1
|
Zhang X, Wu J, Wang M, Chen L, Wang P, Jiang Q, Yang C. The role of gene mutations and immune responses in sensorineural hearing loss. Int Immunopharmacol 2024; 143:113515. [PMID: 39486181 DOI: 10.1016/j.intimp.2024.113515] [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: 09/10/2024] [Revised: 10/12/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Sensorineural hearing loss (SNHL) is a prevalent clinical condition primarily attributed to dysfunction within various components of the auditory pathway, spanning from the inner ear to the auditory cortex. Recent research has illuminated immune and inflammation-mediated disorders of the inner ear as critical contributors to SNHL. Disruptions in the equilibrium of inflammatory mediators, chemokines, the complement system, and inflammatory vesicles within the cochlea provoke aberrations in immune cell activity, fostering a chronic pro-inflammatory milieu that detrimentally affects the structural and functional integrity of the inner ear, culminating in hearing impairment. Specific genetic mutations, especially those affecting auditory structures, play an important role in SNHL. These mutations regulate inflammatory mediators and cellular responses, thereby altering the inflammatory dynamics within the cochlea. This review delves into the pathogenesis of sensorineural hearing loss, emphasizing the impact of genetic alterations, immune responses within the inner ear, and inflammatory mediators on auditory function. It highlights the significance of Transmembrane Serine Protease 3 (TMPRSS3) and connexin gene mutations as pivotal genetic elements in SNHL, underscoring the central role of inflammatory responses in cochlear damage. Furthermore, the paper discusses the promise of gene therapy and targeted molecular interventions, underscoring the necessity for continued exploration into the specific actions of various inflammatory agents to refine personalized therapeutic strategies.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Junyi Wu
- Department of Otolaryngology-Head and Neck Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu Province, China
| | - Maohua Wang
- Department of Otolaryngology, Head and Neck Surgery, The First People's Hospital of Foshan, Hearing and Balance Medical Engineering Technology Center of Guangdong, Foshan, 528000, China
| | - Li Chen
- Department of Otolaryngology-Head and Neck Surgery, The Second People's Hospital of Yibin City, Sichuan Province, 644000, China
| | - Peng Wang
- Department of Otolaryngology-Head and Neck Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Jiangsu Province, 225200, China
| | - Qiao Jiang
- Department of Neurology, Deyang Fifth Hospital, Sichuan Province, 618000, China.
| | - Chunping Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
2
|
Kraujaliene L, Kraujalis T, Snipas M, Verselis VK. An Ala/Glu difference in E1 of Cx26 and Cx30 contributes to their differential anionic permeabilities. J Gen Physiol 2024; 156:e202413600. [PMID: 39302317 PMCID: PMC11415307 DOI: 10.1085/jgp.202413600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Two closely related connexins, Cx26 and Cx30, share widespread expression in the cochlear cellular networks. Gap junction channels formed by these connexins have been shown to have different permeability profiles, with Cx30 showing a strongly reduced preference for anionic tracers. The pore-forming segment of the first extracellular loop, E1, identified by computational studies of the Cx26 crystal structure to form a parahelix and a narrowed region of the pore, differs at a single residue at position 49. Cx26 contains an Ala and Cx30, a charged Glu at this position, and cysteine scanning in hemichannels identified this position to be pore-lining. To assess whether the Ala/Glu difference affects permeability, we modeled and quantified Lucifer Yellow transfer between HeLa cell pairs expressing WT Cx26 and Cx30 and variants that reciprocally substituted Glu and Ala at position 49. Cx26(A49E) and Cx30(E49A) substitutions essentially reversed the Lucifer Yellow permeability profile when accounting for junctional conductance. Moreover, by using a calcein efflux assay in single cells, we observed a similar reduced anionic preference in undocked Cx30 hemichannels and a reversal with reciprocal Ala/Glu substitutions. Thus, our data indicate that Cx26 and Cx30 gap junction channels and undocked hemichannels retain similar permeability characteristics and that a single residue difference in their E1 domains can largely account for their differential permeabilities to anionic tracers. The higher anionic permeability of Cx26 compared with Cx30 suggests that these connexins may serve distinct signaling functions in the cochlea, perhaps reflected in the vastly higher prevalence of Cx26 mutations in human deafness.
Collapse
Affiliation(s)
- Lina Kraujaliene
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Applied Informatics, Kaunas University of Technology, Kaunas, Lithuania
| | - Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania
| | - Vytas K. Verselis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
3
|
Sun T, Li W, Shi K, Zhao Y, Guo D, Wang D. The Role of Connexin26 and Connexin30 in the Mouse Cochlea of Noise-Induced Hearing Loss. Otolaryngol Head Neck Surg 2024. [PMID: 39369436 DOI: 10.1002/ohn.1002] [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: 01/03/2024] [Revised: 08/15/2024] [Accepted: 09/14/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE We aimed to explore the role of connexin26 (Cx26) and connexin30 (Cx30) in the cochlea in noise-induced permanent threshold shifts (PTS) and temporary threshold shift (TTS). STUDY DESIGN Prospective, controlled. SETTING Laboratory. METHODS A mouse model of noise-induced PTS and TTS was constructed. Western blots were used to detect the expression of Cx26 and Cx30 in the cochlea. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to assess the potential biological pathways. RESULTS Both the expression of Cx26 and Cx30 showed a trend of first rising and then falling in noise-induced PTS. The expression of Cx26 increased greatly in the 24 hours noise exposure (P < .05) and reached the highest level in the 4 hours after noise exposure (P < .05), then decreased gradually and returned to the control level on the seventh day after the noise exposure, when compared with the control group. The expression of Cx30 showed a similar trend in noise-induced PTS. However, both the expression of Cx26 and Cx30 showed a trend of first falling and then rising in noise induced TTS. The expression of Cx26/Cx30 reached its lowest level in the 4 hours after noise exposure (P < .05), and then increased to the control level on the second day after noise exposure (P > .05), compared with the control group. The first KEGG and GO pathway may be related with oxidative phosphorylation. CONCLUSION Cx26 and Cx30 may have an effect in noise induced PTS and TTS. Future studies are needed to confirm the results.
Collapse
Affiliation(s)
- Tao Sun
- Department of Clinical Medicine, Henan Medical College, Zhengzhou, Henan, China
- Henan Provincial Health and Wellness Commission Key Laboratory of Hearing Loss Mechanism Research, Zhengzhou, Henan, China
| | - Wenzhen Li
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute of the Chinese University of Hong Kong, Shenzhen, China
| | - Ke Shi
- Henan Provincial Health and Wellness Commission Key Laboratory of Hearing Loss Mechanism Research, Zhengzhou, Henan, China
- Department of Biochemistry and Molecular Biology, Henan Medical College, Zhengzhou, Henan, China
| | - Yue Zhao
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Guo
- Department of Clinical Medicine, Henan Medical College, Zhengzhou, Henan, China
- Henan Provincial Health and Wellness Commission Key Laboratory of Hearing Loss Mechanism Research, Zhengzhou, Henan, China
- The Second Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Dongming Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
4
|
He C, Gai H, Zhao W, Zhang H, Lai L, Ding C, Chen L, Ding J. Advances in the Study of Etiology and Molecular Mechanisms of Sensorineural Hearing Loss. Cell Biochem Biophys 2024; 82:1721-1734. [PMID: 38849694 DOI: 10.1007/s12013-024-01344-3] [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] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Sensorineural hearing loss (SNHL), a multifactorial progressive disorder, results from a complex interplay of genetic and environmental factors, with its underlying mechanisms remaining unclear. Several pathological factors are believed to contribute to SNHL, including genetic factors, ion homeostasis, cell apoptosis, immune inflammatory responses, oxidative stress, hormones, metabolic syndrome, human cytomegalovirus infection, mitochondrial damage, and impaired autophagy. These factors collectively interact and play significant roles in the onset and progression of SNHL. The present review offers a comprehensive overview of the various factors that contribute to SNHL, emphasizes recent developments in understanding its etiology, and explores relevant preventive and intervention measures.
Collapse
Affiliation(s)
- Cairong He
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Hongcun Gai
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Wen Zhao
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Haiqin Zhang
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Lin Lai
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Chenyu Ding
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Lin Chen
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Jie Ding
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
5
|
Wang L, Zhang R, Jiang L, Gao S, Wu J, Jiao Y. Biomaterials as a new option for treating sensorineural hearing loss. Biomater Sci 2024; 12:4006-4023. [PMID: 38979939 DOI: 10.1039/d4bm00518j] [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: 07/10/2024]
Abstract
Sensorineural hearing loss (SNHL) usually involves damage to complex auditory pathways such as inner ear cells and auditory nerves. The highly intricate and nuanced characteristics of these cells render their repair and regeneration extremely challenging, making it difficult to restore hearing to normal levels once it has been compromised. The effectiveness of traditional drugs is so minimal that they provide little help with the treatment. Fortunately, extensive experiments have demonstrated that combining biomaterials with conventional techniques significantly enhances drug effectiveness. This article reviews the research progress of biomaterials in protecting hair cells and the auditory nerve, repairing genes related to hearing, and developing artificial cochlear materials. By organizing the knowledge presented in this article, perhaps new insights can be provided for the clinical management of SNHL.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| | - Ruhe Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Linlan Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| | - Shuyi Gao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| | - Jun Wu
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China.
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuenong Jiao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| |
Collapse
|
6
|
Yeral C, Seneldir L, Karakoc AH, Sap A, Yilmaz O. Unusual phenotype in 35delG mutation: a case report. J Med Case Rep 2024; 18:241. [PMID: 38734626 PMCID: PMC11088767 DOI: 10.1186/s13256-024-04559-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Mutations in the GJB2 gene, which encodes the protein connexin 26 and is involved in inner ear homeostasis, are identified in approximately 50% of patients with autosomal recessive nonsyndromic hearing loss, making it one of the primary causes of prelingual nonsyndromic hearing loss in various populations. The 35delG mutation, one of the most common mutations of the GJB2 gene, usually causes prelingual, bilateral mild to profound, nonprogressive sensorineural hearing loss. CASE PRESENTATION We present an unusual case of an 18-year-old Turkish female with heterozygous 35delG mutation and postlingual, profound-sloping, progressive and fluctuating unilateral sensorineural hearing loss. The phenotype is different from the usual findings. CONCLUSIONS The 35delG mutation causing hearing loss may not always be reflected in the phenotype as expected and therefore may have different audiologic manifestations.
Collapse
Affiliation(s)
- Cem Yeral
- Faculty of Health Sciences, Department of Audiology, İstanbul University-Cerrahpaşa, Istanbul, Türkiye.
| | - Lutfu Seneldir
- Faculty of Medicine, Department of Otolaryngology, Istanbul Medipol University, Istanbul, Türkiye
| | - Arzu Hediye Karakoc
- Faculty of Health Sciences, Department of Audiology, Istanbul Medipol University, Istanbul, Türkiye
| | - Aleyna Sap
- Faculty of Health Sciences, Department of Audiology, Istanbul Medipol University, Istanbul, Türkiye
| | - Oguz Yilmaz
- Faculty of Health Sciences, Department of Audiology, Istanbul Medipol University, Istanbul, Türkiye
| |
Collapse
|
7
|
Tlili A, Mutery AA, Chouchen J. The Segregation of p.Arg68Ter- CLDN14 Mutation in a Syrian Deaf Family, Phenotypic Variations, and Comparative Analysis with the GJB2 Gene. Genes (Basel) 2024; 15:588. [PMID: 38790217 PMCID: PMC11121454 DOI: 10.3390/genes15050588] [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: 04/10/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Hearing impairment, a rare inherited condition, is notably prevalent in populations with high rates of consanguinity. The most common form observed globally is autosomal recessive non-syndromic hearing loss. Despite its prevalence, this genetic disorder is characterized by a substantial genetic diversity, making diagnosis and screening challenging. The emergence of advanced next-generation sequencing (NGS) technologies has significantly advanced the discovery of genes and variants linked to various conditions, such as hearing loss. In this study, our objective was to identify the specific variant causing hearing loss in a family from Syria using clinical exome sequencing. The proband in the family exhibited profound deafness as shown by pure-tone audiometry results. The analysis of the different variants obtained by NGS revealed the presence of a nonsense mutation within the CLDN14 gene. Through Sanger sequencing, we verified that this variant segregates with the disease and was not present in the control population. Moreover, we conducted a comprehensive review of all reported deafness-related CLDN14 mutations and their associated phenotypes. Furthermore, we endeavored to carry out a comparative analysis between the CLDN14 and GJB2 genes, with the objective of identifying potential factors that could explain the notable discrepancy in mutation frequency between these two genes.
Collapse
Affiliation(s)
- Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Abdullah Al Mutery
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Jihen Chouchen
- Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates;
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Ali-Nazari MM, Rahbar N, Haddadzade Niri H, Vasaghi-Gharamaleki B. Mefloquine-Induced Inner Ear Damage and Preventive Effects of Electrical Stimulation: An Electrophysiological Study. Audiol Neurootol 2023; 29:88-95. [PMID: 37607499 DOI: 10.1159/000531788] [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: 02/12/2023] [Accepted: 06/15/2023] [Indexed: 08/24/2023] Open
Abstract
INTRODUCTION Mefloquine is an antimalarial medicine used to prevent and treat malaria. This medicine has some side effects, including ototoxicity. This study, which was designed in two phases, aimed to investigate the side effects of mefloquine and evaluate the preventive effects of electrical stimulation on these side effects. METHODS In the first phase, two doses of mefloquine (50 and 200 μM) were injected into male rats, and after 7 days, they were evaluated by an auditory brainstem response (ABR) test. In the second phase, electrical stimulation was applied for 10 days, and then a toxic dose of mefloquine was injected. Similar to the first phase of the study, the animals were evaluated by an ABR test after 7 days. RESULTS In the first phase, the results showed that a high dose of mefloquine increased the ABR threshold and wave I latency; however, these changes were not observed in the second phase. CONCLUSION Application of electrical stimulation could prevent the ototoxic effects of mefloquine. According to the findings of the present study, electrical stimulation can be used as a preconditioner to prevent the ototoxic effects of mefloquine.
Collapse
Affiliation(s)
- Mohammad Mahdi Ali-Nazari
- Rehabilitation Research Center, Department of Audiology, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran,
| | - Nariman Rahbar
- Rehabilitation Research Center, Department of Audiology, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Haddadzade Niri
- Rehabilitation Research Center, Department of Audiology, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Behnoosh Vasaghi-Gharamaleki
- Rehabilitation Research Center, Department of Rehabilitation Basic Sciences, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Tawk K, Abouzari M. Cx26 heterozygous mutations: role in hyperacusis and vulnerability to noise. Signal Transduct Target Ther 2023; 8:264. [PMID: 37423920 PMCID: PMC10330177 DOI: 10.1038/s41392-023-01525-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/09/2023] [Accepted: 05/28/2023] [Indexed: 07/11/2023] Open
Affiliation(s)
- Karen Tawk
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA, USA
| | - Mehdi Abouzari
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA, USA.
| |
Collapse
|
12
|
Tsao PC, Lin HC, Chiu HY, Chang YC. Maternal, Perinatal, and Postnatal Predisposing Factors of Hearing Loss in Full-Term Children: A Matched Case-Control Study. Neonatology 2023; 120:607-614. [PMID: 37321203 DOI: 10.1159/000530646] [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: 11/03/2022] [Accepted: 03/21/2023] [Indexed: 06/17/2023]
Abstract
INTRODUCTION Studies on risk factors for childhood hearing loss (HL) are usually based on questionnaires or small sample sizes. We conducted a nationwide population-based case-control study to comprehensively analyze the maternal, perinatal, and postnatal risk factors for HL in full-term children. METHODS We retrieved data from three nationwide databases related to maternal characteristics, perinatal comorbidities, and postnatal characteristics and adverse events. We used 1:5 propensity score matching to include 12,873 full-term children with HL and 64,365 age-, sex-, and enrolled year-matched controls. Conditional logistic regression was used to evaluate the risk factors for HL. RESULTS Among the various maternal factors, maternal HL (adjusted odds ratio [aOR]: 8.09, 95% confidence interval [95% CI]: 7.16-9.16) and type 1 diabetes (aOR: 3.79, 95% CI: 1.98-7.24) had the highest odds of childhood hearing impairment. The major perinatal risk factors for childhood hearing impairment included ear malformations (aOR: 58.78, 95% CI: 37.5-92.0) and chromosomal anomalies (aOR: 6.70, 95% CI: 5.25-8.55), and the major postnatal risk factors included meningitis (aOR: 2.08, 95% CI: 1.18-3.67) and seizure (aOR: 3.71, 95% CI: 2.88-4.77). Other factors included acute otitis media, postnatal ototoxic drug use, and congenital infections. CONCLUSIONS Many risk factors for childhood HL identified in our study are preventable, such as congenital infection, meningitis, ototoxic drug use, and some maternal comorbidities. Accordingly, more effort is required to prevent and control the severity of maternal comorbidities during pregnancy, initiate genetic diagnostic evaluation for high-risk children, and aggressive screening for neonatal infections.
Collapse
Affiliation(s)
- Pei-Chen Tsao
- Department of Pediatrics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan,
| | - Hung-Chih Lin
- Division of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Pediatrics, Asia University Hospital, Asia University, Taichung, Taiwan
| | - Hsiao-Yu Chiu
- Division of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan
| | - Yu-Chia Chang
- Department of Long Term Care, College of Health and Nursing, National Quemoy University, Kinmen County, Jinning, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
13
|
Huang SN, Pan YT, Zhou YP, Wang XZ, Mei MJ, Yang B, Li D, Zeng WB, Cheng S, Sun JY, Cheng H, Zhao F, Luo MH. Human Cytomegalovirus IE1 Impairs Neuronal Migration by Downregulating Connexin 43. J Virol 2023; 97:e0031323. [PMID: 37097169 PMCID: PMC10231247 DOI: 10.1128/jvi.00313-23] [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: 02/27/2023] [Accepted: 04/07/2023] [Indexed: 04/26/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a leading cause of congenital birth defects. Though the underlying mechanisms remain poorly characterized, mouse models of congenital CMV infection have demonstrated that the neuronal migration process is damaged. In this study, we evaluated the effects of HCMV infection on connexin 43 (Cx43), a crucial adhesion molecule mediating neuronal migration. We show in multiple cellular models that HCMV infection downregulated Cx43 posttranslationally. Further analysis identified the immediate early protein IE1 as the viral protein responsible for the reduction of Cx43. IE1 was found to bind the Cx43 C terminus and promote Cx43 degradation through the ubiquitin-proteasome pathway. Deletion of the Cx43-binding site in IE1 rendered it incapable of inducing Cx43 degradation. We validated the IE1-induced loss of Cx43 in vivo by introducing IE1 into the fetal mouse brain. Noteworthily, ectopic IE1 expression induced cortical atrophy and neuronal migration defects. Several lines of evidence suggest that these damages result from decreased Cx43, and restoration of Cx43 levels partially rescued IE1-induced interruption of neuronal migration. Taken together, the results of our investigation reveal a novel mechanism of HCMV-induced neural maldevelopment and identify a potential intervention target. IMPORTANCE Congenital CMV (cCMV) infection causes neurological sequelae in newborns. Recent studies of cCMV pathogenesis in animal models reveal ventriculomegaly and cortical atrophy associated with impaired neural progenitor cell (NPC) proliferation and migration. In this study, we investigated the mechanisms underlying these NPC abnormalities. We show that Cx43, a critical adhesion molecule mediating NPC migration, is downregulated by HCMV infection in vitro and HCMV-IE1 in vivo. We provide evidence that IE1 interacts with the C terminus of Cx43 to promote its ubiquitination and consequent degradation through the proteasome. Moreover, we demonstrate that introducing IE1 into mouse fetal brains led to neuronal migration defects, which was associated with Cx43 reduction. Deletion of the Cx43-binding region in IE1 or ectopic expression of Cx43 rescued the IE1-induced migration defects in vivo. Our study provides insight into how cCMV infection impairs neuronal migration and reveals a target for therapeutic interventions.
Collapse
Affiliation(s)
- Sheng-Nan Huang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu-Ting Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yue-Peng Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xian-Zhang Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meng-Jie Mei
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bo Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Dong Li
- Chinese Institute for Brain Research, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wen-Bo Zeng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shuang Cheng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jin-Yan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Han Cheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Fei Zhao
- Chinese Institute for Brain Research, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
14
|
Abrams CK. Mechanisms of Diseases Associated with Mutation in GJC2/Connexin 47. Biomolecules 2023; 13:biom13040712. [PMID: 37189458 DOI: 10.3390/biom13040712] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Connexins are members of a family of integral membrane proteins that provide a pathway for both electrical and metabolic coupling between cells. Astroglia express connexin 30 (Cx30)-GJB6 and Cx43-GJA1, while oligodendroglia express Cx29/Cx31.3-GJC3, Cx32-GJB1, and Cx47-GJC2. Connexins organize into hexameric hemichannels (homomeric if all subunits are identical or heteromeric if one or more differs). Hemichannels from one cell then form cell-cell channels with a hemichannel from an apposed cell. (These are termed homotypic if the hemichannels are identical and heterotypic if the hemichannels differ). Oligodendrocytes couple to each other through Cx32/Cx32 or Cx47/Cx47 homotypic channels and they couple to astrocytes via Cx32/Cx30 or Cx47/Cx43 heterotypic channels. Astrocytes couple via Cx30/Cx30 and Cx43/Cx43 homotypic channels. Though Cx32 and Cx47 may be expressed in the same cells, all available data suggest that Cx32 and Cx47 cannot interact heteromerically. Animal models wherein one or in some cases two different CNS glial connexins have been deleted have helped to clarify the role of these molecules in CNS function. Mutations in a number of different CNS glial connexin genes cause human disease. Mutations in GJC2 lead to three distinct phenotypes, Pelizaeus Merzbacher like disease, hereditary spastic paraparesis (SPG44) and subclinical leukodystrophy.
Collapse
Affiliation(s)
- Charles K Abrams
- Department of Neurology and Rehabilitation, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Mao L, Wang Y, An L, Zeng B, Wang Y, Frishman D, Liu M, Chen Y, Tang W, Xu H. Molecular Mechanisms and Clinical Phenotypes of GJB2 Missense Variants. BIOLOGY 2023; 12:biology12040505. [PMID: 37106706 PMCID: PMC10135792 DOI: 10.3390/biology12040505] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 03/29/2023]
Abstract
The GJB2 gene is the most common gene responsible for hearing loss (HL) worldwide, and missense variants are the most abundant type. GJB2 pathogenic missense variants cause nonsyndromic HL (autosomal recessive and dominant) and syndromic HL combined with skin diseases. However, the mechanism by which these different missense variants cause the different phenotypes is unknown. Over 2/3 of the GJB2 missense variants have yet to be functionally studied and are currently classified as variants of uncertain significance (VUS). Based on these functionally determined missense variants, we reviewed the clinical phenotypes and investigated the molecular mechanisms that affected hemichannel and gap junction functions, including connexin biosynthesis, trafficking, oligomerization into connexons, permeability, and interactions between other coexpressed connexins. We predict that all possible GJB2 missense variants will be described in the future by deep mutational scanning technology and optimizing computational models. Therefore, the mechanisms by which different missense variants cause different phenotypes will be fully elucidated.
Collapse
Affiliation(s)
- Lu Mao
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Yueqiang Wang
- Basecare Medical Device Co., Ltd., Suzhou 215000, China
| | - Lei An
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Beiping Zeng
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Wang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Dmitrij Frishman
- Wissenschaftszentrum Weihenstephan, Technische Universitaet Muenchen, Am Staudengarten 2, 85354 Freising, Germany
| | - Mengli Liu
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Yanyu Chen
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Wenxue Tang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
- Correspondence:
| |
Collapse
|
16
|
Degranulation of Murine Resident Cochlear Mast Cells: A Possible Factor Contributing to Cisplatin-Induced Ototoxicity and Neurotoxicity. Int J Mol Sci 2023; 24:ijms24054620. [PMID: 36902051 PMCID: PMC10003316 DOI: 10.3390/ijms24054620] [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: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Permanent hearing loss is one of cisplatin's adverse effects, affecting 30-60% of cancer patients treated with that drug. Our research group recently identified resident mast cells in rodents' cochleae and observed that the number of mast cells changed upon adding cisplatin to cochlear explants. Here, we followed that observation and found that the murine cochlear mast cells degranulate in response to cisplatin and that the mast cell stabilizer cromoglicic acid (cromolyn) inhibits this process. Additionally, cromolyn significantly prevented cisplatin-induced loss of auditory hair cells and spiral ganglion neurons. Our study provides the first evidence for the possible mast cell participation in cisplatin-induced damage to the inner ear.
Collapse
|
17
|
Wang L, Yi D, Geng Y, Duan T, Tong Z, Chen S, Ning Z, Du Y, Hong X, Li X. Ultrasensitive deafness gene DNA hybridization detection employing a fiber optic Mach-Zehnder interferometer: Enabled by a black phosphorus nanointerface. Biosens Bioelectron 2023; 222:114952. [PMID: 36455374 DOI: 10.1016/j.bios.2022.114952] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
The rapid and efficient detection of deafness gene DNA plays an important role in the clinical diagnosis of deafness diseases. This study demonstrates the ultrasensitive detection of complementary DNA (cDNA) by employing a nanointerface-sensitized fiber optic biosensor. The sensor consists of SMF-TNCF-MMF-SMF (abbreviated as STMS) structure with lateral offset. Besides, it is functionalized with a nanointerface of black phosphorus (BP) to enhance the light-matter interaction and eventually improve the sensing performances. Relying on this nanointerface-sensitized sensor, we successfully realize the in-situ detection of cDNA at concentrations ranging from 1 pM to 1 μM, with a sensitivity of 0.719 nm/lgM. The limit of detection (LOD) is as low as 0.24 pM, which is at least two orders of magnitude lower than those of existing methods. The sensor exhibits the advantages of simple operation, fast response, label-free measurement, excellent repeatability, and high selectivity. Our contribution suggests a convenient approach for deafness gene DNA detection and can be extended for general ultra-low concentration DNA detection applications.
Collapse
Affiliation(s)
- Lina Wang
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Duo Yi
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Youfu Geng
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China.
| | - Tingting Duan
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Zhifeng Tong
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Shaolan Chen
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Zhiyao Ning
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Yu Du
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Xueming Hong
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Sensor Technology, Shenzhen University, Shenzhen, 518060, PR China
| | - Xuejin Li
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, 518172, PR China.
| |
Collapse
|
18
|
Liu LM, Liang C, Chen J, Fang S, Zhao HB. Cx26 heterozygous mutations cause hyperacusis-like hearing oversensitivity and increase susceptibility to noise. SCIENCE ADVANCES 2023; 9:eadf4144. [PMID: 36753545 PMCID: PMC9908021 DOI: 10.1126/sciadv.adf4144] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 06/18/2023]
Abstract
Gap junction gene GJB2 (Cx26) mutations cause >50% of nonsyndromic hearing loss. Its recessive hetero-mutation carriers, who have no deafness, occupy ~10 to 20% of the general population. Here, we report an unexpected finding that these heterozygote carriers have hearing oversensitivity, and active cochlear amplification increased. Mouse models show that Cx26 hetero-deletion reduced endocochlear potential generation in the cochlear lateral wall and caused outer hair cell electromotor protein prestin compensatively up-regulated to increase active cochlear amplification and hearing sensitivity. The increase of active cochlear amplification also increased sensitivity to noise; exposure to daily-level noise could cause Cx26+/- mice permanent hearing threshold shift, leading to hearing loss. This study demonstrates that Cx26 recessive heterozygous mutations are not "harmless" for hearing as previously considered and can cause hyperacusis-like hearing oversensitivity. The data also indicate that GJB2 hetero-mutation carriers are vulnerable to noise and should avoid noise exposure in daily life.
Collapse
Affiliation(s)
- Li-Man Liu
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
- Department of Surgery–Otolaryngology, Yale University Medical School, 310 Cedar Street, New Haven, CT 06510, USA
| | - Chun Liang
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
- Hearing Function Testing Center, Shenzhen Maternity and Child Healthcare Hospital, 3012 Fuqiang Road, Shenzhen 518017, China
| | - Jin Chen
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Shu Fang
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Hong-Bo Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
- Department of Surgery–Otolaryngology, Yale University Medical School, 310 Cedar Street, New Haven, CT 06510, USA
| |
Collapse
|
19
|
Aboagye ET, Adadey SM, Wonkam-Tingang E, Amenga-Etego L, Awandare GA, Wonkam A. Global Distribution of Founder Variants Associated with Non-Syndromic Hearing Impairment. Genes (Basel) 2023; 14:399. [PMID: 36833326 PMCID: PMC9957346 DOI: 10.3390/genes14020399] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The genetic etiology of non-syndromic hearing impairment (NSHI) is highly heterogeneous with over 124 distinct genes identified. The wide spectrum of implicated genes has challenged the implementation of molecular diagnosis with equal clinical validity in all settings. Differential frequencies of allelic variants in the most common NSHI causal gene, gap junction beta 2 (GJB2), has been described as stemming from the segregation of a founder variant and/or spontaneous germline variant hot spots. We aimed to systematically review the global distribution and provenance of founder variants associated with NSHI. The study protocol was registered on PROSPERO, the International Prospective Register of Systematic Reviews, with the registration number "CRD42020198573". Data from 52 reports, involving 27,959 study participants from 24 countries, reporting 56 founder pathogenic or likely pathogenic (P/LP) variants in 14 genes (GJB2, GJB6, GSDME, TMC1, TMIE, TMPRSS3, KCNQ4, PJVK, OTOF, EYA4, MYO15A, PDZD7, CLDN14, and CDH23), were reviewed. Varied number short tandem repeats (STRs) and single nucleotide polymorphisms (SNPs) were used for haplotype analysis to identify the shared ancestral informative markers in a linkage disequilibrium and variants' origins, age estimates, and common ancestry computations in the reviewed reports. Asia recorded the highest number of NSHI founder variants (85.7%; 48/56), with variants in all 14 genes, followed by Europe (16.1%; 9/56). GJB2 had the highest number of ethnic-specific P/LP founder variants. This review reports on the global distribution of NSHI founder variants and relates their evolution to population migration history, bottleneck events, and demographic changes in populations linked with the early evolution of deleterious founder alleles. International migration and regional and cultural intermarriage, coupled to rapid population growth, may have contributed to re-shaping the genetic architecture and structural dynamics of populations segregating these pathogenic founder variants. We have highlighted and showed the paucity of data on hearing impairment (HI) variants in Africa, establishing unexplored opportunities in genetic traits.
Collapse
Affiliation(s)
- Elvis Twumasi Aboagye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra LG Box 54, Ghana
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Samuel Mawuli Adadey
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra LG Box 54, Ghana
| | - Edmond Wonkam-Tingang
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Lucas Amenga-Etego
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra LG Box 54, Ghana
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra LG Box 54, Ghana
| | - Ambroise Wonkam
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- McKusick-Nathans Institute and Department of Genetic Medicine, John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Chiereghin C, Robusto M, Lewis MA, Caetano S, Massa V, Castorina P, Ambrosetti U, Steel KP, Duga S, Asselta R, Soldà G. In-depth genetic and molecular characterization of diaphanous related formin 2 (DIAPH2) and its role in the inner ear. PLoS One 2023; 18:e0273586. [PMID: 36689403 PMCID: PMC9870134 DOI: 10.1371/journal.pone.0273586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Diaphanous related formins are regulatory cytoskeletal protein involved in actin elongation and microtubule stabilization. In humans, defects in two of the three diaphanous genes (DIAPH1 and DIAPH3) have been associated with different types of hearing loss. Here, we investigate the role of the third member of the family, DIAPH2, in nonsyndromic hearing loss, prompted by the identification, by exome sequencing, of a predicted pathogenic missense variant in DIAPH2. This variant occurs at a conserved site and segregated with nonsyndromic X-linked hearing loss in an Italian family. Our immunohistochemical studies indicated that the mouse ortholog protein Diaph2 is expressed during development in the cochlea, specifically in the actin-rich stereocilia of the sensory outer hair cells. In-vitro studies showed a functional impairment of the mutant DIAPH2 protein upon RhoA-dependent activation. Finally, Diaph2 knock-out and knock-in mice were generated by CRISPR/Cas9 technology and auditory brainstem response measurements performed at 4, 8 and 14 weeks. However, no hearing impairment was detected. Our findings indicate that DIAPH2 may play a role in the inner ear; further studies are however needed to clarify the contribution of DIAPH2 to deafness.
Collapse
Affiliation(s)
| | - Michela Robusto
- Experimental Therapeutics Program, IFOM ETS -The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Morag A. Lewis
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Susana Caetano
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Valentina Massa
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milano, Italy
| | | | - Umberto Ambrosetti
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano and Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, UO Audiologia, Milano, Italy
| | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Stefano Duga
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Rosanna Asselta
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Giulia Soldà
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| |
Collapse
|
21
|
Pun R, Kim MH, North BJ. Role of Connexin 43 phosphorylation on Serine-368 by PKC in cardiac function and disease. Front Cardiovasc Med 2023; 9:1080131. [PMID: 36712244 PMCID: PMC9877470 DOI: 10.3389/fcvm.2022.1080131] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Intercellular communication mediated by gap junction channels and hemichannels composed of Connexin 43 (Cx43) is vital for the propagation of electrical impulses through cardiomyocytes. The carboxyl terminal tail of Cx43 undergoes various post-translational modifications including phosphorylation of its Serine-368 (S368) residue. Protein Kinase C isozymes directly phosphorylate S368 to alter Cx43 function and stability through inducing conformational changes affecting channel permeability or promoting internalization and degradation to reduce intercellular communication between cardiomyocytes. Recent studies have implicated this PKC/Cx43-pS368 circuit in several cardiac-associated diseases. In this review, we describe the molecular and cellular basis of PKC-mediated Cx43 phosphorylation and discuss the implications of Cx43 S368 phosphorylation in the context of various cardiac diseases, such as cardiomyopathy, as well as the therapeutic potential of targeting this pathway.
Collapse
Affiliation(s)
- Renju Pun
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Michael H. Kim
- CHI Health Heart Institute, School of Medicine, Creighton University, Omaha, NE, United States
| | - Brian J. North
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States,*Correspondence: Brian J. North,
| |
Collapse
|
22
|
Abbott AC, García IE, Villanelo F, Flores-Muñoz C, Ceriani R, Maripillán J, Novoa-Molina J, Figueroa-Cares C, Pérez-Acle T, Sáez JC, Sánchez HA, Martínez AD. Expression of KID syndromic mutation Cx26S17F produces hyperactive hemichannels in supporting cells of the organ of Corti. Front Cell Dev Biol 2023; 10:1071202. [PMID: 36699003 PMCID: PMC9868548 DOI: 10.3389/fcell.2022.1071202] [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: 10/15/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Some mutations in gap junction protein Connexin 26 (Cx26) lead to syndromic deafness, where hearing impairment is associated with skin disease, like in Keratitis Ichthyosis Deafness (KID) syndrome. This condition has been linked to hyperactivity of connexin hemichannels but this has never been demonstrated in cochlear tissue. Moreover, some KID mutants, like Cx26S17F, form hyperactive HCs only when co-expressed with other wild-type connexins. In this work, we evaluated the functional consequences of expressing a KID syndromic mutation, Cx26S17F, in the transgenic mouse cochlea and whether co-expression of Cx26S17F and Cx30 leads to the formation of hyperactive HCs. Indeed, we found that cochlear explants from a constitutive knock-in Cx26S17F mouse or conditional in vitro cochlear expression of Cx26S17F produces hyperactive HCs in supporting cells of the organ of Corti. These conditions also produce loss of hair cells stereocilia. In supporting cells, we found high co-localization between Cx26S17F and Cx30. The functional properties of HCs formed in cells co-expressing Cx26S17F and Cx30 were also studied in oocytes and HeLa cells. Under the recording conditions used in this study Cx26S17F did not form functional HCs and GJCs, but cells co-expressing Cx26S17F and Cx30 present hyperactive HCs insensitive to HCs blockers, Ca2+ and La3+, resulting in more Ca2+ influx and cellular damage. Molecular dynamic analysis of putative heteromeric HC formed by Cx26S17F and Cx30 presents alterations in extracellular Ca2+ binding sites. These results support that in KID syndrome, hyperactive HCs are formed by the interaction between Cx26S17F and Cx30 in supporting cells probably causing damage to hair cells associated to deafness.
Collapse
Affiliation(s)
- Ana C. Abbott
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile,Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Viña del Mar, Chile
| | - Isaac E. García
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile,Laboratorio de Fisiología Molecular y Biofísica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso, Chile,Centro de Investigaciones en Ciencias Odontológicas y Médicas, CICOM, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Villanelo
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago, Chile,Computational Biology Lab, Centro Basal Ciencia & Vida, Universidad San Sebastián, Santiago, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ricardo Ceriani
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile,Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso, Chile
| | - Jaime Maripillán
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Joel Novoa-Molina
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Cindel Figueroa-Cares
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Tomas Pérez-Acle
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago, Chile,Computational Biology Lab, Centro Basal Ciencia & Vida, Universidad San Sebastián, Santiago, Chile
| | - Juan C. Sáez
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Helmuth A. Sánchez
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile,*Correspondence: Helmuth A. Sánchez, ; Agustín D. Martínez,
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencias de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile,*Correspondence: Helmuth A. Sánchez, ; Agustín D. Martínez,
| |
Collapse
|
23
|
Sarkaria S, Callahan M, Hostoffer Z, Venglarcik J, Hostoffer R. IgM Deficiency Associated With Connexin Mutation in an 18-Year-old Male. THERAPEUTIC ADVANCES IN ALLERGY AND RHINOLOGY 2023; 14:27534030231156116. [PMID: 36968253 PMCID: PMC10035527 DOI: 10.1177/27534030231156116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
IgM deficiency is characterized by remarkably low serum levels of IgM with normal IgG and IgA levels. These patients clinically present with recurrent infections, autoimmune disorders, and malignancies. While unknown, the proposed mechanisms explain the pathophysiology as an issue due to impaired IgG antibody response. The connexin genes encode for gap junctional proteins where mutations can cause hearing deficits and immune dysregulation. We present a unique case of an 18-year-old patient with recurrent sinusitis, diagnosed connexin-26 mutation and an IgM deficiency. An 18-year-old male with chronic sinusitis, Marfanoid joint hypermobility syndrome, and sensorineural hearing loss due to connexin-26 deficiency with bilateral cochlear implants. This patient's mutation is a GJB2 deletion located on chromosome 13 which encodes for the connexin-26 protein. The patient experienced recurrent infections, and serum immunoglobulins showed a normal IgA (84 mg/dL; normal: 70-400 mg/dL), IgG (922 mg/dL; normal: 700–1600 mg/dL) and reduced IgM (26 mg/dL; normal: 40–230 mg/dL) levels. The patient was responsive to Mumps, Measles, Rubella, and Diphtheria vaccinations among others, consistent with SIGMD diagnoses. Antibody responses to polysaccharide antigens were absent. The leukocyte counts were within normal limits. His parents are connexin-26 deficient carriers, and his older brother was diagnosed with SIGMD. Connexin-26 has been identified with multiple immunological mechanisms. Although mutations of this gene have no direct tie to antibody formation in relation to IgM, the presence of these 2 pathologies in 1 patient is intriguing and may suggest a pathophysiologic connection. We describe the first case of connexin mutation with an IgM deficiency in an 18-year-old male.
Collapse
Affiliation(s)
- Sandeep Sarkaria
- University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | | | | | | | - Robert Hostoffer
- University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
- Allergy/Immunology Associates Inc., Mayfield Heights, OH, USA
| |
Collapse
|
24
|
Hotkani MM, Esmaeilpoor MRM, Khadem M, Garmaroudi AA, Inanloorahatloo K. Intermittent white noise exposure is associated with rat cochleae damage and changes in the gene expression. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00317-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Noise, a physical factor in most work environments, has many effects on human health. Exposure to excessive noise can modify the expression of associated genes with NIHL. The aim of this study to elucidate changes in expression of GJB2 and SLC26A4 after exposure to intense noise which are the most frequent causing genes to apparent autosomal recessive non-syndromic hearing loss.
Methods
In this experimental and case–control study, 17 male Wistar rats were randomly divided into exposure groups (n = 12) and without exposure (n = 5). First group was exposed to noise (90–120 dB, 70 Hz–16 kHz, 8 h/day) for 3–6 days. Cochlear biopsies performed 1 h and 1 week post-exposure, relative gene expression levels were calculated using $${2}^{-\Delta \Delta Ct}$$
2
-
Δ
Δ
C
t
. From each group, one ear was stained by hematoxylin and eosin method for histopathological survey. Real-time PCR technique took place, and gene expression data were normalized by GAPDH gene. One-way ANOVA test was performed with a significance level of 0.05 by GraphPad prism software.
Results
Both GJB2 and SLC26A4 in all groups were down-regulated after exposure compared to their controls. Fold changes in the highest times were related to 1 week after 6 days of exposure, 0.052 and 0.015, respectively. Serious damages occurred in different parts of cochlea, and they were more severe after 6 days and 1-week later.
Conclusion
It is expected that if the hearing threshold tests be performed before/after exposure and considering longer post-exposure times, subsequently, the expression of these genes does not return to basal level, and irrecoverable damage to the cochlea, progressive and irreversible ARNSHL will be expected.
Collapse
|
25
|
Jang MW, Lim J, Park MG, Lee JH, Lee CJ. Active role of glia-like supporting cells in the organ of Corti: Membrane proteins and their roles in hearing. Glia 2022; 70:1799-1825. [PMID: 35713516 DOI: 10.1002/glia.24229] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022]
Abstract
The organ of Corti, located in the cochlea in the inner ear, is one of the major sensory organs involved in hearing. The organ of Corti consists of hair cells, glia-like supporting cells, and the cochlear nerve, which work in harmony to receive sound from the outer ear and transmit auditory signals to the cochlear nucleus in the auditory ascending pathway. In this process, maintenance of the endocochlear potential, with a high potassium gradient and clearance of electrolytes and biochemicals in the inner ear, is critical for normal sound transduction. There is an emerging need for a thorough understanding of each cell type involved in this process to understand the sophisticated mechanisms of the organ of Corti. Hair cells have long been thought to be active, playing a primary role in the cochlea in actively detecting and transmitting signals. In contrast, supporting cells are thought to be silent and function to support hair cells. However, growing lines of evidence regarding the membrane proteins that mediate ionic movement in supporting cells have demonstrated that supporting cells are not silent, but actively play important roles in normal signal transduction. In this review, we summarize studies that characterize diverse membrane proteins according to the supporting cell subtypes involved in cochlear physiology and hearing. This review contributes to a better understanding of supporting cell functions and facilitates the development of potential therapeutic tools for hearing loss.
Collapse
Affiliation(s)
- Minwoo Wendy Jang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Mingu Gordon Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - C Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
26
|
The protective effects of systemic dexamethasone on sensory epithelial damage and hearing loss in targeted Cx26-null mice. Cell Death Dis 2022; 13:545. [PMID: 35688810 PMCID: PMC9187686 DOI: 10.1038/s41419-022-04987-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 01/21/2023]
Abstract
Mutations in the GJB2 gene (encoding Connexin26(Cx26)) are the most common cause of hereditary deafness, accounting for about a quarter of all cases. Sensory epithelial damage is considered to be one of the main causes of deafness caused by GJB2 gene mutation. Dexamethasone (DEX) is widely used in the treatment of a variety of inner ear diseases including sudden sensorineural hearing loss (SSNHL), noise-induced hearing loss (NIHL), and deafness caused by ototoxic drugs. Whether DEX has a direct therapeutic effect on hereditary deafness, especially GJB2-related deafness, remains unclear. In this study, we revealed that DEX can effectively prevent hair cell death caused by oxidative stress in cochlear explants. Additionally, two distinct Cx26-null mouse models were established to investigate whether systemic administration of DEX alleviate the cochlear sensory epithelial injury or deafness in these models. In a specific longitudinally Cx26-null model that does not cause deafness, systemic administration of DEX prevents the degeneration of outer hair cells (OHCs) induced by Cx26 knockout. Similarly, in a targeted-Deiter's cells (DCs) Cx26-null mouse model that causes deafness, treatment with DEX can almost completely prevent OHCs loss and alleviates auditory threshold shifts at some frequencies. Additionally, we observed that DEX inhibited the recruitment of CD45-positive cells in the targeted-DCs Cx26-null mice. Taken together, our results suggest that the protective effect of dexamethasone on cochlear sensory epithelial damage and partially rescue auditory function may be related to the regulation of inner ear immune response in Cx26 deficiency mouse models.
Collapse
|
27
|
Qiu Y, Xu K, Xie L, Chen S, Sun Y. The Reduction in Microtubule Arrays Caused by the Dysplasia of the Non-Centrosomal Microtubule-Organizing Center Leads to a Malformed Organ of Corti in the Cx26-Null Mouse. Biomedicines 2022; 10:biomedicines10061364. [PMID: 35740388 PMCID: PMC9219875 DOI: 10.3390/biomedicines10061364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in the GJB2 gene account for approximately 20–50% of all non-syndromic hereditary deafness cases. The malformed organ of Corti (OC) was observed in different Cx26-null mouse models, which was mainly caused by the developmental arrest of pillar cells (PCs). However, the mechanism of developmental abnormalities in PCs caused by Cx26 deletion is still unclear. In this study, the ultrastructure of PCs at different postnatal days was observed in Cx26-null mice. Knockout of cochlear Cx26 led to the malformed assembly of non-centrosomal microtubule-organizing centers (MTOCs) far from the centrosome rather than near the centrosome. Additionally, the microtubule (MT) arrays emitted by abnormal non-centrosomal MTOCs were significantly reduced. In addition, we found that the protein expression of calmodulin-regulated, spectrin-associated protein2 (camsap2), a microtubule minus-end targeting protein associated with the organization of non-centrosomal MTs, was decreased in juvenile PCs in the Cx26-null group. Our results indicated that the malformation of non-centrosomal MTOCs in cochlear PCs might lead to the corresponding MTs’ failure to be captured and anchored in Cx26-null mice, which results in the deformity of OC. Additionally, this abnormal developmental process might be correlated with the reduced expression of camsap2 caused by Cx26 deletion in the early developmental stage.
Collapse
Affiliation(s)
- Yue Qiu
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.Q.); (K.X.); (L.X.)
| | - Kai Xu
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.Q.); (K.X.); (L.X.)
- Department of Otolaryngology, Nanchang University, Nanchang 330006, China
| | - Le Xie
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.Q.); (K.X.); (L.X.)
| | - Sen Chen
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.Q.); (K.X.); (L.X.)
- Correspondence: (S.C.); (Y.S.); Tel.: +86-27-8535-0870 (S.C.); +86-27-8535-1632 (Y.S.)
| | - Yu Sun
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.Q.); (K.X.); (L.X.)
- Correspondence: (S.C.); (Y.S.); Tel.: +86-27-8535-0870 (S.C.); +86-27-8535-1632 (Y.S.)
| |
Collapse
|
28
|
Gao Y, Zhang Q, Zhang S, Yang L, Liu Y, Liu Y, Wang T. A Connexin Gene (GJB3) Mutation in a Chinese Family With Erythrokeratodermia Variabilis, Ichthyosis and Nonsyndromic Hearing Loss: Case Report and Mutations Update. Front Genet 2022; 13:797124. [PMID: 35677558 PMCID: PMC9168653 DOI: 10.3389/fgene.2022.797124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Gap junctions formed by connexins are channels on cytoplasm functioning in ion recycling and homeostasis. Some members of connexin family including connexin 31 are significant components in human skin and cochlea. In clinic, mutations of connexin 31 have been revealed as the cause of a rare hereditary skin disease called erythrokeratodermia variabilis (EKV) and non-syndromic hearing loss (NSHL).Objective: To determine the underlying genetic cause of EKV, ichthyosis and NSHL in three members of a Chinese pedigree and skin histologic characteristics of the EKV patient.Methods: By performing whole exome sequencing (WES), Sanger sequencing and skin biopsy, we demonstrate a Chinese pedigree carrying a mutation of GJB3 with three patients separately diagnosed with EKV, ichthyosis and NSHL.Results: The proband, a 6-year-old Chinese girl, presented with demarcated annular red-brown plaques and hyperkeratotic scaly patches on her trunk and limbs. Her mother has ichthyosis with hyperkeratosis and geographic tongue while her younger brother had NSHL since birth. Mutation analysis revealed all of them carried a heterozygous missense mutation c.293G>A of GJB3. Skin biopsy showed many grain cells with dyskeratosis in the granular layer. Acanthosis, papillomatosis, and a mild superficial perivascular lymphocytic infiltrate were observed.Conclusion: A mutation of GJB3 associated with EKV, ichthyosis and NSHL is reported in this case. The daughter with EKV and the son with NSHL in this Chinese family inherited the mutation from their mother with ichthyosis. The variation of clinical features may involve with genetic, epigenetic and environmental factors.
Collapse
Affiliation(s)
- Yajuan Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Qianli Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shiyu Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Lu Yang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yaping Liu
- Department of Medical Genetics and National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yaping Liu, ; Yuehua Liu, ; Tao Wang,
| | - Yuehua Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
- *Correspondence: Yaping Liu, ; Yuehua Liu, ; Tao Wang,
| | - Tao Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
- *Correspondence: Yaping Liu, ; Yuehua Liu, ; Tao Wang,
| |
Collapse
|
29
|
Connexin Mutations and Hereditary Diseases. Int J Mol Sci 2022; 23:ijms23084255. [PMID: 35457072 PMCID: PMC9027513 DOI: 10.3390/ijms23084255] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 02/01/2023] Open
Abstract
Inherited diseases caused by connexin mutations are found in multiple organs and include hereditary deafness, congenital cataract, congenital heart diseases, hereditary skin diseases, and X-linked Charcot–Marie–Tooth disease (CMT1X). A large number of knockout and knock-in animal models have been used to study the pathology and pathogenesis of diseases of different organs. Because the structures of different connexins are highly homologous and the functions of gap junctions formed by these connexins are similar, connexin-related hereditary diseases may share the same pathogenic mechanism. Here, we analyze the similarities and differences of the pathology and pathogenesis in animal models and find that connexin mutations in gap junction genes expressed in the ear, eye, heart, skin, and peripheral nerves can affect cellular proliferation and differentiation of corresponding organs. Additionally, some dominant mutations (e.g., Cx43 p.Gly60Ser, Cx32 p.Arg75Trp, Cx32 p.Asn175Asp, and Cx32 p.Arg142Trp) are identified as gain-of-function variants in vivo, which may play a vital role in the onset of dominant inherited diseases. Specifically, patients with these dominant mutations receive no benefits from gene therapy. Finally, the complete loss of gap junctional function or altered channel function including permeability (ions, adenosine triphosphate (ATP), Inositol 1,4,5-trisphosphate (IP3), Ca2+, glucose, miRNA) and electric activity are also identified in vivo or in vitro.
Collapse
|
30
|
The Interplay of Cx26, Cx32, Cx37, Cx40, Cx43, Cx45, and Panx1 in Inner-Ear Development of Yotari (dab1−/−) Mice and Humans. Biomedicines 2022; 10:biomedicines10030589. [PMID: 35327391 PMCID: PMC8945117 DOI: 10.3390/biomedicines10030589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 01/01/2023] Open
Abstract
We investigated DAB1-protein deficiency in the inner-ear development of yotari in comparison to humans and wild-type (wt) mice by immunofluorescence for the expression of connexins (Cxs) and the pannexin Panx1. The spatial and temporal dynamics of Cx26, Cx32, Cx37, Cx40, Cx43, Cx45, and Panx1 were determined in the sixth and eighth weeks of human development and at the corresponding mouse embryonic E13.5 and E15.5, in order to examine gap junction intercellular communication (GJIC) and hemichannel formation. The quantification of the area percentage covered by positive signal was performed for the epithelium and mesenchyme of the cochlear and semicircular ducts and is expressed as the mean ± SD. The data were analysed by one-way ANOVA. Almost all of the examined Cxs were significantly decreased in the cochlear and semicircular ducts of yotari compared to wt and humans, except for Cx32, which was significantly higher in yotari. Cx40 dominated in human inner-ear development, while yotari and wt had decreased expression. The Panx1 expression in yotari was significantly lower than that in the wt and human inner ear, except at E13.5 in the mesenchyme of the wt and epithelium and mesenchyme of humans. Our results emphasize the relevance of GJIC during the development of vestibular and cochlear functions, where they can serve as potential therapeutic targets in inner-ear impairments.
Collapse
|
31
|
Lin Q, Guo Q, Zhu M, Zhang J, Chen B, Wu T, Jiang W, Tang W. Application of Nanomedicine in Inner Ear Diseases. Front Bioeng Biotechnol 2022; 9:809443. [PMID: 35223817 PMCID: PMC8873591 DOI: 10.3389/fbioe.2021.809443] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
The treatment of inner ear disorders always remains a challenge for researchers. The presence of various physiological barriers, primarily the blood–labyrinth barrier (BLB), limits the accessibility of the inner ear and hinders the efficacy of various drug therapies. Yet despite recent advances in the cochlea for repair and regeneration, there are currently no pharmacological or biological interventions for hearing loss. Current research focuses on the localized drug-, gene-, and cell-based therapies. Drug delivery based on nanotechnology represents an innovative strategy to improve inner ear treatments. Materials with specific nanostructures not only exhibit a unique ability to encapsulate and transport therapeutics to the inner ear but also endow specific targeting properties to auditory hair cells as well as the stabilization and sustained drug release. Along with this, some alternative routes, like intratympanic drug delivery, can also offer a better means to access the inner ear without exposure to the BLB. This review discusses a variety of nano-based drug delivery systems to the ear for treating inner ear diseases. The main factors affecting the curative efficacy of nanomaterials are also discussed. With a deeper understanding of the link between these crucial factors and the clinical effect of nanomaterials, it paves the way for the optimization of the therapeutic activity of nanocarriers.
Collapse
Affiliation(s)
- Qianyu Lin
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiong Guo
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mingchao Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Juanli Zhang
- Henan Institute of Medical Device Inspection, Zhengzhou, China
| | - Bei Chen
- Department of Otology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tingting Wu
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Wei Jiang, ; Wenxue Tang,
| | - Wenxue Tang
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Wei Jiang, ; Wenxue Tang,
| |
Collapse
|
32
|
Al-Zubidi N, Page JC, Gombos DS, Srivastava A, Appelbaum E, Gidley PW, Chambers MS, Nader ME. Immune-Related Oral, Otologic, and Ocular Adverse Events. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:399-416. [PMID: 34972977 DOI: 10.1007/978-3-030-79308-1_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Emerging immunotherapeutic agents, including immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed cell death protein ligand 1 (PD-L1), have revolutionized cancer treatment. The first immune checkpoint inhibitor (ICI) ipilimumab, an anti-CTLA-4, was approved in 2011. Since then, the US Food and Drug Administration (FDA) has approved more than half a dozen immune checkpoint inhibitors to treat various malignancies. These agents are part of a broader class of chemotherapy agents termed immunotherapy, which selectively target different steps in the immune response cascade to upregulate the body's normal response to cancer. While the effects of traditional chemotherapy are well known, the toxicity profile of emerging immune therapies is not fully elucidated. They have been associated with atypical side effects labeled collectively as immune-related adverse events (irAEs).
Collapse
Affiliation(s)
- Nagham Al-Zubidi
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Cody Page
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan S Gombos
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akanksha Srivastava
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eric Appelbaum
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul W Gidley
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark S Chambers
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marc-Elie Nader
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
33
|
Zhao HB, Liu LM, Yu N, Zhu Y, Mei L, Chen J, Liang C. Efferent neurons control hearing sensitivity and protect hearing from noise through the regulation of gap junctions between cochlear supporting cells. J Neurophysiol 2022; 127:313-327. [PMID: 34907797 PMCID: PMC8759971 DOI: 10.1152/jn.00468.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It is critical for hearing that the descending cochlear efferent system provides a negative feedback to hair cells to regulate hearing sensitivity and protect hearing from noise. The medial olivocochlear (MOC) efferent nerves project to outer hair cells (OHCs) to regulate OHC electromotility, which is an active cochlear amplifier and can increase hearing sensitivity. Here, we report that the MOC efferent nerves also could innervate supporting cells (SCs) in the vicinity of OHCs to regulate hearing sensitivity. MOC nerve fibers are cholinergic, and acetylcholine (ACh) is a primary neurotransmitter. Immunofluorescent staining showed that MOC nerve endings, presynaptic vesicular acetylcholine transporters (VAChTs), and postsynaptic ACh receptors were visible at SCs and in the SC area. Application of ACh in SCs could evoke a typical inward current and reduce gap junctions (GJs) between them, which consequently enhanced the direct effect of ACh on OHCs to shift but not eliminate OHC electromotility. This indirect, GJ-mediated inhibition had a long-lasting influence. In vivo experiments further demonstrated that deficiency of this GJ-mediated efferent pathway decreased the regulation of active cochlear amplification and compromised the protection against noise. In particular, distortion product otoacoustic emission (DPOAE) showed a delayed reduction after noise exposure. Our findings reveal a new pathway for the MOC efferent system via innervating SCs to control active cochlear amplification and hearing sensitivity. These data also suggest that this SC GJ-mediated efferent pathway may play a critical role in long-term efferent inhibition and is required for protection of hearing from noise trauma.NEW & NOTEWORTHY The cochlear efferent system provides a negative feedback to control hair cell activity and hearing sensitivity and plays a critical role in noise protection. We reveal a new efferent control pathway in which medial olivocochlear efferent fibers have innervations with cochlear supporting cells to control their gap junctions, therefore regulating outer hair cell electromotility and hearing sensitivity. This supporting cell gap junction-mediated efferent control pathway is required for the protection of hearing from noise.
Collapse
|
34
|
Arias S, Paradisi I, Hernández A, Kanzler D. Undescribed GJB2 c.35dupG homozygous prelingual distinguished from c.35delG homozygous/compound heterozygous deafs, dwelling a German ancestry Venezuelan isolate. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00159-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Among ten hearing-impaired (HI) families mostly of German descent dwelling the Venezuelan isolate Colonia Tovar, which were initially studied several decades ago to assess the etiology of their profound/prelingual nonsyndromic deafness phenotype, an undescribed genotype/phenotype was found. Forty-eight subjects, including 8 of the still living 143 originally searched with audiograms 4 decades ago, were retested and their DNA collected. A genomic search of 27 loci involved in HI was performed on a randomly chosen prelingual deaf patient. Subsequently, GJB2 sequencing was performed in all subjects from each pedigree. Haplotypes were constructed with five intragenic GJB2 SNPs (rs117685390, rs7994748, rs2274084, rs2274083, and rs3751385). Audiograms performed along 5 decades were compared to evaluate age-related hearing loss in the different genotypes found in the population.
Results
Three prelingual deaf siblings, having the highest recorded symmetrical hearing loss of all the known affected in the isolate, carried the very rare mutation c.35dupG (p.V13Cfs*35) at GJB2 in a homozygous condition. Two additional GJB2 mutations were identified (p.W77R and c.35delG) in the isolate. Allelic disequilibrium in both c.35dupG and p.W77R carriers (with in-phase haplotype T;T;G;A;C) were found, although not so in the 2 other found c.35delG independent haplotypes. A compound heterozygote in trans (c.35delG/c.35dupG) was audiometrically distinguishable from both the c.35dupG and c.35delG homozygotes.
Conclusions
A relatively higher frequency of mutation of c.35dupG found than elsewhere was retrospectively inferred for the ancient population of the Kaiserstuhl region in Germany, having an opposite epidemiological situation to the one found with the contiguous and very frequent c.35delG. Haplotype analysis suggests founder phenomena and independent occurrence, hundreds of generations back in Caucasoid populations for both mutations.
Collapse
|
35
|
Al-janabi AM, Ahmmed HS, Al-Khafaji SM. Connexin 26 (GJB2) gene mutations linked with autosomal recessive non-syndromic sensor neural hearing loss in the Iraqi population. J Med Life 2021; 14:841-846. [PMID: 35126756 PMCID: PMC8811675 DOI: 10.25122/jml-2021-0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/08/2021] [Indexed: 11/19/2022] Open
Abstract
Deafness is a total or partial hearing loss that may appear at any age and with different degrees of severity. Approximately 50% of hearing loss have a genetic origin, and among them, non-syndromic sensorineural deafness represents about 70% of the cases. From them, 80% correspond to autosomal recessive inheritance deafness. Autosomal recessive deafness was not studied enough at the molecular level in Iraq. This study aimed to verify the frequency of three GJB2 mutations in non-syndromic sensorineural deafness in the Iraqi population. The current case-control study was conducted from January 2018 to January 2020. The study included 95 deafness patients (55 males and 40 females) and 110 healthy control group. Age and sex were matched between the two groups. In order to detect c.35delG, 235delC, and 167delT mutations in the GJB2 gene, we employed the PCR-RFLP technique. The c.35delG was the main frequent mutation encountered with the GJB2 gene among patients with autosomal recessive non-syndromic sensorineural hearing loss. Among them, 35 (36.8%) were homozygous, 40 (42.1%) were heterozygous, and 20 (21.1%) were wild genotypes. The second-degree mutation in the GJB2 gene was c.235delC mutation, which from the 95 deaf patients, there were 20 (21.1%) with homozygous, 33 (34.7%) heterozygous, and 42 (44.2%) wild genotypes. None of the 95 deaf patients showed the c.167delT mutation, and no mutations appeared in the control group. Our data concluded that the GJB2 c.35delG and c.235delC gene mutations were the main cause of autosomal recessive non-syndromic sensorineural hearing loss in the Iraqi deaf population.
Collapse
|
36
|
Taukulis IA, Olszewski RT, Korrapati S, Fernandez KA, Boger ET, Fitzgerald TS, Morell RJ, Cunningham LL, Hoa M. Single-Cell RNA-Seq of Cisplatin-Treated Adult Stria Vascularis Identifies Cell Type-Specific Regulatory Networks and Novel Therapeutic Gene Targets. Front Mol Neurosci 2021; 14:718241. [PMID: 34566577 PMCID: PMC8458580 DOI: 10.3389/fnmol.2021.718241] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/17/2021] [Indexed: 11/21/2022] Open
Abstract
The endocochlear potential (EP) generated by the stria vascularis (SV) is necessary for hair cell mechanotransduction in the mammalian cochlea. We sought to create a model of EP dysfunction for the purposes of transcriptional analysis and treatment testing. By administering a single dose of cisplatin, a commonly prescribed cancer treatment drug with ototoxic side effects, to the adult mouse, we acutely disrupt EP generation. By combining these data with single cell RNA-sequencing findings, we identify transcriptional changes induced by cisplatin exposure, and by extension transcriptional changes accompanying EP reduction, in the major cell types of the SV. We use these data to identify gene regulatory networks unique to cisplatin treated SV, as well as the differentially expressed and druggable gene targets within those networks. Our results reconstruct transcriptional responses that occur in gene expression on the cellular level while identifying possible targets for interventions not only in cisplatin ototoxicity but also in EP dysfunction.
Collapse
Affiliation(s)
- Ian A. Taukulis
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Rafal T. Olszewski
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Soumya Korrapati
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Katharine A. Fernandez
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Erich T. Boger
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Tracy S. Fitzgerald
- Mouse Auditory Testing Core Facility, National Institutes of Health, Bethesda, MD, United States
| | - Robert J. Morell
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Lisa L. Cunningham
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Michael Hoa
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
37
|
Abstract
BACKGROUND Keratitis-ichthyosis-deafness (KID) syndrome is characterized by a congenital triad of keratitis, ichthyosis, and deafness, and is most commonly associated with mutations in the gap junction protein beta 2 gene (GJB2) on chromosome 13q11-q12. METHODS Multimodal anterior segment imaging and genetic testing were used to supplement clinical examination findings in the diagnosis and management of a 12-year-old boy with suspected KID syndrome. RESULTS The patient presented with hearing loss, ichthyosis of the face and extremities, and corneal scarring and keratinization. The corneal limbal stem cell population was found to be normal on in vivo confocal microscopy, whereas the basal epithelium of the cornea demonstrated scarring and areas of cellular loss. Screening of GJB2 revealed a presumed pathogenic heterozygous missense mutation, c.148G>A, confirming the diagnosis of KID syndrome. CONCLUSIONS Multimodal imaging including in vivo confocal microscopy suggests that dysfunctional corneal basal epithelium maturation might contribute to the pathophysiology of keratopathy in KID syndrome.
Collapse
|
38
|
Genetic etiology of hereditary hearing loss in the Gulf Cooperation Council countries. Hum Genet 2021; 141:595-605. [PMID: 34338889 DOI: 10.1007/s00439-021-02323-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
The past 30 years have seen an exponential growth concerning the identification of genes and variants responsible for hereditary hearing loss (HL) worldwide. This has led to a huge gain in our understanding of molecular mechanisms of hearing and deafness, which improved diagnosis for populations with hereditary HL. Many communities around the world, especially in the Middle East and North Africa, have a high prevalence of consanguineous marriages. Congenital monogenic conditions, such as recessive HL, are more common in these populations due to high consanguinity rates. Many studies have shown that high rates of consanguinity, endogamy, and first cousin marriages were observed in the six countries of the Gulf Cooperation Council (GCC). The intent of this study is to investigate the etiology of HL in the GCC region. A deep literature review of genes and variants responsible for HL in this region revealed 89 recessive DNA pathogenic variants reported in 138 cases/familial cases. A total of 21 genes responsible for non-syndromic hearing loss (NSHL) and 17 genes associated with syndromic hearing loss (SHL) were reported in cases from the GCC region. Out of 156 reported affected cases, 112 showed HL only, and 44 showed HL associated with other clinical manifestations. This data suggests that in the GCC region 72% of HL forms are non-syndromic and 28% are syndromic. For individuals with NSHL, 66% of variants were detected in four genes (GJB2, OTOF, TMC1 and CDH23), with a predominance of variants located in the GJB2 gene (37.5%). However, among SHL, Usher syndrome was the more frequent as it has been observed in 41% of the reported syndromic GCC cases. Finally, our analysis showed that HL genetics testing and research in the GCC region took advantage of the next generation sequencing (NGS)-based techniques, as approximately 58% of reported variants were identified using this technology.
Collapse
|
39
|
Lin YH, Wu PC, Tsai CY, Lin YH, Lo MY, Hsu SJ, Lin PH, Erdenechuluun J, Wu HP, Hsu CJ, Wu CC, Chen PL. Hearing Impairment with Monoallelic GJB2 Variants: A GJB2 Cause or Non-GJB2 Cause? J Mol Diagn 2021; 23:1279-1291. [PMID: 34325055 DOI: 10.1016/j.jmoldx.2021.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/23/2021] [Accepted: 07/07/2021] [Indexed: 12/26/2022] Open
Abstract
Recessive variants in GJB2 are the most common genetic cause of sensorineural hearing impairment. However, in many patients, only one variant in the GJB2 coding region is identified using conventional sequencing strategy (eg, Sanger sequencing), resulting in nonconfirmative diagnosis. Conceivably, there might be other unidentified pathogenic variants in the noncoding region of GJB2 or other deafness-causing genes in these patients. To address this, a next-generation sequencing-based diagnostic panel targeting the entire GJB2 gene and the coding regions of 158 other known deafness-causing genes was designed and applied to 95 patients with nonsyndromic sensorineural hearing impairment (including 81 Han Taiwanese and 14 Mongolian patients) in whom only a single GJB2 variant had been detected using conventional Sanger sequencing. The panel confirmed the genetic diagnosis in 24 patients (25.3%). Twenty-two of them had causative variants in several deafness-causing genes other than GJB2, including MYO15A, MYO7A, TECTA, POU4F3, KCNQ4, SLC26A4, OTOF, MT-RNR1, MITF, WFS1, and USH2A. The other two patients had causative variants in GJB2, including a Taiwanese patient with a mosaic maternal uniparental disomy c.235delC variant (approximately 69% mosaicism) and a Mongolian patient with compound heterozygous c.35dupG and c.35delG variants, which occurred at the same site. This study demonstrates the utility of next-generation sequencing in clarifying the genetic diagnosis of hearing-impaired patients with nonconfirmative GJB2 genotypes on conventional genetic examinations.
Collapse
Affiliation(s)
- Yi-Hsin Lin
- Department of Otolaryngology, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ping-Che Wu
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Cheng-Yu Tsai
- Department of Otolaryngology, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institutes of Medical Genomic, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yin-Hung Lin
- Department of Otolaryngology, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institutes of Medical Genomic, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ming-Yu Lo
- Department of Otolaryngology, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Jui Hsu
- Graduate Institutes of Medical Genomic, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Hsuan Lin
- Department of Otolaryngology, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Jargalkhuu Erdenechuluun
- Department of Otolaryngology, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia; The EMJJ Otolaryngology Hospital, Ulaanbaatar, Mongolia
| | - Hung-Pin Wu
- Department of Otolaryngology Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chuan-Jen Hsu
- Department of Otolaryngology, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Department of Otolaryngology Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Pei-Lung Chen
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institutes of Medical Genomic, National Taiwan University College of Medicine, Taipei, Taiwan; Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
40
|
Kohrman D, Borges BC, Cassinotti L, Ji L, Corfas G. Axon-glia interactions in the ascending auditory system. Dev Neurobiol 2021; 81:546-567. [PMID: 33561889 PMCID: PMC9004231 DOI: 10.1002/dneu.22813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022]
Abstract
The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.
Collapse
Affiliation(s)
- David Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Luis Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
41
|
Torkamandi S, Bayat S, Mirfakhraie R, Rezaei S, Askari M, Piltan S, Gholami M. Targeted sequencing of CDH23 and GJB2 genes in an Iranian pedigree with Usher syndrome and non-syndromic hearing loss. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
42
|
Fukunaga I, Oe Y, Danzaki K, Ohta S, Chen C, Shirai K, Kawano A, Ikeda K, Kamiya K. Modeling gap junction beta 2 gene-related deafness with human iPSC. Hum Mol Genet 2021; 30:1429-1442. [PMID: 33997905 DOI: 10.1093/hmg/ddab097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
There are >120 forms of non-syndromic deafness associated with identified genetic loci. In particular, mutation of the gap junction beta 2 gene (GJB2), which encodes connexin (CX)26 protein, is the most frequent cause of hereditary deafness worldwide. We previously described an induction method to develop functional CX26 gap junction-forming cells from mouse-induced pluripotent stem cells (iPSCs) and generated in vitro models for GJB2-related deafness. However, functional CX26 gap junction-forming cells derived from human iPSCs or embryonic stem cells (ESCs) have not yet been reported. In this study, we generated human iPSC-derived functional CX26 gap junction-forming cells (iCX26GJCs), which have the characteristics of cochlear supporting cells. These iCX26GJCs had gap junction plaque-like formations at cell-cell borders and co-expressed several markers that are expressed in cochlear supporting cells. Furthermore, we generated iCX26GJCs derived from iPSCs from two patients with the most common GJB2 mutation in Asia, and these cells reproduced the pathology of GJB2-related deafness. These in vitro models may be useful for establishing optimal therapies and drug screening for various mutations in GJB2-related deafness.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Yoko Oe
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Keiko Danzaki
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Sayaka Ohta
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Cheng Chen
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Kyoko Shirai
- Department of Otolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo 1600023, Japan
| | - Atsushi Kawano
- Department of Otolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo 1600023, Japan
| | - Katsuhisa Ikeda
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| |
Collapse
|
43
|
Fukunaga I, Oe Y, Chen C, Danzaki K, Ohta S, Koike A, Ikeda K, Kamiya K. Activin/Nodal/TGF-β Pathway Inhibitor Accelerates BMP4-Induced Cochlear Gap Junction Formation During in vitro Differentiation of Embryonic Stem Cells. Front Cell Dev Biol 2021; 9:602197. [PMID: 33968919 PMCID: PMC8097046 DOI: 10.3389/fcell.2021.602197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in gap junction beta-2 (GJB2), the gene that encodes connexin 26 (CX26), are the most frequent cause of hereditary deafness worldwide. We recently developed an in vitro model of GJB2-related deafness (induced CX26 gap junction-forming cells; iCX26GJCs) from mouse induced pluripotent stem cells (iPSCs) by using Bone morphogenetic protein 4 (BMP4) signaling-based floating cultures (serum-free culture of embryoid body-like aggregates with quick aggregation cultures; hereafter, SFEBq cultures) and adherent cultures. However, to use these cells as a disease model platform for high-throughput drug screening or regenerative therapy, cell yields must be substantially increased. In addition to BMP4, other factors may also induce CX26 gap junction formation. In the SFEBq cultures, the combination of BMP4 and the Activin/Nodal/TGF-β pathway inhibitor SB431542 (SB) resulted in greater production of isolatable CX26-expressing cell mass (CX26+ vesicles) and higher Gjb2 mRNA levels than BMP4 treatment alone, suggesting that SB may promote BMP4-mediated production of CX26+ vesicles in a dose-dependent manner, thereby increasing the yield of highly purified iCX26GJCs. This is the first study to demonstrate that SB accelerates BMP4-induced iCX26GJC differentiation during stem cell floating culture. By controlling the concentration of SB supplementation in combination with CX26+ vesicle purification, large-scale production of highly purified iCX26GJCs suitable for high-throughput drug screening or regenerative therapy for GJB2-related deafness may be possible.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
44
|
Mahfood M, Chouchen J, Kamal Eddine Ahmad Mohamed W, Al Mutery A, Harati R, Tlili A. Whole exome sequencing, in silico and functional studies confirm the association of the GJB2 mutation p.Cys169Tyr with deafness and suggest a role for the TMEM59 gene in the hearing process. Saudi J Biol Sci 2021; 28:4421-4429. [PMID: 34354426 PMCID: PMC8324942 DOI: 10.1016/j.sjbs.2021.04.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
The development of next generation sequencing techniques has facilitated the detection of mutations at an unprecedented rate. These efficient tools have been particularly beneficial for extremely heterogeneous disorders such as autosomal recessive non-syndromic hearing loss, the most common form of genetic deafness. GJB2 mutations are the most common cause of hereditary hearing loss. Amongst them the NM_004004.5: c.506G > A (p.Cys169Tyr) mutation has been associated with varying severity of hearing loss with unclear segregation patterns. In this study, we report a large consanguineous Emirati family with severe to profound hearing loss fully segregating the GJB2 missense mutation p.Cys169Tyr. Whole exome sequencing (WES), in silico, splicing and expression analyses ruled out the implication of any other variants and confirmed the implication of the p.Cys169Tyr mutation in this deafness family. We also show preliminary murine expression analysis that suggests a link between the TMEM59 gene and the hearing process. The present study improves our understanding of the molecular pathogenesis of hearing loss. It also emphasizes the significance of combining next generation sequencing approaches and segregation analyses especially in the diagnosis of disorders characterized by complex genetic heterogeneity.
Collapse
Key Words
- ARNSHL, autosomal recessive non-syndromic hearing loss
- Actb, Actin beta
- BAM, Binary Alignment Map
- BWA, Burrows-Wheeler Aligner
- C1QTNF9, C1q and TNF related 9
- Cx26, Connexin 26
- ESRRAP2, Estrogen-Related Receptor Alpha Pseudogene 2
- GJB2 gene
- GJB2, Gap Junction Protein Beta 2
- HHLA1, HERV-H LTR-Associating 1
- HL, Hearing loss
- KCNQ3, Potassium Voltage-Gated Channel Subfamily Q Member 3
- Missense mutation
- NGS, next generation sequencing
- NSHL, Non-syndromic hearing loss
- Non-syndromic hearing loss
- PROVEAN, Protein Variation Effect Analyzer
- PolyPhen-2, Polymorphism Phenotyping v2
- RFLP, restriction fragment length polymorphism
- ROH, runs of homozygosity
- RT-PCR, reverse transcription PCR
- RT-qPCR, quantitative reverse transcription PCR
- SAM, Sequence Alignment/Map
- SIFT, Sorting Intolerant From Tolerant
- SJL, Swiss Jim Lambert
- SPATA13, Spermatogenesis Associated 13
- ST3GAL1, ST3 Beta-Galactoside Alpha-2,3-Sialyltransferase 1
- TMEM59, Transmembrane Protein 59
- UAE, United Arab Emirates
- VariMAT, Variation and Mutation Annotation Toolkit
- WES, Whole exome sequencing
- Whole exome sequencing
- dpSNP, Single Nucleotide Polymorphism Database
- gEAR, gene Expression Analysis Resource
- gnomAD, genome aggregation database
- qPCR, quantitative PCR
Collapse
Affiliation(s)
- Mona Mahfood
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Jihen Chouchen
- Human Genetics and Stem Cell Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Walaa Kamal Eddine Ahmad Mohamed
- Human Genetics and Stem Cell Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdullah Al Mutery
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Human Genetics and Stem Cell Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Rania Harati
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Human Genetics and Stem Cell Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
45
|
Hearing Loss Caused by HCMV Infection through Regulating the Wnt and Notch Signaling Pathways. Viruses 2021; 13:v13040623. [PMID: 33917368 PMCID: PMC8067389 DOI: 10.3390/v13040623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 01/27/2023] Open
Abstract
Hearing loss is one of the most prevalent sensory disabilities worldwide with huge social and economic burdens. The leading cause of sensorineural hearing loss (SNHL) in children is congenital cytomegalovirus (CMV) infection. Though the implementation of universal screening and early intervention such as antiviral or anti-inflammatory ameliorate the severity of CMV-associated diseases, direct and targeted therapeutics is still seriously lacking. The major hurdle for it is that the mechanism of CMV induced SNHL has not yet been well understood. In this review, we focus on the impact of CMV infection on the key players in inner ear development including the Wnt and Notch signaling pathways. Investigations on these interactions may gain new insights into viral pathogenesis and reveal novel targets for therapy.
Collapse
|
46
|
Wang J, Song Q. Inhibition of connexin 43 induces hearing loss in postnatal mice. Physiol Int 2021. [PMID: 33769954 DOI: 10.1556/2060.2021.00008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/09/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Connexin 43 (Cx43) is the most ubiquitously expressed member of the family of connexins, constituting gap junctions and mediating cell communication, still its role in hearing loss has been little studied. METHODS Immunohistochemistry was used to detect the expression pattern of Cx43. Spiral ganglia neurons (SGNs) and Corti co-culture were utilized to assay the re-innervation of hair cells by newborn SGNs. Gap19 was utilized to inhibit Cx43 hemichannels. Auditory brainstem responses (ABR) and endocochlear potential (E.P.) were measured to confirm the hearing loss. RESULTS The expression of Cx43 in P14 mice was higher than in P0 and P28 (adult) mice, the earlier time point coinciding with the early inner ear development. Additionally, the growth and synapse generation of fibers were inhibited after Gap 19 treatment of the co-cultures of the Corti and SGNs from newborn mice. Furthermore, the inhibition of Cx43 could increase the ABR threshold and decrease E.P. level in postnatal mice, whereas such an effect was not observed in adult mice. CONCLUSION The function of Cx43 is critical during the early development of mouse cochlea but is dispensable in adult mice.
Collapse
Affiliation(s)
- J Wang
- Department of Otolaryngology, Weifang City Hanting District People's Hospital, Weifang 261100, Shandong, China
| | - Q Song
- Department of Otolaryngology, Weifang City Hanting District People's Hospital, Weifang 261100, Shandong, China
| |
Collapse
|
47
|
Wang H, Gao Y, Guan J, Lan L, Yang J, Xiong W, Zhao C, Xie L, Yu L, Wang D, Wang Q. Phenotypic Heterogeneity of Post-lingual and/or Milder Hearing Loss for the Patients With the GJB2 c.235delC Homozygous Mutation. Front Cell Dev Biol 2021; 9:647240. [PMID: 33718389 PMCID: PMC7953049 DOI: 10.3389/fcell.2021.647240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/08/2021] [Indexed: 01/01/2023] Open
Abstract
Objective To report the phenotypic heterogeneity of GJB2 c.235delC homozygotes associated with post-lingual and/or milder hearing loss, and explore the possible mechanism of these unconditional phenotypes. Methods Mutation screening of GJB2 was performed on all ascertained members from Family 1006983 and three sporadic patients by polymerase chain reaction (PCR) amplification and Sanger sequencing. Next generation sequencing (NGS) was successively performed on some of the affected members and normal controls from Family 1006983 to explore additional possible genetic codes. Reverse transcriptase–quantitative PCR was conducted to test the expression of Connexin30. Results We identified a Chinese autosomal recessive hearing loss family with the GJB2 c.235delC homozygous mutation, affected members from which had post-lingual moderate to profound hearing impairment, and three sporadic patients with post-lingual moderate hearing impairment, instead of congenital profound hearing loss. NGS showed no other particular variants. Overexpression of Connexin30 in some of these cases was verified. Conclusion Post-lingual and/or moderate hearing impairment phenotypes of GJB2 c.235delC homozygotes are not the most common phenotype, revealing the heterogeneity of GJB2 pathogenic mutations. To determine the possible mechanism that rescues part of the hearing or postpones onset age of these cases, more cases are required to confirm both Connexin30 overexpression and the existence of modifier genes.
Collapse
Affiliation(s)
- Hongyang Wang
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Gao
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Jing Guan
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Lan Lan
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Ju Yang
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Wenping Xiong
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Cui Zhao
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Linyi Xie
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Lan Yu
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Dayong Wang
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| | - Qiuju Wang
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) Institute of Otolaryngology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,Key Lab of Hearing Impairment Science of Ministry of Education, Beijing, China.,Key Lab of Hearing Impairment Prevention and Treatment of Beijing, Beijing, China
| |
Collapse
|
48
|
Maslova EA, Orishchenko KE, Posukh OL. Functional Evaluation of a Rare Variant c.516G>C (p.Trp172Cys) in the GJB2 (Connexin 26) Gene Associated with Nonsyndromic Hearing Loss. Biomolecules 2021; 11:61. [PMID: 33466560 PMCID: PMC7824951 DOI: 10.3390/biom11010061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/05/2023] Open
Abstract
Mutations in the GJB2 gene encoding transmembrane protein connexin 26 (Cx26) are the most common cause for hearing loss worldwide. Cx26 plays a crucial role in the ionic and metabolic homeostasis in the inner ear, indispensable for normal hearing process. Different pathogenic mutations in the GJB2 gene can affect all stages of the Cx26 life cycle and result in nonsyndromic autosomal recessive (DFNB1) or dominant (DFNA3) deafness and syndromes associating hearing loss with skin disorders. This study aims to elucidate the functional consequences of a rare GJB2 variant c.516G>C (p.Trp172Cys) found with high frequency in deaf patients from indigenous populations of Southern Siberia (Russia). The substitution c.516G>C leads to the replacement of tryptophan at a conserved amino acid position 172 with cysteine (p.Trp172Cys) in the second extracellular loop of Cx26 protein. We analyzed the subcellular localization of mutant Cx26-p.Trp172Cys protein by immunocytochemistry and the hemichannels permeability by dye loading assay. The GJB2 knockout HeLa cell line has been generated using CRISPR/Cas9 genome editing tool. Subsequently, the HeLa transgenic cell lines stably expressing different GJB2 variants (wild type and mutations associated with hearing loss) were established based on knockout cells and used for comparative functional analysis. The impaired trafficking of mutant Cx26-p.Trp172Cys protein to the plasma membrane and reduced hemichannels permeability support the pathogenic effect of the c.516G>C (p.Trp172Cys) variant and its association with nonsyndromic hearing loss. Our data contribute to a better understanding of the role of mutations in the second extracellular loop of Cx26 protein in pathogenesis of deafness.
Collapse
Affiliation(s)
- Ekaterina A. Maslova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (E.A.M.); (K.E.O.)
- Novosibirsk State University, Novosibirsk 630090, Russia
| | - Konstantin E. Orishchenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (E.A.M.); (K.E.O.)
- Novosibirsk State University, Novosibirsk 630090, Russia
| | - Olga L. Posukh
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (E.A.M.); (K.E.O.)
- Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
49
|
Rashid MH. Molecular simulation of the Kv7.4[ΔS269] mutant channel reveals that ion conduction in the cavity is perturbed due to hydrophobic gating. Biochem Biophys Rep 2020; 25:100879. [PMID: 33367117 PMCID: PMC7749434 DOI: 10.1016/j.bbrep.2020.100879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 11/19/2020] [Accepted: 12/09/2020] [Indexed: 12/05/2022] Open
Abstract
Mutations in the voltage-gated potassium channel Kv7.4 (encoded as KCNQ4) lead to the early onset of non-syndromic hearing loss, which is significant during language acquisition. The deletion of the S269 pore residue (genetic Δ mutation) in Kv7.4 has been reported to be associated with hearing loss. So far, there is no mechanistic understanding of how this mutation modulates channel function. To understand the role of S269 in ion conduction, we performed molecular dynamics simulations for both wild type and ΔS269 mutant channels. Simulations indicate that the ΔS269 mutation suppresses the fluctuations in the neighboring Y269 residue and thereby consolidates the ring formed by I307 and F310 residues in the adjacent S6 helixes in the cavity region. We show that the long side chains of I307 near the entrance to the cavity form a hydrophobic gate. Comparison of the free energy profiles of a cavity ion in Kv7.4 and Kv7.4[ΔS269] channels reveals a sizable energy barrier in the latter case, which suppresses ion conduction. Thus the simulation studies reveal that the hydrophobic gate resulting from the ΔS269 mutation appears to be responsible for sensorineural hearing loss. DeltaS269 mutation in the Kv7.4 channel is associated with hearing loss (SNHL). The mutation effects on channel function are studied via MD simulations. DeltaS269 mutation imposes a constriction at the cavity to suppress K+ conductance. Understanding the mutation effects on channel function will help to treat SNHL.
Collapse
Affiliation(s)
- Md Harunur Rashid
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
- Department of Mathematics and Physics, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| |
Collapse
|
50
|
Peeleman N, Verdoodt D, Ponsaerts P, Van Rompaey V. On the Role of Fibrocytes and the Extracellular Matrix in the Physiology and Pathophysiology of the Spiral Ligament. Front Neurol 2020; 11:580639. [PMID: 33193034 PMCID: PMC7653186 DOI: 10.3389/fneur.2020.580639] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
The spiral ligament in the cochlea has been suggested to play a significant role in the pathophysiology of different etiologies of strial hearing loss. Spiral ligament fibrocytes (SLFs), the main cell type in the lateral wall, are crucial in maintaining the endocochlear potential and regulating blood flow. SLF dysfunction can therefore cause cochlear dysfunction and thus hearing impairment. Recent studies have highlighted the role of SLFs in the immune response of the cochlea. In contrast to sensory cells in the inner ear, SLFs (more specifically type III fibrocytes) have also demonstrated the ability to regenerate after different types of trauma such as drug toxicity and noise. SLFs are responsible for producing proteins, such as collagen and cochlin, that create an adequate extracellular matrix to thrive in. Any dysfunction of SLFs or structural changes to the extracellular matrix can significantly impact hearing function. However, SLFs may prove useful in restoring hearing by their potential to regenerate cells in the spiral ligament.
Collapse
Affiliation(s)
- Noa Peeleman
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Dorien Verdoodt
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Vincent Van Rompaey
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Department of Otorhinolaryngology and Head and Neck Surgery, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|