1
|
Fehrmann MLA, Haer-Wigman L, Kremer H, Yntema HG, Thijssen MEG, Mylanus EAM, Huinck WJ, Lanting CP, Pennings RJE. Cochlear Implantation Outcomes in Genotyped Subjects with Sensorineural Hearing Loss. J Assoc Res Otolaryngol 2025:10.1007/s10162-025-00987-0. [PMID: 40268851 DOI: 10.1007/s10162-025-00987-0] [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: 01/16/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
PURPOSE Cochlear implants (CIs) are an effective rehabilitation option for individuals with severe-to-profound sensorineural hearing loss (SNHL). While genetic factors play a significant role in SNHL, the variability in CI outcomes remains unclear. This study evaluated short- and long-term CI outcomes in a large genotyped cohort and investigated correlations with genetic defects and their cochlear site-of-lesion. METHODS This retrospective, single-center, cohort study included 220 subjects (127 females; 299 ears) with pathogenic variants identified in 31 different nuclear genes and in mitochondrial genes. Audiological outcomes were measured pre- and post-implantation. Cochlear site-of-lesion was categorized as pre-synaptic, post-synaptic, or mitochondrial, based on gene function or expression. Multiple regression analysis assessed factors influencing outcomes, including age at implantation, SNHL duration, hearing aid (HA) use, and cochlear site-of-lesion. RESULTS Results showed a median phoneme score of 90%, with better outcomes in early implantation (≤ 6 years). Variability in outcomes was not linked to cochlear site-of-lesion, but to subject-specific factors, such as age at implantation, duration of SNHL, pre-implantation HA use, and CI experience. A model incorporating these subject-specific factors explained 19% of the total variance in outcomes. Poorer outcomes (phoneme scores < 70%) were more common in individuals with prolonged auditory deprivation or older age at implantation. CONCLUSION Genotyped CI recipients demonstrated excellent outcomes, with variability largely attributed to non-genetic factors. These findings show that cochlear implantation is a beneficial type of rehabilitation for most individuals with hereditary SNHL and underscore the importance of early implantation.
Collapse
Affiliation(s)
- M L A Fehrmann
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - L Haer-Wigman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H Kremer
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H G Yntema
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M E G Thijssen
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - E A M Mylanus
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - W J Huinck
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - C P Lanting
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - R J E Pennings
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| |
Collapse
|
2
|
Kim MJ, Simms S, Behnammanesh G, Chen WW, Honkura Y, Suzuki J, Park HJ, Milani M, Katori Y, Bird JE, Ikeda A, Someya S. A mutation in Tmem135 causes progressive sensorineural hearing loss. Hear Res 2025; 459:109221. [PMID: 39970612 DOI: 10.1016/j.heares.2025.109221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 02/08/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Transmembrane protein 135 (TMEM135) is a highly conserved 52 kDa protein with five predicted transmembrane domains that colocalizes with mitochondria and peroxisomes. Previous studies have shown that TMEM135 is involved in mitochondrial dynamics, thermogenesis, and lipid metabolism across multiple tissues and species; however, its role in the inner ear and auditory system remains unknown. We investigated the function of TMEM135 in hearing using wild-type (WT) and Tmem135FUN025/FUN025 (FUN025) mutant mice on a CBA/CaJ background, a normal-hearing mouse strain. Although FUN025 mice displayed normal auditory brainstem response (ABR) thresholds at 1 month, we observed significantly elevated ABR thresholds at 8, 16, and 64 kHz by 3 months, which progressed to profound hearing loss by 12 months. Consistent with our auditory testing results, 13-month-old FUN025 mice exhibited a severe loss of outer hair cells and more modest changes in inner hair cell survival, spiral ganglion neuron density, and stria vascularis integrity in the cochlea. Our results using BaseScope RNA in situ hybridization indicate that TMEM135 is expressed in the inner hair cells, outer hair cells, supporting cells, and stria vascularis. Using Volocity software and Costes colocalization analysis, we found that TMEM135 closely colocalizes with mitochondria in hair cells. Together, these results demonstrate that the FUN025 mutation in Tmem135 causes progressive sensorineural hearing loss, and suggest that TMEM135 is crucial for maintaining key cochlear cell types and normal sensory function in the aging cochlea.
Collapse
MESH Headings
- Animals
- Hearing Loss, Sensorineural/genetics
- Hearing Loss, Sensorineural/metabolism
- Hearing Loss, Sensorineural/pathology
- Hearing Loss, Sensorineural/physiopathology
- Evoked Potentials, Auditory, Brain Stem
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred CBA
- Mutation
- Spiral Ganglion/metabolism
- Spiral Ganglion/pathology
- Hearing/genetics
- Disease Models, Animal
- Auditory Threshold
- Disease Progression
- Phenotype
- Genetic Predisposition to Disease
- Hair Cells, Auditory, Outer/pathology
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/pathology
- Stria Vascularis/metabolism
- Stria Vascularis/pathology
- Age Factors
- Female
- Male
- Acoustic Stimulation
Collapse
Affiliation(s)
- Mi-Jung Kim
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States
| | - Shion Simms
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States
| | - Ghazaleh Behnammanesh
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, United States
| | - Wei-Wen Chen
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| | - Yohei Honkura
- Department of Otolaryngology-Head &Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Suzuki
- Department of Otolaryngology-Head &Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hyo-Jin Park
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States
| | - Marcus Milani
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States
| | - Yukio Katori
- Department of Otolaryngology-Head &Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jonathan E Bird
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, United States
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, WI, United States
| | - Shinichi Someya
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States; Department of Speech, Language, and Hearing Sciences, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
3
|
Sitko AA, Frank MM, Romero GE, Hunt M, Goodrich LV. Lateral olivocochlear neurons modulate cochlear responses to noise exposure. Proc Natl Acad Sci U S A 2025; 122:e2404558122. [PMID: 39854232 PMCID: PMC11789013 DOI: 10.1073/pnas.2404558122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 12/04/2024] [Indexed: 01/26/2025] Open
Abstract
The sense of hearing originates in the cochlea, which detects sounds across dynamic sensory environments. Like other peripheral organs, the cochlea is subjected to environmental insults, including loud, damage-inducing sounds. In response to internal and external stimuli, the central nervous system directly modulates cochlear function through olivocochlear neurons (OCNs), which are located in the brainstem and innervate the cochlear sensory epithelium. One population of OCNs, the lateral olivocochlear (LOC) neurons, target spiral ganglion neurons (SGNs), the primary sensory neurons of the ear. LOCs alter their transmitter expression for days to weeks in response to noise exposure (NE), suggesting that they could tune SGN excitability over long time periods in response to auditory experience. To examine how LOCs affect auditory function after NE, we characterized OCN transcriptional profiles and found transient LOC-specific gene expression changes after NE, including upregulation of multiple neuropeptide-encoding genes. Next, by generating intersectional mouse lines that selectively target LOCs, we chemogenetically ablated LOCs and assayed auditory responses at baseline and after NE. Compared to controls, mice with reduced LOC innervation showed greater NE-induced functional deficits 1 d later and had worse auditory function after a 2-wk recovery period. The number of remaining presynaptic puncta at the SGN synapse with inner hair cells did not differ between control and LOC-ablated animals, suggesting that the primary role of LOCs after NE is likely not to protect but instead to compensate, ensuring that SGN function is enhanced during periods of need.
Collapse
Affiliation(s)
- Austen A. Sitko
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | | | - Mackenzie Hunt
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Lisa V. Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
4
|
Underhill A, Webb S, Grandi FC, Jeng JY, de Monvel JB, Plion B, Carlton AJ, Amariutei AE, Voulgari N, De Faveri F, Ceriani F, Mustapha M, Johnson SL, Safieddine S, Kros CJ, Marcotti W. MYO7A is required for the functional integrity of the mechanoelectrical transduction complex in hair cells of the adult cochlea. Proc Natl Acad Sci U S A 2025; 122:e2414707122. [PMID: 39746042 PMCID: PMC11725811 DOI: 10.1073/pnas.2414707122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
Myosin-VIIA (MYO7A) is an unconventional myosin responsible for syndromic (Usher 1B) or nonsyndromic forms of deafness in humans when mutated. In the cochlea, MYO7A is expressed in hair cells, where it is believed to act as the motor protein tensioning the mechanoelectrical transducer (MET) channels, thus setting their resting open probability (Po). However, direct evidence for this unique role for an unconventional myosin in mature hair cells is lacking. Here, we show that MYO7A has a distinct role in hair cells, being crucial for the structural integrity of hair bundles. Postnatal deletion of Myo7a leads to 87 to 96% reduction in MYO7A from hair cells by postnatal day 20 (P20), without affecting hearing function. During the following week, mice showed progressive decline in both hearing function and MET current amplitude in hair cells without affecting the resting Po and calcium sensitivity of the MET channel. Hair-bundle stiffness was normal at P20 but halved at P30, despite it having a normal staircase morphology and tip links. The reduction of MYO7A in the stereocilia (>87%) increased their vulnerability to sound-induced damage, with significantly more hearing loss and hair bundle deterioration than in control mice. RNA-sequencing identified a downregulation of several stereociliary genes in the Myo7a-deficient cochlea, indicating the presence of indirect compensatory mechanisms. This study reveals that mature hair cells seem to use a MYO7A-independent mechanism to maintain the resting Po of the MET channels. Instead, MYO7A is essential for maintaining the structural and functional integrity of the hair bundles.
Collapse
Affiliation(s)
- Anna Underhill
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Samuel Webb
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Fiorella C. Grandi
- Sorbonne Université, INSERM, Institute de Myologie, Centre de Researche en Myologie, ParisF-75013, France
| | - Jing-Yi Jeng
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Jacques B. de Monvel
- Université de la Cité de Paris, Institut Pasteur, Assistance publique - Hôpitaux de Paris, Inserm, Fondation pour l’audition, CNRS, Instituts Hospitalo-Universitaires reConnect, ParisF-75012, France
| | - Baptiste Plion
- Université de la Cité de Paris, Institut Pasteur, Assistance publique - Hôpitaux de Paris, Inserm, Fondation pour l’audition, CNRS, Instituts Hospitalo-Universitaires reConnect, ParisF-75012, France
| | - Adam J. Carlton
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Ana E. Amariutei
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Niovi Voulgari
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Francesca De Faveri
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Federico Ceriani
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Mirna Mustapha
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
- Neuroscience Institute, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Stuart L. Johnson
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
- Neuroscience Institute, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Saaid Safieddine
- Université de la Cité de Paris, Institut Pasteur, Assistance publique - Hôpitaux de Paris, Inserm, Fondation pour l’audition, CNRS, Instituts Hospitalo-Universitaires reConnect, ParisF-75012, France
| | - Corné J. Kros
- School of Life Sciences, University of Sussex, Falmer, BrightonBN1 9QG, United Kingdom
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, United Kingdom
- Neuroscience Institute, University of Sheffield, SheffieldS10 2TN, United Kingdom
| |
Collapse
|
5
|
Martin HR, Lysakowski A, Eatock RA. The potassium channel subunit K V1.8 ( Kcna10) is essential for the distinctive outwardly rectifying conductances of type I and II vestibular hair cells. eLife 2024; 13:RP94342. [PMID: 39625061 PMCID: PMC11614384 DOI: 10.7554/elife.94342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
In amniotes, head motions and tilt are detected by two types of vestibular hair cells (HCs) with strikingly different morphology and physiology. Mature type I HCs express a large and very unusual potassium conductance, gK,L, which activates negative to resting potential, confers very negative resting potentials and low input resistances, and enhances an unusual non-quantal transmission from type I cells onto their calyceal afferent terminals. Following clues pointing to KV1.8 (Kcna10) in the Shaker K channel family as a candidate gK,L subunit, we compared whole-cell voltage-dependent currents from utricular HCs of KV1.8-null mice and littermate controls. We found that KV1.8 is necessary not just for gK,L but also for fast-inactivating and delayed rectifier currents in type II HCs, which activate positive to resting potential. The distinct properties of the three KV1.8-dependent conductances may reflect different mixing with other KV subunits that are reported to be differentially expressed in type I and II HCs. In KV1.8-null HCs of both types, residual outwardly rectifying conductances include KV7 (Knq) channels. Current clamp records show that in both HC types, KV1.8-dependent conductances increase the speed and damping of voltage responses. Features that speed up vestibular receptor potentials and non-quantal afferent transmission may have helped stabilize locomotion as tetrapods moved from water to land.
Collapse
Affiliation(s)
- Hannah R Martin
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | - Anna Lysakowski
- Department of Anatomy and Cell Biology, University of Illinois at ChicagoChicagoUnited States
| | - Ruth Anne Eatock
- Department of Neurobiology, University of ChicagoChicagoUnited States
| |
Collapse
|
6
|
Fukuda M, Okanishi H, Ino D, Ono K, Ota T, Wakai E, Sato T, Ohta Y, Kikkawa Y, Inohara H, Kanai Y, Hibino H. Protein profile of mouse endolymph suggests a role in controlling cochlear homeostasis. iScience 2024; 27:111214. [PMID: 39563888 PMCID: PMC11574807 DOI: 10.1016/j.isci.2024.111214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/03/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024] Open
Abstract
The cochlea contains two extracellular fluids, perilymph and endolymph. Endolymph exhibits high potential of approximately +80 to +110 mV (depending on species), which sensitizes sensory hair cells. Other properties of this unique fluid remain elusive, owing to its minuscule volume in rodent cochlea. We therefore developed a technique to collect high-purity endolymph from mouse cochleae. Comprehensive proteomic analysis of sampled endolymph using liquid chromatography with mass spectrometry identified 301 proteins, dominated by molecules engaged in immunity and proteostasis. Approximately 30% of these proteins were undetectable in our perilymph. A combination of mass spectrometry and different approaches revealed that, compared to perilymph, endolymph was enriched with α2-macroglobulin, osteopontin, apolipoprotein D, apolipoprotein E, and apolipoprotein J/clusterin. In other cells or tissues, α2-macroglobulin, apolipoprotein E, and apolipoprotein J contribute to the clearance of degraded proteins from extracellular fluid. Altogether, with the proteins described here, endolymph may play a protective role in stabilizing cochlear homeostasis.
Collapse
Affiliation(s)
- Masatoshi Fukuda
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Daisuke Ino
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuya Ono
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeru Ota
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eri Wakai
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Sato
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yumi Ohta
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiaki Kikkawa
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- AMED-CREST, AMED, Osaka 565-0871, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka 565-0871, Japan
| | - Hiroshi Hibino
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- AMED-CREST, AMED, Osaka 565-0871, Japan
| |
Collapse
|
7
|
Velde HM, Vaseghi-Shanjani M, Smits JJ, Ramakrishnan G, Oostrik J, Wesdorp M, Astuti G, Yntema HG, Hoefsloot L, Lanting CP, Huynen MA, Lehman A, Turvey SE, Pennings RJE, Kremer H. Exome variant prioritization in a large cohort of hearing-impaired individuals indicates IKZF2 to be associated with non-syndromic hearing loss and guides future research of unsolved cases. Hum Genet 2024; 143:1379-1399. [PMID: 39406892 PMCID: PMC11522133 DOI: 10.1007/s00439-024-02706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024]
Abstract
Although more than 140 genes have been associated with non-syndromic hereditary hearing loss (HL), at least half of the cases remain unexplained in medical genetic testing. One reason is that pathogenic variants are located in 'novel' deafness genes. A variant prioritization approach was used to identify novel (candidate) genes for HL. Exome-wide sequencing data were assessed for subjects with presumed hereditary HL that remained unexplained in medical genetic testing by gene-panel analysis. Cases in group AD had presumed autosomal dominantly inherited HL (n = 124), and in group AR, presumed autosomal recessive HL (n = 337). Variants in known and candidate deafness genes were prioritized based on allele frequencies and predicted effects. Selected variants were tested for their co-segregation with HL. Two cases were solved by variants in recently identified deafness genes (ABHD12, TRRAP). Variant prioritization also revealed potentially causative variants in candidate genes associated with recessive and X-linked HL. Importantly, missense variants in IKZF2 were found to co-segregate with dominantly inherited non-syndromic HL in three families. These variants specifically affected Zn2+-coordinating cysteine or histidine residues of the zinc finger motifs 2 and 3 of the encoded protein Helios. This finding indicates a complex genotype-phenotype correlation for IKZF2 defects, as this gene was previously associated with non-syndromic dysfunction of the immune system and ICHAD syndrome, including HL. The designed strategy for variant prioritization revealed that IKZF2 variants can underlie non-syndromic HL. The large number of candidate genes for HL and variants therein stress the importance of inclusion of family members for variant prioritization.
Collapse
Affiliation(s)
- Hedwig M Velde
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Maryam Vaseghi-Shanjani
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Jeroen J Smits
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Jaap Oostrik
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Mieke Wesdorp
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Galuh Astuti
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Helger G Yntema
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Lies Hoefsloot
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cris P Lanting
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboudumc, Nijmegen, The Netherlands
| | - Anna Lehman
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Ronald J E Pennings
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Hannie Kremer
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
8
|
Chokr SM, Bui-Tran A, Cramer KS. Loss of C1q alters the auditory brainstem response. Front Cell Neurosci 2024; 18:1464670. [PMID: 39416682 PMCID: PMC11480778 DOI: 10.3389/fncel.2024.1464670] [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: 07/14/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
Neural circuits in the auditory brainstem compute interaural time and intensity differences used to determine the locations of sound sources. These circuits display features that are specialized for these functions. The projection from the ventral cochlear nucleus (VCN) to the medial nucleus of the trapezoid (MNTB) body travels along highly myelinated fibers and terminates in the calyx of Held. This monoinnervating synapse emerges during development as multiple inputs are eliminated. We previously demonstrated that elimination of microglia with a colony stimulating factor-1 inhibitor results in impaired synaptic pruning so that multiple calyceal terminals reside on principal cells of MNTB. This inhibitor also resulted in impaired auditory brainstem responses (ABRs), with elevated thresholds and increased peak latencies. Loss of the microglial fractalkine receptor, CX3CR1, decreased peak latencies in the ABR. The mechanisms underlying these effects are not known. One prominent microglial signaling pathway involved in synaptic pruning and plasticity during development and aging is the C1q-initiated compliment cascade. Here we investigated the classical complement pathway initiator, C1q, in auditory brainstem maturation. We found that C1q expression is detected in the MNTB by the first postnatal week. C1q levels increased with age and were detected within microglia and surrounding the soma of MNTB principal neurons. Loss of C1q did not affect microglia-dependent calyceal pruning. Excitatory and inhibitory synaptic markers in the MNTB and LSO were not altered with C1q deletion. ABRs showed that C1q KO mice had normal hearing thresholds but shortened peak latencies. Altogether this study uncovers the developmental time frame of C1q expression in the sound localization pathway and shows a subtle functional consequence of C1q knockdown.
Collapse
Affiliation(s)
| | | | - Karina S. Cramer
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
9
|
Cheng C, Zhu G, Wang K, Bu C, Li S, Qiu Y, Lu J, Ji X, Hao W, Wang J, Zhu C, Yang Y, Gu Y, Qian X, Yu C, Gao X. Deletion of Luzp2 Does Not Cause Hearing Loss in Mice. Neurosci Bull 2024; 40:1519-1528. [PMID: 38589712 PMCID: PMC11422540 DOI: 10.1007/s12264-024-01202-5] [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: 09/07/2023] [Accepted: 11/19/2023] [Indexed: 04/10/2024] Open
Abstract
Deafness is the prevailing sensory impairment among humans, impacting every aspect of one's existence. Half of congenital deafness cases are attributed to genetic factors. Studies have shown that Luzp2 is expressed in hair cells (HCs) and supporting cells of the inner ear, but its specific role in hearing remains unclear. To determine the importance of Luzp2 in auditory function, we generated mice deficient in Luzp2. Our results revealed that Luzp2 has predominant expression within the HCs and pillar cells. However, the loss of Luzp2 did not result in any changes in auditory threshold. HCs or synapse number and HC stereocilia morphology in Luzp2 knockout mice did not show any notable distinctions. This was the first study of the role of Luzp2 in hearing in mice, and our results provide important guidance for the screening of deafness genes.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Guangjie Zhu
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Kaijian Wang
- Department of Otorhinolaryngology, Qidong People's Hospital, Qidong Liver Cancer Institute, Affiliated Qidong Hospital of Nantong University, Nantong, 226200, China
| | - Chuan Bu
- The First Affiliated Hospital of Kangda College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Siyu Li
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Yue Qiu
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Jie Lu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Xinya Ji
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Wenli Hao
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Junguo Wang
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Chengwen Zhu
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Ye Yang
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Yajun Gu
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Xiaoyun Qian
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Chenjie Yu
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China.
- Research Institute of Otolaryngology, Nanjing, 210008, China.
| | - Xia Gao
- Department of Otolaryngology-Head and Neck Surgery, Drum Tower Hospital, Affiliated Hospital of the Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China.
- Research Institute of Otolaryngology, Nanjing, 210008, China.
| |
Collapse
|
10
|
Martin HR, Lysakowski A, Eatock RA. The potassium channel subunit K V1.8 ( Kcna10) is essential for the distinctive outwardly rectifying conductances of type I and II vestibular hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.21.563853. [PMID: 38045305 PMCID: PMC10690164 DOI: 10.1101/2023.11.21.563853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
In amniotes, head motions and tilt are detected by two types of vestibular hair cells (HCs) with strikingly different morphology and physiology. Mature type I HCs express a large and very unusual potassium conductance, gK,L, which activates negative to resting potential, confers very negative resting potentials and low input resistances, and enhances an unusual non-quantal transmission from type I cells onto their calyceal afferent terminals. Following clues pointing to KV1.8 (KCNA10) in the Shaker K channel family as a candidate gK,L subunit, we compared whole-cell voltage-dependent currents from utricular hair cells of KV1.8-null mice and littermate controls. We found that KV1.8 is necessary not just for gK,L but also for fast-inactivating and delayed rectifier currents in type II HCs, which activate positive to resting potential. The distinct properties of the three KV1.8-dependent conductances may reflect different mixing with other KV subunits that are reported to be differentially expressed in type I and II HCs. In KV1.8-null HCs of both types, residual outwardly rectifying conductances include KV7 (KCNQ) channels. Current clamp records show that in both HC types, KV1.8-dependent conductances increase the speed and damping of voltage responses. Features that speed up vestibular receptor potentials and non-quantal afferent transmission may have helped stabilize locomotion as tetrapods moved from water to land.
Collapse
Affiliation(s)
| | - Anna Lysakowski
- University of Illinois at Chicago, Department of Anatomy and Cell Biology
| | | |
Collapse
|
11
|
Andersen RE, Alkuraya IF, Ajeesh A, Sakamoto T, Mena EL, Amr SS, Romi H, Kenna MA, Robson CD, Wilch ES, Nalbandian K, Piña-Aguilar R, Walsh CA, Morton CC. Chromosomal structural rearrangements implicate long non-coding RNAs in rare germline disorders. Hum Genet 2024; 143:921-938. [PMID: 39060644 PMCID: PMC11294402 DOI: 10.1007/s00439-024-02693-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
In recent years, there has been increased focus on exploring the role the non-protein-coding genome plays in Mendelian disorders. One class of particular interest is long non-coding RNAs (lncRNAs), which has recently been implicated in the regulation of diverse molecular processes. However, because lncRNAs do not encode protein, there is uncertainty regarding what constitutes a pathogenic lncRNA variant, and thus annotating such elements is challenging. The Developmental Genome Anatomy Project (DGAP) and similar projects recruit individuals with apparently balanced chromosomal abnormalities (BCAs) that disrupt or dysregulate genes in order to annotate the human genome. We hypothesized that rearrangements disrupting lncRNAs could be the underlying genetic etiology for the phenotypes of a subset of these individuals. Thus, we assessed 279 cases with BCAs and selected 191 cases with simple BCAs (breakpoints at only two genomic locations) for further analysis of lncRNA disruptions. From these, we identified 66 cases in which the chromosomal rearrangements directly disrupt lncRNAs. In 30 cases, no genes of any other class aside from lncRNAs are directly disrupted, consistent with the hypothesis that lncRNA disruptions could underly the phenotypes of these individuals. Strikingly, the lncRNAs MEF2C-AS1 and ENSG00000257522 are each disrupted in two unrelated cases. Furthermore, we experimentally tested the lncRNAs TBX2-AS1 and MEF2C-AS1 and found that knockdown of these lncRNAs resulted in decreased expression of the neighboring transcription factors TBX2 and MEF2C, respectively. To showcase the power of this genomic approach for annotating lncRNAs, here we focus on clinical reports and genetic analysis of seven individuals with likely developmental etiologies due to lncRNA disruptions.
Collapse
Affiliation(s)
- Rebecca E Andersen
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ibrahim F Alkuraya
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Abna Ajeesh
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tyler Sakamoto
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Elijah L Mena
- Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Sami S Amr
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Hila Romi
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Margaret A Kenna
- Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, USA
| | - Caroline D Robson
- Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, USA
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Ellen S Wilch
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Katarena Nalbandian
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Raul Piña-Aguilar
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Cynthia C Morton
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- University of Manchester, Manchester Center for Audiology and Deafness, Manchester, UK.
| |
Collapse
|
12
|
Andersen RE, Alkuraya IF, Ajeesh A, Sakamoto T, Mena EL, Amr SS, Romi H, Kenna MA, Robson CD, Wilch ES, Nalbandian K, Piña-Aguilar R, Walsh CA, Morton CC. Rare germline disorders implicate long non-coding RNAs disrupted by chromosomal structural rearrangements. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.16.24307499. [PMID: 38946951 PMCID: PMC11213069 DOI: 10.1101/2024.06.16.24307499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
In recent years, there has been increased focus on exploring the role the non-protein-coding genome plays in Mendelian disorders. One class of particular interest is long non-coding RNAs (lncRNAs), which has recently been implicated in the regulation of diverse molecular processes. However, because lncRNAs do not encode protein, there is uncertainty regarding what constitutes a pathogenic lncRNA variant, and thus annotating such elements is challenging. The Developmental Genome Anatomy Project (DGAP) and similar projects recruit individuals with apparently balanced chromosomal abnormalities (BCAs) that disrupt or dysregulate genes in order to annotate the human genome. We hypothesized that rearrangements disrupting lncRNAs could be the underlying genetic etiology for the phenotypes of a subset of these individuals. Thus, we assessed 279 cases with BCAs and selected 191 cases with simple BCAs (breakpoints at only two genomic locations) for further analysis of lncRNA disruptions. From these, we identified 66 cases in which the chromosomal rearrangements directly disrupt lncRNAs. Strikingly, the lncRNAs MEF2C-AS1 and ENSG00000257522 are each disrupted in two unrelated cases. Furthermore, in 30 cases, no genes of any other class aside from lncRNAs are directly disrupted, consistent with the hypothesis that lncRNA disruptions could underly the phenotypes of these individuals. To showcase the power of this genomic approach for annotating lncRNAs, here we focus on clinical reports and genetic analysis of two individuals with BCAs and additionally highlight six individuals with likely developmental etiologies due to lncRNA disruptions.
Collapse
Affiliation(s)
- Rebecca E. Andersen
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ibrahim F. Alkuraya
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Abna Ajeesh
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Tyler Sakamoto
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Elijah L. Mena
- Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sami S. Amr
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hila Romi
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Margaret A. Kenna
- Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA, USA
| | - Caroline D. Robson
- Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA, USA
- Department of Radiology, Boston Children’s Hospital, Boston, MA, USA
| | - Ellen S. Wilch
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Katarena Nalbandian
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Raul Piña-Aguilar
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Cynthia C. Morton
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- University of Manchester, Manchester Center for Audiology and Deafness, UK
| |
Collapse
|
13
|
Polesskaya O, Boussaty E, Cheng R, Lamonte O, Zhou T, Du E, Sanches TM, Nguyen KM, Okamoto M, Palmer AA, Friedman R. Genome-wide association study for age-related hearing loss in CFW mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598304. [PMID: 38915500 PMCID: PMC11195089 DOI: 10.1101/2024.06.10.598304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Age-related hearing impairment is the most common cause of hearing loss and is one of the most prevalent conditions affecting the elderly globally. It is influenced by a combination of environmental and genetic factors. The mouse and human inner ears are functionally and genetically homologous. Investigating the genetic basis of age-related hearing loss (ARHL) in an outbred mouse model may lead to a better understanding of the molecular mechanisms of this condition. We used Carworth Farms White (CFW) outbred mice, because they are genetically diverse and exhibit variation in the onset and severity of ARHL. The goal of this study was to identify genetic loci involved in regulating ARHL. Hearing at a range of frequencies was measured using Auditory Brainstem Response (ABR) thresholds in 946 male and female CFW mice at the age of 1, 6, and 10 months. We obtained genotypes at 4.18 million single nucleotide polymorphisms (SNP) using low-coverage (mean coverage 0.27x) whole-genome sequencing followed by imputation using STITCH. To determine the accuracy of the genotypes we sequenced 8 samples at >30x coverage and used calls from those samples to estimate the discordance rate, which was 0.45%. We performed genetic analysis for the ABR thresholds for each frequency at each age, and for the time of onset of deafness for each frequency. The SNP heritability ranged from 0 to 42% for different traits. Genome-wide association analysis identified several regions associated with ARHL that contained potential candidate genes, including Dnah11, Rapgef5, Cpne4, Prkag2, and Nek11. We confirmed, using functional study, that Prkag2 deficiency causes age-related hearing loss at high frequency in mice; this makes Prkag2 a candidate gene for further studies. This work helps to identify genetic risk factors for ARHL and to define novel therapeutic targets for the treatment and prevention of ARHL.
Collapse
Affiliation(s)
- Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ely Boussaty
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Olivia Lamonte
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Thomas Zhou
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Eric Du
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mika Okamoto
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rick Friedman
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
14
|
Takahashi S, Zhou Y, Cheatham MA, Homma K. The frequency dependence of prestin-mediated fast electromotility for mammalian cochlear amplification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595389. [PMID: 38826260 PMCID: PMC11142200 DOI: 10.1101/2024.05.22.595389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Prestin's voltage-driven motor activity confers sound-elicited somatic electromotility in auditory outer hair cells (OHCs) and is essential for the exquisite sensitivity and frequency selectivity of mammalian hearing. Lack of prestin results in hearing threshold shifts across frequency, supporting the causal association of variants in the prestin-coding gene, SLC26A5 , with human hearing loss, DFNB61. However, cochlear function can tolerate reductions in prestin-mediated OHC electromotility. We found that two deafness-associated prestin variants, p.A100T and p.P119S, do not deprive prestin of its fast motor function but significantly reduce membrane expression, leading to large reductions in OHC electromotility that were only ∼30% of wildtype (WT). Mice harboring these missense variants suffered congenital hearing loss that was worse at high frequencies; however, they retained WT-like auditory brainstem response thresholds at 8 kHz, which is processed at the apex of the mouse cochlea. This observation suggests the increasing importance of prestin-driven cochlear amplification at higher frequencies relevant to mammalian hearing. The observation also suggests the promising clinical possibility that small enhancements of OHC electromotility could significantly ameliorate DFNB61 hearing loss in human patients. SIGNIFICANCE Prestin is abundantly expressed in the auditory outer hair cells and is essential for normal cochlear operation. Hence, reduction of prestin expression is often taken as indicative of reduced cochlear function in diseased or aged ears. However, this assumption overlooks the fact that cochlear function can tolerate large reductions in prestin motor activity. DFNB61 mouse models generated and characterized in this study provide an opportunity to gauge the amount of prestin motor activity needed to sustain normal hearing sensitivity. This knowledge is crucial not only for understanding the pathogenic roles of deafness-associated variants that impair OHC electromotility but also for unraveling how prestin contributes to cochlear amplification.
Collapse
|
15
|
ShilinLi, Hu Y. Identification of four mitochondria-related genes in sepsis based on RNA sequencing technology. BMC Immunol 2024; 25:32. [PMID: 38755528 PMCID: PMC11097488 DOI: 10.1186/s12865-024-00623-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/13/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVES The purpose of this study was to identify and analyze the mitochondrial genes associated with sepsis patients in order to elucidate the underlying mechanism of sepsis immunity and provide new ideas for the clinical treatment of sepsis. METHODS The hospitalized cases of sepsis (n = 20) and systemic inflammatory response syndrome (SIRS) (n = 12) admitted to the Emergency Intensive Care Unit (EICU) of the Affiliated Hospital of Southwest Medical University from January 2019 to December 2019 were collected consecutively. RNA-seq was used to sequence the RNA (mRNA) of peripheral blood cells. Bioinformatics techniques were used to screen and identify differentially expressed RNAs, with an absolute value of fold change (FC) greater than or equal to 1.2 and a false discovery rate (FDR) less than 0.05. At the same time, mitochondrial genes were obtained from the MitoCarta 3.0 database. Differential genes were then intersected with mitochondrial genes. The resulting crossover genes were subjected to GO, KEGG, and PPI analysis. Subsequently, the GSE65682 dataset was downloaded from the GEO database for survival analysis to assess the prognostic value of core genes, and GSE67652 was downloaded for ROC curve analysis to validate the diagnostic value of core genes. Finally, the localization of core genes was clarified through 10X single-cell sequencing. RESULTS The crossing of 314 sepsis differential genes and 1136 mitochondrial genes yielded 28 genes. GO and KEGG analysis showed that the crossover genes were mainly involved in the mitochondrion, mitochondrial matrix, and mitochondrial inner membrane. Survival analysis screened four genes that were significantly negatively associated with the prognosis of sepsis, namely FIS1, FKBP8, GLRX5, and GUK1. A comparison of peripheral blood RNA-seq results between the sepsis group and the SIRS group showed that the expression levels of these four genes were significantly decreased in the sepsis group compared to the SIRS group. ROC curve analysis based on GSE67652 indicates these four genes' high sensitivity and specificity for sepsis detection. Additionally, single-cell RNA sequencing found that the core genes were mainly expressed in macrophages, T cells, and B cells. CONCLUSIONS Mitochondria-related genes (FIS1, FKBP8, GLRX5, GUK1) were underexpressed in the sepsis group, negatively correlated with survival, and mainly distributed in immune cells. This finding may guide studying the immune-related mechanisms of sepsis. This study protocol was reviewed by the Ethics Committee of the Affiliated Hospital of Southwest Medical University (ethics number: KY2018029), the clinical trial registration number is ChiCTR1900021261, and the registration date is February 4, 2019.
Collapse
Affiliation(s)
- ShilinLi
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan, China
| | - Yingchun Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan, China.
| |
Collapse
|
16
|
Kim MJ, Simms S, Behnammanesh G, Honkura Y, Suzuki J, Park HJ, Milani M, Katori Y, Bird JE, Ikeda A, Someya S. A Mutation in Tmem135 Causes Progressive Sensorineural Hearing Loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593414. [PMID: 38766120 PMCID: PMC11100813 DOI: 10.1101/2024.05.09.593414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Transmembrane protein 135 (TMEM135) is a 52 kDa protein with five predicted transmembrane domains that is highly conserved across species. Previous studies have shown that TMEM135 is involved in mitochondrial dynamics, thermogenesis, and lipid metabolism in multiple tissues; however, its role in the inner ear or the auditory system is unknown. We investigated the function of TMEM135 in hearing using wild-type (WT) and Tmem135 FUN025/FUN025 ( FUN025 ) mutant mice on a CBA/CaJ background, a normal-hearing mouse strain. Although FUN025 mice displayed normal auditory brainstem response (ABR) at 1 month, we observed significantly elevated ABR thresholds at 8, 16, and 64 kHz by 3 months, which progressed to profound hearing loss by 12 months. Consistent with our auditory testing, 13-month-old FUN025 mice exhibited a severe loss of outer hair cells and spiral ganglion neurons in the cochlea. Our results using BaseScope in situ hybridization indicate that TMEM135 is expressed in the inner hair cells, outer hair cells, and supporting cells. Together, these results demonstrate that the FUN025 mutation in Tmem135 causes progressive sensorineural hearing loss, and suggest that TMEM135 is crucial for maintaining key cochlear cell types and normal sensory function in the aging cochlea.
Collapse
|
17
|
Zhu G, Huang Y, Zhang L, Yan K, Qiu C, He Y, Liu Q, Zhu C, Morín M, Moreno‐Pelayo MÁ, Zhu M, Cao X, Zhou H, Qian X, Xu Z, Chen J, Gao X, Wan G. Cingulin regulates hair cell cuticular plate morphology and is required for hearing in human and mouse. EMBO Mol Med 2023; 15:e17611. [PMID: 37691516 PMCID: PMC10630877 DOI: 10.15252/emmm.202317611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023] Open
Abstract
Cingulin (CGN) is a cytoskeleton-associated protein localized at the apical junctions of epithelial cells. CGN interacts with major cytoskeletal filaments and regulates RhoA activity. However, physiological roles of CGN in development and human diseases are currently unknown. Here, we report a multi-generation family presenting with autosomal dominant non-syndromic hearing loss (ADNSHL) that co-segregates with a CGN heterozygous truncating variant, c.3330delG (p.Leu1110Leufs*17). CGN is normally expressed at the apical cell junctions of the organ of Corti, with enriched localization at hair cell cuticular plates and circumferential belts. In mice, the putative disease-causing mutation results in reduced expression and abnormal subcellular localization of the CGN protein, abolishes its actin polymerization activity, and impairs the normal morphology of hair cell cuticular plates and hair bundles. Hair cell-specific Cgn knockout leads to high-frequency hearing loss. Importantly, Cgn mutation knockin mice display noise-sensitive, progressive hearing loss and outer hair cell degeneration. In summary, we identify CGN c.3330delG as a pathogenic variant for ADNSHL and reveal essential roles of CGN in the maintenance of cochlear hair cell structures and auditory function.
Collapse
Affiliation(s)
- Guang‐Jie Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Yuhang Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Linqing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Keji Yan
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life SciencesShandong UniversityQingdaoChina
| | - Cui Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Yihan He
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Qing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Chengwen Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Matías Morín
- Servicio de GenéticaHospital Universitario Ramón y Cajal, IRYCISMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades RarasInstituto de Salud Carlos III (CB06/07/0048; CIBERER‐ISCIII)MadridSpain
| | - Miguel Ángel Moreno‐Pelayo
- Servicio de GenéticaHospital Universitario Ramón y Cajal, IRYCISMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades RarasInstituto de Salud Carlos III (CB06/07/0048; CIBERER‐ISCIII)MadridSpain
| | - Min‐Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical ScienceNanjing Medical UniversityNanjingChina
| | - Han Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xiaoyun Qian
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life SciencesShandong UniversityQingdaoChina
| | - Jie Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xia Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Guoqiang Wan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| |
Collapse
|
18
|
Kang M, Kim JA, Song MH, Joo SY, Kim SJ, Jang SH, Lee H, Seong JK, Choi JY, Gee HY, Jung J. Novel Variant in CEP250 Causes Protein Mislocalization and Leads to Nonsyndromic Autosomal Recessive Type of Progressive Hearing Loss. Cells 2023; 12:2328. [PMID: 37759551 PMCID: PMC10528078 DOI: 10.3390/cells12182328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Genetic hearing loss is the most common hereditary sensorial disorder. Though more than 120 genes associated with deafness have been identified, unveiled causative genes and variants of diverse types of hearing loss remain. Herein, we identified a novel nonsense homozygous variant in CEP250 (c.3511C>T; p.Gln1171Ter) among the family members with progressive moderate sensorineural hearing loss in nonsyndromic autosomal recessive type but without retinal degeneration. CEP250 encodes C-Nap1 protein belonging to the CEP protein family, comprising 30 proteins that play roles in centrosome aggregation and cell cycle progression. The nonsense variant in CEP250 led to the early truncating protein of C-Nap1, which hindered centrosome localization; heterologous expression of CEP250 (c.3511C>T) in NIH3T3 cells within cilia expression condition revealed that the truncating C-Nap1 (p.Gln1171Ter) was not localized at the centrosome but was dispersed in the cytosol. In the murine adult cochlea, Cep250 was expressed in the inner and outer hair cells. Knockout mice of Cep250 showed significant hair cell degeneration and progressive hearing loss in auditory brainstem response. In conclusion, a nonsense variant in CEP250 results in a deficit of centrosome localization and hair cell degeneration in the cochlea, which is associated with the progression of hearing loss in humans and mice.
Collapse
Affiliation(s)
- Minjin Kang
- Department of Otorhinolaryngology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.K.); (J.Y.C.)
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
| | - Jung Ah Kim
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Mee Hyun Song
- Department of Otorhinolaryngology Head and Neck Surgery, Myongji Hospital, Hanyang University College of Medicine, Goyang 04763, Republic of Korea
| | - Sun Young Joo
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Se Jin Kim
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Hyun Jang
- Department of Otorhinolaryngology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.K.); (J.Y.C.)
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si 10408, Republic of Korea;
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | - Jae Young Choi
- Department of Otorhinolaryngology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.K.); (J.Y.C.)
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
| | - Heon Yung Gee
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jinsei Jung
- Department of Otorhinolaryngology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.K.); (J.Y.C.)
- Institute for Lee Won Sang Yonsei Ear Science, Seoul 03722, Republic of Korea (S.Y.J.)
| |
Collapse
|
19
|
Sodero AO, Castagna VC, Elorza SD, Gonzalez-Rodulfo SM, Paulazo MA, Ballestero JA, Martin MG, Gomez-Casati ME. Phytosterols reverse antiretroviral-induced hearing loss, with potential implications for cochlear aging. PLoS Biol 2023; 21:e3002257. [PMID: 37619212 PMCID: PMC10449472 DOI: 10.1371/journal.pbio.3002257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 07/18/2023] [Indexed: 08/26/2023] Open
Abstract
Cholesterol contributes to neuronal membrane integrity, supports membrane protein clustering and function, and facilitates proper signal transduction. Extensive evidence has shown that cholesterol imbalances in the central nervous system occur in aging and in the development of neurodegenerative diseases. In this work, we characterize cholesterol homeostasis in the inner ear of young and aged mice as a new unexplored possibility for the prevention and treatment of hearing loss. Our results show that cholesterol levels in the inner ear are reduced during aging, an effect that is associated with an increased expression of the cholesterol 24-hydroxylase (CYP46A1), the main enzyme responsible for cholesterol turnover in the brain. In addition, we show that pharmacological activation of CYP46A1 with the antiretroviral drug efavirenz reduces the cholesterol content in outer hair cells (OHCs), leading to a decrease in prestin immunolabeling and resulting in an increase in the distortion product otoacoustic emissions (DPOAEs) thresholds. Moreover, dietary supplementation with phytosterols, plant sterols with structure and function similar to cholesterol, was able to rescue the effect of efavirenz administration on the auditory function. Altogether, our findings point towards the importance of cholesterol homeostasis in the inner ear as an innovative therapeutic strategy in preventing and/or delaying hearing loss.
Collapse
Affiliation(s)
- Alejandro O. Sodero
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - Valeria C. Castagna
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas (INGEBI-CONICET), Buenos Aires, Argentina
| | - Setiembre D. Elorza
- Laboratorio de Neurobiología, Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas (INIMEC-CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sara M. Gonzalez-Rodulfo
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - María A. Paulazo
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - Jimena A. Ballestero
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mauricio G. Martin
- Laboratorio de Neurobiología, Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas (INIMEC-CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Eugenia Gomez-Casati
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
20
|
Trigila AP, Castagna VC, Berasain L, Montini D, Rubinstein M, Gomez-Casati ME, Franchini LF. Accelerated Evolution Analysis Uncovers PKNOX2 as a Key Transcription Factor in the Mammalian Cochlea. Mol Biol Evol 2023; 40:msad128. [PMID: 37247388 PMCID: PMC10337857 DOI: 10.1093/molbev/msad128] [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: 09/09/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/31/2023] Open
Abstract
The genetic bases underlying the evolution of morphological and functional innovations of the mammalian inner ear are poorly understood. Gene regulatory regions are thought to play an important role in the evolution of form and function. To uncover crucial hearing genes whose regulatory machinery evolved specifically in mammalian lineages, we mapped accelerated noncoding elements (ANCEs) in inner ear transcription factor (TF) genes and found that PKNOX2 harbors the largest number of ANCEs within its transcriptional unit. Using reporter gene expression assays in transgenic zebrafish, we determined that four PKNOX2-ANCEs drive differential expression patterns when compared with ortholog sequences from close outgroup species. Because the functional role of PKNOX2 in cochlear hair cells has not been previously investigated, we decided to study Pknox2 null mice generated by CRISPR/Cas9 technology. We found that Pknox2-/- mice exhibit reduced distortion product otoacoustic emissions (DPOAEs) and auditory brainstem response (ABR) thresholds at high frequencies together with an increase in peak 1 amplitude, consistent with a higher number of inner hair cells (IHCs)-auditory nerve synapsis observed at the cochlear basal region. A comparative cochlear transcriptomic analysis of Pknox2-/- and Pknox2+/+ mice revealed that key auditory genes are under Pknox2 control. Hence, we report that PKNOX2 plays a critical role in cochlear sensitivity at higher frequencies and that its transcriptional regulation underwent lineage-specific evolution in mammals. Our results provide novel insights about the contribution of PKNOX2 to normal auditory function and to the evolution of high-frequency hearing in mammals.
Collapse
Affiliation(s)
- Anabella P Trigila
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Valeria C Castagna
- Facultad de Medicina, Instituto de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lara Berasain
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Dante Montini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Lucía F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
21
|
Moysan L, Fazekas F, Fekete A, Köles L, Zelles T, Berekméri E. Ca 2+ Dynamics of Gap Junction Coupled and Uncoupled Deiters' Cells in the Organ of Corti in Hearing BALB/c Mice. Int J Mol Sci 2023; 24:11095. [PMID: 37446272 DOI: 10.3390/ijms241311095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
ATP, as a paracrine signalling molecule, induces intracellular Ca2+ elevation via the activation of purinergic receptors on the surface of glia-like cochlear supporting cells. These cells, including the Deiters' cells (DCs), are also coupled by gap junctions that allow the propagation of intercellular Ca2+ waves via diffusion of Ca2+ mobilising second messenger IP3 between neighbouring cells. We have compared the ATP-evoked Ca2+ transients and the effect of two different gap junction (GJ) blockers (octanol and carbenoxolone, CBX) on the Ca2+ transients in DCs located in the apical and middle turns of the hemicochlea preparation of BALB/c mice (P14-19). Octanol had no effect on Ca2+ signalling, while CBX inhibited the ATP response, more prominently in the middle turn. Based on astrocyte models and using our experimental results, we successfully simulated the Ca2+ dynamics in DCs in different cochlear regions. The mathematical model reliably described the Ca2+ transients in the DCs and suggested that the tonotopical differences could originate from differences in purinoceptor and Ca2+ pump expressions and in IP3-Ca2+ release mechanisms. The cochlear turn-dependent effect of CBX might be the result of the differing connexin isoform composition of GJs along the tonotopic axis. The contribution of IP3-mediated Ca2+ signalling inhibition by CBX cannot be excluded.
Collapse
Affiliation(s)
- Louise Moysan
- Department of Zoology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Fruzsina Fazekas
- Department of Zoology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Adam Fekete
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - László Köles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Tibor Zelles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, H-1083 Budapest, Hungary
| | - Eszter Berekméri
- Department of Zoology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| |
Collapse
|
22
|
Jones M, Kovacevic B, Ionescu CM, Wagle SR, Quintas C, Wong EYM, Mikov M, Mooranian A, Al-Salami H. The applications of Targeted Delivery for Gene Therapies in Hearing Loss. J Drug Target 2023:1-22. [PMID: 37211674 DOI: 10.1080/1061186x.2023.2216900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/07/2022] [Accepted: 04/09/2023] [Indexed: 05/23/2023]
Abstract
Gene therapies are becoming more abundantly researched for use in a multitude of potential treatments, including for hearing loss. Hearing loss is a condition which impacts an increasing number of the population each year, with significant burdens associated. As such, this review will present the concept that delivering a gene effectively to the inner ear may assist in expanding novel treatment options and improving patient outcomes. Historically, several drawbacks have been associated with the use of gene therapies, some of which may be overcome via targeted delivery. Targeted delivery has the potential to alleviate off-target effects and permit a safer delivery profile. Viral vectors have often been described as a delivery method, however, there is an emerging depiction of the potential for nanotechnology to be used. Resulting nanoparticles may also be tuned to allow for targeted delivery. Therefore, this review will focus on hearing loss, gene delivery techniques and inner ear targets, including highlighting promising research. Targeted delivery is a key concept to permitting gene delivery in a safe effective manner, however, further research is required, both in the determination of genes to use in functional hearing recovery and formulating nanoparticles for targeted delivery.
Collapse
Affiliation(s)
- Melissa Jones
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Christina Quintas
- School of human sciences, University of Western Australia, Crawley 6009, Perth, Western Australia, Australia
| | - Elaine Y M Wong
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21101 Novi Sad, Serbia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, New Zealand
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
23
|
Takahashi S, Zhou Y, Kojima T, Cheatham MA, Homma K. Prestin's fast motor kinetics is essential for mammalian cochlear amplification. Proc Natl Acad Sci U S A 2023; 120:e2217891120. [PMID: 36893263 PMCID: PMC10089206 DOI: 10.1073/pnas.2217891120] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/06/2023] [Indexed: 03/11/2023] Open
Abstract
Prestin (SLC26A5)-mediated voltage-driven elongations and contractions of sensory outer hair cells within the organ of Corti are essential for mammalian cochlear amplification. However, whether this electromotile activity directly contributes on a cycle-by-cycle basis is currently controversial. By restoring motor kinetics in a mouse model expressing a slowed prestin missense variant, this study provides experimental evidence acknowledging the importance of fast motor action to mammalian cochlear amplification. Our results also demonstrate that the point mutation in prestin disrupting anion transport in other proteins of the SLC26 family does not alter cochlear function, suggesting that the potential weak anion transport of prestin is not essential in the mammalian cochlea.
Collapse
Affiliation(s)
- Satoe Takahashi
- Department of Otolaryngology–Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Yingjie Zhou
- Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL60208
| | - Takashi Kojima
- Department of Otolaryngology–Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Mary Ann Cheatham
- Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL60208
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern University, Evanston, IL60208
| | - Kazuaki Homma
- Department of Otolaryngology–Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern University, Evanston, IL60208
| |
Collapse
|
24
|
Frank MM, Sitko AA, Suthakar K, Torres Cadenas L, Hunt M, Yuk MC, Weisz CJC, Goodrich LV. Experience-dependent flexibility in a molecularly diverse central-to-peripheral auditory feedback system. eLife 2023; 12:e83855. [PMID: 36876911 PMCID: PMC10147377 DOI: 10.7554/elife.83855] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/03/2023] [Indexed: 03/07/2023] Open
Abstract
Brainstem olivocochlear neurons (OCNs) modulate the earliest stages of auditory processing through feedback projections to the cochlea and have been shown to influence hearing and protect the ear from sound-induced damage. Here, we used single-nucleus sequencing, anatomical reconstructions, and electrophysiology to characterize murine OCNs during postnatal development, in mature animals, and after sound exposure. We identified markers for known medial (MOC) and lateral (LOC) OCN subtypes, and show that they express distinct cohorts of physiologically relevant genes that change over development. In addition, we discovered a neuropeptide-enriched LOC subtype that produces Neuropeptide Y along with other neurotransmitters. Throughout the cochlea, both LOC subtypes extend arborizations over wide frequency domains. Moreover, LOC neuropeptide expression is strongly upregulated days after acoustic trauma, potentially providing a sustained protective signal to the cochlea. OCNs are therefore poised to have diffuse, dynamic effects on early auditory processing over timescales ranging from milliseconds to days.
Collapse
Affiliation(s)
- Michelle M Frank
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Austen A Sitko
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Kirupa Suthakar
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication DisordersBethesdaUnited States
| | - Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication DisordersBethesdaUnited States
| | - Mackenzie Hunt
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mary Caroline Yuk
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Catherine JC Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication DisordersBethesdaUnited States
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
25
|
IGF-1 Controls Metabolic Homeostasis and Survival in HEI-OC1 Auditory Cells through AKT and mTOR Signaling. Antioxidants (Basel) 2023; 12:antiox12020233. [PMID: 36829792 PMCID: PMC9952701 DOI: 10.3390/antiox12020233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a trophic factor for the nervous system where it exerts pleiotropic effects, including the regulation of metabolic homeostasis. IGF-1 deficiency induces morphological alterations in the cochlea, apoptosis and hearing loss. While multiple studies have addressed the role of IGF-1 in hearing protection, its potential function in the modulation of otic metabolism remains unclear. Here, we report that "House Ear Institute-organ of Corti 1" (HEI-OC1) auditory cells express IGF-system genes that are regulated during their differentiation. Upon binding to its high-affinity receptor IGF1R, IGF-1 activates AKT and mTOR signaling to stimulate anabolism and, concomitantly, to reduce autophagic catabolism in HEI-OC1 progenitor cells. Notably, IGF-1 stimulation during HEI-OC1 differentiation to mature otic cells sustained both constructive metabolism and autophagic flux, possibly to favor cell remodeling. IGF1R engagement and downstream AKT signaling promoted HEI-OC1 cell survival by maintaining redox balance, even when cells were challenged with the ototoxic agent cisplatin. Our findings establish that IGF-1 not only serves an important function in otic metabolic homeostasis but also activates antioxidant defense mechanisms to promote hair cell survival during the stress response to insults.
Collapse
|
26
|
Kim KS, Koo HY, Bok J. Alternative splicing in shaping the molecular landscape of the cochlea. Front Cell Dev Biol 2023; 11:1143428. [PMID: 36936679 PMCID: PMC10018040 DOI: 10.3389/fcell.2023.1143428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
The cochlea is a complex organ comprising diverse cell types with highly specialized morphology and function. Until now, the molecular underpinnings of its specializations have mostly been studied from a transcriptional perspective, but accumulating evidence points to post-transcriptional regulation as a major source of molecular diversity. Alternative splicing is one of the most prevalent and well-characterized post-transcriptional regulatory mechanisms. Many molecules important for hearing, such as cadherin 23 or harmonin, undergo alternative splicing to produce functionally distinct isoforms. Some isoforms are expressed specifically in the cochlea, while some show differential expression across the various cochlear cell types and anatomical regions. Clinical phenotypes that arise from mutations affecting specific splice variants testify to the functional relevance of these isoforms. All these clues point to an essential role for alternative splicing in shaping the unique molecular landscape of the cochlea. Although the regulatory mechanisms controlling alternative splicing in the cochlea are poorly characterized, there are animal models with defective splicing regulators that demonstrate the importance of RNA-binding proteins in maintaining cochlear function and cell survival. Recent technological breakthroughs offer exciting prospects for overcoming some of the long-standing hurdles that have complicated the analysis of alternative splicing in the cochlea. Efforts toward this end will help clarify how the remarkable diversity of the cochlear transcriptome is both established and maintained.
Collapse
Affiliation(s)
- Kwan Soo Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hei Yeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jinwoong Bok,
| |
Collapse
|
27
|
Lindner LA, Derstroff D, Oliver D, Reimann K. Distribution of ciliary adaptor proteins tubby and TULP3 in the organ of Corti. Front Neurosci 2023; 17:1162937. [PMID: 37144094 PMCID: PMC10151737 DOI: 10.3389/fnins.2023.1162937] [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: 02/10/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Tubby-like proteins are membrane-associated adaptors that mediate directional trafficking into primary cilia. In inner ear sensory epithelia, cilia-including the hair cell's kinocilium-play important roles as organizers of polarity, tissue architecture and cellular function. However, auditory dysfunction in tubby mutant mice was recently found to be related to a non-ciliary function of tubby, the organization of a protein complex in sensory hair bundles of auditory outer hair cells (OHCs). Targeting of signaling components into cilia in the cochlea might therefore rather rely on closely related tubby-like proteins (TULPs). In this study, we compared cellular and subcellular localization of tubby and TULP3 in the mouse inner ear sensory organs. Immunofluorescence microscopy confirmed the previously reported highly selective localization of tubby in the stereocilia tips of OHCs and revealed a previously unnoticed transient localization to kinocilia during early postnatal development. TULP3 was detected in the organ of Corti and vestibular sensory epithelium, where it displayed a complex spatiotemporal pattern. TULP3 localized to kinocilia of cochlear and vestibular hair cells in early postnatal development but disappeared subsequently before the onset of hearing. This pattern suggested a role in targeting ciliary components into kinocilia, possibly related to the developmental processes that shape the sensory epithelia. Concurrent with loss from kinocilia, pronounced TULP3 immunolabeling progressively appeared at microtubule bundles in non-sensory Pillar (PCs) and Deiters cells (DC). This subcellular localization may indicate a novel function of TULP proteins associated with the formation or regulation of microtubule-based cellular structures.
Collapse
Affiliation(s)
- Laura A. Lindner
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Dennis Derstroff
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Katrin Reimann
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
- *Correspondence: Katrin Reimann,
| |
Collapse
|
28
|
Xia A, Udagawa T, Quiñones PM, Atkinson PJ, Applegate BE, Cheng AG, Oghalai JS. The impact of targeted ablation of one row of outer hair cells and Deiters' cells on cochlear amplification. J Neurophysiol 2022; 128:1365-1373. [PMID: 36259670 PMCID: PMC9678430 DOI: 10.1152/jn.00501.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
The mammalian cochlea contains three rows of outer hair cells (OHCs) that amplify the basilar membrane traveling wave with high gain and exquisite tuning. The pattern of OHC loss caused by typical methods of producing hearing loss in animal models (noise, ototoxic exposure, or aging) is variable and not consistent along the length of the cochlea. Thus, it is difficult to use these approaches to understand how forces from multiple OHCs summate to create normal cochlear amplification. Here, we selectively removed the third row of OHCs and Deiters' cells in adult mice and measured cochlear amplification. In the mature cochlear epithelia, expression of the Wnt target gene Lgr5 is restricted to the third row of Deiters' cells, the supporting cells directly underneath the OHCs. Diphtheria toxin administration to Lgr5DTR-EGFP/+ mice selectively ablated the third row of Deiters' cells and the third row of OHCs. Basilar membrane vibration in vivo demonstrated disproportionately lower reduction in cochlear amplification by about 13.5 dB. On a linear scale, this means that the 33% reduction in OHC number led to a 79% reduction in gain. Thus, these experimental data describe the impact of reducing the force of cochlear amplification by a specific amount. Furthermore, these data argue that because OHC forces progressively and sequentially amplify the traveling wave as it travels to its peak, the loss of even a relatively small number of OHCs, when evenly distributed longitudinally, will cause a substantial reduction in cochlear amplification.NEW & NOTEWORTHY Normal cochlear physiology involves force production from three rows of outer hair cells to amplify and tune the traveling wave. Here, we used a genetic approach to target and ablate the third row of outer hair cells in the mouse cochlea and found it reduced cochlear amplification by 79%. This means that the loss of even a relatively small number of OHCs, when evenly distributed, causes a substantial reduction in cochlear amplification.
Collapse
Affiliation(s)
- Anping Xia
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California
| | - Tomokatsu Udagawa
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo, Japan
| | - Patricia M Quiñones
- Caruso Department of Otolaryngology-Head & Neck Surgery, University of Southern California, Los Angeles, California
| | - Patrick J Atkinson
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California
| | - Brian E Applegate
- Caruso Department of Otolaryngology-Head & Neck Surgery, University of Southern California, Los Angeles, California
- Department of Biomedical Engineering, Denney Research Center (DRB) 140, University of Southern California, Los Angeles, California
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California
| | - John S Oghalai
- Caruso Department of Otolaryngology-Head & Neck Surgery, University of Southern California, Los Angeles, California
- Department of Biomedical Engineering, Denney Research Center (DRB) 140, University of Southern California, Los Angeles, California
| |
Collapse
|
29
|
Robles-Bolivar P, Bächinger D, Parra-Perez AM, Román-Naranjo P, Escalera-Balsera A, Gallego-Martinez A, Eckhard AH, Lopez-Escamez JA. A novel nonsense variant in the CENPP gene segregates in a Swiss family with autosomal dominant low-frequency sensorineural hearing loss. Eur J Hum Genet 2022; 30:1301-1305. [PMID: 36071244 PMCID: PMC9626507 DOI: 10.1038/s41431-022-01184-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 02/04/2023] Open
Abstract
Low-frequency sensorineural hearing loss (SNHL) is a rare hearing impairment affecting frequencies below 1000 Hz, previously associated with DIAPH1, WSF1, MYO7A, TNC, SLC26A4 or CCDC50 genes. By exome sequencing, we report a novel nonsense variant in CENPP gene, segregating low-frequency SNHL in five affected members in a Swiss family with autosomal dominant inheritance pattern. Audiological evaluation showed up-sloping audiometric configuration with mild-to-moderate losses below 1000 Hz, that progresses to high-frequencies over time. Protein modeling shows that the variant truncates five amino acids at the end, losing electrostatic interactions that alter protein stability. CENPP gene is expressed in the supporting cells of the organ of Corti and takes part as a subunit of the Constitutive Centromere Associated Network in the kinetochore, that fixes the centromere to the spindle microtubules. We report CENPP as a new candidate gene for low-frequency SNHL. Further functional characterization might enable us to elucidate its molecular role in SNHL.
Collapse
Affiliation(s)
- Paula Robles-Bolivar
- Otology & Neurotology Group CTS 495, Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, 28029, Madrid, Spain
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs, Granada, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18014, Granada, Spain
| | - David Bächinger
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alberto M Parra-Perez
- Otology & Neurotology Group CTS 495, Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, 28029, Madrid, Spain
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs, Granada, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18014, Granada, Spain
- Division of Otolaryngology, Department of Surgery, University of Granada, 18011, Granada, Spain
| | - Pablo Román-Naranjo
- Otology & Neurotology Group CTS 495, Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, 28029, Madrid, Spain
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs, Granada, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18014, Granada, Spain
- Division of Otolaryngology, Department of Surgery, University of Granada, 18011, Granada, Spain
| | - Alba Escalera-Balsera
- Otology & Neurotology Group CTS 495, Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, 28029, Madrid, Spain
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs, Granada, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18014, Granada, Spain
| | - Alvaro Gallego-Martinez
- Otology & Neurotology Group CTS 495, Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, 28029, Madrid, Spain
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs, Granada, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18014, Granada, Spain
| | - Andreas H Eckhard
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jose A Lopez-Escamez
- Otology & Neurotology Group CTS 495, Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain.
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, 28029, Madrid, Spain.
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs, Granada, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18014, Granada, Spain.
- Division of Otolaryngology, Department of Surgery, University of Granada, 18011, Granada, Spain.
| |
Collapse
|
30
|
Gao G, Guo S, Zhang Q, Zhang H, Zhang C, Peng G. Kiaa1024L/Minar2 is essential for hearing by regulating cholesterol distribution in hair bundles. eLife 2022; 11:e80865. [PMID: 36317962 PMCID: PMC9714970 DOI: 10.7554/elife.80865] [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: 06/07/2022] [Accepted: 10/31/2022] [Indexed: 12/05/2022] Open
Abstract
Unbiased genetic screens implicated a number of uncharacterized genes in hearing loss, suggesting some biological processes required for auditory function remain unexplored. Loss of Kiaa1024L/Minar2, a previously understudied gene, caused deafness in mice, but how it functioned in the hearing was unclear. Here, we show that disruption of kiaa1024L/minar2 causes hearing loss in the zebrafish. Defects in mechanotransduction, longer and thinner hair bundles, and enlarged apical lysosomes in hair cells are observed in the kiaa1024L/minar2 mutant. In cultured cells, Kiaa1024L/Minar2 is mainly localized to lysosomes, and its overexpression recruits cholesterol and increases cholesterol labeling. Strikingly, cholesterol is highly enriched in the hair bundle membrane, and loss of kiaa1024L/minar2 reduces cholesterol localization to the hair bundles. Lowering cholesterol levels aggravates, while increasing cholesterol levels rescues the hair cell defects in the kiaa1024L/minar2 mutant. Therefore, cholesterol plays an essential role in hair bundles, and Kiaa1024L/Minar2 regulates cholesterol distribution and homeostasis to ensure normal hearing.
Collapse
Affiliation(s)
- Ge Gao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Shuyu Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Quan Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Hefei Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Cuizhen Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Gang Peng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| |
Collapse
|
31
|
Yasuda SP, Miyasaka Y, Hou X, Obara Y, Shitara H, Seki Y, Matsuoka K, Takahashi A, Wakai E, Hibino H, Takada T, Shiroishi T, Kominami R, Kikkawa Y. Two Loci Contribute to Age-Related Hearing Loss Resistance in the Japanese Wild-Derived Inbred MSM/Ms Mice. Biomedicines 2022; 10:biomedicines10092221. [PMID: 36140322 PMCID: PMC9496148 DOI: 10.3390/biomedicines10092221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
An MSM/Ms strain was established using Japanese wild mice, which exhibit resistance to several phenotypes associated with aging, such as obesity, inflammation, and tumorigenesis, compared to common inbred mouse strains. MSM/Ms strain is resistant to age-related hearing loss, and their auditory abilities are sustained for long durations. The age-related hearing loss 3 (ahl3) locus contributes to age-related hearing in MSM/Ms strain. We generated ahl3 congenic strains by transferring a genomic region on chromosome 17 from MSM/Ms mice into C57BL/6J mice. Although C57BL/6J mice develop age-related hearing loss because of the ahl allele of the cadherin 23 gene, the development of middle- to high-frequency hearing loss was significantly delayed in an ahl3 congenic strain. Moreover, the novel age-related hearing loss 10 (ahl10) locus associated with age-related hearing resistance in MSM/Ms strain was mapped to chromosome 12. Although the resistance effects in ahl10 congenic strain were slightly weaker than those in ahl3 congenic strain, slow progression of age-related hearing loss was confirmed in ahl10 congenic strain despite harboring the ahl allele of cadherin 23. These results suggest that causative genes and polymorphisms of the ahl3 and ahl10 loci are important targets for the prevention and treatment of age-related hearing loss.
Collapse
Affiliation(s)
- Shumpei P. Yasuda
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuki Miyasaka
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Division of Experimental Animals, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Xuehan Hou
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Yo Obara
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Hiroshi Shitara
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
- Laboratory for Transgenic Technology, Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuta Seki
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kunie Matsuoka
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Ai Takahashi
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Eri Wakai
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Hiroshi Hibino
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Toyoyuki Takada
- Integrated Bioresource Information Division, RIKEN BioResource Research Center, Tsukuba 305-0074, Japan
| | | | - Ryo Kominami
- Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Yoshiaki Kikkawa
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- Correspondence:
| |
Collapse
|
32
|
Outer Hair Cell Function is Normal in βV Spectrin Knockout Mice. Hear Res 2022; 423:108564. [DOI: 10.1016/j.heares.2022.108564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022]
|
33
|
Lewis MA, Schulte BA, Dubno JR, Steel KP. Investigating the characteristics of genes and variants associated with self-reported hearing difficulty in older adults in the UK Biobank. BMC Biol 2022; 20:150. [PMID: 35761239 PMCID: PMC9238072 DOI: 10.1186/s12915-022-01349-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Age-related hearing loss is a common, heterogeneous disease with a strong genetic component. More than 100 loci have been reported to be involved in human hearing impairment to date, but most of the genes underlying human adult-onset hearing loss remain unknown. Most genetic studies have focussed on very rare variants (such as family studies and patient cohort screens) or very common variants (genome-wide association studies). However, the contribution of variants present in the human population at intermediate frequencies is hard to quantify using these methods, and as a result, the landscape of variation associated with adult-onset hearing loss remains largely unknown. RESULTS Here we present a study based on exome sequencing and self-reported hearing difficulty in the UK Biobank, a large-scale biomedical database. We have carried out variant load analyses using different minor allele frequency and impact filters, and compared the resulting gene lists to a manually curated list of nearly 700 genes known to be involved in hearing in humans and/or mice. An allele frequency cutoff of 0.1, combined with a high predicted variant impact, was found to be the most effective filter setting for our analysis. We also found that separating the participants by sex produced markedly different gene lists. The gene lists obtained were investigated using gene ontology annotation, functional prioritisation and expression analysis, and this identified good candidates for further study. CONCLUSIONS Our results suggest that relatively common as well as rare variants with a high predicted impact contribute to age-related hearing impairment and that the genetic contributions to adult hearing difficulty may differ between the sexes. Our manually curated list of deafness genes is a useful resource for candidate gene prioritisation in hearing loss.
Collapse
Affiliation(s)
- Morag A Lewis
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK.
| | | | - Judy R Dubno
- The Medical University of South Carolina, Charleston, SC, USA
| | - Karen P Steel
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK
| |
Collapse
|
34
|
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: 11] [Impact Index Per Article: 3.7] [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
|
35
|
Population-scale analysis of common and rare genetic variation associated with hearing loss in adults. Commun Biol 2022; 5:540. [PMID: 35661827 PMCID: PMC9166757 DOI: 10.1038/s42003-022-03408-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/23/2022] [Indexed: 12/29/2022] Open
Abstract
To better understand the genetics of hearing loss, we performed a genome-wide association meta-analysis with 125,749 cases and 469,497 controls across five cohorts. We identified 53/c loci affecting hearing loss risk, including common coding variants in COL9A3 and TMPRSS3. Through exome sequencing of 108,415 cases and 329,581 controls, we observed rare coding associations with 11 Mendelian hearing loss genes, including additive effects in known hearing loss genes GJB2 (Gly12fs; odds ratio [OR] = 1.21, P = 4.2 × 10-11) and SLC26A5 (gene burden; OR = 1.96, P = 2.8 × 10-17). We also identified hearing loss associations with rare coding variants in FSCN2 (OR = 1.14, P = 1.9 × 10-15) and KLHDC7B (OR = 2.14, P = 5.2 × 10-30). Our results suggest a shared etiology between Mendelian and common hearing loss in adults. This work illustrates the potential of large-scale exome sequencing to elucidate the genetic architecture of common disorders where both common and rare variation contribute to risk.
Collapse
|
36
|
García-Añoveros J, Clancy JC, Foo CZ, García-Gómez I, Zhou Y, Homma K, Cheatham MA, Duggan A. Tbx2 is a master regulator of inner versus outer hair cell differentiation. Nature 2022; 605:298-303. [PMID: 35508658 PMCID: PMC9803360 DOI: 10.1038/s41586-022-04668-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 03/21/2022] [Indexed: 01/03/2023]
Abstract
The cochlea uses two types of mechanosensory cell to detect sounds. A single row of inner hair cells (IHCs) synapse onto neurons to transmit sensory information to the brain, and three rows of outer hair cells (OHCs) selectively amplify auditory inputs1. So far, two transcription factors have been implicated in the specific differentiation of OHCs, whereas, to our knowledge, none has been identified in the differentiation of IHCs2-4. One such transcription factor for OHCs, INSM1, acts during a crucial embryonic period to consolidate the OHC fate, preventing OHCs from transdifferentiating into IHCs2. In the absence of INSM1, embryonic OHCs misexpress a core set of IHC-specific genes, which we predict are involved in IHC differentiation. Here we find that one of these genes, Tbx2, is a master regulator of IHC versus OHC differentiation in mice. Ablation of Tbx2 in embryonic IHCs results in their development as OHCs, expressing early OHC markers such as Insm1 and eventually becoming completely mature OHCs in the position of IHCs. Furthermore, Tbx2 is epistatic to Insm1: in the absence of both genes, cochleae generate only OHCs, which suggests that TBX2 is necessary for the abnormal transdifferentiation of INSM1-deficient OHCs into IHCs, as well as for normal IHC differentiation. Ablation of Tbx2 in postnatal, largely differentiated IHCs makes them transdifferentiate directly into OHCs, replacing IHC features with those of mature and not embryonic OHCs. Finally, ectopic expression of Tbx2 in OHCs results in their transdifferentiation into IHCs. Hence, Tbx2 is both necessary and sufficient to make IHCs distinct from OHCs and maintain this difference throughout development.
Collapse
Affiliation(s)
- Jaime García-Añoveros
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Department of Neuroscience, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL, USA.,These authors jointly supervised this work: Jaime García-Añoveros, Anne Duggan.,Correspondence and requests for materials should be addressed to Jaime García-Añoveros or Anne Duggan. ;
| | - John C. Clancy
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Chuan Zhi Foo
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Ignacio García-Gómez
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Yingjie Zhou
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Kazuaki Homma
- Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL, USA.,Department of Otolaryngology–Head and Neck Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Mary Ann Cheatham
- Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL, USA.,Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Anne Duggan
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,These authors jointly supervised this work: Jaime García-Añoveros, Anne Duggan.,Correspondence and requests for materials should be addressed to Jaime García-Añoveros or Anne Duggan. ;
| |
Collapse
|
37
|
ASK1 is a novel molecular target for preventing aminoglycoside-induced hair cell death. J Mol Med (Berl) 2022; 100:797-813. [PMID: 35471608 PMCID: PMC9110505 DOI: 10.1007/s00109-022-02188-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 02/07/2022] [Accepted: 03/03/2022] [Indexed: 10/31/2022]
Abstract
Aminoglycoside antibiotics are lifesaving medicines, crucial for the treatment of chronic or drug resistant infections. However, aminoglycosides are toxic to the sensory hair cells in the inner ear. As a result, aminoglycoside-treated individuals can develop permanent hearing loss and vestibular impairment. There is considerable evidence that reactive oxygen species (ROS) production and the subsequent phosphorylation of c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (P38) drives apoptosis in aminoglycoside-treated hair cells. However, treatment strategies that directly inhibit ROS, JNK, or P38 are limited by the importance of these molecules for normal cellular function. Alternatively, the upstream regulator apoptosis signal-regulating kinase 1 (ASK1/MAP3K5) is a key mediator of ROS-induced JNK and P38 activation under pathologic but not homeostatic conditions. We investigated ASK1 as a mediator of drug-induced hair cell death using cochlear explants from Ask1 knockout mice, demonstrating that Ask1 deficiency attenuates neomycin-induced hair cell death. We then evaluated pharmacological inhibition of ASK1 with GS-444217 as a potential otoprotective therapy. GS-444217 significantly attenuated hair cell death in neomycin-treated explants but did not impact aminoglycoside efficacy against P. aeruginosa in the broth dilution test. Overall, we provide significant pre-clinical evidence that ASK1 inhibition represents a novel strategy for preventing aminoglycoside ototoxicity. KEY MESSAGES: • ASK1 is an upstream, redox-sensitive regulator of P38 and JNK, which are known mediators of hair cell death. • Ask1 knockout does not affect hair cell development in vivo, but significantly reduces aminoglycoside-induced hair cell death in vitro. • A small-molecule inhibitor of ASK1 attenuates neomycin-induced hair cell death, and does not impact antibiotic efficacy in vitro. • ASK1 may be a novel molecular target for preventing aminoglycoside-induced hearing loss.
Collapse
|
38
|
Giffen KP, Li Y, Liu H, Zhao XC, Zhang CJ, Shen RJ, Wang T, Janesick A, Chen BB, Gong SS, Kachar B, Jin ZB, He DZ. Mutation of SLC7A14 causes auditory neuropathy and retinitis pigmentosa mediated by lysosomal dysfunction. SCIENCE ADVANCES 2022; 8:eabk0942. [PMID: 35394837 PMCID: PMC8993119 DOI: 10.1126/sciadv.abk0942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 02/22/2022] [Indexed: 05/13/2023]
Abstract
Lysosomes contribute to cellular homeostasis via processes including macromolecule degradation, nutrient sensing, and autophagy. Defective proteins related to lysosomal macromolecule catabolism are known to cause a range of lysosomal storage diseases; however, it is unclear whether mutations in proteins involved in homeostatic nutrient sensing mechanisms cause syndromic sensory disease. Here, we show that SLC7A14, a transporter protein mediating lysosomal uptake of cationic amino acids, is evolutionarily conserved in vertebrate mechanosensory hair cells and highly expressed in lysosomes of mammalian cochlear inner hair cells (IHCs) and retinal photoreceptors. Autosomal recessive mutation of SLC7A14 caused loss of IHCs and photoreceptors, leading to presynaptic auditory neuropathy and retinitis pigmentosa in mice and humans. Loss-of-function mutation altered protein trafficking and increased basal autophagy, leading to progressive cell degeneration. This study implicates autophagy-lysosomal dysfunction in syndromic hearing and vision loss in mice and humans.
Collapse
Affiliation(s)
- Kimberlee P. Giffen
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University/University of Georgia Medical Partnership, Athens, GA 30602, USA
| | - Yi Li
- Beijing Institute of Otorhinolaryngology, Department of Otorhinolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Xiao-Chang Zhao
- Beijing Institute of Otorhinolaryngology, Department of Otorhinolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Chang-Jun Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing 100730, China
| | - Ren-Juan Shen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing 100730, China
| | - Tianying Wang
- Beijing Institute of Otorhinolaryngology, Department of Otorhinolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Amanda Janesick
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, CA 94305, USA
| | - Bo-Bei Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shu-Sheng Gong
- Department of Otorhinolaryngology-Head and Neck Surgery, Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing 100730, China
| | - David Z. He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| |
Collapse
|
39
|
Oh JK, Vargas Del Valle JG, Lima de Carvalho JR, Sun YJ, Levi SR, Ryu J, Yang J, Nagasaki T, Emanuelli A, Rasool N, Allikmets R, Sparrow JR, Izquierdo NJ, Duncan JL, Mahajan VB, Tsang SH. Expanding the phenotype of TTLL5-associated retinal dystrophy: a case series. Orphanet J Rare Dis 2022; 17:146. [PMID: 35365235 PMCID: PMC8973795 DOI: 10.1186/s13023-022-02295-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inherited retinal dystrophies describe a heterogeneous group of retinal diseases that lead to the irreversible degeneration of rod and cone photoreceptors and eventual blindness. Recessive loss-of-function mutations in Tubulin Tyrosine Ligase Like 5 (TTLL5) represent a recently described cause of inherited cone-rod and cone dystrophy. This study describes the unusual phenotypes of three patients with autosomal recessive mutations in TTLL5. Examination of these patients included funduscopic evaluation, spectral-domain optical coherence tomography, short-wavelength autofluorescence, and full-field electroretinography (ffERG). Genetic diagnoses were confirmed using whole exome capture. Protein modeling of the identified variants was performed to explore potential genotype-phenotype correlations. RESULTS Genetic testing revealed five novel variants in TTLL5 in three unrelated patients with retinal dystrophy. Clinical imaging demonstrated features of sectoral cone-rod dystrophy and cone dystrophy, with phenotypic variability seen across all three patients. One patient also developed high-frequency hearing loss during a similar time period as the onset of retinal disease, potentially suggestive of a syndromic disorder. Retinal structure findings were corroborated with functional measures including ffERG findings that supported these diagnoses. Modeling of the five variants suggest that they cause different effects on protein function, providing a potential reason for genotype-phenotype correlation in these patients. CONCLUSIONS The authors report retinal phenotypic findings in three unrelated patients with novel mutations causing autosomal recessive TTLL5-mediated retinal dystrophy. These findings broaden the understanding of the phenotypes associated with TTLL5-mediated retinal disease and suggest that mutations in TTLL5 should be considered as a potential cause of sectoral retinal dystrophy in addition to cone-rod and cone dystrophies.
Collapse
Affiliation(s)
- Jin Kyun Oh
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- State University of New York at Downstate Medical Center, Brooklyn, NY, USA
| | | | - Jose Ronaldo Lima de Carvalho
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Ophthalmology, Hospital das Clinicas de Pernambuco (HCPE) - Empresa Brasileira de Servicos Hospitalares (EBSERH), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Young Joo Sun
- Omics Laboratory, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Sarah R Levi
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joseph Ryu
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jing Yang
- Omics Laboratory, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Takayuki Nagasaki
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andres Emanuelli
- Department of Ophthalmology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - Nailyn Rasool
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Rando Allikmets
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Janet R Sparrow
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Natalio J Izquierdo
- Department of Surgery, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - Jacque L Duncan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Harkness Eye Institute, Columbia University Medical Center, 635 West 165th Street, Box 212, New York, NY, 10032, USA.
| |
Collapse
|
40
|
Cornman RS, Cryan PM. Positively selected genes in the hoary bat ( Lasiurus cinereus) lineage: prominence of thymus expression, immune and metabolic function, and regions of ancient synteny. PeerJ 2022; 10:e13130. [PMID: 35317076 PMCID: PMC8934532 DOI: 10.7717/peerj.13130] [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: 09/23/2021] [Accepted: 02/25/2022] [Indexed: 01/12/2023] Open
Abstract
Background Bats of the genus Lasiurus occur throughout the Americas and have diversified into at least 20 species among three subgenera. The hoary bat (Lasiurus cinereus) is highly migratory and ranges farther across North America than any other wild mammal. Despite the ecological importance of this species as a major insect predator, and the particular susceptibility of lasiurine bats to wind turbine strikes, our understanding of hoary bat ecology, physiology, and behavior remains poor. Methods To better understand adaptive evolution in this lineage, we used whole-genome sequencing to identify protein-coding sequence and explore signatures of positive selection. Gene models were predicted with Maker and compared to seven well-annotated and phylogenetically representative species. Evolutionary rate analysis was performed with PAML. Results Of 9,447 single-copy orthologous groups that met evaluation criteria, 150 genes had a significant excess of nonsynonymous substitutions along the L. cinereus branch (P < 0.001 after manual review of alignments). Selected genes as a group had biased expression, most strongly in thymus tissue. We identified 23 selected genes with reported immune functions as well as a divergent paralog of Steep1 within suborder Yangochiroptera. Seventeen genes had roles in lipid and glucose metabolic pathways, partially overlapping with 15 mitochondrion-associated genes; these adaptations may reflect the metabolic challenges of hibernation, long-distance migration, and seasonal variation in prey abundance. The genomic distribution of positively selected genes differed significantly from background expectation by discrete Kolmogorov-Smirnov test (P < 0.001). Remarkably, the top three physical clusters all coincided with islands of conserved synteny predating Mammalia, the largest of which shares synteny with the human cat-eye critical region (CECR) on 22q11. This observation coupled with the expansion of a novel Tbx1-like gene family may indicate evolutionary innovation during pharyngeal arch development: both the CECR and Tbx1 cause dosage-dependent congenital abnormalities in thymus, heart, and head, and craniodysmorphy is associated with human orthologs of other positively selected genes as well.
Collapse
|
41
|
Langlie J, Finberg A, Bencie NB, Mittal J, Omidian H, Omidi Y, Mittal R, Eshraghi AA. Recent advancements in cell-based models for auditory disorders. BIOIMPACTS 2022; 12:155-169. [PMID: 35411298 PMCID: PMC8905588 DOI: 10.34172/bi.2022.23900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/09/2021] [Accepted: 11/15/2021] [Indexed: 11/24/2022]
Abstract
![]()
Introduction: Cell-based models play an important role in understanding the pathophysiology and etiology of auditory disorders. For the auditory system, models have primarily focused on restoring inner and outer hair cells. However, they have largely underrepresented the surrounding structures and cells that support the function of the hair cells.
Methods: In this article, we will review recent advancements in the evolution of cell-based models of auditory disorders in their progression towards three dimensional (3D) models and organoids that more closely mimic the pathophysiology in vivo.
Results: With the elucidation of the molecular targets and transcription factors required to generate diverse cell lines of the components of inner ear, research is starting to progress from two dimensional (2D) models to a greater 3D approach. Of note, the 3D models of the inner ear, including organoids, are relatively new and emerging in the field. As 3D models of the inner ear continue to evolve in complexity, their role in modeling disease will grow as they bridge the gap between cell culture and in vivo models.
Conclusion: Using 3D cell models to understand the etiology and molecular mechanisms underlying auditory disorders holds great potential for developing more targeted and effective novel therapeutics.
Collapse
Affiliation(s)
- Jake Langlie
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ariel Finberg
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nathalie B. Bencie
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jeenu Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Hossein Omidian
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Rahul Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Adrien A. Eshraghi
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
42
|
Bharadwaj T, Schrauwen I, Rehman S, Liaqat K, Acharya A, Giese APJ, Nouel-Saied LM, Nasir A, Everard JL, Pollock LM, Zhu S, Bamshad MJ, Nickerson DA, Ali RH, Ullah A, Wali A, Ali G, Santos-Cortez RLP, Ahmed ZM, McDermott BM, Ansar M, Riazuddin S, Ahmad W, Leal SM. ADAMTS1, MPDZ, MVD, and SEZ6: candidate genes for autosomal recessive nonsyndromic hearing impairment. Eur J Hum Genet 2022; 30:22-33. [PMID: 34135477 PMCID: PMC8738740 DOI: 10.1038/s41431-021-00913-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 11/08/2022] Open
Abstract
Hearing impairment (HI) is a common disorder of sensorineural function with a highly heterogeneous genetic background. Although substantial progress has been made in the understanding of the genetic etiology of hereditary HI, many genes implicated in HI remain undiscovered. Via exome and Sanger sequencing of DNA samples obtained from consanguineous Pakistani families that segregate profound prelingual sensorineural HI, we identified rare homozygous missense variants in four genes (ADAMTS1, MPDZ, MVD, and SEZ6) that are likely the underlying cause of HI. Linkage analysis provided statistical evidence that these variants are associated with autosomal recessive nonsyndromic HI. In silico analysis of the mutant proteins encoded by these genes predicted structural, conformational or interaction changes. RNAseq data analysis revealed expression of these genes in the sensory epithelium of the mouse inner ear during embryonic, postnatal, and adult stages. Immunohistochemistry of the mouse cochlear tissue, further confirmed the expression of ADAMTS1, SEZ6, and MPDZ in the neurosensory hair cells of the organ of Corti, while MVD expression was more prominent in the spiral ganglion cells. Overall, supported by in silico mutant protein analysis, animal models, linkage analysis, and spatiotemporal expression profiling in the mouse inner ear, we propose four new candidate genes for HI and expand our understanding of the etiology of HI.
Collapse
Affiliation(s)
- Thashi Bharadwaj
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Sakina Rehman
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Khurram Liaqat
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Anushree Acharya
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Arnaud P J Giese
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Liz M Nouel-Saied
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Abdul Nasir
- Synthetic Protein Engineering Lab (SPEL), Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jenna L Everard
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Lana M Pollock
- Case Western Reserve University, Department of Otolaryngology, Head and Neck Surgery, Cleveland, OH, USA
| | - Shaoyuan Zhu
- Case Western Reserve University, Department of Otolaryngology, Head and Neck Surgery, Cleveland, OH, USA
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Raja Hussain Ali
- Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Asmat Ullah
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abdul Wali
- Department of Biotechnology and Informatics, Faculty of Life Sciences and Informatics, BUITEMS, Quetta, Pakistan
| | - Ghazanfar Ali
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Regie Lyn P Santos-Cortez
- Department of Otolaryngology - Head and Neck Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Brian M McDermott
- Case Western Reserve University, Department of Otolaryngology, Head and Neck Surgery, Cleveland, OH, USA
| | - Muhammad Ansar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Maryland, Baltimore, MD, USA
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA.
- Taub Institute for Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
43
|
Trigila AP, Pisciottano F, Franchini LF. Hearing loss genes reveal patterns of adaptive evolution at the coding and non-coding levels in mammals. BMC Biol 2021; 19:244. [PMID: 34784928 PMCID: PMC8594068 DOI: 10.1186/s12915-021-01170-6] [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: 11/20/2020] [Accepted: 10/21/2021] [Indexed: 11/26/2022] Open
Abstract
Background Mammals possess unique hearing capacities that differ significantly from those of the rest of the amniotes. In order to gain insights into the evolution of the mammalian inner ear, we aim to identify the set of genetic changes and the evolutionary forces that underlie this process. We hypothesize that genes that impair hearing when mutated in humans or in mice (hearing loss (HL) genes) must play important roles in the development and physiology of the inner ear and may have been targets of selective forces across the evolution of mammals. Additionally, we investigated if these HL genes underwent a human-specific evolutionary process that could underlie the evolution of phenotypic traits that characterize human hearing. Results We compiled a dataset of HL genes including non-syndromic deafness genes identified by genetic screenings in humans and mice. We found that many genes including those required for the normal function of the inner ear such as LOXHD1, TMC1, OTOF, CDH23, and PCDH15 show strong signatures of positive selection. We also found numerous noncoding accelerated regions in HL genes, and among them, we identified active transcriptional enhancers through functional enhancer assays in transgenic zebrafish. Conclusions Our results indicate that the key inner ear genes and regulatory regions underwent adaptive evolution in the basal branch of mammals and along the human-specific branch, suggesting that they could have played an important role in the functional remodeling of the cochlea. Altogether, our data suggest that morphological and functional evolution could be attained through molecular changes affecting both coding and noncoding regulatory regions. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01170-6.
Collapse
Affiliation(s)
- Anabella P Trigila
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Francisco Pisciottano
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina.,Current address: Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Lucía F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina.
| |
Collapse
|
44
|
Du H, Zhou H, Sun Y, Zhai X, Chen Z, Wang Y, Xu Z. The Rho GTPase Cell Division Cycle 42 Regulates Stereocilia Development in Cochlear Hair Cells. Front Cell Dev Biol 2021; 9:765559. [PMID: 34746154 PMCID: PMC8570139 DOI: 10.3389/fcell.2021.765559] [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: 08/27/2021] [Accepted: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
Stereocilia are actin-based cell protrusions on the apical surface of inner ear hair cells, playing a pivotal role in hearing and balancing sensation. The development and maintenance of stereocilia is tightly regulated and deficits in this process usually lead to hearing or balancing disorders. The Rho GTPase cell division cycle 42 (CDC42) is a key regulator of the actin cytoskeleton. It has been reported to localize in the hair cell stereocilia and play important roles in stereocilia maintenance. In the present work, we utilized hair cell-specific Cdc42 knockout mice and CDC42 inhibitor ML141 to explore the role of CDC42 in stereocilia development. Our data show that stereocilia height and width as well as stereocilia resorption are affected in Cdc42-deficient cochlear hair cells when examined at postnatal day 8 (P8). Moreover, ML141 treatment leads to planar cell polarity (PCP) deficits in neonatal hair cells. We also show that overexpression of a constitutively active mutant CDC42 in cochlear hair cells leads to enhanced stereocilia developmental deficits. In conclusion, the present data suggest that CDC42 plays a pivotal role in regulating hair cell stereocilia development.
Collapse
Affiliation(s)
- Haibo Du
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Hao Zhou
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yixiao Sun
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiaoyan Zhai
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhengjun Chen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.,School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Yanfei Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, China
| |
Collapse
|
45
|
García-Mato Á, Cervantes B, Murillo-Cuesta S, Rodríguez-de la Rosa L, Varela-Nieto I. Insulin-like Growth Factor 1 Signaling in Mammalian Hearing. Genes (Basel) 2021; 12:genes12101553. [PMID: 34680948 PMCID: PMC8535591 DOI: 10.3390/genes12101553] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a peptide hormone belonging to the insulin family of proteins. Almost all of the biological effects of IGF-1 are mediated through binding to its high-affinity tyrosine kinase receptor (IGF1R), a transmembrane receptor belonging to the insulin receptor family. Factors, receptors and IGF-binding proteins form the IGF system, which has multiple roles in mammalian development, adult tissue homeostasis, and aging. Consequently, mutations in genes of the IGF system, including downstream intracellular targets, underlie multiple common pathologies and are associated with multiple rare human diseases. Here we review the contribution of the IGF system to our understanding of the molecular and genetic basis of human hearing loss by describing, (i) the expression patterns of the IGF system in the mammalian inner ear; (ii) downstream signaling of IGF-1 in the hearing organ; (iii) mouse mutations in the IGF system, including upstream regulators and downstream targets of IGF-1 that inform cochlear pathophysiology; and (iv) human mutations in these genes causing hearing loss.
Collapse
Affiliation(s)
- Ángela García-Mato
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Blanca Cervantes
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Silvia Murillo-Cuesta
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| | - Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| |
Collapse
|
46
|
Liu Q, Zhang L, Zhu MS, Wan G. High-throughput screening on cochlear organoids identifies VEGFR-MEK-TGFB1 signaling promoting hair cell reprogramming. Stem Cell Reports 2021; 16:2257-2273. [PMID: 34525385 PMCID: PMC8452601 DOI: 10.1016/j.stemcr.2021.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 01/06/2023] Open
Abstract
Hair cell degeneration is a major cause of sensorineural hearing loss. Hair cells in mammalian cochlea do not spontaneously regenerate, posing a great challenge for restoration of hearing. Here, we establish a robust, high-throughput cochlear organoid platform that facilitates 3D expansion of cochlear progenitor cells and differentiation of hair cells in a temporally regulated manner. High-throughput screening of the FDA-approved drug library identified regorafenib, a VEGFR inhibitor, as a potent small molecule for hair cell differentiation. Regorafenib also promotes reprogramming and maturation of hair cells in both normal and neomycin-damaged cochlear explants. Mechanistically, inhibition of VEGFR suppresses TGFB1 expression via the MEK pathway and TGFB1 downregulation directly mediates the effect of regorafenib on hair cell reprogramming. Our study not only demonstrates the power of a cochlear organoid platform in high-throughput analyses of hair cell physiology but also highlights VEGFR-MEK-TGFB1 signaling crosstalk as a potential target for hair cell regeneration and hearing restoration. Cochlear organoids can be derived from both LGR5+ and LGR5– supporting cells HTS using cochlear organoids identifies regorafenib for hair cell differentiation Regorafenib promotes hair cell reprogramming and maturation in cochlear explants Regorafenib acts via a NOTCH-independent and VEGFR-MEK-TGFB1-dependent mechanism
Collapse
Affiliation(s)
- Qing Liu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical School, Nanjing University, Nanjing 210032, China
| | - Linqing Zhang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical School, Nanjing University, Nanjing 210032, China
| | - Min-Sheng Zhu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical School, Nanjing University, Nanjing 210032, China
| | - Guoqiang Wan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical School, Nanjing University, Nanjing 210032, China; Research Institute of Otolaryngology, No. 321 Zhongshan Road, 210008 Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210032, China; Institute for Brain Sciences, Nanjing University, Nanjing 210032, China.
| |
Collapse
|
47
|
Qi Y, Xiong W, Yu S, Du Z, Qu T, He L, Wei W, Zhang L, Liu K, Li Y, He DZ, Gong S. Deletion of C1ql1 Causes Hearing Loss and Abnormal Auditory Nerve Fibers in the Mouse Cochlea. Front Cell Neurosci 2021; 15:713651. [PMID: 34512267 PMCID: PMC8424102 DOI: 10.3389/fncel.2021.713651] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/21/2021] [Indexed: 01/19/2023] Open
Abstract
Complement C1q Like 1 (C1QL1), a secreted component of C1Q-related protein, is known to play an important role in synaptic maturation, regulation, and maintenance in the central nervous system. C1ql1 is expressed in adult cochlear inner and outer hair cells (IHCs and OHCs) with preferential expression in OHCs. We generated C1ql1 null mice to examine the role of C1QL1 in the auditory periphery. C1ql1-null mice exhibited progressive hearing loss with elevated thresholds of auditory brainstem response and distortion product otoacoustic emission. Confocal microscopy showed that the number of nerve fibers innervating both IHCs and OHCs was significantly reduced. However, spiral ganglion neurons appeared to be normal under electron microscopy. IHC development and survival were not affected by deletion of C1ql1. Voltage-clamp recording and immunocytochmistry combined with confocal microscopy showed C1ql1-null IHCs showed no significant reduction of pre-synaptic proteins and synaptic vesicle release. This is in contrast to significant OHC loss in the KO mice. Our study suggests that C1ql1 is essential for development of hair cell innervation and OHC survival. But maturation of presynaptic machinery in IHCs does not depend on C1QL1.
Collapse
Affiliation(s)
- Yue Qi
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Xiong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shukui Yu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhengde Du
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tengfei Qu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lu He
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Wei
- Department of Otology, Sheng Jing Hospital, China Medical University, Shenyang, China
| | - Lingjun Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - David Z He
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
| | - Shusheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
48
|
Munnamalai V, Sammudin NH, Young CA, Thawani A, Kuhn RJ, Fekete DM. Embryonic and Neonatal Mouse Cochleae Are Susceptible to Zika Virus Infection. Viruses 2021; 13:v13091823. [PMID: 34578404 PMCID: PMC8472928 DOI: 10.3390/v13091823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022] Open
Abstract
Congenital Zika Syndrome (CZS) is caused by vertical transmission of Zika virus (ZIKV) to the gestating human fetus. A subset of CZS microcephalic infants present with reduced otoacoustic emissions; this test screens for hearing loss originating in the cochlea. This observation leads to the question of whether mammalian cochlear tissues are susceptible to infection by ZIKV during development. To address this question using a mouse model, the sensory cochlea was explanted at proliferative, newly post-mitotic or maturing stages. ZIKV was added for the first 24 h and organs cultured for up to 6 days to allow for cell differentiation. Results showed that ZIKV can robustly infect proliferating sensory progenitors, as well as post-mitotic hair cells and supporting cells. Virus neutralization using ZIKV-117 antibody blocked cochlear infection. AXL is a cell surface molecule known to enhance the attachment of flavivirus to host cells. While Axl mRNA is widely expressed in embryonic cochlear tissues susceptible to ZIKV infection, it is selectively downregulated in the post-mitotic sensory organ by E15.5, even though these cells remain infectible. These findings may offer insights into which target cells could potentially contribute to hearing loss resulting from fetal exposure to ZIKV in humans.
Collapse
Affiliation(s)
- Vidhya Munnamalai
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; (V.M.); (C.A.Y.)
- Graduate School of Biomedical Sciences and Engineering, University of Main, Orono, ME 04469, USA
| | - Nabilah H. Sammudin
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
| | - Caryl A. Young
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; (V.M.); (C.A.Y.)
| | - Ankita Thawani
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
| | - Richard J. Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| | - Donna M. Fekete
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Correspondence:
| |
Collapse
|
49
|
Cochlear hair cells of echolocating bats are immune to intense noise. J Genet Genomics 2021; 48:984-993. [PMID: 34393089 DOI: 10.1016/j.jgg.2021.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/15/2021] [Accepted: 06/06/2021] [Indexed: 11/23/2022]
Abstract
Exposure to intense noise can damage cochlear hair cells, leading to hearing loss in mammals. To avoid this constraint, most mammals have evolved in relatively quiet environments. Echolocating bats, however, are naturally exposed to continuous intense sounds from their own and neighboring sonar emissions for maintaining sonar directionality and range. Here, we propose the presence of intense noise resistance in cochlear hair cells of echolocating bats against noise-induced hearing loss (NIHL). To test this hypothesis, we performed noise exposure experiments for laboratory mice, one nonecholocating bat species, and five echolocating bat species. Contrary to nonecholocating fruit bats and mice, the hearing and the cochlear hair cells of echolocating bats remained unimpaired after continuous intense noise exposure. The comparative analyses of cochleae transcriptomic data showed that several genes protecting cochlear hair cells from intense sounds were overexpressed in echolocating bats. Particularly, the experimental examinations revealed that ISL1 overexpression significantly improved the survival of cochlear hair cells. Our findings support the existence of protective effects in cochlear hair cells of echolocating bats against intense noises, which provides new insight into understanding the relationship between cochlear hair cells and intense noises, and preventing or ameliorating NIHL in mammals.
Collapse
|
50
|
Deletion of Clusterin Protects Cochlear Hair Cells against Hair Cell Aging and Ototoxicity. Neural Plast 2021; 2021:9979157. [PMID: 34194490 PMCID: PMC8181089 DOI: 10.1155/2021/9979157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/15/2021] [Accepted: 04/28/2021] [Indexed: 01/06/2023] Open
Abstract
Hearing loss is a debilitating disease that affects 10% of adults worldwide. Most sensorineural hearing loss is caused by the loss of mechanosensitive hair cells in the cochlea, often due to aging, noise, and ototoxic drugs. The identification of genes that can be targeted to slow aging and reduce the vulnerability of hair cells to insults is critical for the prevention of sensorineural hearing loss. Our previous cell-specific transcriptome analysis of adult cochlear hair cells and supporting cells showed that Clu, encoding a secreted chaperone that is involved in several basic biological events, such as cell death, tumor progression, and neurodegenerative disorders, is expressed in hair cells and supporting cells. We generated Clu-null mice (C57BL/6) to investigate its role in the organ of Corti, the sensory epithelium responsible for hearing in the mammalian cochlea. We showed that the deletion of Clu did not affect the development of hair cells and supporting cells; hair cells and supporting cells appeared normal at 1 month of age. Auditory function tests showed that Clu-null mice had hearing thresholds comparable to those of wild-type littermates before 3 months of age. Interestingly, Clu-null mice displayed less hair cell and hearing loss compared to their wildtype littermates after 3 months. Furthermore, the deletion of Clu is protected against aminoglycoside-induced hair cell loss in both in vivo and in vitro models. Our findings suggested that the inhibition of Clu expression could represent a potential therapeutic strategy for the alleviation of age-related and ototoxic drug-induced hearing loss.
Collapse
|