Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.383
Filter
Add more filters

Publication year range
1.
Physiol Rev ; 101(4): 1609-1632, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33769102

ABSTRACT

Tinnitus is a pervasive public health issue that affects ∼15% of the United States population. Similar estimates have also been shown on a global scale, with similar prevalence found in Europe, Asia, and Africa. The severity of tinnitus is heterogeneous, ranging from mildly bothersome to extremely disruptive. In the United States, ∼10-20% of individuals who experience tinnitus report symptoms that severely reduce their quality of life. Due to the huge personal and societal burden, in the last 20 yr a concerted effort on basic and clinical research has significantly advanced our understanding and treatment of this disorder. Yet, neither full understanding, nor cure exists. We know that tinnitus is the persistent involuntary phantom percept of internally generated nonverbal indistinct noises and tones, which in most cases is initiated by acquired hearing loss and maintained only when this loss is coupled with distinct neuronal changes in auditory and extra-auditory brain networks. Yet, the exact mechanisms and patterns of neural activity that are necessary and sufficient for the perceptual generation and maintenance of tinnitus remain incompletely understood. Combinations of animal model and human research will be essential in filling these gaps. Nevertheless, the existing progress in investigating the neurophysiological mechanisms has improved current treatment and highlighted novel targets for drug development and clinical trials. The aim of this review is to thoroughly discuss the current state of human and animal tinnitus research, outline current challenges, and highlight new and exciting research opportunities.


Subject(s)
Neurosciences , Tinnitus/physiopathology , Tinnitus/therapy , Animals , Disease Models, Animal , Hearing Loss/etiology , Hearing Loss/physiopathology , Humans , Quality of Life , Tinnitus/complications
2.
Annu Rev Pharmacol Toxicol ; 64: 211-230, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-37562496

ABSTRACT

Each year over half a million people experience permanent hearing loss caused by treatment with therapeutic drugs with ototoxic side effects. There is a major unmet clinical need for therapies that protect against this hearing loss without reducing the therapeutic efficacy of these lifesaving drugs. At least 17 clinical trials evaluating 10 therapeutics are currently underway for therapies aimed at preventing aminoglycoside- and/or cisplatin-induced ototoxicity. This review describes the preclinical and clinical development of each of these approaches, provides updates on the status of ongoing trials, and highlights the importance of appropriate outcome measures in trial design and the value of reporting criteria in the dissemination of results.


Subject(s)
Hearing Loss , Humans , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Clinical Trials as Topic
3.
Trends Genet ; 40(3): 228-237, 2024 03.
Article in English | MEDLINE | ID: mdl-38161109

ABSTRACT

Age-related hearing loss (ARHL) is a prevalent concern in the elderly population. Recent genome-wide and phenome-wide association studies (GWASs and PheWASs) have delved into the identification of causative variants and the understanding of pleiotropy, highlighting the polygenic intricacies of this complex condition. While recent large-scale GWASs have pinpointed significant SNPs and risk variants associated with ARHL, the detailed mechanisms, encompassing both genetic and epigenetic modifications, remain to be fully elucidated. This review presents the latest advances in association studies, integrating findings from both human studies and model organisms. By juxtaposing historical perspectives with contemporary genomics, we aim to catalyze innovative research and foster the development of novel therapeutic strategies for ARHL.


Subject(s)
Presbycusis , Humans , Aged , Presbycusis/genetics , Presbycusis/epidemiology , Polymorphism, Single Nucleotide/genetics
4.
Am J Hum Genet ; 111(10): 2253-2264, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39241775

ABSTRACT

Gene therapy has made significant progress in the treatment of hereditary hearing loss. However, most research has focused on deafness-related genes that are primarily expressed in hair cells with less attention given to multisite-expressed deafness genes. MPZL2, the second leading cause of mild-to-moderate hereditary deafness, is widely expressed in different inner ear cells. We generated a mouse model with a deletion in the Mpzl2 gene, which displayed moderate and slowly progressive hearing loss, mimicking the phenotype of individuals with DFNB111. We developed a gene replacement therapy system mediated by AAV-ie for efficient transduction in various types of cochlear cells. AAV-ie-Mpzl2 administration significantly lowered the auditory brainstem response and distortion product otoacoustic emission thresholds of Mpzl2-/- mice for at least seven months. AAV-ie-Mpzl2 delivery restored the structural integrity in both outer hair cells and Deiters cells. This study suggests the potential of gene therapy for MPZL2-related deafness and provides a proof of concept for gene therapy targeting other deafness-related genes that are expressed in different cell populations in the cochlea.


Subject(s)
Deafness , Disease Models, Animal , Genetic Therapy , Animals , Mice , Humans , Deafness/genetics , Deafness/therapy , Dependovirus/genetics , Genetic Vectors , Hearing/genetics , Mice, Knockout , Evoked Potentials, Auditory, Brain Stem , Cochlea/metabolism , Cochlea/pathology
5.
Am J Hum Genet ; 111(3): 614-617, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38330941

ABSTRACT

Age-related hearing loss (ARHL) is a major health concern among the elderly population. It is hoped that increasing our understanding of its underlying pathophysiological processes will lead to the development of novel therapies. Recent genome-wide association studies (GWASs) discovered a few dozen genetic variants in association with elevated risk for ARHL. Integrated analysis of GWAS results and transcriptomics data is a powerful approach for elucidating specific cell types that are involved in disease pathogenesis. Intriguingly, recent studies that applied such bioinformatics approaches to ARHL resulted in disagreeing findings as for the key cell types that are most strongly linked to the genetic pathogenesis of ARHL. These conflicting studies pointed either to cochlear sensory epithelial or to stria vascularis cells as the cell types most prominently involved in the genetic basis of ARHL. Seeking to resolve this discrepancy, we integrated the analysis of four ARHL GWAS datasets with four independent inner-ear single-cell RNA-sequencing datasets. Our analysis clearly points to the cochlear sensory epithelial cells as the key cells for the genetic predisposition to ARHL. We also explain the limitation of the bioinformatics analysis performed by previous studies that led to missing the enrichment for ARHL GWAS signal in sensory epithelial cells. Collectively, we show that cochlear epithelial cells, not stria vascularis cells, are the main inner-ear cells related to the genetic pathogenesis of ARHL.


Subject(s)
Presbycusis , Stria Vascularis , Aged , Humans , Stria Vascularis/pathology , Genome-Wide Association Study , Cochlea/pathology , Presbycusis/genetics , Presbycusis/pathology , Epithelium/pathology
6.
Proc Natl Acad Sci U S A ; 121(24): e2311570121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38830095

ABSTRACT

Even a transient period of hearing loss during the developmental critical period can induce long-lasting deficits in temporal and spectral perception. These perceptual deficits correlate with speech perception in humans. In gerbils, these hearing loss-induced perceptual deficits are correlated with a reduction of both ionotropic GABAA and metabotropic GABAB receptor-mediated synaptic inhibition in auditory cortex, but most research on critical period plasticity has focused on GABAA receptors. Therefore, we developed viral vectors to express proteins that would upregulate gerbil postsynaptic inhibitory receptor subunits (GABAA, Gabra1; GABAB, Gabbr1b) in pyramidal neurons, and an enzyme that mediates GABA synthesis (GAD65) presynaptically in parvalbumin-expressing interneurons. A transient period of developmental hearing loss during the auditory critical period significantly impaired perceptual performance on two auditory tasks: amplitude modulation depth detection and spectral modulation depth detection. We then tested the capacity of each vector to restore perceptual performance on these auditory tasks. While both GABA receptor vectors increased the amplitude of cortical inhibitory postsynaptic potentials, only viral expression of postsynaptic GABAB receptors improved perceptual thresholds to control levels. Similarly, presynaptic GAD65 expression improved perceptual performance on spectral modulation detection. These findings suggest that recovering performance on auditory perceptual tasks depends on GABAB receptor-dependent transmission at the auditory cortex parvalbumin to pyramidal synapse and point to potential therapeutic targets for developmental sensory disorders.


Subject(s)
Auditory Cortex , Gerbillinae , Hearing Loss , Animals , Auditory Cortex/metabolism , Auditory Cortex/physiopathology , Hearing Loss/genetics , Hearing Loss/physiopathology , Receptors, GABA-B/metabolism , Receptors, GABA-B/genetics , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Parvalbumins/metabolism , Parvalbumins/genetics , Auditory Perception/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Genetic Vectors/genetics
7.
Proc Natl Acad Sci U S A ; 121(8): e2310561121, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38354264

ABSTRACT

Exposure to loud noise triggers sensory organ damage and degeneration that, in turn, leads to hearing loss. Despite the troublesome impact of noise-induced hearing loss (NIHL) in individuals and societies, treatment strategies that protect and restore hearing are few and insufficient. As such, identification and mechanistic understanding of the signaling pathways involved in NIHL are required. Biological zinc is mostly bound to proteins, where it plays major structural or catalytic roles; however, there is also a pool of unbound, mobile (labile) zinc. Labile zinc is mostly found in vesicles in secretory tissues, where it is released and plays a critical signaling role. In the brain, labile zinc fine-tunes neurotransmission and sensory processing. However, injury-induced dysregulation of labile zinc signaling contributes to neurodegeneration. Here, we tested whether zinc dysregulation occurs and contributes to NIHL in mice. We found that ZnT3, the vesicular zinc transporter responsible for loading zinc into vesicles, is expressed in cochlear hair cells and the spiral limbus, with labile zinc also present in the same areas. Soon after noise trauma, ZnT3 and zinc levels are significantly increased, and their subcellular localization is vastly altered. Disruption of zinc signaling, either via ZnT3 deletion or pharmacological zinc chelation, mitigated NIHL, as evidenced by enhanced auditory brainstem responses, distortion product otoacoustic emissions, and number of hair cell synapses. These data reveal that noise-induced zinc dysregulation is associated with cochlear dysfunction and recovery after NIHL, and point to zinc chelation as a potential treatment for mitigating NIHL.


Subject(s)
Hearing Loss, Noise-Induced , Mice , Animals , Hearing Loss, Noise-Induced/drug therapy , Zinc , Cochlea , Noise/adverse effects , Hearing , Evoked Potentials, Auditory, Brain Stem/physiology , Auditory Threshold
8.
Proc Natl Acad Sci U S A ; 121(15): e2314763121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38557194

ABSTRACT

Although sudden sensorineural hearing loss (SSNHL) is a serious condition, there are currently no approved drugs for its treatment. Nevertheless, there is a growing understanding that the cochlear pathologies that underlie SSNHL include apoptotic death of sensory outer hair cells (OHCs) as well as loss of ribbon synapses connecting sensory inner hair cells (IHCs) and neurites of the auditory nerve, designated synaptopathy. Noise-induced hearing loss (NIHL) is a common subtype of SSNHL and is widely used to model hearing loss preclinically. Here, we demonstrate that a single interventive application of a small pyridoindole molecule (AC102) into the middle ear restored auditory function almost to prenoise levels in a guinea pig model of NIHL. AC102 prevented noise-triggered loss of OHCs and reduced IHC synaptopathy suggesting a role of AC102 in reconnecting auditory neurons to their sensory target cells. Notably, AC102 exerted its therapeutic properties over a wide frequency range. Such strong improvements in hearing have not previously been demonstrated for other therapeutic agents. In vitro experiments of a neuronal damage model revealed that AC102 protected cells from apoptosis and promoted neurite growth. These effects may be explained by increased production of adenosine triphosphate, indicating improved mitochondrial function, and reduced levels of reactive-oxygen species which prevents the apoptotic processes responsible for OHC death. This action profile of AC102 might be causal for the observed hearing recovery in in vivo models.


Subject(s)
Hearing Loss, Noise-Induced , Hearing Loss, Sensorineural , Guinea Pigs , Animals , Hearing , Cochlea , Noise/adverse effects , Hair Cells, Auditory, Outer/physiology , Auditory Threshold
9.
Hum Mol Genet ; 33(19): 1648-1659, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-38981620

ABSTRACT

Hearing loss is the most common congenital sensory deficit worldwide and exhibits high genetic heterogeneity, making molecular diagnoses elusive for most individuals. Detecting novel mutations that contribute to hearing loss is crucial to providing accurate personalized diagnoses, tailored interventions, and improving prognosis. Copy number variants (CNVs) are structural mutations that are understudied, potential contributors to hearing loss. Here, we present the Abnormal Wobbly Gait (AWG) mouse, the first documented mutant exhibiting waltzer-like locomotor dysfunction, hyperactivity, circling behaviour, and profound deafness caused by a spontaneous CNV deletion in cadherin 23 (Cdh23). We were unable to identify the causative mutation through a conventional whole-genome sequencing (WGS) and variant detection pipeline, but instead found a linked variant in hexokinase 1 (Hk1) that was insufficient to recapitulate the AWG phenotype when introduced into C57BL/6J mice using CRISPR-Cas9. Investigating nearby deafness-associated genes revealed a pronounced downregulation of Cdh23 mRNA and a complete absence of full-length CDH23 protein, which is critical for the development and maintenance of inner ear hair cells, in whole head extracts from AWG neonates. Manual inspection of WGS read depth plots of the Cdh23 locus revealed a putative 10.4 kb genomic deletion of exons 11 and 12 that was validated by PCR and Sanger sequencing. This study underscores the imperative to refine variant detection strategies to permit identification of pathogenic CNVs easily missed by conventional variant calling to enhance diagnostic precision and ultimately improve clinical outcomes for individuals with genetically heterogenous disorders such as hearing loss.


Subject(s)
Cadherins , DNA Copy Number Variations , Deafness , Animals , DNA Copy Number Variations/genetics , Cadherins/genetics , Mice , Deafness/genetics , Vestibular Diseases/genetics , Humans , Hexokinase/genetics , Disease Models, Animal , Mice, Inbred C57BL , Whole Genome Sequencing , Phenotype , Cadherin Related Proteins , Mutation
10.
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
11.
J Neurosci ; 44(18)2024 May 01.
Article in English | MEDLINE | ID: mdl-38548338

ABSTRACT

Hearing loss is a major disability in everyday life and therapeutic interventions to protect hearing would benefit a large portion of the world population. Here we found that mice devoid of the protein kinase suppressor of RAS 1 (KSR1) in their tissues (germline KO mice) exhibit resistance to both cisplatin- and noise-induced permanent hearing loss compared with their wild-type KSR1 littermates. KSR1 is a scaffold protein that brings in proximity the mitogen-activated protein kinase (MAPK) proteins BRAF, MEK1/2 and ERK1/2 and assists in their activation through a phosphorylation cascade induced by both cisplatin and noise insults in the cochlear cells. KSR1, BRAF, MEK1/2, and ERK1/2 are all ubiquitously expressed in the cochlea. Deleting the KSR1 protein tempered down the MAPK phosphorylation cascade in the cochlear cells following both cisplatin and noise insults and conferred hearing protection of up to 30 dB SPL in three tested frequencies in male and female mice. Treatment with dabrafenib, an FDA-approved oral BRAF inhibitor, protected male and female KSR1 wild-type mice from both cisplatin- and noise-induced hearing loss. Dabrafenib treatment did not enhance the protection of KO KSR1 mice, providing evidence dabrafenib works primarily through the MAPK pathway. Thus, either elimination of the KSR1 gene expression or drug inhibition of the MAPK cellular pathway in mice resulted in profound protection from both cisplatin- and noise-induced hearing loss. Inhibition of the MAPK pathway, a cellular pathway that responds to damage in the cochlear cells, can prove a valuable strategy to protect and treat hearing loss.


Subject(s)
Cisplatin , Hearing Loss, Noise-Induced , MAP Kinase Signaling System , Mice, Knockout , Protein Kinases , Animals , Cisplatin/toxicity , Mice , Female , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/genetics , Male , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Protein Kinases/metabolism , Protein Kinases/genetics , Mice, Inbred C57BL
12.
J Neurosci ; 44(38)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39147590

ABSTRACT

Ribbon synapses of inner hair cells (IHCs) are uniquely designed for ultrafast and indefatigable neurotransmission of the sound. The molecular machinery ensuring the efficient, compensatory recycling of the synaptic vesicles (SVs), however, remains elusive. This study showed that hair cell knock-out of murine Dmxl2, whose human homolog is responsible for nonsyndromic sensorineural hearing loss DFNA71, resulted in auditory synaptopathy by impairing synaptic endocytosis and recycling. The mutant mice in the C57BL/6J background of either sex had mild hearing loss with severely diminished wave I amplitude of the auditory brainstem response. Membrane capacitance measurements of the IHCs revealed deficiency in sustained synaptic exocytosis and endocytic membrane retrieval. Consistent with the electrophysiological findings, 3D electron microscopy reconstruction showed reduced reserve pool of SVs and endocytic compartments, while the membrane-proximal and ribbon-associated vesicles remain intact. Our results propose an important role of DMXL2 in hair cell endocytosis and recycling of the SVs.


Subject(s)
Endocytosis , Hair Cells, Auditory, Inner , Nerve Tissue Proteins , Synaptic Vesicles , Animals , Female , Male , Mice , Endocytosis/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Exocytosis/physiology , Hair Cells, Auditory, Inner/physiology , Mice, Inbred C57BL , Mice, Knockout , Synaptic Vesicles/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
13.
J Neurosci ; 44(4)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38050104

ABSTRACT

Outer hair cells (OHCs) of the organ of Corti (OoC), acting as bidirectional cellular mechanoelectrical transducers, generate, receive, and exchange forces with other major elements of the cochlear partition, including the sensory inner hair cells (IHCs). Force exchange is mediated via a supporting cell scaffold, including Deiters' (DC) and outer pillar cells (OPC), to enable the sensitivity and exquisite frequency selectivity of the mammalian cochlea and to transmit its responses to the auditory nerve. To selectively activate DCs and OPCs in male and female mice, we conditionally expressed in them a hyperpolarizing halorhodopsin (HOP), a light-gated inward chloride ion pump, and measured extracellular receptor potentials (ERPs) and their DC component (ERPDCs) from the cortilymph, which fills the OoC fluid spaces, and compared the responses with similar potentials from HOP-/- littermates. The compound action potentials (CAP) of the auditory nerve were measured as an indication of IHC activity and transmission of cochlear responses to the CNS. HOP light-activated hyperpolarization of DCs and OPCs suppressed cochlear amplification through changing the timing of its feedback, altered basilar membrane (BM) responses to tones at all measured levels and frequencies, and reduced IHC excitation. HOP activation findings reported here complement recent studies that revealed channelrhodopsin activation depolarized DCs and OPCs and effectively bypassed, rather than blocked, the control of OHC mechanical and electrical responses to sound and their contribution to timed and directed electromechanical feedback to the mammalian cochlea. Moreover, our findings identify DCs and OPCs as potential targets for the treatment of noise-induced hearing loss.


Subject(s)
Hair Cells, Auditory, Outer , Hair Cells, Vestibular , Female , Male , Mice , Animals , Hair Cells, Auditory, Outer/physiology , Optogenetics , Cochlea/physiology , Hair Cells, Auditory, Inner/physiology , Organ of Corti/physiology , Mammals
14.
J Biol Chem ; : 107813, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39322015

ABSTRACT

The formin protein Diaph3 is an actin nucleator that regulates numerous cytoskeleton-dependent cellular processes through the activation of actin polymerization. Expression and activity of Diaph3 is tightly regulated: lack of Diaph3 results in developmental defects and embryonic lethality in mice, while overexpression of Diaph3 causes auditory neuropathy. It is known that Diaph3 homophilic interactions include the intramolecular interaction of its DID-DAD domains and the intermolecular interactions of DD-DD domains or FH2-FH2 domains. However, the physiological significance of these interactions in Diaph3 protein stability and activity is not fully understood. In this study, we show that FH2-FH2 interaction promotes Diaph3 activity, while DID-DAD and DD-DD interactions inhibit Diaph3 activity through distinct mechanisms. DID-DAD interaction is responsible for the autoinhibition of Diaph3 protein, which is disrupted by binding of Rho GTPases. Interestingly, we find that DID-DAD interaction stabilizes the expression of each DID or DAD domain against proteasomal-mediated degradation. Disruption of DID-DAD interaction by RhoA binding or M1041A mutation causes increased Diaph3 activity and accelerated degradation of the activated Diaph3 protein. Further, the activated Diaph3 is ubiquitinated at K1142/1143/1144 lysine residues by the E3 ligase Stub1. Expression of Stub1 is causally related to the stability and activity of Diaph3. Knockdown of Stub1 in mouse cochlea results in hair cell stereocilia defects, neuronal degeneration and hearing loss, resembling the phenotypes of mice overexpressing Diaph3. Thus, our study reports a novel regulatory mechanism of Diaph3 protein expression and activity whereby the active but not inactive Diaph3 is readily degraded to prevent excessive actin polymerization.

15.
Annu Rev Genomics Hum Genet ; 23: 275-299, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35667089

ABSTRACT

Current estimates suggest that nearly half a billion people worldwide are affected by hearing loss. Because of the major psychological, social, economic, and health ramifications, considerable efforts have been invested in identifying the genes and molecular pathways involved in hearing loss, whether genetic or environmental, to promote prevention, improve rehabilitation, and develop therapeutics. Genomic sequencing technologies have led to the discovery of genes associated with hearing loss. Studies of the transcriptome and epigenome of the inner ear have characterized key regulators and pathways involved in the development of the inner ear and have paved the way for their use in regenerative medicine. In parallel, the immense preclinical success of using viral vectors for gene delivery in animal models of hearing loss has motivated the industry to work on translating such approaches into the clinic. Here, we review the recent advances in the genomics of auditory function and dysfunction, from patient diagnostics to epigenetics and gene therapy.


Subject(s)
Deafness , Ear, Inner , Hearing Loss , Animals , Deafness/metabolism , Deafness/therapy , Ear, Inner/metabolism , Genetic Therapy , Genomics , Hearing Loss/genetics , Hearing Loss/therapy , Humans
16.
Am J Hum Genet ; 109(6): 1077-1091, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35580588

ABSTRACT

Hearing loss is one of the top contributors to years lived with disability and is a risk factor for dementia. Molecular evidence on the cellular origins of hearing loss in humans is growing. Here, we performed a genome-wide association meta-analysis of clinically diagnosed and self-reported hearing impairment on 723,266 individuals and identified 48 significant loci, 10 of which are novel. A large proportion of associations comprised missense variants, half of which lie within known familial hearing loss loci. We used single-cell RNA-sequencing data from mouse cochlea and brain and mapped common-variant genomic results to spindle, root, and basal cells from the stria vascularis, a structure in the cochlea necessary for normal hearing. Our findings indicate the importance of the stria vascularis in the mechanism of hearing impairment, providing future paths for developing targets for therapeutic intervention in hearing loss.


Subject(s)
Deafness , Hearing Loss , Animals , Cochlea , Genome-Wide Association Study , Hearing Loss/genetics , Humans , Mice , Stria Vascularis
17.
Hum Genomics ; 18(1): 59, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38844983

ABSTRACT

BACKGROUND: Hereditary hearing loss is a rare hereditary condition that has a significant presence in consanguineous populations. Despite its prevalence, hearing loss is marked by substantial genetic diversity, which poses challenges for diagnosis and screening, particularly in cases with no clear family history or when the impact of the genetic variant requires functional analysis, such as in the case of missense mutations and UTR variants. The advent of next-generation sequencing (NGS) has transformed the identification of genes and variants linked to various conditions, including hearing loss. However, there remains a high proportion of undiagnosed patients, attributable to various factors, including limitations in sequencing coverage and gaps in our knowledge of the entire genome, among other factors. In this study, our objective was to comprehensively identify the spectrum of genes and variants associated with hearing loss in a cohort of 106 affected individuals from the UAE. RESULTS: In this study, we investigated 106 sporadic cases of hearing impairment and performed genetic analyses to identify causative mutations. Screening of the GJB2 gene in these cases revealed its involvement in 24 affected individuals, with specific mutations identified. For individuals without GJB2 mutations, whole exome sequencing (WES) was conducted. WES revealed 33 genetic variants, including 6 homozygous and 27 heterozygous DNA changes, two of which were previously implicated in hearing loss, while 25 variants were novel. We also observed multiple potential pathogenic heterozygous variants across different genes in some cases. Notably, a significant proportion of cases remained without potential pathogenic variants. CONCLUSIONS: Our findings confirm the complex genetic landscape of hearing loss and the limitations of WES in achieving a 100% diagnostic rate, especially in conditions characterized by genetic heterogeneity. These results contribute to our understanding of the genetic basis of hearing loss and emphasize the need for further research and comprehensive genetic analyses to elucidate the underlying causes of this condition.


Subject(s)
Connexin 26 , Exome Sequencing , Hearing Loss , Humans , Male , Female , Hearing Loss/genetics , Hearing Loss/epidemiology , Connexin 26/genetics , Adult , United Arab Emirates/epidemiology , Child , Mutation/genetics , Adolescent , High-Throughput Nucleotide Sequencing , Genetic Testing , Middle Aged , Young Adult , Child, Preschool , Connexins/genetics , Genetic Predisposition to Disease , Heterozygote , Homozygote
18.
Hum Genomics ; 18(1): 73, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956677

ABSTRACT

Knockout of GAS2 (growth arrest-specific protein 2), causes disorganization and destabilization of microtubule bundles in supporting cells of the cochlear duct, leading to hearing loss in vivo. However, the molecular mechanism through which GAS2 variant results in hearing loss remains unknown. By Whole-exome sequencing, we identified a novel heterozygous splicing variant in GAS2 (c.616-2 A > G) as the only candidate mutation segregating with late-onset and progressive nonsyndromic hearing loss (NSHL) in a large dominant family. This splicing mutation causes an intron retention and produces a C-terminal truncated protein (named GAS2mu). Mechanistically, the degradation of GAS2mu via the ubiquitin-proteasome pathway is enhanced, and cells expressing GAS2mu exhibit disorganized microtubule bundles. Additionally, GAS2mu further promotes apoptosis by increasing the Bcl-xS/Bcl-xL ratio instead of through the p53-dependent pathway as wild-type GAS2 does, indicating that GAS2mu acts as a toxic molecule to exacerbate apoptosis. Our findings demonstrate that this novel variant of GAS2 promotes its own protein degradation, microtubule disorganization and cellular apoptosis, leading to hearing loss in carriers. This study expands the spectrum of GAS2 variants and elucidates the underlying pathogenic mechanisms, providing a foundation for future investigations of new therapeutic strategies to prevent GAS2-associated progressive hearing loss.


Subject(s)
Deafness , Pedigree , Adult , Female , Humans , Male , Middle Aged , Apoptosis/genetics , Asian People/genetics , Deafness/genetics , Deafness/pathology , East Asian People/genetics , Exome Sequencing , Genes, Dominant , Microtubules/genetics , Microtubules/metabolism , Mutation/genetics
19.
Cereb Cortex ; 34(3)2024 03 01.
Article in English | MEDLINE | ID: mdl-38494888

ABSTRACT

INTRODUCTION: Previous studies have suggested a correlation between hearing loss (HL) and cortical alterations, but the specific brain regions that may be affected are unknown. METHODS: Genome-wide association study (GWAS) data for 3 subtypes of HL phenotypes, sensorineural hearing loss (SNHL), conductive hearing loss, and mixed hearing loss, were selected as exposures, and GWAS data for brain structure-related traits were selected as outcomes. The inverse variance weighted method was used as the main estimation method. RESULTS: Negative associations were identified between genetically predicted SNHL and brain morphometric indicators (cortical surface area, cortical thickness, or volume of subcortical structures) in specific brain regions, including the bankssts (ß = -0.006 mm, P = 0.016), entorhinal cortex (ß = -4.856 mm2, P = 0.029), and hippocampus (ß = -24.819 cm3, P = 0.045), as well as in brain regions functionally associated with visual perception, including the pericalcarine (ß = -10.009 cm3, P = 0.013). CONCLUSION: Adaptive changes and functional remodeling of brain structures occur in patients with genetically predicted HL. Brain regions functionally associated with auditory perception, visual perception, and memory function are the main brain regions vulnerable in HL.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hearing Loss , Humans , Genome-Wide Association Study , Mendelian Randomization Analysis , Hearing Loss, Sensorineural/diagnostic imaging , Hearing Loss, Sensorineural/genetics
20.
Cereb Cortex ; 34(3)2024 03 01.
Article in English | MEDLINE | ID: mdl-38517177

ABSTRACT

Empathy deficiencies are prevalent among deaf individuals. It has yet to be determined whether they exhibit an ingroup bias in empathic responses. This study employed explicit and implicit empathy tasks (i.e. attention-to-pain-cue [A-P] task and attention-to-nonpain-cue [A-N] task) to explore the temporal dynamics of neural activities when deaf individuals were processing painful/nonpainful stimuli from both ingroup models (deaf people) and outgroup models (hearing people), which aims to not only assist deaf individuals in gaining a deeper understanding of their intergroup empathy traits but also to aid in the advancement of inclusive education. In the A-P task, we found that (i) ingroup priming accelerated the response speed to painful/nonpainful pictures; (ii) the N2 amplitude of painful pictures was significantly more negative than that of nonpainful pictures in outgroup priming trials, whereas the N2 amplitude difference between painful and nonpainful pictures was not significant in ingroup priming trials. For N1 amplitude of the A-N task, we have similar findings. However, this pattern was reversed for P3/late positive component amplitude of the A-P task. These results suggest that the deaf individuals had difficulty in judging whether hearing individuals were in pain. However, their group identification and affective responses could shape the relatively early stage of pain empathy.


Subject(s)
Empathy , Pain , Humans , Pain/psychology , Attention , Reaction Time , Group Processes , Electroencephalography , Evoked Potentials/physiology
SELECTION OF CITATIONS
SEARCH DETAIL