Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.472
Filter
1.
Cell Biol Toxicol ; 40(1): 79, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289208

ABSTRACT

Noise-induced hidden hearing loss (HHL) is a newly uncovered form of hearing impairment that causes hidden damage to the cochlea. Patients with HHL do not have significant abnormalities in their hearing thresholds, but they experience impaired speech recognition in noisy environments. However, the mechanisms underlying HHL remain unclear. In this study, we developed single-cell transcriptome profiles of the cochlea of mice with HHL, detailing changes in individual cell types. Our study revealed a transient threshold shift, reduced auditory brainstem response wave I amplitude, and decreased number of ribbon synapses in HHL mice. Our findings suggest elevated oxidative stress and GDF15 expression in cochlear hair cells of HHL mice. Notably, the upregulation of GDF15 attenuated oxidative stress and auditory impairment in the cochlea of HHL mice. This suggests that a therapeutic strategy targeting GDF15 may be efficacious against HHL.


Subject(s)
Growth Differentiation Factor 15 , Hearing Loss, Noise-Induced , Oxidative Stress , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Animals , Hearing Loss, Noise-Induced/metabolism , Mice , Cochlea/metabolism , Cochlea/pathology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Male , Mice, Inbred C57BL , Evoked Potentials, Auditory, Brain Stem , Noise/adverse effects , Transcriptome/genetics , Disease Models, Animal , Hearing Loss, Hidden
2.
Commun Biol ; 7(1): 1146, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39277687

ABSTRACT

Microtubules are essential for various cellular processes. The functional diversity of microtubules is attributed to the incorporation of various α- and ß-tubulin isotypes encoded by different genes. In this work, we investigated the functional role of ß4B-tubulin isotype (TUBB4B) in hearing and vision as mutations in TUBB4B are associated with sensorineural disease. Using a Tubb4b knockout mouse model, our findings demonstrate that TUBB4B is essential for hearing. Mice lacking TUBB4B are profoundly deaf due to defects in the inner and middle ear. Specifically, in the inner ear, the absence of TUBB4B lead to disorganized and reduced densities of microtubules in pillar cells, suggesting a critical role for TUBB4B in providing mechanical support for auditory transmission. In the middle ear, Tubb4b-/- mice exhibit motile cilia defects in epithelial cells, leading to the development of otitis media. However, Tubb4b deletion does not affect photoreceptor function or cause retinal degeneration. Intriguingly, ß6-tubulin levels increase in retinas lacking ß4B-tubulin isotype, suggesting a functional compensation mechanism. Our findings illustrate the essential roles of TUBB4B in hearing but not in vision in mice, highlighting the distinct functions of tubulin isotypes in different sensory systems.


Subject(s)
Cilia , Cochlea , Tubulin , Animals , Mice , Cilia/metabolism , Cochlea/cytology , Cochlea/metabolism , Cytoskeleton/metabolism , Mice, Knockout , Microtubules/metabolism , Tubulin/metabolism , Tubulin/genetics
3.
J Neuroinflammation ; 21(1): 223, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39277762

ABSTRACT

The activation of the NLRP3 inflammasome has been linked to several inflammatory and autoinflammatory diseases. Despite cases of potential hearing improvement in immune-mediated diseases, direct evidence of the efficacy of targeting this mechanism in the inner ear is still lacking. Previously, we discovered that macrophages are associated with Sensorineural Hearing loss (SNHL) in Chronic Suppurative Otitis Media (CSOM), the leading cause of this permanent hearing loss in the developing world and incurring costs of $4 to $11 billion dollars in the United States. However, the underlying mechanism remained unknown. Here, we investigate how macrophages drive permanent hearing loss in CSOM. We first confirmed the occurrence of NLRP3 inflammasome activation in cochlear macrophages in CSOM. We then revealed that Outer Hair Cells (OHCs) were protected in CSOM by macrophage depletion and subsequently confirmed the same protection in the NLRP3 knockout condition. Furthermore, we showed that therapeutic inhibition of NLRP3 inflammasome activation and downstream inhibition of IL-1ß protects OHCs in CSOM. Collectively, our data demonstrates that the main driver for hearing loss in CSOM is NLRP3 inflammasome activation in cochlear macrophages and this is therapeutically targetable, leading the way for the development of interventions to prevent the leading cause of permanent hearing loss and a costly disease in the developed world.


Subject(s)
Cochlea , Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Otitis Media, Suppurative , Animals , Female , Humans , Male , Mice , Chronic Disease , Cochlea/metabolism , Cochlea/pathology , Disease Models, Animal , Hearing Loss/etiology , Hearing Loss/prevention & control , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
4.
J Gen Physiol ; 156(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39302316

ABSTRACT

Connexins (Cxs) function as gap junction (GJ) channels and hemichannels that mediate intercellular and transmembrane signaling, respectively. Here, we investigated the proximal segment of the first extracellular loop, E1, of two closely related Cxs, Cx26 and Cx30, that share widespread expression in the cochlea. Computational studies of Cx26 proposed that this segment of E1 contains a parahelix and functions in gating. The sequence of the parahelix is identical between Cx26 and Cx30 except for an Ala/Glu difference at position 49. We show through cysteine-scanning and mutational analyses that position 49 is pore-lining and interacts with the adjacent Asp50 residue to impact hemichannel functionality. When both positions 49 and 50 are charged, as occurs naturally in Cx30, the hemichannel function is dampened. Co-expression of Cx30 with Cx26(D50N), the most common mutation associated with keratitis-ichthyosis-deafness syndrome, results in robust hemichannel currents indicating that position 49-50 interactions are relevant in heteromerically assembled hemichannels. Cysteine substitution at position 49 in either Cx26 or Cx30 results in tonic inhibition of hemichannels, both through disulfide formation and high-affinity metal coordination, suggestive of a flexible region of the pore that can narrow substantially. These effects are absent in GJ channels, which exhibit wild-type functionality. Examination of postnatal cochlear explants suggests that Cx30 expression is associated with reduced propagation of Ca2+ waves. Overall, these data identify a pore locus in E1 of Cx26 and Cx30 that impacts hemichannel functionality and provide new considerations for understanding the roles of these connexins in cochlear function.


Subject(s)
Connexin 26 , Connexin 30 , Connexins , Connexin 26/metabolism , Connexin 26/genetics , Animals , Connexin 30/metabolism , Connexin 30/genetics , Humans , Connexins/metabolism , Connexins/genetics , Protein Domains , Gap Junctions/metabolism , Mice , HEK293 Cells , Cochlea/metabolism , Cochlea/physiology
5.
Immunohorizons ; 8(9): 688-694, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39264736

ABSTRACT

Chronic suppurative otitis media (CSOM) is a neglected disease that afflicts 330 million people worldwide and is the most common cause of permanent hearing loss among children in the developing world. Previously, we discovered that outer hair cell (OHC) loss occurred in the basal turn of the cochlea and that macrophages are the major immune cells associated with OHC loss in CSOM. Macrophage-associated cytokines are upregulated. Specifically, CCL-2, an important member of the MCP family, is elevated over time following middle ear infection. CCR2 is a common receptor of the MCP family and the unique receptor of CCL2. CCR2 knockout mice (CCR2-/-) have been used extensively in studies of monocyte activation in neurodegenerative diseases. In the present study, we investigated the effect of CCR2 deletion on the cochlear immune response and OHC survival in CSOM. The OHC survival rate was 84 ± 12.5% in the basal turn of CCR2+/+ CSOM cochleae, compared with was 63 ± 19.9% in the basal turn of CCR2-/- CSOM cochleae (p ≤ 0.05). Macrophage numbers were significantly reduced in CCR2-/- CSOM cochleae compared with CCR2+/+ CSOM cochleae (p ≤ 0.001). In addition, CCL7 was upregulated, whereas IL-33 was downregulated, in CCR2-/- CSOM cochleae. Finally, the permeability of the blood-labyrinth barrier in the stria vascularis remained unchanged in CCR2-/- CSOM compared with CCR2+/+ CSOM. Taken together, the data suggest that CCR2 plays a protective role through cochlear macrophages in the CSOM cochlea.


Subject(s)
Hair Cells, Auditory, Outer , Otitis Media, Suppurative , Receptors, CCR2 , Animals , Female , Male , Mice , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Chronic Disease , Cochlea/metabolism , Cochlea/pathology , Cochlea/immunology , Disease Models, Animal , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Otitis Media, Suppurative/immunology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics
6.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39254648

ABSTRACT

During embryonic development, Wnt signaling influences both proliferation and sensory formation in the cochlea. How this dual nature of Wnt signaling is coordinated is unknown. In this study, we define a novel role for a Wnt-regulated gene, Mybl2, which was already known to be important for proliferation, in determining the size and patterning of the sensory epithelium in the murine cochlea. Using a quantitative spatial analysis approach and analyzing Mybl2 loss-of-function, we show that Mybl2 promoted proliferation in the inner sulcus domain but limited the size of the sensory domain by influencing their adjoining boundary position via Jag1 regulation during development. Mybl2 loss-of-function simultaneously decreased proliferation in the inner sulcus and increased the size of the sensory domain, resulting in a wider sensory epithelium with ectopic inner hair cell formation during late embryonic stages. These data suggest that progenitor cells in the inner sulcus determine boundary formation and pattern the sensory epithelium via MYBL2.


Subject(s)
Cell Proliferation , Cochlea , Jagged-1 Protein , Stem Cells , Animals , Cochlea/embryology , Cochlea/cytology , Cochlea/metabolism , Mice , Epithelium/embryology , Epithelium/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Gene Expression Regulation, Developmental , Wnt Signaling Pathway , Body Patterning/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/cytology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics
7.
Commun Biol ; 7(1): 1078, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223249

ABSTRACT

Macrophages serve as the primary immune cell population and assume a pivotal role in the immune response within the damaged cochleae. Yet, the origin and role of macrophages in response to noise exposure remain controversial. Here, we take advantage of Ccr2RFP/+ Cx3cr1GFP/+ dual-reporter mice to identify the infiltrated and tissue-resident macrophages. After noise exposure, we reveal that activated resident macrophages change in morphology, increase in abundance, and migrate to the region of hair cells, leading to the loss of outer hair cells and the damage of ribbon synapses. Meanwhile, peripheral monocytes are not implicated in the noise-induced hair cell insults. These noise-induced activities of macrophages are abolished by inhibiting TLR4 signaling, resulting in alleviated insults of hair cells and partial recovery of hearing. Our findings indicate cochlear resident macrophages are pro-inflammatory and detrimental players in acoustic trauma and introduce a potential therapeutic target in noise-induced hearing loss.


Subject(s)
Hearing Loss, Noise-Induced , Macrophages , Animals , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Hair Cells, Auditory/pathology , Hair Cells, Auditory/metabolism , Noise/adverse effects , Macrophage Activation , Cochlea/pathology , Cochlea/immunology , Cochlea/metabolism , Male , Mice, Transgenic
8.
Hear Res ; 452: 109109, 2024 10.
Article in English | MEDLINE | ID: mdl-39241555

ABSTRACT

The genes Ocm (encoding oncomodulin) and Slc26a5 (encoding prestin) are expressed strongly in outer hair cells and both are involved in deafness in mice. However, it is not clear if they influence the expression of each other. In this study, we characterise the auditory phenotype resulting from two new mouse alleles, Ocmtm1e and Slc26a5tm1Cre. Each mutation leads to absence of detectable mRNA transcribed from the mutant allele, but there was no evidence that oncomodulin regulates expression of prestin or vice versa. The two mutants show distinctive patterns of auditory dysfunction. Ocmtm1e homozygotes have normal auditory brainstem response thresholds at 4 weeks old followed by progressive hearing loss starting at high frequencies, while heterozygotes show largely normal thresholds until 6 months of age, when signs of worse thresholds are detected. In contrast, Slc26a5tm1Cre homozygotes have stable but raised thresholds across all frequencies tested, 3 to 42 kHz, at least from 4 to 8 weeks old, while heterozygotes have raised thresholds at high frequencies. Distortion product otoacoustic emissions and cochlear microphonics show deficits similar to auditory brainstem responses in both mutants, suggesting that the origin of hearing impairment is in the outer hair cells. Endocochlear potentials are normal in the two mutants. Scanning electron microscopy revealed normal development of hair cells in Ocmtm1e homozygotes but scattered outer hair cell loss even at 4 weeks old when thresholds appeared normal, indicating that there is not a direct relationship between numbers of outer hair cells present and auditory thresholds.


Subject(s)
Alleles , Auditory Threshold , Evoked Potentials, Auditory, Brain Stem , Homozygote , Otoacoustic Emissions, Spontaneous , Phenotype , Sulfate Transporters , Animals , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Mice , Mutation , Heterozygote , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Cochlea/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Mice, Inbred C57BL , Acoustic Stimulation
9.
Proc Natl Acad Sci U S A ; 121(35): e2405217121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39172791

ABSTRACT

Intercellular signaling mediated by evolutionarily conserved planar cell polarity (PCP) proteins aligns cell polarity along the tissue plane and drives polarized cell behaviors during tissue morphogenesis. Accumulating evidence indicates that the vertebrate PCP pathway is regulated by noncanonical, ß-catenin-independent Wnt signaling; however, the signaling components and mechanisms are incompletely understood. In the mouse hearing organ, both PCP and noncanonical Wnt (ncWnt) signaling are required in the developing auditory sensory epithelium to control cochlear duct elongation and planar polarity of resident sensory hair cells (HCs), including the shape and orientation of the stereociliary hair bundle essential for sound detection. We have recently discovered a Wnt/G-protein/PI3K pathway that coordinates HC planar polarity and intercellular PCP signaling. Here, we identify Wnt7b as a ncWnt ligand acting in concert with Wnt5a to promote tissue elongation in diverse developmental processes. In the cochlea, Wnt5a and Wnt7b are redundantly required for cochlear duct coiling and elongation, HC planar polarity, and asymmetric localization of core PCP proteins Fzd6 and Dvl2. Mechanistically, Wnt5a/Wnt7b-mediated ncWnt signaling promotes membrane recruitment of Daple, a nonreceptor guanine nucleotide exchange factor for Gαi, and activates PI3K/AKT and ERK signaling, which promote asymmetric Fzd6 localization. Thus, ncWnt and PCP signaling pathways have distinct mutant phenotypes and signaling components, suggesting that they act as separate, parallel pathways with nonoverlapping functions in cochlear morphogenesis. NcWnt signaling drives tissue elongation and reinforces intercellular PCP signaling by regulating the trafficking of PCP-specific Frizzled receptors.


Subject(s)
Cell Polarity , Wnt Proteins , Wnt Signaling Pathway , Wnt-5a Protein , Animals , Cell Polarity/physiology , Wnt Proteins/metabolism , Wnt Proteins/genetics , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Mice , Wnt Signaling Pathway/physiology , Cochlea/metabolism , Cochlea/cytology , Cochlea/growth & development , Hair Cells, Auditory/metabolism , Frizzled Receptors/metabolism , Frizzled Receptors/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Morphogenesis
10.
Environ Pollut ; 360: 124691, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39134170

ABSTRACT

Pesticides are a group of extensively used man-made chemicals with high toxicity and strong residues, which are closely related to hearing health. Pesticide metabolite 3, 5, 6-Trichloro-2-pyridinol (TCP) exposure leads to neurotoxicity and auditory cell toxicity. However, whether TCP causes damage to hearing in adult mice is not clear. In this study, adult male C57BL/6 mice continuously exposed to TCP for 21 days showed a dose-dependent elevation of hearing threshold. Outer hair cells and spiral neuron cells were lost in a dose-dependent manner. Type I and V of spiral ligament were severely shrunk and stria vascularis were thinned in mice after 50 and 150 mg/kg TCP exposure. Similarly, ROS levels in the cochlea were significantly increased whereas the activities of anti-oxidation enzymes were decreased after TCP exposure. The expression level of Na+/K+ ATPase was decreased, resulting in cochlear potential disruption. Levels of inflammatory factors (TNF-α and IL-1ß), γ-H2AX, and pro-apoptotic-related factors (Bax and cleaved-Caspase 3) were elevated, respectively. These results suggest that TCP can cause oxidative stress, inflammation, and imbalance of cochlear potential in the cochlea, induce cochlear DNA damage and apoptosis, and cause cochlear morphological changes, eventually leading to impaired hearing function.


Subject(s)
Cochlea , Hearing Loss , Mice, Inbred C57BL , Oxidative Stress , Animals , Male , Mice , Cochlea/drug effects , Cochlea/metabolism , Hearing Loss/chemically induced , Oxidative Stress/drug effects , Pesticides/toxicity , Pyridones/toxicity , Reactive Oxygen Species/metabolism
11.
Nutrients ; 16(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39203853

ABSTRACT

Sensorineural hearing loss (SNHL), characterized by damage to the inner ear or auditory nerve, is a prevalent auditory disorder. This study explores the potential of Castanopsis echinocarpa (CAE) as a therapeutic agent for SNHL. In vivo experiments were conducted using zebrafish and mouse models. Zebrafish with neomycin-induced ototoxicity were treated with CAE, resulting in otic hair cell protection with an EC50 of 0.49 µg/mL and a therapeutic index of 1020. CAE treatment improved auditory function and protected cochlear sensory cells in a mouse model after noise-induced hearing loss (NIHL). RNA sequencing of NIHL mouse cochleae revealed that CAE up-regulates genes involved in neurotransmitter synthesis, secretion, transport, and neuronal survival. Real-time qPCR validation showed that NIHL decreased the mRNA expression of genes related to neuronal function, such as Gabra1, Gad1, Slc32a1, CaMK2b, CaMKIV, and Slc17a7, while the CAE treatment significantly elevated these levels. In conclusion, our findings provide strong evidence that CAE protects against hearing loss by promoting sensory cell protection and enhancing the expression of genes critical for neuronal function and survival.


Subject(s)
Gene Expression Regulation , Hearing Loss, Sensorineural , Plant Extracts , Zebrafish , Animals , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/drug therapy , Hearing Loss, Sensorineural/chemically induced , Mice , Plant Extracts/pharmacology , Gene Expression Regulation/drug effects , Disease Models, Animal , Hearing Loss, Noise-Induced/drug therapy , Neurons/drug effects , Neurons/metabolism , Neomycin/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Cochlea/drug effects , Cochlea/metabolism , Ototoxicity/etiology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism
12.
Redox Rep ; 29(1): 2382943, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39092597

ABSTRACT

OBJECTIVES: Diabetes is closely linked to hearing loss, yet the exact mechanisms remain unclear. Cochlear stria vascularis and pericytes (PCs) are crucial for hearing. This study investigates whether high glucose induces apoptosis in the cochlear stria vascularis and pericytes via elevated ROS levels due to oxidative stress, impacting hearing loss. METHODS: We established a type II diabetes model in C57BL/6J mice and used auditory brainstem response (ABR), Evans blue staining, HE staining, immunohistochemistry, and immunofluorescence to observe changes in hearing, blood-labyrinth barrier (BLB) permeability, stria vascularis morphology, and apoptosis protein expression. Primary cultured stria vascularis pericytes were subjected to high glucose, and apoptosis levels were assessed using flow cytometry, Annexin V-FITC, Hoechst 33342 staining, Western blot, Mitosox, and JC-1 probes. RESULTS: Diabetic mice showed decreased hearing thresholds, reduced stria vascularis density, increased oxidative stress, cell apoptosis, and decreased antioxidant levels. High glucose exposure increased apoptosis and ROS content in pericytes, while mitochondrial membrane potential decreased, with AIF and cytochrome C (CytC) released from mitochondria to the cytoplasm. Adding oxidative scavengers reduced AIF and CytC release, decreasing pericyte apoptosis. DISCUSSION: Hyperglycemia may induce mitochondrial apoptosis of cochlear stria vascularis pericytes through oxidative stress.


Subject(s)
Apoptosis Inducing Factor , Apoptosis , Cytochromes c , Hyperglycemia , Mice, Inbred C57BL , Mitochondria , Oxidative Stress , Pericytes , Proto-Oncogene Proteins c-bcl-2 , Reactive Oxygen Species , Stria Vascularis , Animals , Pericytes/metabolism , Pericytes/drug effects , Pericytes/pathology , Stria Vascularis/metabolism , Stria Vascularis/pathology , Mice , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , Cytochromes c/metabolism , Apoptosis Inducing Factor/metabolism , Hyperglycemia/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Male , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Cochlea/metabolism , Cochlea/pathology
13.
J Int Adv Otol ; 20(3): 189-195, 2024 May 23.
Article in English | MEDLINE | ID: mdl-39158163

ABSTRACT

Different organs respond differently to cisplatin (CDDP)-induced toxicity. Oleuropein (OLE) is a natural phenolic antioxidant. The purpose of this study was to determine the potential protective effect of OLE against CDDP-induced ototoxicity by evaluating expression of genes associated with deoxyribonucleic acid (DNA) damage and repair in cochlear cells. House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were treated using CDDP, OLE, and OLE-CDDP. The water-soluble tetrazolium salt assay was used for monitoring cell viability. Deoxyribonucleic acid damage in cells due to the CDDP, OLE, and combination treatments was determined using a flow-cytometric kit. The change in the expression of 84 genes associated with CCDP, OLE, and OLE-CDDP treatments that induced DNA damage was tested using the reverse transcription polymerase chain reaction array. Changes ≥3-fold were considered significant. House Ear Institute-Organ of Corti 1 cell viability was significantly reduced by CDDP. The OLE-CDDP combination restored the cell viability. Cisplatin increased the H2AX ratio, while OLE-CDDP combination decreased it. Some of the DNA damage-associated genes whose expression was upregulated with CDDP were downregulated with OLE-CDDP, while the expression of genes such as Gadd45g and Rev1 was further downregulated. The expression of DNA repair-related Abl1, Dbd2, Rad52, and Trp53 genes was downregulated with CDDP, whereas their expression was upregulated with OLE-CDDP treatment. In cochlear cells, the OLE-CDDP combination downregulated DNA damage-associated gene expression relative to that upregulated mainly by CDDP. The results revealed that OLE has a potential protective effect on CDDP-induced ototoxicity in cochlear cells by altering the expression of DNA damage-related genes.


Subject(s)
Cell Survival , Cisplatin , Cochlea , DNA Damage , Iridoid Glucosides , Ototoxicity , Cisplatin/toxicity , Iridoid Glucosides/pharmacology , DNA Damage/drug effects , Animals , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , Cell Survival/drug effects , Ototoxicity/prevention & control , Mice , Iridoids/pharmacology , Antineoplastic Agents/toxicity , Antioxidants/pharmacology , Humans , Cell Line , Gene Expression/drug effects
14.
Biophys J ; 123(18): 3163-3175, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39014895

ABSTRACT

Sensory hair cells, including the sensorimotor outer hair cells, which enable the sensitive, sharply tuned responses of the mammalian cochlea, are excited by radial shear between the organ of Corti and the overlying tectorial membrane. It is not currently possible to measure directly in vivo mechanical responses in the narrow cleft between the tectorial membrane and organ of Corti over a wide range of stimulus frequencies and intensities. The mechanical responses can, however, be derived by measuring hair cell receptor potentials. We demonstrate that the seemingly complex frequency- and intensity-dependent behavior of outer hair cell receptor potentials could be qualitatively explained by a two degrees of freedom system with local cochlear partition and tectorial membrane resonances strongly coupled by the outer hair cell stereocilia. A local minimum in the receptor potential below the characteristic frequency should always be observed at a frequency where the tectorial membrane mechanical impedance is minimal, i.e., at the presumed tectorial membrane resonance frequency. The tectorial membrane resonance frequency might, however, shift with stimulus intensity in accordance with a shift in the maximum of the tectorial membrane radial mechanical responses to lower frequencies, as observed in experiments.


Subject(s)
Hair Cells, Auditory, Outer , Tectorial Membrane , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/physiology , Animals , Biomechanical Phenomena , Tectorial Membrane/physiology , Tectorial Membrane/metabolism , Cochlea/physiology , Cochlea/metabolism , Membrane Potentials , Models, Biological , Mechanical Phenomena , Stereocilia/metabolism
15.
Hear Res ; 451: 109091, 2024 09 15.
Article in English | MEDLINE | ID: mdl-39067415

ABSTRACT

Sgms1 encodes sphingomyelin synthase 1, an enzyme in the sphingosine-1-phosphate signalling pathway, and was previously reported to underlie hearing impairment in the mouse. A new mouse allele, Sgms1tm1a, unexpectedly showed normal Auditory Brainstem Response thresholds. We found that the Sgms1tm1a mutation led to incomplete knockdown of transcript to 20 % of normal values, which was enough to support normal hearing. The Sgms1tm1b allele was generated by knocking out exon 7, leading to a complete lack of detectable transcript in the inner ear. Sgms1tm1b homozygotes showed largely normal auditory brainstem response thresholds at first, followed by progressive loss of sensitivity until they showed severe impairment at 6 months old. The endocochlear potential was consistently reduced in Sgms1tm1b mutants at 3, 4 and 8 weeks old, to around 80 mV compared with around 120 mV in control littermates. The stria vascularis showed a characteristic irregularity of marginal cell surfaces and patchy loss of Kcnq1 expression at their apical membrane, and expression analysis of the lateral wall suggested that marginal cells were the most likely initial site of dysfunction in the mutants. Finally, significant association of auditory thresholds with DNA markers within and close to the human SGMS1 gene were found in the 1958 Birth Cohort, suggesting that SGMS1 variants may play a role in the range of hearing abilities in the human population.


Subject(s)
Evoked Potentials, Auditory, Brain Stem , Hearing Loss , Stria Vascularis , Transferases (Other Substituted Phosphate Groups) , Animals , Female , Male , Mice , Auditory Threshold , Cochlea/physiopathology , Cochlea/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Hearing/genetics , Hearing Loss/genetics , Hearing Loss/physiopathology , Homozygote , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype , Stria Vascularis/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics
16.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39037017

ABSTRACT

Following up on our previous observation that early B cell factor (EBF) sites are enriched in open chromatin of the developing sensory epithelium of the mouse cochlea, we investigated the effect of deletion of Ebf1 on inner ear development. We used a Cre driver to delete Ebf1 at the otocyst stage before development of the cochlea. We examined the cochlea at postnatal day (P) 1 and found that the sensory epithelium had doubled in size but the length of the cochlear duct was unaffected. We also found that deletion of Ebf1 led to ectopic sensory patches in the Kölliker's organ. Innervation of the developing organ of Corti was disrupted with no obvious spiral bundles. The ectopic patches were also innervated. All the extra hair cells (HCs) within the sensory epithelium and Kölliker's organ contained mechanoelectrical transduction channels, as indicated by rapid uptake of FM1-43. The excessive numbers of HCs were still present in the adult Ebf1 conditional knockout (cKO) animal. The animals had significantly elevated auditory brainstem response thresholds, suggesting that this gene is essential for hearing development.


Subject(s)
Hair Cells, Auditory , Mice, Knockout , Organ of Corti , Trans-Activators , Animals , Trans-Activators/genetics , Trans-Activators/metabolism , Organ of Corti/metabolism , Hair Cells, Auditory/metabolism , Mice , Deafness/genetics , Gene Deletion , Labyrinth Supporting Cells/metabolism , Cochlea/metabolism , Evoked Potentials, Auditory, Brain Stem
17.
Sci Transl Med ; 16(755): eadn0689, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985856

ABSTRACT

Mutations in microRNA-96 (MIR96) cause autosomal dominant deafness-50 (DFNA50), a form of delayed-onset hearing loss. Genome editing has shown efficacy in hearing recovery through intervention in neonatal mice, yet editing in the adult inner ear is necessary for clinical applications, which has not been done. Here, we developed a genome editing therapy for the MIR96 mutation 14C>A by screening different CRISPR systems and optimizing Cas9 expression and the sgRNA scaffold for efficient and specific mutation editing. AAV delivery of the KKH variant of Staphylococcus aureus Cas9 (SaCas9-KKH) and sgRNA to the cochleae of presymptomatic (3-week-old) and symptomatic (6-week-old) adult Mir9614C>A/+ mutant mice improved hearing long term, with efficacy increased by injection at a younger age. Adult inner ear delivery resulted in transient Cas9 expression without evidence of AAV genomic integration, indicating the good safety profile of our in vivo genome editing strategy. We developed a dual-AAV system, including an AAV-sgmiR96-master carrying sgRNAs against all known human MIR96 mutations. Because mouse and human MIR96 sequences share 100% homology, our approach and sgRNA selection for efficient and specific hair cell editing for long-term hearing recovery lay the foundation for the development of treatment for patients with DFNA50 caused by MIR96 mutations.


Subject(s)
Dependovirus , Gene Editing , Hearing Loss , MicroRNAs , Mutation , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Editing/methods , Humans , Mutation/genetics , Hearing Loss/genetics , Hearing Loss/therapy , Dependovirus/genetics , Mice , CRISPR-Cas Systems/genetics , Cochlea/metabolism , Genetic Therapy/methods , RNA, Guide, CRISPR-Cas Systems/genetics , Base Sequence , Hearing
18.
Mech Ageing Dev ; 221: 111964, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39019118

ABSTRACT

Age-related hearing loss (ARHL) is an auditory disease characterized by gradual loss of high-frequency hearing sensitivity. Excessive reactive oxygen species trigger NLRP3-inflammasome activation that may be crucial for ARHL pathogenesis. The antioxidant factor Sestrin2 (SESN2) has been reported to be involved in the remission of oxidative stress and ARHL. However, the mechanism by which SESN2 protects auditory cells in the aging mouse cochlea remains unknown. Here, we observed that ectopic overexpression of SESN2 delayed ARHL, whereas SESN2 knockdown accelerated it. Importantly, we elucidated that SESN2 exerts a hearing-protective effect by inhibiting the production of NLRP3 by acting as a mitophagy agonist. Our study proposes a new theoretical basis for SESN2 prevention of ARHL and provides a novel therapeutic strategy for maintaining SESN2 activity in the aging cochlea.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Presbycusis , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Presbycusis/metabolism , Presbycusis/pathology , Presbycusis/prevention & control , Inflammasomes/metabolism , Mitophagy/physiology , Aging/metabolism , Cochlea/metabolism , Cochlea/pathology , Oxidative Stress , Sestrins
19.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000390

ABSTRACT

The motor protein prestin, found in the inner ear's outer hair cells (OHCs), is responsible for high sensitivity and sharp frequency selectivity in mammalian hearing. Some studies have suggested that prestin could be a serological biomarker for cochlear damage, as OHCs are highly vulnerable to damage from various sources. However, the reported data are inconsistent and lack appropriate negative controls. To investigate whether prestin can be used as a serological biomarker for cochlear damage or stress, we measured prestin quantities in the bloodstreams of mice using ELISA kits from different companies. Wildtype (WT) mice were exposed to different ototoxic treatments, including noise exposure and ototoxic reagents that rapidly kill OHCs. Prestin-knockout (KO) mice were used as a negative control. Our data show that some ELISA kits were not able to detect prestin specifically. The ELISA kit that could detect the prestin protein from cochlear homogenates failed to detect prestin in the bloodstream, despite there being significant damage to OHCs in the cochleae. Furthermore, the optical densities of the serum samples, which correlate to prestin quantities, were significantly influenced by hemolysis in the samples. In conclusion, Prestin from OHCs is not a sensitive and reliable serological biomarker for detecting cochlear damage in mice using ELISA.


Subject(s)
Biomarkers , Hair Cells, Auditory, Outer , Molecular Motor Proteins , Animals , Biomarkers/blood , Mice , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/metabolism , Molecular Motor Proteins/metabolism , Molecular Motor Proteins/genetics , Mice, Knockout , Cochlea/pathology , Cochlea/metabolism , Enzyme-Linked Immunosorbent Assay , Mice, Inbred C57BL
20.
Otol Neurotol ; 45(7): 810-817, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38995724

ABSTRACT

HYPOTHESIS: Transforming growth factor beta-1 (TGFß-1) and connective tissue growth factor (CTGF) are upregulated in the implanted human cochlea. BACKGROUND: Cochlear implantation can lead to insertion trauma and intracochlear new tissue formation, which can detrimentally affect implant performance. TGFß-1 and CTGF are profibrotic proteins implicated in various pathologic conditions, but little is known about their role in the cochlea. The present study aimed to characterize the expression of these proteins in the human implanted cochlea. METHODS: Archival human temporal bones (HTB) acquired from 12 patients with previous CI and histopathological evidence of new tissue formation as well as surgical samples of human intracochlear scar tissue surrounding the explanted CI were used in this study. Histopathologic analysis of fibrosis and osteoneogenesis was conducted using H&E. Protein expression was characterized using immunofluorescence. RNA expression from surgical specimens of fibrotic tissue surrounding the CI was quantified using qRT-PCR. RESULTS: TGFß-1 and CTGF protein expressions were upregulated in the areas of fibrosis and osteoneogenesis surrounding the CI HTB. Similarly, surgical samples demonstrated upregulation of protein and mRNA expression of TGFß-1 and mild upregulation of CTGF compared with control. TGFß-1 was expressed diffusely within the fibrous capsule, whereas CTGF was expressed in the thickened portion toward the modiolus and the fibrosis-osteoneogensis junction. CONCLUSION: To our knowledge, this is the first study to demonstrate increased expression of TGFß-1 and CTGF in the human implanted cochlea and may provide better understanding of the mechanism behind this pathogenic process to better develop future mitigating interventions.


Subject(s)
Cochlea , Connective Tissue Growth Factor , Transforming Growth Factor beta1 , Humans , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cochlea/metabolism , Male , Middle Aged , Female , Cochlear Implantation , Cochlear Implants , Temporal Bone/metabolism , Temporal Bone/pathology , Fibrosis , Aged , Adult
SELECTION OF CITATIONS
SEARCH DETAIL