretions revealed that the OMSCs secrete significant amounts of insulin-like growth factor 1 (IGF-1). Co-culturing OMSCs with organ of Corti explants resulted in a 10-fold increase in IGF-1 level compared to control, and their secretion was 2 to 3 times higher compared to the AMSCs. CONCLUSIONS: This study has shown that OMSCs may mitigate auditory hair cell stereocilia degeneration. Their neuroprotective effects may, at least partially, be ascribed to their enhanced IGF-1 secretory abilities compared to AMSCs.


Subject(s)
Hair Cells, Auditory , Insulin-Like Growth Factor I , Mesenchymal Stem Cells , Animals , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Rats , Hair Cells, Auditory/metabolism , Olfactory Mucosa/cytology , Enzyme-Linked Immunosorbent Assay , Coculture Techniques , Cell Survival , Cells, Cultured , Adipose Tissue/cytology , Mesenchymal Stem Cell Transplantation/methods
4.
J Biomed Mater Res B Appl Biomater ; 112(7): e35439, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923766

ABSTRACT

Sensorineural hearing loss (SNHL) is mainly caused by injury or loss of hair cells (HCs) and associated spiral ganglion neurons (SGNs) in the inner ear. At present, there is still no effective treatment for SNHL in clinic. Recently, advances in organoid bring a promising prospect for research and treatment of SNHL. Meanwhile, three-dimensional (3D) printing provides a tremendous opportunity to construct versatile organoids for tissue engineering and regenerative medicine. In this study, gelatin (Gel), sodium alginate (SA), and polyvinyl alcohol (PVA) were used to fabricate biomimetic scaffold through 3D printing. The organ of Corti derived from neonatal mice inner ear was seeded on the PVA/Gel/SA scaffold to construct organ of Corti organoid. Then, the organ of Corti organoid was used to study the potential protective effects of berberine sulfate on neomycin-juried auditory HCs and SGNs. The results showed that the PVA/Gel/SA biomimetic 3D scaffolds had good cytocompatibilities and mechanical properties. The constructed organoid could maintain organ of Corti activity well in vitro. In addition, the injury intervention results showed that berberine sulfate could significantly inhibit neomycin-induced HC and SGN damage. This study suggests that the fabricated organoid is highly biomimetic to the organ of Corti, which may provide an effective model for drug development, cell and gene therapy for SNHL.


Subject(s)
Berberine , Organ of Corti , Tissue Scaffolds , Animals , Organ of Corti/drug effects , Mice , Berberine/pharmacology , Berberine/chemistry , Tissue Scaffolds/chemistry , Organoids/metabolism , Organoids/drug effects , Printing, Three-Dimensional , Alginates/chemistry , Alginates/pharmacology , Gelatin/chemistry , Gelatin/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Tissue Engineering , Polyvinyl Alcohol/chemistry , Polyvinyl Alcohol/pharmacology , Hearing Loss, Sensorineural , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism
5.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727276

ABSTRACT

In mammals, hearing loss is irreversible due to the lack of the regenerative capacity of the auditory epithelium. However, stem/progenitor cells in mammalian cochleae may be a therapeutic target for hearing regeneration. The ubiquitin proteasome system plays an important role in cochlear development and maintenance. In this study, we investigated the role of ubiquitin C-terminal hydrolase L1 (UCHL1) in the process of the transdifferentiation of auditory supporting cells (SCs) into hair cells (HCs). The expression of UCHL1 gradually decreased as HCs developed and was restricted to inner pillar cells and third-row Deiters' cells between P2 and P7, suggesting that UCHL1-expressing cells are similar to the cells with Lgr5-positive progenitors. UCHL1 expression was decreased even under conditions in which supernumerary HCs were generated with a γ-secretase inhibitor and Wnt agonist. Moreover, the inhibition of UCHL1 by LDN-57444 led to an increase in HC numbers. Mechanistically, LDN-57444 increased mTOR complex 1 activity and allowed SCs to transdifferentiate into HCs. The suppression of UCHL1 induces the transdifferentiation of auditory SCs and progenitors into HCs by regulating the mTOR pathway.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , Signal Transduction , TOR Serine-Threonine Kinases , Ubiquitin Thiolesterase , Animals , Cell Transdifferentiation/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Indoles , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/cytology , Oximes , TOR Serine-Threonine Kinases/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Rats
6.
Stem Cells Transl Med ; 13(7): 661-677, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38709826

ABSTRACT

Loss of cochlear hair cells (HCs) leads to permanent hearing loss in mammals, and regenerative medicine is regarded as an ideal strategy for hearing recovery. Limited genetic and pharmaceutical approaches for HC regeneration have been established, and the existing strategies cannot achieve recovery of auditory function. A promising target to promote HC regeneration is MEK/ERK signaling because dynamic shifts in its activity during the critical stages of inner ear development have been observed. Here, we first showed that MEK/ERK signaling is activated specifically in supporting cells (SCs) after aminoglycoside-induced HC injury. We then selected 4 MEK/ERK signaling inhibitors, and PD0325901 (PD03) was found to induce the transdifferentiation of functional supernumerary HCs from SCs in the neonatal mammalian cochlear epithelium. We next found that PD03 facilitated the generation of HCs in inner ear organoids. Through genome-wide high-throughput RNA sequencing and verification, we found that the Notch pathway is the downstream target of MEK/ERK signaling. Importantly, delivery of PD03 into the inner ear induced mild HC regeneration in vivo. Our study thus reveals the importance of MEK/ERK signaling in cell fate determination and suggests that PD03 might serve as a new approach for HC regeneration.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , MAP Kinase Signaling System , Receptors, Notch , Animals , Cell Transdifferentiation/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , MAP Kinase Signaling System/drug effects , Mice , Receptors, Notch/metabolism , Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Labyrinth Supporting Cells/metabolism
7.
Hear Res ; 447: 109021, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703432

ABSTRACT

Understanding the complex pathologies associated with hearing loss is a significant motivation for conducting inner ear research. Lifelong exposure to loud noise, ototoxic drugs, genetic diversity, sex, and aging collectively contribute to human hearing loss. Replicating this pathology in research animals is challenging because hearing impairment has varied causes and different manifestations. A central aspect, however, is the loss of sensory hair cells and the inability of the mammalian cochlea to replace them. Researching therapeutic strategies to rekindle regenerative cochlear capacity, therefore, requires the generation of animal models in which cochlear hair cells are eliminated. This review discusses different approaches to ablate cochlear hair cells in adult mice. We inventoried the cochlear cyto- and histo-pathology caused by acoustic overstimulation, systemic and locally applied drugs, and various genetic tools. The focus is not to prescribe a perfect damage model but to highlight the limitations and advantages of existing approaches and identify areas for further refinement of damage models for use in regenerative studies.


Subject(s)
Cochlea , Disease Models, Animal , Hair Cells, Auditory , Regeneration , Animals , Hair Cells, Auditory/pathology , Hair Cells, Auditory/metabolism , Mice , Cochlea/pathology , Cochlea/physiopathology , Humans , Hearing , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Noise-Induced/pathology , Hearing Loss/pathology , Hearing Loss/physiopathology , Acoustic Stimulation
8.
Hear Res ; 448: 109035, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763033

ABSTRACT

The sensory epithelia of the auditory and vestibular systems of vertebrates have shared developmental and evolutionary histories. However, while the auditory epithelia show great variation across vertebrates, the vestibular sensory epithelia appear seemingly more conserved. An exploration of the current knowledge of the comparative biology of the amniote utricle, a vestibular sensory epithelium that senses linear acceleration, shows interesting instances of variability between birds and mammals. The distribution of sensory hair cell types, the position of the line of hair bundle polarity reversal and the properties of supporting cells show marked differences, likely impacting vestibular function and hair cell regeneration potential.


Subject(s)
Saccule and Utricle , Animals , Saccule and Utricle/physiology , Biological Evolution , Humans , Birds/physiology , Mammals/physiology , Hair Cells, Vestibular/physiology , Vestibule, Labyrinth/physiology , Hair Cells, Auditory/physiology , Species Specificity , Regeneration
9.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38804528

ABSTRACT

The planar polarized organization of hair cells in the vestibular maculae is unique because these sensory organs contain two groups of cells with oppositely oriented stereociliary bundles that meet at a line of polarity reversal (LPR). EMX2 is a transcription factor expressed by one hair cell group that reverses the orientation of their bundles, thereby forming the LPR. We generated Emx2-CreERt2 transgenic mice for genetic lineage tracing and demonstrate Emx2 expression before hair cell specification when the nascent utricle and saccule constitute a continuous prosensory domain. Precursors labeled by Emx2-CreERt2 at this stage give rise to hair cells located along one side of the LPR in the mature utricle or saccule, indicating that this boundary is first established in the prosensory domain. Consistent with this, Emx2-CreERt2 lineage tracing in Dreher mutants, where the utricle and saccule fail to segregate, labels a continuous field of cells along one side of a fused utriculo-saccular-cochlear organ. These observations reveal that LPR positioning is pre-determined in the developing prosensory domain, and that EMX2 expression defines lineages of hair cells with oppositely oriented stereociliary bundles.


Subject(s)
Cell Lineage , Cell Polarity , Ear, Inner , Homeodomain Proteins , Mice, Transgenic , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Cell Lineage/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Cell Polarity/genetics , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology
10.
Hear Res ; 447: 109013, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718672

ABSTRACT

Cisplatin, a highly effective chemotherapeutic drug for various human cancers, induces irreversible sensorineural hearing loss as a side effect. Currently there are no highly effective clinical strategies for the prevention of cisplatin-induced ototoxicity. Previous studies have indicated that short-term cisplatin ototoxicity primarily affects the outer hair cells of the cochlea. Therefore, preventing the entry of cisplatin into hair cells may be a promising strategy to prevent cisplatin ototoxicity. This study aimed to investigate the entry route of cisplatin into mouse cochlear hair cells. The competitive inhibitor of organic cation transporter 2 (OCT2), cimetidine, and the sensory mechanoelectrical transduction (MET) channel blocker benzamil, demonstrated a protective effect against cisplatin toxicity in hair cells in cochlear explants. Sensory MET-deficient hair cells explanted from Tmc1Δ;Tmc2Δ mice were resistant to cisplatin toxicity. Cimetidine showed an additive protective effect against cisplatin toxicity in sensory MET-deficient hair cells. However, in the apical turn, cimetidine, benzamil, or genetic ablation of sensory MET channels showed limited protective effects, implying the presence of other entry routes for cisplatin to enter the hair cells in the apical turn. Systemic administration of cimetidine failed to protect cochlear hair cells from ototoxicity caused by systemically administered cisplatin. Notably, outer hair cells in MET-deficient mice exhibited no apparent deterioration after systemic administration of cisplatin, whereas the outer hair cells in wild-type mice showed remarkable deterioration. The susceptibility of mouse cochlear hair cells to cisplatin ototoxicity largely depends on the sensory MET channel both ex vivo and in vivo. This result justifies the development of new pharmaceuticals, such as a specific antagonists for sensory MET channels or custom-designed cisplatin analogs which are impermeable to sensory MET channels.


Subject(s)
Antineoplastic Agents , Cimetidine , Cisplatin , Mechanotransduction, Cellular , Organic Cation Transporter 2 , Ototoxicity , Cisplatin/toxicity , Animals , Ototoxicity/prevention & control , Ototoxicity/metabolism , Ototoxicity/physiopathology , Mechanotransduction, Cellular/drug effects , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/antagonists & inhibitors , Cimetidine/pharmacology , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/metabolism , Mice, Inbred C57BL , Mice , Membrane Proteins
11.
Hear Res ; 447: 109024, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735179

ABSTRACT

Delayed loss of residual acoustic hearing after cochlear implantation is a common but poorly understood phenomenon due to the scarcity of relevant temporal bone tissues. Prior histopathological analysis of one case of post-implantation hearing loss suggested there were no interaural differences in hair cell or neural degeneration to explain the profound loss of low-frequency hearing on the implanted side (Quesnel et al., 2016) and attributed the threshold elevation to neo-ossification and fibrosis around the implant. Here we re-evaluated the histopathology in this case, applying immunostaining and improved microscopic techniques for differentiating surviving hair cells from supporting cells. The new analysis revealed dramatic interaural differences, with a > 80 % loss of inner hair cells in the cochlear apex on the implanted side, which can account for the post-implantation loss of residual hearing. Apical degeneration of the stria further contributed to threshold elevation on the implanted side. In contrast, spiral ganglion cell survival was reduced in the region of the electrode on the implanted side, but apical counts in the two ears were similar to that seen in age-matched unimplanted control ears. Almost none of the surviving auditory neurons retained peripheral axons throughout the basal half of the cochlea. Relevance to cochlear implant performance is discussed.


Subject(s)
Auditory Threshold , Cochlear Implantation , Cochlear Implants , Spiral Ganglion , Cochlear Implantation/instrumentation , Cochlear Implantation/adverse effects , Humans , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology , Hair Cells, Auditory, Inner/pathology , Time Factors , Cell Survival , Male , Hearing , Hearing Loss/physiopathology , Hearing Loss/pathology , Hearing Loss/surgery , Hearing Loss/etiology , Female , Hair Cells, Auditory/pathology , Aged , Nerve Degeneration , Middle Aged , Temporal Bone/pathology , Temporal Bone/surgery
13.
Sci Rep ; 14(1): 10910, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740884

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling plays a significant role in multiple biological processes, including inflammation, immunity, and cell death. However, its specific impact on the cochlea remains unclear. In this study, we aimed to investigate the effects of TGF-ß signaling suppression on auditory function and cochlear pathology in mice with kanamycin-induced ototoxicity. Kanamycin and furosemide (KM-FS) were systemically administered to 8-week-old C57/BL6 mice, followed by immediate topical application of a TGF-ß receptor inhibitor (TGF-ßRI) onto the round window membrane. Results showed significant TGF-ß receptor upregulation in spiral ganglion neurons (SGNs) after KM-FA ototoxicity, whereas expression levels in the TGF-ßRI treated group remained unchanged. Interestingly, despite no significant change in cochlear TGF-ß expression after KM-FS ototoxicity, TGF-ßRI treatment resulted in a significant decrease in TGF-ß signaling. Regarding auditory function, TGF-ßRI treatment offered no therapeutic effects on hearing thresholds and hair cell survival following KM-FS ototoxicity. However, SGN loss and macrophage infiltration were significantly increased with TGF-ßRI treatment. These results imply that inhibition of TGF-ß signaling after KM-FS ototoxicity promotes cochlear inflammation and SGN degeneration.


Subject(s)
Kanamycin , Mice, Inbred C57BL , Ototoxicity , Signal Transduction , Spiral Ganglion , Transforming Growth Factor beta , Animals , Kanamycin/toxicity , Signal Transduction/drug effects , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Transforming Growth Factor beta/metabolism , Mice , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Cochlea/metabolism , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Furosemide/pharmacology , Male
14.
Toxicol Appl Pharmacol ; 486: 116947, 2024 May.
Article in English | MEDLINE | ID: mdl-38688426

ABSTRACT

AIMS: SERCA2, one of the P-type pumps encoded by gene ATP2A2, is the only calcium reflux channel of the endoplasmic reticulum (ER) and participates in maintaining calcium homeostasis. The present study was designed to explore SERCA2 expression pattern in auditory hair cells and the possible mechanism underlying the effects of SERCA2 on cisplatin-induced ototoxicity. MAIN METHODS: The SERCA2 expression pattern in cochlea hair cells and HEI-OC1 cells was measured by Western blot (WB) and immunofluorescence staining. The apoptosis and its related factors were detected by TUNEL assay and WB. The expression levels of ER stress-related factors, ATF6, PERK, IRE1α, and GRP78, were measured via WB. As for the determination of SERCA2 overexpression and knockdown, plasmids and lentiviral vectors were constructed, respectively. KEY FINDINGS: We found that SERCA2 was highly expressed in cochlea hair cells and HEI-OC1 cells. Of note, the level of SERCA2 expression in neonatal mice was remarkably higher than that in adult mice. Under the exposure of 30 µM cisplatin, SERCA2 was down-regulated significantly compared with the control group. In addition, cisplatin administration triggered the occurrence of ER stress and apoptosis. Those events were reversed by overexpressing SERCA2. On the contrary, SERCA2 knockdown could aggravate the above processes. SIGNIFICANCE: The findings from the present study disclose, for the first time, that SERCA2 is abundantly expressed in cochlea hair cells, and the suppression of SERCA2 caused by cisplatin could trigger ER homeostasis disruption, thereby implying that SERCA2 might be a promising target to prevent cisplatin-induced cytotoxicity of hair cells.


Subject(s)
Apoptosis , Cisplatin , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Hair Cells, Auditory , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Cisplatin/toxicity , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Endoplasmic Reticulum Stress/drug effects , Mice , Apoptosis/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Cell Line , Antineoplastic Agents/toxicity , Male , Ototoxicity/prevention & control
15.
Sci Rep ; 14(1): 7862, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570547

ABSTRACT

The small muscle protein, X-linked (SMPX) gene encodes a cytoskeleton-associated protein, highly expressed in the inner ear hair cells (HCs), possibly regulating auditory function. In the last decade, several mutations in SMPX have been associated with X-chromosomal progressive non syndromic hearing loss in humans and, in line with this, Smpx-deficient animal models, namely zebrafish and mouse, showed significant impairment of inner ear HCs development, maintenance, and functioning. In this work, we uncovered smpx expression in the neuromast mechanosensory HCs of both Anterior and Posterior Lateral Line (ALL and PLL, respectively) of zebrafish larvae and focused our attention on the PLL. Smpx was subcellularly localized throughout the cytoplasm of the HCs, as well as in their primary cilium. Loss-of-function experiments, via both morpholino-mediated gene knockdown and CRISPR/Cas9 F0 gene knockout, revealed that the lack of Smpx led to fewer properly differentiated and functional neuromasts, as well as to a smaller PLL primordium (PLLp), the latter also Smpx-positive. In addition, the kinocilia of Smpx-deficient neuromast HCs appeared structurally and numerically altered. Such phenotypes were associated with a significant reduction in the mechanotransduction activity of the neuromast HCs, in line with their positivity for Smpx. In summary, this work highlights the importance of Smpx in lateral line development and, specifically, in proper HCs differentiation and/or maintenance, and in the mechanotransduction process carried out by the neuromast HCs. Because lateral line HCs are both functionally and structurally analogous to the cochlear HCs, the neuromasts might represent an invaluable-and easily accessible-tool to dissect the role of Smpx in HCs development/functioning and shed light on the underlying mechanisms involved in hearing loss.


Subject(s)
Hearing Loss , Lateral Line System , Humans , Animals , Mice , Zebrafish/genetics , Zebrafish/metabolism , Lateral Line System/metabolism , Mechanotransduction, Cellular , Hair Cells, Auditory/metabolism , Hearing Loss/genetics , Muscle Proteins/metabolism
16.
Environ Toxicol ; 39(7): 4058-4065, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38661261

ABSTRACT

Platinum-based antineoplastic drugs, including cisplatin, carboplatin, and oxaliplatin, are widely used in the treatment of various cancers. Ototoxicity is a common adverse effect of platinum-based drugs. Ototoxicity leads to irreversible hearing impairment. We hypothesize that different platinum-based drugs exhibit varying ototoxic concentrations, time effects, and ototoxic mechanisms. We tested this hypothesis by using a zebrafish model (pvalb3b: TagGFP) to assess the viability of hair cells collected from zebrafish larvae. Cisplatin, carboplatin, and oxaliplatin were administered at dosages of 100, 200, or 400 µM, and the ototoxic effects of these drugs were assessed 1, 2, or 3 h after administration. Fm4-64 and a TUNEL assay were used to label the membranes of living hair cells and to detect cell apoptosis, respectively. We observed that >50% of hair cells were damaged at 1 h after cisplatin (100 µM) exposure, and this ototoxic effect increased at higher dosages and over time. Owing to the smaller ototoxic effects of carboplatin and oxaliplatin, we conducted higher-strength and longer-duration experiments with these drugs. Neither carboplatin nor oxaliplatin was obviously ototoxic, even at 1600 µM and after 6 h. Moreover, only cisplatin damaged the membranes of the hair cells. Cell apoptosis and significantly increased antioxidant gene expression were observed in only the cisplatin group. In conclusion, cisplatin significantly damages sensory hair cells and has notable dosage and time effects. Carboplatin and oxaliplatin are less ototoxic than cisplatin, likely due to having different ototoxic mechanisms than cisplatin.


Subject(s)
Antineoplastic Agents , Apoptosis , Carboplatin , Cisplatin , Ototoxicity , Oxaliplatin , Zebrafish , Animals , Cisplatin/toxicity , Oxaliplatin/toxicity , Carboplatin/toxicity , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Hair Cells, Auditory/drug effects , Larva/drug effects
17.
J Assoc Res Otolaryngol ; 25(3): 259-275, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38622383

ABSTRACT

PURPOSE: Cisplatin is a low-cost clinical anti-tumor drug widely used to treat solid tumors. However, its use could damage cochlear hair cells, leading to irreversible hearing loss. Currently, there appears one drug approved in clinic only used for reducing ototoxicity associated with cisplatin in pediatric patients, which needs to further explore other candidate drugs. METHODS: Here, by screening 1967 FDA-approved drugs to protect cochlear hair cell line (HEI-OC1) from cisplatin damage, we found that Tedizolid Phosphate (Ted), a drug indicated for the treatment of acute infections, had the best protective effect. Further, we evaluated the protective effect of Ted against ototoxicity in mouse cochlear explants, zebrafish, and adult mice. The mechanism of action of Ted was further explored using RNA sequencing analysis and verified. Meanwhile, we also observed the effect of Ted on the anti-tumor effect of cisplatin. RESULTS: Ted had a strong protective effect on hair cell (HC) loss induced by cisplatin in zebrafish and mouse cochlear explants. In addition, when administered systemically, it protected mice from cisplatin-induced hearing loss. Moreover, antitumor studies showed that Ted had no effect on the antitumor activity of cisplatin both in vitro and in vivo. RNA sequencing analysis showed that the otoprotective effect of Ted was mainly achieved by inhibiting phosphorylation of ERK. Consistently, ERK activator aggravated the damage of cisplatin to HCs. CONCLUSION: Collectively, these results showed that FDA-approved Ted protected HCs from cisplatin-induced HC loss by inhibiting ERK phosphorylation, indicating its potential as a candidate for preventing cisplatin ototoxicity in clinical settings.


Subject(s)
Antineoplastic Agents , Cisplatin , Hearing Loss , Organophosphates , Oxazoles , Zebrafish , Animals , Cisplatin/toxicity , Cisplatin/adverse effects , Mice , Hearing Loss/prevention & control , Hearing Loss/chemically induced , Oxazoles/pharmacology , Organophosphates/toxicity , Antineoplastic Agents/toxicity , United States Food and Drug Administration , Drug Approval , Hair Cells, Auditory/drug effects , United States , Ototoxicity/prevention & control , Ototoxicity/etiology , Humans
18.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682291

ABSTRACT

The planar polarized organization of hair cells in the vestibular maculae is unique because these sensory organs contain two groups of cells with oppositely oriented stereociliary bundles that meet at a line of polarity reversal (LPR). EMX2 is a transcription factor expressed by one hair cell group that reverses the orientation of their bundles, thereby forming the LPR. We generated Emx2-CreERt2 transgenic mice for genetic lineage tracing and demonstrate Emx2 expression before hair cell specification when the nascent utricle and saccule constitute a continuous prosensory domain. Precursors labeled by Emx2-CreERt2 at this stage give rise to hair cells located along one side of the LPR in the mature utricle or saccule, indicating that this boundary is first established in the prosensory domain. Consistent with this, Emx2-CreERt2 lineage tracing in Dreher mutants, where the utricle and saccule fail to segregate, labels a continuous field of cells along one side of a fused utriculo-saccular-cochlear organ. These observations reveal that LPR positioning is pre-determined in the developing prosensory domain, and that EMX2 expression defines lineages of hair cells with oppositely oriented stereociliary bundles.


Subject(s)
Cell Lineage , Cell Polarity , Ear, Inner , Homeodomain Proteins , Mice, Transgenic , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Cell Lineage/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Cell Polarity/genetics , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology
19.
Hear Res ; 446: 109006, 2024 May.
Article in English | MEDLINE | ID: mdl-38583350

ABSTRACT

Hair cells in the cochlear sensory epithelia serve as mechanosensory receptors, converting sound into neuronal signals. The basal sensory epithelia are responsible for transducing high-frequency sounds, while the apex handles low-frequency sounds. Age-related hearing loss predominantly affects hearing at high frequencies and is indicative of damage to the basal sensory epithelia. However, the precise mechanism underlying this site-selective injury remains unclear. In this study, we employed a microscale proteomics approach to examine and compare protein expression in different regions of the cochlear sensory epithelia (upper half and lower half) in 1.5-month-old (normal hearing) and 6-month-old (severe high-frequency hearing loss without hair cell loss) C57BL/6J mice. A total of 2,386 proteins were detected, and no significant differences in protein expression were detected in the upper half of the cochlear sensory epithelia between the two age groups. The expression of 20 proteins in the lower half of the cochlear sensory epithelia significantly differed between the two age groups (e.g., MATN1, MATN4, and AQP1). Moreover, there were 311 and 226 differentially expressed proteins between the upper and lower halves of the cochlear sensory epithelia in 1.5-month-old and 6-month-old mice, respectively. The expression levels of selected proteins were validated by Western blotting. These findings suggest that the spatial differences in protein expression within the cochlear sensory epithelia may play a role in determining the susceptibility of cells at different sites of the cochlea to age-related damage.


Subject(s)
Cochlea , Mice, Inbred C57BL , Presbycusis , Proteomics , Animals , Cochlea/metabolism , Cochlea/pathology , Presbycusis/metabolism , Presbycusis/pathology , Presbycusis/physiopathology , Presbycusis/genetics , Age Factors , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Aging/metabolism , Aging/pathology , Disease Models, Animal , Hearing , Epithelium/metabolism , Male , Mice
20.
Sci Data ; 11(1): 416, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653806

ABSTRACT

Our sense of hearing is mediated by cochlear hair cells, of which there are two types organized in one row of inner hair cells and three rows of outer hair cells. Each cochlea contains 5-15 thousand terminally differentiated hair cells, and their survival is essential for hearing as they do not regenerate after insult. It is often desirable in hearing research to quantify the number of hair cells within cochlear samples, in both pathological conditions, and in response to treatment. Machine learning can be used to automate the quantification process but requires a vast and diverse dataset for effective training. In this study, we present a large collection of annotated cochlear hair-cell datasets, labeled with commonly used hair-cell markers and imaged using various fluorescence microscopy techniques. The collection includes samples from mouse, rat, guinea pig, pig, primate, and human cochlear tissue, from normal conditions and following in-vivo and in-vitro ototoxic drug application. The dataset includes over 107,000 hair cells which have been identified and annotated as either inner or outer hair cells. This dataset is the result of a collaborative effort from multiple laboratories and has been carefully curated to represent a variety of imaging techniques. With suggested usage parameters and a well-described annotation procedure, this collection can facilitate the development of generalizable cochlear hair-cell detection models or serve as a starting point for fine-tuning models for other analysis tasks. By providing this dataset, we aim to give other hearing research groups the opportunity to develop their own tools with which to analyze cochlear imaging data more fully, accurately, and with greater ease.


Subject(s)
Cochlea , Animals , Mice , Guinea Pigs , Humans , Rats , Swine , Hair Cells, Auditory , Microscopy, Fluorescence , Machine Learning
SELECTION OF CITATIONS
SEARCH DETAIL
...