1
|
Zhou Z, Dai J, Chen W, Zhu F, Shen J, Wang P, Zhou X, Fu Y. Targeted sequencing identifies 33 novel mutations in 130 ClinGen curated hearing loss genes among 253 pediatric patients: A retrospective case study. Biomed Rep 2025; 22:100. [PMID: 40322553 PMCID: PMC12046280 DOI: 10.3892/br.2025.1978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Hearing loss (HL) can occur at any age, with hereditary HL being one of the most prevalent congenital disabilities. In the present study, a cohort of pediatric patients with HL was established, comprising 259 individuals at the Children's Hospital of Zhejiang University from 2017-2022. All patients underwent comprehensive diagnostic evaluations, including complete clinical examinations and audiological assessments. Targeted genomic enrichment with massively parallel sequencing was applied to analyze the mutation spectrum of known hearing-loss genes in 253 Chinese children who had positive hearing screening results. Among the 253 patients, 211 (83.40%) exhibited bilateral HL, while 42 (16.60%) had unilateral HL. Targeted sequencing identified 197 variants in 104 genes, yielding a detection rate of 41.1%. A total of 144 genotypes were identified, including 62 heterozygous mutations, 6 hemizygous mutations, 23 homozygous mutations and 48 complex heterozygous mutations. The four most frequently identified genes were GJB2 (26.5%), SLC26A4 (13.5%), MYO15A (6.5%) and USH2A (6.5%). Additionally, 33 novel variants in deafness-associated genes were discovered, comprising 21 novel pathogenic or likely pathogenic variants and 12 variants of uncertain significance. The present results highlight the genetic profile of HL in the Chinese population, with GJB2 being the most prevalent causative gene in early-onset deafness. Furthermore, the current findings provide insight into age- or severity-related gene frequencies for HL. For the genetically unsolved cases, further investigation into digenic inheritance models or other contributing factors is warranted.
Collapse
Affiliation(s)
- Zhiying Zhou
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital Zhejiang University School of Medicine, Key Laboratory for Research on Genetic Defects and Developmental Disorders, Hangzhou, Zhejiang 310051, P.R. China
| | - Jiren Dai
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital Zhejiang University School of Medicine, Key Laboratory for Research on Genetic Defects and Developmental Disorders, Hangzhou, Zhejiang 310051, P.R. China
| | - Wenxin Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital Zhejiang University School of Medicine, Key Laboratory for Research on Genetic Defects and Developmental Disorders, Hangzhou, Zhejiang 310051, P.R. China
| | | | - Jiahua Shen
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital Zhejiang University School of Medicine, Key Laboratory for Research on Genetic Defects and Developmental Disorders, Hangzhou, Zhejiang 310051, P.R. China
| | | | | | - Yong Fu
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital Zhejiang University School of Medicine, Key Laboratory for Research on Genetic Defects and Developmental Disorders, Hangzhou, Zhejiang 310051, P.R. China
| |
Collapse
|
2
|
Saran S, Singh AB, Agrawal A, Misra S, Siddiqui SS, Lohiya A, Misra P. Hearing loss assessment by pure tone audiometry amongst the survivors of intensive care unit: A prospective observational cohort study. J Crit Care 2025; 87:155042. [PMID: 40015086 DOI: 10.1016/j.jcrc.2025.155042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE To assess hearing deficits (HD) through pure tone audiometry (PTA), amongst the survivors of intensive care unit (ICU). METHODS In this prospective observational study, ICU survivors aged 18 years and above were subjected to PTA and were classified into two groups based on PTA findings as those "with HD" and "without HD". Demographic and clinical factors were compared between these groups with a p-value of ≤0.05 considered as significant. RESULTS One hundred and two survivors were enrolled with a median age of 25.5 years (23-30.5), and acute physiology and chronic health evaluation (APACHE II) score of 19 (14-22). Sixty were diagnosed (58.89 %) with HD based on PTA, and forty-two (41.2 %) without HD. More than 80 % of the survivors (52/60) had sensorineural hearing loss (SNHL). Propensity match analysis, after the exact matching of the APACHE-II score between those "with HD" and "without HD", revealed that patients with HD had a longer duration of shock days (mean ± SD) (0.96 ± 1.24 vs 1.68 ± 1.28; p value: 0.022), received higher maximum noradrenaline dose (0.03 v/s 0.06 μg/kg/min; p value: 0.004), longer duration of endotracheal tube (2.04 ± 1.17 vs 3.52 ± 2.06) days; p value:0.009), more days on mechanical ventilation (2.24 ± 1.33 vs 4.44 ± 5.12; p value: 0.011), and length of stay in the ICU (7.2 ± 3.8 vs 9.24 ± 4.68; p value: 0.013) than those without HD. CONCLUSIONS Hearing deficits are present in majority of the survivors of critical illness and audiometric screening is recommended. TRIAL REGISTRATION Clinical trials registry. India (CTRI/2022/01/039539) dated 18.01.2022. https://ctri.nic.in/Clinicaltrials/login.php.
Collapse
Affiliation(s)
- Sai Saran
- Department of Critical Care Medicine, Sanjay Gandhi Post Graduate Medical Sciences (SGPGIMS), Raebareli Road, Lucknow 226014, Uttar Pradesh, India.
| | - Abhishek Bahadur Singh
- Department of Otolaryngology, King George Medical University, Chowk, Lucknow 226003, Uttar Pradesh, India
| | - Avinash Agrawal
- Department of Critical Care Medicine, King George Medical University, Chowk, Lucknow 226003, Uttar Pradesh, India
| | - Saumitra Misra
- Department of Critical Care Medicine, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Suhail Sarwar Siddiqui
- Department of Critical Care Medicine, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Ayush Lohiya
- Department of Public Health, Kalyan Singh Super specialty Cancer Institute & Hospital, Lucknow 226002, Uttar Pradesh, India
| | - Prabhaker Misra
- Department of Biostatistics and Health Information, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| |
Collapse
|
3
|
Bing X, Liu C, Cao X, Li C, Gao X, Zhu F, Wu X, Guo N, Hu H, Xia M, Zhao M. Development of the inner ear and regeneration of hair cells after hearing impairment. FUNDAMENTAL RESEARCH 2025; 5:203-214. [PMID: 40166090 PMCID: PMC11955025 DOI: 10.1016/j.fmre.2023.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/04/2023] [Accepted: 09/06/2023] [Indexed: 04/02/2025] Open
Abstract
Hearing loss, as a sensory disorder, is the most common occurrence among humans, which has received increasing attention from society. It is mainly caused by the damage of inner ear hair cells (HCs) or the degeneration of spiral ganglion neurons. In mammals, cochlear HCs cannot regenerate naturally after injury, leading to irreversible hearing loss. Therefore, HCs are essential for hearing protection. In recent years, the protection of drug-related ototoxicity, inner ear stem cells, gene therapy, new materials, and signal regulation have become important ways to develop regeneration strategies of HCs. An in-depth study of the causes of the occurrence and development of hearing impairment and the regeneration of hearing loss for effective prevention, discovery, and treatment of deafness has great significance. This review aimed to analyze the development of the inner ear and summarize the related factors leading to HCs injury and the research progress of regeneration after injury.
Collapse
Affiliation(s)
- Xin Bing
- Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
| | - Chengcheng Liu
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 271000, China
| | - Xue Cao
- Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
| | - Chengzhilin Li
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 271000, China
| | - Xiaochen Gao
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 271000, China
| | - Fangyuan Zhu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 271000, China
| | - Xinhao Wu
- Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
| | - Na Guo
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 271000, China
| | - Houyang Hu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Department of Otolaryngology, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
- NHC Key Laboratory of Otorhinolaryngology, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 271000, China
| | - Miaoqing Zhao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
4
|
Schubert NMA, Reijntjes DOJ, van Tuinen M, Vijayakumar S, Jones TA, Jones SM, Pyott SJ. Pathophysiological processes underlying hidden hearing loss revealed in Kcnt1/2 double knockout mice. Aging Cell 2024; 23:e14243. [PMID: 39049179 PMCID: PMC11488318 DOI: 10.1111/acel.14243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 07/27/2024] Open
Abstract
Presbycusis is a prevalent condition in older adults characterized by the progressive loss of hearing due to age-related changes in the cochlea, the auditory portion of the inner ear. Many adults also struggle with understanding speech in noise despite having normal auditory thresholds, a condition termed "hidden" hearing loss because it evades standard audiological assessments. Examination of animal models and postmortem human tissue suggests that hidden hearing loss is also associated with age-related changes in the cochlea and may, therefore, precede overt age-related hearing loss. Nevertheless, the pathological mechanisms underlying hidden hearing loss are not understood, which hinders the development of diagnostic biomarkers and effective treatments for age-related hearing loss. To fill these gaps in knowledge, we leveraged a combination of tools, including transcriptomic profiling and morphological and functional assessments, to identify these processes and examine the transition from hidden to overt hearing loss. As a novel approach, we took advantage of a recently characterized model of hidden hearing loss: Kcnt1/2 double knockout mice. Using this model, we find that even before observable morphological pathology, hidden hearing loss is associated with significant alteration in several processes, notably proteostasis, in the cochlear sensorineural structures, and increased susceptibility to overt hearing loss in response to noise exposure and aging. Our findings provide the first insight into the pathophysiology associated with the earliest and, therefore, most treatable stages of hearing loss and provide critical insight directing future investigation of pharmaceutical strategies to slow and possibly prevent overt age-related hearing loss.
Collapse
Affiliation(s)
- Nick M A Schubert
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Graduate School of Medical Sciences Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| | - Daniël O J Reijntjes
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcel van Tuinen
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarath Vijayakumar
- Department of Special Education and Communication Disorders, Barkley Memorial Center, University of Nebraska Lincoln, Lincoln, Nebraska, USA
| | - Timothy A Jones
- Department of Special Education and Communication Disorders, Barkley Memorial Center, University of Nebraska Lincoln, Lincoln, Nebraska, USA
| | - Sherri M Jones
- Department of Special Education and Communication Disorders, Barkley Memorial Center, University of Nebraska Lincoln, Lincoln, Nebraska, USA
| | - Sonja J Pyott
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Graduate School of Medical Sciences Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Tsai CY, Hsu JSJ, Chen PL, Wu CC. Implementing next-generation sequencing for diagnosis and management of hereditary hearing impairment: a comprehensive review. Expert Rev Mol Diagn 2024; 24:753-765. [PMID: 39194060 DOI: 10.1080/14737159.2024.2396866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Sensorineural hearing impairment (SNHI), a common childhood disorder with heterogeneous genetic causes, can lead to delayed language development and psychosocial problems. Next-generation sequencing (NGS) offers high-throughput screening and high-sensitivity detection of genetic etiologies of SNHI, enabling clinicians to make informed medical decisions, provide tailored treatments, and improve prognostic outcomes. AREAS COVERED This review covers the diverse etiologies of HHI and the utility of different NGS modalities (targeted sequencing and whole exome/genome sequencing), and includes HHI-related studies on newborn screening, genetic counseling, prognostic prediction, and personalized treatment. Challenges such as the trade-off between cost and diagnostic yield, detection of structural variants, and exploration of the non-coding genome are also highlighted. EXPERT OPINION In the current landscape of NGS-based diagnostics for HHI, there are both challenges (e.g. detection of structural variants and non-coding genome variants) and opportunities (e.g. the emergence of medical artificial intelligence tools). The authors advocate the use of technological advances such as long-read sequencing for structural variant detection, multi-omics analysis for non-coding variant exploration, and medical artificial intelligence for pathogenicity assessment and outcome prediction. By integrating these innovations into clinical practice, precision medicine in the diagnosis and management of HHI can be further improved.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jacob Shu-Jui Hsu
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| |
Collapse
|
6
|
Semerci Sevimli T, Sevimli M, Ghorbani A, Şahintürk V, Qomi Ekenel E, Ertem T, Demir Cevizlidere B, Altuğ B, Tomsuk Ö, Uysal O, Güneş Bağış S, Avci H, Çemrek F, Ahmadova Z. The analysis of boric acid effect on epithelial-mesenchymal transition of CD133 + CD117 + lung cancer stem cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6791-6802. [PMID: 38536434 PMCID: PMC11422429 DOI: 10.1007/s00210-024-03062-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/18/2024] [Indexed: 09/25/2024]
Abstract
Targeting lung cancer stem cells (LC-SCs) for metastasis may be an effective strategy against lung cancer. This study is the first on epithelial-mesenchymal transition (EMT) properties of boric acid (BA) in LC-SCs. LC-SCs were isolated using the magnetic cell sorting (MACS) method. Tumor-sphere formation and flow cytometry confirmed CSC phenotype. The cytotoxic effect of BA was measured by MTT analysis, and the effect of BA on EMT was examined by migration analysis. The expression levels of ZEB1, SNAIL1, ITGA5, CDH1, ITGB1, VIM, COL1A1, and LAMA5 genes were analyzed by RT-qPCR. E-cadherin, Collagen-1, MMP-3, and Vimentin expressions were analyzed immunohistochemically. Boric acid slightly reduced the migration of cancer cells. Increased expression of transcription factor SNAIL (p < 0.001), but not ZEB1, was observed in LC-SCs. mRNA expression levels of ITGB1 (p < 0.01), ITGA5 (p < 0.001), COL1A1 (p < 0.001), and LAMA5 (p < 0.001) increased; CDH1 and VIM decreased in LC-SCs. Moreover, while E-cadherin (p < 0.001) and Collagen-1 (p < 0.01) immunoreactivities significantly increased, MMP-3 (p < 0.001) and Vimentin (p < 0.01) immunoreactivities decreased in BA-treated LC-SCs. To conclude, the current study provided insights into the efficacy and effects of BA against LC-SCs regarding proliferation, EMT, and cell death for future studies.
Collapse
Affiliation(s)
- Tuğba Semerci Sevimli
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| | - Murat Sevimli
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Aynaz Ghorbani
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Varol Şahintürk
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Emilia Qomi Ekenel
- Graduate School of İnformatics, Middle East Technical University, Ankara, Turkey
| | - Tuğba Ertem
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Bahar Demir Cevizlidere
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Burcugül Altuğ
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Özlem Tomsuk
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Onur Uysal
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Sibel Güneş Bağış
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Hüseyin Avci
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Fatih Çemrek
- Department of Statistics, Faculty of Science and Letters, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Zarifa Ahmadova
- Stem Cell, Cellular Therapy, and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| |
Collapse
|
7
|
Polesskaya O, Boussaty E, Cheng R, Lamonte O, Zhou T, Du E, Sanches TM, Nguyen KM, Okamoto M, Palmer AA, Friedman R. Genome-wide association study for age-related hearing loss in CFW mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598304. [PMID: 38915500 PMCID: PMC11195089 DOI: 10.1101/2024.06.10.598304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Age-related hearing impairment is the most common cause of hearing loss and is one of the most prevalent conditions affecting the elderly globally. It is influenced by a combination of environmental and genetic factors. The mouse and human inner ears are functionally and genetically homologous. Investigating the genetic basis of age-related hearing loss (ARHL) in an outbred mouse model may lead to a better understanding of the molecular mechanisms of this condition. We used Carworth Farms White (CFW) outbred mice, because they are genetically diverse and exhibit variation in the onset and severity of ARHL. The goal of this study was to identify genetic loci involved in regulating ARHL. Hearing at a range of frequencies was measured using Auditory Brainstem Response (ABR) thresholds in 946 male and female CFW mice at the age of 1, 6, and 10 months. We obtained genotypes at 4.18 million single nucleotide polymorphisms (SNP) using low-coverage (mean coverage 0.27x) whole-genome sequencing followed by imputation using STITCH. To determine the accuracy of the genotypes we sequenced 8 samples at >30x coverage and used calls from those samples to estimate the discordance rate, which was 0.45%. We performed genetic analysis for the ABR thresholds for each frequency at each age, and for the time of onset of deafness for each frequency. The SNP heritability ranged from 0 to 42% for different traits. Genome-wide association analysis identified several regions associated with ARHL that contained potential candidate genes, including Dnah11, Rapgef5, Cpne4, Prkag2, and Nek11. We confirmed, using functional study, that Prkag2 deficiency causes age-related hearing loss at high frequency in mice; this makes Prkag2 a candidate gene for further studies. This work helps to identify genetic risk factors for ARHL and to define novel therapeutic targets for the treatment and prevention of ARHL.
Collapse
Affiliation(s)
- Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ely Boussaty
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Olivia Lamonte
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Thomas Zhou
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Eric Du
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mika Okamoto
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rick Friedman
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
8
|
Cai M, Zheng Q, Chen Y, Liu S, Zhu H, Bai B. Insights from the neural guidance factor Netrin-1 into neurodegeneration and other diseases. Front Mol Neurosci 2024; 17:1379726. [PMID: 38638604 PMCID: PMC11024333 DOI: 10.3389/fnmol.2024.1379726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Netrin-1 was initially discovered as a neuronal growth cue for axonal guidance, and its functions have later been identified in inflammation, tumorigenesis, neurodegeneration, and other disorders. We have recently found its alterations in the brains with Alzheimer's disease, which might provide important clues to the mechanisms of some unique pathologies. To provide better understanding of this promising molecule, we here summarize research progresses in genetics, pathology, biochemistry, cell biology and other studies of Netrin-1 about its mechanistic roles and biomarker potentials with an emphasis on clinical neurodegenerative disorders in order to expand understanding of this promising molecular player in human diseases.
Collapse
Affiliation(s)
- Minqi Cai
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| | - Qian Zheng
- Health Management Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yiqiang Chen
- Center for Precision Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Siyuan Liu
- Center for Precision Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Huimin Zhu
- Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
| | - Bing Bai
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Lewis MA, Schulte J, Matthews L, Vaden KI, Steves CJ, Williams FMK, Schulte BA, Dubno JR, Steel KP. Accurate phenotypic classification and exome sequencing allow identification of novel genes and variants associated with adult-onset hearing loss. PLoS Genet 2023; 19:e1011058. [PMID: 38011198 PMCID: PMC10718637 DOI: 10.1371/journal.pgen.1011058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/13/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023] Open
Abstract
Adult-onset progressive hearing loss is a common, complex disease with a strong genetic component. Although to date over 150 genes have been identified as contributing to human hearing loss, many more remain to be discovered, as does most of the underlying genetic diversity. Many different variants have been found to underlie adult-onset hearing loss, but they tend to be rare variants with a high impact upon the gene product. It is likely that combinations of more common, lower impact variants also play a role in the prevalence of the disease. Here we present our exome study of hearing loss in a cohort of 532 older adult volunteers with extensive phenotypic data, including 99 older adults with normal hearing, an important control set. Firstly, we carried out an outlier analysis to identify genes with a high variant load in older adults with hearing loss compared to those with normal hearing. Secondly, we used audiometric threshold data to identify individual variants which appear to contribute to different threshold values. We followed up these analyses in a second cohort. Using these approaches, we identified genes and variants linked to better hearing as well as those linked to worse hearing. These analyses identified some known deafness genes, demonstrating proof of principle of our approach. However, most of the candidate genes are novel associations with hearing loss. While the results support the suggestion that genes responsible for severe deafness may also be involved in milder hearing loss, they also suggest that there are many more genes involved in hearing which remain to be identified. Our candidate gene lists may provide useful starting points for improved diagnosis and drug development.
Collapse
Affiliation(s)
- Morag A. Lewis
- Wolfson Centre for Age-Related Diseases, King’s College London, United Kingdom
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jennifer Schulte
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Lois Matthews
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kenneth I. Vaden
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Claire J. Steves
- Department of Twin Research and Genetic Epidemiology, King’s College London, School of Life Course and Population Sciences, London, United Kingdom
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, King’s College London, School of Life Course and Population Sciences, London, United Kingdom
| | - Bradley A. Schulte
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Judy R. Dubno
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King’s College London, United Kingdom
- The Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
10
|
Borgese N, Guillén-Samander A, Colombo SF, Mancassola G, Di Berardino F, Zanetti D, Carrera P. Combined Presence in Heterozygosis of Two Variant Usher Syndrome Genes in Two Siblings Affected by Isolated Profound Age-Related Hearing Loss. Biomedicines 2023; 11:2657. [PMID: 37893031 PMCID: PMC10604119 DOI: 10.3390/biomedicines11102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Sensorineural age-related hearing loss affects a large proportion of the elderly population, and has both environmental and genetic causes. Notwithstanding increasing interest in this debilitating condition, the genetic risk factors remain largely unknown. Here, we report the case of two sisters affected by isolated profound sensorineural hearing loss after the age of seventy. Genomic DNA sequencing revealed that the siblings shared two monoallelic variants in two genes linked to Usher Syndrome (USH genes), a recessive disorder of the ear and the retina: a rare pathogenic truncating variant in USH1G and a previously unreported missense variant in ADGRV1. Structure predictions suggest a negative effect on protein stability of the latter variant, allowing its classification as likely pathogenic according to American College of Medical Genetics criteria. Thus, the presence in heterozygosis of two recessive alleles, which each cause syndromic deafness, may underlie digenic inheritance of the age-related non-syndromic hearing loss of the siblings, a hypothesis that is strengthened by the knowledge that the two genes are integrated in the same functional network, which underlies stereocilium development and organization. These results enlarge the spectrum and complexity of the phenotypic consequences of USH gene mutations beyond the simple Mendelian inheritance of classical Usher syndrome.
Collapse
Affiliation(s)
- Nica Borgese
- Consiglio Nazionale delle Ricerche Neuroscience Institute, 20854 Vedano al Lambro, Italy;
| | | | - Sara Francesca Colombo
- Consiglio Nazionale delle Ricerche Neuroscience Institute, 20854 Vedano al Lambro, Italy;
- NeuroMi Milan Center for Neuroscience, Milan, Italy
| | - Giulia Mancassola
- Unit of Genomics for Human Disease Diagnosis, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
- Laboratory of Clinical Molecular Genetics, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Federica Di Berardino
- Audiology Unit, Department of Specialistic Surgical Sciences, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.D.B.); (D.Z.)
- Department of Clinical Sciences and Community Health, University of Milano, 20122 Milan, Italy
| | - Diego Zanetti
- Audiology Unit, Department of Specialistic Surgical Sciences, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.D.B.); (D.Z.)
- Department of Clinical Sciences and Community Health, University of Milano, 20122 Milan, Italy
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
- Laboratory of Clinical Molecular Genetics, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
11
|
Azeem A, Julleekeea A, Knight B, Sohail I, Bruyns-Haylett M, Sastre M. Hearing loss and its link to cognitive impairment and dementia. FRONTIERS IN DEMENTIA 2023; 2:1199319. [PMID: 39081997 PMCID: PMC11285555 DOI: 10.3389/frdem.2023.1199319] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/26/2023] [Indexed: 08/02/2024]
Abstract
Hearing loss is an important risk factor for the development of dementia, particularly Alzheimer's disease (AD). Mid-life hearing loss increases the risk of developing dementia by double any other single factor. However, given this strong connection between hearing loss and AD, the mechanisms responsible for this link are still unknown. Data from observational studies relating hearing loss and cognitive impairment, measured with standardized questionnaires, has shown a strong relationship between them. Similar findings have emerged from animal studies, showing that the induction of hearing loss via prolonged loud sound exposure or ear canal blocking, can impair cognitive abilities. Interestingly, patients with age-related hearing impairment exhibit increased phosphorylated tau in the cerebrospinal fluid, but no such relationship has been identified for amyloid-β. In addition, hearing loss predisposes to social isolation precipitating the development of dementia through a supposed reduction in cognitive load and processing requirements. Given this link between hearing loss and dementia, the question arises whether the restoration of hearing might mitigate against the onset or progress of AD. Indeed, there is a growing body of research that suggests that those who wear hearing aids for age-related hearing problems maintain better cognitive function over time than those who do not. These are compelling findings, as they suggest the use of hearing aids has the potential to be a cost-effective treatment for those with hearing loss both prior (for those at high risk for AD) and after the development of symptoms. This review aims to summarize the current theories that relate hearing loss and cognitive decline, present the key findings of animal studies, observational studies and summarize the gaps and limitations that need to be addressed in this topic. Through this, we suggest directions for future studies to tackle the lack of adequately randomized control trials in the field. This omission is responsible for the inability to provide a conclusive verdict on whether to use hearing interventions to target hearing-loss related cognitive decline.
Collapse
Affiliation(s)
- Abdul Azeem
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Arun Julleekeea
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Beth Knight
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Isha Sohail
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | | | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
12
|
Hui D, Mehrabi S, Quimby AE, Chen T, Chen S, Park J, Li B, Ruckenstein MJ, Rader DJ, Ritchie MD, Brant JA, Epstein DJ, Mathieson I. Gene burden analysis identifies genes associated with increased risk and severity of adult-onset hearing loss in a diverse hospital-based cohort. PLoS Genet 2023; 19:e1010584. [PMID: 36656851 PMCID: PMC9888707 DOI: 10.1371/journal.pgen.1010584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/31/2023] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Loss or absence of hearing is common at both extremes of human lifespan, in the forms of congenital deafness and age-related hearing loss. While these are often studied separately, there is increasing evidence that their genetic basis is at least partially overlapping. In particular, both common and rare variants in genes associated with monogenic forms of hearing loss also contribute to the more polygenic basis of age-related hearing loss. Here, we directly test this model in the Penn Medicine BioBank-a healthcare system cohort of around 40,000 individuals with linked genetic and electronic health record data. We show that increased burden of predicted deleterious variants in Mendelian hearing loss genes is associated with increased risk and severity of adult-onset hearing loss. As a specific example, we identify one gene-TCOF1, responsible for a syndromic form of congenital hearing loss-in which deleterious variants are also associated with adult-onset hearing loss. We also identify four additional novel candidate genes (COL5A1, HMMR, RAPGEF3, and NNT) in which rare variant burden may be associated with hearing loss. Our results confirm that rare variants in Mendelian hearing loss genes contribute to polygenic risk of hearing loss, and emphasize the utility of healthcare system cohorts to study common complex traits and diseases.
Collapse
Affiliation(s)
- Daniel Hui
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Shadi Mehrabi
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alexandra E. Quimby
- Department of Otolaryngology–Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tingfang Chen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sixing Chen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joseph Park
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Binglan Li
- Department of Biomedical Data Science, Stanford University, Stanford, California, United States of America
| | | | - Penn Medicine Biobank
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael J. Ruckenstein
- Department of Otolaryngology–Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel J. Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jason A. Brant
- Department of Otolaryngology–Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Otolaryngology–Head and Neck Surgery, Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, United States of America
| | - Douglas J. Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (DJE); (IM)
| | - Iain Mathieson
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (DJE); (IM)
| |
Collapse
|
13
|
Pouyo R, Chung K, Delacroix L, Malgrange B. The ubiquitin-proteasome system in normal hearing and deafness. Hear Res 2022; 426:108366. [PMID: 34645583 DOI: 10.1016/j.heares.2021.108366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/03/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022]
Abstract
Post-translational modifications of proteins are essential for the proper development and function of many tissues and organs, including the inner ear. Ubiquitination is a highly selective post-translational modification that involves the covalent conjugation of ubiquitin to a substrate protein. The most common outcome of protein ubiquitination is degradation by the ubiquitin-proteasome system (UPS), preventing the accumulation of misfolded, damaged, and excess proteins. In addition to proteasomal degradation, ubiquitination regulates other cellular processes, such as transcription, translation, endocytosis, receptor activity, and subcellular localization. All of these processes are essential for cochlear development and maintenance, as several studies link impairment of UPS with altered cochlear development and hearing loss. In this review, we provide insight into the well-oiled machinery of UPS with a focus on its confirmed role in normal hearing and deafness and potential therapeutic strategies to prevent and treat UPS-associated hearing loss.
Collapse
Affiliation(s)
- Ronald Pouyo
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium
| | - Keshi Chung
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium
| | - Laurence Delacroix
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium
| | - Brigitte Malgrange
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium.
| |
Collapse
|
14
|
Outer Hair Cell Function is Normal in βV Spectrin Knockout Mice. Hear Res 2022; 423:108564. [DOI: 10.1016/j.heares.2022.108564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022]
|
15
|
Population-scale analysis of common and rare genetic variation associated with hearing loss in adults. Commun Biol 2022; 5:540. [PMID: 35661827 PMCID: PMC9166757 DOI: 10.1038/s42003-022-03408-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/23/2022] [Indexed: 12/29/2022] Open
Abstract
To better understand the genetics of hearing loss, we performed a genome-wide association meta-analysis with 125,749 cases and 469,497 controls across five cohorts. We identified 53/c loci affecting hearing loss risk, including common coding variants in COL9A3 and TMPRSS3. Through exome sequencing of 108,415 cases and 329,581 controls, we observed rare coding associations with 11 Mendelian hearing loss genes, including additive effects in known hearing loss genes GJB2 (Gly12fs; odds ratio [OR] = 1.21, P = 4.2 × 10-11) and SLC26A5 (gene burden; OR = 1.96, P = 2.8 × 10-17). We also identified hearing loss associations with rare coding variants in FSCN2 (OR = 1.14, P = 1.9 × 10-15) and KLHDC7B (OR = 2.14, P = 5.2 × 10-30). Our results suggest a shared etiology between Mendelian and common hearing loss in adults. This work illustrates the potential of large-scale exome sequencing to elucidate the genetic architecture of common disorders where both common and rare variation contribute to risk.
Collapse
|
16
|
Wonkam A, Adadey SM, Schrauwen I, Aboagye ET, Wonkam-Tingang E, Esoh K, Popel K, Manyisa N, Jonas M, deKock C, Nembaware V, Cornejo Sanchez DM, Bharadwaj T, Nasir A, Everard JL, Kadlubowska MK, Nouel-Saied LM, Acharya A, Quaye O, Amedofu GK, Awandare GA, Leal SM. Exome sequencing of families from Ghana reveals known and candidate hearing impairment genes. Commun Biol 2022; 5:369. [PMID: 35440622 PMCID: PMC9019055 DOI: 10.1038/s42003-022-03326-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022] Open
Abstract
We investigated hearing impairment (HI) in 51 families from Ghana with at least two affected members that were negative for GJB2 pathogenic variants. DNA samples from 184 family members underwent whole-exome sequencing (WES). Variants were found in 14 known non-syndromic HI (NSHI) genes [26/51 (51.0%) families], five genes that can underlie either syndromic HI or NSHI [13/51 (25.5%)], and one syndromic HI gene [1/51 (2.0%)]. Variants in CDH23 and MYO15A contributed the most to HI [31.4% (16/51 families)]. For DSPP, an autosomal recessive mode of inheritance was detected. Post-lingual expression was observed for a family segregating a MARVELD2 variant. To our knowledge, seven novel candidate HI genes were identified (13.7%), with six associated with NSHI (INPP4B, CCDC141, MYO19, DNAH11, POTEI, and SOX9); and one (PAX8) with Waardenburg syndrome. MYO19 and DNAH11 were replicated in unrelated Ghanaian probands. Six of the novel genes were expressed in mouse inner ear. It is known that Pax8-/- mice do not respond to sound, and depletion of Sox9 resulted in defective vestibular structures and abnormal utricle development. Most variants (48/60; 80.0%) have not previously been associated with HI. Identifying seven candidate genes in this study emphasizes the potential of novel HI genes discovery in Africa.
Collapse
Affiliation(s)
- Ambroise Wonkam
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Samuel Mawuli Adadey
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, LG 54, Ghana
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Elvis Twumasi Aboagye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, LG 54, Ghana
| | - Edmond Wonkam-Tingang
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Kevin Esoh
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Kalinka Popel
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Noluthando Manyisa
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Mario Jonas
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Carmen deKock
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Victoria Nembaware
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Diana M Cornejo Sanchez
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Thashi Bharadwaj
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Abdul Nasir
- Department of Molecular Science and Technology, Ajou University, Suwon-si, Republic of Korea
| | - Jenna L Everard
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Magda K Kadlubowska
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Liz M Nouel-Saied
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Anushree Acharya
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, LG 54, Ghana
| | - Geoffrey K Amedofu
- Department of Eye, Ear, Nose, and Throat, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, LG 54, Ghana
| | - Suzanne M Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Centre, New York, NY, 10032, USA.
- Taub Institute for Alzheimer's Disease and the Aging Brain, Columbia University Medical Centre, New York, NY, 10032, USA.
| |
Collapse
|
17
|
Boussaty EC, Friedman RA, Clifford RE. Hearing loss and tinnitus: association studies for complex-hearing disorders in mouse and man. Hum Genet 2022; 141:981-990. [PMID: 34318347 PMCID: PMC8792513 DOI: 10.1007/s00439-021-02317-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/13/2021] [Indexed: 12/29/2022]
Abstract
Genome-wide association studies (GWAS) provide an unbiased first look at genetic loci involved in aging and noise-induced sensorineural hearing loss and tinnitus. The hearing phenotype, whether audiogram-based or self-report, is regressed against genotyped information at representative single nucleotide polymorphisms (SNPs) across the genome. Findings include the fact that both hearing loss and tinnitus are polygenic disorders, with up to thousands of genes, each of effect size of < 0.02. Smaller human GWAS' were able to use objective measures and identified a few loci; however, hundreds of thousands of participants have been required for the statistical power to identify significant variants, and GWAS is unable to assess rare variants with mean allele frequency < 1%. Animal studies are required as well because of inability to access the human cochlea. Mouse GWAS builds on linkage techniques and the known phenotypic differences in auditory function between inbred strains. With the advantage that the laboratory environment can be controlled for noise and aging, the Hybrid Mouse Diversity Panel (HDMP) combines 100 strains sequenced at high resolution. Lift-over regions between mice and humans have identified over 17,000 homologous genes. Since most significant SNPs are either intergenic or in introns, and binding sites between species are poorly preserved between species, expression quantitative trait locus information is required to bring humans and mice into agreement. Transcriptome-wide analysis studies (TWAS) can prioritize putative causal genes and tissues. Diverse species, each making a distinct contribution, carry a synergistic advantage in the quest for treatment and ultimate cure of sensorineural hearing difficulties.
Collapse
Affiliation(s)
- Ely Cheikh Boussaty
- School of Health Sciences, Division of Otolaryngology, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Rick Adam Friedman
- School of Health Sciences, Division of Otolaryngology, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Royce E Clifford
- School of Health Sciences, Division of Otolaryngology, University of California San Diego, La Jolla, San Diego, CA, USA.
- Research Department, VA Hospitals San Diego, San Diego, CA, USA.
- Visiting Scientist, Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA.
| |
Collapse
|
18
|
Transcriptome-Guided Identification of Drugs for Repurposing to Treat Age-Related Hearing Loss. Biomolecules 2022; 12:biom12040498. [PMID: 35454087 PMCID: PMC9028743 DOI: 10.3390/biom12040498] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Age-related hearing loss (ARHL) or presbycusis is a prevalent condition associated with social isolation, cognitive impairment, and dementia. Age-related changes in the cochlea, the auditory portion of the inner ear, are the primary cause of ARHL. Unfortunately, there are currently no pharmaceutical approaches to treat ARHL. To examine the biological processes underlying age-related changes in the cochlea and identify candidate drugs for rapid repurposing to treat ARHL, we utilized bulk RNA sequencing to obtain transcriptomes from the functional substructures of the cochlea—the sensorineural structures, including the organ of Corti and spiral ganglion neurons (OC/SGN) and the stria vascularis and spiral ligament (SV/SL)—in young (6-week-old) and old (2-year-old) C57BL/6 mice. Transcriptomic analyses revealed both overlapping and unique patterns of gene expression and gene enrichment between substructures and with ageing. Based on these age-related transcriptional changes, we queried the protein products of genes differentially expressed with ageing in DrugBank and identified 27 FDA/EMA-approved drugs that are suitable to be repurposed to treat ARHL. These drugs target the protein products of genes that are differentially expressed with ageing uniquely in either the OC/SGN or SV/SL and that interrelate diverse biological processes. Further transcriptomic analyses revealed that most genes differentially expressed with ageing in both substructures encode protein products that are promising drug target candidates but are, nevertheless, not yet linked to approved drugs. Thus, with this study, we apply a novel approach to characterize the druggable genetic landscape for ARHL and propose a list of drugs to test in pre-clinical studies as potential treatment options for ARHL.
Collapse
|
19
|
Elliott KL, Fritzsch B, Yamoah EN, Zine A. Age-Related Hearing Loss: Sensory and Neural Etiology and Their Interdependence. Front Aging Neurosci 2022; 14:814528. [PMID: 35250542 PMCID: PMC8891613 DOI: 10.3389/fnagi.2022.814528] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is a common, increasing problem for older adults, affecting about 1 billion people by 2050. We aim to correlate the different reductions of hearing from cochlear hair cells (HCs), spiral ganglion neurons (SGNs), cochlear nuclei (CN), and superior olivary complex (SOC) with the analysis of various reasons for each one on the sensory deficit profiles. Outer HCs show a progressive loss in a basal-to-apical gradient, and inner HCs show a loss in a apex-to-base progression that results in ARHL at high frequencies after 70 years of age. In early neonates, SGNs innervation of cochlear HCs is maintained. Loss of SGNs results in a considerable decrease (~50% or more) of cochlear nuclei in neonates, though the loss is milder in older mice and humans. The dorsal cochlear nuclei (fusiform neurons) project directly to the inferior colliculi while most anterior cochlear nuclei reach the SOC. Reducing the number of neurons in the medial nucleus of the trapezoid body (MNTB) affects the interactions with the lateral superior olive to fine-tune ipsi- and contralateral projections that may remain normal in mice, possibly humans. The inferior colliculi receive direct cochlear fibers and second-order fibers from the superior olivary complex. Loss of the second-order fibers leads to hearing loss in mice and humans. Although ARHL may arise from many complex causes, HC degeneration remains the more significant problem of hearing restoration that would replace the cochlear implant. The review presents recent findings of older humans and mice with hearing loss.
Collapse
Affiliation(s)
- Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA, United States
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, United States
- *Correspondence: Bernd Fritzsch
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, Montpellier, France
| |
Collapse
|
20
|
Zhou C, Xiao Y, Xie H, Wang J, Liu S. Case Report: Novel Compound Heterozygous Variants in TRIOBP Associated With Congenital Deafness in a Chinese Family. Front Genet 2021; 12:766973. [PMID: 34868251 PMCID: PMC8635749 DOI: 10.3389/fgene.2021.766973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022] Open
Abstract
Autosomal recessive non-syndromic deafness-28 (DFNB28) is characterized by prelingual, profound sensorineural hearing loss (HL). The disease is related to variants of the TRIOBP gene. TRIO and F-actin binding protein (TRIOBP) plays crucial roles in modulating the assembly of the actin cytoskeleton and are responsible for the proper structure and function of stereocilia in the inner ear. This study aimed to identify pathogenic variants in a patient with HL. Genomic DNA obtained from a 33-year-old woman with HL was evaluated using a disease-targeted gene panel. Using next generation sequencing and bioinformatics analysis, we identified two novel TRIOBP c.1170delC (p.S391Pfs*488) and c.3764C > G (p.S1255*) variants. Both parents of the patient were heterozygous carriers of the gene. The two variants have not been reported in general population databases or published literature. The findings of this study will broaden the spectrum of pathogenic variants in the TRIOBP gene.
Collapse
Affiliation(s)
- Cong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yuanyuan Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Hanbing Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Shanling Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Paplou V, Schubert NMA, Pyott SJ. Age-Related Changes in the Cochlea and Vestibule: Shared Patterns and Processes. Front Neurosci 2021; 15:680856. [PMID: 34539328 PMCID: PMC8446668 DOI: 10.3389/fnins.2021.680856] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
Both age-related hearing loss (ARHL) and age-related loss in vestibular function (ARVL) are prevalent conditions with deleterious consequences on the health and quality of life. Age-related changes in the inner ear are key contributors to both conditions. The auditory and vestibular systems rely on a shared sensory organ - the inner ear - and, like other sensory organs, the inner ear is susceptible to the effects of aging. Despite involvement of the same sensory structure, ARHL and ARVL are often considered separately. Insight essential for the development of improved diagnostics and treatments for both ARHL and ARVL can be gained by careful examination of their shared and unique pathophysiology in the auditory and vestibular end organs of the inner ear. To this end, this review begins by comparing the prevalence patterns of ARHL and ARVL. Next, the normal and age-related changes in the structure and function of the auditory and vestibular end organs are compared. Then, the contributions of various molecular mechanisms, notably inflammaging, oxidative stress, and genetic factors, are evaluated as possible common culprits that interrelate pathophysiology in the cochlea and vestibular end organs as part of ARHL and ARVL. A careful comparison of these changes reveals that the patterns of pathophysiology show similarities but also differences both between the cochlea and vestibular end organs and among the vestibular end organs. Future progress will depend on the development and application of new research strategies and the integrated investigation of ARHL and ARVL using both clinical and animal models.
Collapse
Affiliation(s)
- Vasiliki Paplou
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nick M A Schubert
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Research School of Behavioural and Cognitive Neurosciences, Graduate School of Medical Sciences, University of Groningen, Groningen, Netherlands
| | - Sonja J Pyott
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Research School of Behavioural and Cognitive Neurosciences, Graduate School of Medical Sciences, University of Groningen, Groningen, Netherlands
| |
Collapse
|
22
|
Van Heurck R, Carminho-Rodrigues MT, Ranza E, Stafuzza C, Quteineh L, Gehrig C, Hammar E, Guipponi M, Abramowicz M, Senn P, Guinand N, Cao-Van H, Paoloni-Giacobino A. Benefits of Exome Sequencing in Children with Suspected Isolated Hearing Loss. Genes (Basel) 2021; 12:genes12081277. [PMID: 34440452 PMCID: PMC8391342 DOI: 10.3390/genes12081277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/03/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose: Hearing loss is characterized by an extensive genetic heterogeneity and remains a common disorder in children. Molecular diagnosis is of particular benefit in children, and permits the early identification of clinically-unrecognized hearing loss syndromes, which permits effective clinical management and follow-up, including genetic counselling. Methods: We performed whole-exome sequencing with the analysis of a panel of 189 genes associated with hearing loss in a prospective cohort of 61 children and 9 adults presenting mainly with isolated hearing loss. Results: The overall diagnostic rate using exome sequencing was 47.2% (52.5% in children; 22% in adults). In children with confirmed molecular results, 17/32 (53.2%) showed autosomal recessive inheritance patterns, 14/32 (43.75%) showed an autosomal dominant condition, and one case had X-linked hearing loss. In adults, the two patients showed an autosomal dominant inheritance pattern. Among the 32 children, 17 (53.1%) had nonsyndromic hearing loss and 15 (46.7%) had syndromic hearing loss. One adult was diagnosed with syndromic hearing loss and one with nonsyndromic hearing loss. The most common causative genes were STRC (5 cases), GJB2 (3 cases), COL11A1 (3 cases), and ACTG1 (3 cases). Conclusions: Exome sequencing has a high diagnostic yield in children with hearing loss and can reveal a syndromic hearing loss form before other organs/systems become involved, allowing the surveillance of unrecognized present and/or future complications associated with these syndromes.
Collapse
Affiliation(s)
- Roxane Van Heurck
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Maria Teresa Carminho-Rodrigues
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Emmanuelle Ranza
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Caterina Stafuzza
- Ear-Nose-Throat/Head and Neck Surgery Division, Geneva University Hospitals, 1205 Geneva, Switzerland; (C.S.); (P.S.); (N.G.); (H.C.-V.)
| | - Lina Quteineh
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Corinne Gehrig
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Eva Hammar
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Michel Guipponi
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Marc Abramowicz
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
| | - Pascal Senn
- Ear-Nose-Throat/Head and Neck Surgery Division, Geneva University Hospitals, 1205 Geneva, Switzerland; (C.S.); (P.S.); (N.G.); (H.C.-V.)
| | - Nils Guinand
- Ear-Nose-Throat/Head and Neck Surgery Division, Geneva University Hospitals, 1205 Geneva, Switzerland; (C.S.); (P.S.); (N.G.); (H.C.-V.)
| | - Helene Cao-Van
- Ear-Nose-Throat/Head and Neck Surgery Division, Geneva University Hospitals, 1205 Geneva, Switzerland; (C.S.); (P.S.); (N.G.); (H.C.-V.)
| | - Ariane Paoloni-Giacobino
- Division of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland; (R.V.H.); (M.T.C.-R.); (E.R.); (L.Q.); (C.G.); (E.H.); (M.G.); (M.A.)
- Correspondence:
| |
Collapse
|
23
|
Ahmadmehrabi S, Li B, Hui D, Park J, Ritchie M, Rader DJ, Ruckenstein MJ, Epstein DJ, Brant J. A Genome-First Approach to Rare Variants in Dominant Postlingual Hearing Loss Genes in a Large Adult Population. Otolaryngol Head Neck Surg 2021; 166:746-752. [PMID: 34281439 DOI: 10.1177/01945998211029544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To investigate the importance of rare variants in adult-onset hearing loss. STUDY DESIGN Genomic association study. SETTING Large biobank from tertiary care center. METHODS We investigated rare variants (minor allele frequency <5%) in 42 autosomal dominant (DFNA) postlingual hearing loss (HL) genes in 16,657 unselected individuals in the Penn Medicine Biobank. We determined the prevalence of known pathogenic and predicted deleterious variants in subjects with audiometric-proven sensorineural hearing loss. We scanned across known postlingual DFNA HL genes to determine those most significantly contributing to the phenotype. We replicated findings in an independent cohort (UK Biobank). RESULTS While rare individually, when considering the accumulation of variants in all postlingual DFNA genes, more than 90% of participants carried at least 1 rare variant. Rare variants predicted to be deleterious were enriched in adults with audiometric-proven hearing loss (pure-tone average >25 dB; P = .015). Patients with a rare predicted deleterious variant had an odds ratio of 1.27 for HL compared with genotypic controls (P = .029). Gene burden in DIABLO, PTPRQ, TJP2, and POU4F3 were independently associated with sensorineural hearing loss. CONCLUSION Although prior reports have focused on common variants, we find that rare predicted deleterious variants in DFNA postlingual HL genes are enriched in patients with adult-onset HL in a large health care system population. We show the value of investigating rare variants to uncover hearing loss phenotypes related to implicated genes.
Collapse
Affiliation(s)
- Shadi Ahmadmehrabi
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Binglan Li
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Hui
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph Park
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marylyn Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael J Ruckenstein
- Department of Otorhinolaryngology Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas J Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason Brant
- Department of Otorhinolaryngology Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Eckert MA, Harris KC, Lang H, Lewis MA, Schmiedt RA, Schulte BA, Steel KP, Vaden KI, Dubno JR. Translational and interdisciplinary insights into presbyacusis: A multidimensional disease. Hear Res 2021; 402:108109. [PMID: 33189490 PMCID: PMC7927149 DOI: 10.1016/j.heares.2020.108109] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
There are multiple etiologies and phenotypes of age-related hearing loss or presbyacusis. In this review we summarize findings from animal and human studies of presbyacusis, including those that provide the theoretical framework for distinct metabolic, sensory, and neural presbyacusis phenotypes. A key finding in quiet-aged animals is a decline in the endocochlear potential (EP) that results in elevated pure-tone thresholds across frequencies with greater losses at higher frequencies. In contrast, sensory presbyacusis appears to derive, in part, from acute and cumulative effects on hair cells of a lifetime of environmental exposures (e.g., noise), which often result in pronounced high frequency hearing loss. These patterns of hearing loss in animals are recognizable in the human audiogram and can be classified into metabolic and sensory presbyacusis phenotypes, as well as a mixed metabolic+sensory phenotype. However, the audiogram does not fully characterize age-related changes in auditory function. Along with the effects of peripheral auditory system declines on the auditory nerve, primary degeneration in the spiral ganglion also appears to contribute to central auditory system aging. These inner ear alterations often correlate with structural and functional changes throughout the central nervous system and may explain suprathreshold speech communication difficulties in older adults with hearing loss. Throughout this review we highlight potential methods and research directions, with the goal of advancing our understanding, prevention, diagnosis, and treatment of presbyacusis.
Collapse
Affiliation(s)
- Mark A Eckert
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA.
| | - Kelly C Harris
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Hainan Lang
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| | - Morag A Lewis
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Richard A Schmiedt
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Bradley A Schulte
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Karen P Steel
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Kenneth I Vaden
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Judy R Dubno
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| |
Collapse
|
25
|
Oluwole OG, Esoh KK, Wonkam-Tingang E, Manyisa N, Noubiap JJ, Chimusa ER, Wonkam A. Whole exome sequencing identifies rare coding variants in novel human-mouse ortholog genes in African individuals diagnosed with non-syndromic hearing impairment. Exp Biol Med (Maywood) 2021; 246:197-206. [PMID: 32996353 PMCID: PMC7871117 DOI: 10.1177/1535370220960388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/28/2020] [Indexed: 01/08/2023] Open
Abstract
Physiologically, the human and murine hearing systems are very similar, justifying the extensive use of mice in experimental models for hearing impairment (HI). About 340 murine HI genes have been reported; however, whether variants in all human-mouse ortholog genes contribute to HI has been rarely investigated. In humans, nearly 120 HI genes have been identified to date, with GJB2 and GJB6 variants accounting for half of congenital HI cases, of genetic origin, in populations of European and Asian ancestries, but not in most African populations. The contribution of variants in other known genes of HI among the populations of African ancestry is poorly studied and displays the lowest pick-up rate. We used whole exome sequencing (WES) to investigate pathogenic and likely pathogenic (PLP) variants in 34 novel human-mouse orthologs HI genes, in 40 individuals from Cameroon and South Africa diagnosed with non-syndromic hearing impairment (NSHI), and compared the data to WES data of 129 ethnically matched controls. In addition, protein modeling for selected PLP gene variants, gene enrichment, and network analyses were performed. A total of 4/38 murine genes, d6wsu163e, zfp719, grp152 and minar2, had no human orthologs. WES identified three rare PLP variants in 3/34 human-mouse orthologs genes in three unrelated Cameroonian patients, namely: OCM2, c.227G>C p.(Arg76Thr) and LRGI1, c.1657G>A p.(Gly533Arg) in a heterozygous state, and a PLP variant MCPH1, c.2311C>G p.(Pro771Ala) in a homozygous state. In silico functional analyses suggest that these human-mouse ortholog genes functionally co-expressed interactions with well-established HI genes: GJB2 and GJB6. The study found one homozygous variant in MCPH1, likely to explain HI in one patient, and suggests that human-mouse ortholog variants could contribute to the understanding of the physiology of hearing in humans.
Collapse
Affiliation(s)
- Oluwafemi G Oluwole
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Kevin K Esoh
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Edmond Wonkam-Tingang
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Noluthando Manyisa
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Jean Jacques Noubiap
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide and Royal Adelaide Hospital, Adelaide 5000, Australia
| | - Emile R Chimusa
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Ambroise Wonkam
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
- Department of Medicine, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
26
|
Ultrarare heterozygous pathogenic variants of genes causing dominant forms of early-onset deafness underlie severe presbycusis. Proc Natl Acad Sci U S A 2020; 117:31278-31289. [PMID: 33229591 DOI: 10.1073/pnas.2010782117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Presbycusis, or age-related hearing loss (ARHL), is a major public health issue. About half the phenotypic variance has been attributed to genetic factors. Here, we assessed the contribution to presbycusis of ultrarare pathogenic variants, considered indicative of Mendelian forms. We focused on severe presbycusis without environmental or comorbidity risk factors and studied multiplex family age-related hearing loss (mARHL) and simplex/sporadic age-related hearing loss (sARHL) cases and controls with normal hearing by whole-exome sequencing. Ultrarare variants (allele frequency [AF] < 0.0001) of 35 genes responsible for autosomal dominant early-onset forms of deafness, predicted to be pathogenic, were detected in 25.7% of mARHL and 22.7% of sARHL cases vs. 7.5% of controls (P = 0.001); half were previously unknown (AF < 0.000002). MYO6, MYO7A, PTPRQ, and TECTA variants were present in 8.9% of ARHL cases but less than 1% of controls. Evidence for a causal role of variants in presbycusis was provided by pathogenicity prediction programs, documented haploinsufficiency, three-dimensional structure/function analyses, cell biology experiments, and reported early effects. We also established Tmc1 N321I/+ mice, carrying the TMC1:p.(Asn327Ile) variant detected in an mARHL case, as a mouse model for a monogenic form of presbycusis. Deafness gene variants can thus result in a continuum of auditory phenotypes. Our findings demonstrate that the genetics of presbycusis is shaped by not only well-studied polygenic risk factors of small effect size revealed by common variants but also, ultrarare variants likely resulting in monogenic forms, thereby paving the way for treatment with emerging inner ear gene therapy.
Collapse
|
27
|
Brownstein Z, Gulsuner S, Walsh T, Martins FTA, Taiber S, Isakov O, Lee MK, Bordeynik-Cohen M, Birkan M, Chang W, Casadei S, Danial-Farran N, Abu-Rayyan A, Carlson R, Kamal L, Arnþórsson ÁÖ, Sokolov M, Gilony D, Lipschitz N, Frydman M, Davidov B, Macarov M, Sagi M, Vinkler C, Poran H, Sharony R, Samara N, Zvi N, Baris-Feldman H, Singer A, Handzel O, Hertzano R, Ali-Naffaa D, Ruhrman-Shahar N, Madgar O, Sofrin E, Peleg A, Khayat M, Shohat M, Basel-Salmon L, Pras E, Lev D, Wolf M, Steingrimsson E, Shomron N, Kelley MW, Kanaan M, Allon-Shalev S, King MC, Avraham KB. Spectrum of genes for inherited hearing loss in the Israeli Jewish population, including the novel human deafness gene ATOH1. Clin Genet 2020; 98:353-364. [PMID: 33111345 PMCID: PMC8045518 DOI: 10.1111/cge.13817] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022]
Abstract
Mutations in more than 150 genes are responsible for inherited hearing loss, with thousands of different, severe causal alleles that vary among populations. The Israeli Jewish population includes communities of diverse geographic origins, revealing a wide range of deafness-associated variants and enabling clinical characterization of the associated phenotypes. Our goal was to identify the genetic causes of inherited hearing loss in this population, and to determine relationships among genotype, phenotype, and ethnicity. Genomic DNA samples from informative relatives of 88 multiplex families, all of self-identified Jewish ancestry, with either non-syndromic or syndromic hearing loss, were sequenced for known and candidate deafness genes using the HEar-Seq gene panel. The genetic causes of hearing loss were identified for 60% of the families. One gene was encountered for the first time in human hearing loss: ATOH1 (Atonal), a basic helix-loop-helix transcription factor responsible for autosomal dominant progressive hearing loss in a five-generation family. Our results show that genomic sequencing with a gene panel dedicated to hearing loss is effective for genetic diagnoses in a diverse population. Comprehensive sequencing enables well-informed genetic counseling and clinical management by medical geneticists, otolaryngologists, audiologists, and speech therapists and can be integrated into newborn screening for deafness.
Collapse
Affiliation(s)
- Zippora Brownstein
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Suleyman Gulsuner
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, WA, USA
| | - Tom Walsh
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, WA, USA
| | - Fábio Tadeu Arrojo Martins
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shahar Taiber
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Isakov
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ming K. Lee
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, WA, USA
| | - Mor Bordeynik-Cohen
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Maria Birkan
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetic Institute, Rabin Medical Center–Beilinson Hospital, Tel Aviv University Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Weise Chang
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communications Disorders, NIH, Bethesda, MD, USA
| | - Silvia Casadei
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, WA, USA
| | - Nada Danial-Farran
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Genetics Institute, Ha'Emek Medical Center, Afula, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Amal Abu-Rayyan
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Biological Sciences, Bethlehem University, Bethlehem, Palestine
| | - Ryan Carlson
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, WA, USA
| | - Lara Kamal
- Department of Biological Sciences, Bethlehem University, Bethlehem, Palestine
| | - Ásgeir Örn Arnþórsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Meirav Sokolov
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Otolaryngology - Head and Neck Surgery, Schneider Children's Medical Center, Petach Tikva, Israel
| | - Dror Gilony
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Otolaryngology - Head and Neck Surgery, Schneider Children's Medical Center, Petach Tikva, Israel
| | - Noga Lipschitz
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Otolaryngology - Head and Neck Surgery, Sheba Medical Center, Tel Hashomer, Israel
| | - Moshe Frydman
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel
| | - Bella Davidov
- Raphael Recanati Genetic Institute, Rabin Medical Center–Beilinson Hospital, Tel Aviv University Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Michal Macarov
- Department of Human Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Michal Sagi
- Department of Human Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Chana Vinkler
- Institute of Medical Genetics, Wolfson Medical Center, Holon, Israel
| | - Hana Poran
- Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel
| | - Reuven Sharony
- Genetics Institute, Meir Medical Center, Kfar Saba and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Na’ama Zvi
- Department of Human Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Amihood Singer
- Community Genetics Department, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Ophir Handzel
- Department of Otolaryngology Head and Neck Surgery and Maxillofacial Surgery, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ronna Hertzano
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Doaa Ali-Naffaa
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Human Genetics Institute, Lady Davis Carmel Medical Center, Haifa, Israel
| | - Noa Ruhrman-Shahar
- Raphael Recanati Genetic Institute, Rabin Medical Center–Beilinson Hospital, Tel Aviv University Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Ory Madgar
- Department of Otolaryngology - Head and Neck Surgery, Sheba Medical Center, Tel Hashomer, Israel
| | - Efrat Sofrin
- Raphael Recanati Genetic Institute, Rabin Medical Center–Beilinson Hospital, Tel Aviv University Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Amir Peleg
- Human Genetics Institute, Lady Davis Carmel Medical Center, Haifa, Israel
| | - Morad Khayat
- Genetics Institute, Ha'Emek Medical Center, Afula, Israel
| | - Mordechai Shohat
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Institute of Medical Genetics, Maccabi HMO, Rehovot, Israel
| | - Lina Basel-Salmon
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetic Institute, Rabin Medical Center–Beilinson Hospital, Tel Aviv University Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Elon Pras
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel
| | - Dorit Lev
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Institute of Medical Genetics, Wolfson Medical Center, Holon, Israel
| | - Michael Wolf
- Department of Otolaryngology - Head and Neck Surgery, Sheba Medical Center, Tel Hashomer, Israel
| | - Eirikur Steingrimsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matthew W. Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communications Disorders, NIH, Bethesda, MD, USA
| | - Moien Kanaan
- Department of Biological Sciences, Bethlehem University, Bethlehem, Palestine
| | - Stavit Allon-Shalev
- Genetics Institute, Ha'Emek Medical Center, Afula, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Mary-Claire King
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, WA, USA
| | - Karen B. Avraham
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Askew C, Chien WW. Adeno-associated virus gene replacement for recessive inner ear dysfunction: Progress and challenges. Hear Res 2020; 394:107947. [PMID: 32247629 PMCID: PMC7939749 DOI: 10.1016/j.heares.2020.107947] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/08/2023]
Abstract
Approximately 3 in 1000 children in the US under 4 years of age are affected by hearing loss. Currently, cochlear implants represent the only line of treatment for patients with severe to profound hearing loss, and there are no targeted drug or biological based therapies available. Gene replacement is a promising therapeutic approach for hereditary hearing loss, where viral vectors are used to deliver functional cDNA to "replace" defective genes in dysfunctional cells in the inner ear. Proof-of-concept studies have successfully used this approach to improve auditory function in mouse models of hereditary hearing loss, and human clinical trials are on the immediate horizon. The success of this method is ultimately determined by the underlying biology of the defective gene and design of the treatment strategy, relying on intervention before degeneration of the sensory structures occurs. A challenge will be the delivery of a corrective gene to the proper target within the therapeutic window of opportunity, which may be unique for each specific defective gene. Although rescue of pre-lingual forms of recessive deafness have been explored in animal models thus far, future identification of genes with post-lingual onset that are amenable to gene replacement holds even greater promise for treatment, since the therapeutic window is likely open for a much longer period of time. This review summarizes the current state of adeno-associated virus (AAV) gene replacement therapy for recessive hereditary hearing loss and discusses potential challenges and opportunities for translating inner ear gene replacement therapy for patients with hereditary hearing loss.
Collapse
Affiliation(s)
- Charles Askew
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wade W Chien
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, USA; Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
29
|
Keithley EM. Pathology and mechanisms of cochlear aging. J Neurosci Res 2020; 98:1674-1684. [PMID: 31066107 PMCID: PMC7496655 DOI: 10.1002/jnr.24439] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/22/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022]
Abstract
Presbycusis, or age-related hearing loss (ARHL), occurs in most mammals with variations in the age of onset, rate of decline, and magnitude of degeneration in the central nervous system and inner ear. The affected cochlear structures include the stria vascularis and its vasculature, spiral ligament, sensory hair cells and auditory neurons. Dysfunction of the stria vascularis results in a reduced endocochlear potential. Without this potential, the cochlear amplification provided by the electro-motility of the outer hair cells is insufficient, and a high-frequency hearing-loss results. Degeneration of the sensory cells, especially the outer hair cells also leads to hearing loss due to lack of amplification. Neuronal degeneration, another hallmark of ARHL, most likely underlies difficulties with speech discrimination, especially in noisy environments. Noise exposure is a major cause of ARHL. It is well-known to cause sensory cell degeneration, especially the outer hair cells at the high frequency end of the cochlea. Even loud, but not uncomfortable, sound levels can lead to synaptopathy and ultimately neuronal degeneration. Even in the absence of a noisy environment, aged cells degenerate. This pathology most likely results from damage to mitochondria and contributes to degenerative changes in the stria vascularis, hair cells, and neurons. The genetic underpinnings of ARHL are still unknown and most likely involve various combinations of genes. At present, the only effective strategy for reducing ARHL is prevention of noise exposure. If future strategies can improve mitochondrial activity and reduce oxidative damage in old age, these should also bring relief.
Collapse
Affiliation(s)
- Elizabeth M. Keithley
- Division of Otolaryngology ‐ Head and Neck SurgeryUniversity of CaliforniaSan DiegoCalifornia
| |
Collapse
|
30
|
de Bruijn SE, Smits JJ, Liu C, Lanting CP, Beynon AJ, Blankevoort J, Oostrik J, Koole W, de Vrieze E, Cremers CWRJ, Cremers FPM, Roosing S, Yntema HG, Kunst HPM, Zhao B, Pennings RJE, Kremer H. A RIPOR2 in-frame deletion is a frequent and highly penetrant cause of adult-onset hearing loss. J Med Genet 2020; 58:jmedgenet-2020-106863. [PMID: 32631815 PMCID: PMC8120656 DOI: 10.1136/jmedgenet-2020-106863] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/25/2020] [Accepted: 04/01/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Hearing loss is one of the most prevalent disabilities worldwide, and has a significant impact on quality of life. The adult-onset type of the condition is highly heritable but the genetic causes are largely unknown, which is in contrast to childhood-onset hearing loss. METHODS Family and cohort studies included exome sequencing and characterisation of the hearing phenotype. Ex vivo protein expression addressed the functional effect of a DNA variant. RESULTS An in-frame deletion of 12 nucleotides in RIPOR2 was identified as a highly penetrant cause of adult-onset progressive hearing loss that segregated as an autosomal dominant trait in 12 families from the Netherlands. Hearing loss associated with the deletion in 63 subjects displayed variable audiometric characteristics and an average (SD) age of onset of 30.6 (14.9) years (range 0-70 years). A functional effect of the RIPOR2 variant was demonstrated by aberrant localisation of the mutant RIPOR2 in the stereocilia of cochlear hair cells and failure to rescue morphological defects in RIPOR2-deficient hair cells, in contrast to the wild-type protein. Strikingly, the RIPOR2 variant is present in 18 of 22 952 individuals not selected for hearing loss in the Southeast Netherlands. CONCLUSION Collectively, the presented data demonstrate that an inherited form of adult-onset hearing loss is relatively common, with potentially thousands of individuals at risk in the Netherlands and beyond, which makes it an attractive target for developing a (genetic) therapy.
Collapse
Affiliation(s)
- Suzanne E de Bruijn
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Jeroen J Smits
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Chang Liu
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Cornelis P Lanting
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Andy J Beynon
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | | | - Jaap Oostrik
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Wouter Koole
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Erik de Vrieze
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Cor W R J Cremers
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Frans P M Cremers
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Susanne Roosing
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Helger G Yntema
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Henricus P M Kunst
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Bo Zhao
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ronald J E Pennings
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Hannie Kremer
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Khela H, Kenna MA. Genetics of pediatric hearing loss: A functional perspective. Laryngoscope Investig Otolaryngol 2020; 5:511-519. [PMID: 32596495 PMCID: PMC7314484 DOI: 10.1002/lio2.390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/02/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES This article reviews the current role of genetics in pediatric hearing loss (HL). METHODS A review of the current literature regarding the genetic basis of HL in children was performed. RESULTS To date, 119 nonsyndromic genes have been associated with HL. There are also hundreds of syndromic causes that have HL as part of the clinical phenotype. CONCLUSIONS Identifying HL genes coupled with clinical characteristics ("genotype-phenotype") yields a more accurate diagnosis and prognosis. Although the complexity of the auditory apparatus presents challenges, gene therapy is emerging and may be a viable management option in the future.
Collapse
Affiliation(s)
- Harmon Khela
- Summer Scholars Program, Otolaryngology and Communication Enhancement, Boston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Margaret A. Kenna
- Department of Otolaryngology and Communication EnhancementBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
32
|
Li C, Zou H, Xiong Z, Xiong Y, Miyagishima DF, Wanggou S, Li X. Construction and Validation of a 13-Gene Signature for Prognosis Prediction in Medulloblastoma. Front Genet 2020; 11:429. [PMID: 32508873 PMCID: PMC7249855 DOI: 10.3389/fgene.2020.00429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/07/2020] [Indexed: 01/28/2023] Open
Abstract
Background: Recent studies have identified several molecular subgroups of medulloblastoma associated with distinct clinical outcomes; however, no robust gene signature has been established for prognosis prediction. Our objective was to construct a robust gene signature-based model to predict the prognosis of patients with medulloblastoma. Methods: Expression data of medulloblastomas were acquired from the Gene Expression Omnibus (GSE85217, n = 763; GSE37418, n = 76). To identify genes associated with overall survival (OS), we performed univariate survival analysis and least absolute shrinkage and selection operator (LASSO) Cox regression. A risk score model was constructed based on selected genes and was validated using multiple datasets. Differentially expressed genes (DEGs) between the risk groups were identified. Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and protein–protein interaction (PPI) analyses were performed. Network modules and hub genes were identified using Cytoscape. Furthermore, tumor microenvironment (TME) was evaluated using ESTIMATE algorithm. Tumor-infiltrating immune cells (TIICs) were inferred using CIBERSORTx. Results: A 13-gene model was constructed and validated. Patients classified as high-risk group had significantly worse OS than those as low-risk group (Training set: p < 0.0001; Validation set 1: p < 0.0001; Validation set 2: p = 0.00052). The area under the curve (AUC) of the receiver operating characteristic (ROC) analysis indicated a good performance in predicting 1-, 3-, and 5-year OS in all datasets. Multivariate analysis integrating clinical factors demonstrated that the risk score was an independent predictor for the OS (validation set 1: p = 0.001, validation set 2: p = 0.004). We then identified 265 DEGs between risk groups and PPI analysis predicted modules that were highly related to central nervous system and embryonic development. The risk score was significantly correlated with programmed death-ligand 1 (PD-L1) expression (p < 0.001), as well as immune score (p = 0.035), stromal score (p = 0.010), and tumor purity (p = 0.010) in Group 4 medulloblastomas. Correlations between the 13-gene signature and the TIICs in Sonic hedgehog and Group 4 medulloblastomas were revealed. Conclusion: Our study constructed and validated a robust 13-gene signature model estimating the prognosis of medulloblastoma patients. We also revealed genes and pathways that may be related to the development and prognosis of medulloblastoma, which might provide candidate targets for future investigation.
Collapse
Affiliation(s)
- Chang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Han Zou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Zujian Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Danielle F Miyagishima
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, United States.,Department of Genetics, Yale School of Medicine, New Haven, CT, United States
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
33
|
Yamoah EN, Li M, Shah A, Elliott KL, Cheah K, Xu PX, Phillips S, Young SM, Eberl DF, Fritzsch B. Using Sox2 to alleviate the hallmarks of age-related hearing loss. Ageing Res Rev 2020; 59:101042. [PMID: 32173536 PMCID: PMC7261488 DOI: 10.1016/j.arr.2020.101042] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory deficit. ARHL reduces the quality of life of the growing population, setting seniors up for the enhanced mental decline. The size of the needy population, the structural deficit, and a likely research strategy for effective treatment of chronic neurosensory hearing in the elderly are needed. Although there has been profound advancement in auditory regenerative research, there remain multiple challenges to restore hearing loss. Thus, additional investigations are required, using novel tools. We propose how the (1) flat epithelium, remaining after the organ of Corti has deteriorated, can be converted to the repaired-sensory epithelium, using Sox2. This will include (2) developing an artificial gene regulatory network transmitted by (3) large viral vectors to the flat epithelium to stimulate remnants of the organ of Corti to restore hair cells. We hope to unite with our proposal toward the common goal, eventually restoring a functional human hearing organ by transforming the flat epithelial cells left after the organ of Corti loss.
Collapse
Affiliation(s)
- Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
| | - Anit Shah
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
| | - Karen L Elliott
- Department of Biology, CLAS, University of Iowa, Iowa City, USA
| | - Kathy Cheah
- Department of Biochemistry, Hong Kong University, Hong Kong, China
| | - Pin-Xian Xu
- Department of Biochemistry, Hong Kong University, Hong Kong, China
| | - Stacia Phillips
- Department of Biochemistry, Hong Kong University, Hong Kong, China
| | - Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA; Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
| | - Daniel F Eberl
- Department of Biology, CLAS, University of Iowa, Iowa City, USA
| | - Bernd Fritzsch
- Department of Biology, CLAS, University of Iowa, Iowa City, USA.
| |
Collapse
|
34
|
Data-driven segmentation of audiometric phenotypes across a large clinical cohort. Sci Rep 2020; 10:6704. [PMID: 32317648 PMCID: PMC7174357 DOI: 10.1038/s41598-020-63515-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
Pure tone audiograms are used to assess the degree and underlying source of hearing loss. Audiograms are typically categorized into a few canonical types, each thought to reflect distinct pathologies of the ear. Here, we analyzed 116,400 patient records from our clinic collected over a 24-year period and found that standard categorization left 46% of patient records unclassified. To better account for the full spectrum of hearing loss profiles, we used a Gaussian Mixture Model (GMM) to segment audiograms without any assumptions about frequency relationships, interaural symmetry or etiology. The GMM converged on ten types, featuring varying degrees of high-frequency hearing loss, flat loss, mixed loss, and notched profiles, with predictable relationships to patient age and sex. A separate GMM clustering of 15,380 audiograms from the National Health and Nutrition Examination Survey (NHANES) identified six similar types, that only lacked the more extreme hearing loss configurations observed in our patient cohort. Whereas traditional approaches distill hearing loss configurations down to a few canonical types by disregarding much of the underlying variability, an objective probabilistic model that accounted for all of the data identified an organized, but more heterogenous set of audiogram types that was consistent across two large clinical databases.
Collapse
|
35
|
Stavber L, Hovnik T, Kotnik P, Lovrečić L, Kovač J, Tesovnik T, Bertok S, Dovč K, Debeljak M, Battelino T, Avbelj Stefanija M. High frequency of pathogenic ACAN variants including an intragenic deletion in selected individuals with short stature. Eur J Endocrinol 2020; 182:243-253. [PMID: 31841439 PMCID: PMC7087498 DOI: 10.1530/eje-19-0771] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/13/2019] [Indexed: 12/30/2022]
Abstract
CONTEXT Defining the underlying etiology of idiopathic short stature (ISS) improves the overall management of an individual. OBJECTIVE To assess the frequency of pathogenic ACAN variants in selected individuals. DESIGN The single-center cohort study was conducted at a tertiary university children's hospital. From 51 unrelated patients with ISS, the 16 probands aged between 3 and 18 years (12 females) with advanced bone age and/or autosomal dominant inheritance pattern of short stature were selected for the study. Fifteen family members of ACAN-positive probands were included. Exome sequencing was performed in all probands, and additional copy number variation (CNV) detection was applied in selected probands with a distinct ACAN-associated phenotype. RESULTS Systematic phenotyping of the study cohort yielded 37.5% (6/16) ACAN-positive probands, with all novel pathogenic variants, including a 6.082 kb large intragenic deletion, detected by array comparative genomic hybridization (array CGH) and exome data analysis. All variants were co-segregated with short stature phenotype, except in one family member with the intragenic deletion who had an unexpected growth pattern within the normal range (-0.5 SDS). One patient presented with otosclerosis, a sign not previously associated with aggrecanopathy. CONCLUSIONS ACAN pathogenic variants presented a common cause of familial ISS. The selection criteria used in our study were suggested for a personalized approach to genetic testing of the ACAN gene in clinical practice. Our results expanded the number of pathogenic ACAN variants, including the first intragenic deletion, and suggested CNV evaluation in patients with typical clinical features of aggrecanopathy as reasonable. Intra-familial phenotypic variability in growth patterns should be considered.
Collapse
Affiliation(s)
- L Stavber
- Unit for Special Laboratory Diagnostics, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - T Hovnik
- Unit for Special Laboratory Diagnostics, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - P Kotnik
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - L Lovrečić
- Clinical Institute of Medical Genetics, University Medical Centre, Ljubljana, Slovenia
| | - J Kovač
- Unit for Special Laboratory Diagnostics, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - T Tesovnik
- Unit for Special Laboratory Diagnostics, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - S Bertok
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - K Dovč
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - M Debeljak
- Unit for Special Laboratory Diagnostics, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
| | - T Battelino
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - M Avbelj Stefanija
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre, Ljubljana, Slovenia
- Correspondence should be addressed to M Avbelj Stefanija;
| |
Collapse
|
36
|
Wells HRR, Newman TA, Williams FMK. Genetics of age-related hearing loss. J Neurosci Res 2020; 98:1698-1704. [PMID: 31989664 DOI: 10.1002/jnr.24549] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
Age-related hearing loss (ARHL) has recently been confirmed as a common complex trait, that is, it is heritable with many genetic variants each contributing a small amount of risk, as well as environmental determinants. Historically, attempts to identify the genetic variants underlying the ARHL have been of limited success, relying on the selection of candidate genes based on the limited knowledge of the pathophysiology of the condition, and linkage studies in samples comprising related individuals. More recently genome-wide association studies have been performed, but these require very large samples having consistent and reliable phenotyping for hearing loss (HL), and early attempts suffered from lack of reliable replication of their findings. Replicated variants shown associated with ARHL include those lying in genes GRM7, ISG20, TRIOBP, ILDR1, and EYA4. The availability of large biobanks and the development of collaborative consortia have led to a breakthrough over the last couple of years, and many new genetic variants associated with ARHL are becoming available, through the analysis publicly available bioresources and electronic health records. These findings along with immunohistochemistry and mouse models of HL look set to help disentangle the genetic architecture of ARHL, and highlight the need for standardization of phenotyping methods to facilitate data sharing and collaboration across research networks.
Collapse
Affiliation(s)
| | - Tracey A Newman
- CES, Medicine, B85, M55, Life Sciences, University of Southampton, Southampton, UK
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
37
|
Cousins RPC. Medicines discovery for auditory disorders: Challenges for industry. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 146:3652. [PMID: 31795652 DOI: 10.1121/1.5132706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Currently, no approved medicines are available for the prevention or treatment of hearing loss. Pharmaceutical industry productivity across all therapeutic indications has historically been disappointing, with a 90% chance of failure in delivering a marketed drug after entering clinical evaluation. To address these failings, initiatives have been applied in the three cornerstones of medicine discovery: target selection, clinical candidate selection, and clinical studies. These changes aimed to enable data-informed decisions on the translation of preclinical observations into a safe, clinically effective medicine by ensuring the best biological target is selected, the most appropriate chemical entity is advanced, and that the clinical studies enroll the correct patients. The specific underlying pathologies need to be known to allow appropriate patient selection, so improved diagnostics are required, as are methodologies for measuring in the inner ear target engagement, drug delivery and pharmacokinetics. The different therapeutic strategies of protecting hearing or preventing hearing loss versus restoring hearing are reviewed along with potential treatments for tinnitus. Examples of current investigational drugs are discussed to highlight key challenges in drug discovery and the learnings being applied to improve the probability of success of launching a marketed medicine.
Collapse
Affiliation(s)
- Rick P C Cousins
- University College London Ear Institute, University College London, London, WC1X 8EE, United Kingdom
| |
Collapse
|
38
|
Zou S, Mei X, Yang W, Zhu R, Yang T, Hu H. Whole-exome sequencing identifies rare pathogenic and candidate variants in sporadic Chinese Han deaf patients. Clin Genet 2019; 97:352-356. [PMID: 31486067 DOI: 10.1111/cge.13638] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/29/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022]
Abstract
Genetic causes of hearing loss are highly heterogeneous and often ethnically specific. In recent years, a variety of next-generation sequencing (NGS) panels have been developed to target deafness-causative genes. Whole-exome sequencing (WES), on the other hand, was rarely used for genetic testing for deafness. In this study, we performed WES in 38 sporadic Chinese Han deaf patients who have been pre-excluded for mutations in common deafness genes GJB2, SLC26A4 and MT-RNR1. Non-synonymous variants have been filtered based on their minor allele frequencies in public databases and ethnically matched controls. Bi-allelic pathogenic mutations in eight deafness genes, OTOF, TRIOBP, ESPN, HARS2, CDH23, MYO7A, USH1C and TJP2, were identified in 10 patients, with 17 mutations identified in this study not being associated with deafness previously. For the rest 28 patients, possibly bi-allelic rare non-synonymous variants in an averaged 4.7 genes per patient were identified as candidate pathogenic causes for future analysis. Our study showed that WES may provide a unified platform for genetic testing of deafness and enables retro-analyzing when new causative genes are revealed.
Collapse
Affiliation(s)
- Songfeng Zou
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xueshuang Mei
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Weiqiang Yang
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rvfei Zhu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Tao Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Hongyi Hu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
39
|
Gallego-Martinez A, Requena T, Roman-Naranjo P, May P, Lopez-Escamez JA. Enrichment of damaging missense variants in genes related with axonal guidance signalling in sporadic Meniere's disease. J Med Genet 2019; 57:82-88. [PMID: 31494579 DOI: 10.1136/jmedgenet-2019-106159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/17/2019] [Accepted: 08/03/2019] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Meniere's disease (MD) is a rare inner ear disorder with a significant genetic contribution defined by a core phenotype: episodic vertigo, sensorineural hearing loss and tinnitus. It has been mostly described in sporadic cases, familial cases being around 10% of the observed individuals. It is associated with an accumulation of endolymph in the inner ear, but the molecular underpinnings remain largely unknown. The main molecular pathways showing higher differentially expressed genes in the supporting cells of the inner ear are related to cochlea-vestibular innervation, cell adhesion and leucocyte extravasation. In this study, our objective is to find a burden of rare variants in genes that interact with the main signalling pathways in supporting cells of the inner ear in patients with sporadic MD. METHODS We designed a targeted-sequencing panel including genes related with the main molecular pathways in supporting cells and sequenced 860 Spanish patients with sporadic MD. Variants with minor allele frequencies <0.1 in the gene panel were compared with three independent reference datasets. Variants were classified as loss of function, missense and synonymous. Missense variants with a combined annotation-dependent depletion score of >20 were classified as damaging missense variants. RESULTS We have observed a significant burden of damaging missense variants in few key genes, including the NTN4 gene, associated with axon guidance signalling pathways in patients with sporadic MD. We have also identified active subnetworks having an enrichment of rare variants in sporadic MD. CONCLUSION The burden of missense variants in the NTN4 gene suggests that axonal guidance signalling could be a novel pathway involved in sporadic MD.
Collapse
Affiliation(s)
- Alvaro Gallego-Martinez
- Otology & Neurotology Group CTS 495, Genomic Medicine Area, Centro de Genomica e Investigación Oncológica, Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Sur-Alzette, Luxembourg
| | - Teresa Requena
- Otology & Neurotology Group CTS 495, Genomic Medicine Area, Centro de Genomica e Investigación Oncológica, Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Pablo Roman-Naranjo
- Otology & Neurotology Group CTS 495, Genomic Medicine Area, Centro de Genomica e Investigación Oncológica, Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Patrick May
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Sur-Alzette, Luxembourg
| | - Jose A Lopez-Escamez
- Otology & Neurotology Group CTS 495, Genomic Medicine Area, Centro de Genomica e Investigación Oncológica, Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
- Department of Otolaryngology, Instituto de Investigación Biosanitaria, ibs.GRANADA, Hospital Universitario Virgen de las Nieves, Granada, Spain
| |
Collapse
|
40
|
Pater JA, Green J, O'Rielly DD, Griffin A, Squires J, Burt T, Fernandez S, Fernandez B, Houston J, Zhou J, Roslin NM, Young TL. Novel Usher syndrome pathogenic variants identified in cases with hearing and vision loss. BMC MEDICAL GENETICS 2019; 20:68. [PMID: 31046701 PMCID: PMC6498547 DOI: 10.1186/s12881-019-0777-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Background Usher syndrome, the most common form of inherited deaf-blindness, is unlike many other forms of syndromic hereditary hearing loss in that the extra aural clinical manifestations are also detrimental to communication. Usher syndrome patients with early onset deafness also experience vision loss due to progressive retinitis pigmentosa that can lead to legal blindness in their third or fourth decade. Methods Using a multi-omic approach, we identified three novel pathogenic variants in two Usher syndrome genes (USH2A and ADGRV1) in cases initially referred for isolated vision or hearing loss. Results In a multiplex hearing loss family, two affected sisters, the product of a second cousin union, are homozygous for a novel nonsense pathogenic variant in ADGRV1 (c.17062C > T, p.Arg5688*), predicted to create a premature stop codon near the N-terminus of ADGRV1. Ophthalmological examination of the sisters confirmed typical retinitis pigmentosa and prompted a corrected Usher syndrome diagnosis. In an unrelated clinical case, a child with hearing loss tested positive for two novel USH2A splicing variants (c.5777-1G > A, p. Glu1926_Ala1952del and c.10388-2A > G, p.Asp3463Alafs*6) and RNA studies confirmed that both pathogenic variants cause splicing errors. Interestingly, these same USH2A variants are also identified in another family with vision loss where subsequent clinical follow-up confirmed pre-existing hearing loss since early childhood, eventually resulting in a reassigned diagnosis of Usher syndrome. Conclusion These findings provide empirical evidence to increase Usher syndrome surveillance of at-risk children. Given that novel antisense oligonucleotide therapies have been shown to rescue retinal degeneration caused by USH2A splicing pathogenic variants, these solved USH2A patients may now be eligible to be enrolled in therapeutic trials.
Collapse
Affiliation(s)
- Justin A Pater
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Jane Green
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Darren D O'Rielly
- Molecular Diagnostic Laboratory, Eastern Health, Craig L. Dobbin Genetics Research Centre, Faculty of Medicine, Memorial University, 300 Prince Phillip Drive, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
| | - Anne Griffin
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Jessica Squires
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Taylor Burt
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Sara Fernandez
- Provincial Medical Genetics, Craig L. Dobbin Research Centre, Eastern Health, 300 Prince Phillip Drive, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
| | - Bridget Fernandez
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada.,Provincial Medical Genetics, Craig L. Dobbin Research Centre, Eastern Health, 300 Prince Phillip Drive, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
| | - Jim Houston
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Jiayi Zhou
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada
| | - Nicole M Roslin
- The Centre for Applied Genomics, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Terry-Lynn Young
- Craig L. Dobbin Research Centre, Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, Newfoundland & Labrador, AIB 3V6, Canada. .,Molecular Diagnostic Laboratory, Eastern Health, Craig L. Dobbin Genetics Research Centre, Faculty of Medicine, Memorial University, 300 Prince Phillip Drive, St. John's, Newfoundland and Labrador, A1B 3V6, Canada.
| |
Collapse
|
41
|
Eckert MA, Vaden KI, Dubno JR. Age-Related Hearing Loss Associations With Changes in Brain Morphology. Trends Hear 2019; 23:2331216519857267. [PMID: 31213143 PMCID: PMC6585256 DOI: 10.1177/2331216519857267] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/12/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Age-related hearing loss has been associated with varied auditory cortex morphology in human neuroimaging studies. These findings have suggested that peripheral auditory system declines cause changes in brain morphology but could also be due to latent variables that affect the auditory periphery and brain. The current longitudinal study was designed to evaluate these explanations for pure-tone threshold and brain morphology associations. Thirty adults (mean age at Time 1 = 64.12 ± 10.32 years) were studied at two time points (average duration between visits = 2.62 ± 0.81 years). Small- to medium-effect size associations were observed between high-frequency pure-tone thresholds and auditory cortex gray matter volume at each time point. Although there were significant longitudinal changes in low- and high-frequency hearing measures and brain morphology, those longitudinal changes were not significantly correlated across participants. High-frequency hearing measures at Time 1 were significantly related to more lateral ventricle expansion, such that participants with higher measures exhibited larger increases in ventricle size. This ventricle effect was statistically independent of high-frequency hearing associations with auditory cortex morphology. Together, these results indicate that there are at least two mechanisms for associations between age-related hearing loss and brain morphology. Potential explanations for a direct hearing loss effect on brain morphology, as well as latent variables that likely affect both the inner ear and brain, are discussed.
Collapse
Affiliation(s)
- Mark A. Eckert
- Department of Otolaryngology—Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Kenneth I. Vaden
- Department of Otolaryngology—Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Judy R. Dubno
- Department of Otolaryngology—Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|