Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 799
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928480

ABSTRACT

Our study aimed to investigate the role of ferroptosis in sevoflurane-induced hearing impairment and explore the mechanism of the microRNA-182-5p (miR-182-5p)/Glutathione Peroxidase 4 (GPX4) pathway in sevoflurane-induced ototoxicity. Immunofluorescence staining was performed using myosin 7a and CtBP2. Cell viability was assessed using the CCK-8 kit. Fe2+ concentration was measured using FerroOrange and Mi-to-FerroGreen fluorescent probes. The lipid peroxide level was assessed using BODIPY 581/591 C11 and MitoSOX fluorescent probes. The auditory brainstem response (ABR) test was conducted to evaluate the hearing status. Bioinformatics tools and dual luciferase gene reporter analysis were used to confirm the direct targeting of miR-182-5p on GPX4 mRNA. GPX4 and miR-182-5p expression in cells was assessed by qRT-PCR and Western blot. Ferrostatin-1 (Fer-1) pretreatment significantly improved hearing impairment and damage to ribbon synapses in mice caused by sevoflurane exposure. Immunofluorescence staining revealed that Fer-1 pretreatment reduced intracellular and mitochondrial iron overload, as well as lipid peroxide accumulation. Our findings indicated that miR-182-5p was upregulated in sevoflurane-exposed HEI-OC1 cells, and miR-182-5p regulated GPX4 expression by binding to the 3'UTR of GPX4 mRNA. The inhibition of miR-182-5p attenuated sevoflurane-induced iron overload and lipid peroxide accumulation. Our study elucidated that the miR-182-5p/GPX4 pathway was implicated in sevoflurane-induced ototoxicity by promoting ferroptosis.


Subject(s)
Ferroptosis , MicroRNAs , Ototoxicity , Phospholipid Hydroperoxide Glutathione Peroxidase , Sevoflurane , Ferroptosis/drug effects , Ferroptosis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Sevoflurane/adverse effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Animals , Mice , Ototoxicity/metabolism , Ototoxicity/etiology , Signal Transduction/drug effects , Cell Line , Male , Hearing Loss/chemically induced , Hearing Loss/genetics , Hearing Loss/metabolism , Hearing Loss/pathology , Mice, Inbred C57BL , Phenylenediamines/pharmacology , Cyclohexylamines
2.
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
3.
Hear Res ; 449: 109033, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38797036

ABSTRACT

Hearing loss is well known to cause plastic changes in the central auditory system and pathological changes such as tinnitus and hyperacusis. Impairment of inner ear functions is the main cause of hearing loss. In aged individuals, not only inner ear dysfunction but also senescence of the central nervous system is the cause of malfunction of the auditory system. In most cases of hearing loss, the activity of the auditory nerve is reduced, but that of the successive auditory centers is increased in a compensatory way. It has been reported that activity changes occur in the inferior colliculus (IC), a critical nexus of the auditory pathway. The IC integrates the inputs from the brainstem and drives the higher auditory centers. Since abnormal activity in the IC is likely to affect auditory perception, it is crucial to elucidate the neuronal mechanism to induce the activity changes of IC neurons with hearing loss. This review outlines recent findings on hearing-loss-induced plastic changes in the IC and brainstem auditory neuronal circuits and discusses what neuronal mechanisms underlie hearing-loss-induced changes in the activity of IC neurons. Considering the different causes of hearing loss, we discuss age-related hearing loss separately from other forms of hearing loss (non-age-related hearing loss). In general, the main plastic change of IC neurons caused by both age-related and non-age-related hearing loss is increased central gain. However, plastic changes in the IC caused by age-related hearing loss seem to be more complex than those caused by non-age-related hearing loss.


Subject(s)
Auditory Pathways , Inferior Colliculi , Neuronal Plasticity , Neurons , Inferior Colliculi/physiopathology , Animals , Humans , Neurons/pathology , Auditory Pathways/physiopathology , Hearing , Presbycusis/physiopathology , Presbycusis/pathology , Auditory Perception , Age Factors , Hearing Loss/physiopathology , Hearing Loss/pathology , Aging/pathology , Evoked Potentials, Auditory, Brain Stem , Acoustic Stimulation
4.
EBioMedicine ; 104: 105160, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38788630

ABSTRACT

BACKGROUND: Hearing impairment is a common condition in the elderly. However, a comprehensive understanding of its neural correlates is still lacking. METHODS: We recruited 284 elderly adults who underwent structural MRI, magnetic resonance spectroscopy, audiometry, and cognitive assessments. Individual hearing abilities indexed by pure tone average (PTA) were correlated with multiple structural MRI-derived cortical morphological indices. For regions showing significant correlations, mediation analyses were performed to examine their role in the relationship between hearing ability and cognitive function. Finally, the correlation maps between hearing ability and cortical morphology were linked with publicly available connectomic gradient, transcriptomic, and neurotransmitter maps. FINDINGS: Poorer hearing was related to cortical thickness (CT) reductions in widespread regions and gyrification index (GI) reductions in the right Area 52 and Insular Granular Complex. The GI in the right Area 52 mediated the relationship between hearing ability and executive function. This mediating effect was further modulated by glutamate and N-acetylaspartate levels in the right auditory region. The PTA-CT correlation map followed microstructural connectomic hierarchy, were related to genes involved in certain biological processes (e.g., glutamate metabolic process), cell types (e.g., excitatory neurons and astrocytes), and developmental stages (i.e., childhood to young adulthood), and covaried with dopamine receptor 1, dopamine transporter, and fluorodopa. The PTA-GI correlation map was related to 5-hydroxytryptamine receptor 2a. INTERPRETATION: Poorer hearing is associated with cortical thinning and folding reductions, which may be engaged in the relationship between hearing impairment and cognitive decline in the elderly and have different neurobiological substrates. FUNDING: See the Acknowledgements section.


Subject(s)
Cerebral Cortex , Cognition , Magnetic Resonance Imaging , Humans , Aged , Male , Female , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/metabolism , Hearing , Hearing Loss/pathology , Hearing Loss/physiopathology , Hearing Loss/etiology , Connectome , Middle Aged , Brain Mapping , Aged, 80 and over
5.
Stem Cell Res ; 78: 103452, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38815527

ABSTRACT

We report the establishment of a human induced pluripotent stem cell (iPSC) line from a 54-year-old male patient with an A1555G mutation in the mitochondrial 12S ribosomal RNA gene (MTRNR1), associated with sensorineural hearing loss. The established iPSC line expressed stemness markers or undifferentiated state markers. We also demonstrated the capacity of the cells to differentiate into the three germ layers, suggesting its pluripotency and utility in the pathological study of sensorineural hearing loss and drug screening for ear disorders.


Subject(s)
DNA, Mitochondrial , Induced Pluripotent Stem Cells , Mutation , Humans , Induced Pluripotent Stem Cells/metabolism , DNA, Mitochondrial/genetics , Male , Middle Aged , Cell Differentiation , Cell Line , RNA, Ribosomal/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Hearing Loss/genetics , Hearing Loss/pathology
6.
Hear Res ; 447: 109012, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703433

ABSTRACT

Hearing loss is a common side effect of many tumor treatments. However, hearing loss can also occur as a direct result of certain tumors of the nervous system, the most common of which are the vestibular schwannomas (VS). These tumors arise from Schwann cells of the vestibulocochlear nerve and their main cause is the loss of function of NF2, with 95 % of cases being sporadic and 5 % being part of the rare neurofibromatosis type 2 (NF2)-related Schwannomatosis. Genetic variations in NF2 do not fully explain the clinical heterogeneity of VS, and interactions between Schwann cells and their microenvironment appear to be critical for tumor development. Preclinical in vitro and in vivo models of VS are needed to develop prognostic biomarkers and targeted therapies. In addition to VS, other tumors can affect hearing. Meningiomas and other masses in the cerebellopontine angle can compress the vestibulocochlear nerve due to their anatomic proximity. Gliomas can disrupt several neurological functions, including hearing; in fact, glioblastoma multiforme, the most aggressive subtype, may exhibit early symptoms of auditory alterations. Besides, treatments for high-grade tumors, including chemotherapy or radiotherapy, as well as incomplete resections, can induce long-term auditory dysfunction. Because hearing loss can have an irreversible and dramatic impact on quality of life, it should be considered in the clinical management plan of patients with tumors, and monitored throughout the course of the disease.


Subject(s)
Hearing Loss , Hearing , Neuroma, Acoustic , Humans , Neuroma, Acoustic/pathology , Neuroma, Acoustic/physiopathology , Neuroma, Acoustic/complications , Hearing Loss/physiopathology , Hearing Loss/etiology , Hearing Loss/pathology , Animals , Neurilemmoma/pathology , Neurilemmoma/complications , Neurilemmoma/therapy , Vestibulocochlear Nerve/pathology , Vestibulocochlear Nerve/physiopathology , Risk Factors , Neurofibromatosis 2/genetics , Neurofibromatosis 2/complications , Neurofibromatosis 2/pathology , Neurofibromatosis 2/physiopathology , Neurofibromatosis 2/therapy , Meningioma/pathology , Meningioma/physiopathology , Meningioma/complications
7.
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
8.
PLoS One ; 19(5): e0303375, 2024.
Article in English | MEDLINE | ID: mdl-38728348

ABSTRACT

Hearing loss is a pivotal risk factor for dementia. It has recently emerged that a disruption in the intercommunication between the cochlea and brain is a key process in the initiation and progression of this disease. However, whether the cochlear properties can be influenced by pathological signals associated with dementia remains unclear. In this study, using a mouse model of Alzheimer's disease (AD), we investigated the impacts of the AD-like amyloid ß (Aß) pathology in the brain on the cochlea. Despite little detectable change in the age-related shift of the hearing threshold, we observed quantitative and qualitative alterations in the protein profile in perilymph, an extracellular fluid that fills the path of sound waves in the cochlea. Our findings highlight the potential contribution of Aß pathology in the brain to the disturbance of cochlear homeostasis.


Subject(s)
Alzheimer Disease , Cochlea , Disease Models, Animal , Perilymph , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Perilymph/metabolism , Cochlea/metabolism , Cochlea/pathology , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Hearing Loss/metabolism , Hearing Loss/pathology
9.
Int J Mol Sci ; 25(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38791427

ABSTRACT

Age-related hearing loss (HL), or presbycusis, is a complex and heterogeneous condition, affecting a significant portion of older adults and involving various interacting mechanisms. Metabolic presbycusis, a type of age-related HL, is characterized by the dysfunction of the stria vascularis, which is crucial for maintaining the endocochlear potential necessary for hearing. Although attention on metabolic presbycusis has waned in recent years, research continues to identify strial pathology as a key factor in age-related HL. This narrative review integrates past and recent research, bridging findings from animal models and human studies, to examine the contributions of the stria vascularis to age-related HL. It provides a brief overview of the structure and function of the stria vascularis and then examines mechanisms contributing to age-related strial dysfunction, including altered ion transport, changes in pigmentation, inflammatory responses, and vascular atrophy. Importantly, this review outlines the contribution of metabolic mechanisms to age-related HL, highlighting areas for future research. It emphasizes the complex interdependence of metabolic and sensorineural mechanisms in the pathology of age-related HL and highlights the importance of animal models in understanding the underlying mechanisms. The comprehensive and mechanistic investigation of all factors contributing to age-related HL, including cochlear metabolic dysfunction, remains crucial to identifying the underlying mechanisms and developing personalized, protective, and restorative treatments.


Subject(s)
Aging , Presbycusis , Stria Vascularis , Humans , Stria Vascularis/metabolism , Stria Vascularis/pathology , Animals , Presbycusis/metabolism , Presbycusis/pathology , Presbycusis/physiopathology , Aging/metabolism , Aging/physiology , Cochlea/metabolism , Cochlea/pathology , Hearing Loss/metabolism , Hearing Loss/pathology
10.
J Med Genet ; 61(7): 689-698, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38458752

ABSTRACT

BACKGROUND: Plexins are large transmembrane receptors for the semaphorin family of signalling proteins. Semaphorin-plexin signalling controls cellular interactions that are critical during development as well as in adult life stages. Nine plexin genes have been identified in humans, but despite the apparent importance of plexins in development, only biallelic PLXND1 and PLXNA1 variants have so far been associated with Mendelian genetic disease. METHODS: Eight individuals from six families presented with a recessively inherited variable clinical condition, with core features of amelogenesis imperfecta (AI) and sensorineural hearing loss (SNHL), with variable intellectual disability. Probands were investigated by exome or genome sequencing. Common variants and those unlikely to affect function were excluded. Variants consistent with autosomal recessive inheritance were prioritised. Variant segregation analysis was performed by Sanger sequencing. RNA expression analysis was conducted in C57Bl6 mice. RESULTS: Rare biallelic pathogenic variants in plexin B2 (PLXNB2), a large transmembrane semaphorin receptor protein, were found to segregate with disease in all six families. The variants identified include missense, nonsense, splicing changes and a multiexon deletion. Plxnb2 expression was detected in differentiating ameloblasts. CONCLUSION: We identify rare biallelic pathogenic variants in PLXNB2 as a cause of a new autosomal recessive, phenotypically diverse syndrome with AI and SNHL as core features. Intellectual disability, ocular disease, ear developmental abnormalities and lymphoedema were also present in multiple cases. The variable syndromic human phenotype overlaps with that seen in Plxnb2 knockout mice, and, together with the rarity of human PLXNB2 variants, may explain why pathogenic variants in PLXNB2 have not been reported previously.


Subject(s)
Amelogenesis Imperfecta , Intellectual Disability , Pedigree , Humans , Animals , Male , Female , Mice , Intellectual Disability/genetics , Intellectual Disability/pathology , Amelogenesis Imperfecta/genetics , Amelogenesis Imperfecta/pathology , Receptors, Cell Surface/genetics , Nerve Tissue Proteins/genetics , Alleles , Child , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Adult , Mutation/genetics , Adolescent , Child, Preschool , Phenotype
11.
Hum Mol Genet ; 33(10): 905-918, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38449065

ABSTRACT

Mutations in AIFM1, encoding for apoptosis-inducing factor (AIF), cause AUNX1, an X-linked neurologic disorder with late-onset auditory neuropathy (AN) and peripheral neuropathy. Despite significant research on AIF, there are limited animal models with the disrupted AIFM1 representing the corresponding phenotype of human AUNX1, characterized by late-onset hearing loss and impaired auditory pathways. Here, we generated an Aifm1 p.R450Q knock-in mouse model (KI) based on the human AIFM1 p.R451Q mutation. Hemizygote KI male mice exhibited progressive hearing loss from P30 onward, with greater severity at P60 and stabilization until P210. Additionally, muscle atrophy was observed at P210. These phenotypic changes were accompanied by a gradual reduction in the number of spiral ganglion neuron cells (SGNs) at P30 and ribbons at P60, which coincided with the translocation of AIF into the nucleus starting from P21 and P30, respectively. The SGNs of KI mice at P210 displayed loss of cytomembrane integrity, abnormal nuclear morphology, and dendritic and axonal demyelination. Furthermore, the inner hair cells and myelin sheath displayed abnormal mitochondrial morphology, while fibroblasts from KI mice showed impaired mitochondrial function. In conclusion, we successfully generated a mouse model recapitulating AUNX1. Our findings indicate that disruption of Aifm1 induced the nuclear translocation of AIF, resulting in the impairment in the auditory pathway.


Subject(s)
Apoptosis Inducing Factor , Disease Models, Animal , Hearing Loss , Animals , Humans , Male , Mice , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Cell Nucleus/metabolism , Cell Nucleus/genetics , Gene Knock-In Techniques , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/metabolism , Mutation , Protein Transport , Spiral Ganglion/metabolism , Spiral Ganglion/pathology
12.
Brain Behav ; 14(1): e3374, 2024 01.
Article in English | MEDLINE | ID: mdl-38376024

ABSTRACT

INTRODUCTION: Previous studies have reported that hearing loss (HL) is associated with dementia, although the mechanistic underpinnings remain elusive. This study aimed to evaluate the changes in brain metabolism in patients with HL and different types of dementia. METHODS: Patients with cognitive impairment (CI) and HL treated at the university-based memory clinic from May 2016 to October 2021 were included. In total, 108 patients with CI and HL prospectively underwent audiometry, neuropsychological test, magnetic resonance imaging, and 18 F-fluorodeoxyglucose positron emission tomography. Twenty-seven individuals without cognitive impairment and hearing loss were enrolled as a control group. Multivariable regression was performed to evaluate brain regions correlated with each pathology type after adjusting for confounding factors. RESULTS: Multivariable regression analyses revealed that Alzheimer's disease-related CI (ADCI) was associated with hypometabolic changes in the right superior temporal gyrus (STG), right middle temporal gyrus (MTG), and bilateral medial temporal lobe. Lewy body disease-related CI (LBDCI) and vascular CI were associated with hypermetabolic and hypometabolic changes in the ascending auditory pathway, respectively. In the pure ADCI group, the degree of HL was positively associated with abnormal increase of brain metabolism in the right MTG, whereas it was negatively associated with decreased brain metabolism in the right STG in the pure LBDCI group. CONCLUSION: Each dementia type is associated with distinct changes in brain metabolism in patients with HL.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Dizocilpine Maleate/analogs & derivatives , Hearing Loss , Humans , Fluorodeoxyglucose F18/metabolism , Alzheimer Disease/pathology , Brain/pathology , Positron-Emission Tomography , Cognitive Dysfunction/pathology , Hearing Loss/complications , Hearing Loss/metabolism , Hearing Loss/pathology
13.
Am J Case Rep ; 25: e941558, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38163945

ABSTRACT

BACKGROUND Superior semicircular canal dehiscence is an inner-ear pathology which presents with vertigo, disequilibrium, and hearing loss. Although the exact etiology of superior semicircular canal dehiscence is unknown, it is thought that an increase in middle-ear pressure disrupts a thin overlying temporal bone. Superior semicircular canal dehiscence is frequently seen in association with dehiscence of the tegmen tympani, which overlies the middle ear. Here, we present a case report of a 52-year-old Puerto Rican man with vertigo, dizziness, vomiting, and mild hearing loss associated with superior semicircular canal and tegmen tympani dehiscence after performing improper scuba diving techniques. CASE REPORT A 52-year-old Puerto Rican man presented to the emergency department with vertigo, dizziness, vomiting, and mild hearing loss in the right ear. The symptoms began shortly after scuba diving with inadequate decompression techniques on ascent. He was treated with recompression therapy with mild but incomplete improvement in symptoms. Bilateral temporal magnetic resonance imaging was suggestive of segmental dehiscence of the right superior semicircular canal and tegmen tympani. High-resolution computed tomography of the temporal bone confirmed right superior semicircular canal and tegmen tympani dehiscence with an intact left inner ear. CONCLUSIONS The increased inner-ear pressure that occurs during scuba diving can lead to dehiscence of the superior semicircular canal and tegmen tympani, causing vertigo and hearing loss. Performance of improper diving techniques can further increase the risk of dehiscence. Therefore, appropriate radiologic evaluation of the inner ear should be performed in such patients.


Subject(s)
Diving , Hearing Loss , Semicircular Canal Dehiscence , Male , Humans , Middle Aged , Dizziness/complications , Dizziness/pathology , Semicircular Canal Dehiscence/complications , Semicircular Canal Dehiscence/pathology , Diving/adverse effects , Semicircular Canals/diagnostic imaging , Ear, Middle/diagnostic imaging , Vertigo/etiology , Vertigo/pathology , Hearing Loss/complications , Hearing Loss/pathology , Vomiting
14.
Pediatr Radiol ; 54(2): 337-346, 2024 02.
Article in English | MEDLINE | ID: mdl-38182852

ABSTRACT

BACKGROUND: White matter change is a well-known abnormality in congenital cytomegalovirus (cCMV) infection, but grading remains challenging and clinical relevance unclear. OBJECTIVE: To investigate if quantitative measurement of white matter apparent diffusion coefficient (ADC) values in magnetic resonance imaging (MRI) of the neonatal brain can predict outcome in cCMV. MATERIALS AND METHODS: A retrospective, single-center observational study, including patients with cCMV who had a neonatal brain MRI with diffusion-weighted imaging, was performed between 2007 and 2020. Regions of interest were systematically placed in the white matter on the ADC maps. Two pediatric radiologists independently scored additional brain abnormalities. Outcome measures were neonatal hearing and cognitive and motor development. Statistical analysis included simple and penalized elastic net regression. RESULTS: Neonatal brain MRI was evaluated in 255 patients (median age 21 days, 25-75 percentiles: 14-28 days, 121 male). Gyral abnormalities were noted in nine patients (3.5%), ventriculomegaly in 24 (9.4%), and subependymal cysts in 58 (22.7%). General white matter ADC was significantly higher in patients with neonatal hearing loss and cognitive and motor impairment (P< 0.05). For neonatal hearing loss, simple logistic regression using only general white matter was the best prediction model, with a receiver operating characteristic area under the curve (AUC)=0.76. For cognitive impairment, interacting elastic net regression, including other brain abnormalities and frontoparietal white matter ADC, performed best, with AUC=0.89. For motor impairment, interacting elastic net regression, including other brain abnormalities and deep anterior frontal white matter performed best, with AUC=0.73. CONCLUSION: Neonatal white matter ADC was significantly higher in patients with clinical impairments. Quantitative ADC measurement may be a useful tool for predicting clinical outcome in cCMV.


Subject(s)
Brain Diseases , Cytomegalovirus Infections , Hearing Loss , White Matter , Infant, Newborn , Child , Humans , Male , White Matter/diagnostic imaging , Retrospective Studies , Magnetic Resonance Imaging/methods , Diffusion Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Cytomegalovirus Infections/diagnostic imaging , Cytomegalovirus Infections/congenital , Brain Diseases/pathology , Hearing Loss/pathology
15.
World Neurosurg ; 182: e675-e691, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38070740

ABSTRACT

OBJECTIVE: The role of surgical management of arachnoid cyst (AC) of the cerebellopontine angle (CPA) is uncertain. This topic has remained controversial with varying contradictory recommendations in the literature, which is limited to mostly case reports. We aimed to provide a comprehensive summary and analysis of symptoms, operative techniques, outcomes, and recurrence of all available surgical cases of AC of the CPA to date. METHODS: A systematic literature search was performed in May 2022 querying several scientific databases. Inclusion criteria specified all studies and case reports of patients with AC located at the CPA for which any relevant surgical procedures were performed. RESULTS: A total of 55 patients from the literature and 5 treated at our institution were included. Mean patient age was 29 years (range, 0.08-79 years), with nearly twice (1.7×) as many female as male patients (37 female, 22 male). Headaches (35%), hearing loss (30%), vertigo (22%), and ataxia (22%) were the most common presentations. Following surgery, 95% experienced symptom improvement, with complete resolution in 64%. Of patients with hearing loss, 44% reported a return to normal. The rate of mortality was 1.69%, and 10% of tumors recurred (mean follow-up 2.3 years [range, 0-15 years]. CONCLUSIONS: Symptomatic AC of the CPA is rare. It exhibits a proclivity for females and commonly manifests with headache, hearing loss, vertigo, and ataxia. While careful selection for surgical candidacy is needed and intervention should be reserved for patients with severe symptoms, surgical decompression is an effective tool for symptom alleviation and recovery.


Subject(s)
Arachnoid Cysts , Deafness , Hearing Loss , Humans , Male , Female , Infant, Newborn , Infant , Child, Preschool , Child , Adolescent , Young Adult , Adult , Middle Aged , Aged , Cerebellopontine Angle/diagnostic imaging , Cerebellopontine Angle/surgery , Cerebellopontine Angle/pathology , Hearing Loss/etiology , Hearing Loss/surgery , Hearing Loss/pathology , Headache/pathology , Vertigo/etiology , Arachnoid Cysts/diagnostic imaging , Arachnoid Cysts/surgery , Ataxia
16.
J Biol Chem ; 300(1): 105516, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042485

ABSTRACT

Class III myosins localize to inner ear hair cell stereocilia and are thought to be crucial for stereocilia length regulation. Mutations within the motor domain of MYO3A that disrupt its intrinsic motor properties have been associated with non-syndromic hearing loss, suggesting that the motor properties of MYO3A are critical for its function within stereocilia. In this study, we investigated the impact of a MYO3A hearing loss mutation, H442N, using both in vitro motor assays and cell biological studies. Our results demonstrate the mutation causes a dramatic increase in intrinsic motor properties, actin-activated ATPase and in vitro actin gliding velocity, as well as an increase in actin protrusion extension velocity. We propose that both "gain of function" and "loss of function" mutations in MYO3A can impair stereocilia length regulation, which is crucial for stereocilia formation during development and normal hearing. Furthermore, we generated chimeric MYO3A constructs that replace the MYO3A motor and neck domain with the motor and neck domain of other myosins. We found that duty ratio, fraction of ATPase cycle myosin is strongly bound to actin, is a critical motor property that dictates the ability to tip localize within filopodia. In addition, in vitro actin gliding velocities correlated extremely well with filopodial extension velocities over a wide range of gliding and extension velocities. Taken together, our data suggest a model in which tip-localized myosin motors exert force that slides the membrane tip-ward, which can combat membrane tension and enhance the actin polymerization rate that ultimately drives protrusion elongation.


Subject(s)
Actins , Hearing Loss , Myosin Type III , Animals , Actins/genetics , Actins/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Chlorocebus aethiops , COS Cells , Hearing Loss/genetics , Hearing Loss/metabolism , Hearing Loss/pathology , Myosin Type III/genetics , Myosin Type III/metabolism , Myosins/genetics , Myosins/metabolism , Stereocilia , Humans
17.
J Int Adv Otol ; 19(6): 454-460, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38088316

ABSTRACT

ACKGROUND: There is a need to operationalize existing clinical data to support precision medicine in progressive hearing loss (HL). By utilizing enlarged vestibular aqueduct (EVA) and its associated inner ear abnormalities as an exemplar, we model data from a large international cohort, confirm prognostic factors for HL, and explore the potential to generate a prediction model to optimize current management paradigms. METHODS: An international retrospective cohort study. Regression analyses were utilized to model frequency-specific HL and identify prognostic factors for baseline average HL severity and progression. Elastic-net regression and machine learning (ML) techniques were utilized to predict future average HL progression based upon routinely measurable clinical, genetic, and radiological data. RESULTS: Higher frequencies of hearing were lost more severely. Prognostic factors for HL were the presence of incomplete partition type 2 (coefficient 12.95 dB, P=.011, 95% CI 3.0-22 dB) and presence of sac signal heterogeneity (P=.009, 95% CI 0.062-0.429) on magnetic resonance imaging. Elastic-net regression outperformed the ML algorithms (R2 0.32, mean absolute error 11.05 dB) with coefficients for baseline average hearing level and the presence of sac heterogeneity contributing the most to prediction outcomes. CONCLUSION: Incomplete partition type 2 and endolymphatic sac signal heterogeneity phenotypes should be monitored closely for hearing deterioration and need for early audiological rehabilitation/cochlear implant. Preliminary prediction models have been generated using routinely collected health data in EVA. This study showcases how international collaborative research can use exemplar techniques to improve precision medicine in relatively rare disease entities.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hearing Loss , Vestibular Aqueduct , Humans , Retrospective Studies , Prognosis , Hearing Loss/pathology , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/pathology , Vestibular Aqueduct/diagnostic imaging , Vestibular Aqueduct/pathology
18.
Mol Med ; 29(1): 141, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37875793

ABSTRACT

Diabetes mellitus (DM) is a major disease threatening human health and its incidence is increasing year on year. As a chronic complication of DM, hearing loss mostly occurs undetectably. However, the mechanism of this diabetes-related hearing loss (DRHL) remains unclear and there is no effective clinical treatment. Studies of animal or human pathology show that DM causes damage to the blood vessels, spiral ganglion neurons, afferent nerve fibers, the organ of Corti, and the stria vascularis of the inner ear. In recent years, more advances in pathological research have revealed the possible mechanism of DRHL. In addition, a large number of clinical studies suggest that the duration and severity of DM are closely related to the incidence and severity of DRHL. This review focuses on the relationship between DM and hearing loss. The clinical audiological characteristics of diabetic patients, risk factors for DRHL, typical pathology, and potential interventions of DRHL are summarized. This will help reveal the pathogenesis and intervention approaches for DRHL.


Subject(s)
Diabetes Mellitus , Hearing Loss , Animals , Humans , Hearing Loss/epidemiology , Hearing Loss/etiology , Hearing Loss/pathology , Diabetes Mellitus/epidemiology , Stria Vascularis/pathology , Risk Factors
19.
Proc Natl Acad Sci U S A ; 120(34): e2307355120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37552762

ABSTRACT

Hearing loss is highly heterogeneous, but one common form involves a failure to maintain the local ionic environment of the sensory hair cells reflected in a reduced endocochlear potential. We used a genetic approach to ask whether this type of pathology can be reversed, using the Spns2tm1a mouse mutant known to show this defect. By activating Spns2 gene transcription at different ages after the onset of hearing loss, we found that an existing auditory impairment can be reversed to give close to normal thresholds for an auditory brainstem response (ABR), at least at low to mid stimulus frequencies. Delaying the activation of Spns2 led to less effective recovery of ABR thresholds, suggesting that there is a critical period for intervention. Early activation of Spns2 not only led to improvement in auditory function but also to protection of sensory hair cells from secondary degeneration. The genetic approach we have used to establish that this type of hearing loss is in principle reversible could be extended to many other diseases using available mouse resources.


Subject(s)
Anion Transport Proteins , Genetic Therapy , Hearing Loss , Animals , Mice , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss/therapy , Anion Transport Proteins/genetics , Transcriptional Activation , Cochlear Microphonic Potentials , Hair Cells, Auditory/pathology
20.
Hear Res ; 435: 108815, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37263113

ABSTRACT

Animal studies have shown that the supporting-cells surviving in the organ of Corti after cochlear insult can be transdifferentiated into hair cells as a treatment for sensorineural hearing loss. Clinical trials of small-molecule therapeutics have been undertaken, but little is known about how to predict the pattern and degree of supporting-cell survival based on audiogram, hearing loss etiology or any other metric obtainable pre-mortem. To address this, we systematically assessed supporting-cell and hair cell survival, as a function of cochlear location in 274 temporal bone cases from the archives at the Massachusetts Eye and Ear and compared the histopathology with the audiograms and hearing-loss etiologies. Results showed that supporting-cell survival was always significantly greater in the apical half than the basal half of the cochlea, that inner pillars were more robust than outer pillars or Deiters' cells, and that total replacement of all supporting cells with a flat epithelium was rare outside of the extreme basal 20% of the cochlea. Supporting cell survival in the basal half of the cochlea was better correlated with the slope of the audiogram than with the mean high-frequency threshold per se: i.e. survival was better with flatter audiograms than with steeply down-sloping audiograms. Cochlear regions with extensive hair cell loss and exceptional supporting cell survival were most common in cases with hearing loss due to ototoxic drugs. Such cases also tended to have less pathology in other functionally critical structures, i.e. spiral ganglion neurons and the stria vascularis.


Subject(s)
Deafness , Hearing Loss , Humans , Cell Survival , Cochlea/pathology , Hair Cells, Auditory/pathology , Stria Vascularis/pathology , Deafness/pathology , Hearing Loss/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...