1
|
Lv X, Yang C, Li X, Liu Y, Yang Y, Jin T, Chen Z, Jia J, Wang M, Li L. Ferroptosis and hearing loss: from molecular mechanisms to therapeutic interventions. J Enzyme Inhib Med Chem 2025; 40:2468853. [PMID: 39992186 PMCID: PMC11852237 DOI: 10.1080/14756366.2025.2468853] [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: 08/27/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Hearing loss profoundly affects social engagement, mental health, cognition, and brain development, with sensorineural hearing loss (SNHL) being a major concern. Linked to ototoxic medications, ageing, and noise exposure, SNHL presents significant treatment challenges, highlighting the need for effective prevention and regeneration strategies. Ferroptosis, a distinct form of cell death featuring iron-dependent lipid peroxidation, has garnered interest due to its potential role in cancer, ageing, and neuronal degeneration, especially hearing loss. The emerging role of ferroptosis as a crucial mediator in SNHL suggests that it may offer a novel therapeutic target for otoprotection. This review aims to summarise the intricate connection between ferroptosis and SNHL, offering a fresh perspective for exploring targeted therapeutic strategies that could potentially mitigate cochlear cells damage and enhance the quality of life for individuals with hearing impairments.
Collapse
Affiliation(s)
- Xingyi Lv
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyi Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Xianying Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yun Liu
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yu Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Tongyan Jin
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhijian Chen
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Jinjing Jia
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Min Wang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Li Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
2
|
Krishnamoorthy M, Jayasankaran C, Lakshmi S, Sarvani C, Margret JJ, Mahalingam S, Amritkumar P, Subramanyam PV, S SR, Srisailapathy CRS. Clinical Exome Sequencing Identifies, Two Homozygous LOXHD1 Variants in Two Inbred Families With Pre-Lingual Hearing Loss From South India. Ann Hum Genet 2025; 89:114-125. [PMID: 40070250 DOI: 10.1111/ahg.12593] [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: 07/04/2024] [Revised: 12/16/2024] [Accepted: 02/10/2025] [Indexed: 04/11/2025]
Abstract
In recent years, numerous genetic variants have been linked with prelingual hearing loss (HL). Variants in the LOXHD1 gene (lipoxygenase homology domain-1) associated with DFNB77 are highly heterogeneous, with different auditory characteristics varying from stable to progressive and mild to profound. To date, 168 DFNB77 cases have been recorded worldwide. Forty-one hearing-impaired (HI) probands, who were previously excluded for a set of four common deafness-causing genes (viz., GJB2, GJB6, SLC26A4, and CDH23) from 33 HI families, were subjected to clinical exome sequencing (CES) involving 285 genes associated with HL. This was followed by a segregation analysis of the available members in the family. We identified two pathogenic LOXHD1 variants in two unrelated inbred families. One is a novel homozygous pathogenic nonsense variant (c.3999C > A; p.C1333X), whereas the other is a likely pathogenic missense variant (c.6046G > T; p.E2046K). In silico tools such as SIFT, PolyPhen-2, Mutation Taster, CADD, and REVEL scores were used to predict variant pathogenicity. Furthermore, American College of Medical Genetics and Genomics guidelines specific to HL were applied to finally classify a variant as pathogenic or otherwise. The frequency of LOXHD1 variants identified in our study is 4.88% (2/41). This is the first LOXHD1 report associated with non-syndromic HL in South Indian families.
Collapse
Affiliation(s)
- Mathuravalli Krishnamoorthy
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| | - Chandru Jayasankaran
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| | - Sorna Lakshmi
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| | - Chodisetty Sarvani
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
- The Gordon Institute of TAFE Geelong, Geelong, Victoria, Australia
| | - Jeffrey Justin Margret
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| | - Subathra Mahalingam
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| | - Pavithra Amritkumar
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
- Meenakshi Academy of Higher Education and Research (MAHER), Chennai, Tamil Nadu, India
| | - Paridhy Vanniya Subramanyam
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| | - Sarrath Rathnaraajan S
- Consultant ENT Surgeon, Madras ENT Research Foundation (MERF) Pvt Ltd, Chennai, Tamil Nadu, India
| | - C R Srikumari Srisailapathy
- Department of Genetics, Post Graduate Institute of Basic Medical Sciences, University of Madras (Taramani Campus), Chennai, Tamil Nadu, India
| |
Collapse
|
3
|
Morovvati S, Sarband MM, Doostmohammadi S, Rayat S, Emamdjomeh H, Farhadi M, Asghari A, Garshasbi M, Falah M. The clinical and genetic spectrum of twenty-six individuals with hearing loss affected by MYO15A variants. Sci Rep 2025; 15:14320. [PMID: 40275102 PMCID: PMC12022297 DOI: 10.1038/s41598-025-99417-7] [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: 11/14/2024] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
Myosin XVA (MYO15A) is a member of the myosin superfamily that, as a motor protein, plays an essential role in actin polymerization at the tip of the stereocilia in hair cells. Variants in MYO15A are known to be the third most common reason for autosomal recessive non-syndromic hearing loss (ARNSHL). Here, we present twenty-six unrelated families with MYO15A variants from an Iranian cohort. Whole exome sequencing (WES) was performed following a comprehensive medical evaluation. The identified variants were assessed based on the American College of Medical Genetics and Genomics guidelines. Twenty-seven distinct variants linked to MYO15A were identified as contributors to profound ARNSHL. These included ten novel variants and seventeen previously documented variants that co-segregated. Most variants were truncating, with an equal distribution of missense and splicing variants. This research expands the mutational spectrum of MYO15A by introducing ten novel variants and highlights its importance in profound ARNSHL. Moreover, comparing the variants in different domains of MYO15A with previously reported variants in these domains provides more information about the MYO15A protein's role in the hearing process. This information can enhance understanding of the genetic basis of hearing loss and improve future management strategies, including prognosis, prevention, and treatment based on gene modification.
Collapse
Affiliation(s)
- Saeid Morovvati
- Department of Genetics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Samaneh Doostmohammadi
- Faculty of Converging Sciences and Technologies (NBIC), Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sima Rayat
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hessamaldin Emamdjomeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alimohamad Asghari
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoumeh Falah
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Yang SH, Liu J, Quan Y, Lin G, Zhou X, He H, Gan X, Yang T, Cui MY, Du X, Quan X, Gu W, Zhang HY, Wang H, Guan W. The improvement in diagnostic yield of developmental and epileptic encephalopathy by the multi-omics sequential testing method. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167854. [PMID: 40254267 DOI: 10.1016/j.bbadis.2025.167854] [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: 04/23/2024] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Despite traditional panel and Whole Exome Sequencing (WES) assays, the causative factors for 60 % of epilepsy cases remain elusive, mainly due to incomplete detection of variant spectrums, and limited ability to interpret variants. Our research developed the multi-omics method of a comprehensive sequential testing methodology, to enhance the diagnostic yield for the etiology. In this study, we performed sequential multi-omics analyses on a cohort of 236 Chinese patients exhibiting recurrent seizures along with developmental delay or intellectual disability. Our study had devised a comprehensive multi-omics variant analysis methodology in a sequential mode. The initial analytical strategy included WES, CNV-seq and in-house cases evidence. If no pathogenic cause was identified, the subsequent analytical approach in the sequential mode included the analysis of WGS SVs, mitochondrial variations, dynamic mutations, and abnormalities in RNA-seq. Our results revealed that the initial step achieved a diagnostic detection rate of 44.1 % (104 cases). Subsequently, WGS and RNA-seq testing were performed, with 33 familial diagnoses tested positive, representing a 14 % increase. Meanwhile our pipeline has elucidated the pathogenicity classification of 72 variants which are either not yet recorded in the ClinVar database or are classified as VUS. Our study achieved an overall positive diagnostic rate of 58.1 % (137/236). In summary, our pipeline can detect comprehensive variant spectrums and provide a clear interpretation of variations with unclear clinical significance, the multi-omics sequential testing approach significantly improves the rate of genetic diagnosis for epileptic disorders.
Collapse
Affiliation(s)
- Shuang-Hao Yang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; Department of bioinformatics, Chigene (Beijing) Translational Medical Research Center Co. Ltd., Beijing Yizhuang Biomedical Park, 100176, China; Beijing Quanpu Medical Laboratory Co., Ltd., E2, 3rd Floor, No. 88 Kechuang 6th Road, Beijing Yizhuang Biomedical Park, 100176, China
| | - Jiatong Liu
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110004, China
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Guangyu Lin
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaohua Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hua He
- Department of bioinformatics, Chigene (Beijing) Translational Medical Research Center Co. Ltd., Beijing Yizhuang Biomedical Park, 100176, China
| | - Xianfeng Gan
- Department of bioinformatics, Chigene (Beijing) Translational Medical Research Center Co. Ltd., Beijing Yizhuang Biomedical Park, 100176, China
| | - Tuanfeng Yang
- Department of Neurology, Peking University International Hospital, No.1 Life Park Road, Changping District, Beijing 102206, China
| | - Ming-Yang Cui
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Xilong Du
- Department of bioinformatics, Chigene (Beijing) Translational Medical Research Center Co. Ltd., Beijing Yizhuang Biomedical Park, 100176, China; Beijing Quanpu Medical Laboratory Co., Ltd., E2, 3rd Floor, No. 88 Kechuang 6th Road, Beijing Yizhuang Biomedical Park, 100176, China
| | - Xiaofang Quan
- Department of bioinformatics, Chigene (Beijing) Translational Medical Research Center Co. Ltd., Beijing Yizhuang Biomedical Park, 100176, China
| | - Weiyue Gu
- Department of bioinformatics, Chigene (Beijing) Translational Medical Research Center Co. Ltd., Beijing Yizhuang Biomedical Park, 100176, China; Beijing Quanpu Medical Laboratory Co., Ltd., E2, 3rd Floor, No. 88 Kechuang 6th Road, Beijing Yizhuang Biomedical Park, 100176, China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Hua Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110004, China.
| | - WenZheng Guan
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110004, China.
| |
Collapse
|
5
|
Twumasi Aboagye E, Adadey SM, Alves de Souza Rios L, Esoh KK, Wonkam-Tingang E, Xhakaza L, De Kock C, Schrauwen I, Amenga-Etego L, Lang D, Awandare GA, Leal SM, Mowla S, Wonkam A. Bi-Allelic MARVELD2 Variant Identified with Exome Sequencing in a Consanguineous Multiplex Ghanaian Family Segregating Non-Syndromic Hearing Loss. Int J Mol Sci 2025; 26:3337. [PMID: 40244166 PMCID: PMC11989440 DOI: 10.3390/ijms26073337] [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: 02/28/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Genetic studies and phenotypic expansion of hearing loss (HL) for people living in Africa are greatly needed. We evaluated the clinical phenotypes of three affected siblings presenting non-syndromic (NS) HL and five unaffected members of a consanguineous Ghanaian family. Analysis of exome sequence data was performed for all affected and one unaffected family members. In-depth genetic and cellular characterization studies were performed to investigate biological significance of the implicated variant using bioinformatic tools and cell-based experimentation. Audiological examinations showed severe-to-profound, bilateral, symmetrical, and post-lingual onset. The whole-exome sequencing (WES) identified a homozygous frameshift variant: MARVEL domain containing 2 (MARVELD2):c.1058dup;p.(Val354Serfs*5) in all affected siblings. This frameshift variant leads to an early stop codon insertion and predicted to be targeted by nonsense medicated decay (mutant protein predicted to lack conserved C-terminal domain if translated). Cell immunofluorescence and immunocytochemistry studies exposed the functional impact of the mutant protein's expression, stability, localization, protein-protein binding, barrier function, and actin cytoskeleton architecture. The identified variant segregates with NSHL in the index Ghanaian family. The data support this nonsense variant as pathogenic, likely to impact the homeostasis of ions, solutes, and other molecules, compromising membrane barrier and signaling in the inner ear spaces.
Collapse
Affiliation(s)
- Elvis Twumasi Aboagye
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra LG 54, Ghana; (L.A.-E.); (G.A.A.)
| | - Samuel Mawuli Adadey
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Leonardo Alves de Souza Rios
- Department of Pathology, Division of Haematology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (L.A.d.S.R.); (S.M.)
| | - Kevin K. Esoh
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
- McKusick-Nathans Institute & Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Edmond Wonkam-Tingang
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Lettilia Xhakaza
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Carmen De Kock
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Isabelle Schrauwen
- Department of Translational Neurosciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA;
| | - Lucas Amenga-Etego
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra LG 54, Ghana; (L.A.-E.); (G.A.A.)
| | - Dirk Lang
- Department of Human Biology, Division of Cell Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa;
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra LG 54, Ghana; (L.A.-E.); (G.A.A.)
| | - Suzanne M. Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Centre, New York, NY 10032, USA;
- Taub Institute, Columbia University Medical Center, New York, NY 10032, USA
| | - Shaheen Mowla
- Department of Pathology, Division of Haematology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (L.A.d.S.R.); (S.M.)
| | - Ambroise Wonkam
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
- McKusick-Nathans Institute & Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Zafeer MF, Ramzan M, Duman D, Mutlu A, Seyhan S, Kalcioglu MT, Fitoz S, DeRosa BA, Guo S, Dykxhoorn DM, Tekin M. Human organoids for rapid validation of gene variants linked to cochlear malformations. Hum Genet 2025; 144:375-389. [PMID: 39786576 PMCID: PMC12003500 DOI: 10.1007/s00439-024-02723-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025]
Abstract
Developmental anomalies of the hearing organ, the cochlea, are diagnosed in approximately one-fourth of individuals with congenital. The majority of patients with cochlear malformations remain etiologically undiagnosed due to insufficient knowledge about underlying genes or the inability to make conclusive interpretations of identified genetic variants. We used exome sequencing for the genetic evaluation of hearing loss associated with cochlear malformations in three probands from unrelated families deafness. We subsequently generated monoclonal induced pluripotent stem cell (iPSC) lines, bearing patient-specific knockins and knockouts using CRISPR/Cas9 to assess pathogenicity of candidate variants. We detected FGF3 (p.Arg165Gly) and GREB1L (p.Cys186Arg), variants of uncertain significance in two recognized genes for deafness, and PBXIP1(p.Trp574*) in a candidate gene. Upon differentiation of iPSCs towards inner ear organoids, we observed developmental aberrations in knockout lines compared to their isogenic controls. Patient-specific single nucleotide variants (SNVs) showed similar abnormalities as the knockout lines, functionally supporting their causality in the observed phenotype. Therefore, we present human inner ear organoids as a potential tool to validate the pathogenicity of DNA variants associated with cochlear malformations.
Collapse
Affiliation(s)
- Mohammad Faraz Zafeer
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Memoona Ramzan
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Duygu Duman
- Department of Audiology, Ankara University Faculty of Health Sciences, Ankara, Türkiye
- Ankara University Rare Diseases Application and Research Center (NADiR), Ankara, Türkiye
| | - Ahmet Mutlu
- Faculty of Medicine, Department of Otorhinolaryngology, Istanbul Medeniyet University, Istanbul, Türkiye
- Otorhinolaryngology Clinic of Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Türkiye
| | - Serhat Seyhan
- Laboratory of Genetics, Memorial Şişli Hospital, Istanbul, Türkiye
| | - M Tayyar Kalcioglu
- Faculty of Medicine, Department of Otorhinolaryngology, Istanbul Medeniyet University, Istanbul, Türkiye
- Otorhinolaryngology Clinic of Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Türkiye
| | - Suat Fitoz
- Department of Diagnostic Radiology, Ankara University School of Medicine, Ankara, Türkiye
| | - Brooke A DeRosa
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shengru Guo
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, US
| | - Mustafa Tekin
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, US.
- , 1501 NW 10th Avenue, BRB-610 (M860), Miami, FL, 33136, USA.
| |
Collapse
|
7
|
Miolo G, Margiotta F, Murgia A, Puppa LD, Corona G. Advances in Understanding the Molecular Dynamics of Autosomal Dominant Auditory Neuropathy: Unveiling a Novel DIAPH3 Gene Variant Associated With Sensorineural Hearing Loss and Bilateral Vestibular Aqueduct Enlargement. J Audiol Otol 2025; 29:151-157. [PMID: 40040362 DOI: 10.7874/jao.2024.00143] [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: 02/19/2024] [Accepted: 06/21/2024] [Indexed: 03/06/2025] Open
Abstract
Auditory neuropathy is characterized by abnormal neural conduction in the auditory pathway despite normal outer hair cell function, exhibiting substantial genetic heterogeneity and phenotypic variability. We report the case of a 29-year-old male patient with hearing loss, bilateral enlargement of the vestibular aqueduct (EVA), and vestibular system dysfunction. Based on these features, which are tipically indicative of Pendred syndrome, a molecular investigation including the SLC26A4 gene was performed. This analysis identified a novel heterozygous missense variant, c.411A>C, in exon 4 of the DIAPH3 gene, likely associated with autosomal dominant auditory neuropathy. This point mutation results in substituting glutamic acid with aspartic acid at position 137 p.(Glu137Asp), in the functional Rho-GTPase-binding domain of the DIAPH3 protein. Segregation analysis of the parents and two siblings of the proband revealed the variant's de novo origin. According to the American College of Medical Genetics and Genomics criteria, this finding underscores the need to reclassify the variant as likely pathogenic. This is the first evidence of an association between a DIAPH3 variant and hearing loss coupled with bilateral EVA and vestibular system dysfunction. This finding contributes to a better understanding of the phenotypic complexity of disorders grouped within the auditory neuropathy spectrum.
Collapse
Affiliation(s)
- Gianmaria Miolo
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
- Department of Medical Laboratory, Genetics Section, Pordenone Hospital, Pordenone, Italy
| | - Francesco Margiotta
- Unit of Otolaryngology, Department of Specialistic Surgery, Pordenone Hospital, Pordenone, Italy
| | - Alessandra Murgia
- Laboratory of Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Lara Della Puppa
- Oncogenetics and Functional Oncogenomics Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
8
|
Li Y, Sun T, Hu S, Xu H, Zhang T, Liu J, Lu S, Wang B, Dan G. SLC26A4 C.317C > A Variant: Functional Analysis and Patient-Derived Induced Pluripotent Stem Line Development. Mol Genet Genomic Med 2025; 13:e70098. [PMID: 40260864 PMCID: PMC12012755 DOI: 10.1002/mgg3.70098] [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: 10/29/2024] [Revised: 03/06/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND SLC26A4 is the second most common cause of hereditary hearing loss worldwide. This gene predominantly harbors pathogenic variants, including splice, nonsense, and missense. Although missense variants are relatively common, their specific effects on protein function remain unclear. Consequently, there is an urgent need to establish an in vitro system to investigate how these variants impact SLC26A4 protein function. METHODS Genetic testing was conducted to determine the specific types of underlying genetic variants in patients. Following this, we employed plasmid transfection to evaluate the effects of the variants on both protein expression levels and the protein's subcellular localization. Thereafter, we transformed peripheral blood mononuclear cells (PBMCs) from the proband into induced pluripotent stem cells (iPSCs) through Sendai virus-mediated transduction. RESULTS Genetic testing revealed that the proband carried compound heterozygous variants: SLC26A4 c.919-2A > G and c.317C > A. The c.317C > A variant markedly decreased the expression levels of SLC26A4 mRNA and its encoded protein. Additionally, it led to the protein's accumulation in the cytoplasm as aggregates. We successfully reprogrammed peripheral blood mononuclear cells from the proband into induced pluripotent stem cells (iPSCs) and verified that these iPSCs retained their pluripotency, differentiation potential, and genetic integrity. CONCLUSION These results provide important insights into the mechanisms by which SLC26A4 gene variants lead to hearing loss.
Collapse
Affiliation(s)
- Yijing Li
- National Center for International Research in Cell and Gene Therapy, Sino‐British Research Centre for Molecular Oncology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Tao Sun
- Department of Clinical MedicineHenan Medical CollegeZhengzhouChina
- Laboratory of Hearing Loss MechanismHenan Provincial Medical Key LaboratoryZhengzhouChina
| | - Sang Hu
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouChina
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouChina
| | - Teng Zhang
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouChina
| | - Jinlong Liu
- National Center for International Research in Cell and Gene Therapy, Sino‐British Research Centre for Molecular Oncology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Shuangshuang Lu
- National Center for International Research in Cell and Gene Therapy, Sino‐British Research Centre for Molecular Oncology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Bing Wang
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guo Dan
- Laboratory of Hearing Loss MechanismHenan Provincial Medical Key LaboratoryZhengzhouChina
- Department of Basic Clinical MedicineHenan Medical CollegeZhengzhouChina
| |
Collapse
|
9
|
Srinivasan T, Redfield S, Poorvu T, Kenna MA. Management of Prenatal Expanded Genetic Carrier Screening Results for Autosomal Recessive Sensorineural Hearing Loss. Otolaryngol Head Neck Surg 2025; 172:1006-1016. [PMID: 39523564 DOI: 10.1002/ohn.1028] [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: 02/09/2024] [Revised: 09/26/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Expanded carrier screening (ECS) identified couples at-risk to have a baby with an autosomal recessive genetic condition. Several genes implicated in sensorineural hearing loss (SNHL) are included in prenatal or preconception genetics ECS testing. Early identification of SNHL risk may enable prognostication of hearing loss, early educational intervention, and minimization of unnecessary diagnostic testing. We sought to describe cases where ECS enabled early SNHL-risk identification. STUDY DESIGN Retrospective chart review. SETTING Maternal-Fetal Care Center and Otolaryngology department at an academic tertiary hospital. METHODS Medical records of parent-infant dyads with positive ECS results for variants in autosomal recessive SNHL genes were reviewed. Data regarding genetic diagnostic testing, newborn hearing screening, time to HL diagnosis, audiological evaluation, and clinical consultations were compiled. RESULTS Fifteen pregnant with positive ECS results for SNHL were referred for consultation with a pediatric otolaryngologist and genetic counselor. Generally, these couples were highly educated and adequately insured. 14 had pathogenic variants for GJB2 and 1 for USH2A. Four couples pursued prenatal genetic diagnosis via amniocentesis; 11 couples deferred genetic testing to the postnatal period or waited for initial hearing evaluation. Six babies inherited biallelic GJB2 mutations. Four were found to have SNHL on ABR by age 5 weeks and received follow-up management, 1 had a normal hearing evaluation despite being gene-positive, and 1 was lost to follow-up before hearing evaluation. CONCLUSIONS Carrier screening and confirmatory prenatal or neonatal genetic testing provided considerable lead time for early audiometric testing and appropriate intervention services including hearing aid fitting.
Collapse
Affiliation(s)
| | - Shelby Redfield
- Department of Otolaryngology and Communication Enhancement, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Tabitha Poorvu
- Maternal Fetal Care Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Margaret A Kenna
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Otolaryngology and Communication Enhancement, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Alonso AV, Aguado RG, Camerano AM, Enseñat JF, de la Fuente EO, Angulo CM. Hearing and Vestibular Impairment Related to a Variant (c.263G>C) of the COCH Gene. Otolaryngol Head Neck Surg 2025; 172:982-992. [PMID: 39666779 DOI: 10.1002/ohn.1074] [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: 09/19/2024] [Revised: 10/31/2024] [Accepted: 11/16/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVE To ascertain pathogenic variants frequency and type in the COCH gene among Cantabrian patients with nonsyndromic hereditary hearing loss (HL), and to understand their cochleovestibular manifestations. STUDY DESIGN An observational study on patients with postlingual nonsyndromic sensorineural hearing loss (SNHL), who underwent a genetic study using next-generation sequencing (gene panel) in the otolaryngology clinics between January 2019 and December 2023. SETTING Referral center Marqués de Valdecilla University Hospital in Santander (Spain). METHODS A cohort of 248 otolaryngologic clinic-referred patients suspected of genetic SNHL underwent sequencing analysis targeting 231 genes. RESULTS A likely pathogenic or pathogenic variant causing HL was found in 57 (22.8%) patients. Among them, 7 (2.8%) were heterozygous carriers of the c.263G>C variant in the LCCL domain of the COCH gene, included as index cases. Subsequent familial segregation studies were performed. A total of 22 genetically and clinically studied patients were included. All but 3 family members displayed bilateral progressive SNHL starting in adulthood. Thirteen patients reported instability, but none met Meniere's disease criteria. CONCLUSION COCH gene variants are frequent in Cantabria. A variant with pathogenic evidence (c.263G>C in the LCCL domain) was detected. The phenotype observed is similar to a subgroup of patients with other variants described in the same functional domain: progressive SNHL and instability secondary to vestibular hypofunction.
Collapse
Affiliation(s)
- Aida Veiga Alonso
- Department of Otolaryngology, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Rocío González Aguado
- Department of Otolaryngology, Marqués de Valdecilla University Hospital, Santander, Spain
| | | | | | - Esther Onecha de la Fuente
- Department of Genetics, Marqués de Valdecilla University Hospital, Santander, Spain
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Carmelo Morales Angulo
- Department of Otolaryngology, Marqués de Valdecilla University Hospital, Santander, Spain
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Spain
- Department of Otolaryngology, Faculty of Medicine, University of Cantabria, Santander, Cantabria, Spain
| |
Collapse
|
11
|
Carlson RJ, Taiber S, Rubinstein JT. Gene Therapy for Hearing Loss: Which Genes Next? Otol Neurotol 2025; 46:239-247. [PMID: 39951658 DOI: 10.1097/mao.0000000000004423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2025]
Abstract
INTRODUCTION Hearing loss is the most common sensory deficit in humans, and roughly half of childhood-onset sensorineural hearing loss is genetic. Advances in gene therapy techniques have led to the first clinical trials for OTOF-associated hearing loss DFNB9. Therapies for other hearing loss genes are in various stages of development, and therefore a comprehensive evaluation of potential candidate genes can help to prioritize and guide these efforts. METHODS A list of 93 nonsyndromic hearing loss genes with consensus support was generated. Critical factors for evaluation were identified as gene size, timing of cochlear degradation, cell type(s) of primary expression, availability of mouse models and efficacy of adeno-associated virus experiments in those mice, and human hearing loss severity, onset, and prevalence. Each factor was addressed with gene-specific PubMed searches for applicable studies. RESULTS Each gene was evaluated according to the above factors, with favorable results indicating the most promising candidates for gene therapy. Genes that satisfied all the above conditions included TMPRSS3, PCDH15, and TMC1. Other genes, such as LOXHD1 and MYO6, had not yet had gene replacement attempts in a mouse model but otherwise satisfied all conditions and were likewise identified as promising candidates. CONCLUSION Based on this analysis, hearing loss genes vary widely in terms of their favorability for treatment by gene therapy approaches. Targeting development efforts to promising candidates will ensure the highest likelihood of clinical success. Several genes were identified as appealing next targets, signaling an increasing role of gene therapies in hearing loss care moving forward.
Collapse
Affiliation(s)
- Ryan J Carlson
- Departments of Genome Sciences and Medicine, University of Washington, Seattle, Washington, USA
| | - Shahar Taiber
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jay T Rubinstein
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Tshering KC, DiStefano MT, Oza AM, Ajuyah P, Webb R, Edoh E, Broeren E, Ratliff J, Gitau V, Paris K, Aburyyan A, Alexander J, Albano V, Bai D, Booth KTA, Buonfiglio PI, Charfeddine C, Dalamón V, Castillo ID, Moreno-Pelayo MA, Duzkale H, Dorshorst B, Faridi R, Kenna M, Lewis MA, Luo M, Lu Y, Mkaouar R, Matsunaga T, Nara K, Pandya A, Redfield S, Roux I, Schimmenti LA, Schrauwen I, Shaaban S, Shen J, Vona B, Smith RJ, Rehm HL, Azaiez H, Abou Tayoun AN, Amr SS. ClinGen recuration of hearing loss-associated genes demonstrates significant changes in gene-disease validity over time. Genet Med 2025; 27:101392. [PMID: 39987489 DOI: 10.1016/j.gim.2025.101392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
PURPOSE The Clinical Genome Resource (ClinGen) Hearing Loss Gene Curation Expert Panel was assembled in 2016 and has since curated 174 gene-disease relationships (GDRs) using ClinGen's semiquantitative framework. ClinGen mandates the timely recuration of all GDRs classified as Disputed, Limited, Moderate, and Strong every 2 to 3 years. METHODS Thirty-five GDRs met the criteria for recuration within 2 years of original curation. Previous evidence was reevaluated using the latest curation guidelines, and a comprehensive literature review was performed to obtain new evidence. Recurations were approved by the Gene Curation Expert Panel and published on the ClinGen website (www.clinicalgenome.org). RESULTS Eight of 35 GDRs (22%) changed their classification. Two Moderate and 5 Strong GDRs were upgraded to Definitive because of new case evidence. One Strong was subsumed under another Definitive GDR after evaluation of the lumping/splitting of disease entities. Twenty-seven of 35 patients remained unchanged, with little to no new evidence reported. CONCLUSION Genes classified as Moderate and Strong were likely to build evidence and change their classification over time, whereas Limited were unlikely to gain evidence. These findings highlight the critical role of recuration in ensuring that genetic tests and research studies incorporate the most recent evidence into their efforts.
Collapse
Affiliation(s)
| | - Marina T DiStefano
- The Broad Institute of MIT and Harvard, Cambridge, MA; Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Cambridge, MA
| | - Andrea M Oza
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Pamela Ajuyah
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ryan Webb
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Enyonam Edoh
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ellie Broeren
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Julie Ratliff
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Vanessa Gitau
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Kelley Paris
- The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Amal Aburyyan
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Victoria Albano
- Department of Otolaryngology & Communication Enhancement, Boston Children's Hospital, Boston, MA
| | - Donglin Bai
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Kevin T A Booth
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN; Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Paula I Buonfiglio
- Laboratorio de Fisiología y Genética de la Audición. Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cherine Charfeddine
- Laboratory of Biomedical Genomics and Oncogenetics, Institute Pasteur of Tunis, University of Tunis El Manar, Tunis, Tunisia; Université de la Manouba, Institut de Biotechnologie de Sidi-Thabet, Ariana, Tunisia
| | - Viviana Dalamón
- Laboratorio de Fisiología y Genética de la Audición. Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ignacio Del Castillo
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CB06/07/0048; CIBERER-ISCIII), Madrid, Spain
| | - Miguel Angel Moreno-Pelayo
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CB06/07/0048; CIBERER-ISCIII), Madrid, Spain
| | | | | | - Rabia Faridi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Margaret Kenna
- Department of Otolaryngology & Communication Enhancement, Boston Children's Hospital, Boston, MA
| | - Morag A Lewis
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London, United Kingdom
| | - Minjie Luo
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yu Lu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Rahma Mkaouar
- Laboratory of Biomedical Genomics and Oncogenetics, Institute Pasteur of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Tatsuo Matsunaga
- Division of Hearing and Balance Research, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Kiyomitsu Nara
- Division of Hearing and Balance Research, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Arti Pandya
- Division of Genetics and Metabolism, Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC
| | - Shelby Redfield
- Department of Medicine, University of Washington, Seattle, WA
| | - Isabelle Roux
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Lisa A Schimmenti
- Departments of Clinical Genomics, Mayo Clinic Enterprise, Rochester, MN
| | - Isabelle Schrauwen
- Department of Translational Neurosciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ
| | - Sherin Shaaban
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT; ARUP Laboratories, Salt Lake City, UT
| | - Jun Shen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Richard J Smith
- Molecular Otolaryngology and Renal Research Laboratories (MORL), Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA
| | - Heidi L Rehm
- The Broad Institute of MIT and Harvard, Cambridge, MA; Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Cambridge, MA
| | - Hela Azaiez
- Molecular Otolaryngology and Renal Research Laboratories (MORL), Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA
| | - Ahmad N Abou Tayoun
- Genomics Center of Excellence, Al Jalila Children's Specialty Hospital, Dubai Health, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University, Dubai Health, Dubai, United Arab Emirates
| | - Sami S Amr
- Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Cambridge, MA.
| |
Collapse
|
13
|
Jacovas VC, Zelnick M, McNulty S, Ross JE, Khurana N, Pan X, Nieto A, Martin S, McLean B, Elnagheeb MA, Cowan MJ, Puck JM, Hershfield M, Verbsky J, Walter J, Allenspach E, Chan AY, van Oers NSC, Ghosh R, Piazza M, Yuan B, Notarangelo LD, Johnson BA, Chinn IK. The ClinGen Severe Combined Immunodeficiency Disease Variant Curation Expert Panel: Specifications for classification of variants in ADA , DCLRE1C , IL2RG , IL7R , JAK3 , RAG1 , and RAG2. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322033. [PMID: 39990552 PMCID: PMC11844601 DOI: 10.1101/2025.02.11.25322033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Purpose This collaborative study, led by the Clinical Genome Resource Severe Combined Immunodeficiency Disease Variant Curation Expert Panel (ClinGen SCID-VCEP), implemented and adapted the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) guidelines for interpreting germline variants in genes with established relationships to SCID. The effort focused on the 7 most common SCID-related genes identified by SCID newborn screening in North America: ADA , DCLRE1C , IL2RG , IL7R , JAK3 , RAG1 , and RAG2 . Methods The SCID-VCEP conducted a rigorous review of variants that involved database analyses, literature review, and expert feedback to derive gene-specific modifications to the ACMG/AMP guidelines. These specifications were validated using a pilot set of 90 variants. Results: Of these 90 variants, 25 were classified as pathogenic, 21 as likely pathogenic, 14 as variants of uncertain significance (VUS), 18 as likely benign, and 12 as benign. Seventeen variants with conflicting classifications in ClinVar were successfully resolved. The criteria included modifications to 20 of the 28 original ACMG/AMP criteria specific to SCID-related genes. Conclusion The SCID-specific variant curation guidelines developed by the SCID-VCEP will enhance the precision of SCID genetic diagnosis and provide a robust framework for interpreting variants in SCID-related genes, contributing to appropriate treatment of SCID.
Collapse
|
14
|
Han SY, Cho SH, Jung SH, Kang M, Suh MW, Park MK, Lee JH, Lee SY. Genotypes and clinical phenotypes of pediatric patients with NOG variants: Middle ear surgical outcomes from a Tertiary Center in South Korea. Int J Pediatr Otorhinolaryngol 2025; 189:112230. [PMID: 39837070 DOI: 10.1016/j.ijporl.2025.112230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Although NOG variants are linked to congenital stapes fixation and conductive hearing loss (CHL), little is known about middle ear surgery outcomes and the characteristics of accompanying inner ear anomalies. We explored auditory phenotypes in patients with NOG variants, with a focus on the outcomes of middle ear surgery. METHODS This study included 11 patients from five unrelated Korean families harboring NOG variants. Genomic investigations were conducted using whole-exome sequencing and whole-genome sequencing. The clinical phenotypes, including pre- and postoperative audiological profiles, radiological abnormalities, and other comorbidities, were analyzed. RESULTS The average age at genetic testing was 8.2 years (range, 0-13 years). Two previously reported NOG variants (c.509C > T:p.Pro170Leu and c.252dup:p.Glu85ArgfsTer97) and three novel NOG variants, including the c.187G > T: p.Glu63Ter and two cryptic large deletion within the 17q22.2 region, were identified. All patients exhibited non-progressive CHL. Inner ear anomalies were documented in two patients, with variations such as cochlea and vestibular dysplasia. In this study, seven ears of four patients underwent stapedotomy, resulting in a significantly reduced air-bone gap of 10.18 ± 1.48 dB (P = 0.016), with sustained improvement. Conversely, patients carrying p.Pro170Leu variant, which is associated with poor outcomes for middle ear surgery, were excluded from surgical consideration. CONCLUSION We expanded the spectrum of genotypes and auditory phenotypes associated with NOG variants. Surgical intervention for CHL underlying NOG variants elicits favorable outcomes. However, clinicians should consider the potential for poor prognosis in certain NOG variants. Collectively, identifying NOG variants could guide the treatment strategies to improve CHL.
Collapse
Affiliation(s)
- Sang-Yoon Han
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Sung Ho Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Ho Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Myeongsin Kang
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Myung-Whan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Moo Kyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Zhou Y, Yin N, Ji L, Lu X, Yang W, Ye W, Du W, Li Y, Hu H, Mei X. A Novel PTPRQ c.3697del Variant Causes Autosomal Dominant Progressive Hearing Loss in Both Humans and Mice. Clin Genet 2025; 107:208-213. [PMID: 39434500 DOI: 10.1111/cge.14634] [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: 03/31/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
PTPRQ plays an important role in the development of inner ear hair cell stereocilia. While many autosomal recessive variants in PTPRQ have been identified as the pathogenic cause for nonsyndromic hearing loss (DFNB84A), so far only one autosomal dominant PTPRQ variant, c.6881G>A (p.Trp2294*), has been reported for late-onset, mild-to-severe hearing loss (DFNA73). By using targeted next-generation sequencing, this study identified a novel PTPRQ truncating pathogenic variant, c.3697del (p.Leu1233Phefs*11), from a Chinese Han family that co-segregated with autosomal dominant, postlingual, progressive hearing loss. A Ptprq-3700del knock-in mouse model was generated by CRISPR-Cas9 and characterized for its hearing function and inner ear morphology. While the homozygous knock-in mice exhibit profound hearing loss at all frequencies at the age of 3 weeks, the heterozygous mutant mice resemble the human patients in mild, progressive hearing loss from age 3 to 12 weeks, primarily affecting high frequencies. At this stage, the homozygous knock-in mice have a normal hair cell count but disorganized stereocilia. Cochlear proteosome analysis of the homozygous mutant mice revealed differentially expressed genes and pathways involved in oxidative phosphorylation, regulation of angiogenesis and synaptic vesicle cycling. Our study provides a valuable animal model for further functional studies of the pathogenic mechanisms underlying DFNA73.
Collapse
Affiliation(s)
- Yaqi Zhou
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Na Yin
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Lingchao Ji
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xiaochan Lu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Weiqiang Yang
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Weiping Ye
- Department of Obstetrics and Gynecology, Shanghai Putuo Maternity and Infant Hospital, Shanghai, China
| | - Wenhui Du
- Department of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Ya Li
- PKU-Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, Guangdong, China
| | - Hongyi Hu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xueshuang Mei
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
16
|
Au-Yeung CCY, Cheung YT, Cheng JYT, Ip KWH, Lee SD, Yang VYT, Lau AYT, Lee CKC, Chong PKH, Lau KW, van Lunenburg JTJ, Zheng DFD, Ho BHM, Tik C, Ho KKK, Rajaby R, Au CH, Yu MHC, Sung WK. UniVar: A variant interpretation platform enhancing rare disease diagnosis through robust filtering and unified analysis of SNV, INDEL, CNV and SV. Comput Biol Med 2025; 185:109560. [PMID: 39700857 DOI: 10.1016/j.compbiomed.2024.109560] [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: 08/20/2024] [Revised: 11/24/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Interpreting the pathogenicity of genetic variants associated with rare diseases is a laborious and time-consuming endeavour. To streamline the diagnostic process and lighten the burden of variant interpretation, it is crucial to automate variant annotation and prioritization. Unfortunately, currently available variant interpretation tools lack a unified and comprehensive workflow that can collectively assess the clinical significance of these types of variants together: small nucleotide variants (SNVs), small insertions/deletions (INDELs), copy number variants (CNVs) and structural variants (SVs). RESULTS The Unified Variant Interpretation Platform (UniVar) is a free web server tool that offers an automated and comprehensive workflow on annotation, filtering and prioritization for SNV, INDEL, CNV and SV collectively to identify disease-causing variants for rare diseases in one interface, ensuring accessibility for users even without programming expertise. To filter common CNVs/SVs, a diverse SV catalogue has been generated, that enables robust filtering of common SVs based on population allele frequency. Through benchmarking our SV catalogue, we showed that it is more complete and accurate than the state-of-the-art SV catalogues. Furthermore, to cope with those patients without detailed clinical information, we have developed a novel computational method that enables variant prioritization from gene panels. Our analysis shows that our approach could prioritize pathogenic variants as effective as using HPO terms assigned by clinicians, which adds value for cases without specific clinically assigned HPO terms. Lastly, through a practical case study of disease-causing compound heterozygous variants across SNV and SV, we demonstrated the uniqueness and effectiveness in variant interpretation of UniVar, edging over any existing interpretation tools. CONCLUSIONS UniVar is a unified and versatile platform that empowers researchers and clinicians to identify and interpret disease-causing variants in rare diseases efficiently through a single holistic interface and without a prerequisite for HPO terms. It is freely available without login and installation at https://univar.live/.
Collapse
Affiliation(s)
- Cherie C Y Au-Yeung
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Yuen-Ting Cheung
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Joshua Y T Cheng
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ken W H Ip
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Sau-Dan Lee
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Victor Y T Yang
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Amy Y T Lau
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Chit K C Lee
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Peter K H Chong
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - King Wai Lau
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | | | - Damon F D Zheng
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Brian H M Ho
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Crystal Tik
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Kingsley K K Ho
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ramesh Rajaby
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China; Shibuya Laboratory, Division of Medical Data Informatics, Human Genome Center, University of Tokyo, Japan
| | - Chun-Hang Au
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Mullin H C Yu
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Wing-Kin Sung
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China; Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China; Laboratory of Computational Genomics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
17
|
Stark Z, Byrne AB, Sampson MG, Lennon R, Mallett AJ. A guide to gene-disease relationships in nephrology. Nat Rev Nephrol 2025; 21:115-126. [PMID: 39443743 DOI: 10.1038/s41581-024-00900-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
The use of next-generation sequencing technologies such as exome and genome sequencing in research and clinical care has transformed our understanding of the molecular architecture of genetic kidney diseases. Although the capability to identify and rigorously assess genetic variants and their relationship to disease has advanced considerably in the past decade, the curation of clinically relevant relationships between genes and specific phenotypes has received less attention, despite it underpinning accurate interpretation of genomic tests. Here, we discuss the need to accurately define gene-disease relationships in nephrology and provide a framework for appraising genetic and experimental evidence critically. We describe existing international programmes that provide expert curation of gene-disease relationships and discuss sources of discrepancy as well as efforts at harmonization. Further, we highlight the need for alignment of disease and phenotype terminology to ensure robust and reproducible curation of knowledge. These collective efforts to support evidence-based translation of genomic sequencing into practice across clinical, diagnostic and research settings are crucial for delivering the promise of precision medicine in nephrology, providing more patients with timely diagnoses, accurate prognostic information and access to targeted treatments.
Collapse
Affiliation(s)
- Zornitza Stark
- ClinGen, Boston, MA, USA.
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Australian Genomics, Melbourne, Victoria, Australia.
| | - Alicia B Byrne
- ClinGen, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Matthew G Sampson
- ClinGen, Boston, MA, USA
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Department of Paediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel Lennon
- ClinGen, Boston, MA, USA
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, UK
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester, UK
| | - Andrew J Mallett
- ClinGen, Boston, MA, USA.
- Townsville Hospital and Health Service, Townsville, Queensland, Australia.
- College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia.
- Institute for Molecular Bioscience and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
18
|
Maekawa K, Nishio SY, Ishikawa K, Takahashi M, Kumakawa K, Okami M, Yoshimura H, Nakayama J, Teraoka M, Usami SI. Prevalence and Clinical Characteristics of OTOGL-Associated Hearing Loss Identified in a Cohort of 7065 Japanese Patients with Hearing Loss. Genes (Basel) 2025; 16:123. [PMID: 40004452 PMCID: PMC11854685 DOI: 10.3390/genes16020123] [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: 12/26/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hearing loss is one of the most common sensorineural impairments, and approximately 60% of early-onset cases are due to genetic variations. The otogelin-like protein, encoded by the OTOGL gene, is a component of the acellular membranes of the inner ear, such as the tectorial membrane, and is thought to play an important role in cochlear amplification. OTOGL gene variants are a rare cause of hearing loss such as DFNB84B, a mild-to-moderate sensorineural hearing loss presenting in early childhood with autosomal recessive inheritance. In this study, we aim to enhance our comprehension of the phenotypes of hearing loss caused by OTOGL variants. METHODS A total of 7056 Japanese patients with hearing loss were recruited, and based on massively parallel DNA sequencing on 158 target genes, we selected patients with biallelic OTOGL variants. RESULTS Ten affected individuals with OTOGL gene variants were detected, the largest group of patients yet to be reported, and eight of the eleven variants were novel. Our results showed that variations in this gene led to mild-to-moderate non-progressive hearing loss, and the accompanying symptoms, mainly vestibular symptoms, were speculated to present in adulthood. CONCLUSIONS Determination of the phenotypes of genes causative of hearing loss is expected to greatly benefit patients with hearing loss as it can assist in predicting outcomes and lead to appropriate intervention, which, in OTOGL-associated hearing loss cases, is based around the fact that the patients need not be concerned with deterioration in hearing, but require careful follow-up for vestibular symptoms.
Collapse
Affiliation(s)
- Karuna Maekawa
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (K.M.); (S.-y.N.)
| | - Shin-ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (K.M.); (S.-y.N.)
| | - Kotaro Ishikawa
- Department of Otolaryngology, National Rehabilitation Center for Persons with Disabilities, Tokorozawa 359-8555, Japan;
| | - Masahiro Takahashi
- Department of Otorhinolaryngology, International University of Health and Welfare, Mita Hospital, Tokyo 108-8329, Japan;
| | - Kozo Kumakawa
- Department of Otorhinolaryngology, Akasaka Toranomon Clinic, Minato 107-0052, Japan;
| | - Mayuri Okami
- Department of Otorhinolaryngology, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Hidekane Yoshimura
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Jun Nakayama
- Department of Otorhinolaryngology, Shiga University of Medical Science, Otsu 520-2192, Japan;
| | - Masato Teraoka
- Department of Otolaryngology, Head and Neck Surgery, Ehime University Graduate School of Medicine, Toon 791-0295, Japan;
| | - Shin-ichi Usami
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (K.M.); (S.-y.N.)
| |
Collapse
|
19
|
Kumai T, Nishio SY, Moteki H, Katada A, Usami SI. Auditory Neuropathy Caused by a Structural Variation in the OTOF Gene, Identified Using Oxford Nanopore Adaptive Sampling. Genes (Basel) 2025; 16:116. [PMID: 40004445 PMCID: PMC11855208 DOI: 10.3390/genes16020116] [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: 11/30/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The OTOF gene is reported to be the causative gene for non-syndromic recessive sensorineural hearing loss and auditory neuropathy spectrum disorder. About 300 variants have been reported, but there have been no reports to date on copy gain variants. METHODS We identified a copy gain variant in the OTOF gene through short-read next-generation sequencing analysis from one patient with auditory neuropathy. We also performed long-read next-generation sequencing analysis using the Oxford Nanopore Technologies adaptive sampling procedure. RESULTS The four-year-old male carried a duplication of chr2: 26,477,852 to 26,483,106 (a 5254-base duplication including exon 14 to exon 18 of the OTOF gene NM_001287489) and a c.5385C>A single nucleotide variant. We also confirmed that these two variants were located in the trans configuration based on haplotype phasing results using the long-read next-generation sequencing data. CONCLUSIONS This is the first report of an auditory neuropathy patient with a large duplication variant in the OTOF gene. The identified variants were novel, but based on the clinical phenotype of the patient, these variants seem to be the genetic cause of this patient's phenotype. Oxford Nanopore Technologies adaptive sampling is a powerful tool for the analysis of structural variants (particularly for determining the breakpoint and direction) and haplotype phasing.
Collapse
Affiliation(s)
- Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Shin-ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (S.-y.N.); (H.M.)
| | - Hideaki Moteki
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (S.-y.N.); (H.M.)
| | - Akihiro Katada
- Department of Otolaryngology, Asahikawa Red Cross Hospital, Asahikawa 070-0061, Japan;
| | - Shin-ichi Usami
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (S.-y.N.); (H.M.)
| |
Collapse
|
20
|
Maekawa K, Nishio SY, Miyazaki H, Ohta Y, Oishi N, Kasai M, Yamamoto A, Okami M, Wasano K, Sakai A, Usami SI. The Prevalence and Clinical Characteristics of MYO3A-Associated Hearing Loss in 15,684 Hearing Loss Patients. Genes (Basel) 2025; 16:92. [PMID: 39858639 PMCID: PMC11764741 DOI: 10.3390/genes16010092] [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: 12/26/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives:MYO3A belongs to the unconventional myosin superfamily, and the myosin IIIa protein localizes on the tip of the stereocilia of vestibular and cochlear hair cells. Deficiencies in MYO3A have been reported to cause the deformation of hair cells into abnormally long stereocilia with an increase in spacing. MYO3A is a rare causative gene of autosomal recessive sensorineural hearing loss (DFNB30), with only 13 cases reported to date. In this study, we aimed to elucidate the phenotypes caused by MYO3A variations. Methods: Massively parallel DNA sequencing was performed on 15,684 Japanese hearing loss patients (mean age 27.5 ± 23.1 years old, 6574 male, 8612 female and 498 patients for whom information was unavailable), identifying nine candidate patients with MYO3A variants. Results: We identified eight causative MYO3A variants by massively parallel DNA sequencing, including six novel variants, and reported nine individuals possessing MYO3A gene variants, which is the largest group of non-related patients yet to be detected. Our findings confirmed that MYO3A variants cause progressive hearing loss, with its onset varying from birth to the second decade, eventually leading to severe-to-profound hearing loss. Conclusions: We clarified that patients with MYO3A gene variants present with late-onset, progressive hearing loss. Our findings have enabled us to predict the outcomes of hearing loss in patients with candidate MYO3A gene variants and to provide intervention in a timely manner.
Collapse
Affiliation(s)
- Karuna Maekawa
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (K.M.); (S.-y.N.)
| | - Shin-ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (K.M.); (S.-y.N.)
| | - Hiromitsu Miyazaki
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University School of Medicine, Sendai 980-8575, Japan;
| | - Yoko Ohta
- Department of Otorhinolaryngology-Head and Neck Surgery, Tokyo Medical University, Tokyo 160-0023, Japan;
| | - Naoki Oishi
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Misato Kasai
- Department of Otorhinolaryngology, Juntendo University, Tokyo 113-8421, Japan;
| | - Ai Yamamoto
- Department of Otorhinolaryngology, Tokai University School of Medicine, Isehara 259-1193, Japan; (A.Y.); (M.O.); (K.W.)
| | - Mayuri Okami
- Department of Otorhinolaryngology, Tokai University School of Medicine, Isehara 259-1193, Japan; (A.Y.); (M.O.); (K.W.)
| | - Koichiro Wasano
- Department of Otorhinolaryngology, Tokai University School of Medicine, Isehara 259-1193, Japan; (A.Y.); (M.O.); (K.W.)
| | - Akihiro Sakai
- Department of Ear Nose and Throat-Head and Neck Surgery, Wakayama Medical University, Wakayama 641-0012, Japan;
| | - Shin-ichi Usami
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (K.M.); (S.-y.N.)
| |
Collapse
|
21
|
Pan J, Teng H, Liu F, Chen S, Liu Y, Teng Y, Liang D, Li Z, Wu L. Oligogenic effect is associated with the clinical heterogeneity of autosomal dominant deafness-15. Sci Rep 2025; 15:1981. [PMID: 39809934 PMCID: PMC11733205 DOI: 10.1038/s41598-025-85881-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Autosomal dominant deafness-15 which is caused by mutation in the POU4F3 gene, has been reported with a wide degree of clinical heterogeneity, even between intrafamilial members. However, the reason is still elusive. In this study, A four-generation Chinese family with 11 patients manifesting late-onset progressive non-syndromic hearing loss was recruited. The phenotype of hearing loss in this family showed a large variability in terms of onset age and progression speed. A novel mutation (c.706 C > T, p.L236F) was identified by the whole exome sequencing, and its pathogenicity was confirmed by altering the subcellular localization of POU4F3. In addition, we found that two individuals with earlier age of onset and more rapid progression of hearing loss carry additional pathogenic variants in other deafness genes (III-7, STRC:c.4057 C > T; IV-1, GJB2:c.109G > A; CDC14A:c.935G > A). By using the real time quantitative PCR, western blot, luciferase assays and electrophoretic mobility-shift assay, POU4F3 was proved to directly regulate the expression of STRC, GJB2 and CDC14A respectively. ChIP-seq further revealed that POU4F3 can also bind to a series of deafness genes. In summary we expanded the mutation spectrum of POU4F3 by identifying a novel mutation and its pathogenicity. Meanwhile, three genes STRC, GJB2 and CDC14A were validated as POU4F3 new targets, implicating that the variants in the three genes may play a role of genetic modifier to generate a synergistic and enhancement effect on the progression of DFNA15.
Collapse
Affiliation(s)
- Jianyan Pan
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
- Department of Birth Health and Genetics, The Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530000, Guangxi, China
| | - Hua Teng
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Fang Liu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Siyi Chen
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Yaning Liu
- Laboratory of Molecular Genetics, Hunan Jiahui Genetics Hospital, Changsha, 410000, Hunan, China
| | - Yanling Teng
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Desheng Liang
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
- Laboratory of Molecular Genetics, Hunan Jiahui Genetics Hospital, Changsha, 410000, Hunan, China
| | - Zhuo Li
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China.
| | - Lingqian Wu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China.
- Laboratory of Molecular Genetics, Hunan Jiahui Genetics Hospital, Changsha, 410000, Hunan, China.
- Bright Prosperity Institute, Room 1006-2, 10th Floor, Building 1, No. 180 Kecheng Street, Hangzhou, 310000, China.
| |
Collapse
|
22
|
Cao Y, Zhang X, Lan L, Li D, Li J, Xie L, Xiong F, Yu L, Wu X, Wang H, Wang Q. Identification of genetic mechanisms of non-isolated auditory neuropathy with various phenotypes in Chinese families. Orphanet J Rare Dis 2025; 20:11. [PMID: 39780253 PMCID: PMC11715445 DOI: 10.1186/s13023-025-03540-7] [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: 09/09/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Non-isolated auditory neuropathy (AN), or syndromic AN, is marked by AN along with additional systemic manifestations. The diagnostic process is challenging due to its varied symptoms and overlap with other syndromes. This study focuses on two mitochondrial function-related genes which result in non-isolated AN, FDXR and TWNK, providing a summary and enrichment analysis of genes associated with non-isolated AN to elucidate the genotype-phenotype correlation and underlying mechanisms. METHODS Seven independent Chinese Han patients with mutations in FDXR and TWNK underwent comprehensive clinical evaluations, genetic testing, and bioinformatics analyses. Diagnostic assessments included auditory brainstem response and distortion product otoacoustic emissions, supplemented by other examinations. Whole exome sequencing and Sanger sequencing validated genetic findings. Pathogenicity was assessed following American College of Medical Genetics and Genomics guidelines. Genes associated with non-isolated AN were summarized from prior reports, and functional enrichment analysis was conducted using Gene Ontology databases. RESULTS A total of 11 variants linked to non-isolated AN were identified in this study, eight of which were novel. Patients' age of hearing loss onset ranged from 2 to 25 years, averaging 11 years. Hearing loss varied from mild to profound, with 57.1%(4/7) of patients having risk factors and 71.4%(5/7) exhibiting additional systemic symptoms such as muscle weakness, ataxia, and high arches. Functional enrichment analysis revealed that genes associated with non-isolated AN predominantly involve mitochondrial processes, affecting the central and peripheral nervous, musculoskeletal, and visual systems. CONCLUSION This study identifies novel mutations in FDXR and TWNK that contribute to non-isolated AN through mitochondrial dysfunction. The findings highlight the role of mitochondrial processes in non-isolated AN, suggesting potential relevance as biomarkers for neurodegenerative diseases. Further research is required to explore these mechanisms and potential therapies.
Collapse
Affiliation(s)
- Yang Cao
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Xiaolong Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Lan Lan
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Danyang Li
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Jin Li
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Linyi Xie
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Fen Xiong
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Lan Yu
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Xiaonan Wu
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
| | - Hongyang Wang
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China.
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China.
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China.
| | - Qiuju Wang
- Senior Department of Otolaryngology Head and Neck Surgery, The 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China.
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China.
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China.
| |
Collapse
|
23
|
Arai Y, Nishio SY, Goto S, Kobayashi Y, Honkura Y, Ganaha A, Ishikawa K, Oka SI, Futagawa H, Okami M, Takada F, Nagai K, Esaki T, Okano T, Ohta Y, Masuda S, Egusa K, Teraoka M, Sugahara K, Usami SI. Novel OTOG Variants and Clinical Features of Hearing Loss in a Large Japanese Cohort. Genes (Basel) 2025; 16:60. [PMID: 39858607 PMCID: PMC11765236 DOI: 10.3390/genes16010060] [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: 12/06/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The OTOG gene is responsible for autosomal recessive non-syndromic sensorineural hearing loss and is assigned as DFNB18B. To date, 44 causative OTOG variants have been reported to cause non-syndromic hearing loss. However, the detailed clinical features for OTOG-associated hearing loss remain unclear. METHODS In this study, we analyzed 7065 patients with non-syndromic hearing loss (mean age 26.4 ± 22.9 years, 2988 male, 3855 female, and 222 without gender information) using massively parallel DNA sequencing for 158 target deafness genes. We identified the patients with biallelic OTOG variants and summarized the clinical characteristics. RESULTS Among the 7065 patients, we identified 14 possibly disease-causing OTOG variants in 26 probands, with 13 of the 14 variants regarded as novel. Patients with OTOG-associated hearing loss mostly showed congenital or childhood-onset hearing loss. They were considered to show non-progressive, mild-to-moderate hearing loss. There were no symptoms that accompanied the hearing loss in OTOG-associated hearing loss patients. CONCLUSIONS We confirmed non-progressive, mild-to-moderate hearing loss as the clinical characteristics of OTOG-associated hearing loss. These findings will contribute to a better understanding of the clinical features of OTOG-associated HL and will be useful in clinical practice.
Collapse
Affiliation(s)
- Yasuhiro Arai
- Department of Otorhinolaryngology-Head and Neck Surgery, Yokohama City University School of Medicine, Yokohama 236-0004, Japan;
| | - Shin-ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Shinichi Goto
- Department of Otorhinolaryngology Head and Neck Surgery, Hirosaki University School of Medicine, Hirosaki 036-8562, Japan;
| | - Yumiko Kobayashi
- Department of Otorhinolaryngology and Head and Neck Surgery, Iwate Medical University, Morioka 028-3694, Japan;
| | - Yohei Honkura
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University School of Medicine, Sendai 980-8575, Japan;
| | - Akira Ganaha
- Department of Otorhinolaryngology, International University of Health and Welfare, Narita Hospital, Narita 286-0124, Japan;
| | - Kotaro Ishikawa
- Department of Otolaryngology, National Rehabilitation Center for Persons with Disabilities, Tokorozawa 359-8555, Japan;
| | - Shin-ichiro Oka
- Department of Otorhinolaryngology, International University of Health and Welfare, Mita Hospital, Tokyo 108-8329, Japan;
| | - Hiroshi Futagawa
- Department of Medical Genetics, Tokyo Metropolitan Children’s Medical Center, Tokyo 183-8561, Japan
| | - Mayuri Okami
- Department of Otorhinolaryngology, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Fumio Takada
- Department of Medical Genetics and Genomics, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan;
| | - Kyoko Nagai
- TAKASAKI Ear Nose and Throat Clinic, Takasaki 370-0031, Japan;
| | - Tomoko Esaki
- Department of Otolaryngology, Aichi Children’s Health and Medical Center, Obu 474-8710, Japan;
| | - Takayuki Okano
- Department of Otolaryngology, Fujita Health University Bantane Hospital, Nagoya 454-8509, Japan;
| | - Yumi Ohta
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita 565-0871, Japan;
| | - Shin Masuda
- Department of Pediatric Rehabilitation, Hiroshima Prefectural Hospital, Hiroshima 734-8530, Japan;
| | - Kentaro Egusa
- Department of Otorhinolaryngology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima 730-8518, Japan;
| | - Masato Teraoka
- Department of Otolaryngology, Head and Neck Surgery, Ehime University Graduate School of Medicine, Toon 791-0295, Japan;
| | - Kazuma Sugahara
- Department of Otolaryngology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan;
| | - Shin-ichi Usami
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| |
Collapse
|
24
|
Otsuka S, Morimoto C, Nishio SY, Morita S, Kikuchi D, Takahashi M, Kumakawa K, Arai Y, Sano H, Yoshimura H, Yamamoto N, Kondo S, Hasegawa M, Nishi T, Kitahara T, Usami SI. The Heterozygous p.A684V Variant in the WFS1 Gene Is a Mutational Hotspot Causing a Severe Hearing Loss Phenotype. Genes (Basel) 2025; 16:57. [PMID: 39858604 PMCID: PMC11764508 DOI: 10.3390/genes16010057] [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: 12/16/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES A heterozygous mutation in the WFS1 gene is responsible for autosomal dominant non-syndromic hearing loss (DFNA6/14/38) and Wolfram-like syndrome, which is characterized by bilateral sensorineural hearing loss with optic atrophy and/or diabetes mellitus. However, detailed clinical features for the patients with the heterozygous p.A684V variant remain unknown. METHODS We report the clinical details of 14 cases with a heterozygous p.A684V variant in the WFS1 gene identified from target resequencing analysis of 63 previously reported deafness genes by next-generation sequencing of 15,684 hearing loss patients (mean age 27.5 ± 23.1 years old, 6574 male, 8612 female and 498 for whom information was unavailable). RESULTS Among the 14 patients from 13 families with the p.A684V variant, nine were sporadic cases. In addition, we confirmed de novo occurrence of this variant in seven families. This result strongly supports the notion that this variant was located on a mutational hotspot. When comparing previously reported cases of autosomal dominant WFS1 gene-associated hearing loss, most of the patients in this study showed severe-to-profound bilateral sensorineural hearing loss (genotype-phenotype correlation). Two patients had optic atrophy, while the others did not have any other complications. CONCLUSIONS The identified heterozygous p.A684V variant appears to be a hotspot mutation and likely to cause severe-to-profound hearing loss in early childhood. Cochlear implantation is considered favorable in cases of hearing impairment due to this variant.
Collapse
Affiliation(s)
- Shintaro Otsuka
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara 634-8522, Japan; (C.M.); (T.K.)
| | - Chihiro Morimoto
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara 634-8522, Japan; (C.M.); (T.K.)
| | - Shin-ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Shinya Morita
- Department of Otolaryngology-Head and Neck Surgery, Hokkaido University, Sapporo 060-8648, Japan;
| | - Daisuke Kikuchi
- Department of Otolaryngology, Fukushima Medical University, Fukushima 960-1295, Japan;
| | - Masahiro Takahashi
- Department of Otorhinolaryngology, International University of Health and Welfare Mita Hospital, Tokyo 108-8329, Japan;
| | - Kozo Kumakawa
- Department of Otolaryngology, Akasaka Toranomon Clinic, Tokyo 107-0052, Japan;
| | - Yasuhiro Arai
- Department of Otorhinolaryngology, Head and Neck Surgery, Yokohama City University, Yokohama 236-0004, Japan;
| | - Hajime Sano
- Department of Otorhinolaryngology and Head & Neck Surgery, Kitasao University, Sagamihara 252-0375, Japan;
| | - Hidekane Yoshimura
- Department of Otorhinolaryngology-Head and Neck Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Norio Yamamoto
- Department of Otolaryngology, Kobe City Medical Center General Hospital, Kobe 650-0047, Japan;
| | - Shunsuke Kondo
- Department of Otorhinolaryngology, Head and Neck Surgery, University of the Ryukyus, Okinawa 903-0215, Japan;
| | - Mari Hasegawa
- Department of Pediatrics, Nara Medical University, Kashihara 634-8522, Japan;
| | - Tomo Nishi
- Department of Ophthalmology, Nara Medical University, Kashihara 634-8522, Japan;
| | - Tadashi Kitahara
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara 634-8522, Japan; (C.M.); (T.K.)
| | - Shin-ichi Usami
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| |
Collapse
|
25
|
Drackley A, Somerville C, Arnaud P, Baudhuin LM, Hanna N, Kluge ML, Kotzer K, Boileau C, Bronicki L, Callewaert B, Cecchi A, Dietz H, Guo D, Harris S, Jarinova O, Lindsay M, Little L, Loeys B, MacCarrick G, Meester J, Milewicz D, Morisaki T, Morisaki H, Murdock D, Renard M, Richer J, Robert L, Ouzounian M, Van Laer L, De Backer J, Muiño-Mosquera L. Interpretation and classification of FBN1 variants associated with Marfan syndrome: consensus recommendations from the Clinical Genome Resource's FBN1 variant curation expert panel. Genome Med 2024; 16:154. [PMID: 39741318 DOI: 10.1186/s13073-024-01423-3] [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: 01/26/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND In 2015, the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) developed standardized variant curation guidelines for Mendelian disorders. Although these guidelines have been widely adopted, they are not gene- or disease-specific. To mitigate classification discrepancies, the Clinical Genome Resource FBN1 variant curation expert panel (VCEP) was established in 2018 to develop adaptations to the ACMG/AMP criteria for FBN1 in association with Marfan syndrome. METHODS The specific recommendations were developed through literature review, surveys, online expert panel discussions, and pilot testing of a set of 60 different variants. Consensus among experts was considered reached if at least 75% of the members agreed with a given rule specification. The final set of rules received approval from the ClinGen Sequence Variant Interpretation Working Group. RESULTS The developed specifications introduce modifications to 14 of the 28 ACMG/AMP evidence criteria and deem 6 criteria non-applicable. Some of these specifications include refining the minor allele frequency thresholds, creating a FBN1-specific flowchart for PVS1, defining functional domains of the protein, developing a point-based system of counting probands and instances of de novo occurrences, recommending a points-based method of accounting for family segregation data, and clarifying the applicable functional assays that should be considered. To date, this VCEP has curated 120 variants which have been deposited to ClinVar with the 3-star review status. CONCLUSIONS Establishing specific adaptations for FBN1 has provided a framework to foster greater classification concordance among clinical laboratories, ultimately improving clinical care for patients with Marfan syndrome.
Collapse
Affiliation(s)
- A Drackley
- Department of Pathology & Laboratory Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - C Somerville
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - P Arnaud
- Genetics Department, Hôpital Bichat, Université Paris Cité, Paris, France
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
| | - L M Baudhuin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - N Hanna
- Genetics Department, Hôpital Bichat, Université Paris Cité, Paris, France
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
| | - M L Kluge
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - K Kotzer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - C Boileau
- Genetics Department, Hôpital Bichat, Université Paris Cité, Paris, France
| | - L Bronicki
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - B Callewaert
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - A Cecchi
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Dietz
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D Guo
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - S Harris
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - O Jarinova
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - M Lindsay
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - L Little
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - B Loeys
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre of Medical Genetics, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - G MacCarrick
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J Meester
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre of Medical Genetics, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - D Milewicz
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - T Morisaki
- IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo, Japan
| | - H Morisaki
- IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo, Japan
- Department of Medical Genetics, Sakakibara Heart Institute, Fuchu, Tokyo, Japan
| | - D Murdock
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - M Renard
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - J Richer
- Department of Medical Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - L Robert
- Department of Cardiology, Guy's and St Thomas' Foundation Trust, London, UK
| | - M Ouzounian
- Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - L Van Laer
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre of Medical Genetics, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - J De Backer
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Cardiology, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - L Muiño-Mosquera
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France.
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium.
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium.
- Division of Paediatric Cardiology, Department of Paediatrics, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
26
|
Buonfiglio P, Bruque C, Salatino L, Lotersztein V, Pace M, Grinberg S, Elgoyhen A, Plazas P, Dalamón V. In silico and in vivo analyses of a novel variant in MYO6 identified in a family with postlingual non-syndromic hearing loss from Argentina. NAR Genom Bioinform 2024; 6:lqae162. [PMID: 39664812 PMCID: PMC11632615 DOI: 10.1093/nargab/lqae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Hereditary hearing loss stands as the most prevalent sensory disorder, with over 124 non-syndromic genes and approximately 400 syndromic forms of deafness identified in humans. The clinical presentation of these conditions spans a spectrum, ranging from mild to profound hearing loss. The aim of this study was to identify the genetic cause of hearing loss in a family and functionally validate a novel variant identified in the MYO6 gene. After Whole Exome Sequencing analysis, the variant c.2775G>C p.Arg925Ser in MYO6 was detected in a family with postlingual non-syndromic hearing loss. By protein modeling a change in the electrostatic charge of the single alpha helix domain surface was revealed. Through a knockdown phenotype rescue assay in zebrafish, the detrimental effects of the identified variant on the auditory system was determined. These findings underscore the significance of a comprehensive approach, integrating both in silico and in vivo strategies, to ascertain the pathogenicity of this candidate variant. Such an approach has demonstrated its effectiveness in achieving an accurate genetic diagnosis and in promoting a more profound comprehension of the mechanisms that underlie the pathophysiology of hearing.
Collapse
Affiliation(s)
- Paula I Buonfiglio
- Laboratorio de Fisiología y Genética de la Audición, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
| | - Carlos D Bruque
- Unidad de Conocimiento Traslacional Hospitalaria Patagónica, Hospital de Alta Complejidad SAMIC, El Calafate, Provincia de Santa Cruz, 9405, Argentina
| | - Lucía Salatino
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Vanesa Lotersztein
- Servicio de Genética, Hospital Militar Central “Dr. Cosme Argerich”, Ciudad Autónoma de Buenos Aires, C1426, Argentina
| | - Mariela Pace
- Laboratorio de Fisiología y Genética de la Audición, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
| | - Sofia Grinberg
- Laboratorio de Fisiología y Genética de la Audición, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
| | - Ana B Elgoyhen
- Laboratorio de Fisiología y Genética de la Audición, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Paola V Plazas
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Viviana Dalamón
- Laboratorio de Fisiología y Genética de la Audición, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
| |
Collapse
|
27
|
Giesemann A, Schöner-Heinisch A, Götz F, Steinemann D, Lesinski-Schiedat A, Warnecke A, Lanfermann H, Hartmann H, Döring K. Hamartomas of the Tuber Cinereum Associated with X-Linked Deafness Show Signs of Pubertas Tarda Instead of Pubertas Praecox and No Gelastic Seizures-Long-Term Follow-Up of 12 Years. Neuropediatrics 2024; 55:366-374. [PMID: 39084244 DOI: 10.1055/s-0044-1788730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
PURPOSE Hamartomas of tuber cinereum present as ectopic tissue in the hypothalamic region. Clinically, the usual hypothalamic hamartomas manifest themself by gelastic seizures and pubertas praecox. We observed an increased coincidence of the presence of X-linked recessive deafness DFNX2 (DFN3) and a hamartoma of the tuber cinereum. Initially five patients presented with hearing loss in childhood, two additional were already adults, not showing any characteristic symptoms for a hamartoma but signs of delayed puberty. METHODS Seven patients who underwent computed tomography imaging due to a sensorineural hearing loss and had a hamartoma of the tuber cinereum in addition to X-linked deafness DFNX2 (DFN3) were included in a retrospective study. Patients underwent initial neurologic, endocrinologic, and genetic evaluation. Long-term follow-up was performed after 10 to 12 years. RESULTS The average age at the initial exam was 12.9 years (range 4-29). All patients genetically proven nonsyndromic, X-linked deafness associated with the POU3F4 gene. Three out of six patients presented signs of delayed puberty. None of all seven showed any evidence of pubertas praecox or gelastic seizures at mean age of 17 years (range 17-29 years) at any time. CONCLUSION Hamartomas of tuber cinereum are often coincident with DFNX2. Clinically, half of the cases are-in contrary to the usual pubertas praecox-associated with growth hormone deficiency and delayed puberty, in the sense of pubertas tarda, when coincident. Clinicians' and radiologists' knowledge and awareness of this rare combination are crucial to identify children early enough for hormone-sensitive treatment.
Collapse
Affiliation(s)
- Anja Giesemann
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover Niedersachsen, Germany
| | - Anja Schöner-Heinisch
- Institute for Human Genetics, Hannover Medical School, Hannover, Niedersachsen, Germany
| | - Friedrich Götz
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover Niedersachsen, Germany
| | - Doris Steinemann
- Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Niedersachsen, Germany
| | - Anke Lesinski-Schiedat
- Institute for Otorhinolaryngology, Hannover Medical School, Hannover, Niedersachsen, Germany
| | - Athanasia Warnecke
- Institute for Otorhinolaryngology, Hannover Medical School, Hannover, Niedersachsen, Germany
| | - Heinrich Lanfermann
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover Niedersachsen, Germany
| | - Hans Hartmann
- Department of Paediatrics, Hannover Medical School, Hannover, Niedersachsen, Germany
| | - Katja Döring
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover Niedersachsen, Germany
| |
Collapse
|
28
|
Alerasool M, Eslahi A, Vona B, Kahaei MS, Mojaver NK, Rajati M, Pasdar A, Ghasemi MM, Saburi E, Ardehaie RM, Aval MH, Tale MR, Nourizadeh N, Afzalzadeh MR, Niknezhad HT, Mojarrad M. Genetic landscape of hearing loss in prelingual deaf patients of eastern Iran: Insights from exome sequencing analysis. Clin Genet 2024; 106:693-701. [PMID: 39107234 DOI: 10.1111/cge.14599] [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: 04/17/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 08/09/2024]
Abstract
Hearing loss is one of the most prevalent genetic disorders in humans. Locus and allelic heterogeneity cause fundamental challenges in hearing loss genetic diagnosis and management of patients and their families. This study examined the genetic profile of patients with prelingual hearing loss who were referred to the Genetic Foundation of Khorasan Razavi spanning over a decade. Deleterious variants in GJB2 were evaluated through Sanger sequencing among 745 non-syndromic hearing loss patients. Furthermore, exome sequencing was applied in 250 patients with negative GJB2 sequencing results and 30 patients with syndromic hearing loss. The findings revealed a relatively low frequency of GJB2 variants among the studied patients. Exome sequencing successfully identified the genetic causes of hearing loss in 70% of the patients. Moreover, variants in 10 genes, namely SLC26A4, MYO15A, TMPRSS3, TMC1, OTOF, CDH23, PJVK, MYO7A, TECTA, and PCDH15, accounted for 66% of the positive exome sequencing findings in this study. At least three prevalent founder alleles in the hearing-impaired population of eastern Iran were identified. This study emphasizes the efficiency of exome sequencing as a powerful tool for determining the etiology of prelingual hearing loss in the eastern Iranian population.
Collapse
Affiliation(s)
- Masoome Alerasool
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Genetic Laboratory, Genetic Foundation of Khorasan Razavi, Mashhad, Iran
| | - Atieh Eslahi
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Mir Salar Kahaei
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohsen Rajati
- Sinus and Surgical Endoscopic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
| | - Alireza Pasdar
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mehdi Ghasemi
- Sinus and Surgical Endoscopic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
| | - Ehsan Saburi
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mousavi Ardehaie
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Hadadi Aval
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
- Paramedical and Rehabilitation Faculty, University of Medical Sciences, Mashhad, Iran
- Chochlear Implantation Division, Khorasan Cochlear Implant Center, Mashhad, Iran
| | - Mohammad Reza Tale
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
- Chochlear Implantation Division, Khorasan Cochlear Implant Center, Mashhad, Iran
| | - Navid Nourizadeh
- Sinus and Surgical Endoscopic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
| | - Mohammad Reza Afzalzadeh
- Sinus and Surgical Endoscopic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
| | - Hamid Tayarani Niknezhad
- Chochlear Implantation Department, Ear Foundation Research Center, Mashhad, Iran
- Chochlear Implantation Division, Khorasan Cochlear Implant Center, Mashhad, Iran
| | - Majid Mojarrad
- Faculty of Medicine, Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Genetic Laboratory, Genetic Foundation of Khorasan Razavi, Mashhad, Iran
| |
Collapse
|
29
|
Rezaie N, Ghazanfari SS, Mousavikia SM, Mansour Samaei N, Oladnabi M, Sarli A, Khosravi T. A novel frameshift variant in the TMPRSS3 gene causes nonsyndromic hearing loss in a consanguineous family. BMC Med Genomics 2024; 17:283. [PMID: 39614311 PMCID: PMC11607894 DOI: 10.1186/s12920-024-02055-7] [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: 07/16/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Hearing Loss (HL) is the most common sensorineural condition in humans. Mutations in the TMPRSS3 gene (DNFB8/10 locus) have been linked to autosomal recessive non-syndromic hearing loss (ARNSHL). METHODS Whole-exome sequencing (WES) was utilized to identify disease-causing variants in a proband from Iran with ARNSHL who presented clinically with sensorineural, bilateral, and prelingual HL. The pathogenicity and novelty of the identified variant were assessed using various databases. A co-segregation study was also performed to confirm the presence of the variant in the proband's parents. Additionally, the secondary and tertiary structures of the mutant TMPRSS3 protein were predicted using bioinformatics tools. Furthermore, a global mutational spectrum of TMPRSS3 was created and statistically analyzed. The Iranome database was also used to identify other putative mutations in the TMPRSS3 gene in the Iranian population. RESULTS We identified a novel homozygous single nucleotide deletion in TMPRSS3 (c.297delA, p.Asp100ThrfsTer52) in the proband. This is the first report of this mutation in a patient with ARNSHL. Sanger sequencing confirmed that this variant co-segregated from the proband's parents. Bioinformatic tools classified this novel variant as likely pathogenic. Additionally, 49.55% of families with TMPRSS3-related HL patients were shown to have consanguinity, consistent with our study. The Iranome database also revealed the c.268G > A variant as a putative novel mutation in TMPRSS3. CONCLUSION This research expanded the pool of evidence regarding the association between mutations in the TMPRSS3 gene and ARNSHL. The finding confirmed that a single nucleotide deletion caused HL in the proband, suggesting that genetic testing, such as WES, is a robust technique for diagnosing patients with this condition.
Collapse
Affiliation(s)
- Nahid Rezaie
- Department of Medical Genetics, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Seyede Mahsa Mousavikia
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nader Mansour Samaei
- Department of Medical Genetics, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
- Gorgan Congenital Malformations Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
- Department of Cytogenetics, Genome Genetics Laboratory, Gorgan, Golestan, Iran.
| | - Morteza Oladnabi
- Department of Medical Genetics, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
- Gorgan Congenital Malformations Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Abdolazim Sarli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Teymoor Khosravi
- Department of Medical Genetics, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
30
|
Nam J, Jung H, Won D, Gee HY, Choi JY, Jung J. Natural History of Auditory Function in Patients with Alport Syndrome: A Case Series Study. J Clin Med 2024; 13:6639. [PMID: 39597783 PMCID: PMC11594709 DOI: 10.3390/jcm13226639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/17/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Alport syndrome (AS) is a genetic disorder characterized by progressive renal disease, ocular abnormalities, and sensorineural hearing loss. However, the audiological profile of patients with AS remains elusive. Thus, this study aims to evaluate the natural history of auditory function in patients with AS. Methods: Exome or targeted sequencing for deafness genes was performed to confirm the pathogenic variants in patients with AS. Results: We identified fifteen individuals with AS who carried pathogenic variants of COL4A3, COL4A4, or COL4A5. Among fifteen, twelve (80%) showed hematuria, and six (40%) showed proteinuria. The patients exhibited bilateral sensorineural hearing loss, which was progressive and symmetric. The hearing thresholds increased according to age and plateaued at the level of 53 dB HL, indicating the hearing loss did not reach the severe-to-moderate level. The auditory dysfunction showed a distinct natural history depending on the inheritance pattern, but there was no remarkable difference between males and females among X-linked AS. Conclusions: Auditory dysfunction in AS is progressive up to the level of moderate hearing loss. Precise auditory rehabilitation for patients with AS is warranted depending on the inheritance pattern and genetic predisposition.
Collapse
Affiliation(s)
- Juyun Nam
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.N.); (H.J.)
| | - Hyuntaek Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.N.); (H.J.)
| | - Dongju Won
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.N.); (H.J.)
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.N.); (H.J.)
| |
Collapse
|
31
|
Jung J, Joo SY, Min H, Roh JW, Kim KA, Ma JH, Rim JH, Kim JA, Kim SJ, Jang SH, Koh YI, Kim HY, Lee H, Kim BC, Gee HY, Bok J, Choi JY, Seong JK. MYH1 deficiency disrupts outer hair cell electromotility, resulting in hearing loss. Exp Mol Med 2024; 56:2423-2435. [PMID: 39482536 PMCID: PMC11612406 DOI: 10.1038/s12276-024-01338-4] [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: 03/04/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 11/03/2024] Open
Abstract
Myh1 is a mouse deafness gene with an unknown function in the auditory system. Hearing loss in Myh1-knockout mice is characterized by an elevated threshold for the auditory brainstem response and the absence of a threshold for distortion product otoacoustic emission. Here, we investigated the role of MYH1 in outer hair cells (OHCs), crucial structures in the organ of Corti responsible for regulating cochlear amplification. Direct whole-cell voltage-clamp recordings of OHCs revealed that prestin activity was lower in Myh1-knockout mice than in wild-type mice, indicating abnormal OHC electromotility. We analyzed whole-exome sequencing data from 437 patients with hearing loss of unknown genetic causes and identified biallelic missense variants of MYH1 in five unrelated families. Hearing loss in individuals harboring biallelic MYH1 variants was non-progressive, with an onset ranging from congenital to childhood. Three of five individuals with MYH1 variants displayed osteopenia. Structural prediction by AlphaFold2 followed by molecular dynamic simulations revealed that the identified variants presented structural abnormalities compared with wild-type MYH1. In a heterogeneous overexpression system, MYH1 variants, particularly those in the head domain, abolished MYH1 functions, such as by increasing prestin activity and modulating the membrane traction force. Overall, our findings suggest an essential function of MYH1 in OHCs, as observed in Myh1-deficient mice, and provide genetic evidence linking biallelic MYH1 variants to autosomal recessive hearing loss in humans.
Collapse
Affiliation(s)
- Jinsei Jung
- Department of Otorhinolaryngology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea
| | - Sun Young Joo
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyehyun Min
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Won Roh
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Republic of Korea
| | - Kyung Ah Kim
- Department of Nanobioengineering, Incheon National University, Incheon, Korea
| | - Ji-Hyun Ma
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - John Hoon Rim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Ah Kim
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Jin Kim
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyun Jang
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ik Koh
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Republic of Korea
| | - Hye-Youn Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Republic of Korea
| | - Byoung Choul Kim
- Department of Nanobioengineering, Incheon National University, Incheon, Korea
| | - Heon Yung Gee
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea.
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Republic of Korea.
| | - Jinwoong Bok
- Department of Otorhinolaryngology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea.
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jae Young Choi
- Department of Otorhinolaryngology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Institute for Lee Won Sang Yonsei Ear Science, Seoul, Republic of Korea.
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea.
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Velde HM, Vaseghi-Shanjani M, Smits JJ, Ramakrishnan G, Oostrik J, Wesdorp M, Astuti G, Yntema HG, Hoefsloot L, Lanting CP, Huynen MA, Lehman A, Turvey SE, Pennings RJE, Kremer H. Exome variant prioritization in a large cohort of hearing-impaired individuals indicates IKZF2 to be associated with non-syndromic hearing loss and guides future research of unsolved cases. Hum Genet 2024; 143:1379-1399. [PMID: 39406892 PMCID: PMC11522133 DOI: 10.1007/s00439-024-02706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024]
Abstract
Although more than 140 genes have been associated with non-syndromic hereditary hearing loss (HL), at least half of the cases remain unexplained in medical genetic testing. One reason is that pathogenic variants are located in 'novel' deafness genes. A variant prioritization approach was used to identify novel (candidate) genes for HL. Exome-wide sequencing data were assessed for subjects with presumed hereditary HL that remained unexplained in medical genetic testing by gene-panel analysis. Cases in group AD had presumed autosomal dominantly inherited HL (n = 124), and in group AR, presumed autosomal recessive HL (n = 337). Variants in known and candidate deafness genes were prioritized based on allele frequencies and predicted effects. Selected variants were tested for their co-segregation with HL. Two cases were solved by variants in recently identified deafness genes (ABHD12, TRRAP). Variant prioritization also revealed potentially causative variants in candidate genes associated with recessive and X-linked HL. Importantly, missense variants in IKZF2 were found to co-segregate with dominantly inherited non-syndromic HL in three families. These variants specifically affected Zn2+-coordinating cysteine or histidine residues of the zinc finger motifs 2 and 3 of the encoded protein Helios. This finding indicates a complex genotype-phenotype correlation for IKZF2 defects, as this gene was previously associated with non-syndromic dysfunction of the immune system and ICHAD syndrome, including HL. The designed strategy for variant prioritization revealed that IKZF2 variants can underlie non-syndromic HL. The large number of candidate genes for HL and variants therein stress the importance of inclusion of family members for variant prioritization.
Collapse
Affiliation(s)
- Hedwig M Velde
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Maryam Vaseghi-Shanjani
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Jeroen J Smits
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Jaap Oostrik
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Mieke Wesdorp
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Galuh Astuti
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Helger G Yntema
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Lies Hoefsloot
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cris P Lanting
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboudumc, Nijmegen, The Netherlands
| | - Anna Lehman
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Ronald J E Pennings
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Hannie Kremer
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
33
|
Antunes LN, Dias AMM, Schiavo BC, Mendes BCA, Bertola DR, Lezirovitz K, Mingroni-Netto RC. Genetic heterogeneity in autosomal recessive hearing loss: a survey of Brazilian families. Front Genet 2024; 15:1409306. [PMID: 39498320 PMCID: PMC11532063 DOI: 10.3389/fgene.2024.1409306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/12/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Hearing loss is a frequent sensory impairment type in humans, with about 50% of prelingual cases being attributed to genetic factors. Autosomal recessive hearing loss (ARHL) exhibits great locus heterogeneity and is responsible for 70%-80% of hereditary nonsyndromic cases. Methods A total of 90 unrelated Brazilian individuals were selected for having hearing loss of presumably autosomal recessive inheritance, either born from consanguineous marriages or belonging to families with two or more affected individuals in the sibship and most cases were of normal hearing parents. In all cases, common pathogenic variants in GJB2 (c.35delG), GJB6 [del(GJB6-D13S1830) and del(GJB6-D13S1854)] and MT-RNR1 (m.1555A>G) were discarded and most were previously assessed by complete Sanger sequencing of GJB2. Their genetic material was analyzed through next-generation sequencing, targeting 99 hearing loss-related genes and/or whole exome sequencing. Results In 32 of the 90 probands (36,7%) causative variants were identified, with autosomal recessive inheritance confirmed in all, except for two cases due to dominant variants (SIX1 and P2RX2). Thirty-nine different causative variants were found in 24 different known hearing loss-associated genes, among which 10 variants are novel, indicating wide genetic heterogeneity in the sample, after exclusion of common pathogenic variants. Despite the genetic heterogeneity, some genes showed greater contribution: GJB2, CDH23, MYO15A, OTOF, and USH2A. Conclusion The present results confirmed that next-generation sequencing is an effective tool for identifying causative variants in autosomal recessive hearing loss. To our knowledge, this is the first report of next-generation sequencing being applied to a large cohort of pedigrees with presumable autosomal recessive hearing loss in Brazil and South America.
Collapse
Affiliation(s)
- Larissa Nascimento Antunes
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Alex Marcel Moreira Dias
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Beatriz Cetalle Schiavo
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Beatriz C. A. Mendes
- Divisão de Educação e Reabilitação dos Distúrbios da Comunicação da Pontifícia Universidade Católica de São Paulo, São Paulo, Brazil
| | - Debora Romeo Bertola
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Unidade de Genética do Instituto da Criança, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Karina Lezirovitz
- Laboratório de Otorrinolaringologia/LIM32 - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Regina Célia Mingroni-Netto
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Kraatari-Tiri M, Pykälainen T, Pohjola P, Häkli S, Rahikkala E. Clinical and genetic characterisation of childhood-onset sensorineural hearing loss reveal associated phenotypes and enrichment of pathogenic founder mutations in the Finnish population. Int J Audiol 2024:1-6. [PMID: 39422539 DOI: 10.1080/14992027.2024.2402840] [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: 08/11/2023] [Revised: 07/31/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE To examine the clinical and genetic characteristics of childhood-onset bilateral sensorineural hearing loss (SNHL) in Finland. DESIGN Retrospective analysis. STUDY SAMPLE A total of 249 children younger than 18 years were diagnosed with bilateral SNHL in Oulu University Hospital, Finland, from 2017 to 2022. RESULTS Pathogenic or likely pathogenic gene variants or chromosome abnormalities explaining SNHL were identified in 41% (N = 101/249) of children. Likely causative variants were more commonly identified in patients with severe SNHL than in those with moderate or mild SNHL. Our study identified likely causative gene variants in 24 different genes and six different likely causative chromosome abnormalities, demonstrating the genetic heterogeneity of SNHL. Population-enriched founder mutations were identified in the CABP2, CLRN1, MYO7A, SUCLA2, TMC1, and TWNK genes. A significant number of patients had associated phenotypes, including global developmental delay or intellectual disability (16%), language disorder (20%), ophthalmological abnormalities (16%), or malformations other than those involving the ear (10%). CONCLUSIONS SNHL is genetically and clinically heterogeneous. Pathogenic variants in GJB2 were the most common. Several population-enriched variants were identified as causing SNHL in the northern Finnish population. Associated medical phenotypes are common and should be taken into account in patients' follow-up and treatment.
Collapse
Affiliation(s)
- Minna Kraatari-Tiri
- Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland
- Research Unit of Clinical Medicine and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Tyrni Pykälainen
- Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland
- Research Unit of Clinical Medicine and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Otorhinolaryngology and Phoniatrics, Oulu University Hospital, Oulu, Finland
| | - Pia Pohjola
- Department of Genomics, Turku University Hospital, Turku, Finland
| | - Sanna Häkli
- Research Unit of Clinical Medicine and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Otorhinolaryngology and Phoniatrics, Oulu University Hospital, Oulu, Finland
| | - Elisa Rahikkala
- Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland
- Research Unit of Clinical Medicine and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Genomics, Turku University Hospital, Turku, Finland
| |
Collapse
|
35
|
Ahmed AN, Rawlins LE, Khan N, Jan Z, Ubeyratna N, Voutsina N, Azeem A, Khan S, Baple EL, Crosby AH, Saleha S. Expanding the genetic spectrum of hereditary motor sensory neuropathies in Pakistan. BMC Neurol 2024; 24:394. [PMID: 39415096 PMCID: PMC11481789 DOI: 10.1186/s12883-024-03882-y] [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: 12/13/2023] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Hereditary motor and sensory neuropathy (HMSN) refers to a group of inherited progressive peripheral neuropathies characterized by reduced nerve conduction velocity with chronic segmental demyelination and/or axonal degeneration. HMSN is highly clinically and genetically heterogeneous with multiple inheritance patterns and phenotypic overlap with other inherited neuropathies and neurodegenerative diseases. Due to this high complexity and genetic heterogeneity, this study aimed to elucidate the genetic causes of HMSN in Pakistani families using Whole Exome Sequencing (WES) for variant identification and Sanger sequencing for validation and segregation analysis, facilitating accurate clinical diagnosis. METHODS Families from Khyber Pakhtunkhwa with at least two members showing HMSN symptoms, who had not previously undergone genetic analysis, were included. Referrals for genetic investigations were based on clinical features suggestive of HMSN by local neurologists. WES was performed on affected individuals from each family, with Sanger sequencing used to validate and analyze the segregation of identified variants among family members. Clinical data including age of onset were assessed for variability among affected individuals, and the success rate of genetic diagnosis was compared with existing literature using proportional differences and Cohen's h. RESULTS WES identified homozygous pathogenic variants in GDAP1 (c.310 + 4 A > G, p.?), SETX (c.5948_5949del, p.(Asn1984Profs*30), IGHMBP2 (c.1591 C > A, p.(Pro531Thr) and NARS1 (c.1633 C > T, p.(Arg545Cys) as causative for HMSN in five out of nine families, consistent with an autosomal recessive inheritance pattern. Additionally, in families with HMSN, a SETX variant was found to cause cerebellar ataxia, while a NARS1 variant was linked to intellectual disability. Based on American College of Medical Genetics and Genomics criteria, the GDAP1 variant is classified as a variant of uncertain significance, while variants in SETX and IGHMBP2 are classified as pathogenic, and the NARS1 variant is classified as likely pathogenic. The age of onset ranged from 1 to 15 years (Mean = 5.13, SD = 3.61), and a genetic diagnosis was achieved in 55.56% of families with HMSN, with small effect sizes compared to previous studies. CONCLUSIONS This study expands the molecular genetic spectrum of HMSN and HMSN plus type neuropathies in Pakistan and facilitates accurate diagnosis, genetic counseling, and clinical management for affected families.
Collapse
Affiliation(s)
- Asif Naveed Ahmed
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, 26000, Pakistan
| | - Lettie E Rawlins
- Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK.
- Peninsula Clinical Genetics Service, Royal Devon & Exeter Hospital (Heavitree), Exeter, UK.
| | - Niamat Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, 26000, Pakistan
| | - Zakir Jan
- Department of Neurology, Pakistan Institute of Medical Science, Islamabad, 44000, Pakistan
| | - Nishanka Ubeyratna
- Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Nikol Voutsina
- Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Arfa Azeem
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, 26000, Pakistan
| | - Saadullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, 26000, Pakistan
| | - Emma L Baple
- Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Andrew H Crosby
- Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Shamim Saleha
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, 26000, Pakistan.
| |
Collapse
|
36
|
Tian Y, Liu M, Lu Y, Zhao X, Yan Z, Sun Y, Ma J, Tang W, Wang H, Xu H. Exonic Deletions and Deep Intronic Variants of the SLC26A4 Gene Contribute to the Genetic Diagnosis of Unsolved Patients With Enlarged Vestibular Aqueduct. Hum Mutat 2024; 2024:8444122. [PMID: 40225947 PMCID: PMC11919234 DOI: 10.1155/2024/8444122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 04/15/2025]
Abstract
Enlarged vestibular aqueduct (EVA) is a frequently occurring inner ear malformation that associates with sensorineural hearing loss (SNHL), with SLC26A4 being the responsible gene. Based on multiplex PCR enrichment and sequencing of the exonic and flanking regions of the SLC26A4 gene, we developed a panel specifically for EVA and found that up to 95% of EVA patients in our Chinese cohorts carried biallelic SLC26A4 pathogenic variants (M2). In this study, we tried to investigate the genetic etiology of 13 previously undiagnosed EVA patients with monoallelic (M1) or none (M0) SLC26A4 variant using a stepwise approach, including copy number variation (CNV) analysis of multiplex PCR enrichment and next-generation sequencing data, single-molecule real-time (SMRT) sequencing of the whole SLC26A4 gene, whole exome sequencing (WES), and whole genome sequencing (WGS). CNV analysis revealed deletions in Exons 1-3, Exons 5-6, and Exons 9-10 of the SLC26A4 gene in seven patients, and SMRT sequencing identified the same heterozygous deep intronic variant (NM_000441.2:c.304+941C>T) in two patients, resulting in a final diagnosis in 9/13 patients. Notably, the variants of Exons 9-10 deletion and c.304+941C>T have not been reported previously. We further showed that the variant c.304+941C>T led to the exonization of partial AluSz6 element (126 bp) where the variant is located through sequencing of the mRNA extracted from the blood of a heterozygous variant carrier. In conclusion, our stepwise approach improved the diagnosis rate of EVA, expanded the mutational spectrum of the SLC26A4 gene, and highlighted the contribution of exonic deletions and deep intronic variants to EVA.
Collapse
Affiliation(s)
- Yongan Tian
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou 450052, China
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Jingba Road No. 2, Zhengzhou 450014, China
| | - Mengli Liu
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou 450052, China
| | - Yu Lu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiaoyan Zhao
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou 450052, China
| | - Zhiqiang Yan
- Department of Otolaryngology Head and Neck Surgery, Hospital of the 71st Group Army/Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Yi Sun
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Central Theater Command, Wuhan 430070, China
| | - Jingyuan Ma
- Department of Otolaryngology, Henan Provincial People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Wenxue Tang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Jingba Road No. 2, Zhengzhou 450014, China
| | - Haili Wang
- Longhu Laboratory, Zhengzhou University, No. 100, Science Avenue, Zhengzhou 450001, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou 450002, China
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou 450052, China
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Jingba Road No. 2, Zhengzhou 450014, China
| |
Collapse
|
37
|
Usami SI, Nishio SY, Gavilán J, Van de Heyning P, Mertens G, Karltorp E, Skarżyński H, Hagr A, Manoj M, Staecker H, Zernotti ME, Rajan GP, Müller J, Simon F, Anderson I. HEARRING group genetic marker study: genetic background of CI patients. Acta Otolaryngol 2024; 144:542-551. [PMID: 39434647 DOI: 10.1080/00016489.2024.2416082] [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: 09/05/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND While cochlear implantation (CI) and electric acoustic stimulation (EAS) have a positive outcome in most cases, their effectiveness varies depending on the etiology of the hearing loss. Among the various etiologies, genetic factors are the leading cause of hearing loss and may impact CI and EAS outcomes. AIMS/OBJECTIVES To reveal the genetic background of the hearing loss in CI/EAS patients in each ethnic population, we undertook a multi-center study involving the genetic testing of hearing loss in CI/EAS patients from 10 centers. MATERIAL AND METHODS Saliva samples and clinical information for the patients and their family members were obtained and next-generation sequencing analysis using a panel carrying 63 deafness genes was then performed. RESULTS Genetic testing successfully identified the causative gene variants in 54.5% (48/88) of patients with pre-lingual onset hearing loss (onset under 6 years) and in 12% (12/95) of those with late-onset hearing loss (onset at 6 years or more). CONCLUSIONS AND SIGNIFICANCE We clearly indicated that genetic factors are the most common cause of hearing loss regardless of ethnic background. Saliva-based genetic testing is a useful tool for multi-center studies seeking to clarify the genetic causes of hearing loss in CI or EAS patients between countries separated by distance.
Collapse
Affiliation(s)
| | - Shin-Ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Javier Gavilán
- Department of Otorhinolaryngology, La Paz University Hospital, Madrid, Spain
| | - Paul Van de Heyning
- Department of Otorhinolaryngology Head and Neck Surgery, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | - Griet Mertens
- Department of Otorhinolaryngology Head and Neck Surgery, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | - Eva Karltorp
- Cochlear Implant Department, Karolinska University Hospital, Stockholm, Sweden
| | - Henryk Skarżyński
- Department of Teleaudiology and Screening, World Hearing Center of the Institute of Physiology and Pathology of Hearing, Kajetany, Poland
| | - Abdulrahman Hagr
- King Abdullah Ear Specialist Center, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Manikoth Manoj
- ENT Super Speciality Institute and Research Center, Kozhikode, India
| | - Hinrich Staecker
- Center for Hearing and Balance Disorders, Kansas University, Kansas City, KS, USA
| | - Mario E Zernotti
- Catholic University of Córdoba and National University of Córdoba, Córdoba, Argentina
| | - Gunesh P Rajan
- Faculty of Health Sciences and Medicine, University Hospital Lucerne, University of Lucerne, Luzern, Switzerland
- Otolaryngology, Head and Neck Surgery, Division of Surgery, Medical School University of Western Australia, Perth, Australia
| | - Joachim Müller
- Department of Otorhinolaryngology, University Hospital, LMU Munich, Munich, Germany
| | - Florian Simon
- Department of Otorhinolaryngology, University Hospital, LMU Munich, Munich, Germany
| | - Ilona Anderson
- Clinical Research Department, MED-EL GmbH, Innsbruck, Austria
| |
Collapse
|
38
|
Shearer AE. Genetic testing for pediatric sensorineural hearing loss in the era of gene therapy. Curr Opin Otolaryngol Head Neck Surg 2024; 32:352-356. [PMID: 39146193 DOI: 10.1097/moo.0000000000001005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
PURPOSE OF REVIEW To summarize indications, methods, and diagnostic yields for genetic testing for pediatric hearing loss. RECENT FINDINGS Genetic testing has become a cornerstone of clinical care for children with sensorineural hearing loss. Recent studies have shown the efficacy of gene panels and exome sequencing for any child with sensorineural hearing loss. Recent findings have underscored the importance of a diagnosis in clinical care. Clinical trials for gene therapy for hearing loss have begun. SUMMARY Genetic testing has become critical for personalized care for children with hearing loss. Recent studies have shown a 43% overall diagnostic yield for genetic testing for pediatric hearing loss, though the diagnostic yield may range from 10 to 60% depending on clinical features. Syndromic diagnoses comprise 25% of positive genetic tests for pediatric sensorineural hearing loss. While diagnostic yield is lower for children with unilateral or asymmetric sensorineural hearing loss, the likelihood of syndromic hearing loss finding is higher. An early and accurate genetic diagnosis is required for participating in clinical trials for gene therapy for hearing loss.
Collapse
Affiliation(s)
- A Eliot Shearer
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School
- Boston Children's Hospital, Department of Otolaryngology & Communication Enhancement, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Zhao Y, Long Y, Shi T, Ma X, Lian C, Wang H, Xu H, Yu L, Zhao X. Validating the splicing effect of rare variants in the SLC26A4 gene using minigene assay. BMC Med Genomics 2024; 17:233. [PMID: 39334476 PMCID: PMC11430457 DOI: 10.1186/s12920-024-02007-1] [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: 05/28/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The SLC26A4 gene is the second most common cause of hereditary hearing loss in human. The aim of this study was to utilize the minigene assay in order to identify pathogenic variants of SLC26A4 associated with enlarged vestibular aqueduct (EVA) and hearing loss (HL) in two patients. METHODS The patients were subjected to multiplex PCR amplification and next-generation sequencing of common deafness genes (including GJB2, SLC26A4, and MT-RNR1), then bioinformatics analysis was performed on the sequencing data to identify candidate pathogenic variants. Minigene experiments were conducted to determine the potential impact of the variants on splicing. RESULTS Genetic testing revealed that the first patient carried compound heterozygous variants c.[1149 + 1G > A]; [919-2 A > G] in the SLC26A4 gene, while the second patient carried compound heterozygous variants c.[2089 + 3 A > T]; [919-2 A > G] in the same gene. Minigene experiments demonstrated that both c.1149 + 1G > A and c.2089 + 3 A > T affected mRNA splicing. According to the ACMG guidelines and the recommendations of the ClinGen Hearing Loss Expert Panel for ACMG variant interpretation, these variants were classified as "likely pathogenic". CONCLUSIONS This study identified the molecular etiology of hearing loss in two patients with EVA and elucidated the impact of rare variants on splicing, thus contributing to the mutational spectrum of pathogenic variants in the SLC26A4 gene.
Collapse
Affiliation(s)
- Yixin Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
| | - Yan Long
- Department of Clinical Laboratory, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Tao Shi
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
| | - Xin Ma
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
| | - Chengyu Lian
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou, 450052, China
| | - Hanjun Wang
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou, 450052, China
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Daxuebei Road No. 40, Zhengzhou, 450052, China
| | - Lisheng Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China.
| | - Xiaotao Zhao
- Department of Clinical Laboratory, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
40
|
Rhim JW, Kim DK, Han JY, Park J. A sensorineural hearing loss harboring novel compound heterozygous variant in the TRIOBP gene: A case report. Heliyon 2024; 10:e36717. [PMID: 39296067 PMCID: PMC11408809 DOI: 10.1016/j.heliyon.2024.e36717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Background Autosomal recessive non-syndromic deafness-28 (DFNB28; OMIM #609823) specifically refers to prelingual sensorineural hearing loss (SNHL) resulting from homozygous or compound heterozygous mutations in the TRIO- and F-actin-binding protein, TRIOBP gene. In this report, we present a pediatric patient exhibiting novel compound heterozygous deleterious variants in the TRIOBP gene. Methods The auditory brainstem response result revealed both left- and right-sided deafness with a threshold of 20 dB normal hearing level in the proband. A comprehensive trio whole exome sequencing (WES) using the Celemics G-Mendeliome Whole Exome Sequencing Panel was employed. Results The WES analysis revealed compound heterozygous TRIOBP variants in the proband, namely c.1192_1195delCAACinsT/p.Gln398* classified as pathogenic and c.3661C > T/p.Arg1221Trp categorized as a variant of uncertain significance according to American College of Medical Genetics and Genomics guidelines. These variants are considered the most probable cause of the proband's SNHL. Conclusion TRIOBP isoforms are predominantly expressed in the inner ear, contributing to the formation of stereocilia rootlets. Further investigations are required to fully understand the phenotypic variability and establish the pathogenicity of the identified variant in relation to the TRIOBP gene and SNHL.
Collapse
Affiliation(s)
- Jung Woo Rhim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Dong-Kee Kim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea, Seoul, 06591, Republic of Korea
| | - Ji Yoon Han
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Joonhong Park
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, 54907, Republic of Korea
| |
Collapse
|
41
|
Feng H, Huang S, Ma Y, Yang J, Chen Y, Wang G, Han M, Kang D, Zhang X, Dai P, Yuan Y. Genomic and phenotypic landscapes of X-linked hereditary hearing loss in the Chinese population. Orphanet J Rare Dis 2024; 19:342. [PMID: 39272213 PMCID: PMC11396341 DOI: 10.1186/s13023-024-03338-z] [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: 04/02/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Hearing loss (HL) is the most common sensory birth deficit worldwide, with causative variants in more than 150 genes. However, the etiological contribution and clinical manifestations of X-linked inheritance in HL remain unclear within the Chinese HL population. In this study, we focused on X-linked hereditary HL and aimed to assess its contribution to hereditary HL and identify the genotype-phenotype relationship. METHODS We performed a molecular epidemiological investigation of X-linked hereditary HL based on next-generation sequencing and third-generation sequencing in 3646 unrelated patients with HL. We also discussed the clinical features associated with X-linked non-syndromic HL-related genes based on a review of the literature. RESULTS We obtained a diagnostic rate of 52.72% (1922/3646) among our patients; the aggregate contribution of HL caused by genes on the X chromosome in this cohort was ~ 1.14% (22/1922), and POU3F4 variants caused ~ 59% (13/22) of these cases. We found that X-linked HL was congenital or began during childhood in all cases, with representative audiological profiles or typical cochlear malformations in certain genes. Genotypic and phenotypic analyses showed that causative variants in PRPS1 and AIFM1 were mainly of the missense type, suggesting that phenotypic variability was correlated with the different effects that the replaced residues exert on structure and function. Variations in SMPX causing truncation of the protein product were associated with DFNX4, which resulted in typical audiological profiles before and after the age of 10 years, whereas nontruncated proteins typically led to distal myopathy. No phenotypic differences were identified in patients carrying POU3F4 or COL4A6 variants. CONCLUSIONS Our work constitutes a preliminary evaluation of the molecular contribution of X-linked genes in heritable HL (~ 1.14%). The 15 novel variants reported here expand the mutational spectrum of these genes. Analysis of the genotype-phenotype relationship is valuable for X-linked HL precise diagnostics and genetic counseling. Elucidation of the pathogenic mechanisms and audiological profiles of HL can also guide choices regarding treatment modalities.
Collapse
Affiliation(s)
- Haifeng Feng
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
- Chinese PLA Medical School, Beijing, 100853, China
| | - Shasha Huang
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Ying Ma
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Jinyuan Yang
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Yijin Chen
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Guojian Wang
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Mingyu Han
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Dongyang Kang
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Xin Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
| | - Pu Dai
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China.
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China.
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China.
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China.
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China.
| | - Yongyi Yuan
- Senior Department of Otolaryngology Head and Neck Surgery, the 6th Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School,, Beijing, 100853, China.
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China.
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China.
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China.
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China.
| |
Collapse
|
42
|
Choi WH, Cho Y, Cha JH, Lee DH, Jeong JG, Jung SH, Song JJ, Lee JH, Lee SY. Functional pathogenicity of ESRRB variant of uncertain significance contributes to hearing loss (DFNB35). Sci Rep 2024; 14:21215. [PMID: 39261511 PMCID: PMC11390957 DOI: 10.1038/s41598-024-70795-8] [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: 01/24/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
Advances in next-generation sequencing technologies have led to elucidation of sensorineural hearing loss genetics and associated clinical impacts. However, studies on the functional pathogenicity of variants of uncertain significance (VUS), despite their close association with clinical phenotypes, are lacking. Here we identified compound heterozygous variants in ESRRB transcription factor gene linked to DFNB35, specifically a novel splicing variant (NM_004452.4(ESRRB): c.397 + 2T>G) in trans with a missense variant (NM_004452.4(ESRRB): c.1144C>T p.(Arg382Cys)) whose pathogenicity remains unclear. The splicing variant (c.397 + 2T>G) caused exon 4 skipping, leading to premature stop codon formation and nonsense-mediated decay. The p.(Arg382Cys) variant was classified as a VUS due to its particularly higher allele frequency among East Asian population despite disease-causing in-silico predictions. However, functional assays showed that p.(Arg382Cys) variant disrupted key intramolecular interactions, leading to protein instability. This variant also reduced transcriptional activity and altered expression of downstream target genes essential for inner ear function, suggesting genetic contribution to disease phenotype. This study expanded the phenotypic and genotypic spectrum of ESRRB in DFNB35 and revealed molecular mechanisms underlying ESRRB-associated DFNB35. These findings suggest that variants with high allele frequencies can also possess functional pathogenicity, providing a breakthrough for cases where VUS, previously unexplored, could be reinterpreted by elucidating their functional roles and disease-causing characteristics.
Collapse
Affiliation(s)
- Won Hoon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yeijean Cho
- Seoul National University College of Medicine, Seoul, South Korea
| | - Ju Hyuen Cha
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dae Hee Lee
- CTCELLS, Inc., 21, Yuseong-Daero, 1205 Beon-Gil, Yuseong-Gu, Daejeon, Republic of Korea
| | - Jong Gwan Jeong
- CTCELLS, Inc., 21, Yuseong-Daero, 1205 Beon-Gil, Yuseong-Gu, Daejeon, Republic of Korea
| | - Sung Ho Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae-Jin Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
43
|
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
|
44
|
Buonfiglio PI, Izquierdo A, Pace MV, Grinberg S, Lotersztein V, Brun P, Bruque CD, Elgoyhen AB, Dalamón V. Comprehensive Approach for the Genetic Diagnosis of Patients with Waardenburg Syndrome. J Pers Med 2024; 14:906. [PMID: 39338160 PMCID: PMC11433630 DOI: 10.3390/jpm14090906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Waardenburg syndrome (WS) is a common genetic cause of syndromic hearing loss, accounting for 2-5% of congenital cases. It is characterized by hearing impairment and pigmentation abnormalities in the skin, hair, and eyes. Seven genes are associated with WS: PAX3, MITF, EDNRB, EDN3, SOX10, KITLG, and SNAI2. This study investigates the genetic causes of WS in three familial cases. Whole-exome sequencing (WES) was performed to identify single nucleotide variants (SNVs). Copy number variants (CNVs) were analyzed from the WES raw data and through multiplex ligation-dependent probe amplification (MLPA). The study identified one pathogenic SNV and two novel CNVs, corresponding to type I and type II WS patterns in the three families. The SNV, a nonsense variant (c.1198C>T p.Arg400*), was found in MITF and segregated in the affected father. The two CNVs were a deletion of exon 5 in PAX3 in a family with two affected members and a large novel deletion comprising seven genes, including SOX10, in a family with three affected members. These findings confirmed a WS diagnosis through genetic testing. The study emphasizes the importance of integrating multiple genetic testing approaches for accurate and reliable diagnosis, highlighting their role in improving patient management and providing tailored genetic counseling.
Collapse
Affiliation(s)
- Paula Inés Buonfiglio
- Laboratory of Physiology and Genetics of Hearing, Institute of Genetic Engineering and Molecular Biology "Dr. Héctor N. Torres"-National Council of Scientific and Technology (INGEBI-CONICET), Buenos Aires C1428ADN, Argentina
| | - Agustín Izquierdo
- Center for Endocrinological Research "Dr. César Bergadá" (CEDIE)-CONICET, FEI, Endocrinology División, Ricardo Gutiérrez Children's Hospital, Buenos Aires C1425EFD, Argentina
- Translational Medicine Unit, Ricardo Gutiérrez Children's Hospital, Buenos Aires C1425EFD, Argentina
- Patagonian Translational Knowledge Unit, El Calafate SAMIC High Complexity Hospital, El Calafate Z9405, Argentina
| | - Mariela Vanina Pace
- Laboratory of Physiology and Genetics of Hearing, Institute of Genetic Engineering and Molecular Biology "Dr. Héctor N. Torres"-National Council of Scientific and Technology (INGEBI-CONICET), Buenos Aires C1428ADN, Argentina
| | - Sofia Grinberg
- Laboratory of Physiology and Genetics of Hearing, Institute of Genetic Engineering and Molecular Biology "Dr. Héctor N. Torres"-National Council of Scientific and Technology (INGEBI-CONICET), Buenos Aires C1428ADN, Argentina
| | - Vanesa Lotersztein
- Genetics Service, Central Military Hospital Surgeon General "Dr. Cosme Argerich", Buenos Aires C1426, Argentina
| | - Paloma Brun
- "El Cruce" Néstor Carlos Kirchner High Complexity Hospital, Buenos Aires B1888, Argentina
| | - Carlos David Bruque
- Patagonian Translational Knowledge Unit, El Calafate SAMIC High Complexity Hospital, El Calafate Z9405, Argentina
| | - Ana Belén Elgoyhen
- Laboratory of Physiology and Genetics of Hearing, Institute of Genetic Engineering and Molecular Biology "Dr. Héctor N. Torres"-National Council of Scientific and Technology (INGEBI-CONICET), Buenos Aires C1428ADN, Argentina
- Pharmacology Institute, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1121A6B, Argentina
| | - Viviana Dalamón
- Laboratory of Physiology and Genetics of Hearing, Institute of Genetic Engineering and Molecular Biology "Dr. Héctor N. Torres"-National Council of Scientific and Technology (INGEBI-CONICET), Buenos Aires C1428ADN, Argentina
| |
Collapse
|
45
|
Jung J, Jang SH, Won D, Gee HY, Choi JY, Jung J. Clinical Characteristics and Audiological Profiles of Patients with Pathogenic Variants of WFS1. J Clin Med 2024; 13:4851. [PMID: 39200993 PMCID: PMC11355604 DOI: 10.3390/jcm13164851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Mutations in Wolfram syndrome 1 (WFS1) cause Wolfram syndrome and autosomal dominant non-syndromic hearing loss DFNA6/14/38. To date, more than 300 pathogenic variants of WFS1 have been identified. Generally, the audiological phenotype of Wolfram syndrome or DFNA6/14/38 is characterized by low-frequency hearing loss; however, this phenotype is largely variable. Hence, there is a need to better understand the diversity in audiological and vestibular profiles associated with WFS1 variants, as this can have significant implications for diagnosis and management. This study aims to investigate the clinical characteristics, audiological phenotypes, and vestibular function in patients with DFNA6/14/38. Methods: Whole-exome or targeted deafness gene panel sequencing was performed to confirm the pathogenic variants in patients with genetic hearing loss. Results: We identified nine independent families with affected individuals who carried a heterozygous pathogenic variant of WFS1. The onset of hearing loss varied from the first to the fifth decade. On a pure-tone audiogram, hearing loss was symmetrical, and the severity ranged from mild to severe. Notably, either both low-frequency and high-frequency or all-frequency-specific hearing loss was observed. However, hearing loss was non-progressive in all types. In addition, vestibular impairment was identified in patients with DFNA6/14/38, indicating that impaired WFS1 may also affect the vestibular organs. Conclusions: Diverse audiological and vestibular profiles were observed in patients with pathogenic variants of WFS1. These findings highlight the importance of comprehensive audiological and vestibular assessments in patients with WFS1 mutations for accurate diagnosis and management.
Collapse
Affiliation(s)
- Joonho Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
| | - Seung Hyun Jang
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Dongju Won
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
| |
Collapse
|
46
|
Lin Z, Xiang J, Sun X, Song N, Liu X, Cai Q, Yang J, Ye H, Xu J, Zhang H, Peng J, Sun Y, Peng Z. Genome Sequencing Unveils the Role of Copy Number Variants in Hearing Loss and Identifies Novel Deletions With Founder Effect in the DFNB1 Locus. Hum Mutat 2024; 2024:9517114. [PMID: 40225913 PMCID: PMC11918852 DOI: 10.1155/2024/9517114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/16/2024] [Indexed: 04/15/2025]
Abstract
Sensorineural hearing loss is a prevalent disorder with significant genetic involvement, which is often challenging to diagnose due to genetic heterogeneity. Exome sequencing (ES) has been a standard diagnostic tool for sensorineural hearing loss, but its limitations in detecting copy number variants (CNVs) and intronic variants have prompted the exploration of genome sequencing (GS) for improved diagnostic yield. We conducted GS on 46 hearing loss families with previously negative ES results and an additional cohort of 36 patients with a monoallelic pathogenic variant in GJB2 (the most common deafness gene). Additionally, the impact of a previously unrecognized novel 125-kb deletion in the DFNB1 locus on GJB2 expression was assessed using quantitative polymerase chain reaction (qPCR), and haplotype analysis was performed to characterize the deletion. GS diagnosed eight cases (17%, 8/46) in the ES-negative cohort, primarily attributed to CNVs (6/8). Notably, a previously unrecognized 125 kb deletion in the DFNB1 region was identified, affecting GJB2 expression and characterizing it as a founder effect in East Asian. In 47 patients with a monoallelic GJB2 variant, 15% (95% CI, 7.4%-28%) were diagnosed with DFNB1 deletions. Analysis of the gnomAD database revealed the prevalence and ethnic diversity of DFNB1 deletions, with the novel 125 kb deletion emerging as a prominent pathogenic variant in East Asian, non-Finnish European, and admixed American populations. Our study highlights the utility of GS in diagnosing sensorineural hearing loss. The identification of DFNB1 deletions underscores their significant contribution to hearing loss etiology, advocating for their inclusion in routine diagnostic testing. We propose GS as a primary genetic testing approach for patients with hearing loss, offering comprehensive genomic analysis and the potential for improved diagnostic accuracy.
Collapse
Affiliation(s)
- Zibin Lin
- College of Life SciencesUniversity of Chinese Academy of Sciences, Beijing 100049, China
- BGI Genomics, Shenzhen 518083, China
| | - Jiale Xiang
- College of Life SciencesUniversity of Chinese Academy of Sciences, Beijing 100049, China
- BGI Genomics, Shenzhen 518083, China
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and ControlChangsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | | | - Nana Song
- BGI Genomics, Shenzhen 518083, China
| | - Xiaozhou Liu
- Department of OtorhinolaryngologyUnion Hospital of Tongji Medical CollegeHuazhong University of Science and Technology, Wuhan 430022, China
| | - Qinming Cai
- Department of OtorhinolaryngologyUnion Hospital of Tongji Medical CollegeHuazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Yang
- BGI Genomics, Shenzhen 518083, China
| | | | | | | | | | - Yu Sun
- Department of OtorhinolaryngologyUnion Hospital of Tongji Medical CollegeHuazhong University of Science and Technology, Wuhan 430022, China
| | - Zhiyu Peng
- College of Life SciencesUniversity of Chinese Academy of Sciences, Beijing 100049, China
- BGI Genomics, Shenzhen 518083, China
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and ControlChangsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, China
| |
Collapse
|
47
|
Kim BJ, Kim Y, Kim JA, Han JH, Kim MY, Yang HK, Rhee CS, Kang YC, Kim CH, Choi BY. Novel Variant of FDXR as a Molecular Etiology of Postlingual Post-synaptic Auditory Neuropathy Spectrum Disorder via Mitochondrial Dysfunction: Reiteration of the Correlation between Genotype and Cochlear Implantation Outcomes. Clin Exp Otorhinolaryngol 2024; 17:206-216. [PMID: 39104018 PMCID: PMC11375174 DOI: 10.21053/ceo.2024.00184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/06/2024] [Indexed: 08/07/2024] Open
Abstract
OBJECTIVES FDXR encodes mitochondrial ferredoxin reductase, which is associated with auditory neuropathy spectrum disorder (ANSD) and optic atrophy. To date, only two studies have described FDXR-related hearing loss. The auditory rehabilitation outcomes of this disease entity have not been investigated, and the pathophysiological mechanisms remain incompletely understood. Here we report a hearing-impaired individual with co-segregation of the FDXR variant and post-synaptic type ANSD, who underwent cochlear implantation (CI) with favorable outcomes. We suggest a possible pathophysiological mechanism of adult-onset ANSD involving mitochondrial dysfunction. METHODS A 35-year-old woman was ascertained to have ANSD. Exome sequencing identified the genetic cause of hearing loss, and a functional study measuring mitochondrial activity was performed to provide molecular evidence of pathophysiology. Expression of FDXR in the mouse cochlea was evaluated by immunohistochemistry. Intraoperatively, electrically evoked compound action potential (ECAP) responses were measured, and the mapping parameters were adjusted accordingly. Audiological outcomes were monitored for over 1 year. RESULTS In lymphoblastoid cell lines (LCLs) carrying a novel FDXR variant, decreased ATP levels, reduced mitochondrial membrane potential, and increased reactive oxygen species levels were observed compared to control LCLs. These dysfunctions were restored by administering mitochondria isolated from umbilical cord mesenchymal stem cells, confirming the pathogenic potential of this variant via mitochondrial dysfunction. Partial ECAP responses during CI and FDXR expression in the mouse cochlea indicate that FDXR-related ANSD is post-synaptic. As a result of increasing the pulse width during mapping, the patient's CI outcomes showed significant improvement over 1-year post-CI. CONCLUSION A novel FDXR variant associated with mitochondrial dysfunction and post-synaptic ANSD was first identified in a Korean individual. Additionally, 1-year post-CI outcomes were reported for the first time in the literature. Excellent audiologic. RESULTS were obtained, and our. RESULTS reiterate the correlation between genotype and CI outcomes in ANSD.
Collapse
Affiliation(s)
- Bong Jik Kim
- Department of Otorhinolaryngology, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yujin Kim
- Paean Biotechnology Inc., Seoul, Korea
| | - Ju Ang Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin Hee Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Min Young Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hee Kyung Yang
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Chae-Seo Rhee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | | | | | - Byung Yoon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
48
|
王 月, 梁 悦, 黄 碧, 岑 晓, 黄 露, 陈 垲. [Late-onset hereditary hearing loss caused by TMPRSS3 compound heterozygous mutations]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY HEAD AND NECK SURGERY 2024; 38:679-686. [PMID: 39118504 PMCID: PMC11612751 DOI: 10.13201/j.issn.2096-7993.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Indexed: 08/10/2024]
Abstract
Objective:This study aims to identify the genetic etiology underlying late-onset hearing loss in two unrelated Chinese families. Methods:Detailed clinical data of recruited participants of two families were collected and analyzed using next-generation sequencing, combined with Sanger sequencing and bioinformatics tools. Results:Patients in both families manifested as down-sloping audiograms, mainly with severe mid-to-high frequency hearing loss as well as decreased speech recognition rate, both of which occurred during the second decade. Next-generation sequencing panels succeeded in identifying mutations in gene TMPRSS3, and three heterozygous mutations were screened out, among which c. 383T>C was the first reported mutation. In silico functional analysis and molecular modeling defined the five mutations as "pathogenic" or "likely pathogenic" according to official guideline. Conclusion:The novel mutation combinations in TMPRSS3 gene segregated with an exclusive auditory phenotype in the two pedigrees. Our results provided new data regarding the characteristic deafness caused by TMPRSS3 mutations during adolescent period when hearing should be closely monitored.
Collapse
Affiliation(s)
- 月莹 王
- 中山大学附属第一医院耳鼻咽喉科医院 中山大学耳鼻咽喉科学研究所(广州,510080)Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Otolaryngology, Sun Yat-sen University
| | - 悦 梁
- 中山大学附属第一医院耳鼻咽喉科医院 中山大学耳鼻咽喉科学研究所(广州,510080)Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Otolaryngology, Sun Yat-sen University
| | - 碧雪 黄
- 中山大学附属第一医院耳鼻咽喉科医院 中山大学耳鼻咽喉科学研究所(广州,510080)Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Otolaryngology, Sun Yat-sen University
| | - 晓晴 岑
- 中山大学附属第一医院耳鼻咽喉科医院 中山大学耳鼻咽喉科学研究所(广州,510080)Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Otolaryngology, Sun Yat-sen University
| | - 露莎 黄
- 中山大学附属第一医院耳鼻咽喉科医院 中山大学耳鼻咽喉科学研究所(广州,510080)Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Otolaryngology, Sun Yat-sen University
| | - 垲钿 陈
- 中山大学附属第一医院耳鼻咽喉科医院 中山大学耳鼻咽喉科学研究所(广州,510080)Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Otolaryngology, Sun Yat-sen University
| |
Collapse
|
49
|
Alsafh R, Alhashem A, Elsyed A, Yüksel Z, Graiess-Tlili K, Hundallah K, Thabet F, Tabarki B. Multiplex Consanguineous Family Highlights CLASP1 as a Candidate Gene for Lissencephaly. Neurol Genet 2024; 10:e200172. [PMID: 39040917 PMCID: PMC11261580 DOI: 10.1212/nxg.0000000000200172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Background and Objectives Noncentrosomal microtubules are essential cytoskeletal filaments that are important for neurite formation, axonal transport, and neuronal migration. They require stabilization by microtubule minus-end-targeting proteins including the CLASP family of molecules. To date, no human monogenic disorder has been associated with the CLASP1 gene. In this study, we aimed to delineate the clinical and neuroradiologic phenotype associated with biallelic CLASP1 variants. Methods We analyzed clinical characteristics, MRI data, and genotypes of a cohort of 3 patients with homozygous variants in CLASP1. Results Homozygous CLASP1 variant is associated with primary microcephaly, severe neurodevelopmental delay, and early-onset refractory epilepsy. The neuroradiologic phenotype comprises a highly recognizable combination of classic lissencephaly, with the posterior gradient more severe than the anterior gradient, a thin/hypoplastic splenium of the corpus callosum, mild enlargement of the lateral ventricles primarily posteriorly with a squared pattern, and pontine hypoplasia. Discussion This study underscores the role of CLASP1 in brain development and suggests that the identified variant disrupts CLASP1 interaction with the microtubule cytoskeleton, contributing to lissencephaly pathogenesis.
Collapse
Affiliation(s)
- Rawan Alsafh
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Amal Alhashem
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Aly Elsyed
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Zafer Yüksel
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Kalthoum Graiess-Tlili
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Khalid Hundallah
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Farah Thabet
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Brahim Tabarki
- From the Division of Pediatric Neurology (R.A., K.H., B.T.); Division of Genetics (A.A.), Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia; Department of Pediatrics (A.E.), King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Bioscientia Human Genetics (Z.Y.), Bioscientia Institut für Medizinische Diagnostik GmbH, Ingelheim, Germany; Department of Radiology (K.G.-T.), Sahloul University Hospital, Sousse; and Department of Pediatrics (F.T.), Fattouma Bourguiba University Hospital, Monastir, Tunisia
| |
Collapse
|
50
|
Cho SH, Jeong SH, Choi WH, Lee SY. Genomic Landscape of Branchio-Oto-Renal Syndrome through Whole-Genome Sequencing: A Single Rare Disease Center Experience in South Korea. Int J Mol Sci 2024; 25:8149. [PMID: 39125727 PMCID: PMC11311636 DOI: 10.3390/ijms25158149] [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: 07/03/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Branchio-oto-renal (BOR) and branchio-otic (BO) syndromes are characterized by anomalies affecting the ears, often accompanied by hearing loss, as well as abnormalities in the branchial arches and renal system. These syndromes exhibit a broad spectrum of phenotypes and a complex genomic landscape, with significant contributions from the EYA1 gene and the SIX gene family, including SIX1 and SIX5. Due to their diverse phenotypic presentations, which can overlap with other genetic syndromes, molecular genetic confirmation is essential. As sequencing technologies advance, whole-genome sequencing (WGS) is increasingly used in rare disease diagnostics. We explored the genomic landscape of 23 unrelated Korean families with typical or atypical BOR/BO syndrome using a stepwise approach: targeted panel sequencing and exome sequencing (Step 1), multiplex ligation-dependent probe amplification (MLPA) with copy number variation screening (Step 2), and WGS (Step 3). Integrating WGS into our diagnostic pipeline detected structure variations, including cryptic inversion and complex genomic rearrangement, eventually enhancing the diagnostic yield to 91%. Our findings expand the genomic architecture of BOR/BO syndrome and highlight the need for WGS to address the genetic diagnosis of clinically heterogeneous rare diseases.
Collapse
Affiliation(s)
- Sung Ho Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.H.C.); (S.H.J.); (W.H.C.)
| | - Sung Ho Jeong
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.H.C.); (S.H.J.); (W.H.C.)
| | - Won Hoon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.H.C.); (S.H.J.); (W.H.C.)
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.H.C.); (S.H.J.); (W.H.C.)
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| |
Collapse
|