1
|
Rajput K, Akhtar U, Pagarkar W, Rajput S, Walder C, D'Arco F, Cochrane L, Nash R, Bitner-Glindzicz M, Omar R. Etiology of Childhood Profound Sensorineural Hearing Loss: The Role of Hearing Loss Gene Panel Testing. Otolaryngol Head Neck Surg 2024; 171:1518-1525. [PMID: 38822754 DOI: 10.1002/ohn.838] [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: 06/26/2023] [Revised: 04/22/2024] [Accepted: 05/12/2024] [Indexed: 06/03/2024]
Abstract
OBJECTIVE Establishing the cause of hearing loss (HL) is important and rewarding, though not without its challenges. While our ability to identify the etiology for HL has improved with advances in scientific knowledge, a significant proportion of cases remain of unknown etiology. Recent protocol changes within the NHS Genomic Medicine Service support the utilization of the HL gene panel test, rather than individual gene tests. In light of these changes, determining the yield of these more extensive panel tests is important in informing future practice. STUDY DESIGN Retrospective study. SETTING The Cochlear Implant (CI) Department at Great Ormond Street Hospital (GOSH). METHODS Four hundred seventy-six children with profound HL were identified from a database of referrals to the GOSH CI Department. Data on etiology of HL including genetic diagnosis was collected from hospital notes on an electronic patient records system and hospital genetics database. RESULTS We identified a positive result in 163/476 (34%) cases through the gene panel test, representing an additional 19% yield to current level 1 investigations. Genetic HL, including both syndromic (including those not covered by the HL gene panel) and nonsyndromic (209/476, 44%) was the most common etiology in our cohort. Perinatal, intrauterine, ototoxicity, meningitis, and encephalitis categories altogether comprised 97/476 (20%) cases. CONCLUSION Gene panel testing provides significant additional yield over current level 1 investigations which include GJB2 testing only. This has far-reaching implications for how we optimize investigations into HL in children and counsel families, and for future early interventions.
Collapse
Affiliation(s)
- Kaukab Rajput
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Umar Akhtar
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Waheeda Pagarkar
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sarah Rajput
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Claire Walder
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Felice D'Arco
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Lesley Cochrane
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Robert Nash
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Rohani Omar
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- University College London Hospitals NHS Foundation Trust, Royal National ENT and Eastman Dental Hospitals, London, UK
| |
Collapse
|
2
|
Kazemi N, Rezvani Rezvandeh R, Zare Ashrafi F, Shokouhian E, Edizadeh M, Booth KTA, Kahrizi K, Najmabadi H, Mohseni M. A Frameshift Variant in ANKRD24 Implicates Its Role in Human Non-Syndromic Hearing Loss. Clin Genet 2024. [PMID: 39434538 DOI: 10.1111/cge.14635] [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: 09/11/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Hearing loss (HL) is the most prevalent sensorineural disorders, affecting about one in 1000 newborns. Over half of the cases are attributed to genetic factors; however, due to the extensive clinical and genetic heterogeneity, many cases remain without a conclusive genetic diagnosis. The advent of next-generation sequencing methodologies in recent years has greatly helped unravel the genetic etiology of HL by identifying numerous genes and causative variants. Despite this, much remains to be uncovered about the genetic basis of sensorineural hearing loss (SNHL). Here, we report an Iranian consanguineous family with postlingual, moderate-to-severe autosomal recessive SNHL. After first excluding plausible variants in known deafness-causing genes using whole exome sequencing, we reanalyzed the data, using a gene/variant prioritization pipeline established for novel gene discovery for HL. This approach identified a novel homozygous frameshift variant c.1934_1937del; (p.Thr645Lysfs*52) in ANKRD24, which segregated with the HL phenotype in the family. Recently, ANKRD24 has been shown to be a pivotal constituent of the stereocilia rootlet in cochlea hair cells and interacts with TRIOBP, a protein already implicated in human deafness. Our data implicate for the first time, ANKRD24 in human nonsyndromic HL (NSHL) and expands the genetic spectrum of HL.
Collapse
Affiliation(s)
- Negar Kazemi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Raziye Rezvani Rezvandeh
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Farzane Zare Ashrafi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Ebrahim Shokouhian
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Masoud Edizadeh
- Department of Bioinformatics, Genoks Genetic Diagnosis Center, Ankara, Turkey
- Ilyome Bioinformatics, Ankara, Turkey
| | - Kevin T A Booth
- Department of Medical & Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
- Department of Otolaryngology, Head & Neck Surgery, Indiana University, Indianapolis, Indiana, USA
| | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Marzieh Mohseni
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| |
Collapse
|
3
|
Maniaci A, Briglia M, Allia F, Montalbano G, Romano GL, Zaouali MA, H’mida D, Gagliano C, Malaguarnera R, Lentini M, Graziano ACE, Giurdanella G. The Role of Pericytes in Inner Ear Disorders: A Comprehensive Review. BIOLOGY 2024; 13:802. [PMID: 39452111 PMCID: PMC11504721 DOI: 10.3390/biology13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Inner ear disorders, including sensorineural hearing loss, Meniere's disease, and vestibular neuritis, are prevalent conditions that significantly impact the quality of life. Despite their high incidence, the underlying pathophysiology of these disorders remains elusive, and current treatment options are often inadequate. Emerging evidence suggests that pericytes, a type of vascular mural cell specialized to maintain the integrity and function of the microvasculature, may play a crucial role in the development and progression of inner ear disorders. The pericytes are present in the microvasculature of both the cochlea and the vestibular system, where they regulate blood flow, maintain the blood-labyrinth barrier, facilitate angiogenesis, and provide trophic support to neurons. Understanding their role in inner ear disorders may provide valuable insights into the pathophysiology of these conditions and lead to the development of novel diagnostic and therapeutic strategies, improving the standard of living. This comprehensive review aims to provide a detailed overview of the role of pericytes in inner ear disorders, highlighting the anatomy and physiology in the microvasculature, and analyzing the mechanisms that contribute to the development of the disorders. Furthermore, we explore the potential pericyte-targeted therapies, including antioxidant, anti-inflammatory, and angiogenic approaches, as well as gene therapy strategies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Marilena Briglia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Fabio Allia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Laboratory, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mohamed Amine Zaouali
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Avicenne Street, 5019 Monastir, Tunisia;
| | - Dorra H’mida
- Department of Cytogenetics and Reproductive Biology, Farhat Hached Hospital, 4021 Sousse, Tunisia;
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| |
Collapse
|
4
|
Miura A, Nakagawa T, Sogi C, Shima H, Adachi M, Honkura Y, Kikuchi A, Kanno J. Hearing loss with two pathogenic SLC26A4 variants and positive thyroid autoantibody: A case report. Clin Pediatr Endocrinol 2024; 33:219-223. [PMID: 39359669 PMCID: PMC11442696 DOI: 10.1297/cpe.2023-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/19/2024] [Indexed: 10/04/2024] Open
Abstract
SLC26A4 causes Pendred syndrome (PS) and nonsyndromic hearing loss. PS is distinguished based on perchlorate discharge test abnormality, goiter, and hypothyroidism in some patients. The pathophysiology of thyroid dysfunction in PS differs from that of autoimmune thyroid disease, in that it is considered to be caused by an iodide organification defect. It is believed that both diseases may incidentally coexist, and that SLC26A4 may play an important role in the etiology of autoimmune thyroid disease. Herein, we describe a case of a girl with hearing loss who had two pathogenic SLC26A4 variants and tested positive for thyroid peroxidase (TPO) antibody. She was diagnosed with hearing loss and vestibular aqueduct enlargement at the age of 4 yr. Deafness gene screening revealed two pathogenic SLC26A4 variants. As SLC26A4 variants can cause PS, the patient underwent thorough thyroid examination. Her thyroid gland was within the physiological range of mild enlargement. Although thyroid function test results were normal, the patient tested positive for TPO antibody. The patient was diagnosed with "suspected PS" and "suspected Hashimoto's thyroiditis," both of which increase the risk of developing hypothyroidism. Evaluating the comorbidity of Hashimoto's thyroiditis with the SLC26A4 variant in terms of complications is critical.
Collapse
Affiliation(s)
- Akinobu Miura
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
| | - Tomohiro Nakagawa
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
- Department of Pediatrics, Sendai City Hospital, Sendai, Japan
| | - Chisumi Sogi
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
- Department of Pediatrics, JCHO Sendai Hospital, Sendai, Japan
| | - Hirohito Shima
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
| | - Mika Adachi
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Hospital, Sendai, Japan
| | - Yohei Honkura
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Hospital, Sendai, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
| | - Junko Kanno
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
5
|
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
|
6
|
Zheng Q, Xu Z, Li N, Wang Y, Zhang T, Jing J. Age-related hearing loss in older adults: etiology and rehabilitation strategies. Front Neurosci 2024; 18:1428564. [PMID: 39411148 PMCID: PMC11473498 DOI: 10.3389/fnins.2024.1428564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Age-related hearing loss (ARHL) is a prevalent sensory organ disorder among elderly individuals that significantly impacts their cognitive function, psychological well-being, and ability to perform activities of daily living. As the population ages, the number of ARHL patients is increasing. However, the Audiological rehabilitation (AR) status of patients is not promising. In recent years, there has been an increasing focus on the health and rehabilitation of elderly individuals, and significant progress has been made in researching various age-related disorders. However, a unified definition of ARHL in terms of etiology and rehabilitation treatment is still lacking. This study aims to provide a reference for future research on ARHL and the development of AR strategies by reviewing the classification, etiology, and rehabilitation of ARHL.
Collapse
Affiliation(s)
- Qinzhi Zheng
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Zhuo Xu
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Nan Li
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yueying Wang
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ting Zhang
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Jiapeng Jing
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
7
|
Gombojav B, Erdenechuluun J, Makhbal Z, Danshiitsoodol N, Purevdorj E, Jargalmaa M, Batsaikhan T, Lin PH, Lu YS, Lo MY, Tseng HY, Tsai CY, Wu CC. Genetic Basis of Hearing Loss in Mongolian Patients: A Next-Generation Sequencing Study. Genes (Basel) 2024; 15:1227. [PMID: 39336818 PMCID: PMC11431586 DOI: 10.3390/genes15091227] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVE The genetic landscape of sensorineural hearing impairment (SNHI) varies across populations. In Mongolia, previous studies have shown a lower prevalence of GJB2 mutations and a higher frequency of variants in other deafness-related genes. This study aimed to investigate the genetic variants associated with idiopathic SNHI in Mongolian patients. METHODS We utilized the next-generation sequencing for investigating the causative mutations in 99 Mongolian patients with SNHI. RESULTS We identified pathogenic variants in 53 of the 99 SNHI patients (54%), with SLC26A4 being the most frequently mutated gene. The c.919-2A>G variant in SLC26A4 was the most prevalent, accounting for 46.2% of the mutant alleles. In addition, we identified 19 other known and 21 novel mutations in a total of 21 SNHI genes in autosomal recessive or dominant inheritance patterns. CONCLUSIONS Our findings expand the understanding of the genetic landscape of SNHI in Mongolia and highlight the importance of considering population-specific variations in genetic testing and counseling for SNHI.
Collapse
Affiliation(s)
- Bayasgalan Gombojav
- Department of Epidemiology and Biostatistics, School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia
- Healthy Twin Registry of Mongolia, Ulaanbaatar 14210, Mongolia
| | - Jargalkhuu Erdenechuluun
- Department of Otolaryngology, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14170, Mongolia
- The EMJJ Otolaryngology Hospital, Ulaanbaatar 14210, Mongolia
| | - Zaya Makhbal
- The EMJJ Otolaryngology Hospital, Ulaanbaatar 14210, Mongolia
| | - Narandalai Danshiitsoodol
- Healthy Twin Registry of Mongolia, Ulaanbaatar 14210, Mongolia
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 7348551, Japan
| | - Erkhembulgan Purevdorj
- Healthy Twin Registry of Mongolia, Ulaanbaatar 14210, Mongolia
- Department of Genetics, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia
| | | | - Tserendulam Batsaikhan
- Department of Otolaryngology, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14170, Mongolia
- The EMJJ Otolaryngology Hospital, Ulaanbaatar 14210, Mongolia
| | - Pei-Hsuan Lin
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Yue-Sheng Lu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Ming-Yu Lo
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Hsin-Yi Tseng
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Cheng-Yu Tsai
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 30261, Taiwan
| |
Collapse
|
8
|
Boehler NA, Seheult SDI, Wahid M, Hase K, D'Amico SF, Saini S, Mascarenhas B, Bergman ME, Phillips MA, Faure PA, Cheng HYM. A novel copy number variant in the murine Cdh23 gene gives rise to profound deafness and vestibular dysfunction. Hum Mol Genet 2024; 33:1648-1659. [PMID: 38981620 DOI: 10.1093/hmg/ddae095] [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: 02/07/2024] [Revised: 04/10/2024] [Accepted: 05/30/2024] [Indexed: 07/11/2024] Open
Abstract
Hearing loss is the most common congenital sensory deficit worldwide and exhibits high genetic heterogeneity, making molecular diagnoses elusive for most individuals. Detecting novel mutations that contribute to hearing loss is crucial to providing accurate personalized diagnoses, tailored interventions, and improving prognosis. Copy number variants (CNVs) are structural mutations that are understudied, potential contributors to hearing loss. Here, we present the Abnormal Wobbly Gait (AWG) mouse, the first documented mutant exhibiting waltzer-like locomotor dysfunction, hyperactivity, circling behaviour, and profound deafness caused by a spontaneous CNV deletion in cadherin 23 (Cdh23). We were unable to identify the causative mutation through a conventional whole-genome sequencing (WGS) and variant detection pipeline, but instead found a linked variant in hexokinase 1 (Hk1) that was insufficient to recapitulate the AWG phenotype when introduced into C57BL/6J mice using CRISPR-Cas9. Investigating nearby deafness-associated genes revealed a pronounced downregulation of Cdh23 mRNA and a complete absence of full-length CDH23 protein, which is critical for the development and maintenance of inner ear hair cells, in whole head extracts from AWG neonates. Manual inspection of WGS read depth plots of the Cdh23 locus revealed a putative 10.4 kb genomic deletion of exons 11 and 12 that was validated by PCR and Sanger sequencing. This study underscores the imperative to refine variant detection strategies to permit identification of pathogenic CNVs easily missed by conventional variant calling to enhance diagnostic precision and ultimately improve clinical outcomes for individuals with genetically heterogenous disorders such as hearing loss.
Collapse
Affiliation(s)
- Nicholas A Boehler
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| | - Shane D I Seheult
- Department of Psychology, Neuroscience & Behaviour, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Muhammad Wahid
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| | - Kazuma Hase
- Department of Psychology, Neuroscience & Behaviour, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Sierra F D'Amico
- Department of Psychology, Neuroscience & Behaviour, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Shakshi Saini
- Department of Psychology, Neuroscience & Behaviour, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Brittany Mascarenhas
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| | - Matthew E Bergman
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| | - Michael A Phillips
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| | - Paul A Faure
- Department of Psychology, Neuroscience & Behaviour, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
9
|
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
|
10
|
Harada S, Koyama Y, Yoshioka Y, Inohara H, Shimada S. Visualization of Reissner's membrane in the mouse inner ear using highly sensitive magnetic resonance imaging analysis. Biochem Biophys Res Commun 2024; 723:150153. [PMID: 38820624 DOI: 10.1016/j.bbrc.2024.150153] [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: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024]
Abstract
Although research on hearing loss, including the identification of causative genes, has become increasingly active, the pathogenic mechanism of hearing loss remains unclear. One of the reasons for this is that the structure of the inner ear of mice, which is commonly used as a genetically modified animal model, is too small and complex, making it difficult to accurately capture abnormalities and dynamic changes in vivo. Especially, Reissner's membrane is a very important structure that separates the perilymph and endolymph of the inner ear. This malformation or damage induces abnormalities in hearing and balance. Until now, imaging analyses, such as magnetic resonance imaging (MRI) and computed tomography, are performed to investigate the inner ear structure in vivo; however, it has been difficult to analyze the small inner ear structure of mice owing to resolution. Therefore, there is an urgent need to develop an image analysis method that can accurately capture the structure of the inner ear of mice including Reissner's membrane, both dynamically and statically. This study aimed to investigate whether it is possible to accurately capture the structure (e.g., Reissner's membrane) and abnormalities of the inner ear of mice using an 11.7 T MRI. By combining two types of MRI methods, in vivo and ex vivo, we succeeded for the first time in capturing the fine structure of the normal mouse inner ear, such as the Reissner's membrane, and inflammatory lesions of otitis media mouse models in detail and accurately. In the future, we believe that understanding the state of Reissner's membrane during living conditions will greatly contribute to the development of research on inner ear issues, such as hearing loss.
Collapse
Affiliation(s)
- Shotaro Harada
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, 541-8567, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita 565-0871, Japan.
| | - Yoshichika Yoshioka
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology (NICT) and Osaka University, Osaka 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan.
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan.
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, 541-8567, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, 565-0871, Japan.
| |
Collapse
|
11
|
Guan C, Shaikh M, Warnecke A, Vona B, Albert JT. A burden shared: The evolutionary case for studying human deafness in Drosophila. Hear Res 2024; 450:109047. [PMID: 38896942 DOI: 10.1016/j.heares.2024.109047] [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: 03/09/2024] [Revised: 05/09/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
Hearing impairment is the most prevalent sensory disease in humans and can have dramatic effects on the development, and preservation, of our cognitive abilities and social interactions. Currently 20 % of the world's population suffer from a form of hearing impairment; this is predicted to rise to 25 % by 2050. Despite this staggering disease load, and the vast damage it inflicts on the social, medical and economic fabric of humankind, our ability to predict, or prevent, the loss of hearing is very poor indeed. We here make the case for a paradigm shift in our approach to studying deafness. By exploiting more forcefully the molecular-genetic conservation between human hearing and hearing in morphologically distinct models, such as the fruit fly Drosophila melanogaster, we believe, a deeper understanding of hearing and deafness can be achieved. An understanding that moves beyond the surface of the 'deafness genes' to probe the underlying bedrock of hearing, which is shared across taxa, and partly shared across modalities. When it comes to understanding the workings (and failings) of human sensory function, a simple fruit fly has a lot to offer and a fly eye might sometimes be a powerful model for a human ear. Particularly the use of fly avatars, in which specific molecular (genetic or proteomic) states of humans (e.g. specific patients) are experimentally reproduced, in order to study the corresponding molecular mechanisms (e.g. specific diseases) in a controlled yet naturalistic environment, is a tool that promises multiple unprecedented insights. The use of the fly - and fly avatars - would benefit humans and will help enhance the power of other scientific models, such as the mouse.
Collapse
Affiliation(s)
- Chonglin Guan
- Sensory Physiology & Behaviour Group, Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, 26111 Oldenburg, Germany; Cluster of Excellence Hearing4all, Carl von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, 26111 Oldenburg, Germany
| | - Muhammad Shaikh
- Ear Institute, University College London, 332 Gray's Inn Road, London, WC1 × 8EE, UK
| | - Athanasia Warnecke
- Hannover Medical School, Department of Otorhinolaryngology, Head & Neck Surgery, Hannover, Germany; Cluster of Excellence Hearing4all, MHH Hannover, Germany
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
| | - Joerg T Albert
- Sensory Physiology & Behaviour Group, Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, 26111 Oldenburg, Germany; Cluster of Excellence Hearing4all, Carl von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, 26111 Oldenburg, Germany; Ear Institute, University College London, 332 Gray's Inn Road, London, WC1 × 8EE, UK.
| |
Collapse
|
12
|
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
|
13
|
Yan D, Nawab A, Smeal M, Liu XZ. Etiologic Diagnosis of Genetic Hearing Loss in an Ethnically Diverse Deafness Cohort. Audiol Neurootol 2024:1-10. [PMID: 39182490 DOI: 10.1159/000540202] [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: 10/18/2023] [Accepted: 06/25/2024] [Indexed: 08/27/2024] Open
Abstract
INTRODUCTION Hearing loss is a common sensory disorder that impacts patients across the lifespan. Many genetic variants have been identified that contribute to non-syndromic hearing loss. Yet, genetic testing is not routinely administered when hearing loss is diagnosed, particularly in adults. In this study, genetic testing was completed in patients with known hearing loss. METHODS A total of 104 patients who were evaluated for hearing loss were enrolled and received genetic testing. RESULTS Of those 104 patients, 39 had available genetic testing, 20 had one missing allele, and 45 yielded no genetic diagnosis. Of the 39 cases with genetic testing data, 24 were simplex cases, and 15 were multiplex cases. A majority of patients presented with an autosomal recessive inheritance pattern (n = 32), 26 of whom presented with congenital hearing loss. 38% of cases were positive for GJB2 mutation with c.35delG being the most common pathogenic variant. These findings are consistent with previous literature suggesting GJB2 mutations are the most common causes of non-syndromic hearing loss. CONCLUSION Given the frequency of genetic variants in patients with hearing loss, genetic testing should be considered a routine part of the hearing loss work-up, particularly as gene therapies are studied and become more widely available. LAY SUMMARY Many genetic variants have been identified that contribute to non-syndromic hearing loss. Given the frequency of genetic variants in patients with hearing loss, genetic testing should be considered a routine part of the hearing loss work-up.
Collapse
Affiliation(s)
- Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Aria Nawab
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA,
| | - Molly Smeal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Xue-Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
14
|
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
|
15
|
Toure M, Amalou G, Raise IA, Mobio NMA, Malki A, Barakat A. First report of an Ivorian family with nonsyndromic hearing loss caused by GJB2 compound heterozygous variants. Ann Hum Genet 2024. [PMID: 39092543 DOI: 10.1111/ahg.12574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
The primary etiology of congenital hearing loss is attributed to genetic factors, with GJB2 identified as a pivotal gene across diverse ethnic groups. Additionally, nonsyndromic hearing loss is predominantly inherited in an autosomal recessive manner. We used Sanger sequencing to analyze GJB2 in 17 deaf children from 13 unrelated Ivory Coast families. One family had two children born with severe congenital deafness and exhibited pathogenic compound heterozygous variants. These variants included a nonsense substitution (c.132G > A or p.Trp44Ter) and a newly discovered duplication of 7 base pairs (c.205_211dupTTCCCCA or p.Ser72ProfsTer32). Segregation testing confirmed these variants, marking the first identification of GJB2 in an Ivorian family with congenital hearing loss.
Collapse
Affiliation(s)
- Madoussou Toure
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
- Ben M'Sik Faculty of science, Hassan II University of Casablanca, Casablanca, Morocco
| | - Ghita Amalou
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Imane Ait Raise
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - N'kan Max Ange Mobio
- ENT department at the University Hospital Medical Center of Treichville, Abidjan, Ivory Coast
| | - Abderrahim Malki
- Ben M'Sik Faculty of science, Hassan II University of Casablanca, Casablanca, Morocco
| | - Abdelhamid Barakat
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
16
|
Gogal RA, Nessler AJ, Thiel AC, Bernabe HV, Corrigan Grove RA, Cousineau LM, Litman JM, Miller JM, Qi G, Speranza MJ, Tollefson MR, Fenn TD, Michaelson JJ, Okada O, Piquemal JP, Ponder JW, Shen J, Smith RJH, Yang W, Ren P, Schnieders MJ. Force Field X: A computational microscope to study genetic variation and organic crystals using theory and experiment. J Chem Phys 2024; 161:012501. [PMID: 38958156 PMCID: PMC11223778 DOI: 10.1063/5.0214652] [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: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
Force Field X (FFX) is an open-source software package for atomic resolution modeling of genetic variants and organic crystals that leverages advanced potential energy functions and experimental data. FFX currently consists of nine modular packages with novel algorithms that include global optimization via a many-body expansion, acid-base chemistry using polarizable constant-pH molecular dynamics, estimation of free energy differences, generalized Kirkwood implicit solvent models, and many more. Applications of FFX focus on the use and development of a crystal structure prediction pipeline, biomolecular structure refinement against experimental datasets, and estimation of the thermodynamic effects of genetic variants on both proteins and nucleic acids. The use of Parallel Java and OpenMM combines to offer shared memory, message passing, and graphics processing unit parallelization for high performance simulations. Overall, the FFX platform serves as a computational microscope to study systems ranging from organic crystals to solvated biomolecular systems.
Collapse
Affiliation(s)
- Rose A. Gogal
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Aaron J. Nessler
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Andrew C. Thiel
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Hernan V. Bernabe
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Rae A. Corrigan Grove
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, USA
| | - Leah M. Cousineau
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Jacob M. Litman
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Jacob M. Miller
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Guowei Qi
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Matthew J. Speranza
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Mallory R. Tollefson
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, USA
| | - Timothy D. Fenn
- Analytical Development, LEXEO Therapeutics, New York, New York 10010, USA
| | - Jacob J. Michaelson
- Department of Psychiatry, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, USA
| | - Okimasa Okada
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | | | - Jay W. Ponder
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, USA
| | - Richard J. H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, USA
| | | | - Pengyu Ren
- Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA
| | | |
Collapse
|
17
|
Frohne A, Vrabel S, Laccone F, Neesen J, Roesch S, Dossena S, Schoefer C, Frei K, Parzefall T. Mutational spectrum in patients with dominant non-syndromic hearing loss in Austria. Eur Arch Otorhinolaryngol 2024; 281:3577-3586. [PMID: 38400873 PMCID: PMC11211180 DOI: 10.1007/s00405-024-08492-5] [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: 11/09/2023] [Accepted: 01/17/2024] [Indexed: 02/26/2024]
Abstract
PURPOSE Hearing loss (HL) is often monogenic. The clinical importance of genetic testing in HL may further increase when gene therapy products become available. Diagnoses are, however, complicated by a high genetic and allelic heterogeneity, particularly of autosomal dominant (AD) HL. This work aimed to characterize the mutational spectrum of AD HL in Austria. METHODS In an ongoing prospective study, 27 consecutive index patients clinically diagnosed with non-syndromic AD HL, including 18 previously unpublished cases, were analyzed using whole-exome sequencing (WES) and gene panels. Novel variants were characterized using literature and bioinformatic means. Two additional Austrian medical centers provided AD HL mutational data obtained with in-house pipelines. Other Austrian cases of AD HL were gathered from literature. RESULTS The solve rate (variants graded as likely pathogenic (LP) or pathogenic (P)) within our cohort amounted to 59.26% (16/27). MYO6 variants were the most common cause. One third of LP/P variants were truncating variants in haploinsufficiency genes. Ten novel variants in HL genes were identified, including six graded as LP or P. In one cohort case and one external case, the analysis uncovered previously unrecognized syndromic presentations. CONCLUSION More than half of AD HL cases analyzed at our center were solved with WES. Our data demonstrate the importance of genetic testing, especially for the diagnosis of syndromic presentations, enhance the molecular knowledge of genetic HL, and support other laboratories in the interpretation of variants.
Collapse
Affiliation(s)
- Alexandra Frohne
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department for Cell and Developmental Biology, Center of Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Sybille Vrabel
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Franco Laccone
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Juergen Neesen
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Sebastian Roesch
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Christian Schoefer
- Department for Cell and Developmental Biology, Center of Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Klemens Frei
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Danube Private University, Steiner Landstraße 124, 3500, Krems a.d. Donau, Austria
| | - Thomas Parzefall
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
18
|
Lee NK, Uhler KM, Yoon PJ, Santos-Cortez RLP. Clinical Genetic Testing for Hearing Loss: Implications for Genetic Counseling and Gene-Based Therapies. Biomedicines 2024; 12:1427. [PMID: 39062005 PMCID: PMC11274279 DOI: 10.3390/biomedicines12071427] [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: 02/26/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/28/2024] Open
Abstract
Genetic factors contribute significantly to congenital hearing loss, with non-syndromic cases being more prevalent and genetically heterogeneous. Currently, 150 genes have been associated with non-syndromic hearing loss, and their identification has improved our understanding of auditory physiology and potential therapeutic targets. Hearing loss gene panels offer comprehensive genetic testing for hereditary hearing loss, and advancements in sequencing technology have made genetic testing more accessible and affordable. Currently, genetic panel tests available at a relatively lower cost are offered to patients who face financial barriers. In this study, clinical and audiometric data were collected from six pediatric patients who underwent genetic panel testing. Known pathogenic variants in MYO15A, GJB2, and USH2A were most likely to be causal of hearing loss. Novel pathogenic variants in the MYO7A and TECTA genes were also identified. Variable hearing phenotypes and inheritance patterns were observed amongst individuals with different pathogenic variants. The identification of these variants contributes to the continually expanding knowledge base on genetic hearing loss and lays the groundwork for personalized treatment options in the future.
Collapse
Affiliation(s)
- Nam K. Lee
- Department of Otolaryngology—Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristin M. Uhler
- Department of Physical Medicine and Rehabilitation, Children’s Hospital Colorado, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
- Department of Pediatric Otolaryngology, Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Patricia J. Yoon
- Department of Otolaryngology—Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatric Otolaryngology, Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Regie Lyn P. Santos-Cortez
- Department of Otolaryngology—Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
19
|
Andersen RE, Alkuraya IF, Ajeesh A, Sakamoto T, Mena EL, Amr SS, Romi H, Kenna MA, Robson CD, Wilch ES, Nalbandian K, Piña-Aguilar R, Walsh CA, Morton CC. Rare germline disorders implicate long non-coding RNAs disrupted by chromosomal structural rearrangements. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.16.24307499. [PMID: 38946951 PMCID: PMC11213069 DOI: 10.1101/2024.06.16.24307499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
In recent years, there has been increased focus on exploring the role the non-protein-coding genome plays in Mendelian disorders. One class of particular interest is long non-coding RNAs (lncRNAs), which has recently been implicated in the regulation of diverse molecular processes. However, because lncRNAs do not encode protein, there is uncertainty regarding what constitutes a pathogenic lncRNA variant, and thus annotating such elements is challenging. The Developmental Genome Anatomy Project (DGAP) and similar projects recruit individuals with apparently balanced chromosomal abnormalities (BCAs) that disrupt or dysregulate genes in order to annotate the human genome. We hypothesized that rearrangements disrupting lncRNAs could be the underlying genetic etiology for the phenotypes of a subset of these individuals. Thus, we assessed 279 cases with BCAs and selected 191 cases with simple BCAs (breakpoints at only two genomic locations) for further analysis of lncRNA disruptions. From these, we identified 66 cases in which the chromosomal rearrangements directly disrupt lncRNAs. Strikingly, the lncRNAs MEF2C-AS1 and ENSG00000257522 are each disrupted in two unrelated cases. Furthermore, in 30 cases, no genes of any other class aside from lncRNAs are directly disrupted, consistent with the hypothesis that lncRNA disruptions could underly the phenotypes of these individuals. To showcase the power of this genomic approach for annotating lncRNAs, here we focus on clinical reports and genetic analysis of two individuals with BCAs and additionally highlight six individuals with likely developmental etiologies due to lncRNA disruptions.
Collapse
Affiliation(s)
- Rebecca E. Andersen
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ibrahim F. Alkuraya
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Abna Ajeesh
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Tyler Sakamoto
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Elijah L. Mena
- Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sami S. Amr
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hila Romi
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Margaret A. Kenna
- Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA, USA
| | - Caroline D. Robson
- Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA, USA
- Department of Radiology, Boston Children’s Hospital, Boston, MA, USA
| | - Ellen S. Wilch
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Katarena Nalbandian
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Raul Piña-Aguilar
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Cynthia C. Morton
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- University of Manchester, Manchester Center for Audiology and Deafness, UK
| |
Collapse
|
20
|
Tlili A, Mahfood M, Al Mutery A, Chouchen J. Genetic analysis of 106 sporadic cases with hearing loss in the UAE population. Hum Genomics 2024; 18:59. [PMID: 38844983 PMCID: PMC11157727 DOI: 10.1186/s40246-024-00630-8] [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: 02/03/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Hereditary hearing loss is a rare hereditary condition that has a significant presence in consanguineous populations. Despite its prevalence, hearing loss is marked by substantial genetic diversity, which poses challenges for diagnosis and screening, particularly in cases with no clear family history or when the impact of the genetic variant requires functional analysis, such as in the case of missense mutations and UTR variants. The advent of next-generation sequencing (NGS) has transformed the identification of genes and variants linked to various conditions, including hearing loss. However, there remains a high proportion of undiagnosed patients, attributable to various factors, including limitations in sequencing coverage and gaps in our knowledge of the entire genome, among other factors. In this study, our objective was to comprehensively identify the spectrum of genes and variants associated with hearing loss in a cohort of 106 affected individuals from the UAE. RESULTS In this study, we investigated 106 sporadic cases of hearing impairment and performed genetic analyses to identify causative mutations. Screening of the GJB2 gene in these cases revealed its involvement in 24 affected individuals, with specific mutations identified. For individuals without GJB2 mutations, whole exome sequencing (WES) was conducted. WES revealed 33 genetic variants, including 6 homozygous and 27 heterozygous DNA changes, two of which were previously implicated in hearing loss, while 25 variants were novel. We also observed multiple potential pathogenic heterozygous variants across different genes in some cases. Notably, a significant proportion of cases remained without potential pathogenic variants. CONCLUSIONS Our findings confirm the complex genetic landscape of hearing loss and the limitations of WES in achieving a 100% diagnostic rate, especially in conditions characterized by genetic heterogeneity. These results contribute to our understanding of the genetic basis of hearing loss and emphasize the need for further research and comprehensive genetic analyses to elucidate the underlying causes of this condition.
Collapse
Affiliation(s)
- Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Building W8 Room 107, P.O. Box: 27272, Sharjah, United Arab Emirates.
- Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates.
| | - Mona Mahfood
- Department of Applied Biology, College of Sciences, University of Sharjah, Building W8 Room 107, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Abdullah Al Mutery
- Department of Applied Biology, College of Sciences, University of Sharjah, Building W8 Room 107, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Jihen Chouchen
- Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
21
|
Sun F, Xiao M, Ji D, Zheng F, Shi T. Deciphering potential causative factors for undiagnosed Waardenburg syndrome through multi-data integration. Orphanet J Rare Dis 2024; 19:226. [PMID: 38844942 PMCID: PMC11155130 DOI: 10.1186/s13023-024-03220-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/19/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Waardenburg syndrome (WS) is a rare genetic disorder mainly characterized by hearing loss and pigmentary abnormalities. Currently, seven causative genes have been identified for WS, but clinical genetic testing results show that 38.9% of WS patients remain molecularly unexplained. In this study, we performed multi-data integration analysis through protein-protein interaction and phenotype-similarity to comprehensively decipher the potential causative factors of undiagnosed WS. In addition, we explored the association between genotypes and phenotypes in WS with the manually collected 443 cases from published literature. RESULTS We predicted two possible WS pathogenic genes (KIT, CHD7) through multi-data integration analysis, which were further supported by gene expression profiles in single cells and phenotypes in gene knockout mouse. We also predicted twenty, seven, and five potential WS pathogenic variations in gene PAX3, MITF, and SOX10, respectively. Genotype-phenotype association analysis showed that white forelock and telecanthus were dominantly present in patients with PAX3 variants; skin freckles and premature graying of hair were more frequently observed in cases with MITF variants; while aganglionic megacolon and constipation occurred more often in those with SOX10 variants. Patients with variations of PAX3 and MITF were more likely to have synophrys and broad nasal root. Iris pigmentary abnormality was more common in patients with variations of PAX3 and SOX10. Moreover, we found that patients with variants of SOX10 had a higher risk of suffering from auditory system diseases and nervous system diseases, which were closely associated with the high expression abundance of SOX10 in ear tissues and brain tissues. CONCLUSIONS Our study provides new insights into the potential causative factors of WS and an alternative way to explore clinically undiagnosed cases, which will promote clinical diagnosis and genetic counseling. However, the two potential disease-causing genes (KIT, CHD7) and 32 potential pathogenic variants (PAX3: 20, MITF: 7, SOX10: 5) predicted by multi-data integration in this study are all computational predictions and need to be further verified through experiments in follow-up research.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Dong Ji
- Department of Otolaryngology, Head and Neck Surgery, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Feng Zheng
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, the Institute of Biomedical Sciences and the School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Beijing Advanced Innovation Center, for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing, 100083, China.
| |
Collapse
|
22
|
Justin Margret J, Jayasankaran C, Amritkumar P, Azaiez H, Srisailapathy CRS. Unraveling the Genetic Basis of Combined Deafness and Male Infertility Phenotypes through High-Throughput Sequencing in a Unique Cohort from South India. ADVANCED GENETICS (HOBOKEN, N.J.) 2024; 5:2300206. [PMID: 38884051 PMCID: PMC11170077 DOI: 10.1002/ggn2.202300206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/15/2024] [Indexed: 06/18/2024]
Abstract
The co-occurrence of sensorineural hearing loss and male infertility has been reported in several instances, suggesting potential shared genetic underpinnings. One such example is the contiguous gene deletion of CATSPER2 and STRC genes, previously associated with deafness-infertility syndrome (DIS) in males. Fifteen males with both hearing loss and infertility from southern India after exclusion for the DIS contiguous gene deletion and the FOXI1 gene mutations are subjected to exome sequencing. This resolves the genetic etiology in four probands for both the phenotypes; In the remaining 11 probands, two each conclusively accounted for deafness and male infertility etiologies. Genetic heterogeneity is well reflected in both phenotypes. Four recessive (TRIOBP, SLC26A4, GJB2, COL4A3) and one dominant (SOX10) for the deafness; six recessive genes (LRGUK, DNAH9, ARMC4, DNAH2, RSPH6A, and ACE) for male infertility can be conclusively ascribed. LRGUK and RSPH6A genes are implicated earlier only in mice models, while the ARMC4 gene is implicated in chronic destructive airway diseases due to primary ciliary dyskinesia. This study would be the first to document the role of these genes in the male infertility phenotype in humans. The result suggests that deafness and infertility are independent events and do not segregate together among the probands.
Collapse
Affiliation(s)
- Jeffrey Justin Margret
- Department of Genetics Dr. ALM Post Graduate Institute of Basic Medical Sciences University of Madras Taramani Campus Chennai 600 113 India
- Department of Pediatrics Louisiana State University Health Sciences Center Shreveport LA 71103 USA
| | - Chandru Jayasankaran
- Department of Genetics Dr. ALM Post Graduate Institute of Basic Medical Sciences University of Madras Taramani Campus Chennai 600 113 India
- Department of Personalized Health Care Roche Products India Pvt., Ltd. Bengaluru Karnataka 560 025 India
| | - Pavithra Amritkumar
- Department of Genetics Dr. ALM Post Graduate Institute of Basic Medical Sciences University of Madras Taramani Campus Chennai 600 113 India
- Meenakshi Academy of Higher Education and Research (MAHER) Chennai 600 078 India
| | - Hela Azaiez
- Department of Otolaryngology Carver College of Medicine University of Iowa Iowa City Iowa 52242 USA
| | - C R Srikumari Srisailapathy
- Department of Genetics Dr. ALM Post Graduate Institute of Basic Medical Sciences University of Madras Taramani Campus Chennai 600 113 India
| |
Collapse
|
23
|
Chen PY, Yang TW, Tseng YS, Tsai CY, Yeh CS, Lee YH, Lin PH, Lin TC, Wu YJ, Yang TH, Chiang YT, Hsu JSJ, Hsu CJ, Chen PL, Chou CF, Wu CC. Machine learning-based longitudinal prediction for GJB2-related sensorineural hearing loss. Comput Biol Med 2024; 176:108597. [PMID: 38763069 DOI: 10.1016/j.compbiomed.2024.108597] [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: 12/12/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Recessive GJB2 variants, the most common genetic cause of hearing loss, may contribute to progressive sensorineural hearing loss (SNHL). The aim of this study is to build a realistic predictive model for GJB2-related SNHL using machine learning to enable personalized medical planning for timely intervention. METHOD Patients with SNHL with confirmed biallelic GJB2 variants in a nationwide cohort between 2005 and 2022 were included. Different data preprocessing protocols and computational algorithms were combined to construct a prediction model. We randomly divided the dataset into training, validation, and test sets at a ratio of 72:8:20, and repeated this process ten times to obtain an average result. The performance of the models was evaluated using the mean absolute error (MAE), which refers to the discrepancy between the predicted and actual hearing thresholds. RESULTS We enrolled 449 patients with 2184 audiograms available for deep learning analysis. SNHL progression was identified in all models and was independent of age, sex, and genotype. The average hearing progression rate was 0.61 dB HL per year. The best MAE for linear regression, multilayer perceptron, long short-term memory, and attention model were 4.42, 4.38, 4.34, and 4.76 dB HL, respectively. The long short-term memory model performed best with an average MAE of 4.34 dB HL and acceptable accuracy for up to 4 years. CONCLUSIONS We have developed a prognostic model that uses machine learning to approximate realistic hearing progression in GJB2-related SNHL, allowing for the design of individualized medical plans, such as recommending the optimal follow-up interval for this population.
Collapse
Affiliation(s)
- Pey-Yu Chen
- Department of Otolaryngology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City, Taiwan; Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ta-Wei Yang
- Graduate Institute of Networking and Multimedia, National Taiwan University, Taipei, Taiwan
| | - Yi-Shan Tseng
- Department of Computer Science & Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Cheng-Yu Tsai
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chiung-Szu Yeh
- Department of Computer Science & Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Yen-Hui Lee
- Department of Otolaryngology, National Taiwan University Biomedical Park Hospital, Hsinchu County, Taiwan; Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu City, Taiwan; Hearing and Speech Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Hsuan Lin
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Chun Lin
- Department of Computer Science & Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Jen Wu
- Department of Computer Science & Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Ting-Hua Yang
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Ting Chiang
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jacob Shu-Jui Hsu
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chuan-Jen Hsu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Department of Otorhinolaryngology-Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Fu Chou
- Department of Computer Science & Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan.
| |
Collapse
|
24
|
Zhou S, Chen M, Pei J, Zhang C, Ren X, Li J, Sa Y, Zhu B, Li Y. Distribution of mitochondrial MT-RNR1, MT-TL1, MT-TS1, MT-TK and MT-TE genes variants associated with hearing loss in Southwestern China. Int J Pediatr Otorhinolaryngol 2024; 181:111979. [PMID: 38739980 DOI: 10.1016/j.ijporl.2024.111979] [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: 02/26/2024] [Revised: 04/21/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Maternally inherited hearing loss has been associated with mitochondrial genes, including MT-RNR1, MT-TL1, MT-TS1, MT-TK and MT-TE. Among these genes, MT-RNR1 is known to be a hotspot for pathogenic variants related to aminoglycoside ototoxicity and nonsyndromic hearing loss. However, the frequency and spectrum of variants in these genes, particularly in multi-ethnic hearing loss patients from Southwestern China, are still not fully understood. METHODS In this study, we enrolled 460 hearing loss patients from various ethnic backgrounds (Han, Yi, Dai, Hani, etc.) in Southwestern China. Next-generation sequencing was used to analyze the mitochondrial MT-RNR1, MT-TL1, MT-TS1, MT-TK and MT-TE genes. Subsequently, bioinformatical methods were employed to evaluate the identified variants. RESULTS Among the patients with hearing loss, we identified 70 variants in MT-RNR1 (78.6 %, 55/70), MT-TL1 (4.3 %, 3/70), MT-TS1 (4.3 %, 3/70), MT-TK (7.1 %, 5/70) and MT-TE (5.7 %, 4/70) genes. We found that 15 variants were associated with hearing loss, including m.1555 A > G and m.1095 T > C. Additionally, we discovered three reported mitochondrial variants (m.676 G > A, m.7465 insC, and m.7474 A > G) newly correlated with hearing loss. Notably, certain pathogenic variants, such as m.1555 A > G, displayed non-consistent distributions among the multi-ethnic patients with hearing loss. Furthermore, the number of variants associated with hearing loss was higher in the Sinitic group (n = 181) and Tibeto-Burman group (n = 215) compared to the Kra-Dai group (n = 38) and Hmong-Mien group (n = 26). CONCLUSIONS This present study revealed the distribution of mitochondrial variants linked to hearing loss across various ethnic groups in Southwestern China. These data suggest a potential correlation between the distribution of mitochondrial variants associated with hearing loss and ethnic genetic backgrounds.
Collapse
Affiliation(s)
- Shiyu Zhou
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Menglan Chen
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiahong Pei
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Chen Zhang
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiaofei Ren
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jingyu Li
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yaliang Sa
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| | - Baosheng Zhu
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China; National Health Commission Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, Kunming, Yunnan, China.
| | - Yunlong Li
- Department of Medical Genetics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China; National Health Commission Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, Kunming, Yunnan, China.
| |
Collapse
|
25
|
Bodenbender JP, Marino V, Philipp J, Tropitzsch A, Kernstock C, Stingl K, Kempf M, Haack TB, Zuleger T, Mazzola P, Kohl S, Weisschuh N, Dell'Orco D, Kühlewein L. Comprehensive analysis of two hotspot codons in the TUBB4B gene and associated phenotypes. Sci Rep 2024; 14:10551. [PMID: 38719929 PMCID: PMC11078972 DOI: 10.1038/s41598-024-61019-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Our purpose was to elucidate the genotype and ophthalmological and audiological phenotype in TUBB4B-associated inherited retinal dystrophy (IRD) and sensorineural hearing loss (SNHL), and to model the effects of all possible amino acid substitutions at the hotspot codons Arg390 and Arg391. Six patients from five families with heterozygous missense variants in TUBB4B were included in this observational study. Ophthalmological testing included best-corrected visual acuity, fundus examination, optical coherence tomography, fundus autofluorescence imaging, and full-field electroretinography (ERG). Audiological examination included pure-tone and speech audiometry in adult patients and auditory brainstem response testing in a child. Genetic testing was performed by disease gene panel analysis based on genome sequencing. The molecular consequences of the substitutions of residues 390 and 391 on TUBB4B and its interaction with α-tubulin were predicted in silico on its three-dimensional structure obtained by homology modelling. Two independent patients had amino acid exchanges at position 391 (p.(Arg391His) or p.(Arg391Cys)) of the TUBB4B protein. Both had a distinct IRD phenotype with peripheral round yellowish lesions with pigmented spots and mild or moderate SNHL, respectively. Yet the phenotype was milder with a sectorial pattern of bone spicules in one patient, likely due to a genetically confirmed mosaicism for p.(Arg391His). Three patients were heterozygous for an amino acid exchange at position 390 (p.(Arg390Gln) or p.(Arg390Trp)) and presented with another distinct retinal phenotype with well demarcated pericentral retinitis pigmentosa. All showed SNHL ranging from mild to severe. One additional patient showed a variant distinct from codon 390 or 391 (p.(Tyr310His)), and presented with congenital profound hearing loss and reduced responses in ERG. Variants at codon positions 390 and 391 were predicted to decrease the structural stability of TUBB4B and its complex with α-tubulin, as well as the complex affinity. In conclusion, the twofold larger reduction in heterodimer affinity exhibited by Arg391 substitutions suggested an association with the more severe retinal phenotype, compared to the substitution at Arg390.
Collapse
Affiliation(s)
- Jan-Philipp Bodenbender
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Valerio Marino
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| | - Julia Philipp
- Department of Otolaryngology-Head & Neck Surgery, Hearing Research Center, University of Tübingen Medical Center, Tübingen, Germany
| | - Anke Tropitzsch
- Department of Otolaryngology-Head & Neck Surgery, Hearing Research Center, University of Tübingen Medical Center, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Christoph Kernstock
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Katarina Stingl
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Melanie Kempf
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Tobias B Haack
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Theresia Zuleger
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Pascale Mazzola
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Susanne Kohl
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Nicole Weisschuh
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| | - Laura Kühlewein
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany.
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
26
|
Hu SW, Lv J, Wang Z, Tang H, Wang H, Wang F, Wang D, Zhang J, Zhang L, Cao Q, Chen Y, Gao Z, Han Y, Wang W, Li GL, Shu Y, Li H. Engineering of the AAV-Compatible Hair Cell-Specific Small-Size Myo15 Promoter for Gene Therapy in the Inner Ear. RESEARCH (WASHINGTON, D.C.) 2024; 7:0341. [PMID: 38665848 PMCID: PMC11045262 DOI: 10.34133/research.0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/21/2024] [Indexed: 04/28/2024]
Abstract
Adeno-associated virus (AAV)-mediated gene therapy is widely applied to treat numerous hereditary diseases in animal models and humans. The specific expression of AAV-delivered transgenes driven by cell type-specific promoters should further increase the safety of gene therapy. However, current methods for screening cell type-specific promoters are labor-intensive and time-consuming. Herein, we designed a "multiple vectors in one AAV" strategy for promoter construction in vivo. Through this strategy, we truncated a native promoter for Myo15 expression in hair cells (HCs) in the inner ear, from 1,611 bp down to 1,157 bp, and further down to 956 bp. Under the control of these 2 promoters, green fluorescent protein packaged in AAV-PHP.eB was exclusively expressed in the HCs. The transcription initiation ability of the 2 promoters was further verified by intein-mediated otoferlin recombination in a dual-AAV therapeutic system. Driven by these 2 promoters, human otoferlin was selectively expressed in HCs, resulting in the restoration of hearing in treated Otof -/- mice for at least 52 weeks. In summary, we developed an efficient screening strategy for cell type-specific promoter engineering and created 2 truncated Myo15 promoters that not only restored hereditary deafness in animal models but also show great potential for treating human patients in future.
Collapse
Affiliation(s)
- Shao Wei Hu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Jun Lv
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Zijing Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Honghai Tang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Hui Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Fang Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Daqi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Juan Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Longlong Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Qi Cao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Yuxin Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Ziwen Gao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Yu Han
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Wuqing Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Geng-lin Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| | - Huawei Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,
Fudan University, Shanghai, 200031, China
- Institute of Biomedical Science,
Fudan University, Shanghai, 200032, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200032, China
| |
Collapse
|
27
|
Yan X, Wang J, Yang W, Li L, Shen T, Geng J, Zhang Q, Zhong M, Xiong W, Bu F, Lu Y, Zhao Y, Cheng J, Yuan H. Molecular diagnosis, clinical evaluation and phenotypic spectrum of Townes-Brocks syndrome: insights from a large Chinese hearing loss cohort. J Med Genet 2024; 61:459-468. [PMID: 38296632 DOI: 10.1136/jmg-2023-109579] [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: 08/17/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Townes-Brocks syndrome (TBS) is a rare genetic disorder characterised by multiple malformations. Due to its phenotypic heterogeneity and rarity, diagnosis and recognition of TBS can be challenging and there has been a lack of investigation of patients with atypical TBS in large cohorts and delineation of their phenotypic characteristics. METHODS We screened SALL1 and DACT1 variants using next-generation sequencing in the China Deafness Genetics Consortium (CDGC) cohort enrolling 20 666 unrelated hearing loss (HL) cases. Comprehensive clinical evaluations were conducted on seven members from a three-generation TBS family. Combining data from previously reported cases, we also provided a landscape of phenotypes and genotypes of patients with TBS. RESULTS We identified five novel and two reported pathogenic/likely pathogenic (P/LP) SALL1 variants from seven families. Audiological features in patients differed in severity and binaural asymmetry. Moreover, previously undocumented malformations in the middle and inner ear were detected in one patient. By comprehensive clinical evaluations, we further provide evidence for the causal relationship between SALL1 variation and certain endocrine abnormalities. Penetrance analysis within familial contexts revealed incomplete penetrance among first-generation patients with TBS and a higher disease burden among their affected offspring. CONCLUSION This study presents the first insight of genetic screening for patients with TBS in a large HL cohort. We broadened the phenotypic-genotypic spectrum of TBS and our results supported an underestimated prevalence of TBS. Due to the rarity and phenotypic heterogeneity of rare diseases, broader spectrum molecular tests, especially whole genome sequencing, can improve the situation of underdiagnosis and provide effective recommendations for clinical management.
Collapse
Affiliation(s)
- Xiaohong Yan
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jing Wang
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wen Yang
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Linke Li
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Tian Shen
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jia Geng
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Mingjun Zhong
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wenyu Xiong
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Fengxiao Bu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yu Lu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yu Zhao
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jing Cheng
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Huijun Yuan
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Han JH, Bae SH, Joo SY, Kim JA, Kim SJ, Jang SH, Won D, Gee HY, Choi JY, Jung J, Kim SH. Characterization of Vestibular Phenotypes in Patients with Genetic Hearing Loss. J Clin Med 2024; 13:2001. [PMID: 38610765 PMCID: PMC11012556 DOI: 10.3390/jcm13072001] [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: 02/27/2024] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Background: The vestibular phenotypes of patients with genetic hearing loss are poorly understood. Methods: we performed genetic testing including exome sequencing and vestibular function tests to investigate vestibular phenotypes and functions in patients with genetic hearing loss. Results: Among 627 patients, 143 (22.8%) had vestibular symptoms. Genetic variations were confirmed in 45 (31.5%) of the 143 patients. Nineteen deafness genes were linked with vestibular symptoms; the most frequent genes in autosomal dominant and recessive individuals were COCH and SLC26A4, respectively. Vestibular symptoms were mostly of the vertigo type, recurrent, and persisted for hours in the genetically confirmed and unconfirmed groups. Decreased vestibular function in the caloric test, video head impulse test, cervical vestibular-evoked myogenic potential, and ocular vestibular-evoked myogenic potential was observed in 42.0%, 16.3%, 57.8%, and 85.0% of the patients, respectively. The caloric test revealed a significantly higher incidence of abnormal results in autosomal recessive individuals than in autosomal dominant individuals (p = 0.011). The genes, including SLC26A4, COCH, KCNQ4, MYH9, NLRP3, EYA4, MYO7A, MYO15A, and MYH9, were heterogeneously associated with abnormalities in the vestibular function test. Conclusions: In conclusion, diverse vestibular symptoms are commonly concomitant with genetic hearing loss and are easily overlooked.
Collapse
Affiliation(s)
- Ji Hyuk Han
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.H.H.); (J.Y.C.)
| | - Seong Hoon Bae
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.H.H.); (J.Y.C.)
| | - Sun Young Joo
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (H.Y.G.)
| | - Jung Ah Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (H.Y.G.)
| | - Se Jin Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (H.Y.G.)
| | - Seung Hyun Jang
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (H.Y.G.)
| | - 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; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (H.Y.G.)
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.H.H.); (J.Y.C.)
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.H.H.); (J.Y.C.)
| | - Sung Huhn Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.H.H.); (J.Y.C.)
| |
Collapse
|
29
|
Kriukelis R, Gabbett MT, Beswick R, McInerney-Leo AM, Driscoll C, Liddle K. The congenital hearing phenotype in GJB2 in Queensland, Australia: V37I and mild hearing loss predominates. Eur J Hum Genet 2024:10.1038/s41431-024-01584-0. [PMID: 38486023 DOI: 10.1038/s41431-024-01584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
GJB2 was originally identified in severe, non-syndromic sensorineural hearing loss (SNHL), but was subsequently associated with mild and moderate SNHL. Given the increasing utilisation of genetic testing pre-conceptually, prenatally, and neonatally, it is crucial to understand genotype-phenotype correlations. This study evaluated the nature and frequency of GJB2 variants in an Australian paediatric population with varying degrees of SNHL ascertained through newborn hearing screening. Audiograms from individuals with GJB2 variants and/or a GJB6 deletion (GJB6-D13S11830) were retrospectively reviewed (n = 127). Two-thirds were biallelic (homozygous/compound heterozygous) for pathogenic/likely pathogenic variants of GJB2 and/or GJB6 (n = 80). The most frequent variant was c.109 G > A, followed by c.35delG and c.101 T > C. Compared to biallelic carriage of other GJB2 variants, c.109 G > A positive individuals (homozygous/compound heterozygous) were more likely to have mild HL at their initial and latest audiograms (p = 0.0004). Biallelic carriage of c.35delG was associated with moderately-severe or greater SNHL at both initial and latest audiograms (p = 0.007). The c.101 T > C variant presented with milder SNHL and U-shaped audiograms (p = 0.02). In this agnostically identified cohort, mild SNHL predominated in GJB2/GJB6 carriers in contrast to previous studies targeting individuals with significant loss. Consequently, c.109 G > A, associated with milder phenotypes, was the most frequent. This study provides valuable data to support prognostic confidence in genetic counselling.
Collapse
Affiliation(s)
| | - Michael T Gabbett
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rachael Beswick
- University of Queensland Centre for Children's Health Research, South Brisbane, QLD, Australia
- Healthy Hearing Program, Children's Health Queensland Hospital and Health Service, Brisbane, QLD, Australia
- School of Health and Rehabilitation Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Aideen M McInerney-Leo
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, QLD, Australia
| | - Carlie Driscoll
- School of Health and Rehabilitation Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Karen Liddle
- Queensland Children's Hospital, South Brisbane, QLD, Australia.
- University of Queensland Centre for Children's Health Research, South Brisbane, QLD, Australia.
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, QLD, Australia.
| |
Collapse
|
30
|
Korleski MJ, Stefanac SJ, Inglehart MR. Dentists' considerations concerning treating patients from the Deaf and Hard of Hearing community: A national survey. J Dent Educ 2024; 88:278-288. [PMID: 37921409 DOI: 10.1002/jdd.13405] [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: 09/04/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
OBJECTIVES Research shows that adults who were Deaf or Hard of Hearing (HoH) had poorer oral health than adults who did not belong to this community. The objectives were to assess dentists' education, knowledge, attitudes, and professional behavior related to treating patients from the Deaf or HoH community and the relationships between these constructs. METHODS A total of 207 members of the American Dental Association and the Michigan Dental Association responded to a mailed or web-based survey concerning their education, knowledge, attitudes, and professional behavior related to treating patients from the Deaf or HoH community. RESULTS On average, the respondents disagreed that they were well educated in classroom-based, clinical, or community-based dental school settings (five-point answer scale with 1 = disagree strongly; mean = 2.29/2.27/2.35) or by their professional organization (mean = 2.00) about treating Deaf or HoH patients. However, the more recently the respondents had graduated from dental school, the better they described their education about this topic (r = 0.29; p < 0.001). Additionally, 45.9% agreed/strongly agreed that they would like to attend a continuing education course about this topic; 68.9% agreed/agreed strongly that negative consequences for patients' general health can occur; and 61.1% that patients cannot be well educated about oral hygiene if Deaf or HoH patients do not have appropriate interpretive support in dental offices. The better dentists were educated about this topic, the more knowledge they had (r = 0.50; p < 0.001). On average, the respondents agreed more strongly that they were comfortable treating adult patients who communicated orally than patients using American Sign Language (4.02 vs. 3.25; p < 0.001). CONCLUSIONS These findings show that efforts are needed to improve dental school and continuing education curricula about dental treatment for Deaf and HoH patients. The more recently the respondents had graduated, the more positively they described their education. Increased dental school and continuing education efforts are still urgently needed.
Collapse
Affiliation(s)
- Michael J Korleski
- General Practice Residency Program, University of California, San Francisco, California, USA
| | - Stephen J Stefanac
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Marita R Inglehart
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
- Department of Psychology, College of Literature, Science & Arts, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Naddafnia H, Noormohammadi Z, Irani S, Salahshoorifar I. Whole Exome Sequencing of Non-Syndromic Hearing Loss Patients. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:453-461. [PMID: 38894825 PMCID: PMC11182469 DOI: 10.18502/ijph.v53i2.14930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/18/2023] [Indexed: 06/21/2024]
Abstract
Background Hearing loss is the second most common disease after mental retardation in Iran. Autosomal recessive non-syndromic hearing loss (ARNSHL) is an extreme and highly heterogeneous disease, for which more than 70 genes have been identified. Considering the frequency of family marriage as well as the importance of ARNSHL in Iran, we evaluated the genetic factors involved in this type of deafness. Methods We performed the whole exome sequencing (WES) of eight Iranian subjects with severe nonsyndromic hearing loss selected from 110 well-characterized subjects with non-syndromic hearing loss from 2017-2019. The patients with mutated GJB2 and GJB6 genes were excluded from the study. Results The use of the whole exome sequencing method revealed 10 different mutations in 7 genes, including SLC26A4 (c.1234G>T), FGF3 (c.45DelC, c.466T>C), ADGRV1 (c.12528-2A>C, c.16226-16227insAGTC), OTOG (c.7454delG), OTOF (c.3570+2T>C), ESPN (c.992G>A), OTOA (c.2359G>T, c.2353A>C). Seven new variants were observed in seven families including SLC26A4 (c.1234G>T), FGF3 (c.45DelC), ADGRV1 (c.12528-2A>C), OTOG (c.7454delG), ADGRV1 (c.16226-16227insAGTC), OTOF (c.3570+2T>C). Conclusion The causal mutation of ARNSHL was found in all patients using the WES. Meta-analysis studies can help to identify common mutations causing deafness in any population to facilitate identification of carriers and subjects with deafness.
Collapse
Affiliation(s)
- Hossein Naddafnia
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Iman Salahshoorifar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
32
|
De Rosa MA, Bernardi MT, Kleppe S, Walz K. Hearing Loss: Genetic Testing, Current Advances and the Situation in Latin America. Genes (Basel) 2024; 15:178. [PMID: 38397168 PMCID: PMC10888486 DOI: 10.3390/genes15020178] [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: 12/27/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Congenital hearing loss is the most common birth defect, estimated to affect 2-3 in every 1000 births, with ~50-60% of those related to genetic causes. Technological advances enabled the identification of hundreds of genes related to hearing loss (HL), with important implications for patients, their families, and the community. Despite these advances, in Latin America, the population with hearing loss remains underdiagnosed, with most studies focusing on a single locus encompassing the GJB2/GJB6 genes. Here we discuss how current and emerging genetic knowledge has the potential to alter the approach to diagnosis and management of hearing loss, which is the current situation in Latin America, and the barriers that still need to be overcome.
Collapse
Affiliation(s)
- Maria Agustina De Rosa
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (M.A.D.R.); (M.T.B.)
| | - Maria T. Bernardi
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (M.A.D.R.); (M.T.B.)
| | - Soledad Kleppe
- Department of Clinical Pediatrics, Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires C1199ABB, Argentina;
| | - Katherina Walz
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (M.A.D.R.); (M.T.B.)
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, 1501 NW 10th Avenue, BRB-418 (M-860), Miami, FL 33136, USA
| |
Collapse
|
33
|
Martini A, Cozza A, Di Pasquale Fiasca VM. The Inheritance of Hearing Loss and Deafness: A Historical Perspective. Audiol Res 2024; 14:116-128. [PMID: 38391767 PMCID: PMC10886121 DOI: 10.3390/audiolres14010010] [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: 11/19/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
If the term "genetics" is a relatively recent proposition, introduced in 1905 by English biologist William Bateson, who rediscovered and spread in the scientific community Mendel's principles of inheritance, since the dawn of human civilization the influence of heredity has been recognized, especially in agricultural crops and animal breeding. And, later, in familial dynasties. In this concise review, we outline the evolution of the idea of hereditary hearing loss, up to the current knowledge of molecular genetics and epigenetics.
Collapse
Affiliation(s)
- Alessandro Martini
- Padova University Research Center "International Auditory Processing Project in Venice (I-APPROVE)", Department of Neurosciences, University of Padua, 35128 Padua, Italy
| | - Andrea Cozza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy
| | | |
Collapse
|
34
|
Kun L, Jiexiang H, Hua L, Junlin H, Yijun R, Lixian Z, Mingqiao C. Genetic screening of 15 hearing loss variants in 77,647 neonates with clinical follow-up. Mol Genet Genomic Med 2024; 12:e2324. [PMID: 38037722 PMCID: PMC10767602 DOI: 10.1002/mgg3.2324] [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: 07/11/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND To analyze the genotype distribution and frequency of hearing loss genes in newborn population and evaluate the clinical value of genetic screening policy in China. METHODS Genetic screening for hearing loss was offered to 84,029 neonates between March 2019 and December 2021, of whom 77,647 newborns accepted the screening program with one-year follow-up. The genotyping of 15 hot spot variants in GJB2, GJB3, SLC26A4, and MT-RNR1 was performed on microarray platform. RESULTS A total of 3.05% (2369/77,647) newborns carried at least one genetic hearing loss-associated variant, indicated for early preventive management. The carrier frequency of GJB2 gene was the highest, at 1.48% (1147/77,647), followed by SLC26A4 gene at 1.07% (831/77,647), and GJB3 gene at 0.23% (181/77,647). GJB2 c.235delC variant and SLC26A4 IVS7-2A>G variant were the most common allelic variants with allele frequency of 0.6304% (979/155,294) and 0.3992% (620/155,294), respectively. 10 children are identified as homozygous or compound heterozygous for pathogenic variants (4 in GJB2, 6 in SLC26A4), and 7 of these infants had passed the hearing screening. Following up of the genetically screened newborns revealed that genetic screening detected more hearing-impaired infants than hearing screening alone. Genetic screening helped identify the infants who had passed the initial hearing screening, and reduced time for diagnosis and intervention of hearing aid. In addition, we identified 234 newborns (0.30%, 234/77,647) susceptible to preventable aminoglycoside antibiotic ototoxicity undetectable by hearing screening. CONCLUSION We performed the largest-scale neonatal carrier screening for hearing loss genes in Southeast China. Our results indicated that genetic screening is an important complementation to conventional hearing screening. Our practice and experience may facilitate the application and development of neonatal genetic screening policy in mainland China.
Collapse
Affiliation(s)
- Lin Kun
- Prenatal Diagnosis Center, The Affiliated Hospital of Putian UniversityPutian UniversityPutianChina
- Newborn Screening CenterPutian Maternity and Child Health Care HospitalPutianChina
| | - Huang Jiexiang
- Prenatal Diagnosis Center, The Affiliated Hospital of Putian UniversityPutian UniversityPutianChina
| | - Lin Hua
- Prenatal Diagnosis Center, The Affiliated Hospital of Putian UniversityPutian UniversityPutianChina
| | - Han Junlin
- Department of PediatricsPutian Maternity and Child Health Care HospitalPutianChina
| | - Ruan Yijun
- Department of PediatricsPutian Maternity and Child Health Care HospitalPutianChina
| | - Zhang Lixian
- Newborn Screening CenterPutian Maternity and Child Health Care HospitalPutianChina
| | - Chen Mingqiao
- Prenatal Diagnosis Center, The Affiliated Hospital of Putian UniversityPutian UniversityPutianChina
- Newborn Screening CenterPutian Maternity and Child Health Care HospitalPutianChina
- Department of PediatricsPutian Maternity and Child Health Care HospitalPutianChina
| |
Collapse
|
35
|
Liu S, Zhong M, Huang Y, Zhang Q, Chen T, Xu X, Peng W, Wang X, Feng X, Kang L, Lu Y, Cheng J, Bu F, Yuan H. Quantitative thresholds for variant enrichment in 13,845 cases: improving pathogenicity classification in genetic hearing loss. Genome Med 2023; 15:116. [PMID: 38111038 PMCID: PMC10726519 DOI: 10.1186/s13073-023-01271-7] [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: 07/21/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND The American College of Medical Genetics and Genomics (ACMG)/Association for Molecular Pathology (AMP) guidelines recommend using variant enrichment among cases as "strong" evidence for pathogenicity per the PS4 criterion. However, quantitative support for PS4 thresholds from real-world Mendelian case-control cohorts is lacking. METHODS To address this gap, we evaluated and established PS4 thresholds using data from the Chinese Deafness Genetics Consortium. A total of 9,050 variants from 13,845 patients with hearing loss (HL) and 6,570 ancestry-matched controls were analyzed. Positive likelihood ratio and local positive likelihood ratio values were calculated to determine the thresholds corresponding to each strength of evidence across three variant subsets. RESULTS In subset 1, consisting of variants present in both cases and controls with an allele frequency (AF) in cases ≥ 0.0005, an odds ratio (OR) ≥ 6 achieved strong evidence, while OR ≥ 3 represented moderate evidence. For subset 2, which encompassed variants present in both cases and controls with a case AF < 0.0005, and subset 3, comprising variants found only in cases and absent from controls, we defined the PS4_Supporting threshold (OR > 2.27 or allele count ≥ 3) and the PS4_Moderate threshold (allele count ≥ 6), respectively. Reanalysis applying the adjusted PS4 criteria changed the classification of 15 variants and enabled diagnosis of an additional four patients. CONCLUSIONS Our study quantified evidence strength thresholds for variant enrichment in genetic HL cases, highlighting the importance of defining disease/gene-specific thresholds to improve the precision and accuracy of clinical genetic testing.
Collapse
Affiliation(s)
- Sihan Liu
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Mingjun Zhong
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yu Huang
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Qian Zhang
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Ting Chen
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xiaofei Xu
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Wan Peng
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xiaolu Wang
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xiaoshu Feng
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Lu Kang
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yu Lu
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Jing Cheng
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Fengxiao Bu
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, 610000, China.
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China.
| | - Huijun Yuan
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, 610000, China.
- Institute of Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
36
|
Tesolin P, Santin A, Morgan A, Lenarduzzi S, Rubinato E, Girotto G, Spedicati B. Which Came First? When Usher Syndrome Type 1 Couples with Neuropsychiatric Disorders. Audiol Res 2023; 13:989-995. [PMID: 38131811 PMCID: PMC10740809 DOI: 10.3390/audiolres13060086] [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: 09/20/2023] [Revised: 11/17/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
Usher syndrome (USH) is an autosomal recessive disorder characterized by sensorineural hearing loss (HL), retinopathy, and vestibular areflexia, with variable severity. Although a high prevalence of behavioural and mental disorders in USH patients has been reported, few studies on these psychiatric and psychological issues have been conducted. This work describes the case of a 16-year-old boy affected by congenital bilateral sensorineural HL, presenting a suddenly altered behaviour concomitant with a decrease in visual acuity. To establish a molecular diagnosis, Whole-Exome Sequencing analysis was performed, detecting a pathogenetic homozygous variant (c. 5985C>A, p.(Tyr1995*)) within the CDH23 gene. CDH23 is a known USH type 1 causative gene, recently associated with schizophrenia-like symptoms and bipolar disorders. To date, no studies have provided evidence of a direct genotype-phenotype correlation between USH patients carrying CDH23 variants and mental/behavioural issues; however, considering the multiple biological functions of CDH23, it can be hypothesised that it could have a pleiotropic effect. Overall, this study highlights the relevance of a continuous clinical evaluation of USH patients, to monitor not only the disease progression, but to early detect any psychological or behavioural alterations, thus allowing a rapid implementation of therapeutic strategies aimed at improving their quality of life and well-being.
Collapse
Affiliation(s)
- Paola Tesolin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (P.T.); (G.G.); (B.S.)
| | - Aurora Santin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (P.T.); (G.G.); (B.S.)
| | - Anna Morgan
- Institute for Maternal and Child Health—I.R.C.C.S. “Burlo Garofolo”, 34137 Trieste, Italy (S.L.); (E.R.)
| | - Stefania Lenarduzzi
- Institute for Maternal and Child Health—I.R.C.C.S. “Burlo Garofolo”, 34137 Trieste, Italy (S.L.); (E.R.)
| | - Elisa Rubinato
- Institute for Maternal and Child Health—I.R.C.C.S. “Burlo Garofolo”, 34137 Trieste, Italy (S.L.); (E.R.)
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (P.T.); (G.G.); (B.S.)
- Institute for Maternal and Child Health—I.R.C.C.S. “Burlo Garofolo”, 34137 Trieste, Italy (S.L.); (E.R.)
| | - Beatrice Spedicati
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (P.T.); (G.G.); (B.S.)
- Institute for Maternal and Child Health—I.R.C.C.S. “Burlo Garofolo”, 34137 Trieste, Italy (S.L.); (E.R.)
| |
Collapse
|
37
|
Dmitriev DA, Shilov BV, Polunin MM, Zadorozhny AD, Lagunin AA. Predicting the Impact of OTOF Gene Missense Variants on Auditory Neuropathy Spectrum Disorder. Int J Mol Sci 2023; 24:17240. [PMID: 38139069 PMCID: PMC10743402 DOI: 10.3390/ijms242417240] [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: 11/06/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Auditory neuropathy spectrum disorder (ANSD) associated with mutations of the OTOF gene is one of the common types of sensorineural hearing loss of a hereditary nature. Due to its high genetic heterogeneity, ANSD is considered one of the most difficult hearing disorders to diagnose. The dataset from 270 known annotated single amino acid substitutions (SAV) related to ANSD was created. It was used to estimate the accuracy of pathogenicity prediction using the known (from dbNSFP4.4) method and a new one. The new method (ConStruct) for the creation of the protein-centric classification model is based on the use of Random Forest for the analysis of missense variants in exons of the OTOF gene. A system of predictor variables was developed based on the modern understanding of the structure and function of the otoferlin protein and reflecting the location of changes in the tertiary structure of the protein due to mutations in the OTOF gene. The conservation values of nucleotide substitutions in genomes of 100 vertebrates and 30 primates were also used as variables. The average prediction of balanced accuracy and the AUC value calculated by the 5-fold cross-validation procedure were 0.866 and 0.903, respectively. The model shows good results for interpreting data from the targeted sequencing of the OTOF gene and can be implemented as an auxiliary tool for the diagnosis of ANSD in the early stages of ontogenesis. The created model, together with the results of the pathogenicity prediction of SAVs via other known accurate methods, were used for the evaluation of a manually created set of 1302 VUS related to ANSD. Based on the analysis of predicted results, 16 SAVs were selected as the new most probable pathogenic variants.
Collapse
Affiliation(s)
- Dmitry A. Dmitriev
- Department of Bioinformatics, Medico-Biological Faculty, Pirogov Russian National Research Medical University, Moscow 117997, Russia; (D.A.D.); (B.V.S.); (A.D.Z.)
| | - Boris V. Shilov
- Department of Bioinformatics, Medico-Biological Faculty, Pirogov Russian National Research Medical University, Moscow 117997, Russia; (D.A.D.); (B.V.S.); (A.D.Z.)
| | - Michail M. Polunin
- Department of Otorhinolaryngology, Faculty of Pediatrics, Pirogov Russian National Research Medical University, Moscow 117997, Russia;
| | - Anton D. Zadorozhny
- Department of Bioinformatics, Medico-Biological Faculty, Pirogov Russian National Research Medical University, Moscow 117997, Russia; (D.A.D.); (B.V.S.); (A.D.Z.)
| | - Alexey A. Lagunin
- Department of Bioinformatics, Medico-Biological Faculty, Pirogov Russian National Research Medical University, Moscow 117997, Russia; (D.A.D.); (B.V.S.); (A.D.Z.)
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Hearing loss is the most common sensory deficit and in young children sensorineural hearing loss is most frequently genetic in etiology. Hearing aids and cochlear implant do not restore normal hearing. There is significant research and commercial interest in directly addressing the root cause of hearing loss through gene therapies. This article provides an overview of major barriers to cochlear gene therapy and recent advances in preclinical development of precision treatments of genetic deafness. RECENT FINDINGS Several investigators have recently described successful gene therapies in many common forms of genetic hearing loss in animal models. Elegant strategies that do not target a specific pathogenic variant, such as mini gene replacement and mutation-agnostic RNA interference (RNAi) with engineered replacement, facilitate translation of these findings to development of human therapeutics. Clinical trials for human gene therapies are in active recruitment. SUMMARY Gene therapies for hearing loss are expected to enter clinical trials in the immediate future. To provide referral for appropriate trials and counseling regarding benefits of genetic hearing loss evaluation, specialists serving children with hearing loss such as pediatricians, geneticists, genetic counselors, and otolaryngologists should be acquainted with ongoing developments in precision therapies.
Collapse
Affiliation(s)
- Miles J. Klimara
- Molecular Otolaryngology & Renal Research Laboratories, Department of Otolaryngology – Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Richard J.H. Smith
- Molecular Otolaryngology & Renal Research Laboratories, Department of Otolaryngology – Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
39
|
Yamamoto N, Balciuniene J, Hartman T, Diaz-Miranda MA, Bedoukian E, Devkota B, Lawrence A, Golenberg N, Patel M, Tare A, Chen R, Schindler E, Choi J, Kaur M, Charles S, Chen J, Fanning EA, Dechene E, Cao K, Jill MR, Rajagopalan R, Bayram Y, Dulik MC, Germiller J, Conlin LK, Krantz ID, Luo M. Comprehensive Gene Panel Testing for Hearing Loss in Children: Understanding Factors Influencing Diagnostic Yield. J Pediatr 2023; 262:113620. [PMID: 37473993 DOI: 10.1016/j.jpeds.2023.113620] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/17/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVE To evaluate factors influencing the diagnostic yield of comprehensive gene panel testing (CGPT) for hearing loss (HL) in children and to understand the characteristics of undiagnosed probands. STUDY DESIGN This was a retrospective cohort study of 474 probands with childhood-onset HL who underwent CGPT between 2016 and 2020 at a single center. Main outcomes and measures included the association between clinical variables and diagnostic yield and the genetic and clinical characteristics of undiagnosed probands. RESULTS The overall diagnostic yield was 44% (209/474) with causative variants involving 41 genes. While the diagnostic yield was high in the probands with congenital, bilateral, and severe HL, it was low in those with unilateral, noncongenital, or mild HL; cochlear nerve deficiency; preterm birth; neonatal intensive care unit admittance; certain ancestry; and developmental delay. Follow-up studies on 49 probands with initially inconclusive CGPT results changed the diagnostic status to likely positive or negative outcomes in 39 of them (80%). Reflex to exome sequencing on 128 undiagnosed probands by CGPT revealed diagnostic findings in 8 individuals, 5 of whom had developmental delays. The remaining 255 probands were undiagnosed, with 173 (173/255) having only a single variant in the gene(s) associated with autosomal recessive HL and 28% (48/173) having a matched phenotype. CONCLUSION CGPT efficiently identifies the genetic etiologies of HL in children. CGPT-undiagnosed probands may benefit from follow-up studies or expanded testing.
Collapse
Affiliation(s)
- Nobuko Yamamoto
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA; Division of Otolaryngology, Department of Surgical Specialties, National Center for Children's Health and Development, Tokyo, Japan; Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Jorune Balciuniene
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; PerkinElmer Genomics, Pittsburgh, PA
| | - Tiffiney Hartman
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Maria Alejandra Diaz-Miranda
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Emma Bedoukian
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Batsal Devkota
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Audrey Lawrence
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Netta Golenberg
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Maha Patel
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Archana Tare
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Robert Chen
- Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Emma Schindler
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jiwon Choi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Maninder Kaur
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sarah Charles
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jiani Chen
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Elizabeth A Fanning
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Elizabeth Dechene
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Kajia Cao
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Murrell R Jill
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Ramakrishnan Rajagopalan
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Yavuz Bayram
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Matthew C Dulik
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - John Germiller
- Division of Pediatric Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Otorhinolaryngology, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Laura K Conlin
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Ian D Krantz
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Roberts Individualized Medical Genetics Center (RIMGC), Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Minjie Luo
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
40
|
Tropitzsch A, Schade-Mann T, Gamerdinger P, Dofek S, Schulte B, Schulze M, Fehr S, Biskup S, Haack TB, Stöbe P, Heyd A, Harre J, Lesinski-Schiedat A, Büchner A, Lenarz T, Warnecke A, Müller M, Vona B, Dahlhoff E, Löwenheim H, Holderried M. Variability in Cochlear Implantation Outcomes in a Large German Cohort With a Genetic Etiology of Hearing Loss. Ear Hear 2023; 44:1464-1484. [PMID: 37438890 PMCID: PMC10583923 DOI: 10.1097/aud.0000000000001386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/04/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVES The variability in outcomes of cochlear implantation is largely unexplained, and clinical factors are not sufficient for predicting performance. Genetic factors have been suggested to impact outcomes, but the clinical and genetic heterogeneity of hereditary hearing loss makes it difficult to determine and interpret postoperative performance. It is hypothesized that genetic mutations that affect the neuronal components of the cochlea and auditory pathway, targeted by the cochlear implant (CI), may lead to poor performance. A large cohort of CI recipients was studied to verify this hypothesis. DESIGN This study included a large German cohort of CI recipients (n = 123 implanted ears; n = 76 probands) with a definitive genetic etiology of hearing loss according to the American College of Medical Genetics (ACMG)/Association for Molecular Pathology (AMP) guidelines and documented postoperative audiological outcomes. All patients underwent preoperative clinical and audiological examinations. Postoperative CI outcome measures were based on at least 1 year of postoperative audiological follow-up for patients with postlingual hearing loss onset (>6 years) and 5 years for children with congenital or pre/perilingual hearing loss onset (≤6 years). Genetic analysis was performed based on three different methods that included single-gene screening, custom-designed hearing loss gene panel sequencing, targeting known syndromic and nonsyndromic hearing loss genes, and whole-genome sequencing. RESULTS The genetic diagnosis of the 76 probands in the genetic cohort involved 35 genes and 61 different clinically relevant (pathogenic, likely pathogenic) variants. With regard to implanted ears (n = 123), the six most frequently affected genes affecting nearly one-half of implanted ears were GJB2 (21%; n = 26), TMPRSS3 (7%; n = 9), MYO15A (7%; n = 8), SLC26A4 (5%; n = 6), and LOXHD1 and USH2A (each 4%; n = 5). CI recipients with pathogenic variants that influence the sensory nonneural structures performed at or above the median level of speech performance of all ears at 70% [monosyllable word recognition score in quiet at 65 decibels sound pressure level (SPL)]. When gene expression categories were compared to demographic and clinical categories (total number of compared categories: n = 30), mutations in genes expressed in the spiral ganglion emerged as a significant factor more negatively affecting cochlear implantation outcomes than all clinical parameters. An ANOVA of a reduced set of genetic and clinical categories (n = 10) identified five detrimental factors leading to poorer performance with highly significant effects ( p < 0.001), accounting for a total of 11.8% of the observed variance. The single strongest category was neural gene expression accounting for 3.1% of the variance. CONCLUSIONS The analysis of the relationship between the molecular genetic diagnoses of a hereditary etiology of hearing loss and cochlear implantation outcomes in a large German cohort of CI recipients revealed significant variabilities. Poor performance was observed with genetic mutations that affected the neural components of the cochlea, supporting the "spiral ganglion hypothesis."
Collapse
Affiliation(s)
- Anke Tropitzsch
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Hearing Center, Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Center for Rare Hearing Disorders, Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
- Neurosensory Center, Departments of Otolaryngology—Head & Neck Surgery and Ophthalmology, University of Tübingen Medical Center, Tübingen, Germany
| | - Thore Schade-Mann
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Hearing Center, Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
| | - Philipp Gamerdinger
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Hearing Center, Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
| | - Saskia Dofek
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
| | - Björn Schulte
- CeGaT GmbH und Praxis für Humangenetik Tübingen, Tübingen, Germany
| | - Martin Schulze
- CeGaT GmbH und Praxis für Humangenetik Tübingen, Tübingen, Germany
| | - Sarah Fehr
- CeGaT GmbH und Praxis für Humangenetik Tübingen, Tübingen, Germany
| | - Saskia Biskup
- CeGaT GmbH und Praxis für Humangenetik Tübingen, Tübingen, Germany
| | - Tobias B. Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Petra Stöbe
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Andreas Heyd
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
| | - Jennifer Harre
- Department of Otorhinolaryngology—Head & Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation, Hannover, Germany
| | - Anke Lesinski-Schiedat
- Department of Otorhinolaryngology—Head & Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation, Hannover, Germany
| | - Andreas Büchner
- Department of Otorhinolaryngology—Head & Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation, Hannover, Germany
| | - Thomas Lenarz
- Department of Otorhinolaryngology—Head & Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otorhinolaryngology—Head & Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation, Hannover, Germany
| | - Marcus Müller
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Neurosensory Center, Departments of Otolaryngology—Head & Neck Surgery and Ophthalmology, University of Tübingen Medical Center, Tübingen, Germany
| | - Barbara Vona
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Neurosensory Center, Departments of Otolaryngology—Head & Neck Surgery and Ophthalmology, University of Tübingen Medical Center, Tübingen, Germany
| | - Ernst Dahlhoff
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Neurosensory Center, Departments of Otolaryngology—Head & Neck Surgery and Ophthalmology, University of Tübingen Medical Center, Tübingen, Germany
| | - Hubert Löwenheim
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Neurosensory Center, Departments of Otolaryngology—Head & Neck Surgery and Ophthalmology, University of Tübingen Medical Center, Tübingen, Germany
| | - Martin Holderried
- Department of Otolaryngology—Head & Neck Surgery, University of Tübingen Medical Center, Tübingen, Germany
- Department of Medical Development and Quality Management, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
41
|
Chiang YT, Lin PH, Lo MY, Chen HL, Lee CY, Tsai CY, Lin YH, Tsai SF, Liu TC, Hsu CJ, Chen PL, Hsu JSJ, Wu CC. Genetic Factors Contribute to the Phenotypic Variability in GJB2-Related Hearing Impairment. J Mol Diagn 2023; 25:827-837. [PMID: 37683890 DOI: 10.1016/j.jmoldx.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 09/10/2023] Open
Abstract
Recessive variants in GJB2 are the most important genetic cause of sensorineural hearing impairment (SNHI) worldwide. Phenotypes vary significantly in GJB2-related SNHI, even in patients with identical variants. For instance, patients homozygous for the GJB2 p.V37I variant, which is highly prevalent in the Asian populations, usually present with mild-to-moderate SNHI; yet severe-to-profound SNHI is occasionally observed in approximately 10% of p.V37I homozygotes. To investigate the genomic underpinnings of the phenotypic variability, we performed next-generation sequencing of GJB2 and other deafness genes in 63 p.V37I homozygotes with extreme phenotypic severities. Additional pathogenic variants of other deafness genes were identified in five of the 35 patients with severe-to-profound SNHI. Furthermore, case-control association analyses were conducted for 30 unrelated p.V37I homozygotes with severe-to-profound SNHI against 28 p.V37I homozygotes with mild-to-moderate SNHI, and 120 population controls from the Taiwan Biobank. The severe-to-profound group exhibited a higher frequency of the crystallin lambda 1 (CRYL1) variant (rs14236), located upstream of GJB2, than the mild-to-moderate and Taiwan Biobank groups. Our results demonstrated that pathogenic variants in other deafness genes and a possible modifier, the CRYL1 rs14236 variant, may contribute to phenotypic variability in GJB2-realted SNHI, highlighting the importance of comprehensive genomic surveys to delineate the genotype-phenotype correlations.
Collapse
Affiliation(s)
- Yu-Ting Chiang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Hsuan Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Otolaryngology Head and Neck Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Yu Lo
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Lin Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Surgical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Chen-Yu Lee
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Cheng-Yu Tsai
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yin-Hung Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shih-Feng Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Tien-Chen Liu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chuan-Jen Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Otolaryngology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Otolaryngology Head and Neck Surgery, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jacob Shu-Jui Hsu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Chen-Chi Wu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| |
Collapse
|
42
|
Davis KN, Qu PP, Ma S, Lin L, Plastini M, Dahl N, Plazzi G, Pizza F, O’Hara R, Wong WH, Hallmayer J, Mignot E, Zhang X, Urban AE. Mutations in human DNA methyltransferase DNMT1 induce specific genome-wide epigenomic and transcriptomic changes in neurodevelopment. Hum Mol Genet 2023; 32:3105-3120. [PMID: 37584462 PMCID: PMC10586194 DOI: 10.1093/hmg/ddad123] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 08/17/2023] Open
Abstract
DNA methyltransferase type 1 (DNMT1) is a major enzyme involved in maintaining the methylation pattern after DNA replication. Mutations in DNMT1 have been associated with autosomal dominant cerebellar ataxia, deafness and narcolepsy (ADCA-DN). We used fibroblasts, induced pluripotent stem cells (iPSCs) and induced neurons (iNs) generated from patients with ADCA-DN and controls, to explore the epigenomic and transcriptomic effects of mutations in DNMT1. We show cell type-specific changes in gene expression and DNA methylation patterns. DNA methylation and gene expression changes were negatively correlated in iPSCs and iNs. In addition, we identified a group of genes associated with clinical phenotypes of ADCA-DN, including PDGFB and PRDM8 for cerebellar ataxia, psychosis and dementia and NR2F1 for deafness and optic atrophy. Furthermore, ZFP57, which is required to maintain gene imprinting through DNA methylation during early development, was hypomethylated in promoters and exhibited upregulated expression in patients with ADCA-DN in both iPSC and iNs. Our results provide insight into the functions of DNMT1 and the molecular changes associated with ADCA-DN, with potential implications for genes associated with related phenotypes.
Collapse
Affiliation(s)
- Kasey N Davis
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - Ping-Ping Qu
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - Shining Ma
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Ling Lin
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Center for Narcolepsy, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Melanie Plastini
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - Niklas Dahl
- Department of Immunology, Genetics and Pathology Sciences for Life Laboratory, Uppsala University BMC, Uppsala 75122, Sweden
| | - Giuseppe Plazzi
- IRCCS—Istituto delle Scienze Neurologiche di Bologna, Bologna 40139, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Fabio Pizza
- IRCCS—Istituto delle Scienze Neurologiche di Bologna, Bologna 40139, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40126, Italy
| | - Ruth O’Hara
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Wing Hung Wong
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Joachim Hallmayer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Emmanuel Mignot
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Center for Narcolepsy, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Xianglong Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - Alexander E Urban
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto CA 94304, USA
| |
Collapse
|
43
|
Posukh OL, Maslova EA, Danilchenko VY, Zytsar MV, Orishchenko KE. Functional Consequences of Pathogenic Variants of the GJB2 Gene (Cx26) Localized in Different Cx26 Domains. Biomolecules 2023; 13:1521. [PMID: 37892203 PMCID: PMC10604905 DOI: 10.3390/biom13101521] [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: 09/15/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
One of the most common forms of genetic deafness has been predominantly associated with pathogenic variants in the GJB2 gene, encoding transmembrane protein connexin 26 (Cx26). The Cx26 molecule consists of an N-terminal domain (NT), four transmembrane domains (TM1-TM4), two extracellular loops (EL1 and EL2), a cytoplasmic loop, and a C-terminus (CT). Pathogenic variants in the GJB2 gene, resulting in amino acid substitutions scattered across the Cx26 domains, lead to a variety of clinical outcomes, including the most common non-syndromic autosomal recessive deafness (DFNB1A), autosomal dominant deafness (DFNA3A), as well as syndromic forms combining hearing loss and skin disorders. However, for rare and poorly documented variants, information on the mode of inheritance is often lacking. Numerous in vitro studies have been conducted to elucidate the functional consequences of pathogenic GJB2 variants leading to amino acid substitutions in different domains of Cx26 protein. In this work, we summarized all available data on a mode of inheritance of pathogenic GJB2 variants leading to amino acid substitutions and reviewed published information on their functional effects, with an emphasis on their localization in certain Cx26 domains.
Collapse
Affiliation(s)
- Olga L. Posukh
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.M.); (V.Y.D.); (M.V.Z.); (K.E.O.)
- Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ekaterina A. Maslova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.M.); (V.Y.D.); (M.V.Z.); (K.E.O.)
- Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Valeriia Yu. Danilchenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.M.); (V.Y.D.); (M.V.Z.); (K.E.O.)
- Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Marina V. Zytsar
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.M.); (V.Y.D.); (M.V.Z.); (K.E.O.)
| | - Konstantin E. Orishchenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.M.); (V.Y.D.); (M.V.Z.); (K.E.O.)
- Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
44
|
Daneshi A, Garshasbi M, Farhadi M, Falavarjani KG, Vafaee-Shahi M, Almadani N, Zabihi M, Ghalavand MA, Falah M. Genetic insights into PHARC syndrome: identification of a novel frameshift mutation in ABHD12. BMC Med Genomics 2023; 16:235. [PMID: 37803361 PMCID: PMC10557151 DOI: 10.1186/s12920-023-01682-w] [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: 04/08/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Mutations in ABHD12 (OMIM: 613,599) are associated with polyneuropathy, hearing loss, ataxia, retinitis pigmentosa, and cataract (PHARC) syndrome (OMIM: 612674), which is a rare autosomal recessive neurodegenerative disease. PHARC syndrome is easily misdiagnosed as other neurologic disorders, such as retinitis pigmentosa, Charcot-Marie-Tooth disease, and Refsum disease, due to phenotype variability and slow progression. This paper presents a novel mutation in ABHD12 in two affected siblings with PHARC syndrome phenotypes. In addition, we summarize genotype-phenotype information of the previously reported patients with ABHD12 mutation. METHODS Following a thorough medical evaluation, whole-exome sequencing was done on the proband to look for potential genetic causes. This was followed by confirmation of identified variant in the proband and segregation analysis in the family by Sanger sequencing. The variants were interpreted based on the American College of Medical Genetics and Genomics (ACMG) guidelines. RESULTS A novel pathogenic homozygous frameshift variant, NM_001042472.3:c.601dup, p.(Val201GlyfsTer4), was identified in exon 6 of ABHD12 (ACMG criteria: PVS1 and PM2, PM1, PM4, PP3, and PP4). Through Sanger sequencing, we showed that this variant is co-segregated with the disease in the family. Further medical evaluations confirmed the compatibility of the patients' phenotype with PHARC syndrome. CONCLUSIONS Our findings expand the spectrum of mutations in the ABHD12 and emphasize the significance of multidisciplinary diagnostic collaboration among clinicians and geneticists to solve the differential diagnosis of related disorders. Moreover, a summary based on mutations found so far in the ABHD12 gene did not suggest a clear genotype-phenotype correlation for PHARC syndrome.
Collapse
Affiliation(s)
- Ahmad Daneshi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Khalil Ghasemi Falavarjani
- Eye Research Centre, Five Senses Health Institute, School of Medicine, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vafaee-Shahi
- Pediatric Growth and Development Research Center, Institute of Endocrinology and metabolism, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Navid Almadani
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - MohammadSina Zabihi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Ghalavand
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
- 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, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Sumalde AAM, Yang IV, Yarza TKL, Tobias-Grasso CAM, Tantoco MLC, Davidson E, Chan AL, Azamian MS, Cruz TLG, Lalani SR, Reyes-Quintos MRT, Cutiongco-de la Paz EM, Santos-Cortez RLP, Chiong CM. Lack of Methylation Changes in GJB2 and RB1 Non-coding Regions of Cochlear Implant Patients with Sensorineural Hearing Loss. ACTA MEDICA PHILIPPINA 2023; 57:116-120. [PMID: 37990697 PMCID: PMC10662870 DOI: 10.47895/amp.v57i9.5200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Objective Recent advances in epigenetic studies continue to reveal novel mechanisms of gene regulation and control, however little is known on the role of epigenetics in sensorineural hearing loss (SNHL) in humans. We aimed to investigate the methylation patterns of two regions, one in RB1 and another in GJB2 in Filipino patients with SNHL compared to hearing control individuals. Methods We investigated an RB1 promoter region that was previously identified as differentially methylated in children with SNHL and lead exposure. Additionally, we investigated a sequence in an enhancer-like region within GJB2 that contains four CpGs in close proximity. Bisulfite conversion was performed on salivary DNA samples from 15 children with SNHL and 45 unrelated ethnically-matched individuals. We then performed methylation-specific real-time PCR analysis (qMSP) using TaqMan® probes to determine percentage methylation of the two regions. Results Using qMSP, both our cases and controls had zero methylation at the targeted GJB2 and RB1 regions. Conclusion Our study showed no changes in methylation at the selected CpG regions in RB1 and GJB2 in the two comparison groups with or without SNHL. This may be due to a lack of environmental exposures to these target regions. Other epigenetic marks may be present around these regions as well as those of other HL-associated genes.
Collapse
Affiliation(s)
- Angelo Augusto M. Sumalde
- College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Otolaryngology – Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus (CU-AMC), Aurora, Colorado, USA
- Department of Otolaryngology-Head and Neck Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Ivana V. Yang
- Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus (CU-AMC), Aurora, Colorado, USA
| | - Talitha Karisse L. Yarza
- Philippine National Ear Institute, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Newborn Hearing Screening Reference Center, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | | | - Ma. Leah C. Tantoco
- Department of Otolaryngology-Head and Neck Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
- Philippine National Ear Institute, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Newborn Hearing Screening Reference Center, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Elizabeth Davidson
- Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus (CU-AMC), Aurora, Colorado, USA
| | - Abner L. Chan
- College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Otolaryngology-Head and Neck Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Mahshid S. Azamian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Teresa Luisa G. Cruz
- College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Otolaryngology-Head and Neck Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Seema R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Maria Rina T. Reyes-Quintos
- Department of Otolaryngology-Head and Neck Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
- Philippine National Ear Institute, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Newborn Hearing Screening Reference Center, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Eva Maria Cutiongco-de la Paz
- National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Philippine Genome Center, UP Diliman Campus, Quezon City, Philippines
| | - Regie Lyn P. Santos-Cortez
- Department of Otolaryngology – Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus (CU-AMC), Aurora, Colorado, USA
| | - Charlotte M. Chiong
- College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Otolaryngology-Head and Neck Surgery, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
- Philippine National Ear Institute, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Newborn Hearing Screening Reference Center, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
46
|
Borgese N, Guillén-Samander A, Colombo SF, Mancassola G, Di Berardino F, Zanetti D, Carrera P. Combined Presence in Heterozygosis of Two Variant Usher Syndrome Genes in Two Siblings Affected by Isolated Profound Age-Related Hearing Loss. Biomedicines 2023; 11:2657. [PMID: 37893031 PMCID: PMC10604119 DOI: 10.3390/biomedicines11102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Sensorineural age-related hearing loss affects a large proportion of the elderly population, and has both environmental and genetic causes. Notwithstanding increasing interest in this debilitating condition, the genetic risk factors remain largely unknown. Here, we report the case of two sisters affected by isolated profound sensorineural hearing loss after the age of seventy. Genomic DNA sequencing revealed that the siblings shared two monoallelic variants in two genes linked to Usher Syndrome (USH genes), a recessive disorder of the ear and the retina: a rare pathogenic truncating variant in USH1G and a previously unreported missense variant in ADGRV1. Structure predictions suggest a negative effect on protein stability of the latter variant, allowing its classification as likely pathogenic according to American College of Medical Genetics criteria. Thus, the presence in heterozygosis of two recessive alleles, which each cause syndromic deafness, may underlie digenic inheritance of the age-related non-syndromic hearing loss of the siblings, a hypothesis that is strengthened by the knowledge that the two genes are integrated in the same functional network, which underlies stereocilium development and organization. These results enlarge the spectrum and complexity of the phenotypic consequences of USH gene mutations beyond the simple Mendelian inheritance of classical Usher syndrome.
Collapse
Affiliation(s)
- Nica Borgese
- Consiglio Nazionale delle Ricerche Neuroscience Institute, 20854 Vedano al Lambro, Italy;
| | | | - Sara Francesca Colombo
- Consiglio Nazionale delle Ricerche Neuroscience Institute, 20854 Vedano al Lambro, Italy;
- NeuroMi Milan Center for Neuroscience, Milan, Italy
| | - Giulia Mancassola
- Unit of Genomics for Human Disease Diagnosis, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
- Laboratory of Clinical Molecular Genetics, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Federica Di Berardino
- Audiology Unit, Department of Specialistic Surgical Sciences, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.D.B.); (D.Z.)
- Department of Clinical Sciences and Community Health, University of Milano, 20122 Milan, Italy
| | - Diego Zanetti
- Audiology Unit, Department of Specialistic Surgical Sciences, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.D.B.); (D.Z.)
- Department of Clinical Sciences and Community Health, University of Milano, 20122 Milan, Italy
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
- Laboratory of Clinical Molecular Genetics, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
47
|
Hu S, Sun Q, Xu F, Jiang N, Gao J. Age-related hearing loss and its potential drug candidates: a systematic review. Chin Med 2023; 18:121. [PMID: 37730634 PMCID: PMC10512576 DOI: 10.1186/s13020-023-00825-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Age-related hearing loss (ARHL) is one of the main illnesses afflicting the aged population and has a significant negative impact on society, economy, and health. However, there is presently no appropriate therapeutic treatment of ARHL due to the absence of comprehensive trials. OBJECTIVES The goal of this review is to systematically evaluate and analyze recent statistics on the pathologic classifications, risk factors, treatment strategies, and drug candidates of ARHL, including that from traditional Chinese medicine (TCM), to provide potential new approaches for preventing and treating ARHL. METHODS Literature related to ARHL was conducted in databases such as PubMed, WOS, China National Knowledge Infrastructure (CNKI), and Wanfang from the establishment of the database to Jan, 2023. The pathology, causal factor, pathophysiological mechanism, treatment strategy, and the drug candidate of ARHL were extracted and pooled for synthesis. RESULTS Many hypotheses about the etiology of ARHL are based on genetic and environmental elements. Most of the current research on the pathology of ARHL focuses on oxidative damage, mitochondrial dysfunction, inflammation, cochlear blood flow, ion homeostasis, etc. In TCM, herbs belonging to the kidney, lung, and liver meridians exhibit good hearing protection. Seven herbs belonging to the kidney meridian, 9 belonging to the lung meridian, and 4 belonging to the liver meridian were ultimately retrieved in this review, such as Polygonum multiflorum Thunb., Panax ginseng C.A. Mey, and Pueraria lobata (Willd.) Ohwi. Their active compounds, 2,3,4',5-Tetrahydroxystilbene-2-O-D-glucoside, ginsenoside Rb1, and puerarin, may act as the molecular substance for their anti-ARHL efficacy, and show anti-oxidative, neuroprotective, anti-inflammatory, anti-apoptotic, or mitochondrial protective effects. CONCLUSION Anti-oxidants, modulators of mitochondrial function, anti-inflammation agents, vasodilators, K+ channel openers, Ca2+ channel blockers, JNK inhibitors, and nerve growth factors/neurotrophic factors all contribute to hearing protection, and herbs are an important source of potential anti-ARHL drugs.
Collapse
Affiliation(s)
- Shiyu Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Qingru Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Fei Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Ninghua Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| |
Collapse
|
48
|
Isgrig K, Cartagena-Rivera AX, Wang HJ, Grati M, Fernandez KA, Friedman TB, Belyantseva IA, Chien W. Combined AAV-mediated gene replacement therapy improves auditory function in a mouse model of human DFNB42 deafness. Mol Ther 2023; 31:2783-2795. [PMID: 37481704 PMCID: PMC10492026 DOI: 10.1016/j.ymthe.2023.07.014] [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: 02/08/2023] [Revised: 05/30/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023] Open
Abstract
Hearing loss is a common disorder affecting nearly 20% of the world's population. Recently, studies have shown that inner ear gene therapy can improve auditory function in several mouse models of hereditary hearing loss. In most of these studies, the underlying mutations affect only a small number of cell types of the inner ear (e.g., sensory hair cells). Here, we applied inner ear gene therapy to the Ildr1Gt(D178D03)Wrst (Ildr1w-/-) mouse, a model of human DFNB42, non-syndromic autosomal recessive hereditary hearing loss associated with ILDR1 variants. ILDR1 is an integral protein of the tricellular tight junction complex and is expressed by diverse inner ear cell types in the organ of Corti and the cochlear lateral wall. We simultaneously applied two synthetic adeno-associated viruses (AAVs) with different tropism to deliver Ildr1 cDNA to the Ildr1w-/- mouse inner ear: one targeting the organ of Corti (AAV2.7m8) and the other targeting the cochlear lateral wall (AAV8BP2). We showed that combined AAV2.7m8/AAV8BP2 gene therapy improves cochlear structural integrity and auditory function in Ildr1w-/- mice.
Collapse
Affiliation(s)
- Kevin Isgrig
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Hong Jun Wang
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Mhamed Grati
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Katharine A Fernandez
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Inna A Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Wade Chien
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA; Department of Otolaryngology - Head & Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Vona B, Regele S, Rad A, Strenzke N, Pater JA, Neumann K, Sturm M, Haack TB, Am Zehnhoff-Dinnesen AG. Unraveling haplotype errors in the DFNA33 locus. Front Genet 2023; 14:1214736. [PMID: 37671045 PMCID: PMC10475583 DOI: 10.3389/fgene.2023.1214736] [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: 05/02/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
Genetic heterogeneity makes it difficult to identify the causal genes for hearing loss. Studies from previous decades have mapped numerous genetic loci, providing critical supporting evidence for gene discovery studies. Despite widespread sequencing accessibility, many historically mapped loci remain without a causal gene. The DFNA33 locus was mapped in 2009 and coincidentally contains ATP11A, a gene recently associated with autosomal dominant hearing loss and auditory neuropathy type 2. In a rare opportunity, we genome-sequenced a member of the original family to determine whether the DFNA33 locus may also be assigned to ATP11A. We identified a deep intronic variant in ATP11A that showed evidence of functionally normal splicing. Furthermore, we re-assessed haplotypes from the originally published DFNA33 family and identified two double recombination events and one triple recombination event in the pedigree, a highly unlikely occurrence, especially at this scale. This brief research report also serves as a call to the community to revisit families who have previously been involved in gene mapping studies, provide closure, and resolve these historical loci.
Collapse
Affiliation(s)
- Barbara Vona
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Sabrina Regele
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, University of Münster, Münster, Germany
| | - Aboulfazl Rad
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Department of Otolaryngology and Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
| | - Justin A. Pater
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- Health Sciences Centre, Faculty of Medicine, Memorial University, St. John’s, NL, Canada
| | - Katrin Neumann
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, University of Münster, Münster, Germany
| | - Marc Sturm
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Tobias B. Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
50
|
Lachgar-Ruiz M, Morín M, Martelletti E, Ingham NJ, Preite L, Lewis MA, Serrão de Castro LS, Steel KP, Moreno-Pelayo MÁ. Insights into the pathophysiology of DFNA44 hearing loss associated with CCDC50 frameshift variants. Dis Model Mech 2023; 16:dmm049757. [PMID: 37165931 PMCID: PMC10445743 DOI: 10.1242/dmm.049757] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 05/02/2023] [Indexed: 05/12/2023] Open
Abstract
Non-syndromic sensorineural hearing loss (SNHL) is the most common sensory disorder, and it presents a high genetic heterogeneity. As part of our clinical genetic studies, we ascertained a previously unreported mutation in CCDC50 [c.828_858del, p.(Asp276Glufs*40)] segregating with hearing impairment in a Spanish family with SNHL associated with the autosomal dominant deafness locus DFNA44, which is predicted to disrupt protein function. To gain insight into the mechanism behind DFNA44 mutations, we analysed two Ccdc50 presumed loss-of-function mouse mutants, which showed normal hearing thresholds up to 6 months of age, indicating that haploinsufficiency is unlikely to be the pathogenic mechanism. We then carried out in vitro studies on a set of artificial mutants and on the p.(Asp276Glufs*40) and p.(Phe292Hisfs*37) human mutations, and determined that only the mutants containing the six-amino-acid sequence CLENGL as part of their aberrant protein tail showed an abnormal distribution consisting of perinuclear aggregates of the CCDC50 protein (also known as Ymer). Therefore, we conclude that the CLENGL sequence is necessary to form these aggregates. Taken together, the in vivo and in vitro results obtained in this study suggest that the two identified mutations in CCDC50 exert their effect through a dominant-negative or gain-of-function mechanism rather than by haploinsufficiency.
Collapse
Affiliation(s)
- María Lachgar-Ruiz
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS and Biomedical Network Research Centre on Rare Diseases (CIBERER), km 9.100, 28034 Madrid, Spain
| | - Matías Morín
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS and Biomedical Network Research Centre on Rare Diseases (CIBERER), km 9.100, 28034 Madrid, Spain
| | - Elisa Martelletti
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Neil J. Ingham
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Lorenzo Preite
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Morag A. Lewis
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Luciana Santos Serrão de Castro
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS and Biomedical Network Research Centre on Rare Diseases (CIBERER), km 9.100, 28034 Madrid, Spain
| | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Miguel Ángel Moreno-Pelayo
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS and Biomedical Network Research Centre on Rare Diseases (CIBERER), km 9.100, 28034 Madrid, Spain
| |
Collapse
|