Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.433
Filtrar
1.
Cells ; 13(9)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38727276

RESUMEN

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.


Asunto(s)
Transdiferenciación Celular , Células Ciliadas Auditivas , Transducción de Señal , Serina-Treonina Quinasas TOR , Ubiquitina Tiolesterasa , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Transdiferenciación Celular/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/citología , Ratones , Células Laberínticas de Soporte/metabolismo , Células Laberínticas de Soporte/citología , Indoles , Oximas
2.
Sci Rep ; 14(1): 10910, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740884

RESUMEN

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.


Asunto(s)
Kanamicina , Ratones Endogámicos C57BL , Ototoxicidad , Transducción de Señal , Ganglio Espiral de la Cóclea , Factor de Crecimiento Transformador beta , Animales , Kanamicina/toxicidad , Transducción de Señal/efectos de los fármacos , Ototoxicidad/etiología , Ototoxicidad/metabolismo , Ototoxicidad/patología , Factor de Crecimiento Transformador beta/metabolismo , Ratones , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/patología , Cóclea/metabolismo , Cóclea/efectos de los fármacos , Cóclea/patología , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Furosemida/farmacología , Masculino
3.
Redox Rep ; 29(1): 2341470, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38629504

RESUMEN

Cisplatin is widely employed in clinical oncology as an anticancer chemotherapy drug in clinical practice and is known for its severe ototoxic side effects. Prior research indicates that the accumulation of reactive oxygen species (ROS) plays a pivotal role in cisplatin's inner ear toxicity. Hesperidin is a flavanone glycoside extracted from citrus fruits that has anti-inflammatory and antioxidant effects. Nonetheless, the specific pharmacological actions of hesperidin in alleviating cisplatin-induced ototoxicity remain elusive. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a critical mediator of the cellular oxidative stress response, is influenced by hesperidin. Activation of Nrf2 was shown to have a protective effect against cisplatin-induced ototoxicity. The potential of hesperidin to stimulate Nrf2 in attenuating cisplatin's adverse effects on the inner ear warrants further investigation. This study employs both in vivo and in vitro models of cisplatin ototoxicity to explore this possibility. Our results reveal that hesperidin mitigates cisplatin-induced ototoxicity by activating the Nrf2/NQO1 pathway in sensory hair cells, thereby reducing ROS accumulation, preventing hair cell apoptosis, and alleviating hearing loss.


Asunto(s)
Antineoplásicos , Hesperidina , Ototoxicidad , Humanos , Cisplatino/toxicidad , Hesperidina/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Ototoxicidad/tratamiento farmacológico , Ototoxicidad/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Línea Celular , Antineoplásicos/toxicidad , Células Ciliadas Auditivas/metabolismo , Apoptosis
4.
Hear Res ; 446: 109006, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583350

RESUMEN

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.


Asunto(s)
Cóclea , Ratones Endogámicos C57BL , Presbiacusia , Proteómica , Animales , Cóclea/metabolismo , Cóclea/patología , Presbiacusia/metabolismo , Presbiacusia/patología , Presbiacusia/fisiopatología , Presbiacusia/genética , Factores de Edad , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Modelos Animales de Enfermedad , Audición , Epitelio/metabolismo , Masculino , Ratones
5.
Elife ; 122024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38651641

RESUMEN

Inhibitory G alpha (GNAI or Gαi) proteins are critical for the polarized morphogenesis of sensory hair cells and for hearing. The extent and nature of their actual contributions remains unclear, however, as previous studies did not investigate all GNAI proteins and included non-physiological approaches. Pertussis toxin can downregulate functionally redundant GNAI1, GNAI2, GNAI3, and GNAO proteins, but may also induce unrelated defects. Here, we directly and systematically determine the role(s) of each individual GNAI protein in mouse auditory hair cells. GNAI2 and GNAI3 are similarly polarized at the hair cell apex with their binding partner G protein signaling modulator 2 (GPSM2), whereas GNAI1 and GNAO are not detected. In Gnai3 mutants, GNAI2 progressively fails to fully occupy the sub-cellular compartments where GNAI3 is missing. In contrast, GNAI3 can fully compensate for the loss of GNAI2 and is essential for hair bundle morphogenesis and auditory function. Simultaneous inactivation of Gnai2 and Gnai3 recapitulates for the first time two distinct types of defects only observed so far with pertussis toxin: (1) a delay or failure of the basal body to migrate off-center in prospective hair cells, and (2) a reversal in the orientation of some hair cell types. We conclude that GNAI proteins are critical for hair cells to break planar symmetry and to orient properly before GNAI2/3 regulate hair bundle morphogenesis with GPSM2.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go , Células Ciliadas Auditivas , Morfogénesis , Animales , Ratones , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/fisiología , Polaridad Celular , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética
6.
Sci Rep ; 14(1): 7862, 2024 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570547

RESUMEN

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.


Asunto(s)
Pérdida Auditiva , Sistema de la Línea Lateral , Humanos , Animales , Ratones , Pez Cebra/genética , Pez Cebra/metabolismo , Sistema de la Línea Lateral/metabolismo , Mecanotransducción Celular , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/genética , Proteínas Musculares/metabolismo
7.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38673858

RESUMEN

Hearing loss represents a multifaceted and pervasive challenge that deeply impacts various aspects of an individual's life, spanning psychological, emotional, social, and economic realms. Understanding the molecular underpinnings that orchestrate hearing loss remains paramount in the quest for effective therapeutic strategies. This review aims to expound upon the physiological, biochemical, and molecular aspects of hearing loss, with a specific focus on its correlation with diabetes. Within this context, phytochemicals have surfaced as prospective contenders in the pursuit of potential adjuvant therapies. These compounds exhibit noteworthy antioxidant and anti-inflammatory properties, which hold the potential to counteract the detrimental effects induced by oxidative stress and inflammation-prominent contributors to hearing impairment. Furthermore, this review offers an up-to-date exploration of the diverse molecular pathways modulated by these compounds. However, the dynamic landscape of their efficacy warrants recognition as an ongoing investigative topic, inherently contingent upon specific experimental models. Ultimately, to ascertain the genuine potential of phytochemicals as agents in hearing loss treatment, a comprehensive grasp of the molecular mechanisms at play, coupled with rigorous clinical investigations, stands as an imperative quest.


Asunto(s)
Antioxidantes , Células Ciliadas Auditivas , Pérdida Auditiva Sensorineural , Estrés Oxidativo , Fitoquímicos , Estrés Oxidativo/efectos de los fármacos , Humanos , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Pérdida Auditiva Sensorineural/metabolismo , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Muerte Celular/efectos de los fármacos , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
8.
Sci Rep ; 14(1): 6670, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509148

RESUMEN

Age-related hearing loss (ARHL) is a debilitating disorder for millions worldwide. While there are multiple underlying causes of ARHL, one common factor is loss of sensory hair cells. In mammals, new hair cells are not produced postnatally and do not regenerate after damage, leading to permanent hearing impairment. By contrast, fish produce hair cells throughout life and robustly regenerate these cells after toxic insult. Despite these regenerative abilities, zebrafish show features of ARHL. Here, we show that aged zebrafish of both sexes exhibited significant hair cell loss and decreased cell proliferation in all inner ear epithelia (saccule, lagena, utricle). Ears from aged zebrafish had increased expression of pro-inflammatory genes and significantly more macrophages than ears from young adult animals. Aged zebrafish also had fewer lateral line hair cells and less cell proliferation than young animals, although lateral line hair cells still robustly regenerated following damage. Unlike zebrafish, African turquoise killifish (an emerging aging model) only showed hair cell loss in the saccule of aged males, but both sexes exhibit age-related changes in the lateral line. Our work demonstrates that zebrafish exhibit key features of auditory aging, including hair cell loss and increased inflammation. Further, our finding that aged zebrafish have fewer lateral line hair cells yet retain regenerative capacity, suggests a decoupling of homeostatic hair cell addition from regeneration following acute trauma. Finally, zebrafish and killifish show species-specific strategies for lateral line homeostasis that may inform further comparative research on aging in mechanosensory systems.


Asunto(s)
Oído Interno , Peces Killi , Sistema de la Línea Lateral , Perciformes , Animales , Masculino , Femenino , Pez Cebra/genética , Células Ciliadas Auditivas/metabolismo , Regeneración/genética , Mamíferos
9.
Biochem Pharmacol ; 222: 116115, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38460910

RESUMEN

In recent years, extensive research has been conducted on the pathogenesis of sensorineural hearing loss (SNHL). Apoptosis and necrosis have been identified to play important roles in hearing loss, but they cannot account for all hearing loss. Autophagy, a cellular process responsible for cell self-degradation and reutilization, has emerged as a significant factor contributing to hearing loss, particularly in cases of autophagy deficiency. Autophagy plays a crucial role in maintaining cell health by exerting cytoprotective and metabolically homeostatic effects in organisms. Consequently, modulating autophagy levels can profoundly impact the survival, death, and regeneration of cells in the inner ear, including hair cells (HCs) and spiral ganglion neurons (SGNs). Abnormal mitochondrial autophagy has been demonstrated in animal models of SNHL. These findings indicate the profound significance of comprehending autophagy while suggesting that our perspective on this cellular process holds promise for advancing the treatment of SNHL. Thus, this review aims to clarify the pathogenic mechanisms of SNHL and the role of autophagy in the developmental processes of various cochlear structures, including the greater epithelial ridge (GER), SGNs, and the ribbon synapse. The pathogenic mechanisms of age-related hearing loss (ARHL), also known as presbycusis, and the latest research on autophagy are also discussed. Furthermore, we underscore recent findings on the modulation of autophagy in SNHL induced by ototoxic drugs. Additionally, we suggest further research that might illuminate the complete potential of autophagy in addressing SNHL, ultimately leading to the formulation of pioneering therapeutic strategies and approaches for the treatment of deafness.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Animales , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Pérdida Auditiva Sensorineural/metabolismo , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/metabolismo , Modelos Animales de Enfermedad , Autofagia
10.
Elife ; 132024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38334748

RESUMEN

Two calcium-binding proteins, CaBP1 and CaBP2, cooperate to keep calcium channels in the hair cells of the inner ear open.


Asunto(s)
Calcio , Células Ciliadas Auditivas , Calcio/metabolismo , Células Ciliadas Auditivas/metabolismo , Canales de Calcio/metabolismo , Calcio de la Dieta , Células Ciliadas Auditivas Internas/metabolismo , Proteínas de Unión al Calcio/metabolismo
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167024, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38242180

RESUMEN

Oxidative stress is the common mechanism of sensorineural hearing loss (SNHL) caused by many factors, such as noise, drugs and ageing. Here, we used tert-butyl hydroperoxide (t-BHP) to cause oxidative stress damage in HEI-OC1 cells and in an in vitro cochlear explant model. We observed lipid peroxidation, iron accumulation, mitochondrial shrinkage and vanishing of mitochondrial cristae, which caused hair cell ferroptosis, after t-BHP exposure. Moreover, the number of TUNEL-positive cells in cochlear explants and HEI-OC1 cells increased significantly, suggesting that t-BHP caused the apoptosis of hair cells. Administration of deferoxamine (DFOM) significantly attenuated t-BHP-induced hair cell loss and disordered hair cell arrangement in cochlear explants as well as HEI-OC1 cell death, including via apoptosis and ferroptosis. Mechanistically, we found that DFOM treatment reduced t-BHP-induced lipid peroxidation, iron accumulation and mitochondrial pathological changes in hair cells, consequently mitigating apoptosis and ferroptosis. Moreover, DFOM treatment alleviated GSH depletion caused by t-BHP and activated the Nrf2 signalling pathway to exert a protective effect. Furthermore, we confirmed that the protective effect of DFOM mainly depended on its ability to chelate iron by constructing Fth1 knockout (KO), TfR1 KO and Nrf2 KO HEI-OC1 cell lines using CRISPR/Cas9 technology and a Flag-Fth1 (overexpression) HEI-OC1 cell line using the FlpIn™ System. Our findings suggest that DFOM is a potential drug for SNHL treatment due to its ability to inhibit apoptosis and ferroptosis by chelating iron and scavenging reactive oxygen species (ROS).


Asunto(s)
Deferoxamina , Ototoxicidad , Humanos , terc-Butilhidroperóxido/toxicidad , terc-Butilhidroperóxido/metabolismo , Deferoxamina/farmacología , Ototoxicidad/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Células Ciliadas Auditivas/metabolismo , Hierro/metabolismo
12.
Hear Res ; 443: 108962, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38295585

RESUMEN

Nestin expression is associated with pluripotency. Growing evidence suggests nestin is involved in hair cell development. The objective of this study was to investigate the morphology and role of nestin-expressing cells residing in the early postnatal murine inner ear. A lineage-tracing nestin reporter mouse line was used to further characterize these cells. Their cochleae and vestibular organs were immunostained and whole-mounted for cell counting. We found Nestin-expressing cells present in low numbers throughout the inner ear. Three morphotypes were observed: bipolar, unipolar, and globular. Mitotic activity was noted in nestin-expressing cells in the cochlea, utricle, saccule, and crista. Nestin-expressing cell characteristics were then observed after hair cell ablation in two mouse models. First, a reporter model demonstrated nestin expression in a significantly higher proportion of hair cells after hair cell ablation than in control cochleae. However, in a lineage tracing nestin reporter mouse, none of the new hair cells which repopulated the organ of Corti after hair cell ablation expressed nestin, nor did the nestin-expressing cells change in morphotype. In conclusion, Nestin-expressing cells were identified in the cochlea and vestibular organs. After hair cell ablation, nestin-expressing cells did not react to the insult. However, a small number of nestin-expressing cells in all inner ear tissues exhibited mitotic activity, supporting progenitor cell potential, though perhaps not involved in hair cell regeneration.


Asunto(s)
Cóclea , Vestíbulo del Laberinto , Animales , Ratones , Cóclea/metabolismo , Células Ciliadas Auditivas/metabolismo , Nestina/genética , Nestina/metabolismo , Sáculo y Utrículo/metabolismo , Vestíbulo del Laberinto/metabolismo
13.
Mol Ther ; 32(3): 800-817, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38243601

RESUMEN

Hearing loss is a major health concern affecting millions of people worldwide with currently limited treatment options. In clarin-2-deficient Clrn2-/- mice, used here as a model of progressive hearing loss, we report synaptic auditory abnormalities in addition to the previously demonstrated defects of hair bundle structure and mechanoelectrical transduction. We sought an in-depth evaluation of viral-mediated gene delivery as a therapy for these hearing-impaired mice. Supplementation with either the murine Clrn2 or human CLRN2 genes preserved normal hearing in treated Clrn2-/- mice. Conversely, mutated forms of CLRN2, identified in patients with post-lingual moderate to severe hearing loss, failed to prevent hearing loss. The ectopic expression of clarin-2 successfully prevented the loss of stereocilia, maintained normal mechanoelectrical transduction, preserved inner hair cell synaptic function, and ensured near-normal hearing thresholds over time. Maximal hearing preservation was observed when Clrn2 was delivered prior to the loss of transducing stereocilia. Our findings demonstrate that gene therapy is effective for the treatment of post-lingual hearing impairment and age-related deafness associated with CLRN2 patient mutations.


Asunto(s)
Células Ciliadas Auditivas , Pérdida Auditiva , Humanos , Animales , Ratones , Células Ciliadas Auditivas/metabolismo , Audición , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Estereocilios/metabolismo , Suplementos Dietéticos
14.
Mol Biol Rep ; 51(1): 217, 2024 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-38281217

RESUMEN

BACKGROUND: In lower vertebrates like fish, the inner ear and lateral line hair cells (HCs) can regenerate after being damaged by proliferation/differentiation of supporting cells (SCs). However, the HCs of mouse cochlear could only regenerate within one to two weeks after birth but not for adults. METHODS AND RESULTS: To better understand the molecular foundations, we collected several public single-cell RNA sequencing (scRNAseq) data of mouse cochleae from E14 to P33 and extracted the prosensory and supporting cells specifically. Gene Set Enrichment Analysis (GSEA) results revealed a down-regulation of genes in Notch signaling pathway during postnatal stages (P7 and P33). We also identified 107 time-course co-expression genes correlated with developmental stage and predicated that EZH2 and KLF15 may be the key transcriptional regulators for these genes. Expressions of candidate target genes of EZH2 and KLF15 were also found in supporting cells of the auditory epithelia in chick and the neuromasts in zebrafish. Furthermore, inhibiting EZH2 suppressed regeneration of hair cells in zebrafish neuromasts and altered expressions of some developmental stage correlated genes. CONCLUSIONS: Our results extended the understanding for molecular basis of hair cell regeneration ability and revealed the potential role of Ezh2 in it.


Asunto(s)
Análisis de Expresión Génica de una Sola Célula , Animales , Ratones , Proliferación Celular/genética , Células Ciliadas Auditivas/metabolismo , Transducción de Señal/fisiología
15.
Nat Commun ; 15(1): 526, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38228630

RESUMEN

The inner ear is the hub where hair cells (HCs) transduce sound, gravity, and head acceleration stimuli to the brain. Hearing and balance rely on mechanosensation, the fastest sensory signals transmitted to the brain. The mechanoelectrical transducer (MET) channel is the entryway for the sound-balance-brain interface, but the channel-complex composition is not entirely known. Here, we report that the mouse utilizes Piezo1 (Pz1) and Piezo2 (Pz2) isoforms as MET-complex components. The Pz channels, expressed in HC stereocilia, and cell lines are co-localized and co-assembled with MET complex partners. Mice expressing non-functional Pz1 and Pz2 at the ROSA26 locus have impaired auditory and vestibular traits that can only be explained if the Pzs are integral to the MET complex. We suggest that Pz subunits constitute part of the MET complex and that interactions with other MET complex components yield functional MET units to generate HC MET currents.


Asunto(s)
Oído Interno , Células Ciliadas Auditivas Internas , Animales , Ratones , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas/metabolismo , Estereocilios/metabolismo , Oído Interno/metabolismo , Audición , Mecanotransducción Celular , Mamíferos/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo
17.
Chin Med J (Engl) ; 137(6): 651-656, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-37407223

RESUMEN

ABSTRACT: Cochlear spiral ganglion neurons (SGNs) are bipolar ganglion cells and are the first neurons in the auditory transduction pathway. They transmit complex acoustic information from hair cells to second-order sensory neurons in the cochlear nucleus for sound processing. Injury to SGNs causes largely irreversible hearing impairment because these neurons are highly differentiated cells and cannot regenerate, making treatment of sensorineural hearing loss (SNHL) arising from SGN injury difficult. When exposed to ototoxic drugs or damaging levels of noise or when there is loss of neurotrophic factors (NTFs), aging, and presence of other factors, SGNs can be irreversibly damaged, resulting in SNHL. It has been found that NTFs and stem cells can induce regeneration among dead spiral ganglion cells. In this paper, we summarized the present knowledge regarding injury, protection, and regeneration of SGNs.


Asunto(s)
Pérdida Auditiva Sensorineural , Ganglio Espiral de la Cóclea , Humanos , Ganglio Espiral de la Cóclea/metabolismo , Neuronas , Cóclea , Células Ciliadas Auditivas/metabolismo
18.
Tissue Eng Part B Rev ; 30(1): 15-28, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37440318

RESUMEN

The incidence and prevalence of hearing loss is increasing globally at an accelerated pace. Hair cells represent the sensory receptors of auditory and vestibular systems. Hair cell absence, loss or degeneration due to congenital diseases, trauma, toxicity, infection or advancing age, results in disabling hearing loss. Regenerative medicine approaches consisting in stem cell-based hair cell rescue or regeneration, gene therapy, as well as cell and tissue engineering are expected to dramatically improve the therapeutic arsenal available for addressing hearing loss. Current strategies that are using different stem cell types to rescue or to induce hair cell proliferation and regeneration are presented. Gene and cell therapy methods that modulates transdifferentiation of surrounding cell types into hair cells are presented, together with their specific advantages and limitations. Several modalities for improving therapeutic targeting to the inner ear such as nanoparticle-mediated cell and gene delivery are introduced. Further steps in building more relevant high-throughput models for testing novel drugs and advanced therapies are proposed as a modality to accelerate translation to clinical settings.


Asunto(s)
Sordera , Pérdida Auditiva , Humanos , Regeneración , Pérdida Auditiva/terapia , Pérdida Auditiva/metabolismo , Sordera/metabolismo , Sordera/terapia , Células Ciliadas Auditivas/metabolismo , Células Madre
19.
J Appl Toxicol ; 44(2): 235-244, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37650462

RESUMEN

Gentamicin (GM) is one of the commonly used antibiotics in the aminoglycoside class but is ototoxic, which constantly impacts the quality of human life. Pyrroloquinoline quinone (PQQ) as a redox cofactor produced by bacteria was found in soil and foods that exert an antioxidant and redox modulator. It is well documented that the PQQ can alleviate inflammatory responses and cytotoxicity. However, our understanding of PQQ in ototoxicity remains unclear. We reported that PQQ could protect against GM-induced ototoxicity in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells in vitro. To evaluate reactive oxygen species (ROS) production and mitochondrial function, ROS and JC-1 staining, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR) measurements in living cells, mitochondrial dynamics analysis was performed. GM-mediated damage was performed by reducing the production of ROS and inhibiting mitochondria biogenesis and dynamics. PQQ ameliorated the cellular oxidative stress and recovered mitochondrial membrane potential, facilitating the recovery of mitochondrial biogenesis and dynamics. Our in vitro findings improve our understanding of the GM-induced ototoxicity with therapeutic implications for PQQ.


Asunto(s)
Gentamicinas , Ototoxicidad , Humanos , Gentamicinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Cofactor PQQ/farmacología , Cofactor PQQ/uso terapéutico , Cofactor PQQ/metabolismo , Ototoxicidad/etiología , Ototoxicidad/prevención & control , Ototoxicidad/metabolismo , Células Ciliadas Auditivas/metabolismo , Antibacterianos/metabolismo , Apoptosis
20.
Neurobiol Aging ; 134: 115-125, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38056217

RESUMEN

KCNMA1 encodes the K+ potassium channel α-subunit that plays a significant role in the auditory system. Our previous studies indicated that KCNMA1 is associated with age-related hearing loss(AHL). However, the detailed mechanism of KCNMA1 involvement in auditory age-related degradation has not been fully clarified. Therefore, we explored the expression of KCNMA1 in the peripheral auditory of 2-month-old and 12-month-old mice by Western blotting and immunofluorescence. The results of animal experiments showed that KCNMA1 expression was decreased in 12-month-old mice compared with 2-month-old mice, whereas the ferroptosis level was increased. To verify the role of KCNMA1 in AHL, we downregulated KCNMA1 in HEI-OC1 cells by transfecting shRNA. After downregulation, the ferroptosis level was increased and the aging process was accelerated. Furthermore, the aging process was affected by the expression of ferroptosis. In conclusion, these results revealed that KCNMA1 is associated with the aging process in auditory hair cells by regulating ferroptosis, which deepens our understanding of age-related hearing loss.


Asunto(s)
Ferroptosis , Presbiacusia , Animales , Ratones , Regulación hacia Abajo , Ferroptosis/genética , Células Ciliadas Auditivas/metabolismo , Presbiacusia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...